You are on page 1of 736

Hematology 2021

AMERICAN SOCIETY OF HEMATOLOGY EDUCATION PROGRAM


63rd ASH® Annual Meeting and Exposition

Editors
Adam Cuker, MD, MS
Executive Editor, Hematology 2021
Associate Professor of Medicine
Perelman School of Medicine
University of Pennsylvania
Philadelphia, PA

Mario Cazzola, MD
Deputy Editor, Hematology 2021
Research Group Leader on Myeloid Neoplasms
Fondazione IRCCS Policlinico San Matteo & University of Pavia
Pavia, Italy

Ann LaCasce, MD, MSc


Deputy Editor, Hematology 2021
Associate Professor of Medicine
Dana-Farber Cancer Institute
Boston, MA

Stella Chou, MD
Deputy Editor, Hematology 2021
Associate Professor of Pediatrics
The Children’s Hospital of Philadelphia
University of Pennsylvania
Philadelphia, PA

David Garcia, MD
Special Section Editor, Hematology 2021
Professor of Medicine
University of Washington
Seattle, WA

AMERICAN SOCIETY OF HEMATOLOGY • WASHINGTON, DC

Prakash Singh Shekhawat


©2021 by the American Society of Hematology
ISSN 1520–4383 (online)

Hematology, the ASH Education Program, is published Committee on Educational Affairs


annually by the American Society of Hematology (ASH) in Marc J. Kahn (‘21) (Chair)
one volume per year.
Anjali Advani, MD (‘21)
All business correspondence and purchase and reprint Jennifer R. Brown, MD, PhD (‘21)
requests should be addressed to the American Society of
Eldad Dann, MD (‘21)
Hematology, 2021 L Street, NW, Suite 900, Washington, DC
20036, USA; phone: 202–776–0544; fax: 202–776–0545; Faith Davies, MD (‘24)
e-mail: ASH@hematology.org. James Foran, MD (‘22)
Nobuko Hijiya, MD (‘24)
Hematology, the ASH Education Program, is available on the
Internet at https://ashpublications.org/hematology. Margaret Kasner, MD (‘22)
Kah Poh (Melissa) Loh, MD (‘23)
©2021 by The American Society of Hematology. All rights
reserved. Copyright is not claimed in any works of the United Martha P. Mims, MD (‘23)
States Government. Except as expressly permitted in this Casey L. O’Connell, MD (‘23)
statement, no part of this publication may be used (as here Heather A. O’Leary, PhD (‘23)
in after defined) in any form or by any means now or hereafter Adriana Seber, MD, MS (‘21)
known, electronic or mechanical, without permission in
Andrew D. Zelenetz, MD, PhD (‘21)
writing from the Publisher, The American Society of
Hematology. For purposes of this notice, the term “use”
Ex Offcio Members
includes but is not limited to reproduction, photocopying,
storage in a retrieval system, translation, and transmittal. The Martin S. Tallman, MD (‘21) – President
Publisher hereby consents to the use of this publication or Jane N. Winter, MD (‘22) – President-Elect
any part hereof for any noncommercial educational purpose Robert A. Brodsky, MD (‘23) – Vice President
within the health field such as classroom instruction and
clinical and residency training. This publication or any part Liaisons
thereof may be used for educational purposes at confer-
Jessica Altman, MD (‘21) – Senior Executive Editor, ASH-SAP8;
ences, continuing education courses, and other educational
Junior Editor, ASH-SAP7
activity, provided no fee or other compensation is charged,
therefore. All materials so used must acknowledge the Adam Cuker, MD (‘22) – Executive Editor, Hematology; Senior
Publisher’s copyright therein as “©2021 by The American Executive Editor, ASH-SAP7
Society of Hematology.” When requesting the Publisher’s Laura M. De Castro, MD (‘23) – Member, Committee on
permission to use this publication or any part thereof, please Practice
contact the Copyright Clearance Center at 222 Rosewood Cynthia E. Dunbar, MD (‘24) – Secretary
Drive, Danvers, MA 01923, USA; phone: 978–750–8400; fax: Alison Loren, MD (‘21) – 2021 Education Program Co-Chair
978–750–4470; or The American Society of Hematology
Anita Rajasekhar, MD (‘21) – 2021 Education Program Co-Chair
Publishing at 2021 L Street, NW, Suite 900, Washington, DC
20036, USA; phone: 202–776–0544; fax: 202–776–0545. Cover: Computer illustration of eosinophilia showing a view
inside a blood vessel with numerous eosinophils. Source:
Article Citations
Kateryna Kon/Science Photo Library.
Cite articles in this volume by listing Author(s), Title,
Hematology Am Soc Hematol Educ Program. 2021;2021: This activity is supported by educational grants from
beginning page number-ending page number. AstraZeneca; Jazz Pharmaceuticals, Inc.; Merck & Co., Inc.;
Novartis Pharmaceuticals; Pfizer Inc.; and Vertex Pharma-
Hematology Rights and Permission ceuticals Inc.
To request permission to reprint or reuse Hematology
articles, figures, or tables, please visit the Hematology page
at the Copyright Clearance Center (https://marketplace.
copyright.com/rs-ui-web/mp/details/journal/123161909).

The publisher disclaims responsibility for opinions expressed


by the authors.

ii
Contents

ix Editors’ Message
x Reviewers
xi Continuing Medical Education Information

ALL: New Directions for Adult Patients


1 Relapsed ALL: CAR T vs transplant vs novel therapies
NOELLE V. FREY

7 Acute lymphoblastic leukemia in older adults: curtain call for conventional chemotherapy?
MARLISE R. LUSKIN

AML: So Many Options, So Little Time


16 What to use to treat AML: the role of emerging therapies
FELICITAS THOL

24 Whom should we treat with novel agents? Specifc indications for specifc and challenging
populations
LINDSAY WILDE AND MARGARET KASNER

Challenges in Multiple Myeloma Treatment


30 High-risk multiple myeloma: how to treat at diagnosis and relapse?
MARÍA-VICTORIA MATEOS, BORJA PUERTAS MARTÍNEZ, AND VERÓNICA GONZÁLEZ-CALLE

37 Minimal residual disease in multiple myeloma—why, when, where


ANDREW J. YEE AND NOOPUR RAJE

46 Treatment of older adult or frail patients with multiple myeloma


SHAKIRA J. GRANT, CIARA L. FREEMAN, AND ASHLEY E. ROSKO

CLL: Extending Survival


55 Upfront therapy: the case for continuous treatment
CONSTANTINE S. TAM

59 Frontline treatment in CLL: the case for time-limited treatment


VINCENT LÉVY, ALAIN DELMER, AND FLORENCE CYMBALISTA

68 Is there a role for anti-CD20 antibodies in CLL?


HARSH R. SHAH AND DEBORAH M. STEPHENS

Clotting and Bleeding Conundrums


76 Unexplained arterial thrombosis: approach to diagnosis and treatment
JORI E. MAY AND STEPHAN MOLL

85 Cryptogenic oozers and bruisers


KRISTI J. SMOCK AND KAREN A. MOSER

Prakash Singh Shekhawat


Contents | iii
92 Clots in unusual places: lots of stress, lim­ited data, crit­i­cal deci­sions
CAROL MATHEW AND MARC ZUMBERG

100 EVIDENCE-BASED MINIREVIEW


Should war­fa­rin or a direct oral anti­co­ag­u­lant be used in patients presenting with throm­bo­sis in the
splanch­nic or cere­bral veins?
CAROL MATHEW AND MARC ZUMBERG

CML: Success Breeds More Success


106 TKI dis­con­tin­u­at­ ion in CML: how do we make more patients eli­gi­ble? How do we increase the
chances of a suc­cess­ful treat­ment-free remis­sion?
ANDREAS HOCHHAUS AND THOMAS ERNST

113 Lifelong TKI ther­apy: how to man­age car­dio­vas­cu­lar and other risks
MICHAEL J. MAURO

122 Blast and accel­er­ated phase CML: room for improve­ment


JOAN HOW, VINAYAK VENKATARAMAN, AND GABRIELA SORIANO HOBBS

Coagulation Laboratory Potpourri


129 Direct oral anti­co­ag­u­lant (DOAC) inter­fer­ence in hemo­sta­sis assays
KAREN A. MOSER AND KRISTI J. SMOCK

What Do Aplastic Anemia, PNH, and “Hypoplastic” Leukemia Have in Common?


134 The clin­i­cal and lab­o­ra­tory eval­u­a­tion of patients with suspected hypocellular mar­row fail­ure
SIOBÁN KEEL AND AMY GEDDIS

143 When does a PNH clone have clin­i­cal sig­nif­i­cance?


DARIA V. BABUSHOK

153 When to worry about inherited bone mar­row fail­ure and mye­loid malig­nancy pre­dis­po­si­tion
syn­dromes in the set­ting of a hypocellular mar­row
ANUPAMA NARLA

Defeating Diffuse, Double-Hit, and Dogged Non-Hodgkin Lymphoma


157 Double-hit lym­phoma: opti­miz­ing ther­apy
KIERON DUNLEAVY

164 Relapsed dis­ease: off-the-shelf immunotherapies vs cus­tom­ized engineered prod­ucts


REEM KARMALI

Emerging Therapies and Considerations for Sickle Cell Disease


174 Gene ther­apy for sickle cell dis­ease: where we are now?
JULIE KANTER AND COREY FALCON

181 Hematopoietic cell trans­plan­ta­tion for sickle cell dis­ease: updates and future direc­tions
LAKSHMANAN KRISHNAMURTI

190 EVIDENCE-BASED MINIREVIEW


In young chil­dren with severe sickle cell dis­ease, do the ben­e­fits of HLA-iden­ti­cal sib­ling donor HCT
out­weigh the risks?
NIKETA SHAH AND LAKSHMANAN KRISHNAMURTI

196 Clinical trial con­sid­er­ations in sickle cell dis­ease: patient-reported out­comes, data ele­ments,
and the stake­holder engage­ment frame­work
SHERIF M. BADAWY

iv  |  Hematology 2021  |  ASH Education Program


Hemophilia Update: Our Cup Runneth Over
206 How do we opti­mally uti­lize fac­tor con­cen­trates in per­sons with hemo­philia?
MING Y. LIM

215 EVIDENCE-BASED MINIREVIEW


For over­weight or obese per­sons with hemo­philia A, should fac­tor VIII dos­ing be based on ideal or
actual body weight?
NICOLETTA MACHIN AND MING Y. LIM

219 Factor-mimetic and rebalancing ther­a­pies in hemo­philia A and B: the end of fac­tor con­cen­trates?
PATRICK ELLSWORTH AND ALICE MA

226 Hemophilia gene ther­apy: ush­er­ing in a new treat­ment par­a­digm?


LINDSEY A. GEORGE

Hodgkin Lymphoma: Cure and Optimal Survivorship


234 Controversies in the man­age­ment of early-stage Hodgkin lym­phoma
KRISTIE A. BLUM

240 How to choose first sal­vage ther­apy in Hodgkin lym­phoma: tra­di­tional che­mo­ther­apy vs novel agents
JULIA DRIESSEN, SANNE H. TONINO, ALISON J. MOSKOWITZ, AND MARIE JOSÉ KERSTEN

247 Double-refrac­tory Hodgkin lym­phoma: tack­ling relapse after brentuximab vedotin and check­point
inhib­i­tors
NARENDRANATH EPPERLA AND MEHDI HAMADANI

How Can We Ensure That Everyone Who Needs a Transplant Can Get One?
254 Allogeneic hema­to­poi­etic cell trans­plan­ta­tion for older patients
RICHARD J. LIN AND ANDREW S. ARTZ

264 Increasing access to allo­ge­neic hema­to­poi­etic cell trans­plant: an inter­na­tional per­spec­tive


VANDERSON ROCHA, GIANCARLO FATOBENE, AND DIETGER NIEDERWIESER, FOR THE BRAZILIAN SOCIETY OF BONE
MARROW TRANSPLANTATION AND THE WORLDWIDE NETWORK FOR BLOOD AND MARROW TRANSPLANTATION

275 Increasing access to allotransplants in the United States: the impact of race, geog­ra­phy, and
socio­eco­nom­ics
SANGHEE HONG AND NAVNEET S. MAJHAIL

Immunology 101: What the Practicing Hematologist Needs to Know


281 Immunology 101: fun­da­men­tal immu­nol­ogy for the prac­tic­ing hema­tol­o­gist
SHANNON A. CARTY

287 How immu­no­de­fi­ciency can lead to malig­nancy


SUNG-YUN PAI, KATHRYN LURAIN, AND ROBERT YARCHOAN

296 Modified T cells as ther­a­peu­tic agents


NATHAN SINGH

Indolent Lymphomas
303 What fac­tors guide treat­ment selec­tion in myco­sis fungoides and Sezary syn­drome?
YOUN H. KIM

313 Follicular lym­phoma: is there an opti­mal way to define risk?


CARLA CASULO

320 Does MRD have a role in the man­age­ment of iNHL?


ILARIA DEL GIUDICE, IRENE DELLA STARZA, AND ROBIN FOÀ

Inherited Red Cell Disorders Beyond Hemoglobinopathies


331 Diagnosis and clin­i­cal man­age­ment of red cell mem­brane dis­or­ders
THEODOSIA A. KALFA

Prakash Singh Shekhawat


Contents | v
341 Diagnosis and clin­i­cal man­age­ment of enzymopathies
LUCIO LUZZATTO

353 Diamond-Blackfan ane­mia


LYDIE M. DA COSTA, ISABELLE MARIE, AND THIERRY M. LEBLANC

It Takes a Village: Maximizing Supportive Care and Minimizing Toxicity During Childhood Leukemia
Therapy
361 Fungal diag­nos­tic test­ing and ther­apy: nav­i­gat­ing the neutropenic period in chil­dren with high-
risk leu­ke­mia
BRIAN T. FISHER

368 Minimizing car­diac tox­ic­ity in chil­dren with acute mye­loid leu­ke­mia


HARI K. NARAYAN, KELLY D. GETZ, AND KASEY J. LEGER

376 Managing ther­apy-asso­ci­ated neu­ro­tox­ic­ity in chil­dren with ALL


DEEPA BHOJWANI, RAVI BANSAL, AND ALAN S. WAYNE

Let the CHIP(s) Fall Where They May? Burdens and Benefits of Diagnosing Clonal Hematopoiesis
384 CHIP: is clonal hema­to­poi­e­sis a sur­ro­gate for aging and other dis­ease?
LUKASZ P. GONDEK

390 Mechanisms of somatic trans­for­ma­tion in inherited bone mar­row fail­ure syn­dromes


HARUNA BATZORIG CHOIJILSUREN, YEJI PARK, AND MOONJUNG JUNG

399 When are idi­o­pathic and clonal cytopenias of unknown sig­nif­i­cance (ICUS or CCUS)?
AFAF E. W. G. OSMAN

Management Strategies for Sickle Cell Disease


405 Optimizing man­age­ment of sickle cell dis­ease in patients under­go­ing sur­gery
CHARITY I. OYEDEJI AND IAN J. WELSBY

411 Treatment dilem­mas: strat­eg ­ ies for priapism, chronic leg ulcer dis­ease, and pul­mo­nary
hyper­ten­sion in sickle cell dis­ease
ROBERTA C.G. AZBELL AND PAYAL CHANDARANA DESAI

MDS: Beyond a One-Size-Fits-All Approach


418 Have we reached a molec­ul­ar era in myelodysplastic syn­dromes?
MARIA TERESA VOSO AND CARMELO GURNARI

428 Lower risk but high risk


AMY E. DeZERN

435 EVIDENCE-BASED MINIREVIEW


Molecular pre­ci­sion and clin­i­cal uncer­tainty: should molec­u­lar pro­fil­ing be rou­tinely used to guide
risk strat­i­fi­ca­tion in MDS?
DAN­IEL R. RICHARDSON AND AMY E. DeZERN

439 Oral hypomethylating agents: beyond con­ve­nience in MDS


ELIZABETH A. GRIFFITHS

MPN: New Directions


710 MPN and throm­bo­sis was hard enough . . . ​now there’s COVID-19 throm­bo­sis too
ANNA FALANGA

448 EVIDENCE-BASED MINIREVIEW


Are DOACs an alter­na­tive to vita­min K antag­on
­ ists for treat­ment of venous throm­bo­em­bo­lism in
patients with MPN?
FRANCESCA SCHIEPPATI AND ANNA FALANGA

453 Novel ther­a­pies vs hema­to­poi­etic cell trans­plan­ta­tion in mye­lo­fi­bro­sis: who, when, how?
JAMES ENGLAND AND VIKAS GUPTA

463 Running inter­feron inter­fer­ence in treating PV/ET: meet­ing unmet needs


ELIZABETH A. TRAXLER AND ELIZABETH O. HEXNER

vi  |  Hematology 2021  |  ASH Education Program


Multidisciplinary Hematologic Disorders: What Is the Hematologist’s Role?
469 Hereditary hem­or­rhagic tel­an­gi­ec­ta­sia (HHT): a prac­ti­cal guide to man­age­ment
ADRIENNE M. HAMMILL, KATIE WUSIK, AND RAJ S. KASTHURI

478 Chronic throm­bo­em­bolic pul­mo­nary hyper­ten­sion: anticoagulation and beyond


KARLYN A. MARTIN AND MICHAEL J. CUTTICA

485 Rebalanced hemo­sta­sis in liver dis­ease: a mis­un­der­stood coagulopathy


LARA N. ROBERTS

Neutropenia: Doing More with Less


492 Diagnosis and ther­a­peu­tic deci­sion-mak­ing for the neutropenic patient
JAMES A. CONNELLY AND KELLY WALKOVICH

504 Understanding neutropenia sec­ond­ary to intrin­sic or iat­ro­genic immune dysregulation


KELLY WALKOVICH AND JAMES A. CONNELLY

514 Impaired myelopoiesis in con­gen­i­tal neutropenia: insights into clonal and malig­nant hema­to­poi­e­sis
JULIA T. WARREN AND DAN­IEL C. LINK

Perioperative Consultative Hematology: Can You Clear My Patient for Surgery?


521 Perioperative con­sul­ta­tive hema­tol­ogy: can you clear my patient for a pro­ce­dure?
ALLISON ELAINE BURNETT, BISHOY RAGHEB, AND SCOTT KAATZ

529 How to man­age bleed­ing dis­or­ders in aging patients need­ing sur­gery


MOUHAMED YAZAN ABOU-ISMAIL AND NATHAN T. CONNELL

536 Heparin-induced throm­bo­cy­to­pe­nia and car­dio­vas­cu­lar sur­gery


ALLYSON M. PISHKO AND ADAM CUKER

Pregnancy in Special Populations: Challenges and Solutions


545 How to eval­u­ate and treat the spec­trum of TMA syn­dromes in preg­nancy
MARIE SCULLY

552 Pregnancy in spe­cial pop­u­la­tions: chal­lenges and solu­tions prac­ti­cal aspects of man­ag­ing von
Willebrand dis­ease in preg­nancy
OZLEM TURAN AND REZAN ABDUL KADIR

559 Prevention and man­age­ment of venous throm­bo­em­bo­lism in preg­nancy: cut­ting through the
prac­tice var­i­a­tion
LESLIE SKEITH

Survivorship After Allogeneic Hematopoietic Cell Transplant


570 Psychosocial and finan­cial issues after hema­to­poi­etic cell trans­plan­ta­tion
DAVID BUCHBINDER AND NANDITA KHERA

578 Noninfectious com­pli­ca­tions of hema­to­poi­etic cell trans­plan­ta­tion


KIRSTEN M. WILLIAMS

587 Infectious com­pli­ca­tions and vac­cines


PER LJUNGMAN

The Changing Landscape of α- and β-Thalassemia Diagnosis and Treatment


592 Advances in the man­age­ment of α-thal­as­se­mia major: rea­sons to be opti­mis­tic
PAULINA HORVEI, TIPPI MACKENZIE, AND SANDHYA KHARBANDA

600 β-Thalassemia: evolv­ing treat­ment options beyond trans­fu­sion and iron che­la­tion
ARIELLE L. LANGER AND ERICA B. ESRICK

607 Optimal strat­e­gies for car­rier screen­ing and pre­na­tal diag­no­sis of α- and β-thal­as­se­mia
CHERYL MENSAH AND SUJIT SHETH
Prakash Singh Shekhawat
Contents | vii
The COVID Crash: Lessons Learned from a World on Pause
614 How to rec­og­nize and man­age COVID-19-asso­ci­ated coagulopathy
GLO­RIA F. GERBER AND SHRUTI CHATURVEDI

621 COVID-19 and throm­bo­sis: searching for evi­dence


BRIGHT THILAGAR, MOHAMMAD BEIDOUN, RUBEN RHOADES, AND SCOTT KAATZ

628 Passive immune ther­a­pies: another tool against COVID-19


LISE J. ESTCOURT

Update in Graft-versus-Host Disease


642 Acute GVHD: think before you treat
LAURA F. NEWELL AND SHERNAN G. HOLTAN

648 Updates in chronic graft-ver­sus-host dis­ease


BETTY K. HAMILTON

655 What else do I need to worry about when treating graft-ver­sus-host dis­ease?
AREEJ EL-JAWAHRI

What’s New in Plasma Cell Disorders?


662 Advances in MGUS diag­no­sis, risk strat­i­fi­ca­tion, and man­age­ment: intro­duc­ing myeloma-defining
genomic events
OLA LANDGREN

673 Smoldering mul­ti­ple mye­loma: evolv­ing diag­nos­tic cri­te­ria and treat­ment strat­e­gies
ALISSA VISRAM, JOSELLE COOK, AND RAHMA WARSAME

682 AL amy­loid­osis: untangling new ther­a­pies


SUSAN BAL AND HEATHER LANDAU

What’s New in the Transfusion Management of Sickle Cell Disease?


689 How to avoid the prob­lem of eryth­ro­cyte alloimmunization in sickle cell dis­ease
FRANCE PIRENNE, A­LINE FLOCH, AND ANOOSHA HABIBI

696 Indications for transfusion in the management of sickle cell disease


HYOJEONG HAN, LISA HENSCH, AND VENÉE N. TUBMAN

704 Management of hemo­lytic trans­fu­sion reac­tions


JEANNE E. HENDRICKSON AND ROSS M. FASANO

viii  |  Hematology 2021  |  ASH Education Program


Editors’ Message

Welcome to the 63rd annual meeting of the American Society of Hematology. Two years
after the first cases of COVID-19 were reported, we are learning to co-exist with the virus.
Many of us are thrilled to be able to attend a scientific congress in person for the first time
since the pandemic began. Others are grateful for the option of virtual participation, which
greatly expands access to the meeting and its cutting-edge content for hematologists
around the world.
This marks our third and final year as Editors of Hematology. During our term, we have
made significant changes to the book. For starters, Hematology is now electronic-only;
gone are the days of schlepping a heavy tome home from the ASH meeting. We have also
sought to make the book more readable and visually appealing. All chapters are based on
clinical cases and include visual abstracts. We have increased the number of tables and
figures per chapter while reducing the amount of text.
Nonetheless, at its core, Hematology remains the same great resource we inherited
three years ago. As always, you can count on high quality content spanning the breadth of
malignant and nonmalignant hematology, based on a state-of-the-art Education Program
developed by Education Program Co-Chairs, Dr. Alison Loren and Dr. Anita Rajasekhar.
This book is the culmination of the efforts of many individuals and their dedication to
ASH, including the authors, reviewers, and ASH staff (Ken April, Glenn Landis, Michelle Lee,
and Joanna Robertson). We hope that you enjoy reading Hematology as much as we have
enjoyed editing it these last three years.

Adam Cuker, MD Ann S. LaCasce, MD Mario Cazzola, MD Stella T. Chou, MD David Garcia, MD

Prakash Singh Shekhawat


ix
Reviewers

The editors would like to thank the session chairs and the reviewers who worked hard to ensure the reliability of the material
presented in Hematology 2021:

Jeremy S. Abramson Erica B. Esrick Ola Landgren Rachel E. Rau


Ranjana Advani Andrew M. Evens Sophie Lanzkron Farhad Ravandi
Walter Ageno Anna Falanga Stephanie J. Lee Ulrike M. Reiss
Hanny Al-Samkari Emmanuel J. Favaloro Georg Lenz Lawrence Rice
Jennifer Andrews Courtney D. Fitzhugh Jeffrey M. Lipton Heesun J. Rogers
Joseph H. Antin Angela G. Fleischman Mark R. Litzow Cristhiam M. Rojas
Andrew Artz Mary E. Flowers Marlise R. Luskin Hernandez
Daria V. Babushok Gianluca Gaidano Samuel E. Lux David M. Ross
Andrea Bacigalupo Guillermo Garcia-Manero Alice Ma Bruce S. Sachais
Tiziano Barbui Rebecca A. Gardner Ivan Maillard Marie Scully
Renato Bassan Alex Gatt Navneet S. Majhail John F. Seymour
Shannon Bates James N. George Michael Makris Nirmish Shah
Lisa Baumann Kreuziger Lukasz P. Gondek Luca Malcovati Urvi A. Shah
Emily Baumrin Rachael F. Grace Karlyn Martin Bronwen E. Shaw
Neel S. Bhatt John G. Gribben Paul J. Martin Shalini Shenoy
Deepa Bhojwani Elizabeth A. Griffiths Maria-Victoria Mateos Sujit Sheth
Kristie A. Blum Anna B. Halpern Patrick T. McGann Akiko Shimamura
Niccolo Bolli Betty K. Hamilton Neha Mehta-Shah Michelle Sholzberg
Robert A. Brodsky Catherine P.M. Hayward Reid W. Merryman B. Douglas Smith
Andrew Campbell Jeanne Elise Hendrickson Ruben A. Mesa Kristi J. Smock
Shannon Ann Carty Alex F. Herrera Brian Miller Gerald A. Soff
Carla Casulo Jens Hillengass Alison R. Moliterno Jean Soulier
Mario Cazzola Jo Howard Philippe Moreau Brady L. Stein
Shruti Chaturvedi Scott F. Huntington Alison J. Moskowitz Sean R. Stowell
Alan R. Cohen E. Leila Jerome Clay Michael F. Murphy Kendra L. Sweet
Raymond L. Comenzo Theodosia A. Kalfa Peter E. Newburger Felicitas Thol
Nathan T. Connell Manali Kamdar Diane Nugent Judith Trotman
Catherine Cordonnier Julie Kanter Susan M. O’Brien Saad Z. Usmani
Jennifer L. Crombie Margaret Kasner Kristen M. O’Dwyer Adrianna Vlachos
Stacy E Croteau Sioban B. Keel Vivian G. Oehler Oksana Volod
Geoffrey D.E. Cuvelier Nandita Khera Rebecca L. Olin Kelly Walkovich
Anita D’Souza Miriam Kimpton Ashok Pai Mark C. Walters
Michael R. DeBaun Melanie Ann Kirby-Allen Sung-Yun Pai Theodore E. Warkentin
Matteo G. Della Porta Frederikus A. Klok Christopher Jordan Patriquin William George Wierda
Amy E. DeZern Barbara A. Konkle Claire S. Philipp Jennifer J. Wilkes
Courtney D. DiNardo Peter Kouides Joseph Pidala Marcin W. Wlodarski
James Douketis Austin Kulesekararaj Uwe Platzbecker Jasmine Zain
Kieron Dunleavy Shaji K. Kumar Daniel A. Pollyea Alberto Zanella
Christopher C. Dvorak Peter Kurre Steven Pugliese
Matthew Ehrhardt Janet L. Kwiatkowski Margaret V. Ragni

x
Continuing Medical Education Information

Hematology: the ASH Education Program, is an annual publica- ABIM Maintenance of Certifcation
tion that provides practicing hematologists with invaluable infor- Successful completion of this CME activi-
mation on the most important areas of clinical progress. ty enables the participant to earn up to 40
Hematology 2021 is a peer-reviewed collection of articles Knowledge Points points in the American Board of Internal Med-
written by the 2021 ASH Education Program speakers and the icine’s (ABIM) Maintenance of Certification (MOC) program. Par-
Ham-Wasserman Lecturer. The papers showcase groundbreak- ticipants will earn MOC points equivalent to the amount of CME
ing advances and new concepts in 32 different fields. Every year, credits claimed for the activity. It is the CME activity provider’s
the periodical provides an updated and comprehensive review responsibility to submit participant completion information to
of each of the topics covered in the annual meeting education ACCME for the purpose of granting ABIM MOC credit.
sessions.
Claiming CME Credits and ABIM points
Educational objectives The estimated time to complete this educational activity is 40
1. Employ the knowledge gained regarding the diagnosis and hours. To claim CME credit, users must complete an evaluation
treatment of malignant and nonmalignant hematologic dis- of the product and a test of medical knowledge, both of which
orders to improve patient care. are accessed through ASH Academy On Demand (academy.hema
tology.org). There is a one-time processing fee for claiming CME/
2. Discuss the state-of-the-art therapeutics in hematology.
MOC credit. Users with scores of 80% or better on these self-
3. Analyze the potential contribution of novel, not-yet-approved assessment modules are eligible to claim credit for the activity.
modalities of therapy to current evidence-based manage- To facilitate claiming of credit, the test for this product is
ment of malignant and nonmalignant hematologic disorders. divided into two subtests, one of which focuses on malignant
hematology content with the other focusing on nonmalignant
Date of release content. Successful completion of each test earns the user 20
December 2021 AMA Category 1 PRA CreditsTM. Users claim CME and/or ABIM
MOC credit for each test individually. You can take one or both
Date of expiration
tests, depending on your CME and MOC needs.
On December 31, 2022, the ability to earn Continuing Medical
Each test consists of 25 questions. The 25 questions can be
Education credit and American Board of Internal Medicine Main-
answered in one sitting, or a user can save their progress and
tenance of Certification Medical Knowledge points for this prod-
return to complete the test at a later time.
uct expires. This is the last date for users to claim credit for this
product. For questions about credit, please contact the ASH The Malignant Hematology Test covers information present-
Education Department at cme@hematology.org or call toll-free ed in the following sections:
866-828-1231 within United States only; 1-202-776-0544 interna-

ALL: New Directions for Adult Patients
tionally.

AML: So Many Options, So Little Time

Challenges in Multiple Myeloma Treatment
Accreditation and Credit Designation

CLL: Extending Survival
The American Society of Hematology is accred-

CML: Success Breeds More Success
ited by the Accreditation Council for Continuing
Medical Education to provide continuing medical •
Defeating Diffuse, Double-Hit and Dogged Non-Hodgkin
education for physicians. Lymphoma
The American Society of Hematology designates this endur- • Hodgkin Lymphoma: Cure and Optimal Survivorship
ing material for a maximum of 40 AMA PRA Category 1 Credits . TM • How Can We Ensure That Everyone Who Needs a Transplant
Physicians should claim only the credit commensurate with the Can Get One?
extent of their participation in the activity. • Immunology 101: What the Practicing Hematologist Needs to
Physicians who participate in this CME activity but are not Know
licensed in the United States are also eligible for AMA Category • Indolent Lymphomas
1 PRA CreditTM. To earn these credits, readers must pass two on- • It Takes a Village: Maximizing Supportive Care and Minimizing
Prakash
line tests (malignant and nonmalignant) Singh
based on Shekhawat
chapters from Toxicity During Childhood Leukemia Therapy
the book. • MDS: Beyond a One-Size-Fits-All Approach
xi
• MPN: New Directions • Let the CHIP(s) Fall Where They May? Burdens and Benefits
• Normal and Leukemic Stem Cells of Diagnosing Clonal Hematopoiesis
• Survivorship After Allogeneic Hematopoietic Cell Transplant • Management Strategies for Sickle Cell Disease
• Update in Graft vs Host Disease • Multi-Disciplinary Hematologic Disorders: What Is the Hema-
• What’s New in Plasma Cell Disorders? tologist’s Role?
• Neutropenia: Doing More with Less
The Nonmalignant Hematology Test covers information presented
• Perioperative Consultative Hematology: Can You Clear My
in the following sections:
Patient for Surgery?
• Clotting and Bleeding Conundrums • Pregnancy in Special Populations: Challenges and Solutions
• Coagulation Laboratory Potpourri • The Changing Landscape of Alpha and Beta-Thalassemia
• Curative Therapies and Considerations for Sickle Cell Disease Diagnosis and Treatment
• Dameshek’s Riddle in Practice: Is this Aplastic Anemia, Parox- • The COVID Crash: Lessons Learned from a World on Pause
ysmal Nocturnal Hemoglobinuria, or Hypoplastic Leukemia? • What’s New in the Transfusion Management of Sickle Cell
• Hemophilia Update: Our Cup Runneth Over ­Disease?
• Inherited Red Cell Disorders Beyond Hemoglobinopathies

xii 
ALL: NEW DIRECTIONS FOR ADULT PATIENTS

Relapsed ALL: CAR T vs transplant vs novel


therapies

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/1/1851774/1frey.pdf by guest on 13 December 2021


Noelle V. Frey
Cell Therapy and Transplant Program, Abramson Cancer Center, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA

Chimeric antigen receptor T-cell therapy targeting CD19 (CART19) has expanded the treatment options for patients
with relapsed/refractory (r/r) B-cell acute lymphoblastic leukemia (ALL). The approval of tisagenlecleucel for pediatric
and young adult patients with r/r ALL has allowed broader access for some patients, but the treatment of older adults
is available (at the time of this writing) only within a clinical trial. High remission rates have been consistently observed
with varied CART19 products and treatment platforms, but durability of remissions and thus the potential role of a con-
solidative allogeneic stem cell transplant (SCT) is more uncertain and likely to vary by product and population treated.
The immunologic characteristics of CARTs that confer high response rates also account for the life-threatening toxicities
of cytokine release syndrome and immune effector cell–associated neurotoxicity syndrome, the severity of which also
varies by patient and disease characteristics and product. Further considerations informing a decision to treat include
feasibility of leukapheresis and timeline of manufacture, alternative treatment options available, and the appropriate-
ness of a potential consolidative allogeneic SCT. Advances in the field are under way to improve rate and duration of
responses and to mitigate toxicity.

LEARNING OBJECTIVES
• Understand the efficacy and toxicity outcomes of CART19 in r/r ALL and how they vary by product, patient, and
disease-related factors
• Understand factors that inform a decision to consolidate a recipient of CART19 in CR with allogeneic SCT

Efficacy outcomes of CART19 in relapsed (by 1-month postinfusion), are often minimal residual dis-
and refractory ALL ease (MRD) negative, and are not discriminated by muta-
Response tional status or number and type of prior therapies. The
Autologous T cells engineered to express a chimeric anti- trial populations represent heavily pretreated patients, with
gen receptor T-cell therapy targeted to CD19 (CART19) have over a third of patients in some studies having relapsed
consistently shown high complete remission (CR) rates after prior allogeneic stem cell transplant (SCT).1–3,6,8,9 Impor-
(62%-95%) in adult and pediatric patients with relapsed or tantly, several studies have shown CART19 cells tracking
refractory (r/r) acute lymphoblastic leukemia (ALL).1–12 The into the central nervous system (CNS) with responses seen
chimeric antigen receptor (CAR) T cells used in the studies in patients with CNS disease.2,5,10,11 Although the outcomes
summarized in Table 1 are made from a patient’s T cells col- discussed here are focused on recipients of CART19, CARTs
lected through leukapheresis that are then transduced with to other targets (specifically CD22) have been shown to
a CAR targeting CD19 using a replication-incompetent ret- be effective.13–16 In a large series of pediatric and adoles-
rovirus or lentivirus. The CARs include a costimulatory mol- cent young adult (AYA) patients (N=58), many of whom had
ecule, which is either CD28 or 41BB depending on product. relapsed after CART19 therapy (N=51), anti-CD22 CAR ther-
Patients typically receive lymphodepleting chemotherapy apy induced a CR in 70% of patients.14
(commonly cyclophosphamide and fludarabine) prior to
CART19 infusion. Importantly, despite differences in patient Survival and durability of response
populations, clinical trial procedures, CAR molecules, and Median overall survival (OS) in most studies using CART19
manufacturing differences, high initial response rates are is beyond 1 year and importantly is noted within some
maintained. Across studies, remissions are achieved quickly reports to vary by dose level or other changes to study
Prakash Singh Shekhawat
CAR T for ALL | 1
Table 1. Outcomes of CART19 in patients with relapsed and refrac­tory acute lym­pho­blas­tic leu­ke­mia

Prior
Reference CART domain No. treated Median age, y blinatumomab, % Prior SCT, % CR, % CRS ICANS
Adult patients
  Frey et al1 41BB 35 34 (21-70) 31 37 69 94% total 40% total
9% grades 4-5 6% grade 3
  Hay et al3 41BB 53 39 (20-76) 20 43 85 75% total 23% total
19% grades 3-4
  Park et al9 CD28 53 44 (23-64) 25 36 83 85% total 42% grades 3-4
26% severe
1 grade 5
  Shah et al12 CD28 55 40 (28-52) 45 42 71 89% total 60% total
25% grade 3 or 23% grades 3-4

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/1/1851774/1frey.pdf by guest on 13 December 2021


higher 1 grade 5
Combined pedi­at­ric and adult patients
  Jiang et al4 41BB 58 28 (10-65) NA 5 88 38% grades 3-5 16% grades 2-3
 Ortíz-Maldonado 41BB 38 24 (3-67) 26 87 85 13.2% grades 3-5 2.6% grade3 or
et al8 higher
  Wang et al16 41BB 23 42 (10-67) NA 0 83 100% total 13% total
18% grade 3
  Maude et al5 41BB 30 14 (5-60) 10 60 90 100% total 43% total
27% severe
Pediatric and AYA patients
  Gardner et al2 41BB 45 12.2 (1.3-25.3) 14 62 93 93% total 49% total
21% severe 21% severe
  Maude et al6 41BB 75 11 (3-23) 0 61 81 77% total 13% grade 3
25% grade 4
  Shah et al11 CD28 50 13.5 (4.3-30.4) 10 40 62 70% total 20% total
22% grades 3-4 8% severe
NA, not available.

design through­ out the clin­i­


cal trial. Multicenter stud­ ies using higher pro­ por­
tion of patients (20%-45%) on CART19 stud­ ies
con­sis­tent prod­ucts reflect more gen­er­al­iz­able out­comes. In the hav­ing received blinatumomab (see Table 1).1,3,9,12 In the recently
piv­otal ELIANA trial lead­ing to approval of tisagenlecleucel for published ZUMA-3 mul­ti­cen­ter trial, 45% of the 55 adult patients
pedi­at­ric and young adult patients, 75 patients were treated, treated with KTE-X19 had received blinatumomab. Although
with a CR rate of 81%. Median OS was not reached, with event- prior expo­sure to CD19-targeted agents may affect risk of sub­se­
free sur­vival (EFS) and OS at 12 months of 50% and 76%, respec­ quent CD19– relapse, ini­tial response rates do not seem greatly
tively. For respond­ers, the median dura­tion of remis­sion was not affected. The Children’s Hospital of Philadelphia reported out­
reached.6 In the mul­ti­cen­ter ZUMA-3 study treating 55 adults comes from 166 patients treated with CART19 at their insti­tu­tion.
with r/r ALL with KTE-X19, CR rate was 71% and median OS was The CR rate was 93%, and 67 patients ulti­mately relapsed, 39 with
18.2 months. The median dura­tion of remis­sion for respond­ers CD19– dis­ease due to anti­gen escaper. Prior ther­apy with blina-
was 12.8 months.12 Another mul­ti­cen­ter study treated 38 adult tumomab was asso­ci­ated with a higher risk for CD19– relapse.17
and pedi­at­ric patients with CART19 with a CR rate of 71%, with
OS and pro­gres­sion-free sur­vival of 67% and 47%, respec­tively, at Importance of per­sis­tence for dura­ble remis­sions
1 year. The median dura­tion of response was 14.8 months.8 Several ALL tri­ als using CART19 prod­ ucts containing a 4-1BB
costimulatory domain have shown a strong cor­re­la­tion between
Impact of prior blinatumomab on effi­cacy CART19 per­ sis­
tence (often represented by B-cell aplasia, a
Blinatumomab is a bispecific T cell engag­ing a sin­gle-chain anti­ bio­log­i­cal sur­ro­gate for func­tional per­sis­tence) and CD19+
body con­struct linking CD3+ T cells with CD19+ B cells. There relapses.1-3,6,7,18 In an anal­

sis of out­
comes with tisagenlecleu-
is a log­i­cal con­cern that due to its sim­i­lar mech­a­nism of action cel in patients with ALL on the ELIANA trial, the patients with
and tar­get, prior treat­ment with blinatumomab could adversely CD19+ relapses had a more rapid loss of CART per­ sis­
tence
affect out­comes from CART19. For this rea­son, early CART19 stud­ com­ pared with those with dura­ ble remis­ sions. Of inter­
est,
ies, such as the ELIANA study, prohibited prior treat­ment with patients who devel­oped a CD19– relapse had per­sis­tence sim­
blinatumomab or other CD19-targeted ther­ a­
pies.6 Since these i­lar to those who had sustained responses with mech­a­nism of
ini­
tial stud­ ies, the use of blinatumomab has increased sig­ nif­i­ relapse due to anti­gen escape.18 In another pedi­at­ric study using
cantly, espe­cially in adults, which is reflected in a pro­gres­sively a dif­fer­ent 4-1BBCART19 prod­uct at Seattle Children’s, a lon­ger

2  |  Hematology 2021  |  ASH Education Program


dura­tion of B-cell aplasia cor­re­lated sig­nif­i­cantly with the dura­ in CR, an improve­ment when com­pared ret­ro­spec­tively with
bil­ity of remis­sion.2 Another study found the median per­ sis­ out­comes from CART19 alone.13
tence of 41BBCART19 was shorter for those with a CD19+ relapse
(2.5 months) as opposed to a CD19– relapse (6 months).4 Role of consolidative SCT
Several stud­ies using CD28-CART19 prod­ucts have shown For a patient with­out an ante­ced­ent SCT with an MRD-CR after
lim­ited per­sis­tence, but a cor­re­la­tion of out­comes with per­sis­ CART19, a crit­i­cal ques­tion is whether to con­sol­i­date that remis­
tence is less clear.9,11,12 In the ZUMA-3 trial using CD28-CART19 sion with trans­plant if the patient is med­i­cally fit to con­sider this
(KTE-X19), CARTs were no lon­ger detect­able in 79% of patients approach. As always when con­sid­er­ing SCT, treat­ment-related
with evaluable sam­ples by 6 months, and B-cell recov­ery had mor­bid­ity and mor­tal­ity needs to be bal­anced against risk of
occurred in all­evaluable ongo­ing respond­ers at 12 months. relapse. Another con­sid­er­ation is that the CARTs with func­tional
The expe­ ri­
ence at Memorial Sloane Kettering Cancer Cen- per­sis­tence would be destroyed by SCT, los­ing their ben­e­fit of
ter using their CD28-CART19 showed median per­sis­tence of ongo­ing tumor sur­veil­lance. There are no stud­ies to date that
14 days (range, 7-138 days), and dura­tion of per­sis­tence did not ran­dom­ ize patients after CART ther­ apy to allo­ ge­ neic SCT or
cor­re­late with sur­vival.9 In 1 study, how­ever, from the National obser­va­tion. Furthermore, the deci­sion is not likely gen­er­al­iz­able

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/1/1851774/1frey.pdf by guest on 13 December 2021


Cancer Institute treating 50 pedi­at­ric and AYA patients with a across dif­fer­ent CART19 prod­ucts. For exam­ple, patients tak­ing
CD28-CART19 prod­uct, remis­sions were dura­ble only with con- 4-1BBCART19 prod­ucts have bet­ter per­sis­tence (with poten­tially
solidative SCT, which the authors hypoth­e­size may be due to lon­ger dis­ease-free inter­vals, although more data are needed)
lim­ited per­sis­tence.11 than recip­i­ents of CD28-CART19 prod­ucts. There is con­sid­er­able
Given the cor­re­la­tion between 4-1BBCART19 per­sis­tence and var­i­a­tion among clin­i­cal tri­als and ret­ro­spec­tive ana­ly­ses regard­
CD19+ dura­bil­ity of remis­sions, it is vital to under­stand cor­re­ ing the poten­tial ben­e­fit of a consolidative SCT, and again, no
lates with per­sis­tence using these CART19 prod­ucts. The group ran­dom­ized tri­als have for­mally addressed this ques­tion.
at the Fred Hutchinson Cancer Research Center found that the
addi­tion of fludarabine to cyclo­phos­pha­mide lymphodepletion The role of consolidative SCT in CD28-CARTs
improved per­sis­tence and dis­ease-free sur­vival.19 The impact of Recently, the National Cancer Institute reported long-term fol­
anti­gen load on per­sis­tence has var­ied across stud­ies. The group low-up of 50 chil­dren and young adults (median age, 13.5 years;
from Seattle Children’s found a pos­i­tive cor­re­la­tion, with >15% range, 4.3-30.4 years) treated with their CD28-CART19 prod­
bone mar­row blasts cor­re­lat­ing with prolonged per­sis­tence.2 uct who clearly benefited from a consolidative SCT. Of the
Conversely, the group from did not find an asso­ ci­
a­tion with 28 patients who achieved an MRD-CR, 21 (75%) proceeded to
low dis­ease bur­den and CD19+ relapses.17 Evaluation of T cells SCT with a median OS of 70.2 months. The cumu­la­tive inci­dence
in apher­e­sis and manufactured 4-1BBCART19 prod­ucts iden­ti­fied of relapse after SCT was 9.5% at 24 months. All patients who did
phe­no­typ­i­cal and func­tional attri­butes of CAR CD8 T cells that not pro­ceed to SCT relapsed at a median of 152 (range, 94-394)
cor­re­lated with per­sis­tence.20 days.11 However, data from Memorial Sloane Kettering Cancer
Most of the stud­ies in Table 1 use CARs containing a murine Center pro­vided long-term out­comes from 51 adults (median
domain that may be a tar­ get for immune-medi­ ated rejec­ tion. dura­tion, 18 months) treated with a dif­ fer­
ent CD28-CART19.
The devel­op­ment of a CART19 prod­uct using a CAR containing a Of the 32 patients in MRD-CR, no dif­fer­ence in OS was seen in
­human­ized anti-CD19 scFv domain may bypass this ­rejec­tion, patients who did or did not receive SCT after CARTs.9 Recently,
resulting in improved per­ sis­tence and relapsed free sur­ vival results from the ZUMA-3 mul­ti­cen­ter trial using KTE-X19 (a CART19
(RFS).7,10 A recent report of out­comes in pedi­at­ric and AYA patients with CD28) have been reported. In this study, 55 patients were
with r/r ALL treated with a human­ized CART19 based on the back­ treated and 39 patients (71%) achieved CR. Ten patients (18%)
bone of CTL019 (tisagenlecleucel) has shown excel­lent responses. proceeded to SCT, and with sen­si­tiv­ity ana­ly­ses, the median
In 41 CART19-​naive patients, CR rate was 98%, and RFS at 1 year dura­tion of remis­sion was unchanged by SCT con­sol­i­da­tion.12
was 84%. Similar to prior stud­ies, ear­lier B-cell recov­ery as a time-
depen­­dent covariate cor­re­lated with worse RFS. In an explor­atory The role of consolidative SCT in 4-1BB CARTs
anal­y­sis, time to B-cell recov­ery was com­pared with a his­tor­i­cal It is clear that a sub­set of patients with ini­tial response to some
cohort of CTL019 recip­i­ents, and there was a trend toward a lower 41BBCART19 prod­ucts has ongo­ing dura­ble remis­sions with­out
cumu­la­tive inci­dence of B-cell recov­ery by 6 months (15% vs 29%), consolidative SCT, which cor­re­lates with per­sis­tence.2,5-8 Even
but it did not reach sta­tis­ti­cal sig­nif­i­cance.7 when a par­tic­u­lar CART19 prod­uct has been shown to have good
func­tional per­sis­tence, how­ever, that per­sis­tence is not observed
Risk of CD19– relapse across all­patients treated, and CD19+ relapses remain a sig­nif­i­
CD19– relapses hap­pen when CD19 anti­gen loss occurs through cant chal­lenge that may be miti­gated by SCT in some patients.1
muta­tion or epi­ge­netic alter­ations, likely in preexisting leu­ke­mia In addi­tion, even if one antic­i­pates and observes ongo­ing per­
subclones.21-23 An attrac­tive approach to limit the inci­dence of sis­tence, CD19– relapses from anti­ gen escape occur despite
CD19– relapse is to infuse CARTs that tar­get more than 1 anti­gen CART19 per­sis­tence. Only a minor­ity of the young adult and pedi­
such as CD19 with either CD20 or CD22. One approach is to gen­ at­ric patients with relapsed ALL treated with tisagenlecleucel
er­ate a sin­gu­lar CART prod­uct that can tar­get more than 1 anti­ on the ELIANA study received a consolidative SCT. The EFS of
gen vs coadministering 2 dis­tinct prod­ucts with dif­fer­ent tar­gets 50% at 12 months is there­fore rep­re­sen­ta­tive of remis­sion dura­
either con­cur­rently or sequen­tially.13,16,24-26 In 1 study, 20 chil­dren bil­ity with this 4-1BBCART19 prod­uct.6 When CTL019 (the pre­
with r/r ALL who achieved an MRD-CR with CART19 were infused cur­sor to tisagenlecleucel) was used to treat older adults, those
with CART22 at a median of 1.65 months after CART19 infu­sion.13 who proceeded to SCT in MRD-CR had an improved EFS com­
No patients received consolidative SCT, and at 1 year, 85% were pared with those who were not, although dura­ ble remis­sions
Prakash Singh Shekhawat
CAR T for ALL  |  3
were seen in each group.1 Similarly, in another study using a dif­ ited. Current man­age­ment strat­e­gies are based on inter­ven­tion
fer­ent 4-1BBCART19, there was a trend toward an improved EFS with cor­ti­co­ste­roids. Risk fac­tors for severe ICANS are less clear
in patients bridged to SCT (P = .088). In another study using a but cor­re­late with prod­uct used, higher inter­leu­kin 6 lev­els, high
41BBCART19 for r/r ALL, 21 of the 47 patients who achieved MRD- dis­ease bur­den, and more severe sys­temic CRS.9,12,19 CART19 cells
CR were bridged to SCT. Although there was no dif­fer­ence in OS read­ily cross the blood-brain bar­rier (BBB, a poten­tial ben­e­fit for
between the 2 groups, EFS and RFS were sig­nif­i­cantly prolonged dis­ease targeting as discussed above) and are detect­able in the
in the SCT group.4 cere­bral spi­nal fluid (CSF) in most treated patients. Their pres­
ence in the CSF, how­ever, does not pre­dict for tox­ic­ity.5 Further
Toxicity stud­ies are needed to deter­mine the mech­a­nism of action, risk
Due to their mech­a­nism of action, CART19 is asso­ci­ated with fac­tors, and opti­mal man­age­ment strat­e­gies of neu­ro­logic tox­ic­
cyto­kine release syn­drome (CRS) and immune effec­tor cell– ity after CAR T-cell ther­apy.
asso­ci­ated neu­ro­tox­ic­ity syn­drome (ICANS), which are both
poten­tially life-threat­en­ing.27 The inci­dence of these com­pli­ca­ Role of CART19 vs other ther­a­pies
tions across selected tri­als for adult ALL is sum­ma­rized in Table 1. Clinicians may have sev­eral sal­vage options avail­­able to treat a

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/1/1851774/1frey.pdf by guest on 13 December 2021


It should be noted that dif­fer­ent grad­ing scales were used patient with r/r ALL. Treatment deci­sions need to be indi­vid­u­al­
across tri­als, chal­leng­ing any com­par­i­son of tox­ic­ity. ized based on prior ther­apy, goals of ther­apy, role of poten­tial
consolidative SCT, and fea­tures of the CART19 prod­uct avail­­able
CRS (see Table 2). Comparing out­comes of CART19 stud­ies with out­
Fever, hemo­dy­namic insta­bil­ity, and hyp­oxia are the core clin­i­ comes from inotuzumab (INO-VATE study) and blinatumomab
cal fea­tures of CRS and used in the American Society for Trans- (TOWER study) should be done with cau­tion given dif­fer­ences
plantation and Cellular Therapy con­sen­sus grad­ing scale.28 The in trial design, patient pop­u­la­tions, dis­ease bur­den, and role
inci­dence of CRS is high across all­CART19 stud­ies com­pared of consolidative SCT in the dif­fer­ent stud­ies. In addi­tion, most
with other B-cell malig­nan­cies. The syn­drome can be self-lim­ited effi­cacy out­comes reported from CART19 stud­ies are for those
(requir­ing only sup­port­ive care with anti­py­ret­ics and intra­ve­nous who made it to infu­sion, discounting fail­ures of treat­ment from
flu­ids) or more seri­ous, requir­ing inter­ven­tion with tocilizumab manufactur­ing or inabil­ity to tol­er­ate the treat­ment delay that
(an anti­body to the inter­leu­kin 6 recep­tor) or cor­ti­co­ste­roids. The is inher­ent in autol­og­ ous CART19 treat­ment. Randomized con­
biol­ogy and risk fac­tors for CRS are well under­stood and con­tinue trolled tri­als are lacking and needed to appro­pri­ately com­pare
to inform man­age­ment strat­e­gies, and fur­ther approaches to out­comes. Acknowledging these lim­i­ta­tions, out­comes from
mit­i­gate CRS are being explored in clin­i­cal tri­als. In ALL, dis­ease mature mul­ti­cen­ter CART19 tri­als (CR = 71%-81%; median OS of
bur­den is a strong pre­dic­tor for the sever­ity of CRS.2,5,9,12,19 Some 11 months not reached; median dura­tion of CR = 12.8 months not
cen­ters have adopted a risk-adapted approach in which a lower reached) com­pare favor­ably with out­comes with blinatumomab
dose of cells is given for patients with higher dis­ease bur­dens.9,19 (CR = 35.1%; median OS = 7.7 months, median dura­tion of CR = 7.3
Others have used more inten­ sive cyto-reduc­ tion for patients months) and inotuzumab (CR = 80.7%; median OS = 7.7 months;
with higher dis­ease bur­dens.10 Several cen­ters are eval­u­at­ing the median dura­tion of CR = 4.7 months).6,8,12,30,31
ben­efi ­ t of inter­ven­ing with tocilizumab for ear­lier grades of CRS
com­pared with the approach in early clin­ic ­ al tri­als that treated at
more severe CRS due to con­cerns that ear­lier inter­ven­tion may Table 2. Challenges of CART19 and solu­tions under inves­ti­ga­tion
mit­i­gate response. In 1 study from the Children’s Hospital of Phil-
adelphia, patients were assigned to high and low (<40%) tumor Challenge Solutions under inves­ti­ga­tion
bur­den cohorts. Those with high dis­ease bur­den received tocili- Relapse
zumab for per­sis­tent fevers. Of 70 patients treated, 15 received
  CD19– relapse due to anti­gen Dual targeted approach (CD19 and
early tocilizumab, which, when com­pared ret­ro­spec­tively with a escape CD22)
sim­i­lar group from an ear­lier study, showed a reduc­tion in grade   CD19+ relapse due to loss of 41BB costimulatory domain
4 CRS from 50% to 27%.29 Seattle Children’s reported their expe­ per­sis­tence Humanized CARTs
ri­ence with ear­lier tocilizumab inter­ven­tion resulting in a lower    Immune medi­ ated rejec­tion Manufacturing changes to select
   Exhaustive phe­ no­type for nonexhaustive phe­no­type
inci­dence of severe CRS with­out an impact on response.2 We
and oth­ers have used a frac­tion­ated dos­ing scheme in which the Logistics
total planned CART dose is infused over 3 days. This approach,   Window for leukapheresis Collect and store cells early in
in which sub­se­quent doses are held for early signs of CRS, allows   Disease con­trol dur­ing dis­ease course
for real-time dose mod­i­fi­ca­tion in response to tox­ic­ity with­out manufactur­ing Develop more rapid manufactur­ing
time
an impact on effi­cacy.1,8 “Off-the-shelf” or allo­ge­neic CARTs
Toxicity
Neurologic tox­ic­ity
The ASTCT con­sen­sus grad­ing for ICANS requires assess­ment   CRS and ICANS Earlier inter­ven­tion with
of a patient’s immune effec­tor cell–asso­ci­ated enceph­a­lop­a­thy tocilizumab or cor­ti­co­ste­roids
Fractionated dos­ing scheme: dose
score, level of con­scious­ness, sei­zure activ­ity, focal motor weak­ mod­i­fi­ca­tion in response to early
ness, and cere­bral edema.28 Similar to CRS, neu­ro­logic events tox­ic­ity
occur within the first few weeks of ther­ apy and have been Dose mod­i­fi­ca­tions by dis­ease
reported in all­the stud­ies sum­ma­rized in Table 1. Our under­stand­ bur­den
Novel anticytokine approaches
ing and abil­ity to mit­i­gate neu­ro­logic tox­ic­ity after CART19 is lim­

4  |  Hematology 2021  |  ASH Education Program


CART19, inotuzumab, blinatumomab, and che­mo­ther­apy in Real-world expe­ri­ence with CART19 for ALL
patients who are not refrac­tory all­have the poten­tial to suc­cess­ There is only 1 CART19 prod­uct approved in the United States
fully induce an MRD– remis­sion that can serve as a suc­cess­ful and Europe for r/r B ALL, although more prod­ucts are likely to be
bridge to SCT. Although suc­cess­ful, there is no evi­dence that an approved for this indi­ca­tion over the next few years. Tisagenle-
MRD– remis­sion from a CART prod­uct vs che­mo­ther­apy or other cleucel is approved to treat patients up to 25 years of age with
targeted approach improves RFS after SCT. One report com­ B-cell ALL that is refrac­tory or in sec­ond or greater relapse. This
pared out­comes from patients with r/r ALL who achieved MRD– approval is based on out­comes from the mul­ti­cen­ter phase 2
CR with CART19 or che­mo­ther­apy and were bridged to SCT. No ELIANA study discussed above.6
dif­fer­ence in cumu­la­tive inci­dence of relapse (11.1% vs 12.8%) or Real-world reg­is­try data col­lected after the com­mer­cial­i­za­tion
nonrelapse mor­tal­ity was iden­ti­fied.32 of tisagenlecleucel have sub­stan­ti­ated these out­comes and rep­
The tox­ic­ity of a spe­cific CART19 prod­uct needs to be con­ re­sent a larger num­ber and more diverse group of patients than
sid­ered on an indi­vid­u­al­ized basis and used to inform treat­ment those treated in the piv­otal trial. Information from 255 pedi­at­ric
deci­sions with CART19 vs another avail­­able prod­uct. Patients and young adult patients with relapsed ALL obtained from the
with high dis­ease bur­den, advanced age, and comorbid car­ Center for International Bone Marrow Transplant Registry found a

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/1/1851774/1frey.pdf by guest on 13 December 2021


dio­vas­cu­lar dis­ease, for exam­ple, may not tol­er­ate antic­i­pated CR rate of 86% with an EFS and OS of 68.6% and 88.5%, respec­
severe CRS or ICANS, and another approach may be more tively, at 6 months.34 This com­pares favor­ably to the ELIANA trial,
appro­pri­ate. On the other hand, a patient with min­i­mal dis­ease which had a CR rate of 81% and an EFS and OS of 72% and 87%,
bur­den would be antic­i­pated to have lower risk of these toxic- respec­tively, at 6 months. Importantly, treat­ment in the real world
ities from CART19. Other spe­cific fac­tors may influ­ence a cli­ni­ has been shown to be safe, with fewer patients hav­ing grade 3
cian’s deci­sion to treat with CART19. For a patient with a low or higher CRS (16% vs 48%) and sim­i­lar num­bers hav­ing grade
like­li­hood or desire to pro­ceed to an SCT if a CR is obtained, 3 or higher neu­ro­tox­ic­ity (9% vs 12.7%).34 The lower inci­dence in
CART19 prod­ucts that have been shown to have dura­ble remis­ CRS may reflect prog­ress in man­age­ment or less advanced dis­
sions with­out consolidative SCT may be favored over other ease at time of treat­ment. In addi­tion to val­i­dat­ing effi­cacy and
prod­ucts. A patient with CNS dis­ease his­tory may ben­e­fit from safety out­comes, real-world data have pro­vided infor­ma­tion on
CART19, which has been shown to cross the BBB and treat dis­ pop­u­la­tions not eli­gi­ble for the reg­is­tra­tion trial. For exam­ple, 6%
ease in the CSF. of patients from the Center for International Bone Marrow Trans-
The logis­tics of autol­o­gous CAR T-cell ther­apy, includ­ing plant Registry were youn­ger than 3 years, and 15% of patients had
iden­ti­fy­ing an appro­pri­ate win­dow to per­form leukapheresis prior blinatumomab; both sub­groups would have been excluded
and the need to con­trol dis­ease while cells are being manufac- from ELIANA.
tured, con­fer a sig­nif­i­cant dis­ad­van­tage com­pared with other
approaches to care, such as inotuzumab or blinatumomab. Conclusion
Several inves­ti­ga­tions are under way to explore the poten­ At the time of this writ­ing, there is only 1 CAR T-cell prod­uct,
tial ben­e­fit of using allo­ge­neic CARTs from healthy donors in tisagenlecleucel, approved to treat r/r ALL for patients 25 years
whom the poten­tial for graft-ver­sus-host dis­ease is abro­gated and youn­ger, but more approv­als over the next few years are
by T cell receptor knock­out with gene editing tech­niques. In antic­

pated. Treatment with CART19 yields high and dura­ ble
early stud­ies with this approach, responses have required high response rates for adult and pedi­ at­
ric patients with r/r ALL.
immu­no­sup­pres­sive ther­apy to min­i­mize immune-medi­ated The deci­sion to treat a patient with CART19 depends on sev­eral
rejec­tion.33 patient-, dis­
ease-, and prod­ uct-related fac­tors in con­
junc­
tion

Table 3. Clinical sce­nar­ios that favor CART19 vs other ther­a­pies

Clinical sce­nario CARTs vs alter­na­tive approach


Recent CNS dis­ease Consider CART19 due to abil­ity to cross BBB and treat the CNS com­
part­ment. Patients treated with che­mo­ther­apy, blinatumomab, or
inotuzumab are at risk for extramedullary relapse or pro­gres­sion.
Allogeneic SCT not pos­si­ble or desir­able CART19 should be con­sid­ered if prod­uct with dura­ble remis­sion with­out
  Relapse occurs after prior SCT SCT is avail­­able.
  High risk for com­pli­ca­tions from SCT
  No donor option
  Patient defers SCT
Rapidly pro­gres­sive dis­ease CART19 may not be fea­si­ble due to need to iden­tify a win­dow for
leukapheresis and con­trol dis­ease while awaiting man­u­fac­ture. Other
approaches should be con­sid­ered.
Frail or older patient with comorbid dis­ease and high dis­ease bur­den Patient may not tol­er­ate antic­i­pated severe CRS or ICANS with CART19.
Consider alter­na­tive approaches.
Frail or older patient with comorbid dis­ease and min­i­mal dis­ease bur­den Patient is likely to do well with blinatumomab, inotuzumab, and CART19.
With CART19, side effects of lymphodepletion need to be con­sid­ered,
although low CART-related tox­ic­ity is antic­i­pated. Decision may be
influ­enced by dura­bil­ity of remis­sions observed with CART19 prod­uct
avail­­able.
Prakash Singh Shekhawat
CAR T for ALL  |  5
with con­sid­er­ation of other ther­a­pies avail­­able and the poten­ 15. Singh N, Frey NV, Engels B, et al. Antigen-inde­pen­dent acti­va­tion enhances
tial role of a consolidative SCT. Approaches are being explored the effi­cacy of 4-1BB-costimulated CD22 CAR T cells. Nat Med. 2021;27(5):
­842-850.
to min­i­mize CD19+ and CD19– relapses and pre­vent or mit­i­gate 16. Wang N, Hu X, Cao W, et al. Efficacy and safety of CAR19/22 T-cell cock­
tox­ic­ity, which will con­tinue to affect the treat­ment par­a­digm for tail ther­apy in patients with refrac­tory/relapsed B-cell malig­nan­cies. Blood.
ALL over time (see Table 3). 2020;135(1):17-27.
17. Pillai V, Muralidharan K, Meng W, et al. CAR T-cell ther­apy is effec­tive for
CD19-dim B-lym­pho­blas­tic leu­ke­mia but is impacted by prior blinatum-
Conflict-of-inter­est dis­clo­sure
omab ther­apy. Blood Adv. 2019;3(22):3539-3549.
Noelle V. Frey: Novartis Research Funding. Kite Pharmaceuticals 18. Mueller KT, Waldron E, Grupp SA, et al. Clinical phar­ma­col­ogy of tisagen-
and Syndax Consultancy. lecleucel in B-cell acute lym­pho­blas­tic leu­ke­mia. Clin Cancer Res. 2018;24
­(24):6175-6184.
Off-label drug use 19. Turtle CJ, Hanafi LA, Berger C, et al. CD19 CAR-T cells of defined CD4+:CD8+
com­po­si­tion in adult B cell ALL patients. J Clin Invest. 2016;126:2123-2138.
Noelle V. Frey: nothing to disclose. 20. Finney OC, Brakke HM, Rawlings-Rhea S, et  al. CD19 CAR T cell prod­uct
and dis­ease attri­butes pre­dict leu­ke­mia remis­sion dura­bil­ity. J Clin Invest.
Correspondence 2019;129(5):2123-2132.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/1/1851774/1frey.pdf by guest on 13 December 2021


Noelle V. Frey, Hospital of the University of Pennsylvania, Abramson 21. Jacoby E, Nguyen SM, Fountaine TJ, et  al. CD19 CAR immune pres­ sure
induces B-pre­cur­sor acute lym­pho­blas­tic leu­kae­mia lin­e­age switch expos­
Cancer Center, Perelman Center for Advanced Medicine, 3400
ing inher­ent leukaemic plas­tic­ity. Nat Commun. 2016;7:12320.
Civic Center Boulevard, Philadelphia, PA 19104; e-mail: noelle​­.frey 22. Rabilloud T, Potier D, Pankaew S, et al. Single-cell pro­fil­ing identifies pre-
@pennmedicine​­.upenn​­.edu. existing CD19-neg­a­tive subclones in a B-ALL patient with CD19-neg­a­tive
relapse after CAR-T ther­apy. Nat Commun. 2021;12:865.
References 23. Singh N, Lee YG, Shestova O, et al. Impaired death recep­tor sig­nal­ing in
1. Frey NV, Shaw PA, Hexner EO, et al. Optimizing chi­me­ric anti­gen recep­tor leu­ke­mia causes anti­gen-inde­pen­dent resis­tance by induc­ing CAR T-cell
T-cell ther­apy for adults with acute lym­pho­blas­tic leu­ke­mia. J Clin Oncol. dys­func­tion. Cancer Discov. 2020;10(4):552-567.
2020;38(5):415-422. 24. Qin H, Ramakrishna S, Nguyen S, et al. Preclinical devel­op­ment of biva­lent
2. Gardner RA, Finney O, Annesley C, et al. Intent-to-treat leu­ke­mia remis­sion chi­me­ric anti­gen recep­tors targeting both CD19 and CD22. Mol Ther Onco-
by CD19 CAR T cells of defined for­mu­la­tion and dose in chil­dren and young lytics. 2018;11:127-137.
adults. Blood. 2017;129(25):3322-3331. 25. Schneider D, Xiong Y, Wu D, et  al. A tan­dem CD19/CD20 CAR lentiviral
3. Hay KA, Gauthier J, Hirayama AV, et al. Factors asso­ci­ated with dura­ble EFS vec­tor drives on-tar­get and off-tar­get anti­gen mod­u­la­tion in leu­ke­mia cell
in adult B-cell ALL patients achiev­ing MRD-neg­a­tive CR after CD19 CAR lines. J Immunother Cancer. 2017;5:42.
T-cell ther­apy. Blood. 2019;133(15):1652-1663. 26. Shah NN, Johnson BD, Schneider D, et al. Bispecific anti-CD20, anti-CD19
4. Jiang H, Li C, Yin P, et  al. Anti-CD19 chi­me­ric anti­gen recep­tor-mod­i­fied CAR T cells for relapsed B cell malig­nan­cies: a phase 1 dose esca­la­tion and
T-cell ther­apy bridg­ing to allo­ge­neic hema­to­poi­etic stem cell trans­plan­ta­ expan­sion trial. Nat Med. 2020;26(10):1569-1575.
tion for relapsed/refrac­tory B-cell acute lym­pho­blas­tic leu­ke­mia: an open- 27. Frey NV. Chimeric anti­gen recep­tor T cells for acute lym­pho­blas­tic leu­ke­
label prag­matic clin­i­cal trial. Am J Hematol. 2019;94(10):1113-1122. mia. Am J Hematol. 2019;94(suppl 1):S24-S27.
5. Maude SL, Frey N, Shaw PA, et  al. Chimeric anti­gen recep­tor T cells for 28. Lee DW, Santomasso BD, Locke FL, et  al. ASTCT con­sen­sus grad­ing for
sustained remis­sions in leu­ke­mia. N Engl J Med. 2014;371(16):1507-1517. cyto­kine release syn­drome and neu­ro­logic tox­ic­ity asso­ci­ated with
6. Maude SL, Laetsch TW, Buechner J, et  al. Tisagenlecleucel in chil­ dren immune effec­tor cells. Biol Blood Marrow Transplant. 2019;25(4):625-638.
and young adults with B-cell lym­pho­blas­tic leu­ke­mia. N Engl J Med. 29. Kadauke S, Myers RM, Li Y, et al. Risk-adapted pre­emp­tive tocilizumab to
2018;378(5):439-448. pre­vent severe cyto­kine release syn­drome after CTL019 for pedi­at­ric B-cell
7. Myers RM, Li Y, Barz Leahy A, et  al. Humanized CD19-targeted chi­me­ric acute lym­pho­blas­tic leu­ke­mia: a pro­spec­tive clin­i­cal trial. J Clin Oncol.
anti­gen recep­tor (CAR) T cells in CAR-naive and CAR-exposed chil­dren and 2021;39(8):920-930.
young adults with relapsed or refrac­tory acute lym­pho­blas­tic leu­ke­mia. 30. Kantarjian H, Stein A, Gokbuget N, et al. Blinatumomab ver­sus che­mo­ther­
J Clin Oncol. 2021;39(27):3044-3055. apy for advanced acute lym­pho­blas­tic leu­ke­mia. N Engl J Med. 2017;376:
8. Ortíz-Maldonado V, Rives S, Castellà M, et al. CART19-BE-01: a mul­ti­cen­ter ­836-847.
trial of ARI-0001 cell ther­apy in patients with CD19+ relapsed/refrac­tory 31. Kantarjian HM, DeAngelo DJ, Stelljes M, et  al. Inotuzumab ozogamicin
malig­nan­cies. Mol Ther. 2021;29(2):636-644. ver­sus stan­dard ther­apy for acute lym­pho­blas­tic leu­ke­mia. N Engl J Med.
9. Park JH, Rivière I, Gonen M, et al. Long-term fol­low-up of CD19 CAR ther­apy 2016;375(8):740-753.
in acute lym­pho­blas­tic leu­ke­mia. N Engl J Med. 2018;378(5):449-459. 32. Zhao YL, Liu DY, Sun RJ, et al. Integrating CAR T-cell ther­apy and trans­plan­
10. Wang J, Mou N, Yang Z, et al. Efficacy and safety of human­ized anti-CD19- ta­tion: com­par­i­sons of safety and long-term effi­cacy of allo­ge­neic hema­to­
CAR-T ther­apy fol­low­ing inten­sive lymphodepleting che­mo­ther­apy for poi­etic stem cell trans­plan­ta­tion after CAR T-cell or che­mo­ther­apy-based
refrac­tory/relapsed B acute lym­pho­blas­tic leu­kae­mia. Br J Haematol. com­plete remis­sion in B-cell acute lym­pho­blas­tic leu­ke­mia. Front Immunol.
2020;191(2):212-222. 2021;12:605766.
11. Shah NN, Lee DW, Yates B, et al. Long-term fol­low-up of CD19-CAR T-cell 33. Ben­ja­min R, Graham C, Yallop D, et al. Genome-edited, donor-derived allo­
ther­apy in chil­dren and young adults with B-ALL. J Clin Oncol. 2021;39(15): ge­neic anti-CD19 chi­me­ric anti­gen recep­tor T cells in pae­di­at­ric and adult
­1650-1659. B-cell acute lym­pho­blas­tic leu­kae­mia: results of two phase 1 stud­ies. Lan-
12. Shah BD, Ghobadi A, Oluwole OO, et al. KTE-X19 for relapsed or refrac­tory cet. 2020;396:1885-1894.
adult B-cell acute lym­pho­blas­tic leu­kae­mia: phase 2 results of the sin­gle- 34. Pasquini MC, Hu ZH, Curran K, et al. Real-world evi­dence of tisagenlec-
arm, open-label, multicentre ZUMA-3 study. Lancet. 2021;398(10299):491- leucel for pedi­at­ric acute lym­pho­blas­tic leu­ke­mia and non-Hodgkin lym­
502. phoma. Blood Adv. 2020;4(21):5414-5424.
13. Pan J, Zuo S, Deng B, et al. Sequential CD19-22 CAR T ther­apy induces sus-
tained remis­sion in chil­dren with r/r B-ALL. Blood. 2020;135(5):387-391.
14. Shah NN, Highfill SL, Shalabi H, et al. CD4/CD8 T-cell selec­tion affects chi­
me­ric anti­gen recep­tor (CAR) T-cell potency and tox­ic­ity: updated results
from a phase I anti-CD22 CAR T-cell trial. J Clin Oncol. 2020;38(17):1938- © 2021 by The Amer­i­can Society of Hematology
1950. DOI 10.1182/hema­tol­ogy.2021000225

6  |  Hematology 2021  |  ASH Education Program


ALL: NEW DIRECTIONS FOR ADULT PATIENTS

Acute lymphoblastic leukemia in older adults:


curtain call for conventional chemotherapy?

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/7/1852080/7luskin.pdf by guest on 13 December 2021


Marlise R. Luskin
Dana-Farber Cancer Institute, Department of Medical Oncology, Division of Leukemia, Boston, MA

Unlike younger adults with acute lymphoblastic leukemia (ALL), older adults are rarely cured due to a combination of
intrinsic disease resistance and treatment-related toxicities. Novel therapeutics such as inotuzumab ozogamicin, blinatu-
momab, venetoclax, and ABL kinase inhibitors have high activity in ALL and are well tolerated by older adults. Frontline
treatment regimens for older adults using novel therapeutics with reduction or omission of conventional chemotherapy
are being developed with early results demonstrating high remission rates and lower toxicity, but long-term efficacy and
toxicity data are lacking. Collaboration between academic and pharmaceutical stakeholders is needed to develop clini-
cal trials to define the optimal treatment regimens for older adults with ALL.

LEARNING OBJECTIVES
• Understand the role of novel therapeutics vs CC in initial treatment of older adults with Ph-negative ALL
• Understand the role of tyrosine kinase inhibitor treatment with or without CC or novel therapeutics in
initial treatment of adults with Ph-positive ALL

Outcomes of older adults in the era of conventional due to the use of pediatric-like therapy, with more than
chemotherapy 70% now achieving long-term survival, but older adults
For most of history, acute lymphoblastic leukemia (ALL) aged 55 to 60 or more years have consistently fared more
has been an uncompromising, deadly illness regardless of poorly, with less than 20% cured (Table 1).4–9 Older patients
age, comorbidities, or social circumstance. Then, in 1948, experience more toxicity, leading to dose reductions,
Sidney Farber announced 5 temporary remissions among treatment delays, and high rates of early death and treat-
16 children with acute leukemia treated with the folic acid ment-related mortality in remission.4–7,11,12 In addition, high-
antagonist, aminopterin.1 Now, after decades of effort by risk genetic features are more common, leading to fewer
numerous clinicians, scientists, and cooperative groups, remissions and frequent relapse.10,16 Age-based dose mod-
more than 90% of children in resourced settings are cured ifications and prospective studies of CC designed specifi-
of ALL with chemotherapy.2 Although celebrated, these cally for older adults have not improved outcomes.4,5,12–14,17
pediatric ALL chemotherapy regimens are notable for There remains no accepted standard-of-care CC regimen
length, complexity, and toxicity. for older adults with ALL.
Traditionally thought of as a pediatric disease, approxi- Clinicians caring for older adults with ALL perceive lim-
mately half of ALL diagnoses occur in younger (18–49 years) ited benefit of CC. A 2019 US Medicare analysis revealed
and older (≥50 years) adults in roughly equal porportions.3 that only 53.5% of patients with ALL aged 66 or more years
Unfortunately, the outstanding outcomes in children received any treatment within 90 days of diagnosis.18 A
have not been replicated in older cohorts. The standard 2017 US Surveillance, Epidemiology, and End Results analy-
approach to treating ALL in adults has been conventional sis reported a median overall survival (OS) of just 4 months
chemotherapy (CC) programs adapted from pediatric among adults 60 years or older diagnosed with ALL since
schedules but with more myelosuppressive agents and a 1980, with minimal progress over 3 decades (3-year OS
deemphasis on the noncytotoxic agents that feature prom- increasing from 10% to 16%).19 In summary, the chemother-
inently in pediatric regimens (corticosteroids, vincristine, apy regimens that cure children are not a good match for
and asparaginase). Results in young adults have improved older adults.10,16

Prakash Singh Shekhawat


ALL curtain call chemo | 7
Table 1.  Outcomes of older adults treated with CC, select tri­als (see also Gökbuget10)

Trial Age, y N CR, % Early ­mor­tal­ity,% OS, % (95% CI) Death in CR, among those achiev­ing CR, %
Adult tri­als, out­comes by age
  CALGB 91115 All 185 85 8 3 years: 43 (36-50) 8
≥60 35 77 17 17 (9-31) —
30-59 42 84 — 40 (30-51) —
<30 39 90 — 57 (46-68) —
  ECOG 2993/UKALL XII7,11 55-65 100 73 18 5 years: 21 (12-20) 23
<55 1814 93 4 41 (39-43) —
 Hyper-CVAD 12
≥60 122 84 10 5 years: 20 34
<60 409 92 2 48 7

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/7/1852080/7luskin.pdf by guest on 13 December 2021


Prospectively designed for older adults
  DFCI reg­i­men13 51-75 30 67 13 2 years: 52 (33-68) —
60-75 13 76 — 70 (42-86)
51-59 17 58 — 31 (10-55)
 GMALL14 55-85 268 76 14 5 years: 23 6
55-65 — — 7 — —
65-75 — — 14 — —
≥75 — — 37 — —
  PETHEMA ALLOLD0715 >55 56 74 13 32 (at last fol­low-up, 3
median 11.4 months)
DFCI, Dana-Farber Cancer Institute.

Novel approaches to Philadelphia chro­mo­some–neg­a­tive of older adults will allow toxic CC to be de-esca­lated or omit­ted,
ALL in older adults resulting in both improved response rates and less tox­ic­ity.

IO plus CC
Investigators at MD Anderson Cancer Center (MDACC) have been
CASE 1
test­ing IO plus mini-hyper-CVD (cyclophosphamide, vincristine,
A 78-year-old man with cor­o­nary artery dis­ease, hyper­ten­sion, dexamethasone), a lower-inten­sity CC reg­i­men, in older adults
and hyper­ lip­
id­
emia devel­ oped sev­ eral weeks of fatigue and (≥60 years) with untreated Philadelphia chro­mo­some–neg­a­tive
weight loss. Prior to ill­ness, he vis­ited the gym reg­u­larly engag­ (Ph–) CD22+ B-ALL in a sin­gle-cen­ter phase 2 trial (NCT01371630,
ing in vig­or­ous exer­cise. Laboratory work revealed pan­cy­to­pe­nia Table 2). In the orig­i­nal design, IO was admin­is­tered on day 3 of
(white blood cell count, 2.9 K/µL; hemo­glo­bin, 11.2 g/dL; plate­ the first 4 of 8 planned CC cycles followed by 36 months of POMP
lets, 86 K/µL) with 7% cir­cu­lat­ing blasts. He was diag­nosed with (6-mercaptopurine, vincristine, methotrexate, prednisone) main­
B-cell acute lymphoblastic leukemia (B-ALL). Immunophenotype te­nance. In 2018, MDACC inves­ti­ga­tors reported high response
was CD45+, CD34+, HLA-DR+, TdT+, CD19+ (var­ i­
able), CD22+, rates (98% overall response rate [ORR], 96% minimal residual
CD10−, and CD20−. Karyotype was com­plex with no evi­dence disease-negativity [MRD-negativity]) and no early mor­tal­ity in
of the Philadelphia chro­mo­some. Next-gen­er­a­tion sequenc­ing the first 52 patients treated.22 An update of the trial in 2020 (with
revealed a path­o­genic muta­tion in TP53. In 2019, he was referred 70 patients enrolled) was nota­ble for con­tin­ued high response
to our aca­demic cen­ter. rates (98% ORR, 96% MRD negativity, with no early deaths) and
an encour­ag­ing 3-year com­plete remis­sion (CR) dura­tion and OS
of 79% and 56%, respec­tively.23
The devel­op­ment of novel che­mo­ther­apy agents—blinatu- Notable toxicities asso­ ci­
ated with the IO plus mini-hyper-
momab and inotuzumab ozogamicin (IO)—for relapsed B-ALL CVD reg­i­men have included fre­quent prolonged throm­bo­cy­to­
has led to a reimagining of the treat­ment for older adults with pe­nia (81%), hep­a­to­tox­ic­ity (17% grade ≥3 hyperbilirubinemia,
ALL. IO is a CD22 mono­clo­nal anti­body cova­lently linked to cali- 9% veno-occlu­sive dis­ease), and infec­tions (41% dur­ing induc­
cheamicin, a cyto­toxic agent (anti­body–drug con­ju­gate). Blina- tion, 70% dur­ing con­sol­i­da­tion).23 Death in remis­ sion due to
tumomab is a bispecific T-cell engager targeting CD19 and CD3. treat­ment-related tox­ic­ity has been the major chal­lenge. At the
Each is now approved by the US Food and Drug Administration most recent update, 34% (24/70) of responding patients had
(FDA) for relapsed and refrac­tory (R/R) ALL based on phase 3 died in CR due to sep­sis, veno-occlu­sive dis­ease, or sec­ond­ary
ran­dom­ized trial data show­ing supe­ri­or­ity of the novel agent mye­loid malig­nan­cies. Mortality was more com­mon in patients
to sal­vage cyto­toxic che­mo­ther­apy in the R/R set­ting.20,21 It is 70 years or older who com­prised ~40% of enrolled patients. In
hoped that using IO and blinatumomab in the first-line treat­ment response to an ini­tial expe­ri­ence with tox­ic­ity, pro­to­col mod­i­

8  |  Hematology 2021  |  ASH Education Program


Table 2.  Novel approaches to Ph– ALL in older adults: published and ongo­ing tri­als*

Regimen-related
Reference Regimen Phase N Line Age, y deaths Response Survival
Kantarjian et al IO + mini-hyper- 2 70 First ≥60 0% early mor­tal­ity 98% ORR Continuous CR
(2018)22 CVD (Blina 34% mor­tal­ity in 88% CR (96% MRD 3 years: 79%
Short et al (2020)23 con­sol­i­da­tion) remis­sion neg­a­tive) Median CCR NR
NCT01371630 OS
3 years: 56%
Median OS 62
months
EWALL-INO IO + mild-inten­sity 2 — First ≥55 — — —
NCT03249870 che­mo­ther­apy
CC con­sol­i­da­tion
Stelljes et al IO induc­tion 2 36 First ≥56 0% early mor­tal­ity 100% CR/CRi EFS

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/7/1852080/7luskin.pdf by guest on 13 December 2021


(2020)24 CC con­sol­i­da­tion (78% MRD 1 year: 87% (95%
GMALL (INITIAL-1) neg­a­tive) CI, 70-100)
NCT03460522 OS
1 year: 87% (95%
CI, 70-100)
Jain et al (2019)25 Venetoclax + 1b/2 19 R/R (8) First (11) All ages ≥60 0% early mor­tal­ity 91% CR (100% —
DFCI/MDACC IST mini-hyper-CVD MRD neg­a­tive)
NCT03319901
Advani et al Blina induc­tion 2 29 First ≥65 0% 66% CR/CRi (92% DFS
(2018)26 Blina con­sol­i­da­tion MRD neg­a­tive) 1 year: 56% (95%
SWOG 1318 CI, 58-90)
NCT02143414 OS
1 year: 65% (95%
CI, 43-80)
Alliance 041703 IO induc­tion 2 — R/R First All ages ≥60 Results pend­ing Results pend­ing Results pend­ing
NCT03739814 Blina con­sol­i­da­tion
*Results at most recent pub­li­ca­tion.
Blina, blinatumomab; CCR, con­tin­u­ous com­plete remis­sion; CRi, complete remission with incomplete count recovery; NR, not reached.

fi­
ca­tions have included IO dose frac­ tion­
ation and reduc­ tion dose-limiting toxicities (DLTs) or early mor­tal­ity), with venetoclax
(to address liver tox­ic­ity) and a dras­tic decrease in the num­ber 600 mg daily declared the recommended phase 2 dose.25 Nota-
of recommended CC cycles (orig­i­nally 8, decreased to 4 for bly, there was no evi­dence of prolonged cytopenias or liver tox­ic­
patients aged 60-69 years and to 2 for patients aged ≥70 years). ity. The reg­i­men was par­tic­u­larly effec­tive in newly diag­nosed pa-
The Euro­pean phase 2 EWALL (European Working Group for tients, with 91% (10/11) achiev­ing an MRD-neg­a­tive CR, includ­ing
Adult ALL)-INO study (NCT03249870) is also explor­ing IO in com­ 6 patients with a TP53 muta­tion. Because 9 of 10 respond­ers were
bi­na­tion with low-inten­sity che­mo­ther­apy for untreated patients con­sol­i­dated with allo­ge­neic hematopoietic stem cell transplant
55 years or older with CD22+ ALL. Patients receive IO plus low- (HSCT), there are lim­ited data on late tox­ic­ity. The trial has now
inten­sity che­mo­ther­apy induc­tion for 2 cycles followed by IO- moved to phase 2 (with venetoclax 400  mg) and is cur­ rently
free che­mo­ther­apy con­sol­i­da­tion and main­te­nance. enroll­ing adults 60 years or older with newly diag­nosed Ph– ALL.
In con­trast to the com­bi­na­tion approach taken by MDACC A dis­tinct advan­tage of this reg­i­men is that it can be offered to
and EWALL, the German Multicenter Study Group for Adult patients with both B- and T-cell ALL, in con­trast to approaches
Acute Lymphoblastic Leukemia (GMALL) study group is study­ that rely on B-lin­e­age restricted antibodies and anti­body–drug
ing a sequen­ tial approach. In the phase 2 INITIAL-1 study con­ju­gates. Venetoclax is not approved by the FDA for ALL.
(NCT03460522), untreated older adults (aged >55 years) receive
3 cycles of IO monotherapy followed by CC con­sol­i­da­tion (Table Closing the cur­tain on che­mo­ther­apy for Ph– ALL?
2). A 100% CR was reported among the first 31 patients treated Given the effi­cacy of novel agents, a che­mo­ther­apy-free approach
(78% MRD neg­a­tive), with the major­ity (29/31) ­able to com­plete may be pos­si­ble. The US National Clinical Trial Network (NCTN)
all 3 IO induc­tion cycles with no early deaths.24 Long-term effi­ trial Southwest Oncology Group (SWOG) 1318 phase 2 study of
cacy and tox­ic­ity of the reg­i­men are not yet known. blinatumomab induc­tion followed by blinatumomab con­sol­i­da­
tion for newly diag­nosed Ph– B-ALL (aged >65 years) reported a
Venetoclax plus CC 66% CR rate (92% MRD neg­a­tive) among the first 29 patients with
The BCL2 inhib­i­tor venetoclax, an agent with dem­on­strated pre­ no early deaths (NCT02143414, Table 2).26 Another phase 2 NCTN
clin­i­cal and clin­i­cal activ­ity in relapsed ALL, is also being stud­ied in trial (Alliance 041703 NCT03739814) is inves­ti­gat­ing IO induc­tion
the front­line in com­bi­na­tion with mini-hyper-CVD (NCT03319901, followed by blinatumomab con­sol­i­da­tion (Table 2). This approach
Table 2).27,28 A phase 1b study of venetoclax plus mini-hyper-CVD seeks to use each novel agent to max­i­mum advan­tage. IO is ac-
for relapsed/refractory (R/R) (n = 8) and newly diag­ nosed pa- tive regard­less of dis­ease bur­den, mak­ing it an ideal induc­tion
tients 60 years or older (n = 11) with ALL Prakash Singhsafety
dem­on­strated Shekhawat
(no agent, whereas blinatumomab is more effec­tive and less toxic in

ALL cur­tain call chemo | 9


set­tings of lower dis­ease bur­den and thus a good agent for con­ mer­ase chain reac­tion shows p210 tran­script more than 50%
sol­i­da­tion ther­apy.15,29 international scale (IS).

Encore for che­mo­ther­apy


Blinatumomab and IO achieve high response rates in the R/R set­ The Philadelphia chro­mo­some is pres­ent in approx­i­ma­tely
ting but with short dura­bil­ity.20,21 Although the dura­bil­ity of sin­gle- half of ALLs in older adults.31 Historically asso­ ci­
ated with an
agent novel ther­apy in the upfront set­ting is not known, relapses adverse prog­no­sis, the devel­op­ment of ABL kinase inhib­i­tors has
driven by selec­tive pres­sure would be expected. It is hypoth­e­ resulted in fre­quent cures in youn­ger adults and in older adults
sized that low-inten­sity CC inte­grated into induc­tion and/or con­ has enabled prolonged remis­sions, negat­ing the adverse prog­
sol­id
­ a­tion (as being pur­sued by EWALL-INO and GMALL INITIAL-1 no­sis.32-34 Older adults with ALL are ideal can­di­dates for low-
stud­ies) may con­tinue to ben­e­fit older patients receiv­ing novel inten­sity CC and che­mo­ther­apy-free tyro­sine kinase inhib­i­tor
agents by apply­ ing broader anti­ leu­ke­
mic pres­ sure and dimin- (TKI)–based pro­to­cols.
ishing oppor­ tu­ni­
ties for clonal escape. Novel com­ bi­
na­tion ap-
proaches (such as being stud­ied in A041703) may offer the best TKI plus CC

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/7/1852080/7luskin.pdf by guest on 13 December 2021


of both worlds via a mul­ti­pronged attack with­out che­mo­ther­apy The EWALL has stud­ied TKI plus low-inten­sity CC in adults 55
toxicities. MDACC has added blinatumomab con­ sol­

da­tion to years or older (NCT028889777, Table 3). Second-gen­er­a­tion TKIs
their IO plus mini-hyper-CVD pro­to­col. Venetoclax plus IO will also were com­bined with low-inten­sity induc­tion and con­sol­i­da­tion
soon be tested in R/R B-ALL due to non­over­lap­ping toxicities and CC in the con­sec­u­tive EWALL-PH01 (dasatinib) and EWALL-PH02
pre­clin­i­cal evi­dence for syn­ergy.30 If shown to be safe, it may be (nilotinib) pro­to­cols. Both pro­to­cols resulted in high CR rates
use­ful as a front­line reg­i­men. For T-cell ALL, a che­mo­ther­apy-free (>95%) with­out induc­tion mor­tal­ity. Long-term sur­vival was par­
option does not yet exist. More research to iden­tify ther­a­peu­tic tic­u­larly encour­ag­ing in the EWALL-PH02, in which allo­ge­neic
vulnerabilities and novel ther­ap ­ eu­tics for this sub­type is needed. HSCT was pur­sued in 30% (24/79) of patients (4-year OS, 47%; 61%
and 39% in transplanted vs nontransplanted, respec­tively).35,36
Importantly, the Group for Research on Adult Acute Lymphoblas-
tic Leukemia (GRAAPH)-2005 trial, which enrolled youn­ger adults
CASE 1 (con­tin­ued) (18-59 years), dem­on­strated that reduced-inten­sity CC induc­tion
The patient was treated with venetoclax plus mini-hyper-CVD was asso­ci­ated with fewer induc­tion deaths and equiv­a­lent effi­
on an inves­ti­ga­tional pro­to­col and achieved an MRD-neg­a­tive cacy com­pared with stan­dard-inten­sity CC induc­tion, question-
CR after 1 cycle. He com­pleted 4 cycles of venetoclax plus ing the ben­e­fit of inten­sive CC induc­tion for any patient (regard­
mini-hyper-CVD, after which he was bridged to venetoclax less of age) with Ph+ ALL in the TKI era.37
plus POMP main­te­nance due to tox­ic­ity from CC. He con­tin­
ued POMP main­te­nance for 2 years and remains in an ongo­ing Curtain call on che­mo­ther­apy for Ph± ALL?
remis­sion. (See Figure 1 for approach to Ph– ALL in older adults.) The Ital­ian Gruppo Italiano Malattie EMatologiche dell’Adulto
(GIMEMA) group has pioneered a che­mo­ther­apy-free induc­tion
approach for Ph+ ALL (Table 3). Imatinib plus cor­ti­co­ste­roids
induced uni­ver­sal hema­to­logic remis­sions, although with lim­
Novel approaches to Ph+ ALL in older adults
ited depth (only 1 of 29 achieved a com­plete molec­u­lar remis­
sion) and dura­bil­ity (median dura­tion of hema­to­logic response
of 8 months).38 Subsequently, the more potent TKI dasatinib
CASE 2 was com­bined with cor­ti­co­ste­roids, with more patients achiev­
A 73-year-old woman with a his­tory of hyper­ten­sion and rheu­ ing deep molec­u­lar remis­sions (52.1% achieved BCR-ABL level
ma­toid arthri­tis was asymp­tom­atic but found to have an abnor­ <10−3 by day 85).39,40 The orig­i­nal GIMEMA stud­ies were induc­
mal com­plete blood count on rou­tine test­ing: white blood cells tion-only stud­ies, with sub­se­quent con­sol­i­da­tion ther­apy for
(19 K/µL) with 39% cir­cu­lat­ing blasts that are CD45+, CD34+, each patient dic­tated by the treating phy­si­cian. The US NCTN
HLA-DR+, TdT+, CD19+, CD10+, and CD20−. Karyotype shows Alliance 10701 phase 2 study (NCT01256398, Table 3) tested a
t(9;22) with addi­tional abnor­mal­i­ties. Reverse tran­scrip­tion poly­ che­mo­ther­apy-free dasatinib plus ste­roid induc­tion followed

• Careful assessment of comorbidities and performance status.


• Formal geriatric assessment if able.
• Enroll in clinical trial of novel agent therapy, wherever possible.
• If treating with conventional chemotherapy, apply dose reductions as appropriate
(particularly for asparaginase).
• Ensure CNS prophylaxis is administered per regimen.
• Monitor response per protocol and include MRD assessment (flow cytometry or
molecular).
• Consider referring for allogeneic HSCT if fit and high-risk genetics or persistent
MRD.

Figure 1.  How I treat Ph– ALL in older adults.

10  |  Hematology 2021  |  ASH Education Program


Table 3.  Novel approaches to Ph+ ALL in older adults: published and ongo­ing tri­als*

Regimen-
Reference Regimen Phase N Line Age, y related deaths Response Survival
Rousselot et al Dasatinib 140 mg 2 71 First ≥55 4% (3/71) 96% CR 27% (95% CI, 17-37)
(2016)35 daily + low- induc­tion 65% 3-log ­ 5-year EFS
EWALL PH-01 inten­sity CC 12% (6/71) reduc­tion in 36% (95% CI, 25-47)
NCT028889777 treat­ment- BCR-ABL 5-year OS
related ­ 7 received HSCT
mor­tal­ity 75% of relapses T315I
in CR
Ottmann et al Nilotinib 400 mg 2 79 First ≥55 1% (1/79) 94.4% CR 42% 4-year EFS
(2018)36 twice daily + induc­tion 47% 4-year OS (61%
EWALL PH-02 low-inten­sity 11 died in CR (6 transplanted, 39%
NCT028889777 CC after HSCT) nontransplanted)

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/7/1852080/7luskin.pdf by guest on 13 December 2021


24 received alloHSCT, 3
received autoHSCT
Vignetti et al Imatinib 800 mg 2 29 First >60 0% induc­tion 100% CR 48% (95% CI, 28-69)
(2007)38 daily + 2 died in CR 1/27 CMR 1-year DFS
GIMEMA LAL0101-B pred­ni­sone 74% (95% CI, 54-94)
(induc­tion 1-year OS
pro­to­col only)
Foà et al (2011)39 Dasatinib 70 mg 2 53 First ≥18 0% induc­tion 100% CR 69% (95% 61-79)
GIMEMA LAL1205 twice daily + (22%> 60) 52.1% 3-log 20-month OS
pred­ni­sone reduc­tion in 51% (95% CI, 44-59)
(induc­tion BCR-ABL by 20-month DFS
pro­to­col only) day 85
Martinelli et al Ponatinib 45 mg 2 42 First >60 or unfit 0% induc­tion 95.5% CR 87.5% (95% CI, 76.5%-
(2017)40 daily + 1 death in 45% CMR at 24 99.9%) 1-year OS
GIMEMA LAL1811 pred­ni­sone fol­low-up weeks
NCT01641107 (induc­tion pro­to­ ponatinib
col only)
Luskin et al (2019)42 Dasatinib 140 + 1 8 First >50 0% induc­tion 100% CR Not reported
DFCI IST asciminib (dose
NCT03595917 esca­la­tion)
(induc­tion
pro­to­col only)
Foà et al (2020)43 Dasatinib 140 + 2 63 First ≥18 1.6% induc­tion 95% CR With median fol­low-up
GIMEMA LAL2116 pred­ni­sone (median 54) (1 patient) 18 months
NCT02744768 Blinatumomab No reg­i­men 95% (95% CI, 90-100) OS
con­sol­i­da­tion related 88% (95% CI, 80-97) DFS
deaths in CR
Wieduwilt et al Dasatinib 140 + 2 64 First ≥18 (median 0% induc­tion 97% CR 43% 3-year DFS
(2018)41 dexa­meth­a­ 60) deaths (55% allo, 43% auto, 46%
NCTN/Alliance 10701 sone induc­tion chemo)
NCT01256398 CC con­sol­i­da­tion 55% 3-year OS (63% ≤ 60
Allo- vs autoHSCT years, 49%> 60 years)
(75% allo, 71% auto, 55%
chemo)
Relapse
25% allo, 43% auto, 37%
chemo
NCTN/EA9181 TKI + pred­ni­sone 3 — First ≥18-75 Results Results Results pend­ing
NCT04530565 induc­tion pend­ing pend­ing
TKI + hyper-
CVAD vs TKI +
blinatumomab
con­sol­i­da­tion
SWOG 1318 Dasatinib + 2 — First ≥65 Results Results Results pend­ing
NCT02143414 pred­ni­sone pend­ing pend­ing
induc­tion
Dasatinib +
blinatumomab
con­sol­i­da­tion
*Results at most recent pub­li­ca­tion.
CMR, complete molecular response; DFS, dis­ease-free sur­vival; EFS, event-free sur­vival.
Prakash Singh Shekhawat
ALL cur­tain call chemo | 11
by dasatinib plus CC con­sol­i­da­tion and then trans­plant (allo­ge­ early results (NCT02744768, Table 3).43 The US NCTN SWOG
neic vs autol­o­gous HSCT or che­mo­ther­apy based on avail­abil­ity 1318 Cohort 2 (NCT02143414) is study­ing a sim­i­lar approach of
of an human leukocyte antigen (HLA)-matched donor and fit­ dasatinib-ste­roid induc­tion followed by blinatumomab con­sol­
ness).41 Deep remis­sions were con­sis­tently obtained (97% CR, i­da­tion. A study of ponatinib-ste­roid induc­tion followed by bli-
75% ulti­mately achiev­ing a com­plete molec­u­lar response) with natumomab con­ sol­

da­
tion is also planned (NCT04722848). An
3-year OS of 55% (49% in patients aged >60 years). open ques­tion is whether blinatumomab con­sol­i­da­tion should
Although the GRAAPH-2005 study showed that inten­sive CC be applied to all­patients or used in a risk-adapted man­ner. Fre-
induc­tion was not advan­ta­geous com­pared with lower-inten­sity quency of cen­tral ner­vous sys­tem (CNS) relapse will also need to
CC induc­tion in the con­text of TKIs, the ben­e­fit of adding CC to be mon­i­tored over time in patients treated entirely with TKIs and
TKI-based con­sol­i­da­tion, par­tic­u­larly in patients bridg­ing to allo­ novel agents (with only intrathecal (IT) che­mo­ther­apy admin­is­
ge­neic HSCT, is not known. tered to pre­vent CNS relapse).
Other active agents in ALL, includ­ing IO and venetoclax, also
Approaches to pre­vent relapse in Ph± ALL require eval­u­a­tion as part of treat­ment of Ph+ ALL. The Alliance
Despite improve­ments with TKI-based ther­apy, relapse remains 041703 is plan­ning to add an arm for Ph+ patients test­ing an IO

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/7/1852080/7luskin.pdf by guest on 13 December 2021


com­mon, with T315I-driven relapses a par­tic­u­lar men­ace.35,39 Ap- plus ponatinib induc­tion.
proaches to address this issue include use of ponatinib, which The US NCTN is cur­rently accru­ing to EA9181 (NCT04530565),
is active against T315I; dual ABL kinase inhi­bi­tion with dasatinib a phase 3 study of patients up to age 70 years who receive
and asciminib; and addi­tion of novel agent con­sol­i­da­tion with che­mo­ther­apy-​free induc­tion of TKI plus ste­roids and then are
blinatumomab. ran­ dom­ ized to TKI (dasatinib or ponatinib) plus hyper-​
The GIMEMA group has extended its che­mo­ther­apy-free induc­ cyclo­ phosphamide, vincristine, doxorubicin, dexamethasone
tion approach to ponatinib plus ste­roids in older adults (aged ≥60 (CVAD) vs TKI plus blinatumomab con­sol­i­da­tion to com­pare CC
years), with encour­ ag­ing early results (NCT01641107, Table 3).40 vs novel agent con­sol­i­da­tion. Eligible patients undergo trans­
Although tox­ic­ity appears rea­son­able with dose reduc­tions (only plan­ta­tion when MRD neg­a­tive, and all­patients receive high-
15/42 remained at 45 mg daily at week 24), the risk of car­dio­vas­cu­ dose meth­o­trex­ate for CNS pro­phy­laxis.
lar tox­ic­ity with ponatinib remains a con­cern in older patients. The
planned EWALL-03 study (NCT04688983) is ran­dom­iz­ing patients
to imatinib vs ponatinib in com­bi­na­tion with low-inten­sity CC and CASE 2 (con­tin­ued)
may pro­vide insight regard­ing risk/ben­e­fit ratio of later-gen­er­a­tion The patient was treated with dasatinib and asciminib on a clin­
TKIs. A “risk-adapted” approach using early response mile­stones to i­cal trial. She tol­er­ated ther­apy well ini­tially but had an inad­e­
select which patients require esca­la­tion of ther­apy to more potent quate response after 3 cycles of ther­apy (CR but MRD+ by flow
(and more toxic) later-gen­er­at­ ion TKIs would be attrac­tive. cytom­et­ry, BCR/ABL 6.3% IS). She required mul­ti­ple dasatinib
Combination ABL kinase inhi­bi­tion is being explored in a phase dose reduc­tions due to recur­rent pleu­ral effu­sions. She was
1 study treating newly diag­nosed adults 50 years or older with transitioned to ponatinib 30  mg daily and con­tin­ues in remis­
Ph+ ALL with dasatinib and asciminib, a novel myristoyl bind­ing sion (BCR-ABL p210 tran­script unde­tect­able) now 2 years from
pocket inhib­i­tor (NCT03595917, Table 3). The hypoth­e­sis is that diag­no­sis. (See Figure 2 for approach to Ph– ALL in older adults.)
dual ABL inhi­bi­tion may induce deeper remis­sions and pre­vent
T315I relapses. Thus far, the reg­i­men appears well tol­er­ated, with
asymp­ tom­ atic amy­
lase and lipase ele­ va­tions being the most Additional issues in advanc­ing care for older adults
nota­ble adverse event.42 This study is ongo­ing to estab­lish tol­er­ with ALL
a­bil­ity and recommended dose. The out­look for older adults diag­nosed with ALL is improv­ing
The GIMEMA D-ALBA trial added dasatinib-blinatumomab rap­idly. Additional issues fac­ing the field are sum­ma­rized in
con­sol­i­da­tion to dasatinib-ste­roid induc­tion with prom­is­ing Figure 3.

 Careful assessment of comorbidities and performance status.


 Formal geriatric assessment if able.
 Enroll in clinical trial of novel agent therapy, wherever possible.
 I prefer a chemotherapy-free TKI-based induction using imatinib or dasatinib, or
follow published regimen (Table 3).
 Ensure CNS prophylaxis is administered per regimen.
 Monitor response per protocol and include MRD assessment (BCR-ABL
transcript).
 Escalate treatment (later-generation TKI or additional novel agents) if
inadequate response.
 Refer for consideration of allogeneic HSCT if fit.
Figure 2.  How I treat Ph+ ALL in older adults.

12  |  Hematology 2021  |  ASH Education Program


• Assess and study MRD and other surrogate end points.
• Nonrestrictive inclusion criteria.
• Include quality-of-life and geriatric assessment measures.
• Design strategies to increase recruitment of participants from underrepresented
groups including nonwhite and rural patients.
• Academic and pharmaceutical industry collaborations.
Figure 3.  Important components for ALL clinical trials in older adults.

Curability of older adults 042001). The objec­tive of this trial will be to estab­lish a novel
Although sur­vival is poor among older adults treated with CC, agent–based bench­mark reg­i­men to which other novel agent
a few fit older patients may tol­er­ate and be cured by CC reg­ reg­i­mens can be sub­se­quently com­pared (Figure 4).

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/7/1852080/7luskin.pdf by guest on 13 December 2021


i­mens.5,11,12,44,45 In con­trast, there is no evi­dence yet that novel
agent approaches are cura­tive, although they may induce dura­ Surrogate end points and risk-adapted approaches
ble MRD-neg­ a­tive remis­sions. Patients responding to novel Early response end points (such as MRD neg­a­tiv­ity) should be
approaches may become eli­gi­ble for cura­tive-poten­tial allo­ge­ stud­ied to con­firm validity as sur­ro­gates for sur­vival in older
neic HSCT, but the role of HSCT is not clear and has not been adults. This could facil­i­tate rapid com­ple­tion of tri­als as well as
con­firmed to be supe­rior to CC for older adults with Ph– ALL devel­op­ment of risk-adapted approaches.
in first CR in the CC era.46 The Alliance/CALGB 10701 study has
attempted to address this ques­tion for Ph+ ALL by assigning Equity
adult patients (aged 18-69 years) to reduced intensity condition- Novel agents are expen­sive and likely to increase disparities be-
ing (RIC) allo­ge­neic HSCT vs autol­o­gous HSCT based on avail­ tween resourced and underresourced set­tings. Advocacy for im-
abil­ity of a fully matched donor (Table 3).41 Although 3-year OS proved access is needed.
was sim­i­lar between those receiv­ing allo­ge­neic HSCT and other
con­sol­i­da­tion approaches, patients assigned to the allo­ge­neic Representative enroll­ment and geri­at­ric assess­ment
approach were less likely to relapse. Almost every older adult has a med­i­cal prob­lem other than ALL.
Trial eli­gi­bil­ity should be designed to allow rep­re­sen­ta­tive enroll­
Need for ran­dom­ized com­par­i­son ment. Unnecessarily restric­ tive inclu­sion cri­te­
ria that exclude
Results of novel agent–based reg­i­mens tested in nonrandom- patients based on advanced age, mildly com­ pro­
mised organ
ized stud­ies are extremely encour­ag­ing, with strong evi­dence func­tion, or “clin­i­cally insig­nif­i­cant” con­cur­rent malig­nan­cies
for supe­ri­or­ity com­pared with his­tor­i­cal CC reg­i­mens based should be avoided. Geriatric assess­ment (GA) should be rou­tinely
on high response rates and low tox­ic­ity. The next step will be incor­po­rated into ALL tri­als of older adults (with finan­cial sup­port
to for­mally com­pare a novel and a “stan­dard” CC approach in to ensure com­ple­tion). GA has been shown to be fea­si­ble in coop­
Ph– ALL, and thus the NCTN is design­ing a ran­dom­ized phase er­a­tive group can­cer tri­als, includ­ing in an Alliance trial for acute
2 study of IO plus mini-hyper-CVD (the novel reg­i­men with the mye­loid leu­ke­mia.47,48 The phase 2 NCTN trial in devel­ op­ment
most expe­ri­ence to date) vs age-adjusted hyper-CVAD (Alliance plans to incor­po­rate GA into its study design.

Negative

winner

Figure 4. Prakash Singh Shekhawat


Treating Ph-negative B-cell ALL in older adults: future trial landscape.

ALL cur­tain call chemo | 13


Quality-of-life assess­ment 8. DeAngelo DJ, Stevenson KE, Dahlberg SE, et al. Long-term out­come of a
pedi­at­ric-inspired reg­i­men used for adults aged 18-50 years with newly
Quality-of-life end points should be given impor­tance. Incremen-
diag­nosed acute lym­pho­blas­tic leu­ke­mia. Leukemia. 2015;29(3):526-534.
tal improve­ments in effi­cacy may not be “worth it” to an older 9. Stock W, Luger SM, Advani AS, et  al. A pedi­at­ric reg­i­men for older ado­
adult if it is asso­ci­ated with sig­nif­i­cant tox­ic­ity and treat­ment les­cents and young adults with acute lym­pho­blas­tic leu­ke­mia: results of
com­plex­ity. Similarly, new reg­i­mens with equiv­a­lent responses CALGB 10403. Blood. 2019;133(14):1548-1559.
but bet­ter tol­er­a­bil­ity may war­rant fur­ther devel­op­ment. 10. Gökbuget N. Treatment of older patients with acute lym­pho­blas­tic leu­ke­
mia. Hematology Am Soc Hematol Educ Program. 2016;2016(1):573-579.
11. Sive JI, Buck G, Fielding A, et al. Outcomes in older adults with acute lym­
Cooperation to clob­ber the con­ven­tional approach pho­ blas­
tic leu­
kae­mia (ALL): results from the inter­ na­
tional MRC UKALL
By far the big­gest chal­lenge in adult ALL will be to effi­ciently XII/ECOG2993 trial. Br J Haematol. 2012;157(4):463-471.
design and con­duct log­ic ­ al, sequen­tial stud­ies to answer key 12. O’Brien S, Thomas DA, Ravandi F, Faderl S, Pierce S, Kantarjian H. Results
of the hyperfractionated cyclo­phos­pha­mide, vin­cris­tine, doxo­ru­bi­cin, and
ques­tions and make sure patient care “keeps up” with sci­en­
dexa­meth­a­sone reg­i­men in elderly patients with acute lym­pho­cytic leu­ke­
tific advances. In a rare dis­ease, this is no small task. Trials will mia. Cancer. 2008;113(8):2097-2101.
need to be devel­oped col­lab­o­ra­tively, opened widely with eq- 13. Fathi AT, DeAngelo DJ, Stevenson KE, et  al. Phase 2 study of inten­si­fied
uity con­sid­ered, and conducted effi­ciently so that ques­tions che­mo­ther­apy and allo­ge­neic hema­to­poi­etic stem cell trans­plan­ta­tion for

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/7/1852080/7luskin.pdf by guest on 13 December 2021


can be iter­a­tively asked and answered for the bet­ter­ment of older patients with acute lym­pho­blas­tic leu­ke­mia. Cancer. 2016;122(15):​
2379-2388.
patients. In the United States, a NCTN work­ing group involv­ing
14. Gokbuget N, Beck J, Bruggemann M. Moderate inten­sive che­mo­ther­apy
rep­re­sen­ta­tives from Alliance, ECOG, and SWOG has formed to includ­ing CNS-pro­phy­laxis with lipo­so­mal cytarabine is fea­si­ble and effec­
coor­di­nate efforts. tive in older patients with Ph-neg­a­tive acute lym­pho­blas­tic leu­ke­mia (ALL):
results of a pro­spec­tive trial from the Ger­man Multicenter Study Group for
Conclusion Adult ALL (GMALL). Blood. 2012;120(21):1493.
15. DeAngelo DJ, Advani AS, Marks DI, et  al. Inotuzumab ozogamicin for
An older adult diag­nosed with ALL in 2021 has more prom­is­ing relapsed/refrac­tory acute lym­pho­blas­tic leu­ke­mia: out­comes by dis­ease
treat­ment options than an adult diag­nosed even 5 years ago. The bur­den. Blood Cancer J. 2020;10(8):81.
first task is to expe­di­tiously replace the unac­cept­able tra­di­tional 16. Gökbuget N. How I treat older patients with ALL. Blood. 2013;122(8):1366-
CC approaches with novel approaches via prac­tice chang­ing 1375.
17. Ribera J-M, García O, Oriol A, et al; PETHEMA Group, Span­ish Society of
clin­i­cal tri­als. Then we must work to fur­ther define the opti­mal
Hematology. Feasibility and results of sub­type-ori­ented pro­to­cols in older
treat­ment for all­ patients con­sid­er­ing indi­vid­ual (age, comorbid- adults and fit elderly patients with acute lym­pho­blas­tic leu­ke­mia: results
ities, and frailty) and dis­ease (immunophenotype, genet­ics, re- of three pro­spec­tive par­al­lel tri­als from the PETHEMA group. Leuk Res.
sponse mile­stones) fea­tures and per­sonal pref­er­ences. 2016;41:12-20.
18. Kim C, Molony JT, Chia VM, Kota VK, Katz AJ, Li S. Patient char­ac­ter­is­tics,
treat­ment pat­terns, and mor­tal­ity in elderly patients newly diag­nosed with
Conflict-of-inter­est dis­clo­sure ALL. Leuk Lymphoma. 2019;60(6):1462-1468.
Marlise R. Luskin: dis­closes no rel­e­vant con­flict of inter­est. 19. Geyer MB, Hsu M, Devlin SM, Tallman MS, Douer D, Park JH. Overall sur­vival
among older US adults with ALL remains low despite mod­est improve­ment
Off-label drug use since 1980: SEER anal­y­sis. Blood. 2017;129(13):1878-1881.
20. Kantarjian HM, DeAngelo DJ, Stelljes M, et  al. Inotuzumab ozogamicin
Marlise R. Luskin: only clinical trials discussed.
ver­sus stan­dard ther­apy for acute lym­pho­blas­tic leu­ke­mia. N Engl J Med.
2016;375(8):740-753.
Correspondence 21. Kantarjian H, Stein A, Gökbuget N, et al. Blinatumomab ver­sus che­mo­ther­
Marlise R. Luskin, Dana-Farber Cancer Institute, Department of apy for advanced acute lym­pho­blas­tic leu­ke­mia. N Engl J Med. 2017;376(9):​
Medical Oncology, Division of Leukemia, 450 Brookline Ave, Dana 836-847.
22. Kantarjian H, Ravandi F, Short NJ, et al. Inotuzumab ozogamicin in com­bi­
2056, Bos­ton, MA 02215; e-mail: marlise_luskin@dfci​­.harvard​­.edu. na­tion with low-inten­sity che­mo­ther­apy for older patients with Philadel-
phia chro­mo­some-neg­a­tive acute lym­pho­blas­tic leu­kae­mia: a sin­gle-arm,
References phase 2 study. Lancet Oncol. 2018;19(2):240-248.
1. Farber S, Diamond LK. Temporary remis­sions in acute leu­ke­mia in chil­dren 23. Short NJ, Kantarjian HM, Ravandi F, et  al. Reduced-inten­sity che­mo­ther­
pro­duced by folic acid antag­o­nist, 4-aminopteroyl-glutamic acid. N Engl J apy with mini-hyper-CVD plus inotuzumab ozogamicin, with or with­out
Med. 1948;238(23):787-793. blinatumomab, in older adults with newly diag­nosed Philadelphia chro­mo­
2. Pui C-H, Yang J-J, Hunger S-P, et al. Childhood acute lym­pho­blas­tic leu­ke­ some-neg­a­tive acute lym­pho­blas­tic leu­ke­mia: results from a phase II study.
mia: prog­ress through col­lab­o­ra­tion. J Clin Oncol. 2015;33(27):2938-2948. Blood. 2020;136(suppl 1):1014.
3. Surveillance, Epidemiology, and End Results (SEER) Program. Cancer stat 24. Stelljes M, Raffel S, Wasch R, et al. First results of an open label phase II
facts: leu­ke­mia—acute lym­pho­cytic leu­ke­mia (ALL). 2020. Accessed 2 July study to eval­u­ate the effi­cacy and safety of inotuzumab ozogamicin for
2021. https:​­/​­/seer​­.cancer​­.gov​­/statfacts​­/html​­/alyl​­.html. induc­tion ther­apy followed by con­ven­tional che­mo­ther­apy based con­sol­
4. Larson RA, Dodge RK, Burns CP, et  al. A five-drug remis­sion induc­tion i­da­tion and main­te­nance ther­apy in patients aged 56 years and older with
reg­i­men with inten­sive con­sol­i­da­tion for adults with acute lym­pho­blas­tic acute lym­pho­blas­tic leu­ke­mia (INITIAL-1 trial). Blood. 2020;136(suppl 1):267.
leu­ke­mia: can­cer and leu­ke­mia group B study 8811. Blood. 1995;85(8):2025- 25. Jain N, Stevenson K, Winer ES, et al. A mul­ti­cen­ter phase I study com­bin­
2037. ing venetoclax with mini-hyper-CVD in older adults with untreated and
5. Larson RA, Dodge RK, Linker CA, et  al. A ran­dom­ized con­trolled trial of relapsed/refrac­tory acute lym­pho­blas­tic leu­ke­mia. Blood. 2019;134(suppl
filgrastim dur­ing remis­sion induc­tion and con­sol­i­da­tion che­mo­ther­apy 1):3867.
for adults with acute lym­pho­blas­tic leu­ke­mia: CALGB study 9111. Blood. 26. Advani AS, Moseley A, O’Dwyer KM, et al. Results of SWOG 1318: a phase
1998;92(5):1556-1564. 2 trial of blinatumomab followed by POMP (pred­ni­sone, vin­cris­tine, meth­
6. Kantarjian H, Thomas D, O’Brien S, et  al. Long-term fol­low-up results of o­trex­ate, 6-mer­cap­to­pu­rine) main­te­nance in elderly patients with newly
hyperfractionated cyclo­phos­pha­mide, vin­cris­tine, doxo­ru­bi­cin, and dexa­ diag­nosed Philadelphia chro­mo­some neg­a­tive B-cell acute lym­pho­blas­tic
meth­as­ one (hyper-CVAD), a dose-inten­sive reg­i­men, in adult acute lym­ leu­ke­mia. Blood. 2018;132(suppl 1):33.
pho­cytic leu­ke­mia. Cancer. 2004;101(12):2788-2801. 27. Chonghaile TN, Roderick JE, Glenfield C, et al. Maturation stage of T-cell
7. Rowe JM, Buck G, Burnett AK, et al; ECOG; MRC/NCRI Adult Leukemia acute lym­pho­blas­tic leu­ke­mia deter­mines BCL-2 ver­sus BCL-XL depen­
Working Party. Induction ther­apy for adults with acute lym­pho­blas­tic leu­ dence and sen­si­tiv­ity to ABT-199. Cancer Discov. 2014;4(9):1074-1087.
ke­mia: results of more than 1500 patients from the inter­na­tional ALL trial: 28. Lacayo NJ, Pullarkat VA, Stock W, et al. Safety and effi­cacy of venetoclax
MRC UKALL XII/ECOG E2993. Blood. 2005;106(12):3760-3767. in com­bi­na­tion with navitoclax in adult and pedi­at­ric relapsed/refrac­tory

14  |  Hematology 2021  |  ASH Education Program


acute lym­pho­blas­tic leu­ke­mia and lym­pho­blas­tic lym­phoma. Blood. 40. Martinelli G, Piciocchi A, Papayannidis C, et al. First report of the Gimema
2019;134(suppl 1):285. LAL1811 phase II pro­spec­tive study of the com­bi­na­tion of ste­roids with
29. Gökbuget N, Dombret H, Bonifacio M, et  al. Blinatumomab for min­i­mal ponatinib as front­ line ther­apy of elderly or unfit patients with Phila-
resid­ual dis­ease in adults with B-cell pre­cur­sor acute lym­pho­blas­tic leu­ke­ delphia chro­mo­some-pos­i­tive acute lym­pho­blas­tic leu­ke­mia. Blood.
mia. Blood. 2018;131(14):1522-1531. 2017;130(suppl 1):99.
30. Kirchhoff H, Karsli U, Schoenherr C, et al. Venetoclax and dexa­meth­a­sone 41. Wieduwilt M, Yin J, Wetzler M, et al. A phase II study of dasatinib and dexa­
synergize with inotuzumab ozogamicin-induced DNA dam­age sig­nal­ing in meth­a­sone as pri­mary ther­apy followed by trans­plan­ta­tion for adults with
B-lin­e­age ALL. Blood. 2021;137(19):2657-2661. newly diag­nosed Ph/BCR-ABL-pos­i­tive acute lym­pho­blas­tic leu­ke­mia
31. Chiaretti S, Vitale A, Cazzaniga G, et al. Clinico-bio­log­i­cal fea­tures of 5202 (Ph+ ALL): final results Alliance/CALGB study 10701. Blood. 2018;132(suppl
patients with acute lym­pho­blas­tic leu­ke­mia enrolled in the Ital­ian AIEOP 1):309.
and GIMEMA pro­ to­cols and strat­ i­
fied in age cohorts. Haematologica. 42. Luskin M, Murakami MA, Stevenson KE, et al. A phase I study of asciminib
2013;98(11):1702-1710. (ABL001) in com­bi­na­tion with dasatinib and pred­ni­sone for untreated BCR-
32. Fielding AK. Curing Ph+ ALL: assessing the rel­a­tive con­tri­bu­tions of che­ ABL1-pos­i­tive ALL in older adults. Blood. 2019;134(suppl 1):3879.
mo­ther­apy, TKIs, and allo­ge­neic stem cell trans­plant. Hematology Am Soc 43. Foà R, Bassan R, Vitale A, et al; GIMEMA Investigators. Dasatinib-blinatum-
Hematol Educ Program. 2019;2019(1):24-29. omab for Ph-pos­i­tive acute lym­pho­blas­tic leu­ke­mia in adults. N Engl J Med.
33. Ravandi F. How I treat Philadelphia chro­mo­some-pos­i­tive acute lym­pho­ 2020;383(17):1613-1623.
blas­tic leu­ke­mia. Blood. 2019;133(2):130-136. 44. Derman BA, Streck M, Wynne J, et  al. Efficacy and tox­ic­ity of reduced

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/7/1852080/7luskin.pdf by guest on 13 December 2021


34. Ribera J-M, García O, Fernández-Abellán P, et al; PETHEMA Group. Lack of vs. stan­dard dose pegylated asparaginase in adults with Philadelphia
neg­a­tive impact of Philadelphia chro­mo­some in older patients with acute chro­mo­some-neg­a­tive acute lym­pho­blas­tic leu­ke­mia. Leuk Lymphoma.
lym­pho­blas­tic leu­kae­mia in the thyrosine kinase inhib­i­tor era: com­par­i­son 2020;61(3):614-622.
of two pro­spec­tive par­al­lel pro­to­cols. Br J Haematol. 2012;159(4):485-488. 45. Martell MP, Atenafu EG, Minden MD, et  al. Treatment of elderly patients
35. Rousselot P, Coudé MM, Gokbuget N, et al; Euro­ pean Working Group with acute lym­pho­blas­tic leu­kae­mia using a pae­di­at­ric-based pro­to­col. Br
on Adult ALL (EWALL) group. Dasatinib and low-inten­sity che­mo­ther­apy J Haematol. 2013;163(4):458-464.
in elderly patients with Philadelphia chro­mo­some-pos­i­tive ALL. Blood. 46. Wolach O, Stevenson KE, Wadleigh M, et al. Allogeneic trans­plan­ta­tion is
2016;128(6):774-782. not supe­rior to che­mo­ther­apy in most patients over 40 years of age with
36. Ottmann OG, Pfeifer H, Cayuela JM. Nilotinib (Tasigna®) and low inten­sity Philadelphia-neg­a­tive acute lym­pho­blas­tic leu­ke­mia in first remis­sion. Am J
che­mo­ther­apy for first-line treat­ment of elderly patients with BCR-ABL1- Hematol. 2016;91(8):793-799.
pos­i­tive acute lym­pho­blas­tic leu­ke­mia: final results of a pro­spec­tive mul­ti­ 47. Hurria A, Cirrincione CT, Muss HB, et al. Implementing a geri­at­ric assess­
cen­ter trial (EWALL-PH02). Blood. 2018;132(suppl 1):31. ment in coop­er­a­tive group clin­i­cal can­cer tri­als: CALGB 360401. J Clin
37. Chalandon Y, Thomas X, Hayette S, et al; Group for Research on Adult Oncol. 2011;29(10):1290-1296.
Acute Lymphoblastic Leukemia (GRAALL). Randomized study of reduced- 48. Klepin HD, Ritchie E, Major-Elechi B, et  al. Geriatric assess­ment among
inten­sity che­mo­ther­apy com­bined with imatinib in adults with Ph-pos­i­tive older adults receiv­ing inten­sive ther­apy for acute mye­loid leu­ke­mia: report
acute lym­pho­blas­tic leu­ke­mia. Blood. 2015;125(24):3711-3719. of CALGB 361006 (Alliance). J Geriatr Oncol. 2020;11(1):107-113.
38. Vignetti M, Fazi P, Cimino G, et al. Imatinib plus ste­roids induces com­plete
remis­sions and prolonged sur­vival in elderly Philadelphia chro­mo­some-
pos­i­tive patients with acute lym­pho­blas­tic leu­ke­mia with­out addi­tional
che­mo­ ther­apy: results of the Gruppo Italiano Malattie Ematologiche
dell’Adulto (GIMEMA) LAL0201-B pro­to­col. Blood. 2007;109(9):3676-3678.
39. Foà R, Vitale A, Vignetti M, et al; GIMEMAAcute Leukemia Working Party. Dasat-
inib as first-line treat­ment for adult patients with Philadelphia chro­mo­some- © 2021 by The Amer­i­can Society of Hematology
pos­i­tive acute lym­pho­blas­tic leu­ke­mia. Blood. 2011;118(25):6521-6528. DOI 10.1182/hema­tol­ogy.2021000226

Prakash Singh Shekhawat


ALL cur­tain call chemo | 15
AML: SO MANY OPTIONS, SO LITTLE TIME

What to use to treat AML: the role


of emerging therapies

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/16/1851427/16thol.pdf by guest on 13 December 2021


Felicitas Thol
Department of Hematology, Hemostasis, Oncology, and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany

The development and approval of novel substances have resulted in substantial improvements in the treatment of acute
myeloid leukemia (AML). In the current era of novel treatment options, genetic and molecular testing at the time of diag-
nosis and relapse becomes increasingly relevant. Midostaurin in combination with intensive chemotherapy is the standard
of care as upfront therapy in younger AML patients with mutated fms-related tyrosine kinase 3 (FLT3). Gilteritinib, a second-
generation FLT3 inhibitor, represents a key drug for relapsed/refractory (R/R) FLT3-mutated AML patients. Targeted therapy
has also been developed for patients with mutated isocitrate dehydrogenase 1 (IDH1) and IDH2. The US Food and Drug Admin-
istration (FDA) approved ivosidenib as a monotherapy for newly diagnosed older adult IDH1-mutated patients and enasid-
enib for R/R IDH2-mutated AML patients. CPX-351, a liposomal formulation of daunorubicin and cytarabine, has become an
important upfront treatment strategy for fit patients with therapy-related AML or AML with myelodysplasia-related changes
that are generally challenging to treat. The antibody drug conjugate gemtuzumab ozogamicin was approved in combination
with intensive therapy for patients with newly diagnosed (FDA/European Medicines Agency [EMA]) as well as R/R CD33+
AML. The combination of venetoclax, an oral selective B-cell leukemia/lymphoma-2 inhibitor, with hypomethylating agents
or low-dose AraC (LDAC) has changed the treatment landscape and prognosis for older adult patients very favorably. The
addition of glasdegib, a small-molecule hedgehog inhibitor, to LDAC is another example of novel options in older patients.
Further substances have shown promising results in early clinical trials.

LEARNING OBJECTIVES
• Learn about the indications and efficacy of newly approved drugs in AML
• Become familiar with a treatment algorithm for newly diagnosed and relapsed AML patients

CLINICAL CASE with hypomethylating agents (HMAs) for older adult AML
A 66-year-old man with no significant underlying health con- patients (Figure 1). Novel mechanisms of action underlie
ditions developed leukocytosis, anemia, and thrombocyto- these recently approved drugs. Some of the new treat-
penia. Bone marrow biopsy revealed an infiltration of 60% ment strategies were designed for or have shown the most
myeloblasts. Immunophenotypically, the blasts were CD34+, benefit in a distinct genetic group of patients. Therefore,
CD13+, and CD33+. He was diagnosed with acute myeloid molecular and cytogenetic analysis becomes increasingly
leukemia (AML). Mutational screening showed an FLT3-ITD relevant for the application of novel drugs. In addition, ge-
(allelic ratio 0.6), isocitrate dehydrogenase 2 (IDH2), and netic risk stratification at the time of diagnosis is an essen-
TP53 mutation. The cytogenetic analysis revealed a normal tial element for guiding the decision regarding whether
karyotype. allogeneic hematopoietic stem cell transplantation (HSCT)
What are the treatment options for this patient in the is recommended in the first complete remission (CR).1 In
era of novel therapies? 2017 the European LeukemiaNet (ELN) recommended muta-
tional screening for FLT3-ITD (including allelic ratio), NPM1,
CEBPA (biallelic status), TP53, ASXL1, and RUNX1 in addi-
tion to cytogenetic analysis for the allocation to one of the
Intensive therapy with novel agents three prognostic categories.1 Based on the risk-benefit ratio
In recent years, treatment strategies for AML have evolved for allogeneic HSCT, patients in the adverse and possibly
beyond “7+3” for younger patients and past monotherapy in the intermediate group are candidates for allogeneic

16 | Hematology 2021 | ASH Education Program


Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/16/1851427/16thol.pdf by guest on 13 December 2021
Figure 1. Frontline treatment algorithm in AML patients considering recently approved substances. mut, mutated.

HSCT in first CR. In our patient three impor­tant muta­tions were trans­plan­ta­tion was not taken into account. After the com­ple­
iden­ti­fied (FLT3-ITD, IDH2, TP53). The detec­tion of FLT3-ITD in tion of con­sol­i­da­tion ther­apy, midostaurin or pla­cebo was given
this patient allows the appli­ ca­tion of front­ line-targeted ther­ for up to 12 cycles as main­te­nance ther­apy. A land­mark anal­y­sis
apy with midostaurin. Midostaurin is an oral first-gen­ er­a­
tion at the end of con­sol­i­da­tion ther­apy with high-dose cytarabine
tyro­sine kinase inhib­i­tor (TKI) and is now the stan­dard of care showed no sig­nif­i­cant dif­fer­ences in OS and EFS between both
in FLT3-mutated AML patients who can receive inten­sive ther­ treat­ment arms. Due to these incon­clu­sive results with respect
apy. Its approval by the US Food and Drug Administration (FDA) to the quan­ti­ta­tive effect of main­te­nance ther­apy, the effi­cacy
and the Euro­pean Medicines Agency (EMA) was based on the of the con­tin­ued treat­ment after con­sol­i­da­tion ther­apy is cur­
RATIFY trial, a large inter­na­tional phase 3 trial that ran­dom­ized rently unclear.3 The favor­able safety pro­file of midostaurin was
FLT3-mutated patients to treat­ment with midostaurin vs pla­cebo con­firmed in the RADIUS-X expanded access pro­gram.4
in com­bi­na­tion with inten­sive ther­apy.2 Both over­all sur­vival (OS) Another novel treat­ment option for newly diag­nosed AML
and event-free sur­vival (EFS) were sig­nif­i­cantly lon­ger in the mi- patients with CD33 expres­ sion on leu­ke­mic blasts is gemtu-
dostaurin com­pared to the pla­cebo arm with­out dif­fer­ences in zumab ozogamicin (GO) in com­bi­na­tion with 7 + 3.5 GO com­bines
severe adverse events. The 4-year OS was 51.4% in the midostau- a CD33-directed anti­body with the che­mo­ther­apy agent cali-
rin arm vs 44.3% in the pla­cebo arm. Of note, the sur­vival ben­e­fit cheamicin. CD33 is a suit­able tar­get in AML because it is highly
in the midostaurin arm was observed when looking at the whole expressed in most AML cells and much less on nor­mal hema­to­
cohort. In a sep­a­rate anal­y­sis of molec­u­lar sub­groups (FLT3-ITD poi­etic cells. While prior data have been conflicting, espe­cially
high or low, FLT3-TKD), the sur­vival advan­tage in the midostau- with regard to tox­ic­ity, the French ALFA-0701 trial was designed
rin arm did not reach sta­tis­ti­cal sig­nif­i­cance, which is likely due to look directly at the effect of GO on the sur­ vival of older
to the sub­groups being under­pow­ered.2 The sur­vival advan­tage adults under­go­ing inten­sive ther­apy.6 In this ran­dom­ized, open-
observed in the midostaurin arm for the whole cohort was due label phase 3 study, 280 older patients (aged 50-70 years) were
to a sig­nif­i­cantly lower cumu­la­tive inci­dence of relapse (CIR) if either treated with GO (n = 140) on day 1, 4, and 7 or with­out
Prakash Singh Shekhawat
AML ther­apy with newly approved sub­stances  | 17
GO (n = 140) dur­ing the 7 + 3 induc­tion ther­apy.6 Patients achiev­ as out­pa­tients sug­gests that the admin­is­tra­tion of CPX-351 with­
ing CR were treated with 2 cycles of con­sol­i­da­tion ther­apy with out planned admis­sion is safe and might lead to a decreased
or with­out GO according to their ini­tial ran­dom­i­za­tion. The EFS uti­li­za­tion of health care resources.15
at 2 years was 17.1% in the con­trol arm vs 40.8% in the GO arm. Now we return to our patient. Based on the data above,
Two-year OS was also sig­nif­i­cantly bet­ter in the GO (53.2%) vs he was treated with 7 + 3 and midostaurin. He achieved a CR
stan­dard arm (41.9%; P = 0.037).6 A meta-anal­y­sis by Hills et  al. after induc­tion ther­apy. Importantly, as he was allo­cated to the
looked at the effi­cacy of GO in 5 ran­dom­ized tri­als involv­ing adverse prog­nos­tic group (FLT3-ITD high with­out NPM1 muta­
3325 patients.7 Interestingly, the abso­lute sur­vival ben­e­fit was tion as well as the pres­ence of TP53 muta­tion), it was recom-
most appar­ent in the patient group with favor­able cyto­ge­net­ mended that he undergo allo­ge­neic HSCT in first CR. However,
ics (20.7%), followed by patients with inter­me­di­ate cyto­ge­net­ics for those patients in CR who can­not com­plete inten­sive ther­apy
(5.7%).7 GO had no ben­e­fit in patients with adverse cyto­ge­net­ after induc­tion ther­apy, main­te­nance ther­apy with CC-486 has
ics.7 In the AMLSG 09-09 trial, NPM1-mutated AML patients were become another novel option. CC-486 is an oral HMA that is not
ran­dom­ized to receive inten­sive che­mo­ther­apy with or with­out bioequivalent to inject­able azacitidine and has shown effi­cacy in
GO. In this trial the EFS was not sig­nif­i­cantly dif­fer­ent between patients who have devel­oped resis­tance to inject­able HMAs in

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/16/1851427/16thol.pdf by guest on 13 December 2021


treat­ment arms.8 However, the early death rate dur­ing induc­tions prior stud­ies.16,17 In the phase 3 QUAZAR AML-001 trial, 472 AML
was higher, while the CIR was lower in the GO arm. This is in line patients (aged 55 to 86 years) were ran­dom­ized to receive CC-
with the find­ing that the addi­tion of GO leads to a bet­ter reduc­ 486 (238 patients) or pla­cebo (234 patients).18 Median OS as well
tion in NPM1 mutant tran­script lev­els across all­treat­ment cycles.9 as EFS was sig­nif­i­cantly lon­ger in the CC-486 arm (24.7 months,
Subgroup anal­y­sis revealed that the addi­tion of GO showed a 10.2 months, respec­tively) com­pared to the pla­cebo arm (14.8
sig­nif­i­cant ben­e­fit for youn­ger, FLT3-ITD-neg­a­tive, and female months, 4.8 months). Hematological tox­ic­ity with neutropenia
patients.8 The FDA and the EMA approved GO in com­bi­na­tion was more com­mon in the CC-486 arm (41% in the CC-486 arm vs
with inten­sive che­mo­ther­apy for patients with newly diag­nosed 24% in the pla­cebo arm). Based on the QUAZAR AML-001 results,
CD33+ AML. Considering the data outlined above, GO might be the FDA and EMA granted approval for CC-486 as a main­te­nance
best suited for patients with favor­able cyto­ge­net­ics and with­out ther­apy for adult AML patients in CR/CR with incom­plete hema­
an increased risk for early mor­tal­ity due to under­ly­ing infec­tion. to­logic recov­ery (CRi) after inten­sive induc­tion ther­apy who are
For patients with newly diag­nosed sec­ond­ary AML (s-AML), unable to com­plete inten­sive cura­tive ther­apy (eg, allo­ge­neic
ther­apy-related AML (tAML), or AML with myelodysplasia-related HSCT). CC-486 should not be substituted for inject­able azacit-
changes (AML-MRC), treat­ment with CPX-351, a lipo­so­mal for­mu­ idine because there are dif­fer­ences in phar­ma­co­ki­netic prop­er­
la­tion of dau­no­ru­bi­cin and cytarabine in a fixed com­bi­na­tion, is ties between the oral and the inject­able ver­sion.
now avail­­able.10 In the ini­tial phase 2 trial com­par­ing CPX-351 with We still lack suf­fi­cient data about the effi­cacy of com­bin­ing
stan­dard inten­sive treat­ment in older AML patients, only the sub­ the above novel agents with each other.
group of patients with sAML and adverse cyto­ge­net­ics showed
a sig­nif­i­cantly higher response rate (57.6% in the CPX-351 arm Nonintensive ther­apy with novel agents
vs 31.6% in the stan­dard arm).11 Based on these results, a large Treatment has remained chal­ leng­ ing for older adult patients
phase 3 trial was ini­ti­ated in older (aged 60-75 years) patients who can­not receive inten­sive ther­apy.19 Hypomethylating agents
with high-risk AML or sAML.10 Here the median OS was 9.56 in the such as azacitidine and decitabine have become impor­tant treat­
CPX-351 arm and 5.95 months in the 7 + 3 arm. The over­all remis­ ment strat­eg ­ ies for older patients and have shown sur­vival ben­
sion rate was also sig­nif­i­cantly bet­ter with CPX-351 (47.7%) vs the e­fits when com­pared to low-dose AraC (LDAC).20 However, the
stan­dard arm (33.3%).10 Of note, 34% of patients in the CPX-351 median OS for patients treated with azacitidine in a phase 3 trial
arm under­went allo­ge­neic HSCT, com­pared to 25% in the 7 + 3 was 10.4 months and only 7.1 months in a large pop­u­la­tion-based
arm. In an explor­atory land­mark sur­vival anal­y­sis from the time study.21 In light of the fact that HMA monotherapy does not result
of trans­ plant, out­ comes were more favor­ able in the CPX-351 in long-term remis­sions, clin­i­cal tri­als com­bin­ing HMAs (and LDAC)
arm. This is in line with results from an Ital­ian com­pas­sion­ate-use with venetoclax have dem­ on­strated impres­ sive sur­
vival data.
pro­gram for CPX-351. Here, CIR was reduced in 71 older adult Venetoclax is an oral, highly selec­tive small-mol­e­cule B-cell leu­
AML patients when allo­ge­neic HSCT was performed in the first ke­mia/lym­phoma-2 inhib­it­or with poor effi­cacy as a sin­gle agent
CR, with a very favor­able over­all out­come fol­low­ing trans­plan­ in AML.22 However, in vitro and in vivo stud­ies have shown syn­er­
ta­tion.12 The encour­ag­ing results of the phase 3 trial resulted in gism between che­mo­ther­apy and venetoclax, supporting the use
FDA and EMA approval of CPX-351 for this dis­tinct sub­group of of venetoclax com­bi­na­tions in AML. One mech­a­nism of syn­er­gism
AML patients. In order to iden­tify these patients with AML with between azacitidine and venetoclax is through tran­ scrip­tional
MRC, cyto­ge­netic anal­y­sis is indis­pens­able.13 Real-life expe­ri­ence induc­tion of the proapoptotic BH3-only pro­tein NOXA by azaciti-
from a French ret­ro­spec­tive study looked at 103 sAML, tAML, or dine.23,24 The VIALE-C study, an inter­na­tional phase 3 ran­dom­ized
AML-MRC patients treated with CPX-351.14 The over­all response dou­ble-blind trial, com­pared venetoclax vs pla­cebo in com­bi­na­
rate (ORR) was 59% after induc­tion with an OS of 16.1 months tion with LDAC in older (≥75 years) or youn­ger patients unfit for
at a median fol­low-up time of 8.6 months. Importantly, patients inten­sive ther­apy.25 At a median fol­low-up of 17.5 months, patients
with ASXL1 and RUNX1 muta­tions, who are in the ELN unfa­vor­able treated with venetoclax and LDAC (VEN-LDAC) achieved a sig­nif­i­
prog­nos­tic group, showed sim­i­lar response rates as wild-type cantly lon­ger median OS com­pared to patients receiv­ing pla­cebo
patients in con­trast to patients with mutated TP53.14 The safety and LDAC (PBO-LDAC) (8.4 vs 4.1 months). Similarly, the CR rates
pro­file was very favor­able for CPX-351, with a low rate of alo­pe­cia as well as EFS were bet­ter in the VEN-LDAC arm com­pared to the
(11%) and gas­tro­in­tes­ti­nal tox­ic­ity (50%). Interestingly, a ret­ro­ PBO-LDAC arm (EFS, 4.9 months vs 2.1 months). The com­bi­na­
spec­tive study involv­ing 25 AML patients who received CPX-351 tion of azacitidine and venetoclax (VEN-AZA) yielded even more

18  |  Hematology 2021  |  ASH Education Program


encour­ag­ing results (VIALE-A trial).26 Here, older, pre­vi­ously un- ing that this molec­u­lar sub­group, espe­cially, ben­e­fits from adding
treated AML patients who were inel­i­gi­ble for stan­dard induc­tion venetoclax to azacitidine treat­ment.28 The FDA has approved vene-
received VEN-AZA or pla­cebo and azacitidine (PBO-AZA).26 The toclax in com­bi­na­tion with HMAs or LDAC for newly diag­nosed
median OS was 14.7 months in the VEN-AZA arm vs 9.6 months in AML patients ≥75 years of age or patients with comorbidities pre­
the PBO-AZA arm at a median fol­low-up of 20.5 months. Thus, the clud­ing inten­sive induc­tion ther­apy (the EMA has approved vene-
median OS in the VEN-AZA arm was lon­ger than reported in any toclax only in combination with HMAs). As HMA/VEN has evolved
other prior trial for front­line, older adult AML patients. In addi­tion, as a stan­dard of care (in countries with approval), this com­bi­na­
the CR rate, at 36.7% vs 17.9%, was also more favor­able with the tion also rep­re­sents the back­bone of tri­als with novel sub­stances.
venetoclax com­bi­na­tion. Grade ≥3 throm­bo­cy­to­pe­nia and neu- Unfortunately, a ret­ ro­
spec­ tive anal­
y­sis sug­gests that once pa-
tropenia were more com­mon in patients treated with venetoclax. tients become unre­spon­sive to HMA/VEN the prog­no­sis is very
Likewise, febrile neutropenia occurred in 42% of patients in the poor.29 This might be related to the acqui­si­tion of high-risk cyto­
VEN-AZA arm vs 19% of patients in the PBO-AZA arm. Thus, it is ge­netic and molec­u­lar fea­tures (eg, muta­tions in TP53, N/KRAS,
essen­tial to observe the patient closely for hema­to­log­i­cal tox­ic­ity and/or KIT).29 For patients who are older but lack comorbidities,
in the form of cytopenias and fol­low rec­om­men­da­tions for dose the ques­tion arises as to whether inten­sive ther­apy or HMA/VEN

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/16/1851427/16thol.pdf by guest on 13 December 2021


adjust­ments when treating patients with venetoclax com­bi­na­ treat­ments are the wiser treat­ment choice. To date, there is no
tions.27 Dose adjust­ments also become nec­es­sary when comed- ran­dom­ized trial com­par­ing inten­sive che­mo­ther­apy with HMA-
ications have strong CYP3A4 inhib­i­tory activ­ity (eg, anti­fun­gals, VEN head-to-head. However, a ret­ ro­spec­
tive study com­ pared
fluoroquinolones).27 Recent data sug­gest that IDH1- and IDH2-mu- out­comes of patients with inten­sive che­mo­ther­apy vs decit-
tated AML patients achieve impres­sive response rates, suggest- abine over 10 days in com­bi­na­tion with venetoclax (DEC10-VEN).

Figure 2. Treatment of relapsed/refractory AML patients considering recently novel substances. dDLI, donor lymphocyte infusion;
BSC, best supportive care; HU, hydroxyurea; mut, mutated.
Prakash Singh Shekhawat
AML ther­apy with newly approved sub­stances  | 19
Table 1. Recently approved drugs for AML

Significant find­ings in clin­i­cal Issues need­ing spe­cial


Drug Indication Route of admin­is­tra­tion
tri­als atten­tion
Midostaurin Newly diag­nosed FLT3 Oral Phase 3 trial (RATIFY): Careful admin­is­tra­tion of
Rydapt® mut AML in com­bi­ inten­sive che­mo­ther­apy + comedications with strong
na­tion with inten­sive midostaurin vs pla­cebo: CYP3A4 inhib­i­tory activ­ity
ther­apy (FDA, EMA) 4-year OS: 51.4% midostaurin
vs 44.3% pla­cebo
CPX-351 Newly diag­nosed tAML, IV Phase 3 trial: Side effects sim­il­ar to stan­
Vyxeos® AML-MRC (FDA, EMA) median OS 9.6 (CPX-351) vs 6.0 dard che­mo­ther­apy but less
months (stan­dard che­mo­ alo­pe­cia
ther­apy)
Gemtuzumab Newly diag­nosed CD33+ IV Phase 3 trial (ALFA-0701): Hepatotoxicity (includ­ing
ozogamicin AML (FDA, EMA) in inten­sive che­mo­ther­apy +/− veno-occlu­sive dis­ease),

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/16/1851427/16thol.pdf by guest on 13 December 2021


Mylotarg® com­bi­na­tion with GO: infu­sion-related reac­tions,
inten­sive ther­apy, R/R 2-year OS: 53.2% GO vs 41.9% infec­tion
CD33+ AML in com­bi­ stan­dard
na­tion with inten­sive
ther­apy (FDA)
Venetoclax Newly diag­nosed AML Oral VIALE-A trial: Hematological tox­ic­ity (fol­
Venclexta® in patients ≥75 years VEN-AZA vs pla­cebo: low recommended dose
or with comorbidities median OS 14.7 (venetoclax) adjust­ments), neutropenic
pre­clud­ing inten­sive vs 9.6 months (pla­cebo) fever, dose adjust­ments if
ther­apy; in com­bi­ VIALE-C trial: comedications with strong
na­tion with LDAC or LDAC-VEN vs pla­cebo: CYP3A4 inhib­i­tory activ­ity,
HMA (FDA) median OS 8.4 (venetoclax) tumor lysis syn­drome (but
vs 4.1 months (pla­cebo) much rarer than in CLL)
Glasdegib Newly diag­nosed AML Oral Phase 2 BRIGHT AML 1003 trial: Musculoskeletal pain
Daurismo® in patients ≥75 years LDAC +/− glasdegib:
or with comorbidities median OS was 8.8 (LDAC-
pre­clud­ing inten­sive glasdegib) vs 4.9 months
ther­apy; in com­bi­na­ (LDAC alone)
tion with LDAC (FDA,
EMA)
CC-486 Maintenance ther­apy Oral Phase 3 QUAZAR AML-001: GI tox­ic­ity, neutropenia
Onureg® AML patients in CC-486 vs pla­cebo:
CR/CRi after inten­sive median OS 24.7 (CC-486) vs
induc­tion who are 14.8 months (pla­cebo)
unable to com­plete
inten­sive cura­tive
ther­apy
Enasidenib R/R IDH2 mut AML as Oral Phase 1/2 trial: Check IDH2 muta­tional sta­tus
Idhifa® monotherapy (FDA) ORin 40.3% of patients, OS at time of R/R dis­ease,
9.3 months IDH dif­fer­en­ti­a­tion syn­drome,
indi­rect hyperbilirubinemia
Ivosidenib Newly diag­nosed or Oral Phase 1b: Check IDH1 muta­tional sta­tus
Tibsovo® R/R IDH1 mut AML as monotherapy CR in 22% and at time of diag­no­sis and
monotherapy(FDA) OR in 42% of patients R/R dis­ease,
IDH dif­fer­en­ti­a­tion syn­drome,
indi­rect hyperbilirubinemia
Gilteritinib R/R FLT3 mut AML as Oral Phase 3 (ADMIRAL) trial: Check FLT3 muta­tional sta­tus
Xospata® monotherapy (FDA, gilteritinib vs sal­vage che­mo­ at time of R/R dis­ease
EMA) ther­apy:
CR/CRi 34% (gilteritinib) vs
15.4% (sal­vage ther­apy);
OS 9.3 (gilteritinib) vs 5.6
months (sal­vage ther­apy)
CLL, chronic lym­pho­cytic leu­ke­mia; CRi, CR with incom­plete hema­to­logic recov­ery; IV, intra­ve­nous; mut, mutated; OR, over­all response.

20  |  Hematology 2021  |  ASH Education Program


The CR/CRi rates and relapse rates were more favor­able in the For IDH2-mutated AML patients, the FDA (not the EMA)
DEC10-VEN arm. In addi­tion, OS was sig­nif­i­cantly lon­ger in DEC10- approved enasidenib, an oral inhib­i­tor of mutated IDH2, as a
VEN-treated patients.30 In sum­mary, the intro­duc­tion of venetoclax monotherapy for R/R AML patients.39,40 The approval was based
has mark­edly changed the sur­vival out­look for older AML patients. on a phase 1/2 trial.29 Here, enasidenib monotherapy achieved
Glasdegib, a small-mol­e­cule hedge­hog inhib­i­tor, is another an ORR of 40.3%, with a median dura­tion of response of 5.8
drug that has received approval by the FDA and the EMA for months and a median OS of 9.3 months in R/R IDH2-mutated
older, newly diag­nosed AML patients (≥75 years) or patients with AML patients.40
comorbidities that pre­clude inten­sive ther­apy. The approval was GO also received approval by the FDA (not the EMA) for R/R
based on a phase 2, ran­dom­ized, open-label, mul­ti­cen­ter study patients with CD33+ AML and can be added to inten­sive ther­apy.
that com­pared LDAC vs LDAC/glasdegib. Median OS was 8.8 So if our patient relapses after HSCT with the same molec­u­lar
months with the com­bi­na­tion com­pared to 4.9 months with LDAC pro­file (FLT3-ITD pos, IDH2 mut), targeted ther­apy is avail­­able
alone.31 Of note, a head-to-head com­par­i­son between VEN/LDAC with gilteritinib or enasidinib (Figure 2).
or VEN/HMA and LDAC/glasdegib is cur­rently miss­ing.
For one molec­u­lar sub­group of patients, targeted ther­apy is Further direc­tions

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/16/1851427/16thol.pdf by guest on 13 December 2021


now avail­­able: older AML patients (≥75 years) or AML patients Various agents cur­rently undergo eval­u­a­tion in early clin­i­cal AML
with sig­ nif­i­
cant comorbidities who carry a muta­ tion in IDH1. tri­
als. Magrolimab is a mono­ clo­ nal anti-CD47 anti­body. CD47
Ivosidenib is an oral, targeted agent that inhib­its mutant IDH1 is known to be a mac­ro­phage immune check­point that func­
and con­se­quently blocks the pro­duc­tion of the oncometabolite tions as a “don’t eat me” sig­nal to can­cer so that magrolimab
2-hydroxyglutarate. Ivosidenib has been stud­ied as monother- can pro­mote phago­cy­to­sis of leu­ke­mic cells. Magrolimab was
apy in IDH1-mutated AML patients in a phase 1b trial.32 Of the 258 com­bined with azacitidine in a phase 1b trial enroll­ing 52 AML
patients in the trial, the major­ity had R/R AML (179 patients). In patients.41 In this trial, 65% of patients achieved an objec­tive
the pri­mary effi­cacy cohort, 21.6% of patients achieved CR and response, with 44% of patients achiev­ing a CR. Magrolimab re-
41.6% an over­all response, with a median dura­tion of response ceived orphan drug des­ig­na­tion by the FDA for myelodysplastic
of 9.3 and 6.5 months, respec­tively. The IDH dif­fer­en­ti­a­tion syn­ syndrome (MDS) and AML and by the EMA for AML in 2020. APR-
drome is a dis­tinct side effect of IDH inhib­i­tors and requires a 246 is a prom­is­ing agent devel­oped for patients with mutated
phy­si­cian’s aware­ness. It was observed in 3.9% of patients in this TP53, as it restores its func­tion as a tumor sup­pres­sor gene.42
trial. The FDA (but not the EMA) approved ivosidenib for older Early tri­als have com­bined APR-246 with azacitidine in MDS and
(≥75 years) patients or patients with sig­nif­i­cant comorbidities AML. In a phase 1/2 trial, the ORR was 64% with a 36% CR rate
as a monotherapy for newly diag­ nosed, IDH1-mutant as well in TP53-mutated AML patients.43 These encour­ag­ing results have
as R/R AML. In a phase 1b trial, the com­bi­na­tion of azacitidine been con­firmed in a sec­ond study show­ing a CR rate of 56%
with ivosidenib was well tol­er­ated, and patients showed dura­ble at 6 cycles. APR-246 has received orphan drug and fast-track
remis­sions.33 Thus, we eagerly await the results of the pla­cebo- des­ig­na­tions from the FDA for MDS and orphan drug des­ig­na­
con­trolled phase 3 trial with this com­bi­na­tion. tion from the EMA for AML and MDS. Menin is a prom­is­ing tar­
get for AML patients with mixed-lineage leukemia trans­lo­ca­tions,
Treatment of AML in relapse and menin inhib­i­tors are cur­rently being stud­ied in clin­i­cal tri­
Due to clonal evo­lu­tion, it is advis­able to repeat muta­tional pro­ als.44 Many more sub­stances are in early devel­op­ment for AML
fil­ing at the time of relapse.34 While some muta­tions are sta­ble patients. Importantly, clin­i­cal tri­als study­ing novel agents with
dur­ing dis­ease pro­gres­sion, oth­ers are lost or gained at the time new com­bi­na­tions are also ongo­ing (eg, IDH inhib­i­tors with che­
of relapse.35 Interestingly, the fol­low-up data of the RATIFY trial mo­ther­apy, FLT3 inhib­i­tors with HMAs). Finally, yet impor­tantly,
showed that 46% of patients became FLT3-ITD-neg­a­tive at the advance­ment in mea­sur­able resid­ual dis­ease mon­i­tor­ing is also
time of dis­ease resis­tance or pro­gres­sion.36 For patients who show likely to influ­ence our treat­ment deci­sions and have a pos­i­tive
an FLT3 muta­tion in relapse, gilteritinib, an oral, sec­ond-gen­er­a­ impact on out­come.45 In sum­mary, the treat­ment land­scape has
tion TKI, is now avail­­able. Gilteritinib was com­pared to sal­vage evolved remark­ably over the last 5 years with the approval of a
ther­apy in a large ran­dom­ized phase 3 trial (ADMIRAL).37,38 Here, num­ber of new drugs (Table 1). This has direct impli­ca­tions for
patients were ran­dom­ized to receive gilteritinib vs sal­vage che­ our 66-year-old patient.
mo­ther­apy according to local inves­ti­ga­tors’ choice. Gilteritinib
was asso­ci­ated with higher CR/CRi rates (34% vs 15.4%). This pos­ Conflict-of-inter­est dis­clo­sure
i­tive effect trans­lated into a lon­ger OS in the gilteritinib arm (9.3 Felicitas Thol: Advisory Board: Celgene, Novartis, Jazz, Abbvie,
vs 5.6 months).38 Importantly, severe adverse events were less fre­ Astellas, Pfizer.
quent in the gilteritinib arm. While only 5.7% of patients in the
ADMIRAL trial received midostaurin dur­ing front­line treat­ment, a Off-label drug use
ret­ro­spec­tive study of 13 med­i­cal cen­ters ana­lyzed the effi­cacy Felicitas Thol: nothing to disclose.
of gilteritinib in R/R AML patients pre­vi­ously treated with a TKI.
In this patient pop­u­la­tion, the CR rates were 58% with an OS of Correspondence
7.8 months, suggesting that gilteritinib is no less effec­tive after Felicitas Thol, Department of Hematology, Hemostasis, Oncol-
pre­vi­ous TKI treat­ment. This is rel­e­vant, as in the RATIFY trial 11% ogy, and Stem Cell Transplantation, Hannover Medical School,
of patients devel­oped ITD clones resis­tant to midostaurin dur­ing Carl-Neuberg Str 1, 30625 Hannover, Germany; e-mail: thol​
dis­ease pro­gres­sion.36 ­.felicitas@mh-hannover​­.de.

Prakash Singh Shekhawat


AML ther­apy with newly approved sub­stances  | 21
References 21. Zeidan AM, Wang R, Wang X, et al. Clinical out­comes of older patients with
1. Döhner H, Estey E, Grimwade D, et al. Diagnosis and man­age­ment of AML AML receiv­ing hypomethylating agents: a large pop­u­la­tion-based study in
in adults: 2017 ELN rec­om­men­da­tions from an inter­na­tional expert panel. the United States. Blood Adv. 2020;4(10):2192-2201.
Blood. 2017;129(4):424-447. 22. Konopleva M, Pollyea DA, Potluri J, et  al. Efficacy and bio­log­i­cal cor­
2. Stone RM, Mandrekar SJ, Sanford BL, et  al. Midostaurin plus che­mo­ther­ re­lates of response in a phase II study of venetoclax monotherapy in
apy for acute mye­ loid leu­ ke­mia with a FLT3 muta­ tion. N Engl J Med. patients with acute mye­log­e­nous leu­ke­mia. Cancer Discov. 2016;6(10):
2017;377(5):454-464. 1106-1117.
3. Larson RA, Mandrekar SJ, Huebner LJ, et al. Midostaurin reduces relapse in 23. Jin S, Cojocari D, Purkal JJ, et al. 5-azacitidine induces NOXA to prime AML
FLT3-mutant acute mye­loid leu­ke­mia: the Alliance CALGB 10603/RATIFY trial cells for venetoclax-medi­ated apo­pto­sis. Clin Cancer Res. 2020;26(13):3371-
[published online 2 March 2021]. Leukemia. 3383.
4. Roboz GJ, Strickland SA, Litzow MR, et al. Updated safety of midostaurin 24. Cojocari D, Smith BN, Purkal JJ, et al. Pevonedistat and azacitidine upreg-
plus che­mo­ther­apy in newly diag­nosed FLT3 muta­tion-pos­i­tive acute mye­ ulate NOXA (PMAIP1) to increase sen­si­tiv­ity to venetoclax in pre­clin­i­cal
loid leu­ke­mia: the RADIUS-X expanded access pro­gram. Leuk Lymphoma. mod­els of acute mye­loid leu­ke­mia [published online 15 April 2021]. Hae-
2020;61(13):3146-3153. matologica.
5. Thol F, Schlenk RF. Gemtuzumab ozogamicin in acute mye­loid leu­ke­mia 25. Wei AH, Strickland SA Jr, Hou JZ, et al. Venetoclax com­bined with low-dose
revisited. Expert Opin Biol Ther. 2014;14(8):1185-1195. cytarabine for pre­vi­ously untreated patients with acute mye­loid leu­ke­mia:
6. Castaigne S, Pautas C, Terré C, et al; Acute Leukemia French Association. results from a phase Ib/II study. J Clin Oncol. 2019;37(15):1277-1284.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/16/1851427/16thol.pdf by guest on 13 December 2021


Effect of gemtuzumab ozogamicin on sur­vival of adult patients with de- 26. DiNardo CD, Jonas BA, Pullarkat V, et al. Azacitidine and venetoclax in pre­
novo acute mye­ loid leu­kae­ mia (ALFA-0701): a randomised, open-label, vi­ously untreated acute mye­loid leu­ke­mia. N Engl J Med. 2020;383(7):617-
phase 3 study. Lancet. 2012;379(9825):1508-1516. 629.
7. Hills RK, Castaigne S, Appelbaum FR, et al. Addition of gemtuzumab ozo- 27. DiNardo CD, Wei AH. How I treat acute mye­loid leu­ke­mia in the era of new
gamicin to induc­tion che­mo­ther­apy in adult patients with acute mye­loid drugs. Blood. 2020;135(2):85-96.
leu­kae­mia: a meta-anal­y­sis of indi­vid­ual patient data from randomised con­ 28. Pollyea DA, Dinardo CD, Arellano ML, et al. Results of venetoclax and azac-
trolled tri­als. Lancet Oncol. 2014;15(9):986-996. itidine com­bi­na­tion in che­mo­ther­apy inel­i­gi­ble untreated patients with
8. Schlenk RF, Paschka P, Krzykalla J, et  al. Gemtuzumab ozogamicin in acute mye­loid leu­ke­mia with IDH 1/2 muta­tions. Paper presented at: ASH
NPM1-mutated acute mye­loid leu­ke­mia: early results from the pro­spec­tive Annual Meeting; (Virtual); 5-8 December 2020.
ran­dom­ized AMLSG 09-09 phase III study. J Clin Oncol. 2020;38(6):623- 29. Maiti A, Rausch CR, Cortes JE, et  al. Outcomes of relapsed or refrac­tory
632. acute mye­loid leu­ke­mia after front­line hypomethylating agent and veneto-
9. Kapp-Schwoerer S, Weber D, Corbacioglu A, et al. Impact of gemtuzumab clax reg­i­mens. Haematologica. 2021;106(3):894-898.
ozogamicin on MRD and relapse risk in patients with NPM1-mutated AML: 30. Maiti A, Qiao W, Sasaki K, et  al. Venetoclax with decitabine vs inten­sive
results from the AMLSG 09-09 trial. Blood. 2020;136(26):3041-3050. che­mo­ther­apy in acute mye­loid leu­ke­mia: a pro­pen­sity score matched
10. Lancet JE, Uy GL, Cortes JE, et al. CPX-351 (cytarabine and dau­no­ru­bi­cin) anal­y­sis strat­i­fied by risk of treat­ment-related mor­tal­ity. Am J Hematol.
lipo­some for injec­tion ver­sus con­ven­tional cytarabine plus dau­no­ru­bi­cin in 2021;96(3):282-291.
older patients with newly diag­nosed sec­ond­ary acute mye­loid leu­ke­mia. J 31. Cortes JE, Heidel FH, Hellmann A, et  al. Randomized com­par­i­son of low
Clin Oncol. 2018;36(26):2684-2692. dose cytarabine with or with­out glasdegib in patients with newly diag­
11. Lancet JE, Cortes JE, Hogge DE, et al. Phase 2 trial of CPX-351, a fixed 5:1 nosed acute mye­ loid leu­ke­
mia or high-risk myelodysplastic syn­ drome.
molar ratio of cytarabine/dau­no­ru­bi­cin, vs cytarabine/dau­no­ru­bi­cin in Leukemia. 2019;33(2):379-389.
older adults with untreated AML. Blood. 2014;123(21):3239-3246. 32. DiNardo CD, Stein EM, de Botton S, et  al. Durable remis­sions with ivo-
12. Guolo F, Fianchi L, Minetto P, et al. CPX-351 treat­ment in sec­ond­ary acute sidenib in IDH1-mutated relapsed or refrac­ tory AML. N Engl J Med.
mye­lo­blas­tic leu­ke­mia is effec­tive and improves the fea­si­bil­ity of allo­ge­neic 2018;378(25):2386-2398.
stem cell trans­plan­ta­tion: results of the Ital­ian com­pas­sion­ate use pro­gram. 33. DiNardo CD, Stein AS, Stein EM, et al. Mutant isocitrate dehy­dro­ge­nase 1 inhib­
Blood Cancer J. 2020;10(10):96. i­tor ivosidenib in com­bi­na­tion with azacitidine for newly diag­nosed acute
13. Arber DA, Orazi A, Hasserjian R, et al. The 2016 revi­sion to the World Health mye­loid leu­ke­mia. J Clin Oncol. 2021;39(1):57-65.
Organization clas­si­fi­ca­tion of mye­loid neo­plasms and acute leu­ke­mia. 34. Thol F, Schlenk RF, Heuser M, Ganser A. How I treat refrac­tory and early
Blood. 2016;127(20):2391-2405. relapsed acute mye­loid leu­ke­mia. Blood. 2015;126(3):319-327.
14. Chiche E, Rahmé R, Bertoli S, et al. Real-life expe­ri­ence with CPX-351 and 35. Cocciardi S, Dolnik A, Kapp-Schwoerer S, et  al. Clonal evo­lu­tion pat­
impact on the out­come of high-risk AML patients: a multicentric French terns in acute mye­ loid leu­
ke­
mia with NPM1 muta­ tion. Nat Commun.
cohort. Blood Adv. 2021;5(1):176-184. 2019;10(1):2031.
15. Keiffer G, Hughes K, Zhan T, Wilde L, Palmisiano N, Kasner M. Outpatient 36. Schmalbrock LK, Dolnik A, Cocciardi S, et al. Clonal evo­lu­tion of acute mye­
Vyxeos induc­ tion with­ out planned admis­ sion for select patients with loid leu­ke­mia with FLT3-ITD muta­tion under treat­ment with midostaurin.
sec­ond­ary acute mye­loid leu­ke­mia (sAML) is safe and yields healthcare Blood. 2021;137(22):3093-3104.
resource sav­ings. Paper presented at: ASH Annual Meeting; (Virtual); 5-8 37. Perl A, Martinelli G, Cortes JE, et al. Gilteritinib sig­nif­i­cantly pro­longs over­
December 2020. all sur­vival in patients with FLT3-mutated (FLT3mut+) relapsed/refrac­tory
16. Garcia-Manero G, Gore SD, Kambhampati S, et  al. Efficacy and safety of (R/R) acute mye­loid leu­ke­mia (AML): results from the Phase III ADMIRAL
extended dos­ing sched­ules of CC-486 (oral azacitidine) in patients with trial. Abstract presented at: AACR Annual Meeting; 29 March-April 3 2019.
lower-risk myelodysplastic syn­dromes. Leukemia. 2016;30(4):889-896. Abstract CT184.
17. Garcia-Manero G, Almeida A, Giagounidis A, et al. Design and ratio­nale of 38. Perl AE, Martinelli G, Cortes JE, et  al. Gilteritinib or che­ mo­ther­
apy for
the QUAZAR Lower-Risk MDS (AZA-MDS-003) trial: a ran­dom­ized phase relapsed or refrac­tory FLT3-mutated AML. N Engl J Med. 2019;381(18):1728-
3 study of CC-486 (oral azacitidine) plus best sup­port­ive care vs pla­cebo 1740.
plus best sup­port­ive care in patients with IPSS lower-risk myelodysplas- 39. Stein EM, DiNardo C, Altman JK, et  al. Safety and effi­cacy of AG-221, a
tic syn­dromes and poor prog­no­sis due to red blood cell trans­fu­sion- potent inhib­i­tor of mutant IDH2 that pro­motes dif­fer­en­ti­a­tion of mye­loid
depen­dent ane­mia and throm­bo­cy­to­pe­nia. BMC Hematol. 2016;16:12. cells in patients with advanced hema­to­logic malig­nan­cies: results of a
18. Wei AH, Döhner H, Pocock C, et al; QUAZAR AML-001 Trial Investigators. phase 1/2 trial. Abstract presented at: ASH 57th Annual Meeting and Expo-
Oral azacitidine main­te­nance ther­apy for acute mye­loid leu­ke­mia in first sition; 5-8 Decem­ber 2015; Orlando, FL.
remis­sion. N Engl J Med. 2020;383(26):2526-2537. 40. Stein EM, DiNardo CD, Pollyea DA, et al. Enasidenib in mutant IDH2 relapsed
19. Arellano M, Carlisle JW. How I treat older patients with acute mye­loid leu­ or refrac­tory acute mye­loid leu­ke­mia. Blood. 2017;130(6):722-731.
ke­mia. Cancer. 2018;124(12):2472-2483. 41. Sallman DA, Asch AS, Al Malki MM, et al. The first-in-class anti-CD47 anti­
20. Dombret H, Seymour JF, Butrym A, et  al. International phase 3 study of body magrolimab (5F9) in com­bi­na­tion with azacitidine is effec­tive in
azacitidine vs con­ven­tional care reg­i­mens in older patients with newly MDS and AML patients: ongo­ing phase 1b results. Blood. 2019;134(suppl 1):
diag­nosed AML with >30% blasts. Blood. 2015;126(3):291-299. 569.

22  |  Hematology 2021  |  ASH Education Program


42. Zhang Q , Bykov VJN, Wiman KG, Zawacka-Pankau J. APR-246 reacti- 45. Schuurhuis GJ, Heuser M, Freeman S, et al. Minimal/mea­sur­able resid­ual dis­
vates mutant p53 by targeting cys­ te­
ines 124 and 277. Cell Death Dis. ease in AML: a con­sen­sus doc­u­ment from the Euro­pean LeukemiaNet MRD
2018;9(5):439. Working Party. Blood. 2018;131(12):1275-1291.
43. Sallman DA, DeZern AE, Garcia-Manero G, et al. Eprenetapopt (APR-246)
and azacitidine in TP53-mutant myelodysplastic syn­dromes. J Clin Oncol.
2021;39(14):1584-1594.
44. Krivtsov AV, Evans K, Gadrey JY, et al. A menin-MLL inhib­i­tor induces spe­cific
chro­ma­tin changes and erad­i­cates dis­ease in mod­els of MLL-rearranged leu­ © 2021 by The Amer­i­can Society of Hematology
ke­mia. Cancer Cell. 2019;36(6):660-673.e11673e11. DOI 10.1182/hema­tol­ogy.2021000309

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/16/1851427/16thol.pdf by guest on 13 December 2021

Prakash Singh Shekhawat


AML ther­apy with newly approved sub­stances  | 23
AML: SO MANY OPTIONS, SO LITTLE TIME

Whom should we treat with novel agents?


Specific indications for specific and challenging
populations

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/24/1851553/24wilde.pdf by guest on 13 December 2021


Lindsay Wilde and Margaret Kasner
Medical Oncology, Thomas Jefferson University, Philadelphia, PA

A relative wealth of new therapies for acute myeloid leukemia (AML) have led to a rapid shift in treatment paradigms for
this disease. Understanding whom, when, and how to treat is more complex than ever before. Here we explore whom
to treat with these available new therapies, focusing on special patient populations that include older adults, those with
relapsed disease, and those with TP53-mutated AML. These high-risk subgroups are some of the most challenging to care
for, but novel treatments are providing them with new hope.

LEARNING OBJECTIVES
• Learn the indications for newly approved and emerging drugs in AML in challenging populations
• Become familiar with a treatment algorithm for older, relapsed, and TP53-mutated AML patients

Introduction becomes more difficult to treat and cure. Five-year overall


The recent approval of a variety of novel therapies for survival (OS) for patients diagnosed between the ages of
acute myeloid leukemia (AML) has provided more treat- 60 and 64 is 22%, while that of patients diagnosed over the
ment options for patients than ever before. However, age of 80 is 1.3%.2 The advent of newer, less toxic treat-
identifying whom to treat, and how, can be challenging. ments may help to improve this disparity.
Key elements in making these decisions include assessing Outcomes for older adults with AML are poor because of
patient characteristics and obtaining comprehensive AML many complex reasons (Figure 1) First, patient factors such
profiling, including cytogenetic and molecular data. Even as comorbidities and decreased baseline performance
with this information, the optimal therapeutic path is not status (PS) may prevent patients from receiving optimal
always clear. Here we highlight the complexities of select- therapy or put them at higher risk of complications. The
ing whom to treat with novel AML therapies. number of comorbidities increases with age and corre-
lates with an increase in AML-related and all-cause mortal-
ity.3 Older patients are also more likely to present with an
ECOG PS of 2 or 3, and decreased PS is associated with an
CLINICAL CASE increase in early death.4 Older patients with decreased PS
An 86-year-old man with a past medical history of hyper- are more likely to experience early mortality than younger
tension and gastroesophageal reflux disease developed patients with a similar PS, indicating the complex interplay
leukocytosis, anemia, and thrombocytopenia. A bone mar- between these 2 factors.5,6
row biopsy revealed 60% myeloblasts. Immunophenotyp- Second, disease-related factors also contribute to
ically, the blasts were CD34+, CD13+, and CD33+. He was poorer outcomes in older patients. Such patients are more
diagnosed with AML. Next-generation sequencing showed likely to have disease with high-risk chromosomal abnor-
mutations in ASXL1 and IDH1. Findings from the karyotype malities (−5 or 5q−, −7 or 7q−, or 17p deletion), and the
were normal. incidence of having high-risk mutations in genes such as
With a median age of 68 years at the time of diagno- ASXL1, RUNX1, and TP53 also increases with age.7 By Euro-
sis, AML is primarily a disease of older adults.1 However, pean LeukemiaNet criteria, older patients more often
the incidence of AML not only increases with age but also have high-risk disease, and survival for these patients is

24 | Hematology 2020 | ASH Education Program


Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/24/1851553/24wilde.pdf by guest on 13 December 2021
Figure 1.  Factors leading to a poor prognosis in older AML patients.

decreased com­pared to youn­ger patients with sim­i­larly high-risk toclax (AZA-VEN). This lower-inten­sity treat­ment was shown in
dis­ease.8,9 Older patients also more fre­quently have dis­ease that the phase 3, mul­ti­cen­ter, ran­dom­ized, dou­ble-blind, pla­cebo-
is inher­ently che­mo­ther­apy-resis­tant due to overexpression of con­trolled VIALE-A trial to improve OS when com­pared to azac-
the MDR1 mul­ti­drug-resis­tance gene.10 itidine plus pla­cebo (14.7 months vs 9.6 months; haz­ard ratio
Finally, sev­ eral fac­tors influ­
ence the type and inten­ sity of [HR], 0.66; 95% CI, 0.52-0.85; P < .001) in patients over 75 or
treat­ments offered to older patients, lead­ing to decreased sur­ in youn­ger patients with sig­nif­i­cant comorbidities.14 Moreover,
vival. Older patients are less likely to be offered up-front treat­ the com­ pos­ite com­ plete remis­sion (CR) rate was 66.4% for
ment for AML and are less likely to undergo allo­ge­neic stem cell patients in the AZA-VEN group com­pared to 28.3% in the con­
trans­plant.11,12 This is despite the fact that stud­ies have dem­on­ trol group, and rates of red blood cell and plate­let trans­fu­sion
strated that treat­ment of any inten­sity improves sur­vival in this inde­pen­dence were 59.8% and 68.5%, respec­tively. Regarding
patient pop­u­la­tion.11 The rea­sons for these disparities include safety, rates of grade ≥3 neutropenia and neutropenic fever
phy­si­cian bias regard­ing the treat­ment of older patients, care­ were higher in the AZA-VEN group; how­ever, 30-day mor­tal­ity
giver issues, and other socio­eco­nomic inequities. was sim­i­lar between the 2 groups (7% with AZA-VEN and 6%
Given the urgent need to address disparities in out­comes with AZA-pla­cebo).
for older indi­vid­u­als with AML, in 2020 the Amer­i­can Society of Molecular sub­group anal­y­sis of the VIALE-A study sug­gests
Hematology published guide­lines for treating these patients.13 that muta­tions in IDH1 or IDH2 may con­fer a par­tic­u­larly favor­
The guide­lines are based on a sys­tem­atic review of the lit­er­ able response to AZA-VEN. A pooled anal­y­sis of the phase 1b and
a­ture and con­sider many of the pre­vi­ously discussed fac­tors. phase 3 stud­ies showed that CR/complete remission with par-
Included is a rec­om­men­da­tion to offer anti­leu­ke­mic ther­apy tial hematologic recovery (CRh) for patients with an IDH1 muta­
over best sup­port­ive care and to attempt inten­sive ther­apy tion was 59% with AZA-VEN vs 9% with AZA-pla­cebo. CR/CRh
when pos­si­ble. As newer ther­a­pies con­tinue to emerge, these for patients with an IDH2 muta­tion was 80% with AZA-VEN vs 6%
guide­lines must be adapted and mod­i­fied, par­tic­u­larly with with AZA-pla­cebo.15 This improve­ment in remis­sion rates trans­
regard to inten­sity of treat­ment. Ultimately, opti­mal ther­apy for lated into an improve­ment in dura­tion of response and OS. This
older adults with AML is that which pro­vi­des the best depth of dif­
fered from other molec­ u­
lar sub­sets, such as FLT3-mutated
response, imparts the least amount of tox­ic­ity, and meets the or TP53-mutated dis­ ease, where responses were bet­ ter with
goals of the indi­vid­ual patient. AZA-VEN, but OS was not affected.
Recent efforts to develop safer and more effec­tive treat­ The oral IDH1 inhib­i­tor ivosidenib is another FDA-approved
ments for older adults with AML are begin­ning to pay off. Lead- front­line ther­apy for older patients with AML. This treat­ ment
ing the way has been the US Food and Drug Administration was ini­tially stud­ied in a phase 1, mul­ti­cen­ter, open-label, dose-
(FDA)-approved front­line com­bi­na­tion of azacitidine and vene- esca­la­tion, dose-expan­sion study in patients with AML with an
Prakash Singh Shekhawat
Therapies for dif­fi­cult AML pop­u­la­tions | 25
Newly diagnosed
AML in an older
pa ent

Clinical trial*

No targetable
Targetable muta on TP53 muta on
muta on

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/24/1851553/24wilde.pdf by guest on 13 December 2021


HMA + venetoclax
HMA + venetoclax
FLT3 IDH1 IDH2 magrolimab +/- HMA
LDAC + glasdegib
APR-246 + aza

HMA + venetoclax
HMA + venetoclax HMA + venetoclax
FLT3 inhibitor +/-
Ivosidenib +/- HMA Enasidenib +/- HMA
HMA

Figure 2.  Algorithm for the treatment of newly diagnosed AML in the older patient.

IDH1 muta­tion.16 The median age in the study was 76.5 years, and
56% of patients were over 75. The CR/CRh rate was 42%, with CLINICAL CASE
a median time to response of 2.8 months. The median fol­low-up An 81-year-old woman was treated with AZA-VEN for newly diag­
was 23.5 months, with a median OS of 12.6 months. The util­ity nosed AML. After 2 cycles, her dis­ease went into CR. She remained
of ivosidenib as a sin­gle agent is lim­ited, but it may be a good on this treat­ment for 7 cycles before her dis­ease relapsed. Repeat
option for patients unable to receive venetoclax or tol­er­ate com­ molec­u­lar test­ing showed a new FLT3-ITD muta­tion and a new
bi­na­tion treat­ment. A study of azacitidine, venetoclax, and ivos- muta­tion in IDH2.
idenib is under­way (NCT03471260), and the results, if pos­i­tive, In spite of the advances made in the first-line treat­ment of
will increase the value of this agent. AML, approx­i­ma­tely 50% of patients expe­ri­ence a relapse.19,20
The phase 2 BRIGHT AML 1003 study assessed the com­bi­na­ The treat­ment options for relapsed or refrac­tory (R/R) dis­ease
tion of low-dose cytarabine and glasdegib, a hedge­hog path­ remain lim­ited, and 5-year OS for these patients is approx­i­ma­tely
way inhib­i­tor, vs low-dose cytarabine alone.17 The median age 10%.21 Outcomes after first-line treat­ment with a hypomethylat-
in the glasdegib arm was 77 and in the low-dose cytarabine ing agent and venetoclax are par­tic­ul­arly poor, with a median
arm, 75. The addi­tion of glasdegib improved median OS (8.8 vs sur­vival of 2.4 months.22 At the time of relapse, molec­u­lar test­
4.6 months; HR, 0.51; 80% CI, 0.39-0.67; P = .0004). This led to ing should be repeated to assess for the pres­ence of tar­get­able
the FDA approval of glasdegib plus low-dose cytarabine for the muta­tions. Due to clonal evo­lu­tion, pre­vi­ously iden­ti­fi­able tar­
treat­ment of older patients with AML. However, given the other gets may be lost, or the emer­gence of subclones may lead to the
FDA-approved treat­ment options, which appear to be more pres­ence of new tar­gets.23
effi­ca­cious and equally as safe, the clin­ic
­ al util­ity of this treat­ Several targeted ther­a­pies have been approved for the treat­
ment is ques­tion­able. Subsequent sub­set anal­y­sis indi­cated ment of R/R AML. For patients with FLT3-mutated dis­ease, gilter-
slightly improved sur­vival in patients with sec­ond­ary AML as itinib is a potent oral FLT3 inhib­i­tor that can be used as a sin­gle
opposed to de novo dis­ease.18 Though no head-to-head com­ agent. In the ran­dom­ized phase 3 ADMIRAL study, gilteritinib
par­i­son exists, this sur­vival was shorter than that of patients improved OS when com­pared to inves­ti­ga­tor-choice che­mo­
treated with AZA-VEN in VIALE-A.14,18 ther­apy (9.3 months vs 5.6 months; two-sided P < .001).24 About
Numerous tri­als eval­u­at­ing targeted ther­a­pies, com­bi­na­tion 34% of patients receiv­ing gilteritinib had a CR/compelete remis-
ther­a­pies, and novel agents for the front­line treat­ment of older sion with incomplete count recovery com­pared to 15.3% in the
patients with AML are under­way. In addi­tion, geri­at­ric assess­ che­mo­ther­apy group (risk dif­fer­ence, 18.6 per­cent­age points;
ment tools are being devel­oped and eval­ua ­ ted with the hope of 95% CI, 9.8-27.4). Given its mod­est effi­cacy as a sin­gle agent,
improv­ing treat­ment selec­tion, side effects, and patient qual­ity gilteritinib is being stud­ied in com­bi­na­tion with other ther­a­pies
of life. These advances will con­tinue to have a pos­i­tive impact on in the relapsed and front­line set­ting. Early data on the com­bi­na­
the lives of older patients with AML. (See Figure 2 for a treat­ment tion of gilteritinib and venetoclax for relapsed FLT3-mutated AML
algo­rithm for older patients.) look prom­is­ing.25

26  |  Hematology 2021  |  ASH Education Program


Relapsed AML in
an older paent

Clinical trial*

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/24/1851553/24wilde.pdf by guest on 13 December 2021


No targetable Targetable
mutaon mutaon

Gemtuzumab
ozogamicin FLT3 IDH1 IDH2
HMA + venetoclax

Gilterinib +/- Ivosidenib +/- Enasidenib +/-


HMA HMA HMA

Figure 3.  Algorithm for the treatment of relapsed older AML patients.

The pre­vi­ously men­tioned IDH1 inhib­i­tor, ivosidenib, is also new can­di­dates for targeted treat­ments con­tinue to be iden­
approved for the treat­ment of R/R AML with an IDH1 muta­tion. ti­fied. (See Figure 3 for a treat­ment algo­rithm for patients with
In a large, sin­gle-arm phase 1 study, ivosidenib was shown to be relapsed dis­ease.)
safe and well tol­er­ated and to induce a CR or CRh in 30.4% of
patients.26 Similarly, the IDH2 inhib­i­tor enasidenib is approved for
the treat­ment of R/R AML with a muta­tion in IDH2. This approval
was based on the phase 1/2 study that showed a CR rate of CLINICAL CASE
19.3% in this pop­u­la­tion.27 However, the phase 3 study for this A 78-year-old woman with a past med­i­cal his­tory of hyper­ten­
drug failed to meet the pri­mary OS end point.28 Combination sion devel­oped leu­ko­cy­to­sis, ane­mia, and throm­bo­cy­to­pe­nia.
stud­ies with enasidenib are under­way. A bone mar­row biopsy revealed 60% mye­lo­blasts. Immunophe-
Finally, the some­what con­tro­ver­sial CD33-targeted anti­body- notypically, the blasts were CD34+, CD13+, and CD33+. She was
drug con­ju­gate gemtuzumab ozogamicin (GO) is also approved diag­nosed with AML. Next-gen­er­a­tion sequenc­ing showed a
for patients with AML that is R/R. First approved in 2000, GO muta­tion in TP53.
was vol­un­tar­ily with­drawn from the US mar­ket in 2010 due to TP53, an essen­tial tumor sup­pres­sor gene, is mutated in up
safety con­cerns. Reapproval was granted in 2017 after fur­ther to 10% of AML cases.31 It is found more fre­quently in ther­apy-
review of the data. In the ALFA 9801 trial, GO, which was given as related and com­plex-kar­yo­type AML. This muta­tional sta­tus is
a sin­gle agent or in com­bi­na­tion with che­mo­ther­apy to patients an impor­tant prog­nos­tic fac­tor that con­sis­tently cor­re­sponds to
with R/R dis­ease, dem­on­strated a 39% CR rate.29 Pooled anal­y­sis extremely poor out­comes, includ­ing low response rates to tra­
of the data from 3 other phase 2 stud­ies showed a slightly lower di­tional cyto­toxic che­mo­ther­apy and rare instances of long-term
over­all remis­sion rate of 26%.30 There is inter­est in com­bi­na­tion sur­vival after allo­ge­neic trans­plan­ta­tion.32,33
ther­apy with GO and stud­ies in this area are ongo­ing. Unfortunately, the novel agents approved for the treat­ment
Although these agents rep­re­sent prog­ress for the treat­ment of AML, includ­ing ther­apy related and com­plex kar­yo­type, have
of patients with R/R AML, response rates and long-term sur­vival not led to sig­nif­i­cant improve­ment in the sur­vival of patients with
are still lim­ited, and clin­i­cal tri­als remain the pre­ferred option TP53-mutated AML. For exam­ple, a 2018 study eval­u­ated this sub­
at first relapse or after the use of targeted ther­ a­
pies. Many group of patients in the reg­is­tra­tion trial of CPX-351 vs 7 + 3 in
novel agents are under inves­ti­ga­tion, includ­ing small mol­e­cule older adults with newly diag­nosed high-risk or sec­ond­ary AML.34
inhib­i­tors, immunotherapies, and com­bi­na­tion ther­a­pies, and Patients with the muta­tion had a median sur­vival of 5.7 months
Prakash Singh Shekhawat
Therapies for dif­fi­cult AML pop­u­la­tions | 27
in the CPX-351 arm and 5.1 months in the 7 + 3 arm, dem­on­strat­ Finally, the ongo­ing trial GLAD-AML (NCT03798678) addresses
ing no ben­e­fit. A sub­se­quent small study by Kim et al,35 which this poor-risk pop­ul­a­tion in a ran­dom­ized phase 2 study com­bin­
looked at the real-world use of CPX-351 in 53 patients, 30% of ing glasdegib with DEC5 or DEC10 in patients with newly diag­
whom had TP53-mutated AML, showed more encour­ag­ing results nosed poor-risk AML.
in this sub­set, with an overall response rate (ORR) of 57% (8/14)
and a 63% measurable residual disease neg­a­tiv­ity rate by next Conclusion
generation sequencing. However, these ret­ro­spec­tive data should Understanding who will ben­e­fit from cur­rent and future novel
be interpreted cau­tiously. Skepticism about the effi­cacy of CPX-351 AML ther­ap ­ ies is an evolv­ing area. Patient age and fit­ness, dis­
for TP53-­mutated AML remains. ease biol­ogy, and treat­ment mech­a­nism of action must all­be
The pre­vi­ously discussed VIALE-A trial showed sig­nif­i­cantly taken into con­sid­er­ation when choos­ing whom to treat and how.
higher rates of com­ pos­
ite remis­ sion for patients with TP53- In gen­eral, even low-inten­sity treat­ment pro­vi­des a sur­vival ben­
mutated AML who received azacytadine and venetoclax than e­fit over best sup­port­ive care to patients of all­ages and molec­
those in the con­trol group (55.3% vs 0%, respec­tively; 95% CI, u­lar and cyto­ge­netic sub­groups. Trials looking to build on recent
38.3-71.4; P < .001).14 However, this did not trans­late to an increase improve­ments in out­comes are ongo­ing, includ­ing some with

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/24/1851553/24wilde.pdf by guest on 13 December 2021


in OS. Disappointingly, com­bin­ing venetoclax with 10 days of dou­blet and trip­let ther­apy as well as oth­ers looking at drug
decitabine improved nei­ther response rates nor OS.36 The addi­ sequenc­ing, MRD erad­i­ca­tion, and main­te­nance.
tion of venetoclax to more aggres­sive ther­apy (FLAG-IDA) also
failed to show improve­ment in the TP53-mutated AML sub­group.37 Conflict-of-inter­est dis­clo­sure
Hope for this poor-risk pop­u­la­tion likely lies in the use of Lindsay Wilde: no con­flicts to dis­close.
novel agents. For exam­ple, magrolimab is a first-in-class mac­ Margaret Kasner: research funding: Astellas, Diachi, Gilead, Jazz,
ro­phage immune check­ point inhib­ i­
tor targeting CD47. CD47 Telios, Ono, Otsuka, Pfizer; hon­or­ aria: Kite, Jazz, Ono.
is a “do not eat me” sig­nal overexpressed in mul­ti­ple can­cers,
includ­ing AML.38 Its acti­ va­ tion leads to mac­ ro­
phage immune Off-label drug use
eva­ sion. Magrolimab elim­ i­
na­tes tumor cells through mac­ ro­ Lindsay Wilde: no discussion of off-label drug use outside of the
phage phago­cy­to­sis. Data were presented at ASH 2020 from setting of clinical trials.
a trial using magrolimab plus azacitidine in patients with newly Margaret Kasner: no discussion of off-label drug use outside of
diag­nosed AML who were inel­i­gi­ble for inten­sive che­mo­ther­apy. the setting of clinical trials.
The study pri­mar­ily enrolled patients with TP53-mutated AML
(65%, as they were pref­er­en­tially enrolled after an early pro­to­col Correspondence
amend­ment).39 The com­bi­na­tion induced a 71% ORR and 48% Margaret Kasner, Thomas Jefferson University, Medical Oncology,
CR rate in this high-risk sub­group. Moreover, the median OS for 834 Chestnut St, Ste 315, Philadelphia, PA 19107; e-mail: margaret​
these patients was 12.9 months. Based upon these data and the ­.kasner@jefferson​­.edu.
high unmet need in this pop­u­la­tion, a ran­dom­ized phase 3 trial
of magrolimab plus azacitidine vs AZA-VEN or inten­sive induc­tion References
as a front­line ther­apy for TP53-mutant AML is pre­par­ing to begin 1. National Cancer Institute. SEER pre­lim­i­nary can­cer inci­dence rate esti­ma­
tes for 2017, and diag­no­sis years 2000 to 2017. Accessed 2019. https:​­/​­/seer​
recruit­ment (NCT04778397). ­.cancer​­.gov​­/statistics​­/preliminary​­-estimates​­/.
Another novel agent that tar­gets TP53 more spe­cif­i­cally is 2. Shallis RM, Wang R, Davidoff A, Ma X, Zeidan AM. Epidemiology of acute
APR-246, a meth­yl­ated PRIMA-1 ana­logue. It is a first-in-class small mye­loid leu­ke­mia: recent prog­ress and endur­ing chal­lenges. Blood Rev.
mol­e­cule that selec­tively induces apo­pto­sis in TP53 mutant can­ 2019;36:70-87.
3. Mohammadi M, Cao Y, Glimelius I, Bottai M, Eloranta S, Smedby KE. The
cer cells.40 The agent is spon­ta­ne­ously converted into the active
impact of comorbid dis­ease his­tory on all­-cause and can­cer-spe­cific mor­
spe­ cies meth­ y­
lene quinuclidinone, which cova­ lently binds to tal­ity in mye­loid leu­ke­mia and mye­loma—a Swed­ish pop­u­la­tion-based
cys­te­ine res­i­dues in mutant p53, thereby pro­duc­ing ther­mo­ study. BMC Cancer. 2015;15:850.
dy­namic sta­bi­li­za­tion of the pro­tein and shifting equi­lib­rium 4. Appelbaum FR, Gundacker H, Head DR, et al. Age and acute mye­loid leu­
toward a func­tional con­for­ma­tion. Two early phase stud­ies were ke­mia. Blood. 2006;107(9):3481-3485.
5. Klepin HD, Estey E, Kadia T. More ver­sus less ther­apy for older adults with
recently published assessing the com­bi­na­tion of APR-246 and acute mye­loid leu­ke­mia: new per­spec­tives on an old debate. Am Soc Clin
azacitidine. A phase 1b/2 study in patients with TP53-mutated Oncol Educ Book. 2019;39:421-432.
myelo dysplasic syndrome (MDS) or low-blast per­cent­age AML 6. Bhatt VR, Chen B, Gyawali B, Lee SJ. Socioeconomic and health sys­tem fac­
showed the treat­ment to be safe and well tol­er­ated.41 The ORR tors asso­ci­ated with lower uti­li­za­tion of hema­to­poi­etic cell trans­plan­ta­tion
in older patients with acute mye­loid leu­ke­mia. Bone Marrow Transplant.
was 88%, with a CR rate of 61% for the MDS cohort and 50% for
2018;53(10):1288-1294.
the AML; median OS was 10.8 months (95% CI, 8.1-13.4). A sim­i­ 7. Fröhling S, Schlenk RF, Kayser S, et al; Ger­man-Austrian AML Study Group.
lar phase 2 study of APR-246 plus azacitidine included patients Cytogenetics and age are major deter­mi­nants of out­come in inten­sively
with MDS, low-blast AML, and AML with blasts >30%.42 Response treated acute mye­loid leu­ke­mia patients older than 60 years: results from
rates and OS for the MDS and low-blast AML cohorts were sim­i­lar AMLSG trial AML HD98-B. Blood. 2006;108(10):3280-3288.
8. Prassek VV, Rothenberg-Thurley M, Sauerland MC, et al. Genetics of acute
to the pre­vi­ously discussed trial. However, response rates and mye­loid leu­ke­mia in the elderly: muta­tion spec­trum and clin­i­cal impact
sur­vival in patients with AML with >30% blasts were lower (ORR in inten­sively treated patients aged 75 years or older. Haematologica.
50%, CR 0%, OS 3 months). These stud­ies are encour­ag­ing for 2018;103(11):1853-1861.
patients with MDS or low-blast AML but sug­gest that trip­let ther­ 9. Herold T, Rothenberg-Thurley M, Grunwald VV, et al. Validation and refine­
ment of the revised 2017 Euro­pean LeukemiaNet genetic risk strat­i­fic ­ a­tion
apy or other com­bi­na­tions with APR-246 should be explored in
of acute mye­loid leu­ke­mia. Leukemia. 2020;34(12):3161-3172.
AML with higher blast counts. One such study looking at APR-246 10. Leith CP, Kopecky KJ, Godwin J, et  al. Acute mye­ loid leu­ke­
mia in the
plus azacitidine and venetoclax is ongo­ing (NCT04214860). elderly: assess­ment of mul­ti­drug resis­tance (MDR1) and cyto­ge­net­ics distin-

28  |  Hematology 2021  |  ASH Education Program


guishes bio­logic sub­groups with remark­ably dis­tinct responses to stan­dard 29. Merabet F, Boissel N, Reman O, et al. Outcome of relapsed AML patients
che­ mo­ ther­apy. A Southwest Oncology Group Study. Blood. 1997;89(9): aged between 50 and 70: the ALFA group expe­ri­ence (ALFA 9801 study).
3323-3329. Blood. 2007;110(11):1857.
11. Medeiros BC, Satram-Hoang S, Hurst D, Hoang KQ , Momin F, Reyes C. 30. Larson RA, Sievers EL, Stadtmauer EA, et al. Final report of the effi­cacy and
Big data anal­ y­sis of treat­ ment pat­ terns and out­ comes among elderly safety of gemtuzumab ozogamicin (Mylotarg) in patients with CD33-pos­i­
acute mye­ loid leu­ke­mia patients in the United States. Ann Hematol. tive acute mye­loid leu­ke­mia in first recur­rence. Cancer. 2005;104(7):1442-
2015;94(7):1127-1138. 1452.
12. Muffly L, Pasquini MC, Martens M, et al. Increasing use of allo­ge­neic hema­ 31. Papaemmanuil E, Gerstung M, Bullinger L, et al. Genomic clas­si­fi­ca­tion and
to­poi­etic cell trans­plan­ta­tion in patients aged 70 years and older in the prog­no­sis in acute mye­loid leu­ke­mia. N Engl J Med. 2016;374(23):2209-
United States. Blood. 2017;130(9):1156-1164. 2221.
13. Sekeres MA, Guyatt G, Abel G, et al. Amer­i­can Society of Hematology 2020 32. Kadia TM, Jain P, Ravandi F, et al. TP53 muta­tions in newly diag­nosed acute
guide­lines for treating newly diag­nosed acute mye­loid leu­ke­mia in older mye­loid leu­ke­mia: clinicomolecular char­ac­ter­is­tics, response to ther­apy,
adults. Blood Adv. 2020;4(15):3528-3549. and out­comes. Cancer. 2016;122(22):3484-3491.
14. DiNardo CD, Jonas BA, Pullarkat V, et al. Azacitidine and venetoclax in pre­vi­ 33. Najima Y, Sadato D, Harada Y, et al. Prognostic impact of TP53 muta­tion,
ously untreated acute mye­loid leu­ke­mia. N Engl J Med. 2020;383(7):617-629. monosomal kar­yo­type, and prior mye­loid dis­or­der in nonremission acute
15. Pollyea DA, Dinardo CD, Arellano ML, et al. Results of venetoclax and azacit- mye­loid leu­ke­mia at allo-HSCT. Bone Marrow Transplant. 2021;56(2):334-
idine com­bi­na­tion in che­mo­ther­apy inel­i­gi­ble untreated patients with acute 346.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/24/1851553/24wilde.pdf by guest on 13 December 2021


mye­loid leu­ke­mia with IDH 1/2 muta­tions. Blood. 2020;136(suppl 1):5-7. 34. Goldberg AD, Talati C, Desai P, et  al. TP53 muta­ tions pre­
dict poorer
16. Roboz GJ, DiNardo CD, Stein EM, et al. Ivosidenib induces deep dura­ble responses to CPX-351 in acute mye­loid leu­ke­mia. Blood. 2018;132(suppl
remis­sions in patients with newly diag­nosed IDH1-mutant acute mye­loid 1):1433.
leu­ke­mia. Blood. 2020;135(7):463-471. 35. Kim GY, Koprivnikar JL, Testi R, et al. Treatment with CPX-351 induces deep
17. Cortes JE, Heidel FH, Hellmann A, et  al. Randomized com­par­i­son of low responses and TP53 muta­tion clear­ance in patients with t-AML and AML
dose cytarabine with or with­out glasdegib in patients with newly diag­ MRC, includ­ing youn­ger patients and those with pre-existing MPNs: a real-
nosed acute mye­ loid leu­ ke­
mia or high-risk myelodysplastic syn­ drome. world expe­ri­ence. Blood. 2020;136(suppl 1):49-50.
Leukemia. 2019;33(2):379-389. 36. Kim K, Maiti A, Kadia TM, et al. Outcomes of TP53-mutant acute mye­loid
18. Hueser M, Robak T, Montesinos P, et al. Glasdegib (GLAS) plus low-dose leu­ke­mia with venetoclax and decitabine. Blood. 2020;136(suppl 1):33-36.
cytarabine (LDAC) in AML or MDS: BRIGHT AML 1003 final report and 4-year 37. Lachowiez C, Konopleva M, Kadia TM, et al. Interim anal­y­sis of the phase
over­all sur­vival (OS) fol­low-up. J Clin Oncol. 2020;38(suppl 15):7509. 1b/2 study of the BCL-2 inhib­i­tor venetoclax in com­bi­na­tion with stan­dard
19. Zhu C-Y, Chen G-F, Zhou W, et al. Outcome and prog­nos­tic fac­tors of high- inten­sive AML induc­tion/con­sol­i­da­tion ther­apy with FLAG-IDA in patients
risk acute mye­loid leu­ke­mia after allo­ge­neic hema­to­poi­etic stem cell trans­ with newly diag­nosed or relapsed/refrac­tory AML. Blood. 2020;136(suppl
plan­ta­tion. Ann Transplant. 2019;24:328-340. 1):18-20.
20. Oliva EN, Franek J, Patel D, Zaidi O, Nehme SA, Almeida AM. The real-world 38. Chao MP, Takimoto CH, Feng DD, et al. Therapeutic targeting of the mac­
inci­dence of relapse in acute mye­loid leu­ke­mia (AML): a sys­tem­atic lit­er­a­ ro­phage immune check­point CD47 in mye­loid malig­nan­cies. Front Oncol.
ture review (SLR). Blood. 2018;132(suppl 1):5188. 2019;9:1380.
21. Ganzel C, Sun Z, Cripe LD, et al. Very poor long-term sur­vival in past and 39. Sallman DA, Asch A, Kambhampati S, et al. The first-in-class anti-CD47 anti­
more recent stud­ies for relapsed AML patients: the ECOG-ACRIN expe­ri­ body magrolimab com­bined with azacitidine is well-tol­er­ated and effec­
ence. Am J Hematol. 2018;93(8):1074-1081. tive in AML patients: phase 1b results. Blood. 2019;134(suppl 1):569.
22. Abhishek M, Caitlin RR, Jorge EC, et al. Outcomes of relapsed or refrac­tory 40. Sallman DA. To tar­get the untargetable: elu­ci­da­tion of syn­ergy of APR-
acute mye­loid leu­ke­mia after front­line hypomethylating agent and veneto- 246 and azacitidine in TP53 mutant myelodysplastic syn­dromes and acute
clax reg­i­mens. Haematologica. 2020;106(3):894-898. mye­loid leu­ke­mia. Haematologica. 2020;105(6):1470-1472.
23. Vosberg S, Greif PA. Clonal evo­lu­tion of acute mye­loid leu­ke­mia from diag­ 41. Sallman DA, DeZern AE, Garcia-Manero G, et al. Eprenetapopt (APR-246)
no­sis to relapse. Genes Chromosomes Cancer. 2019;58(12):839-849. and azacitidine in TP53-mutant myelodysplastic syn­dromes. J Clin Oncol.
24. Perl AE, Martinelli G, Cortes JE, et al. Gilteritinib or che­mo­ther­apy for relapsed 2021;39(14):1584-1594.
or refrac­tory FLT3-mutated AML. N Engl J Med. 2019;381(18):1728-1740. 42. Cluzeau T, Sebert M, Rahmé R, et  al. Eprenetapopt plus azacitidine in
25. Perl AE, Daver NG, Pratz KW, et al. Venetoclax in com­bi­na­tion with gilteri- TP53-mutated myelodysplastic syn­dromes and acute mye­loid leu­ke­mia: a
tinib in patients with relapsed/refrac­tory acute mye­loid leu­ke­mia: a phase phase II study by the groupe fran­co­phone des myélodysplasies (GFM). J
1b study. Blood. 2019;134(suppl 1):3910. Clin Oncol. 2021;39(14):1575-1583.
26. DiNardo CD, Stein EM, de Botton S, et al. Durable remis­sions with ivosidenib
in IDH1-mutated relapsed or refrac­tory AML. N Engl J Med. 2018;378(25):
2386-2398.
27. Stein EM, DiNardo CD, Pollyea DA, et al. Enasidenib in mutant IDH2 relapsed
or refrac­tory acute mye­loid leu­ke­mia. Blood. 2017;130(6):722-731.
28. Bristol Meyers Squibb. Bristol Meyers Squibb pro­vi­des update on phase 3
IDHENTIFY trial in patients with relapsed or refrac­tory acute mye­loid leu­ke­ © 2021 by The Amer­i­can Society of Hematology
mia. News release. 24 August 2020. DOI 10.1182/hema­tol­ogy.2021000228

Prakash Singh Shekhawat


Therapies for dif­fi­cult AML pop­u­la­tions | 29
CHALLENGES IN MULTIPLE MYELOMA TREATMENT

High-risk multiple myeloma: how to treat


at diagnosis and relapse?

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/30/1851766/30mateos.pdf by guest on 13 December 2021


María-Victoria Mateos, Borja Puertas Martínez, and Verónica González-Calle
Hospital Universitario de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), Centro de Investigación del Cancer (IBMCC-USAL,
CSIC), Salamanca, Spain

Patients with multiple myeloma have experienced a great improvement in survival over the past century because of the
introduction of novel therapeutic strategies. However, a subgroup of patients with poorer outcomes than expected is
considered high risk and identified by the presence of patient- and disease-based factors such as frailty, extramedullary
disease, cytogenetic abnormalities, or even relapses occurring earlier than expected according to the baseline factors.
Although the management of patients with high-risk features is not well established because of the lack of specific trials
in this subgroup of patients and because of their underrepresentation in the clinical trials, treatment should be planned
on 2 pillars: (1) poor prognosis with the presence of high-risk features can be at least improved or even abrogated by
achieving a deep and sustained response over time, and (2) this can most likely be obtained through using the best ther-
apeutic options and in a response-adapted way. Some clinical trials that have been planned or are ongoing include only
patients with high-risk features, using the most effective therapies (proteasome inhibitors, immunomodulatory drugs,
and anti-CD38 monoclonal antibodies) as well as chimeric antigen receptor T cells and T-cell engagers that will unravel
what the best therapeutic approach will be to overcome the poor prognosis of the presence of high-risk features.

LEARNING OBJECTIVES
• Identify high-risk myeloma based on patient-, disease-, or outcome-based factors
• Be able to define the key objectives to overcome poor prognosis with the presence of high-risk features
• Define the best therapeutic strategy for patients with high-risk features

CLINICAL CASE maintenance therapy, relapse occurred with reappearance


A 48-year-old man with newly diagnosed (ND), Revised of the M-component in urine. He was included in a clini-
International Staging System (R-ISS) III (ISS III plus del(17p)), cal trial and treated with B-cell maturation antigen (BCMA)
Bence-Jones κ multiple myeloma (MM) sought treat- chimeric antigen receptor T (CAR-T) cells, and a new sCR
ment and consultation for MM that had been diagnosed and MRD negativity were achieved. The patient continues
in another institution. The patient had an active lifestyle, in follow-up.
and the workup showed mild anemia and small lytic lesions
in the pelvis and femora as myeloma-defining events. His
Eastern Cooperative Oncology Group performance status How do we define high-risk patients with MM?
was 1. He was treated with 6 induction cycles of lenalid- Table 1 summarizes the most relevant patient- and disease-
omide, bortezomib, and dexamethasone (RVd), achieving based factors to define high-risk patients.
a very good partial response, followed by high-dose mel-
phalan and autologous stem cell transplantation (HDM- Patient-based factors
ASCT), achieving stringent complete remission (sCR) with Frailty
minimal residual disease (MRD) positivity. He rejected For a long time, chronological age influenced treatment
a second ASCT and proceeded to consolidation with 2 decisions, and the outcome was poor for the elderly. The
cycles of lenalidomide, carfilzomib, and dexamethasone, International Myeloma Working Group (IMWG), through
achieving sCR and MRD negativity. Maintenance with lena- a pooled analysis including 869 ND elderly patients en-
lidomide was prescribed. Twelve months after starting rolled in clinical trials, built a simplified geriatric score

30 | Hematology 2021 | ASH Education Program


Table 1. Patient- and dis­ease-based fac­tors for the iden­ti­fi­ca­tion bor­ing +1q had a shorter median pro­gres­sion-free sur­vival (PFS;
of high-risk MM 41.9 months) and OS (not reached) com­pared with those with­out
+1q (PFS of 65.1 months, P  =  .002 and OS not reached, P  =  .003).
High-risk fea­tures Definition The neg­a­tive impact on sur­vival with +1q may be more pro­found
Patient-based fac­tors if there is ampli­fi­ca­tion of 1q, defined by the pres­ence of 4 or
more cop­ies of chro­mo­some 1q (median PFS of 25.1 months) or in
  Frailty sta­tus IMWG frailty score
asso­ci­a­tion with other high-risk CAs (median PFS of 34.6 months).
Modified IMWG frailty score
R-MCI The poor prog­no­sis of patients who have +1q with a coexisting
GAH dele­tion of chro­mo­some 1p (del(1p)) has also been described.6
Although it is well accepted that del(17p) is the CA with more
Disease-based fac­tors
prog­nos­tic impact in MM, some ques­tions are under debate: (1)
  Aggressiveness in the Extramedullary dis­ease (no bone- What is the opti­mal thresh­old to pre­dict poor prog­no­sis? (2)
clin­i­cal pre­sen­ta­tion related plasmacytomas)
Plasma cell leu­ke­mia
Is TP53 an opti­mal molec­u­lar tar­get? (3) What about mono- or
biallelic dele­tion and/or inac­ti­va­tion of TP53 through muta­

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/30/1851766/30mateos.pdf by guest on 13 December 2021


LDH ele­vated
tions? Patients with a “dou­ble-hit” biallelic inac­ti­va­tion of TP53
  Cytogenetic ­ del(17p), t(4;14), t(14;16), amp1q, del(1p)
abnor­mal­i­ties are at high risk, espe­cially if this abnor­mal­ity coex­ists with ISS
3 (1.5-year PFS of 33%) in ND patients.7 The Intergroupe Franco-
 Mutations TP53
phone du Myélome (IFM) has recently reported in a large series
  Biochemical ­ LDH ele­vated of patients with NDMM that the pres­ence of iso­lated del(17p) was
abnor­mal­i­ties β2-microglobulin ≥5.5 mg/L
also asso­ci­ated with a poor out­come, although the poorest out­
Albumin lev­els ≤3.5 mg/L
come was reported for the dou­ble-hit patients.8
Prognostic scores In sum­mary, the opti­mal iden­ti­fi­ca­tion of high-risk MM based
 R-ISS R-ISS III: beta2-microglobulin ≥5.5 mg/L on CA is under con­struc­tion, but in clin­i­cal prac­tice, the CA rec-
plus either LDH ele­vated or high-risk ommended by the IMWG, together with abnor­mal­i­ties of chro­
CA (del(17p), t(4;14), or t(14;16))
mo­some 1 and muta­tional sta­tus of TP53, if pos­si­ble, would be
GAH, geri­at­ric assess­ment in hema­tol­ogy; R-MCI, Revised Myeloma nec­es­sary to define the risk at base­line. At the moment of the
Comorbidity Index. relapse, it would be opti­mal to repeat these eval­u­a­tions because
of the clonal evo­lu­tion and the poten­tial acqui­si­tion of new CAs
not detect­able at base­line.
based on age, comorbidities, and cog­ni­tive and phys­i­cal con­
di­tions to dis­tin­guish among fit (score   =   0), inter­me­di­ate fit­ness R-ISS
(score  =  1), and frailty (score ≥ 2). Frail patients showed a sig­nif­ The ISS risk model, includ­ ing albu­min and β2-microglobulin
i­cantly shorter over­all sur­vival (OS; 57% at 3 years) than unfit lev­els, was improved with the incor­po­ra­tion of 2 well-known
(76% at 3 years) and fit patients (84% at 3 years).1 Many clin­i­cal dis­ease-related prog­nos­tic bio­mark­ers, CA and serum lac­tate
tri­als are using an IMWG-mod­i­fied frailty model to iden­tify frail dehy­dro­ge­nase (LDH) lev­els, which are asso­ci­ated with a higher
patients because of the impor­tance of their iden­ti­fi­ca­tion for pro­lif­er­a­tive activ­ity, resulting in the R-ISS model.
the treat­ment deci­sion-mak­ing pro­cess.2 The R-ISS emerged from a large series of patients with NDMM,
and 3 sub­groups were defined: R-ISS I (n  =  871), includ­ing ISS
Disease-based fac­tors stage I, no high-risk CA, and nor­mal LDH level with a 5-year OS
Features asso­ci­ated with dis­ease aggres­sive­ness rate of 82%; R-ISS III (n  =  295), includ­ing ISS stage III and high-
The pres­ence of extramedullary dis­ease (EMD) or plasma cell leu­ risk CA or high LDH level with a 5-year OS of 40%; and R-ISS II
ke­mia (PCL), pri­mary or sec­ond­ary, is infre­quent, but these are (n  =  1894), includ­ing all­ the other pos­si­ble com­bi­na­tions and with
con­sid­ered high-risk fea­tures not only because the plasma cells a 5-year OS of 62%.9 This stag­ing sys­tem is still valid, although
escape from the bone mar­row envi­ron­ment but also because there are some lim­i­ta­tions: (1) most patients were assigned to
patients with EMD or PCL are dif­fi­cult to treat, with poor out­ the R-ISS II, includ­ing those with high-risk CA but not ISS III, (2)
comes with the cur­rent ther­a­pies (3-year sur­vival rate is 35% for some other high-risk CAs such as +1q were not included, and (3)
EMD3 and median OS is 12 months for PCL4). other, more com­plex geno­mic abnor­mal­i­ties such as muta­tions
or inac­ti­va­tion of TP53 were not con­sid­ered.
Cytogenetic abnor­mal­i­ties
The IMWG cur­ rently rec­ om­ mends the detec­ tion of t(4;14), Functional high-risk patients
t(14;16) and del (17/17p) in selected plasma cells by inter­phase In addi­tion to the above high-risk fea­tures, how do we rec­og­nize
fluo­res­cent in situ hybrid­iza­tion for the iden­ti­fi­ca­tion of high-risk those patients with no appar­ent high risk at diag­no­sis but who
patients.5 In newly diag­nosed MM (NDMM), the pres­ence of at prog­ress within the first 12 to 18 months after an opti­mal first line
least 1 high-risk cyto­ge­netic abnor­mal­ity (CA) is asso­ci­ated with of ther­apy? These patients are func­tional high risk with poor prog­
a median OS of 24.5 months, sig­nif­i­cantly shorter than the 50.5 no­sis, and fur­ther inves­ti­ga­tions are required to unravel if there is
months observed when there are not any CAs (P  <  .001). This is a clonal selec­tion or just an inad­e­quate eval­u­a­tion at diag­no­sis.
far from com­plete and requires being updated.
The gain of the long arm of chro­mo­some 1 (+1q) is a fre­quent What is the opti­mal man­age­ment for patients
CA observed in approx­i­ma­tely 30% of NDMMs and asso­ci­ated with high-risk fea­tures?
with poor out­ come. A ret­ ro­ spec­
tive study conducted in 201 If the iden­ti­fi­ca­tion of high-risk patients with MM is chal­leng­ing,
patients with NDMM treated with RVd Prakash
reported that Singh Shekhawat
patients har­ its man­age­ment is not easy either. So far, only a few clin­i­cal

Management of high-risk mye­loma  | 31


tri­als have been spe­cif­i­cally conducted in this pop­u­la­tion be- methasone) has been a ­ ble to sig­nif­i­cantly improve the out­come
cause the def­i­ ni­tion is het­ ero­
ge­
neous. In addi­ tion, high-risk of frail patients com­pared with VMP or Rd alone, according to a
sub­groups in clin­i­cal tri­als are quite small to gen­er­ate solid rec­ mod­i­fied IMWG frailty index (Table 2). In the relapsed-refrac­tory
om­men­da­tions. set­ting, other subanalyses of phase 3 clin­i­cal tri­als have reported
In 2021, we know that poor prog­no­sis with the pres­ence of how carfilzomib at dif­fer­ent doses and sched­ules or the com­
high-risk fea­tures can be at least improved or even abro­gated by bi­na­tion of pomalidomide-dexa­meth­a­sone plus isatuximab is
the achiev­ing a deep and sustained response over time, which fea­si­ble and ­able to improve the out­come of frail patients.15,16 Al-
can most likely be obtained through the use of novel ther­ap ­ eu­tic though this infor­ma­tion is obtained from clin­i­cal tri­als, the good
options.10 tox­ic­ity pro­file of all­novel agents makes it pos­si­ble to main­tain
At least 3 large meta-ana­ly­ses sup­port the use of MRD for ther­apy in the frail pop­u­la­tion.
mon­i­tor­ing the response in MM because of its prog­nos­tic value.
The most recent one included pub­ li­
ca­
tions up to June 2019, Management of patients with high-risk CA or R-ISS III
show­ing that the achieve­ment of unde­tect­able MRD improved with approved drugs
PFS (haz­ard ratio [HR], 0.33) and OS (HR, 0.45) in com­par­i­son Proteasome inhib­i­tors (PIs), immu­no­mod­u­la­tory drugs, and cor­

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/30/1851766/30mateos.pdf by guest on 13 December 2021


with the pres­ence of MRD. Moreover, its prog­nos­tic impact was ti­co­ste­roids are the key treat­ment ele­ments of patients with MM
sustained across the dif­fer­ent sub­groups, includ­ing those with patients with high-risk CA. For trans­plant-eli­gi­ble patients with
some high-risk fea­ tures such as elderly patients with NDMM, NDMM, the ques­ tion about bortezomib or carfilzomib as the
relapsed/refrac­tory patients, or even the pres­ence of high-risk opti­mal PI for high-risk patients remains under debate because
CA.11 the only phase 3 ran­dom­ized trial com­par­ing bortezomib with
Of note, the higher the sen­si­tiv­ity thresh­old for the MRD eval­ carfilzomib did not show any dif­fer­ence, but it did only include
u­ a­
tion and the lon­ ger the sustained unde­ tect­able MRD over patients with t(4;14).17
time, the higher the prog­nos­tic value. The use of moAbs (monoclonal antibodies) targeting SLAMF7
In addi­tion, the achieve­ment of unde­tect­able MRD can con­ and CD38 also has been eval­ua ­ ted in this set­ting. Although the
vert risk assess­ment in MM into some­thing dynamic, and the high addi­ tion of elotuzumab showed no sig­ nif­i­
cant ben­ e­
fit when
risk at base­line can be over­come when unde­tect­able MRD is com­bined with RVd in the phase 2 SWOG-1211 study,18 the addi­
achieved. In the PETHEMA/GEM2012MENOS65 trial, 458 patients tion of daratumumab has been shown to improve the out­come
with NDMM had lon­gi­tu­di­nal assess­ment of MRD after 6 induc­ of patients with NDMM and RRMM (relapsed refractory multiple
tion cycles with RVd, autol­o­gous trans­plan­ta­tion, and 2 con­sol­ myeloma) with high-risk CA in a recent meta-anal­ y­sis.19 HDM-
i­da­tion courses with RVd. The 3-year PFS rate for patients with ASCT con­tin­ues to be the stan­dard of care in high-risk NDMM
R-ISS I, II, or III was com­pa­ra­ble (95%, 94%, and 88%) if MRD was because of its capac­ity to achieve a higher unde­tect­able MRD
unde­tect­able after treat­ment. By con­trast, out­comes were pro­ rate, and tan­dem trans­plant is even con­sid­ered for this pop­u­la­
gres­sively poor for patients with R-ISS I, II, and III when MRD was tion based on the pos­i­tive data from the EMN02 trial, con­firmed
detect­able, with a 3-year PFS of 62%, 53%, and 28%, respec­ in the STAMINA trial at least in terms of PFS.20,21 However, tan­dem
tively, and anal­o­gous results were observed when OS was con­ trans­plant may not be nec­es­sary with the intro­duc­tion of moABs,
sid­ered. Similarly, out­come of patients with high-risk CA was espe­cially with the intro­duc­tion of cell ther­apy. Maintenance with
abro­gated when unde­tect­able MRD was achieved.12 lenalidomide is the stan­dard of care to improve the out­come of
One addi­tional aspect needs to be incor­po­rated in the MRD high-risk patients com­pared with obser­va­tion, but it should be
assess­ment: the MRD eval­u­a­tion out­side of the bone mar­row improved through the addi­tion of PIs or moABs, with pre­lim­i­nary
through the use of func­tional imag­ing tools such as pos­i­tron pos­i­tive data.22,23
emis­sion tomog­ra­phy/com­put­er­ized tomog­ra­phy.13 Deauville In the nontransplant-eli­gi­ble, high-risk patients with NDMM,
scores to focal lesions less than 4 and bone mar­ row uptake daratumumab, lenalidomide, and dexa­meth­a­sone would be the
show­ing the liver back­ground (Deauville score <4) have been first choice based on the results reported in the MAIA trial, with
iden­ti­fied as the best cut­off to define pos­i­tron emis­sion tomog­ a median PFS of 45.3 months com­pared with 29.6 months in the
ra­phy/com­put­er­ized tomog­ra­phy neg­a­tiv­ity after ther­apy and Rd arm (HR, 0.57).24
com­plete met­ab ­ olic response, as they have been described in In the RR (relapse or refractory) set­ting, the same approach
at least 2 clin­i­cal tri­als, the FORTE and CASSIOPETT substudy of is valid, and the com­bi­na­tion of choice for patients with high-
CASSIOPEIA trial. risk CA would be those with the higher likely prob­ab ­ il­ity of
achiev­ing unde­tect­able MRD (Table 2).
Management of frail patients Of note, the novel drug melflufen flufenamide has shown
The gen­eral approach described above is fea­si­ble for frail pa- prom­is­ing effi­cacy in 45 patients with EMD (extramedullary dis-
tients, but tol­er­ab
­ il­ity and qual­ity of life are cru­cial to deliver ease) included in the HORIZON trial, with an over­all response
treat­ments in order to reach depth responses. At least 1 clin­i­cal of 24% vs 30% in patients with­out EMD.25 Selinexor also may
trial has been conducted in unfit and frail patients with NDMM have a role in treating patients with del(17p) based on its
according to the IMWG frailty index, using ixazomib and dara- mech­an ­ ism of action and avail­­able evi­dence in some clin­i­cal
tumumab plus very low dose of dexa­meth­a­sone.14 Preliminary tri­als.26 Belantamb mafodotin, a BCMA-con­ju­gated moAb, pro­
results are encour­ag­ing, with 1-year OS rates of 96% and 74% for duced a response rate of 33%, which is sim­i­lar to that reported
unfit and frail patients, respec­tively. Subgroup anal­y­sis recently in patients with high-risk cyto­ge­net­ics.27 Further stud­ies are
conducted in the phase 3 tri­als ALCYONE and MAIA have also required to con­firm this effi­cacy. Table 2 shows the effi­cacy
shown how the addi­tion of daratumumab to either bortezomib, reported in patients with high-risk CA in the most rel­e­vant clin­
mel­pha­lan, and pred­ni­sone (VMP) or Rd (lenalidomida and dexa- i­cal tri­als in patients with NDMM and RRMM.

32  |  Hematology 2021  |  ASH Education Program


Table 2. Clinical stud­ies ongo­ing in patients with high-risk fea­tures

Registration num­ber Study design Population


ND high-risk MM
NCT03104842 Isatuximab-KRd as induc­tion, con­sol­i­da­tion, Transplant eli­gi­ble or inel­i­gi­ble
and main­te­nance del(17p) in ≥10% of puri­fied cells and/or t(4;14) and/or >3 cop­ies +1q21
ISS II or III
NCT03756896 Carfilzomib, pomalidomide, and Transplant eli­gi­ble achiev­ing at least par­tial response
dexa­meth­a­sone as main­te­nance after Presence of del(17p), t(4;14), t(14;16), t(14;20)
HDM-ASCT PCL at diag­no­sis
NCT04025450 Chidamide (HDAC inhib­i­tor)–lenalidomide, Transplant eli­gi­ble and inel­i­gi­ble
bortezomib, and dexa­meth­a­sone as Presence of del(17p), t(4;14), t(14;16), t(14;20)
induc­tion R-ISS III
IgD/IgE

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/30/1851766/30mateos.pdf by guest on 13 December 2021


Extramedullary plasmacytomas
Peripheral plasma cells by flow cytom­e­try ≥0.165%
NCT02128230 Induction with mel­pha­lan 20–KTD-PACE Transplant eli­gi­ble
followed by mel­pha­lan 80–KTd-PACE plus
ASCT and KTD-PACE con­sol­i­da­tion and GEP70 risk score of ≥0.66
KRd main­te­nance (1 year) and Kd (1 year)
NCT03549442 BCMA CAR-T + huCART19 in dif­fer­ent ISS III or R-ISS III or
sched­ules Metaphase kar­yo­type with >3 abnor­mal­i­ties except hyperdiploidy
Failure to achieve par­tial response or bet­ter to ini­tial ther­apy based on
PI and IMiD
NCT04196491 BCMA CAR-T bb2121 (ide-cel) (150-800  ×  106) R-ISS III
followed by lenalidomide as main­te­nance
NCT04436029 Autologous CD8+ T cells expressing an anti- High-risk patients who com­pleted pretransplant induc­tion
BCMA chi­me­ric anti­gen recep­tor antimyeloma treat­ment
NCT04133636 BCMA CAR-T JNJ-68284528 (cilta-cel) Less than com­plete response after first-line treat­ment and trans­plant
followed by lenalidomide main­te­nance followed or not by con­sol­i­da­tion
NCT04133636 BCMA CAR-T JNJ-68284528 (cilta-cel) Noneligible for trans­plant patients with ISS III
followed by lenalidomide and
daratumumab main­te­nance
Relapsed-refrac­tory high-risk MM
NCT03601078 BCMA CAR-T bb2121 (150-450  ×  106) R-ISS III and
PD <18 months after the first-line treat­ment includ­ing induc­tion,
trans­plant, and lenalidomide main­te­nance
PD <18 months since date of start ini­tial ther­apy, which must con­tain PI,
IMiD, and dexa­meth­a­sone
Less thanVGPR after induc­tion, includ­ing PI, IMiD, and dexa­meth­a­sone
and trans­plant (between 70 and 110 days after trans­plant)
NCT04133636 BCMA CAR-T JNJ-68284528 (cilta-cel) One prior line includ­ing PI, IMiD, and PD within the first 12 months after
trans­plant or the first-line treat­ment for nontransplant eli­gi­ble
NCT03104270 Elotuzumab in com­bi­na­tion with More than 2 prior lines, includ­ing PI and IMiD and
pomalidomide, carfilzomib, and del(17p), t(14;16), t(14:20)
dexa­meth­a­sone PCL
Extramedullary dis­ease
Doubling in lev­els of MM mark­ers in the past 3 months
Refractoriness to their most recent lenalidomide-containing reg­i­men
and PI-based reg­i­men
Renal fail­ure with CrCl between 15 and 30  mL  ×  min­ute
cilta-cel, ciltacabtagene autoleucel; CrCL, cre­at­i­nine clear­ance; GEP70, gene expression profiling-70; HDAC, histone deacetylase; ide-cel,
idecabtagene vicleucel; IMiD, immu­no­mod­u­la­tory drug; Kd, carfilzomib and dexa­meth­a­sone; KRd, carfilzomib, lenalidomide and dexa­meth­a­sone;
KTD-PACE, carfilzomib, tha­lid­o­mide, dexa­meth­a­sone, cis­platin, doxo­ru­bi­cin, cyclo­phos­pha­mide, and etoposide; PD, pro­gres­sion dis­ease; VGPR,
very good par­tial response.

Management of func­tional high-risk patients vi­ous ther­apy, show­ing that the addi­tion of carfilzomib to Rd
Although no spe­cific tri­als were performed until very recent- or daratumumab to Rd vs Rd in the ASPIRE and POLLUX tri­
ly, new tri­als with BCMA-targeted CAR-T cells have focused als resulted in a sig­nif­i­cant ben­e­fit for the tri­ple com­bi­na­tion
on this sub­group of patients (Table 3). Some sub­group anal­ com­pared with Rd. A sim­i­lar effect has been recently reported
y­sis in phase 3 tri­als focused on early relapses, defined as with the addi­tion of daratumumab to bortezomib or carfilzo-
those occur­ring within the first 12 to 18 months after the pre­ mib28-31 (Table 2).
Prakash Singh Shekhawat
Management of high-risk mye­loma  | 33
Table 3.  Efficacy of cur­rent treat­ment approaches for patients with NDMM with high-risk fea­tures (defined as del(17p), t(4;14), or t(14;16))

Transplant-eli­gi­ble NDMM
Clinical trial SWOG-1211 Cas­si­o­peia Forte Griffin
Population High risk* ITT High risk ITT High risk ITT High risk
Treatment EloVRd vs VRd DVTd vs VTD DVTd vs VTD KRd-T/KRd12 KRd-T/KRd12 DRVd vs RVd DRVd vs RVd
PFS (m)/HR 31 vs 34/0.96 NR/0.47 NR/0.67 NR/0.64 NR/0.51 NR/NA NR/NA
Transplant-inel­i­gi­ble NDMM
Clinical trial SWOG ALCYONE MAIA
Population ITT High risk ITT High risk Frail patients ITT High risk Frail patients
Treatment VRd- > Rd vs Rd VRd- > Rd vs Rd DVMP vs VMP DVMP vs VMP DVMP vs VMP DRd vs Rd DRd vs Rd DRd vs Rd
PFS (m)/HR 43 vs 30/0.74 38 vs 16/NA 36 vs 18/0.50 18 vs 18/0.78 33 vs 19/0.51 NR vs 34/0.54 45 vs 29/0.57 NR vs 30/0.62
Relapsed-refrac­tory MM
Clinical trial POLLUX ASPIRE ELOQUENT-2 TOURMALINE-MM1

34  |  Hematology 2021  |  ASH Education Program


Population ITT High risk Early relapse ITT High risk Early relapse ITT High risk ITT High risk
Treatment DRd vs Rd DRd vs Rd KRd vs Rd KRd vs Rd EloRd vs Rd EloRd vs Rd IRd vs Rd IRd vs Rd
PFS (m)/HR 44.5 vs 17.5/0.44 26.8 vs 8.3/0.37 0.38 26.3 vs 17.3/0.69 23 vs 13.9/0.7 21.4 vs 10.7/0.7 19.4 vs 14.9/0.70 NA/0.72(del17p) 20.6 vs 14.7/0.74 21.4 vs 9.7/0.54
NA/0.56 (t(4;14)
Clinical trial CASTOR ENDEAVOR CANDOR IKEMA
Population ITT High risk ITT High risk ITT High risk Early relapse ITT High risk
Treatment DVd vs Vd DVd vs Vd Kd vs Vd Kd vs Vd DKd vs Kd DKd vs Kd IsaKd vs Kd IsaKd vs Kd
PFS (m)/HR 16.7 vs 7.1/0.31 12.6 vs 6.2/0.41 18.7 vs 9.4/0.53 8.8 vs 6.0/0.7 28.6 vs 15.9/0.59 15.6 vs 5.6/0.49 CRrate 28 vs 3% NR vs 19.1/0.53 NA/0.72
Clinical trial OPTIMISMM BOSTON ICARIA ELOQUENT-3
Population ITT High risk ITT High risk ITT High risk ITT High risk
Treatment PVd vs Vd PVd vs Vd SVd vs Vd SVd vs Vd IsaPd vs Pd IsaPd vs Pd EloPd vs Pd EloPd vs Pd
PFS (m)/HR 11.2 vs 7.1/0.61 NA/0.56 11.2 vs 5.8/0.61 NA/0.67 11.5 vs 6.4/0.59 NA/0.66 10.3 vs 4.7/0.54 0.52
Clinical trial STORM HORIZON DREAMM-2 KARMMA-1
Population ITT High risk ITT High risk ITT High risk ITT High risk
Treatment Sd Sd Melflufen-dex Melflufen-dex Belamaf Belamaf Ide-cel Ide-cel
PFS (m)/HR 3.7 3.3* and 4.6* 4.2 3.0 3.9 2.1 8.8 10.4
*High-risk def­i­ni­tion: gene expres­sion pro­fil­ing high risk, t(14;16), t(14;20), del(17p), amp1q21, plasma cell leu­ke­mia, ele­vated serum LDH (2  ×  upper limit of nor­mal).
Belamaf, belantamaf mafodotin; dex, dexa­meth­a­sone; DKd, daratumumab, carfilzomib, and dexa­meth­a­sone; DRd, daratumumab, lenalidomide, and dexa­meth­a­sone; DRVd, daratumumab,
lenalidomide, bortezomib, and dexa­meth­a­sone; DVd, daratumumab, bortezomib, and dexa­meth­a­sone; DVMP, daratumumab, bortezomib, mel­pha­lan, and pred­ni­sone; DVTd, daratumumab,
bortezomib, tha­lid­o­mide, and dexa­meth­a­sone; Elo, elotuzumab; EloPd, elotuzumab, pomalidomide, and dexa­meth­a­sone; EloRd, elotuzumab, lenalidomide, and dexa­meth­a­sone; Ide-cel,
idecabtagene vicleucel; IRd, ixazomib, lenalidomide, and dexa­meth­a­sone; IsaKd, isatuximab, carfilzomib, and dexa­meth­a­sone; IsaPd, isatuximab, pomalidomide, and dexa­meth­a­sone; ITT,
inten­tion-to-treat pop­u­la­tion; Kd, carfilzomib and dexa­meth­a­sone; KRd, carfilzomib, lenalidomide, and dexa­meth­a­sone; KRd-T, carfilzomib, lenalidomide, and dexa­meth­a­sone followed by
trans­plant; KRd 12, KRd for 12 cycles; m, months; NA, not avail­­able; NR, not reached; PVd, pomalidomide, bortezomib, and dexa­meth­a­sone; Rd, lenalidomide and dexa­meth­a­sone; RVd,
lenalidomide, bortezomib, and dexa­meth­a­sone; Sd, selinexor and dexa­meth­a­sone; SVd, selinexor, bortezomib, and dexa­meth­a­sone; VTd, bortezomib, tha­lid­om ­ ide, and dexa­meth­a­sone.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/30/1851766/30mateos.pdf by guest on 13 December 2021


Management of high-risk patients with CAR-T cell ther­apy References
BCMA is an attrac­tive and exten­sively stud­ied tar­get for immu­no­ 1. Palumbo A, Bringhen S, Mateos M-V, et al. Geriatric assess­ment pre­dicts sur­
vival and toxicities in elderly mye­loma patients: an International Myeloma
ther­apy in MM. BCMA-targeting CAR-T cells have dem­on­strated
Working Group report. Blood. 2015;125(13):2068-2074.
fast, high, and deep responses in patients with RRMM. The sam­ 2. Facon T, Dimopoulos MA, Meuleman N, et  al. A sim­ pli­
fied frailty scale
ple sizes across the dif­fer­ent tri­als are so far rather small but have pre­dicts out­comes in trans­plant-inel­i­gi­ble patients with newly diag­
included a great pro­por­tion of patients with high-risk fea­tures, nosed mul­ti­ple mye­loma treated in the FIRST (MM-020) trial. Leukemia.
such as high-risk CA, R-ISS III, EMD, or high tumor bur­den.32,33 Ide- 2020;34(1):224-233.
3. Usmani SZ, Heuck C, Mitchell A, et  al. Extramedullary dis­ease por­tends
cel (idecabtagene vicleucel), indeed, already has been approved
poor prog­no­sis in mul­ti­ple mye­loma and is over-represented in high-risk
by US Food and Drug Administration for RRMM after at least 4 dis­ease even in the era of novel agents. Haematologica. 2012;97(11):1761–
rounds of ther­apy, includ­ing PIs, immu­no­mod­u­la­tory drugs, and 1767.
anti-CD38, and has been eval­u­ated in 128 patients with RRMM 4. Gonsalves WI, Rajkumar SV, Go RS, et  al. Trends in sur­vival of patients
after a median of 6 prior lines (84% tri­ple refrac­tory). The ORR with pri­mary plasma cell leu­ke­mia: a pop­u­la­tion-based anal­y­sis. Blood.
2014;124(6):907-912.
(overall response rate) was 73%, includ­ing a com­plete remis­ 5. Sonneveld P, Avet-Loiseau H, Lonial S, et al. Treatment of mul­ti­ple mye­loma
sion rate of 33% and a median PFS of 8.8 months. These effi­cacy

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/30/1851766/30mateos.pdf by guest on 13 December 2021


with high-risk cyto­ ge­net­
ics: a con­sen­sus of the International Myeloma
data were sustained in patients with high-risk fea­tures, such as Working Group. Blood. 2016;127(24):2955-2962.
EMD (n  =  50), high-risk CA (n  =  45), or high tumor bur­den (n  =  65)34 6. Schmidt TM, Barwick BG, Joseph N, et  al. Gain of chro­ mo­
some 1q is
asso­ci­ated with early pro­gres­sion in mul­ti­ple mye­loma patients treated
(Table 2). Many other BCMA-targeted CAR-T cells are under
with lenalidomide, bortezomib, and dexa­ meth­a­
sone. Blood Cancer J.
inves­ti­ga­tion, and some clin­i­cal tri­als are focused in patients with 2019;9(12):94.
high-risk fea­tures (Table 3). If results are pos­i­tive, CAR-T cell ther­ 7. Corre J, Munshi NC, Avet-Loiseau H. Risk fac­tors in mul­ti­ple mye­loma: is it
apy will rap­idly move as the first choice in patients with high-risk time for a revi­sion? Blood. 2021;137(1):16-19.
fea­tures. 8. Corre J, Perrot A, Caillot D, et al. del(17p) with­out TP53 muta­tion con­fers a
poor prog­no­sis in inten­sively treated newly diag­nosed patients with mul­
Beyond BCMA-targeted CAR-T cells, there are other ther­ ti­ple mye­loma. Blood. 2021;137(9):1192-1195.
a­peu­tic options, such as bispecific moABs targeting not only 9. Palumbo A, Avet-Loiseau H, Oliva S, et al. Revised inter­na­tional stag­ing sys­
BCMA but also GPRC5D, FcRH5, and oth­ers, under eval­u­a­tion in tem for mul­ti­ple mye­loma: a report from International Myeloma Working
patients with RRMM, and their effi­cacy will be also eval­u­ated in Group. J Clin Oncol. 2015;33(26):2863-2869.
10. Burgos L, Puig N, Cedena M-T, et al. Measurable resid­ual dis­ease in mul­ti­ple
patients with high-risk fea­tures.
mye­loma: ready for clin­i­cal prac­tice? J Hematol Oncol. 2020;13(1):82.
In sum­mary, in 2021, the iden­ti­fi­ca­tion of high-risk patients 11. Munshi NC, Avet-Loiseau H, Anderson KC, et  al. A large meta-anal­ y­sis
with MM con­tin­ues being an unmet med­i­cal need, and the def­ establishes the role of MRD neg­a­tiv­ity in long-term sur­vival out­comes in
i­ni­tion should be revisited. Genomics will help us to improve patients with mul­ti­ple mye­loma. Blood Adv. 2020;4(23):5988-5999.
the iden­ti­fi­ca­tion of these patients, as well as the ther­a­peu­tic 12. Paiva B, Puig N, Cedena M-T, et al; GEM (Grupo Español de Mieloma)/
PETHEMA (Programa Para el Estudio de la Terapéutica en Hemopatías
advances, to find the best option for them. Considering the
Malignas) Cooperative Study Group. Measurable resid­ual dis­ease by next-
achieve­ ment of unde­ tect­
able and sustained MRD can abro­ gen­er­a­tion flow cytom­e­try in mul­ti­ple mye­loma. J Clin Oncol. 2020;38(8):
gate the poor prog­no­sis of high-risk fea­tures, the man­age­ment ­784-792.
of these patients should be response adapted and deter­mined 13. Zamagni E, Nanni C, Dozza L, et  al. Standardization of 18F-FDG-PET/CT
from expo­si­tion to sequen­tial treat­ments based on drugs with a according to Deauville cri­te­ria for met­a­bolic com­plete response def­i­ni­tion
in newly diag­nosed mul­ti­ple mye­loma. J Clin Oncol. 2021;39(2):116-125.
new and dif­fer­ent mech­a­nism of action. International effort and 14. Stege CAM, Nasserinejad K, van der Spek E, et al. Efficacy and tol­er­a­bil­ity
research in this regard are required. of ixazomib, daratumumab and low dose dexa­meth­a­sone (Ixa Dara dex) in
unfit and frail newly diag­nosed mul­ti­ple mye­loma (NDMM) patients; results
Conflict-of-inter­est dis­clo­sure of the interim effi­cacy anal­y­sis of the phase II HOVON 143 study. Blood.
María-Victoria Mateos: has received hon­o­raria derived from lec­ 2019;134(suppl 1):695-695.
15. Facon T, Niesvizky R, Mateos M-V, et  al. Efficacy and safety of carfilzo-
tures and advi­sory boards from Janssen, BMS-Celgene, Amgen, mib-based reg­ i­
mens in frail patients with relapsed and/or refrac­ tory
Takeda, Abbvie, Sanofi, Oncopeptides, Adaptive, Roche, Pfizer, ­mul­ti­ple mye­loma. Blood Adv. 2020;4(21):5449-5459.
Regeneron, GSK, Bluebird Bio, and Sea-Gen. 16. Schjesvold FH, Richardson PG, Attal M, et  al. Efficacy of isatuximab with
Borja Puertas Martínez: no com­pet­ing finan­cial inter­ests to pomalidomide and dexa­meth­a­sone in elderly patients with relapsed/refrac­
tory mul­ti­ple mye­loma: ICARIA-MM sub­group anal­y­sis. Blood. 2019;134
declare.
(suppl 1):1893-1893.
Verónica González-Calle: has received hon­o­raria from Janssen 17. Kumar SK, Jaco­bus SJ, Cohen AD, et al. Carfilzomib or bortezomib in com­
and Celgene, received research funding from Janssen, and pro­ bi­na­tion with lenalidomide and dexa­meth­a­sone for patients with newly
vided con­sul­ting or an advi­sory role for Prothena and Janssen. diag­nosed mul­ti­ple mye­loma with­out inten­tion for imme­di­ate autol­o­gous
stem-cell trans­plan­ta­tion (ENDURANCE): a multicentre, open-label, phase
Off-label drug use 3, randomised, con­trolled trial. Lancet Oncol. 2020;21(10):1317-1330.
18. Usmani SZ, Hoering A, Ailawadhi S, et al; SWOG1211 Trial Investigators. Bor-
María-Victoria Mateos: nothing to disclose.
tezomib, lenalidomide, and dexa­meth­a­sone with or with­out elotuzumab in
Borja Puertas Martínez: nothing to disclose. patients with untreated, high-risk mul­ti­ple mye­loma (SWOG-1211): pri­mary
Verónica González-Calle: nothing to disclose. anal­y­sis of a randomised, phase 2 trial. Lancet Haematol. 2021;8(1):e45–
e54.
Correspondence 19. Giri S, Grimshaw A, Bal S, et al. Evaluation of daratumumab for the treat­
María-Victoria Mateos, Hospital Universitario de Salamanca, Insti- ment of mul­ti­ple mye­loma in patients with high-risk cyto­ge­netic fac­tors: a
tuto de Investigación Biomédica de Salamanca (IBSAL), Centro sys­tem­atic review and meta-anal­y­sis. JAMA Oncol. 2020;6(11):1759-1765.
20. Cavo M, Gay F, Beksac M, et  al. Autologous haematopoietic stem-cell
de Investigación del Cancer (IBMCC-USAL, CSIC), CIBER-ONC trans­plan­ta­tion ver­sus bortezomib-mel­pha­lan-pred­ni­sone, with or with­
num­ber CB16/12/00233, Paseo San Vicente 58-182, 37007 Sala- out bortezomib-lenalidomide-dexa­meth­a­sone con­sol­i­da­tion ther­apy, and
manca, Spain; e-mail: mvmateos@usal​­.es. lenalidomide main­te­nance for newly diag­nosed mul­ti­ple mye­loma

Prakash Singh Shekhawat


Management of high-risk mye­loma  | 35
(EMN02/​HO95): a multicentre, randomised, open-label, phase 3 study. 29. Dimopoulos MA, San-Miguel J, Belch A, et  al. Daratumumab plus lenalid-
Lancet Haematol. 2020;7(6):e456-e468. omide and dexa­meth­a­sone ver­sus lenalidomide and dexa­meth­a­sone in
21. Hari P, Pasquini MC, Stadtmauer EA, et  al. Long-term fol­low-up of BMT relapsed or refrac­ tory mul­
ti­
ple mye­ loma: updated anal­y­sis of POLLUX.
CTN 0702 (STaMINA) of postautologous hema­to­poi­etic cell trans­plan­ta­tion Haematologica. 2018;103(12):2088-2096.
(autoHCT) strat­eg ­ ies in the upfront treat­ment of mul­ti­ple mye­loma (MM). 30. Spencer A, Lentzsch S, Weisel K, et al. Daratumumab plus bortezomib and
J Clin Oncol. 2020;38(15, suppl):8506. dexa­meth­a­sone ver­sus bortezomib and dexa­meth­a­sone in relapsed or
22. Voorhees PM, Kaufman JL, Laubach J, et al. Daratumumab, lenalidomide, refrac­tory mul­ti­ple mye­loma: updated anal­y­sis of CASTOR. Haematolog-
bortezomib, and dexa­meth­a­sone for trans­plant-eli­gi­ble newly diag­nosed ica. 2018;103(12):2079-2087.
mul­ti­ple mye­loma: the GRIFFIN trial. Blood. 2020;136(8):936-945. 31. Weisel K, Geils GF, Karlin L, et al. Carfilzomib, dexa­meth­a­sone, and dara-
23. Gay F, Musto P, Rota Scalabrini D, et al. Survival anal­y­sis of newly diag­nosed tumumab ver­sus carfilzomib and dexa­meth­a­sone in relapsed or refrac­
trans­plant-eli­gi­ble mul­ti­ple mye­loma patients in the ran­dom­ized Forte trial. tory mul­ti­ple mye­loma: sub­group anal­y­sis of the phase 3 can­dor study in
Blood. 2020;136(suppl 1):35-37. patients with early or late relapse. Blood. 2020;136(suppl 1):37-38.
24. Facon T, Kumar S, Plesner T, et al. MAIA Trial Investigators. Daratumumab 32. Munshi NC, Anderson LD Jr, Shah N, et  al. Idecabtagene vicleucel in
plus lenalidomide and dexa­ meth­ as­one for untreated mye­ loma. N Engl relapsed and refrac­tory mul­ti­ple mye­loma. N Engl J Med. 2021;384(8):705–
J Med. 2019;380(22):2104-2115. 716.
25. Richardson PG, Oriol A, Larocca A, et al. HORIZON (OP-106) Investigators. 33. Madduri D, Berdeja JG, Usmani SZ, et al. CARTITUDE-1: phase 1b/2 study of
Melflufen and dexa­meth­a­sone in heavily pretreated relapsed and refrac­ ciltacabtagene autoleucel, a B-cell mat­u­ra­tion anti­gen-directed chi­me­ric

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/30/1851766/30mateos.pdf by guest on 13 December 2021


tory mul­ti­ple mye­loma. J Clin Oncol. 2021;39(7):757-767. anti­gen recep­tor T cell ther­apy, in relapsed/refrac­tory mul­ti­ple mye­loma.
26. Chari A, Vogl DT, Gavriatopoulou M, et al. Oral selinexor-dexa­meth­a­sone Blood. 2020;136(suppl 1):22-25.
for tri­ple-class refrac­tory mul­ti­ple mye­loma. N Engl J Med. 2019;381(8):727– 34. Raje NS, Siegel DS, Jagannath S, et  al. Idecabtagene vicleucel (ide-cel,
738. bb2121) in relapsed and refrac­tory mul­ti­ple mye­loma: ana­ly­ses of high-risk
27. Cohen AD, Trudel S, Lonial S, et al. DREAMM-2: sin­gle-agent belantamab sub­groups in the KarMMa study. Blood. 2020;136(suppl 1):37-38.
mafodotin (GSK2857916) in patients with relapsed/refrac­ tory mul­ti­
ple
mye­loma (RRMM) and high-risk (HR) cyto­ge­net­ics. J Clin Oncol. 2020;38(15,
suppl):8541.
28. Mateos M-V, Goldschmidt H, San-Miguel J, et al. Carfilzomib in relapsed or
refrac­tory mul­ti­ple mye­loma patients with early or late relapse fol­low­ing
prior ther­apy: a sub­group anal­y­sis of the ran­dom­ized phase 3 ASPIRE and © 2021 by The Amer­i­can Society of Hematology
ENDEAVOR tri­als. Hematol Oncol. 2018;36(2):463-470. DOI 10.1182/hema­tol­ogy.2021000229

36  |  Hematology 2021  |  ASH Education Program


CHALLENGES IN MULTIPLE MYELOMA TREATMENT

Minimal residual disease in multiple


myeloma: why, when, where

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/37/1851756/37yee.pdf by guest on 13 December 2021


Andrew J. Yee1,2 and Noopur Raje1,2
Center for Multiple Myeloma, Massachusetts General Hospital Cancer Center, Boston, MA; and 2Harvard Medical School, Boston, MA
1

Improvements in multiple myeloma therapy have led to deeper responses that are beyond the limit of detection by his-
torical immunohistochemistry and conventional flow cytometry in bone marrow samples. In parallel, more sensitive tech-
niques for assessing minimal residual disease (MRD) through next-generation flow cytometry and sequencing have been
developed and are now routinely available. Deep responses when measured by these assays correspond with improved
outcomes and survival. We review the data supporting MRD testing as well as its limitations and how it may fit in with
current and future clinical practice.

LEARNING OBJECTIVES
• Understand the role of MRD status in prognosis in multiple myeloma
• Explore how to apply MRD testing in clinical practice and its limitations

(daratumumab and isatuximab). With the increasing use of


CLINICAL CASE these agents, especially in 3- and 4-drug combinations,
A 57-year-old woman with multiple myeloma (MM) has responses have substantially deepened in newly diagnosed
completed initial therapy with lenalidomide, bortezomib, patients in whom complete responses (CRs) are routinely
and dexamethasone, followed by high-dose melphalan and achieved, for example, from historically 10% with thalido-
autologous stem cell transplant (auto-SCT). She initially pre- mide and dexamethasone1 to 95% in a recently reported
sented with anemia with hemoglobin of 6.1g/dL, and the combination of daratumumab, carfilzomib, lenalidomide,
disease was staged as Revised International Staging System and dexamethasone (dara-KRd) without the use of high-
stage II. Fluorescence in situ hybridization (FISH) showed dose melphalan.2 Similar trends are also seen in relapsed
gain of 1q. She is on maintenance therapy with lenalidomide disease, especially with the advent of anti-B-cell maturation
and ixazomib. Laboratory studies showed no monoclonal antigen (BCMA) directed therapies. To better assess these
protein, and the serum free light chain ratio was normal. She improving responses, MRD testing using more sensitive
mentions that she went to a patient education session and tools has emerged. This review provides an overview of
heard about “minimal residual disease (MRD) testing.” She is MRD assessment in MM and highlights the practical aspects
interested in having MRD testing performed. of MRD testing.

Importance of depth of response


Introduction Intuitively, the depth of response with myeloma therapy cor-
Response assessment in MM has traditionally relied on relates with long-term outcomes. The relationship between
measuring the monoclonal protein in the serum and urine CR and progression-free survival (PFS) and overall survival
and plasma cell involvement in the bone marrow and, (OS) has been consistently demonstrated in a meta-analysis
more recently, serum free light chains. The past 2 decades of trials using intensive therapy combined with older3 or
have seen tremendous progress in the treatment of MM contemporary therapies.4 Moreover, an analysis of 344
with the approval and adoption of effective agents such patients treated by the Grupo Español de Mieloma (GEM)
as proteasome inhibitors (bortezomib, carfilzomib, ixazo- and Programa Para el Estudio de la Terapéutica en Hemo-
mib), immunomodulatory drugs (lenalidomide, pomalido- patías Malignas (PETHEMA) groups noted differences in sur-
mide), and, more recently, anti-CD38 monoclonal antibodies vival between CR, near CR, and very good partial response.5
Prakash Singh Shekhawat
Minimal residual disease in multiple myeloma | 37
Table 1. Comparison of MRD test­ing with next-gen­er­a­tion flow cytom­e­try and next-gen­er­a­tion sequenc­ing

Characteristic Next-gen­er­a­tion flow cytom­e­try Next-gen­er­a­tion sequenc­ing


Requires base­line sam­ple No Yes, in order to iden­tify clone to track
Sensitivity <10 −5
<10−6
Sample require­ments >5 mil­lion cells (more cells to increase sen­si­tiv­ity to 10−6) <2 mil­lion cells
Sample processing Requires fresh sam­ple For Clonoseq track­ing: fresh or fro­zen sam­ple
Assessment for hemo­di­lu­tion Yes Not pos­si­ble
Turnaround time 2-4 days 7 days
Commercial avail­abil­ity Reference or expe­ri­enced in-house lab­o­ra­tory Clonoseq (Adaptive); LymphoTrack for in-house use
Table adapted from Kumar et al. 9

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/37/1851756/37yee.pdf by guest on 13 December 2021


Similar obser­va­tions with CR hold true in older patients who are patients man­aged with inten­sive ther­apy and where MRD was
not eli­gi­ble for high-dose ther­apy.6 This raises the ques­tion of mostly assessed by older, less sen­si­tive, flow cytom­et­ry (10−4),
whether fur­ther gains may be seen with even deeper responses the recent meta-anal­ y­
sis extends on prior obser­ va­tions to
such as MRD-neg­a­tive dis­ease. It should be noted that this rela­ include older, trans­ plant-inel­

gi­
ble patients and patients with
tion­ship between response and out­comes gen­er­ally holds true, relapsed dis­ease.23 Compared with MRD-pos­i­tive dis­ease, MRD-
pro­vided that the method of achiev­ing this depth of response neg­a­tive sta­tus showed improved PFS (haz­ard ratio, 0.33; 95% CI,
is tol­er­ated well. However, the Eastern Cooperative Oncology 0.29-0.37) and OS (haz­ard ratio, 0.45; 95% CI, 0.39-0.51) across
Group study of high-dose dexa­meth­a­sone with lenalidomide7 and mul­ti­ple patient pop­u­la­tions, includ­ing in relapsed dis­ease and
the BELLINI trial of venetoclax with bortezomib and dexa­meth­a­ high-risk dis­ease.23 Importantly, MRD sta­tus can strat­ify patients
sone8 are instruc­tive for illus­trat­ing that deeper responses are not in CR, where OS was 112 vs 82 months for MRD-negative vs
always asso­ci­ated with improve­ments in sur­vival. MRD-positive patients, respec­tively.22
The term min­i­mal resid­ual dis­ease con­ven­tion­ally refers to dis­ Given these find­ings, MRD sta­tus is increas­ingly used as an
ease in the bone mar­row space. Measurement of min­i­mal dis­ease in end point when com­par­ing dif­fer­ent reg­i­mens, espe­cially now
the bone mar­row is rel­e­vant as it com­monly serves as the res­er­voir that reg­im ­ ens are increas­ingly achiev­ing deeper responses. The
of dis­ease relapse. Methods cur­rently used to detect MRD include International Myeloma Working Group9 and Bone Marrow Trans-
mul­ti­pa­ram­e­ter, next-gen­er­a­tion flow cytom­e­try (NGF) and next- plant Clinical Trials Network (BMT CTN)24 have pro­vided guid­ance
gen­er­a­tion sequenc­ing (NGS) (Table 1). More than 10 years ago, around def­i­ni­tions of MRD and per­for­mance (Table 2), with the
flow cytom­e­try was the first tech­nique to eval­u­ate MRD, in which IMWG recommending a sen­si­tiv­ity of 10−5. The use of MRD as a
sen­si­tiv­ity was 10−4 ini­tially.10,11 The sen­ si­
tiv­
ity has improved to sur­ro­gate end point for reg­u­la­tory pur­poses is an area of active
2 × 10−6, and the EuroFlow con­sor­tium has stan­dard­ized the meth­ dis­cus­sion25 and is being addressed by a con­sor­tium of aca­demic
od­ol­ogy.12 NGS has also emerged in par­al­lel for mea­sur­ing MRD, in groups and phar­ma­ceu­ti­cal part­ners, the International Indepen-
which immu­no­glob­u­lin gene seg­ments are ampli­fied using con­ dent Team for Endpoint Approval of Myeloma MRD.26-28
sen­sus prim­ers and sequenced.13 Currently, the sen­si­tiv­ity of the The depth of MRD neg­a­tive sta­tus is also impor­tant. This was
Adaptive Clonoseq plat­form (pre­vi­ously known as LymphoSIGHT) ini­tially shown with flow cytom­et­ ry with sen­si­tiv­ity down to 10−4
is 6.77 × 10−7 with 20 µg DNA from 1 mL of bone mar­row aspi­rate.14,15 and where each log reduc­tion in MRD trans­lated into improve­
The con­cor­dance between NGF and NGS is high. It exceeded ment in median OS.29 In the Francophone du Myélome 2009
80% when exam­ined in the FORTE16 and CASSIOPEIA17 tri­als in study of upfront vs deferred auto-SCT after ini­tial ther­apy with
newly diag­nosed patients. There was sim­i­larly high con­cor­dance, lenalidomide, bortezomib, and dexa­meth­a­sone, MRD sta­tus was
85.8%18 and 92.9%,19 when com­par­ing NGF with NGS from a dif­ assessed by flow cytom­et­ ry in all­patients, and a sub­set of these
fer­ent plat­form, LymphoTrack. The choice of assay used for MRD patients was eval­ua ­ ted by more sen­si­tive NGS.30,31 Patients who
is based on avail­abil­ity and insti­tu­tional pref­er­ence. NGS by the were a ­ ble to achieve MRD neg­a­tive sta­tus at 10−6 by NGS, which
Clonoseq assay is com­mer­cially avail­­able through Adaptive, and is deeper than the recommended IMWG thresh­old of 10−5, had
in Jan­ua ­ ry 2019, Medi­care announced cov­er­age of this test. NGF supe­rior out­comes in PFS and OS com­pared with MRD-pos­i­tive
is also com­mer­cially avail­­able, for exam­ple, through Mayo Clinic sta­tus (Figure 1).31 Moreover, the study showed dif­fer­ences in
ref­er­ence lab­o­ra­tory. A con­sid­er­ation with NGS is that it requires out­comes between 10−6, 10−5, and 10−4. Prior to starting main­
a base­line sam­ple to pro­vide a trackable sequence; NGF does not te­nance ther­apy, patients who were MRD neg­a­tive had sim­i­lar
require a base­line sam­ple. In 1 series, a trackable sequence for PFS whether they received trans­plant upfront or not, although
NGS could not be iden­ti­fied in 7.8% of sam­ples.20 NGF also has the patients in the trans­plant arm were more likely to be MRD neg­a­
advan­tage of assessing for hemo­di­lu­tion by looking for mast cell, tive (29.8% vs 20.5%). Of note is that in the IFM 2009 study, MRD
eryth­ro­blast, and B-cell pre­cur­sor pop­u­la­tions.12 Finally, from a assess­ments were after com­ple­tion of ini­tial ther­apy, prior to
research per­spec­tive, NGF may be a ­ ble to eval­u­ate the bone mar­ main­te­nance ther­apy; the effect of high-dose ther­apy in patients
row micro­en­vi­ron­ment, which may have prog­nos­tic rel­e­vance. who were already MRD neg­at­ ive prior to high-dose ther­apy was
Several meta-ana­ly­ses have con­sis­tently shown that depth not addressed. Nevertheless, as long as a deep, MRD-neg­at­ive
of response beyond CR cor­re­lates with improve­ment in OS.21-23 response is achieved, the method of achiev­ing the response may
Although the ini­tial meta-ana­ly­ses focused on trans­plant-eli­gi­ble not be as impor­tant. For exam­ple, the CASSIOPEIA study eval­u­ated

38  |  Hematology 2021  |  ASH Education Program


Table 2. IMWG MRD cri­te­ria

Response Criteria
Sustained MRD neg­a­tive MRD neg­a­tiv­ity in the mar­row (NGF or NGS, or both) and by imag­ing (PET CT), con­firmed min­i­mum of 1 year apart.
Subsequent eval­u­a­tions can be used to fur­ther spec­ify the dura­tion of neg­a­tiv­ity (eg, MRD neg­a­tive at 5 years).
Flow MRD neg­a­tive Absence of phe­no­typ­i­cally aber­rant clonal plasma cells by NGF on bone mar­row aspi­rates using the EuroFlow stan­
dard oper­a­tion pro­ce­dure for MRD detec­tion in mul­ti­ple mye­loma (or val­i­dated equiv­a­lent method) with a min­i­mum
sen­si­tiv­ity of 1 in 105 nucle­ated cells or higher.
Sequencing MRD neg­a­tive Absence of clonal plasma cells by NGS on bone mar­row aspi­rate in which the pres­ence of a clone is defined as fewer
than 2 iden­ti­cal sequenc­ing reads obtained after DNA sequenc­ing of bone mar­row aspi­rates using the LymphoSIGHT
plat­form (or val­i­dated equiv­a­lent method) with a min­i­mum sen­si­tiv­ity of 1 in 105 nucle­ated cells or higher.
Imaging plus MRD neg­a­tive MRD neg­a­tiv­ity as defined by NGF or NGS plus dis­ap­pear­ance of every area of increased tracer uptake found at base­
line or a pre­ced­ing PET CT or decrease to less than medi­as­ti­nal blood pool SUV or decrease to less than that of
surrounding nor­mal tis­sue.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/37/1851756/37yee.pdf by guest on 13 December 2021


Table adapted from Kumar et al.9
SUV, standard uptake value.

daratumumab, bortezomib, tha­lid­o­mide, and dexa­meth­a­sone espe­cially impor­tant in high-risk dis­ease defined by gene expres­
(dara-VTd) vs VTd in newly diag­nosed patients under­go­ing high- sion pro­fil­ing.37
dose mel­pha­lan and auto-SCT. Patients who achieved both CR It is well established with tra­di­tional response cri­te­ria that
and MRD-neg­a­tive sta­tus had sim­i­lar PFS, irrespective of treat­ dura­bil­ity of response is a pow­er­ful prog­nos­tic fac­tor38,39 and that
ment arm (although higher-qual­ity responses were more com­ loss of CR is asso­ci­ated with infe­rior sur­vival.40 Durability of MRD-
mon in the dara-VTd arm).32 Similar find­ings were observed in the neg­a­tive sta­tus is sim­i­larly impor­tant. This was dem­on­strated
FORTE study, in which out­comes of patients with MRD-neg­a­tive recently in the POLLUX and CASTOR stud­ies, which eval­u­ated
dis­ease sustained for 1 year were sim­i­lar, irrespective of the ini­tial daratumumab with lenalidomide and dexa­meth­a­sone (dara-Rd)
treat­ment KRd vs 12 cycles of KRd with­out auto-SCT vs carfilzo- or daratumumab with bortezomib and dexa­meth­a­sone, respec­
mib, cyclo­phos­pha­mide, and dexa­meth­a­sone with auto-SCT.33 tively, using NGS at 10−5 sen­si­tiv­ity.41 Patients with sustained MRD
In patients with high-risk dis­ease, achiev­ing an MRD-neg­a­tive neg­a­tiv­ity over 12 months had the best out­comes, irrespective of
response may be even more impor­tant. An anal­y­sis of the PETHEMA/ the treat­ment arm, although this was more likely to be achieved
GEM2012MENOS65 trial showed that MRD-neg­ a­
tive responses in the daratumumab-containing com­bi­na­tion. Similar find­ings of
were ­able to over­come poor prog­nos­tic fea­tures at diag­no­sis, improved out­comes were seen with sustained MRD neg­a­tiv­ity
includ­ing Revised International Staging System stage III.34,35 Similar over 6 or 12 months in newly diag­nosed, trans­plant-inel­i­gi­ble
obser­va­tions were seen for patients with high-risk cyto­ge­net­ics patients in the ALCYONE (daratumumab, bortezomib, mel­pha­
in IFM 2009 and ear­lier PETHEMA/GEM tri­als.31,36 The find­ings with lan, and pred­ni­sone [dara-VMP] vs VMP) and MAIA (dara-Rd vs
MRD extend on pre­vi­ous obser­va­tions where achiev­ing CR was Rd) tri­als.42 Reflecting these obser­va­tions, the IMWG defi­nes

Figure 1. Progression-free survival according to MRD level at the start of maintenance in IFM 2009. Progression-free survival
improves with each log reduction in MRD in IFM 2009 in patients who achieved at least a very good partial response. Figure adapted
from Perrot et al.31 Prakash Singh Shekhawat
Minimal resid­ual dis­ease in mul­ti­ple mye­loma  |  39
a sep­a­rate response cat­e­gory of “sustained MRD neg­a­tive,” in other patients with MRD-neg­a­tive dis­ease.55,56 The chal­lenge at
which assess­ments by mar­row and by imag­ing are con­firmed at this time is how to pro­spec­tively iden­tify these patients in whom
least 1 year apart.9 If 1 year is bet­ter, 2 years may be even bet­ter: an MRD-neg­a­tive response is not as crit­i­cal.
this was dem­on­strated in patients with sustained MRD neg­a­tiv­ity Perhaps the most obvi­ous lim­i­ta­tion for MRD assess­ment is
(by NGF at 10−5) for 2 years in a trial of patients on lenalidomide the require­ment for a bone mar­row aspi­ra­tion pro­ce­dure. This
main­te­nance.43 Moreover, in this study, loss of MRD neg­at­iv­ity has moti­vated inves­ti­gat­ing “liq­uid biop­sies,” using the same
was actu­ally worse than sustained MRD pos­i­tiv­ity. tools on the periph­ eral blood. Indeed, anal­ y­
sis of periph­eral
There have been sev­eral stud­ies exam­in­ing the pat­terns of blood pro­vi­des a sys­temic assess­ment and avoids the pit­falls
loss of MRD neg­a­tiv­ity and its clin­i­cal rel­e­vance.44-46 For exam­ of het­ero­ge­ne­ity in bone mar­row sam­pling. Methods involv­ing
ple, MRD pro­gres­sion by flow cytom­e­try with sen­si­tiv­ity at 10−4 the periph­eral blood may allow for detecting and mon­i­tor­ing
or by allele-spe­cific oli­go­nu­cle­o­tide poly­mer­ase chain reac­tion extramedullary dis­ease that is missed by focus­ing on the bone
with sen­si­tiv­ity of 10−5 in a series of patients on lenalidomide mar­row. Using the same NGF meth­od­ol­ogy opti­mized in bone
main­te­nance antic­i­pated bio­chem­i­cal relapse by 4 months and mar­row on periph­eral blood, the sen­si­tiv­ity is less.57 Forty per­
clin­i­cal relapse by 9 to 10 months.45 Similarly, in a ret­ro­spec­tive cent of patients with bone mar­row that was MRD pos­i­tive were

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/37/1851756/37yee.pdf by guest on 13 December 2021


study using NGS (10−6), molec­u­lar relapse by MRD eval­u­a­tion was neg­a­tive in the periph­eral blood; all­ patients with cir­cu­lat­ing
­able to pre­dict clin­i­cal relapse.46 Serial MRD test­ing was a ­ ble to plasma cells were MRD pos­i­tive in the bone mar­row. Similarly,
pre­dict clin­i­cal relapse in 9 of 10 cases, and relapse by IMWG NGS has been explored on periph­eral blood.58 Of patients with
cri­te­ria occurred at a median of 13 months (range, 1-28 months) pos­i­tive bone mar­row MRD tests, the test was neg­a­tive in plasma
fol­low­ing molec­u­lar relapse. These find­ings raise the ques­tion 69% of the time. This may reflect the lower cir­cu­lat­ing DNA bur­
of whether ini­ti­at­ing treat­ment at the time of molec­u­lar relapse den in periph­eral blood. Other approaches under devel­op­ment
rather than waiting for bio­chem­i­cal or clin­ic ­ al relapse could alter include anal­y­sis of cir­cu­lat­ing free tumor DNA using targeted
the nat­u­ral his­tory of the dis­ease (see Relapse from MRD Nega- muta­tion detec­tion59 or whole-genome low-pass sequenc­ing.60
tivity as Indication for Treatment study below). Mass spec­trom­e­try is now being used to mea­sure mono­clo­
nal gammopathy in the periph­eral blood. There are 2 forms of
Limitations of MRD assess­ment mass spec­trom­e­try: matrix-assisted laser desorp­tion ion­i­za­tion
An inher­ent lim­i­ta­tion in MRD assess­ment is its reli­ance on mea­ time-of-flight mass spec­ trom­ et­ry (MALDI-TOF-MS) and liq­ uid
sur­ing dis­ease in the bone mar­row. This assess­ment focuses on chro­ma­tog­ra­phy quad­ru­pole time-of-flight mass spec­trom­e­try
plasma cells and does not take into account the bone mar­row (LC-MS).61 MALDI-TOF-MS has a sen­si­tiv­ity of less than 0.01 g/dL62
micro­en­vi­ron­ment,47 which may play a role in shap­ing prog­no­ and has replaced con­ven­tional serum pro­tein elec­tro­pho­re­
sis. From a prac­ti­cal per­spec­tive, bone mar­row involve­ment may sis at some insti­tu­tions. LC-MS has even more sen­si­tiv­ity than
not be uni­form, such as in the case of macrofocal dis­ease,48 and MALDI-TOF-MS, down to 0.005 g/dL, but has lower through­put.63
per­haps most impor­tant, extramedullary dis­ease may also be Moreover, mass spec­trom­et­ry can dis­tin­guish between “false-
pres­ent. For exam­ple, in the IMAgerie du JEune Myélome study pos­i­tive” bands on pro­tein elec­tro­pho­re­sis from ther­a­peu­tic
of the Intergroupe Francophone du Myélome 2009 trial, 26% of mono­clo­nal antibodies such as daratumumab vs the under­ly­ing
patients with MRD-neg­a­tive dis­ease by flow cytom­e­try (sen­si­tiv­ dis­ease.64 Given the increased sen­si­tiv­ity of mass spec­trom­e­try
ity 10−4) had pos­i­tive pos­i­tron emis­sion tomog­ra­phy (PET) com­ of the periph­eral blood, stud­ies are com­par­ing the per­for­mance
puted tomog­ra­phy (CT) find­ings.49 Similar find­ings were seen of mass spec­trom­e­try with MRD performed on bone mar­row by
in the CASSIOPET substudy of CASSIOPEIA, in which 10.5% of NGS or NGF, for exam­ple, in the Stem Cell Transplant in Myeloma
patients who were neg­a­tive by NGF at 10−5 had pos­it­ ive PET CT.50 Incorporating Novel Agents65 and in GEM2012MENOS65 tri­als.66
This dis­crep­ancy is rel­e­vant, as patients who were MRD neg­a­ In one study, LC-MS was esti­mated to be even more sen­si­tive
tive but PET CT pos­i­tive had sim­i­lar out­comes to patients who than NGS at 10−5 and could be used as a screen for MRD.67
were MRD pos­i­tive. The dis­crep­ancy between MRD neg­a­tiv­ity
and imag­ing is higher in patients with relapsed dis­ease, 50% Applying MRD to clin­i­cal prac­tice
vs 12% in newly diag­nosed patients in 1 series.51 Overall, as was The data establishing depth of response by MRD test­ ing and
seen in CASSIOPET, patients who are “dou­ble neg­a­tive” on MRD out­comes are robust, and clin­i­cal tri­als now rou­tinely incor­po­
and imag­ing tended to have the best out­comes, suggesting that rate MRD test­ing to bench­mark per­for­mance. MRD test­ing is also
these 2 modal­i­ties com­ple­ment each other. The Deauville scale being used to strat­ify patients in clin­i­cal tri­als. For exam­ple, the
used in lym­phoma has been applied to MM to stan­dard­ize “met­ ECOG Effective Quadruplet Utilization after Treatment Evaluation
a­bolic response” cri­te­ria by PET and was an inde­pen­dent pre­dic­ trial (NCT04566328) ran­dom­izes patients after ini­tial ther­apy with
tor for improved PFS and OS out­comes.52 dara-Rd to either con­sol­id ­ a­tion with addi­tional dara-Rd or add-
Moreover, inher­ent to this dis­cus­sion is the het­ero­ge­ne­ity of ing bortezomib to dara-Rd, and the study stratifies by MRD sta­
MM, in which the depth of response may not be as impor­tant in tus. However, apply­ing MRD test­ing to patient care is evolv­ing.
all­ patients. This was pre­vi­ously rec­og­nized with CR, in which Indeed, the ques­tions raised when this topic was ini­tially cov­ered
patients with a his­tory of monoclonal gammopathy of undeter- in this edu­ca­tion pro­gram 4 years ago con­tinue to be rel­e­vant
mined significance53 or with a MGUS-like gene expres­sion pro­file54 now.68 There are ongo­ing tri­als to help answer this ques­tion (Fig-
had lower CR rates with a tan­dem trans­plant reg­i­men in Total ure 2). We acknowl­edge that there is sig­nif­i­cant var­i­abil­ity in MRD
Therapy 2 but supe­rior out­comes. The cyclin D2 molec­u­lar sub­ use in clin­ic
­ al prac­tice. At this time, there are no pro­spec­tive,
type, which char­ac­ter­is­ti­cally includes patients with t(11;14), has ran­dom­ized data in which the infor­ma­tion from MRD test­ing can
the low­est and slowest cumu­la­tive inci­dence of response, yet has guide treat­ment deci­sions. Nevertheless, if a bone mar­row biopsy
com­pa­ra­ble out­comes with MRD-pos­i­tive dis­ease com­pared with is being performed to con­firm a CR, send­ing the aspi­rate for MRD

40  |  Hematology 2021  |  ASH Education Program


MRDA
Consolidation MRDB
+
Initial therapy + Next line
− Maintenance
or observation
− Observation

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/37/1851756/37yee.pdf by guest on 13 December 2021


Primary Sponsor MRD
Identifier MRD Treatment Study population
outcome modality

A. MRD testing to guide treatment after initial therapy

Randomization to arms
including R maint, RVd
consolidation + R maint,
+ Standard risk
R-isa maint, or isa-RVd
RADAR consolidation + R isa patients after auto PFS Univ. of Leeds NGF
maint SCT (10−5)

− Isa maint

Consolidation with dara-


+
RVd Following induction Univ. of
NCT04140162 MRD NGS
with dara-R Michigan
− Maint dara-R

+ Dara-R
NCT03901963 NGS
After auto-SCT MRD Janssen
AURIGA (10−5)
− R

+ Additional dara-KRd
NCT03224507 After dara-KRd and Univ. of NGS
MRD −5
MASTER auto SCT Alabama (10 )
Observation (after MRD

negative × 2)

B. MRD testing to guide treatment on maintenance therapy


+ Dara-Kd MRD negative
NCT04513639 patients
Relapse from randomized to arm
where patients are Oslo Univ. NGF
MRD Negativity PFS
− Continue observation followed by MRD Hospital (10−5)
as Indication for
every 4 months. (In
Treatment other arm, patients
are treated with
Figure 2. Examples of trials evaluating MRD to guide treatment. Treatment decisions with MRD may be broadly divided into
2 categories: (A) following initial therapy to guide treatment intensification or consolidation in patients with MRD positive disease
or (B) in patients on maintenance therapy, to guide discontinuation of treatment. Some trials are also examining “early” initiation of
therapy with the appearance of MRD-positive disease. D, dexamethasone; isa, isatuximab; K, carfilzomib; maint, maintenance; PD,

Prakash Singh Shekhawat


Minimal resid­ual dis­ease in mul­ti­ple mye­loma  |  41
dara-Kd at PD
by conventional
criteria.)
+ Not applicable MRD NGF
Patients on 3 years Memorial
NCT04221178 negative −5
(10 )
− Discontinue maint of continuous maint Sloan Kettering
at 1 year
Continue maint (off NGS
+ −6
protocol) (10 and
NCT04108624 Patients on maint Univ. of
MRD exploring
MRD2STOP therapy and CR Chicago −7
− Discontinue maint 10 ) and
PET CT
Continue previously After 2 years of
+
assigned maint therapy maintenance
NCT04071457
− Randomization to (previously NGS
DRAMMATIC OS SWOG

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/37/1851756/37yee.pdf by guest on 13 December 2021


continue vs stopping randomized to
SWOG S1803
previously assigned dara-R vs R
maint therapy following auto-SCT)
+ Continue dara-R
− If MRD negative After 24 months of European
NCT03710603 NGS
sustained for 12 maint therapy with PFS Myeloma −5
PERSEUS (10 )
months: discontinue dara-R Network
dara; continue R
+ Dara Patients after 1-2
Polish NGF
NCT03697655 − Observation prior lines of Event-free
Myeloma −5
(10 )
PREDATOR therapy who are survival
Consortium
MRD negative

Figure 2. (continued)
progressive disease; PREDATOR, Pre-emptive Daratumumab Therapy of Minimal Residual Disease Reappearance or Biochemical
Relapse in Multiple Myeloma; R, lenalidomide; RADAR, Risk-Adapted Therapy Directed According to Response; REMNANT, Relapse
from MRD Negativity as Indication for Treatment; V, bortezomib.

test­ing is appro­pri­ate, as it may pro­vide prog­nos­tic infor­ma­tion as inten­sify ther­apy above what was pre­vi­ously planned in patients
well as estab­lish a ref­er­ence point for sub­se­quent MRD test­ing that who have not achieved an opti­mal response. Because mye­loma
may con­firm sus­tain­abil­ity of response. To increase the sen­si­tiv­ity, ther­apy is con­tin­u­ous, responses may improve over time. In a ret­
the oper­at­or should pri­or­i­tize the first pull for MRD test­ing, given ro­spec­tive study of a real-world prac­tice of patients on lenalid-
hemo­di­lu­tion with sub­se­quent pulls.69 Finally, if MRD assess­ment is omide main­te­nance, 34.3% of patients who were MRD pos­i­tive
being performed, for com­plete­ness, it may be impor­tant to also (with MRD assess­ment according to local prac­tice) after induc­tion
assess for extramedullary dis­ease with imag­ing such as PET CT. treat­ment achieved MRD-neg­a­tive sta­tus dur­ing main­te­nance
ther­apy,71 suggesting that a change in ther­apy may not be oblig­a­
Timing of high-dose mel­pha­lan and SCT tory. The AURIGA study (NCT NCT03901963) is exam­in­ing the role
This has been a core ques­tion over the years for trans­plant- of adding daratumumab to lenalidomide main­te­nance to eval­u­ate
eli­gi­ble patients and con­tin­ues to be an ongo­ing area of debate. the ben­e­fit of adding addi­tional ther­apy to deepen a response.
Despite the IFM 2009 trial show­ing sig­nif­i­cant improve­ment in
PFS, the lack of improve­ment in OS30 spurs this ongo­ing debate, Can we de-esca­late treat­ment?
includ­ing with the FORTE trial.33 If out­comes of patients with Current prac­tice is to treat until pro­gres­sion with a com­bi­na­tion
MRD-neg­a­tive and espe­cially sustained MRD-neg­a­tive dis­ease of induc­tion and main­te­nance ther­apy. But can patients step off
are com­pa­ra­ble, does it mat­ter if this is achieved with­out high- this “tread­mill” of con­tin­uo
­ us ther­apy to avoid the adverse events
dose mel­pha­lan? Although the IFM and FORTE stud­ies incor­po­ and bur­den of chronic ther­apy? There are sev­eral tri­als exam­in­
rated MRD test­ing, this was after com­ple­tion of ini­tial ther­apy. ing de-esca­la­tion of ther­apy. There is an ongo­ing phase 2 study
These stud­ies did not eval­u­ate MRD find­ings before high-dose in newly diag­nosed, trans­plant eli­gi­ble patients, the Monoclonal
mel­pha­lan to inform deci­sion mak­ing. Antibody-Based Sequential Therapy for Deep Remission in Multi-
ple Myeloma study (NCT03224507).72 Patients undergo induc­tion
Should ther­apy change to deepen response? ther­apy with dara-KRd, followed by auto-SCT. Patients who are
This is another open ques­tion in the mye­loma field. Attempts at MRD neg­a­tive by NGS (10−5) after auto-SCT discontinue treat­ment,
answer­ing this ques­tion, before the avail­abil­ity of MRD assess­ whereas patients who are MRD pos­i­tive con­tinue to undergo con­
ments, include the Myeloma XI study of risk-adapted inten­si­fi­ sol­id
­ a­tion with dara-KRd for up to 2 cycles until MRD neg­a­tive.
ca­tion,70 which showed that the addi­tion of cyclo­phos­pha­mide, In the PERSEUS trial (NCT03710603), patients on main­ te­
nance
bortezomib, and dexa­meth­a­sone in patients with sub­op­ti­mal with daratumumab and lenalidomide who are MRD neg­a­tive can
responses improved PFS. However, cur­ rent prac­ tice does not discontinue daratumumab and con­tinue on lenalidomide. In the

42  |  Hematology 2021  |  ASH Education Program


DRAMMATIC trial (SWOG 1803), patients who are MRD neg­a­tive Noopur Raje has consulted for Amgen, BMS, Bluebird, GSK, Jans-
after ini­
tial ther­
apy are ran­dom­ized to con­ tinue the assigned sen, and Karyopharm; served on sci­en­tific advi­sory board for Car-
main­te­nance vs stop­ping assigned main­te­nance ther­apy. ibou and Immuneel; and received research funding from Bluebird.

Should ther­apy change if a patient becomes MRD pos­i­tive? Off-label drug use
An ongo­ing ques­tion is the opti­mal tim­ing of treating relapsed Andrew J. Yee: no off-label drug use discussed.
dis­ease. Patients who are treated at the time of bio­ chem­ i­
cal Noopur Raje: no off-label drug use discussed.
rather than clin­i­cal relapse have bet­ter out­comes, as seen in a
sub­group anal­y­sis of the ENDEAVOR trial (with the caveat that this Correspondence
study was not designed to answer this spe­cific ques­tion).73 Could Noopur Raje, Mas­sa­chu­setts General Hospital, 55 Fruit Street,
the out­comes of patients be bet­ter when treated at relapse, with Bos­ton, MA 02114; e-mail: nraje@mgh.harvard.edu.
an even lower bur­den of dis­ease, by MRD? As noted pre­vi­ously,
the appear­ance of MRD-pos­i­tive dis­ease may her­ald bio­chem­i­ References
cal or clin­i­cal relapse sev­eral months later. The REMNANT study 1. Cavo M, Zamagni E, Tosi P, et al; Bologna 2002 study. Superiority of tha­

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/37/1851756/37yee.pdf by guest on 13 December 2021


lid­o­mide and dexa­meth­a­sone over vin­cris­tine-doxorubicindexamethasone
(NCT04513639) will help answer this ques­tion.74 Patients who are
(VAD) as pri­mary ther­apy in prep­a­ra­tion for autol­o­gous trans­plan­ta­tion for
MRD neg­a­tive after induc­tion ther­apy are ran­dom­ized to start mul­ti­ple mye­loma. Blood. 2005;106(1):35-39.
treat­ment at the time of MRD relapse vs at the time of pro­gres­sive 2. Landgren O, Hultcrantz M, Diamond B, et  al. Safety and effec­tive­ness of
dis­ease according to IMWG cri­te­ria. However, a lim­i­ta­tion in treat- weekly carfilzomib, lenalidomide, dexa­ meth­a­
sone, and daratumumab
ing patients for relapse by MRD cri­te­ria is that cur­rent clin­i­cal tri­als com­bi­na­tion ther­apy for patients with newly diag­nosed mul­ti­ple mye­loma:
the MANHATTAN nonrandomized clin­i­cal trial. JAMA Oncol. 2021;7(6):862-
gen­er­ally require mea­sur­able dis­ease, and MRD pos­i­tiv­ity is not 868.
con­sid­ered mea­sur­able to be eli­gi­ble for the trial. Consequently, 3. van de Velde HJ, Liu X, Chen G, Cakana A, Deraedt W, Bayssas M. Com-
this may limit treat­ment to stan­dard-of-care options instead of plete response cor­ re­
lates with long-term sur­ vival and pro­ gres­
sion-free
the poten­tially more inno­va­tive ther­a­pies under inves­ti­ga­tion. sur­vival in high-dose ther­ apy in mul­ ti­
ple mye­ loma. Haematologica.
2007;92(10):1399-1406.
4. van de Velde H, Londhe A, Ataman O, et al. Association between com­plete
response and out­comes in trans­plant-eli­gi­ble mye­loma patients in the era
of novel agents. Eur J Haematol. 2017;98(3):269-279.
5. Martinez-Lopez J, Blade J, Mateos M-V, et al; Grupo Español de MM;
CLINICAL CASE (Con­t in­u ed) Programa para el Estudio de la Terapé utica en Hemopatía Maligna. Long-
Our patient under­went MRD test­ing using Adaptive NGS and was term prog­nos­tic sig­nif­i­cance of response in mul­ti­ple mye­loma after stem
MRD neg­a­tive. She had MRD test­ing seri­ally for 2 years and was cell trans­plan­ta­tion. Blood. 2011;118(3):529-534.
6. Gay F, Larocca A, Wijermans P, et al. Complete response cor­re­lates with
neg­a­tive on both occa­sions. However, even with the sustained long-term pro­gres­sion-free and over­all sur­vival in elderly mye­loma treated
MRD neg­a­tiv­ity, she pre­ferred to con­tinue with lenalidomide and with novel agents: anal­y­sis of 1175 patients. Blood. 2011;117(11):3025-
ixazomib main­te­nance. Testing for MRD at 5 years after auto-SCT 3031.
resulted in a pos­i­tive test, albeit at a low level of 0 to 1 × 10−6. With 7. Rajkumar SV, Jaco­bus S, Callander NS, et al; Eastern Cooperative Oncology
Group. Lenalidomide plus high-dose dexa­meth­a­sone ver­sus lenalidomide
this new find­ing, she had fur­ther workup includ­ing whole-body
plus low-dose dexa­meth­a­sone as ini­tial ther­apy for newly diag­nosed mul­
low-dose CT, which did not show any new find­ings. She has opted ti­ple mye­loma: an open-label randomised con­trolled trial. Lancet Oncol.
to con­tinue the cur­rent reg­i­men, with a ten­ta­tive plan of repeat­ 2010;11(1):29-37.
ing MRD test­ing in 6 months. This case illus­trates some of the chal­ 8. Kumar SK, Harrison SJ, Cavo M, et al. Venetoclax or pla­cebo in com­bi­na­tion
lenges with MRD test­ing, as nei­ther the repeated neg­a­tive results with bortezomib and dexa­meth­a­sone in patients with relapsed or refrac­
tory mul­ti­ple mye­loma (BELLINI): a randomised, dou­ble-blind, multicentre,
nor the new low pos­i­tive result prompted a change in treat­ment. phase 3 trial. Lancet Oncol. 2020;21(12):1630-1642.
9. Kumar S, Paiva B, Anderson KC, et al. International Myeloma Working Group
con­sen­sus cri­te­ria for response and min­i­mal resid­ual dis­ease assess­ment in
Conclusion mul­ti­ple mye­loma. Lancet Oncol. 2016;17(8):e328-e346.
10. Rawstron AC, Orfao A, Beksac M, et al; Euro­pean Myeloma Network. Report
Overall, the field is for­tu­nate that newer treat­ments are lead­ing to
of the Euro­pean Myeloma Network on multiparametric flow cytom­e­try in
unprec­e­dented depths of response that require newer meth­ods mul­ti­ple mye­loma and related dis­or­ders. Haematologica. 2008;93(3):431-
such as NGS or NGF to mea­sure their effect. What was once a test 438.
restricted to spe­cial­ized research set­tings or clin­i­cal tri­als is now 11. Paiva B, Vidriales M-B, Cerveró J, et al; GEM (Grupo Español de MM)/PETH-
read­ily avail­­able for any patient. Although there are matur­ing data EMA (Programa para el Estudio de la Terapéutica en Hemopatías Malignas)
Cooperative Study Groups. Multiparameter flow cytometric remis­sion is
on how it adds new prog­nos­tic power, there is a lag in the data the most rel­e­vant prog­nos­tic fac­tor for mul­ti­ple mye­loma patients who
for how to effec­tively use the test to make treat­ment deci­sions. undergo autol­o­gous stem cell trans­plan­ta­tion. Blood. 2008;112(10):4017-
Ongoing tri­als will pro­vide data and guid­ance on how to incor­po­ 4023.
rate MRD test­ing into clin­i­cal prac­tice to tai­lor ther­apy. Moreover, 12. Flores-Montero J, Sanoja-Flores L, Paiva B, et  al. Next gen­er­a­tion flow for
highly sen­si­tive and stan­dard­ized detec­tion of min­i­mal resid­ual dis­ease in
the incor­po­ra­tion of func­tional imag­ing and liq­uid biop­sies will
mul­ti­ple mye­loma. Leukemia. 2017;31(10):2094-2103.
pro­vide less inva­sive ways of eval­u­at­ing dis­ease bur­den. 13. Martinez-Lopez J, Lahuerta JJ, Pepin F, et  al. Prognostic value of deep
sequenc­ing method for min­i­mal resid­ual dis­ease detec­tion in mul­ti­ple
Conflict-of-inter­est dis­clo­sure mye­loma. Blood. 2014;123(20):3073-3079.
14. Ching T, Duncan ME, Newman-Eerkes T, et al. Analytical eval­u­a­tion of the
Andrew J. Yee has consulted for Adaptive, Amgen, BMS, GSK,
clonoSEQ assay for establishing mea­sur­able (min­i­mal) resid­ual dis­ease in
Janssen, Karyopharm, Oncopeptides, Sanofi, and Takeda and has acute lym­pho­blas­tic leu­ke­mia, chronic lym­pho­cytic leu­ke­mia, and mul­ti­ple
received clin­i­cal trial sup­port from Adaptive, Amgen, BMS, Jans- mye­loma. BMC Cancer. 2020;20(1):612.
sen, and Takeda. 15. Clonoseq. Clonoseq assay [tech­ni­cal infor­ma­tion]. Clonoseq; 2021.
Prakash Singh Shekhawat
Minimal resid­ual dis­ease in mul­ti­ple mye­loma  |  43
16. Oliva S, Genuardi E, Belotti A, et al. Minimal resid­ual dis­ease eval­ua ­ ­tion by 36. Lahuerta J-J, Paiva B, Vidriales M-B, et al; GEM (Grupo Español de Mieloma)/
mul­ti­pa­ram­e­ter flow cytom­e­try and next gen­er­a­tion sequenc­ing in the Forte PETHEMA (Programa para el Estudio de la Terapéutica en Hemopatías
trial for newly diag­nosed mul­ti­ple mye­loma patients. Blood. 2019;134(suppl 1): Malignas) Cooperative Study Group. Depth of response in mul­ti­ple mye­
4322. loma: a pooled anal­y­sis of three PETHEMA/GEM clin­ic ­ al tri­als. J Clin Oncol.
17. Avet-Loiseau H, Bene MC, Wuilleme S, et  al. Concordance of post- 2017;35(25):2900-2910.
con­sol­i­da­tion min­i­mal resid­ual dis­ease rates by multiparametric flow 37. Haessler J, Shaughnessy JD Jr, Zhan F, et al. Benefit of com­plete response in
cytom­e­try and next-gen­er­a­tion sequenc­ing in CASSIOPEIA. Clin Lym- mul­ti­ple mye­loma lim­ited to high-risk sub­group iden­ti­fied by gene expres­
phoma Myeloma Leuk. 2019;19(10):e3-e4. sion pro­fil­ing. Clin Cancer Res. 2007;13(23):7073-7079.
18. Medina A, Puig N, Flores-Montero J, et al. Comparison of next-gen­er­a­tion 38. Barlogie B, Anaissie E, Haessler J, et  al. Complete remis­ sion sustained
sequenc­ing (NGS) and next-gen­er­a­tion flow (NGF) for min­i­mal resid­ual 3 years from treat­ment ini­ti­a­tion is a pow­er­ful sur­ro­gate for extended sur­
dis­ease (MRD) assess­ment in mul­ti­ple mye­loma. Blood Cancer J. 2020;10(10): vival in mul­ti­ple mye­loma. Cancer. 2008;113(2):355-359.
108. 39. Sidana S, Tandon N, Dispenzieri A, et al. Relapse after com­plete response in
19. Ho C, Syed M, Roshal M, et al. Routine eval­u­at­ ion of min­i­mal resid­ual dis­ newly diag­nosed mul­ti­ple mye­loma: impli­ca­tions of dura­tion of response
ease in mye­ loma using next-gen­ er­

tion sequenc­ ing clonality test­ ing: and pat­terns of relapse. Leukemia. 2019;33(3):730-738.
fea­si­bil­ity, chal­lenges, and direct com­par­i­son with high-sen­si­tiv­ity flow 40. Hoering A, Crowley J, Shaughnessy JD Jr, et al. Complete remis­sion in mul­ti­
cytom­e­try. J Mol Diagn. 2021;23(2):181-199. ple mye­loma exam­ined as time-depen­dent var­ia ­ ble in terms of both onset
20. Hussaini MO, Srivastava J, Lee LW, et al. Moffitt can­cer cen­ter 2-year sin­gle- and dura­tion in total ther­apy pro­to­cols. Blood. 2009;114(7):1299-1305.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/37/1851756/37yee.pdf by guest on 13 December 2021


insti­tu­tion expe­ri­ence with next-gen­er­a­tion sequenc­ing min­i­mal resid­ual 41. Avet-Loiseau H, San-Miguel J, Casneuf T, et al. Evaluation of sustained min­
dis­ease detec­tion: clin­i­cal util­ity, appli­ca­tion, and cor­re­la­tion with out­ i­mal resid­ual dis­ease neg­a­tiv­ity with daratumumab-com­bi­na­tion reg­i­mens
comes in plasma cell and lym­phoid malig­nan­cies. Blood. 2019;134(suppl in relapsed and/or refrac­tory mul­ti­ple mye­loma: anal­y­sis of POLLUX and
1):4654. CASTOR. J Clin Oncol. 2021;39(10):1139-1149.
21. Landgren O, Devlin S, Boulad M, Mailankody S. Role of MRD sta­tus in rela­ 42. San-Miguel JF, Avet-Loiseau H, Paiva B, et al. Sustained min­i­mal resid­ual dis­
tion to clin­i­cal out­comes in newly diag­nosed mul­ti­ple mye­loma patients: a ease neg­a­tiv­ity with daratumumab in newly diag­nosed mul­ti­ple mye­loma:
meta-anal­y­sis. Bone Marrow Transplant. 2016;51(12):1565-1568. MAIA and ALCYONE [published online 16 July 2021]. Blood.
22. Munshi NC, Avet-Loiseau H, Rawstron AC, et  al. Association of min­i­mal 43. Diamond B, Korde N, Lesokhin AM, et al. Dynamics of min­i­mal resid­ual dis­
resid­ual dis­ease with supe­rior sur­vival out­comes in patients with mul­ti­ple ease in patients with mul­ti­ple mye­loma on con­tin­uo ­ us lenalidomide main­te­
mye­loma: a meta-anal­y­sis. JAMA Oncol. 2017;3(1):28-35. nance: a sin­gle-arm, sin­gle-cen­tre, phase 2 trial. Lancet Haematol. 2021;8(6):
23. Munshi NC, Avet-Loiseau H, Anderson KC, et  al. A large meta-anal­ y­sis e422-e432.
establishes the role of MRD neg­a­tiv­ity in long-term sur­vival out­comes in 44. Ferrero S, Ladetto M, Drandi D, et  al. Long-term results of the GIMEMA
patients with mul­ti­ple mye­loma. Blood Adv. 2020;4(23):5988-5999. VEL-03-096 trial in MM patients receiv­ing VTD con­sol­i­da­tion after ASCT:
24. Costa LJ, Derman BA, Bal S, et al. International har­mo­ni­za­tion in performing MRD kinet­ics’ impact on sur­vival. Leukemia. 2015;29(3):689-695.
and reporting min­i­mal resid­ual dis­ease assess­ment in mul­ti­ple mye­loma 45. Oliva S, Gambella M, Gilestro M, et al. Minimal resid­ual dis­ease after trans­
tri­als. Leukemia. 2021;35(1):18-30. plan­ta­tion or lenalidomide-based con­sol­i­da­tion in mye­loma patients: a
25. Anderson KC, Auclair D, Adam SJ, et al. Minimal resid­ual dis­ease in mye­ pro­spec­tive anal­y­sis. Oncotarget. 2017;8(4):5924-5935.
loma: appli­ca­tion for clin­i­cal care and new drug reg­is­tra­tion [published 46. Martinez-Lopez J, Wong SW, Shah N, et  al. Clinical value of mea­sur­able
online 28 July 2021]. Clin Cancer Res. resid­ual dis­ease test­ing for assessing depth, dura­tion, and direc­tion of
26. Gormley NJ, Farrell AT, Pazdur R. Minimal resid­ual dis­ease as a poten­tial response in mul­ti­ple mye­loma. Blood Adv. 2020;4(14):3295-3301.
sur­ro­gate end point-lin­ger­ing ques­tions. JAMA Oncol. 2017;3(1):18-20. 47. Görgün GT, Whitehill G, Anderson JL, et  al. Tumor-pro­mot­ing immune-
27. Anderson KC, Auclair D, Kelloff GJ, et al. The role of min­i­mal resid­ual dis­ease sup­pres­sive mye­loid-derived sup­pres­sor cells in the mul­ti­ple mye­loma
test­ing in mye­loma treat­ment selec­tion and drug devel­op­ment: cur­rent micro­en­vi­ron­ment in humans. Blood. 2013;121(15):2975-2987.
value and future appli­ca­tions. Clin Cancer Res. 2017;23(15):3980-3993. 48. Rasche L, Buros A, Weinhold N, et al. The clin­i­cal impact of macrofocal dis­
28. Hol­stein SA, Al-Kadhimi Z, Costa LJ, et  al. Summary of the third annual ease in mul­ti­ple mye­loma dif­fers between pre­sen­ta­tion and relapse. Blood.
blood and mar­row trans­plant clin­i­cal tri­als net­work mye­loma inter­group 2016;128(22):4431.
work­shop on min­i­mal resid­ual dis­ease and immune pro­fil­ing. Biol Blood 49. Moreau P, Attal M, Caillot D, et al. Prospective eval­u­a­tion of mag­netic res­o­nance
Marrow Transplant. 2020;26(1):e7-e15. imag­ing and [(18)F]fluorodeoxyglucose pos­i­tron emis­sion tomog­ra­phy-
29. Rawstron AC, Gregory WM, de Tute RM, et al. Minimal resid­ual dis­ease in com­puted tomog­ra­phy at diag­no­sis and before main­te­nance ther­apy in
mye­loma by flow cytom­e­try: inde­pen­dent pre­dic­tion of sur­vival ben­e­fit symp­tom­atic patients with mul­ti­ple mye­loma included in the IFM/DFCI
per log reduc­tion. Blood. 2015;125(12):1932-1935. 2009 trial: results of the IMAJEM study. J Clin Oncol. 2017;35(25):2911-2918.
30. Attal M, Lauwers-Cances V, Hulin C, et al; IFM 2009 Study. Lenalidomide, 50. Moreau P, Zweegman S, Perrot A, et al. Evaluation of the prog­nos­tic value
bortezomib, and dexa­meth­a­sone with trans­plan­ta­tion for mye­loma. N Engl of pos­i­tron emis­sion tomog­ra­phy-com­puted tomog­ra­phy (PET-CT) at
J Med. 2017;376(14):1311-1320. diag­no­sis and fol­low-up in trans­plant-eli­gi­ble newly diag­nosed mul­ti­ple
31. Perrot A, Lauwers-Cances V, Corre J, et al. Minimal resid­ual dis­ease neg­a­ mye­loma (TE NDMM) patients treated in the phase 3 Cas­si­op ­ eia Study:
tiv­ity using deep sequenc­ing is a major prog­nos­tic fac­tor in mul­ti­ple mye­ results of the Cassiopet Companion Study. Blood. 2019;134(suppl 1):692.
loma. Blood. 2018;132(23):2456-2464. 51. Rasche L, Alapat D, Kumar M, et  al. Combination of flow cytom­e­try and
32. Avet-Loiseau H, Moreau P, Attal M, et al. Efficacy of daratumumab (DARA)+ func­tional imag­ing for mon­i­tor­ing of resid­ual dis­ease in mye­loma. Leuke-
bortezomib/tha­lid­o­mide/dexa­meth­a­sone (D-VTd) in trans­plant-eli­gi­ble mia. 2019;33(7):1713-1722.
newly diag­nosed mul­ti­ple mye­loma (TE NDMM) based on min­i­mal resid­ 52. Zamagni E, Nanni C, Dozza L, et  al. Standardization of 18F-FDG-PET/CT
ual dis­ease (MRD) sta­tus: anal­y­sis of the CASSIOPEIA trial. J Clin Oncol. according to Deauville cri­te­ria for met­a­bolic com­plete response def­i­ni­tion
2019;37(suppl_15):8017-8017. in newly diag­nosed mul­ti­ple mye­loma. J Clin Oncol. 2021;39(2):116-125.
33. Oliva S, Genuardi E, Petrucci MT, et  al. Impact of min­i­mal resid­ual dis­ 53. Pineda-Roman M, Bolejack V, Arzoumanian V, et al. Complete response in
ease (MRD) by mul­ti­pa­ram­e­ter flow cytom­e­try (MFC) and next-gen­er­a­tion mye­loma extends sur­vival with­out, but not with his­tory of prior mono­clo­
sequenc­ing (NGS) on out­come: results of newly diag­nosed trans­plant- nal gammopathy of unde­ter­mined sig­nif­i­cance or smoul­der­ing dis­ease. Br
eli­gi­ble mul­ti­ple mye­loma (MM) patients enrolled in the forte trial. Blood. J Haematol. 2007;136(3):393-399.
2020;136(suppl 1):44-45. 54. Zhan F, Barlogie B, Arzoumanian V, et  al. Gene-expres­sion sig­na­ture of
34. Paiva B, Puig N, Cedena M-T, et al; GEM (Grupo Español de Mieloma)/ benign mono­clo­nal gammopathy evi­dent in mul­ti­ple mye­loma is linked to
PETHEMA (Programa Para el Estudio de la Terapéutica en Hemopatías good prog­no­sis. Blood. 2007;109(4):1692-1700.
Malignas) Cooperative Study Group. Measurable resid­ual dis­ease by next- 55. Weinhold N, Heuck CJ, Rosenthal A, et  al. Clinical value of molec­ u­
lar
gen­er­a­tion flow cytom­e­try in mul­ti­ple mye­loma. J Clin Oncol. 2020;38(8): subtyping mul­ti­ple mye­loma using gene expres­sion pro­fil­ing. Leukemia.
784-792. 2016;30(2):423-430.
35. Goicoechea I, Puig N, Cedena M-T, et al. Deep MRD pro­fil­ing defi­nes out­ 56. Schinke C, Hoering A, Wang H, et al. The prog­nos­tic value of the depth
come and unveils dif­fer­ent modes of treat­ment resis­tance in stan­dard- and of response in mul­ti­ple mye­loma depends on the time of assess­ment, risk
high-risk mye­loma. Blood. 2021;137(1):49-60. sta­tus and molec­u­lar sub­type. Haematologica. 2017;102(8):e313-e316.

44  |  Hematology 2021  |  ASH Education Program


57. Sanoja-Flores L, Flores-Montero J, Puig N, et  al. Blood mon­i­tor­ing of cir­ 67. Derman BA, Stefka AT, Jiang K, et al. Measurable resid­ual dis­ease assessed
cu­lat­ing tumor plasma cells by next gen­er­a­tion flow in mul­ti­ple mye­loma by mass spec­trom­e­try in periph­eral blood in mul­ti­ple mye­loma in a phase
after ther­apy. Blood. 2019;134(24):2218-2222. II trial of carfilzomib, lenalidomide, dexa­meth­a­sone and autol­o­gous stem
58. Mazzotti C, Buisson L, Maheo S, et al. Myeloma MRD by deep sequenc­ing cell trans­plan­ta­tion. Blood Cancer J. 2021;11(2):19.
from cir­cu­lat­ing tumor DNA does not cor­re­late with results obtained in the 68. Davies FE. Is molec­u­lar remis­sion the goal of mul­ti­ple mye­loma ther­apy?
bone mar­row. Blood Adv. 2018;2(21):2811-2813. Hematology Am Soc Hematol Educ Program. 2017;2017(1):205-211.
59. Mithraprabhu S, Morley R, Khong T, et al. Monitoring tumour bur­den and 69. Manasanch EE, Salem DA, Yuan CM, et al. Flow cytometric sen­si­tiv­ity and
ther­a­peu­tic response through anal­y­sis of cir­cu­lat­ing tumour DNA and extra­ char­ac­ter­is­tics of plasma cells in patients with mul­ti­ple mye­loma or its pre­
cel­lu­lar RNA in mul­ti­ple mye­loma patients. Leukemia. 2019;33(8):2022- cur­sor dis­ease: influ­ence of biopsy site and anticoagulation method. Leuk
2033. Lymphoma. 2015;56(5):1416-1424.
60. Guo G, Raje NS, Seifer C, et al. Genomic dis­cov­ery and clonal track­ing in 70. Jackson GH, Davies FE, Pawlyn C, et al; UK NCRI Haematological Oncol-
mul­ti­ple mye­loma by cell-free DNA sequenc­ing. Leukemia. 2018;32(8):1838- ogy Clinical Studies Group. Response-adapted inten­si­fi­ca­tion with cyclo­
1841. phos­pha­mide, bortezomib, and dexa­meth­a­sone ver­sus no inten­si­fi­ca­tion
61. Murray DL, Puig N, Kristinsson S, et al. Mass spec­trom­e­try for the eval­u­ in patients with newly diag­ nosed mul­ ti­
ple mye­ loma (Myeloma XI): a
a­tion of mono­clo­nal pro­teins in mul­ti­ple mye­loma and related dis­or­ders: multicentre, open-label, randomised, phase 3 trial. Lancet Haematol.
an International Myeloma Working Group Mass Spectrometry Committee 2019;6(12):e616-e629.
Report. Blood Cancer J. 2021;11(2):24. 71. Alonso R, Cedena M-T, Wong S, et al. Prolonged lenalidomide main­te­nance

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/37/1851756/37yee.pdf by guest on 13 December 2021


62. Mills JR, Kohlhagen MC, Dasari S, et  al. Comprehensive assess­ ment of ther­apy improves the depth of response in mul­ti­ple mye­loma. Blood Adv.
M-pro­teins using nanobody enrich­ment cou­pled to MALDI-TOF mass spec­ 2020;4(10):2163-2171.
trom­e­try. Clin Chem. 2016;62(10):1334-1344. 72. Costa LJ, Chhabra S, Godby KN, et al. Daratumumab, carfilzomib, lenalid-
63. Barnidge DR, Dasari S, Botz CM, et al. Using mass spec­trom­e­try to mon­i­tor omide and dexa­meth­a­sone (dara-KRd) induc­tion, autol­o­gous trans­plan­ta­
mono­clo­nal immu­no­glob­u­lins in patients with a mono­clo­nal gammopathy. tion and post-trans­plant, response-adapted, mea­sur­able resid­ual dis­ease
J Proteome Res. 2014;13(3):1419-1427. (MRD)-based dara-Krd con­ sol­ i­
da­
tion in patients with newly diag­ nosed
64. Mills JR, Kohlhagen MC, Willrich MAV, Kourelis T, Dispenzieri A, Murray DL. mul­ti­ple mye­loma (NDMM). Blood. 2019;134(suppl 1):860.
A uni­ver­sal solu­tion for elim­i­nat­ing false pos­i­tives in mye­loma due to 73. Moreau P, Siegel DS, Goldschmidt H, et al. Subgroup anal­y­sis of patients
ther­a­peu­tic mono­clo­nal anti­body inter­fer­ence. Blood. 2018;132(6):670- with bio­chem­i­cal or symp­tom­atic relapse at the time of enroll­ment in the
672. endeavor study. Blood. 2018;132(suppl 1):3243.
65. Dispenzieri A, Krishnan AY, Arendt B, et al. MASS-FIX ver­sus stan­dard meth­ 74. Rasmussen A-M, Askeland FB, Schjesvold F. The next step for MRD in mye­
ods to pre­dict for PFS and OS among mul­ti­ple mye­loma patients par­tici­ loma? treating MRD relapse after first line treat­ment in the REMNANT study.
pat­ing on the STAMINA trial. J Clin Oncol. 2021;39(15, suppl):8009. Hemato. 2020;1(2):36-48.
66. Puig N, Paiva B, Contreras T, et al. Analysis of min­i­mal resid­ual dis­ease in bone
mar­row by NGF and in periph­eral blood by mass spec­trom­e­try in newly
diag­nosed mul­ti­ple mye­loma patients enrolled in the GEM2012MENOS65 © 2021 by The Amer­i­can Society of Hematology
clin­i­cal trial. J Clin Oncol. 2021;39(15, suppl):8010. DOI 10.1182/hema­tol­ogy.2021000230

Prakash Singh Shekhawat


Minimal resid­ual dis­ease in mul­ti­ple mye­loma  |  45
CHALLENGES IN MULTIPLE MYELOMA TREATMENT

Treatment of older adult or frail patients


with multiple myeloma

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/46/1852113/46grant.pdf by guest on 13 December 2021


Shakira J. Grant,1,* Ciara L. Freeman,2,* and Ashley E. Rosko3
1
Department of Medicine, Division of Hematology, The University of North Carolina at Chapel Hill, Chapel Hill, NC; Lineberger Comprehensive
Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC; 2Division of Medical Oncology, University of British Columbia,
Vancouver, BC, Canada; and 3Division of Hematology, The Ohio State University, Columbus, OH
*Joint frst authorship.

Older adults with multiple myeloma (MM) are a growing population, and personalizing treatment based on disease and
health status is imperative. Similar to MM staging systems that provide disease-related prognostic information, myeloma-
specific frailty tools can better identify subgroups at greatest risk for treatment-related toxicity and early treatment dis-
continuation, as well as predict overall survival. Several myeloma-specific validated tools are well studied. Although these
fitness/frailty scores have shaped our understanding of the heterogeneity among older adults with myeloma, the applica-
tion of such scores in treatment decision making (ie, transplant considerations, relapse) is an unmet need. Here we outline
how to incorporate frailty assessments in the evaluation of older adults with MM in the clinical setting with consideration
of other factors such as patient preferences, treatment risks/benefits, life expectancy, and disease biology.

LEARNING OBJECTIVES
• Frailty can be objectively characterized in older adults with MM using validated scoring systems.
• Disease and patient factors will influence treatment decisions (health status, biology, risks/benefts, and shared
decision making).

CLINICAL CASE taining her independence and quality of life are her top
A 70-year-old woman with newly diagnosed standard-risk, priorities. She wants to know if there are tools to gauge her
Revised International Staging System (R-ISS) III, immuno- health and would like the best therapy for her personally.
globulin G κ multiple myeloma (MM) presents for consul-
tation. Until 4 months ago, she lived independently, had
an active lifestyle, exercised 3 times weekly, and served Myeloma fitness/frailty scores can inform
as the lead volunteer coordinator for a nonproft organi- treatment tolerance
zation. She was also independent in all activities of daily Predicting treatment-related toxicity is especially critical
living (ADLs) and instrumental activities of daily living for older adults with myeloma due to the heterogeneity
(IADLS). She now reports severe lower back pain that that exists in aging. Differences in how individuals age
limits her mobility, fatigue, and shortness of breath after arise from age-associated losses in physical and cognitive
walking 1 block that has limited her function. She has well- function and the additive impact of medical comorbidities,
controlled diabetes mellitus (glipizide) and hypertension which becomes more prevalent with advancing age.1,2 Pre-
(amlodipine, lisinopril) and reports taking several over-the- vious studies have shown that performance status (Karnof-
counter vitamins to maintain her vitality. She is widowed sky, ECOG) is incapable of fully capturing the ftness level
and lives alone. Her Eastern Cooperative Oncology Group of an older adult.3 Furthermore, although staging systems
(ECOG) performance status is 2. Positron emission tomo- such as the ISS and R-ISS can provide valuable disease-
graphy/computed tomography confrms diffuse lytic lesions related prognostic information, these staging systems are
in the humeri, femora, pelvis, and spine along with multiple incapable of fully discerning the subgroup of patients at
compression fractures involving T6 to T8. Her estimated greatest risk for treatment-related toxicity and early treat-
glomerular fltration rate is 55. She emphasizes that main- ment discontinuation.4,5

46 | Hematology 2021 | ASH Education Program


Table 1. Myeloma fit­ness/frailty risk scores

Frailty score Geriatric domains Biologic marker Cytogenetics included? Score range Interpretation
IMWG6 ADLs None No 0-2 0 (fit)
IADLs 1 (inter­me­di­ate-fit)
CCI 2 (frail)
R-MCI7 Age None Yes 0-9 0-3 (fit)
Fried frailty 4-6 (inter­me­di­ate- fit)
Lung func­tion 7-9 (frail)
Renal func­tion
Karnofsky per­for­mance sta­tus
Facon frailty scale8 Age None No 0-1 0-1 (nonfrail)
CCI ≥2 ≥2 (frail)
ECOG per­for­mance sta­tus9

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/46/1852113/46grant.pdf by guest on 13 December 2021


Myeloma Research Age CRP No <−0.256 <−0.256 (low risk)
Alliance risk pro­file10 WHO per­for­mance sta­tus9 ISS −0.256-−0.0283 −0.256 to −0.0283
>−0.0283 (medium risk)
>−0.0283 (high risk)
Mayo frailty index11 Age NT- proBNP No 0-3 0 (stage 1)
WHO per­for­mance sta­tus9 1 (stage 2)
2 (stage 3)
3 (stage 4)
Ancona vul­ner­a­bil­ity CCI None No 0-2 0 (low)
score12 WHO per­for­mance sta­tus9 1 (inter­me­di­ate)
2 (high)
CRP, C-reac­tive pro­tein; NT-proBNP, N-terminal pro B-type Natriuretic Peptide; WHO, World Health Organization.

To account for some of these aging-related def­i­cits, the Inter- frailty score devel­oped from age, CCI, and ECOG.8 Using this
national Myeloma Working Group (IMWG) pro­ posed a scor­ ing pre­dic­tive score, patients con­sid­ered frail (score ≥2) had infe­rior
sys­tem for mye­loma patient frailty that pre­dicts treat­ment-related out­comes, espe­cially OS. This sim­pli­fied score was sub­se­quently
tox­ic­ity and sur­vival, using age, the Katz ADL, the Lawton IADL, exter­nally val­i­dated by Stege et al16 using data from par­tic­i­pants
and the Charlson Comorbidity Index (CCI)6 (Table 1). In the clin­i­ enrolled in HOVON87/NMSG18. For many cen­ters, lung func­tion
cal vignette, the patient has an IMWG score of 1 = inter­me­di­ate-fit. test­ing and for­mal frailty scor­ing (ie, Fried frailty phe­no­type) may
Among the inter­me­di­ate-fit sub­group, the 3-year over­all sur­vival be out of scope of prac­tice. Notwithstanding, frailty scor­ing is
(OS) was 76% (haz­ard ratio [HR], 1.61; 95% CI, 1.02-2.56; P = .042), typ­i­cally col­lected by patients as self-report, and func­tional test­
the cumu­la­tive inci­dence of grade 3 or higher nonhematologic ing such as gait speed requires no cost; these instru­ments are
adverse events at 12 months was 16.7% (HR, 1.23; 95% CI, 0.89-1.71; quick, valid, and cost-effec­tive and inform sur­vival.
P = .217), and the cumu­la­tive inci­dence of treat­ment dis­con­tin­u­a­
tion at 12 months was 20.8% (HR, 1.41; 95% CI, 1.00-2.01; P = .052).6 Rationale for con­sid­er­ing nontransplant strat­e­gies
in older patients with mye­loma
After discussing her frailty score, the patient inquires At this time, the opti­mal tool for assessing frailty for treat­ment
whether addi­tional frailty scales exist and whether deci­sions (ie, trans­plant and nontransplant) has yet to be deter­
these apply to newly diag­nosed MM mined. Despite the wide­spread use of the IMWG frailty score
Additional frailty scores have been devel­oped incor­po­rat­ing age, in clin­i­cal and research set­tings, it has its lim­i­ta­tions. Namely,
comorbidities, phys­i­cal per­for­mance, and bio­mark­ers (Table 1). the use of chro­no­logic age can detract from the con­cept of
Engelhardt et al7 found that age, mye­ loma cyto­ge­
net­ics, frailty bio­logic/func­tional age, and CCI is likely to exclude mye­loma-
(Fried phe­no­type), per­for­mance sta­tus, and pul­mo­nary and re- spe­cific comorbidities. Recommendations for lon­gi­tu­di­nal assess­
nal func­tion (Revised Myeloma Comorbidity Index [R-MCI]) were ments of fit­ness and frailty have been based on con­sen­sus expert
prog­ nos­ tic for OS in a pro­ spec­ tive cohort of 801 patients opin­ion.17-19 Although these fit­ ness/frailty scores have shaped
with newly diag­nosed MM. Of the 801 evaluable patients, 30.8% our under­stand­ing of the het­ero­ge­ne­ity among older adults with
were con­sid­ered fit, 55.7% inter­me­di­ate-fit, and 13.5% frail.7 In mye­loma, the appli­ca­tion of such scores in the clin­i­cal set­ting
the der­i­va­tion cohort, 552 patients clas­si­fied as frail had a near requires con­sid­er­ation of other fac­tors such as patient pref­er­
10-fold greater risk of dying than those con­sid­ered fit (HR, 9.57; ences, life expec­tancy, and dis­ease biol­ogy.
95% CI, 6.52-14.03).7 In our clin­i­cal vignette, we are not pro­vided For patients in whom can­di­dacy of trans­plant is a con­cern
with data to esti­mate the patient’s Fried frailty phe­no­type (re- due to frailty, there are a num­ber of options. The results from
quires gait speed, grip strength, weight, self-reported exhaus­ a sin­gle-cen­ter, sin­gle-arm phase 2 study enroll­ing trans­plant-
tion, and phys­i­cal activ­ity level).13 Neither are we pro­vided with inel­i­gi­ble adults with newly diag­nosed MM found the trip­let reg­
pul­mo­nary func­tion test data, and as such, we can­not cal­cu­late i­men of reduced-inten­sity lenalidomide, sub­cu­ta­ne­ous bortezo-
this patient’s R-MCI score. A ret­ro­spec­tive anal­y­sis of 1618 trial mib, and dexa­meth­a­sone to be tol­er­a­ble and effec­tive.20 Despite
par­tic­i­pants enrolled in the FIRST trial14,15 reported on a sim­pli­fied 62% of par­tic­i­pants expe­ri­enc­ing low-grade sen­sory neu­rop­a­thy,
Prakash Singh Shekhawat
Frailty in mul­ti­ple mye­loma  |  47
Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/46/1852113/46grant.pdf by guest on 13 December 2021
Figure 1. PFS and OS in matched population aged 64 to 70 years treated with or without ASCT, from myeloma XI subanalysis (with
permission).36

only 1 patient expe­ri­enced grade 3 or higher neu­rop­a­thy symp­ (HR, 0.53; P < .0001). The lat­est update of the MAIA study reported
toms. After a 60-month fol­low-up, the median pro­gres­sion-free an esti­mated 48-month PFS of 60%.22 There are numer­ous non-
sur­vival (PFS) was 41.9 months, and the median OS has still not transplant reg­i­mens19,23,24; other alter­na­tives could include daratu-
been reached. Based on this study’s find­ings, reduced-inten­sity mumab, bortezomib, melphalan, prednisone (D-VMP); borezomib,
lenalidomide, sub­cu­ta­ne­ous bortezomib, and dexa­meth­a­ lenalidomide, dexamethasone (VRD); cyclophosphadmide, bor-
sone could be con­sid­ered for select trans­plant-inel­i­gi­ble older tezomib, dexamethasone (CyborD). The most com­mon induc­tion
adults who may be at higher risk for treat­ment-related neu­rop­ strat­egy for those aged less than 65 to 74 years, in the United
a­thy. The excel­lent out­comes achieved with the upfront use of States, is VRD front­line ther­apy.25 Studies are cur­rently under way
daratumumab, lenalidomide, and dexa­meth­a­sone (DRd) in this eval­u­at­ing whether fit­ness-based or risk-adapted ther­apy assign­
patient pop­u­la­tion rival those achieved by induc­tion, trans­plant, ments in mye­ loma improve dis­ ease- and patient-related out­
and main­te­nance. This rec­om­men­da­tion is supported by find­ings comes26-30 and to under­stand whether mye­loma ther­a­pies result
from the MAIA trial,21 a phase 3 mul­ti­cen­ter study in which 44% of in lon­gi­tu­di­nal changes in an indi­vid­ual’s fit­ness/frailty sta­tus.
enrolled par­tic­i­pants were 75 years or older and dem­on­strated
a lon­ger median PFS favor­ing the daratumumab-containing arm. Rationale for con­sid­er­ing trans­plant in the older patients
Among those treated with lenalidomide and dexa­ meth­ a­
sone with mye­loma
alone, the median PFS was 34.4 months vs not reached for those Autologous stem cell trans­plant (ASCT), even in the era of novel
who received daratumumab, lenalidomide, and dexa­meth­a­sone agents, con­tin­ues to be a part of the ther­a­peu­tic par­a­digm with

48  |  Hematology 2021  |  ASH Education Program


clear improve­ments in PFS when incor­po­rated in the upfront set­
ting.31 Historically, the bulk of evi­dence to sup­port the ongo­ing use
of high-dose mel­pha­lan was gen­er­ated from tri­als enroll­ing youn­
ger patients, typ­i­cally 65 years or youn­ger, which excludes almost
two-thirds of patients with newly diag­nosed mye­loma.32 Although
Euro­pean guide­lines con­tinue to rec­om­mend lim­it­ing ASCT to pa-
tients youn­ger than 70 years, there is increas­ing evi­dence to sup­
port the safe use of ASCT in older patients, with empha­sis placed
on bio­log­i­cal age rather than chro­no­log­i­cal age.33,34
A meta-anal­y­sis attempting to quan­tify the ben­e­fit of upfront
ASCT in older patients showed a sig­nif­i­cant improve­ment in OS
(HR, 0.44; 95% CI, 0.34-0.58; P < .001), but authors commented
on the pau­city of evi­dence directly com­par­ing ASCT with non-
ASCT approaches in this age group.35 Another subanalysis of the

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/46/1852113/46grant.pdf by guest on 13 December 2021


large phase 3 mye­loma XI trial attempted to use an overlapping
cohort of age-matched patients (64-70 years) to com­pare out­
comes and tox­ic­ity for those enrolled in the ASCT and non-ASCT
arms.36 Importantly, the deci­sion to enroll in the trans­plant arm
was not ran­dom­ized but based on inves­ti­ga­tor dis­cre­tion and is
a cen­tral lim­i­ta­tion of this study. The study yielded a sig­nif­i­cant
advan­tage in favor of upfront ASCT with improve­ments in PFS
(HR, 0.41; P < .0001) and OS (HR, 0.51; P < .0001), after adjusting
for poten­tial base­line covariates (frailty, response to induc­tion)
(Figure 1). Interestingly, no dif­fer­ences in tox­ic­ity were observed
com­ pared with the cohort youn­ ger than 65 years who also
under­went ASCT. These results echo those reported by a large
anal­y­sis of 2092 patients 70 years or older from the Center for
International Blood and Marrow Transplant Research data­base,
show­ing iden­ti­cal out­comes for patients who were ­able to tol­er­
ate a 200-mg/m2 dose com­pared with youn­ger patients.34 Again,
no dif­fer­ences were seen in nonrelapse mor­tal­ity, but patients
who received dose reduc­ tions (140 
mg/m2) did have infe­ rior
sur­vival esti­ma­tes, thought sec­ond­ary to comorbidities such as
renal fail­ure or frailty. Although the evi­dence is not ideal, an open
dis­cus­sion about the pros and cons of ASCT is warranted, tak­ing
into account the patient’s own pref­er­ences and goals for treat­
ment. Patients with mye­loma in gen­eral do tend to pri­or­i­tize PFS
but also place high value on qual­ity of life, the side effects of
treat­ment, and poten­tial impact that treat­ments have on their
care­giv­ers.37,38

Establishing the risk/ben­e­fit ratio for an older patient with Figure 2. Considerations when evaluating older patients for
mye­loma consideration of ASCT. PS, performance status.
For the treating cli­ ni­
cian, the chal­ lenge is how best to iden­
tify older patients who will derive the greatest ben­e­fit from up-
front ASCT. As discussed, avail­­able frailty scores can assist with The hema­to­poi­etic cell trans­plan­ta­tion spe­cific comorbid-
deci­sion mak­ing, pro­vid­ing an objec­tive assess­ment of the ini­ ity index (HCT-CI) was orig­ i­
nally devel­
oped in the set­ ting of
tial and chang­ing “fit­ness” of patients with mye­loma under­go­ allotransplant but sub­se­quently eval­u­ated in a cohort of 1156
ing ther­apy.17 Although con­sid­ered a stan­dard, it is impor­tant to patients with mye­loma and found to be pre­dic­tive of out­come.39
remem­ber that the IMWG Frailty Score was derived from a co- Patients with a score of 1 to 2 and more than 2 (com­pared with
hort of patients deemed to be trans­ plant inel­i­
gi­
ble.6 Similarly, HCT-CI of 0) or a Karnofsky performance status (KPS) less than
the recently published sim­pli­fied frailty score8 and UK Myeloma 90 had infe­rior OS. It is note­wor­thy that treat­ment-related mor­
Research Alliance Risk Profile were also derived and val­i­dated in tal­ity was low at 2% and equiv­a­lent for patients with an HCT-CI
a cohort of trans­plant-inel­i­gi­ble patients.10 The R-MCI was derived score of 0 or more than 2. The main cause of treat­ment-related
from a cohort includ­ing 343 patients 65 years or older and 383 who mor­tal­ity was dis­ease relapse and pro­gres­sion. Again, only 23%
under­went ASCT, but the exact over­lap between these 2 groups is of this cohort was 65 years or older, and even fewer were 70
unclear.7 Nonetheless, given that those des­ig­nated as “frail” with years or older (n = 72, 6%). Regardless, in the absence of a bet­
these mea­sures are more likely to expe­ri­ence sig­nif­i­cant toxicities ter alter­na­tive, it remains a recommended method of eval­u­at­ing
to induc­tion ther­apy, the addi­tional tox­ic­ity of an ASCT in those patients pretransplant, with sug­ges­tion to con­sider alter­na­tive
iden­ti­fied as such would likely out­weigh the poten­tial ben­e­fit.6 approaches in those with an HCT-CI more than 2 or KPS less
Prakash Singh Shekhawat
Frailty in mul­ti­ple mye­loma  |  49
Table 2. Relapsed mul­ti­ple mye­loma: early and late relapse stud­ies

Sel/Dex Melflufen/Dex Belantamab-mafadotin Idecabtagene


VPd (OPTIMISMM) Anti-CD38 Anti-SLAMF7 (STORM) (HORIZON OP-106) (DREAMM-2) vicleucel
KRd (ASPIRE) DVd (CASTOR) ERd (ELOQUENT-2) Sel/Pom/Dex Melflufen/Bort/Dex Belamaf/Pom/Dex Ciltacabtagene
(STOMP arm1) (ANCHOR OP-104) (ALGONQUIN) autoleucel
IRd (TOURMALINE-MM1) DRd (POLLUX) EPd (ELOQUENT-3) Sel/Carf/Dex Melflufen/Dara/Dex Belamaf/Bort/Dex Orvacabtagene
(STOMP arm6) (ANCHOR OP-104) (DREAMM-6armB) autoleucel
SVd (BOSTON) DKd (CANDOR) Sel/Dara/Dex P-BCMA-101
(STOMP arm5)
DPd (APOLLO)
Isa-Pd (ICARIA)
Isa-Kd (IKEMA)

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/46/1852113/46grant.pdf by guest on 13 December 2021


Table mod­i­fied from Tables 2 to 5 in Nathwani et al.43
Carf,  carfilzomib; Dara, daratumumab; Dex, dexamethasone; DKd, daratumumab, carfilzomib, dexamethasone; DPd, daratumumab, pomalidomide,
dexamethasone; DVd, daratumumab, bortezomib, dexamethasone; EPd, elotuzumab, pomalidomide, dexamethasone; ERd, elotuzumab,
lenalidomide, dexamethasone; IRd, ixazomib, lenalidomide, dexamethasone; Isa, isatuximab; Pom, pomalidomide; Sel, selinexor; SVd, selinexor,
bortezomib, dexamethasone; VPd, bortezomib, pomalidomide, dexamethasone.

than 90.40,41 Other fac­tors that will also influ­ence the dis­cus­sion poten­tial ben­e­fits as high­lighted above. Nonetheless, after a long
around incor­po­ra­tion of ASCT include the biol­ogy of the dis­ease, dis­cus­sion about the rel­a­tive pros and cons, on the basis of per­
prog­nos­tic risk, response to induc­tion, and the wishes of the sonal pref­er­ence, she opted for a non-ASCT strat­egy.
patient and their social sit­ua
­ ­tion (Figure 2). For fit older patients who decline upfront ASCT, there are a
num­ber of options. The excel­lent out­comes achieved with the
upfront use of DRd in this patient pop­u­la­tion rival those achieved
by induc­tion, trans­plant, and main­te­nance. The lat­est update of
CLINICAL CASE (induc­t ion strat­e­g ies) the MAIA study reported an esti­mated 48-month PFS of 60%.22
This patient had a nor­mal echo and pulmonary function test, Other alter­na­tives could include D-VMP, VRD, or CyborD. The
achieved a very good partial response with 3 cycles of induc­tion most com­mon induc­tion strat­egy for those aged less than 65 to
with VRD, and had no high-risk cyto­ge­net­ics. Her ECOG was 1 and 74 years, in the United States, is VRD front­line ther­apy and was
KPS 80% at her pretransplant appoint­ment. Although her renal sub­se­quently recommended for this patient.25
func­tion improved, she still scored as inter­me­di­ate-fit according
to the R-MCI based on a glomerular filtration rate less than 60,
but HCT-CI was low risk based on the higher thresh­old for renal
func­tion (score 1). She had a sup­port­ive care­giver in good health CLINICAL CASE (relapsed MM)
who had a solid under­stand­ing of the risks and ben­e­fits. She was The patient tol­er­ated VRD induc­tion ther­apy and was in a very good
offered upfront ASCT given the low poten­tial risks of tox­ic­ity and partial response and con­ tin­
ued on lenalidomide main­ te­
nance.

Table 3. Carfilzomib-based stud­ies: com­par­i­son of PFS and OS by age and fit­ness49

ASPIRE50 ENDEAVOR51 ARROW52


PFS <70 KRd 28.6 Fit KRd 31.4 <65 Kd NE* Fit Kd NE* <65 1 × Kd 12.2 Fit 1 × Kd 15.7
(median,
Rd 17.6 Rd 18.9 Vd 9.5 Vd 12.1 2 × Kd 5.6 2 × Kd 5.7
months)
≥70 KRd 23.8 Frail KRd 24.1 65-74 Kd 15.6 Frail Kd 18.7 65-74 1 × Kd 9.2 Frail 1 × Kd 10.3
Rd 15.9 Vd 9.5 Vd 6.6 2 × Kd 8.4 2 × Kd 6.6
Rd 16.0 ≥75 Kd 18.7 ≥75 1 × Kd 12.2
Vd 8.9 2 × Kd 9.5
OS <70 Not given Fit KRd 55.6 <65 Not given Fit Kd NE* <65 Not given Fit Not given
(median,
Rd 43.3 Vd 42.2
months)
≥70 Frail KRd 36.4 65-74 Frail Kd 33.6 65-74 Frail
Rd 26.2 ≥75 Vd 21.8 ≥75
*not estimable.
Frailty assess­ment based on the IMWG frailty index.6,53
Rd, lenalidomide and dexa­meth­a­sone; Vd, bortezomib and dexa­meth­a­sone.

50  |  Hematology 2021  |  ASH Education Program


Table 4. Carfilzomib-based stud­ies: com­par­i­son of grade >3 treatment emergent adverse event (TEAE) and car­diac events by age and fit­ness49

ASPIRE50 ENDEAVOR51 ARROW52


Grade ≥3 TEAE, n (%) <70 KRd 236 (81.7) Fit KRd 102 (89) <65 Kd 152 (68.2) Fit Kd 91 (83) <65 1 × Kd 61 (59.2) Fit 1 × Kd 33 (55)
Rd 215 (77.6) Rd 96 (84) Vd 131 (63) Vd 76 (64) 2 × Kd 58 (56.3) 2 × Kd 41 (62)
≥70 KRd 92 (89.3) Frail KRd 86 (93) 65-74 Kd 124 (76.1) Frail Kd 142 (85) 65-74 1 × Kd 62 (68.9) Frail 1 × Kd 64 (81)
Rd 99 (88.4) Rd 94 (94) Vd 128 (69.9) Vd 125 (79) 2 × Kd 64 (63.4) 2 × Kd 42 (70)
≥75 Kd 63 (81.8) ≥75 1 × Kd 38 (84.4)
Vd 46 (70.8) 2 × Kd 23 (74.2)
Grade ≥3 car­diac fail­ure, <70 KRd 6 (2.1) Fit KRd 5 (4) <65 Kd 6 (2.7) Fit Kd 4 (4) <65 1 × Kd 1 (1) Fit 1 × Kd 1 (2)
n (%)
Rd 5 (1.8) Rd 2 (2) Vd 3 (1.4) Vd 2 (2) 2 × Kd 6 (5.8) 2 × Kd 1 (2)
≥70 KRd 9 (8.7) Frail KRd 9 (10) 65-74 Kd 8 (4.9) Frail Kd 15 (9) 65-74 1 × Kd 5 (5.6) Frail 1 × Kd 3 (4)
Rd 2 (1.8) Rd 1 (1) Vd 3 (1.6) Vd 7 (4) 2 × Kd 2 (2) 2 × Kd 5 (8)

Prakash Singh Shekhawat


≥75 Kd 8 (10.4) ≥75 1 × Kd 1 (2.2)
Vd 2 (3.1) 2 × Kd 2 (6.5)
Grade ≥3 ische­mic heart dis­ <70 KRd 8 (2.8) Fit KRd 4 (3) <65 Not given Fit Kd 2 (2) <65 1 × Kd 1 (1) Fit 1 × Kd 1 (1)
ease, n (%)
Rd 7 (2.5) Rd 3 (3) Vd 2 (<1) 2 × Kd 0 2 × Kd 0
≥70 KRd 5 (4.9) Frail KRd 7 (8) 65-74 Frail Kd 8 (5) 65-74 1 × Kd 0 Frail 1 × Kd 0
Rd 1 (0.9) Rd 1 (1) Vd 6 (4) 2 × Kd 1 (1) 2 × Kd 1 (2)
≥75 ≥75 1 × Kd 1 (2.2)
2 × Kd 1 (3.2)
6,54
Frailty assess­ment based on the IMWG frailty index.

Frailty in mul­ti­ple mye­loma  |  51


Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/46/1852113/46grant.pdf by guest on 13 December 2021
She returned to volunteering and remained inde­pen­dent. After 3 Most patients ben­e­fit from a mono­clo­nal anti­body, in com­
years on main­te­nance ther­apy, at age 73 years, the patient devel­ bi­na­tion, at first relapse. Daratumaumb, an anti-CD38 mono­clo­
oped acom­mu­nity-acquired pneu­mo­nia and was hos­pi­tal­ized for nal anti­body, can be offered in com­bi­na­tion with a PI or IMiD at
1 week. She then reported progressing back pain and was iden­ first relapse or as monotherapy among dis­ease that is dou­ble-
ti­fied to have a new ver­te­bral com­pres­sion defor­mity. Serologic refrac­tory or fol­low­ing third line of treat­ment. Triple-drug ther­apy
test­ing revealed a rise in her κ light chains and immu­no­glob­u­lin G for frail patients at relapse may require dose mod­i­fi­ca­tions. As 1
κ immu­no­glob­u­lin pro­tein. exam­ple, in the ICARIA study,47 eval­u­at­ing isatuximab, pomalido-
mide, and dexa­meth­a­sone vs pomalidomide and dexa­meth­a­sone,
43% of patients required dose reduc­tions of pomalidomide and
Relapsed MM 33% of patients required dose reduc­tions in dexa­meth­a­sone with
When MM relapses, this is a chal­leng­ing time for any patient and tri­ple ther­apy. However, patients ran­dom­ized to the isatuximab,
is fur­ther con­founded if the patient’s health sta­tus is frail, clin­i­cal pomalidomide, and dexa­meth­a­sone arm had a sub­stan­tially lon­
health is ten­u­ous due to ill­ness, or there is impair­ment in end- ger dura­tion of ther­apy com­pared with pomalidomide and dexa­
organ func­tion. Importantly, frailty is a dynamic state, and a reas- meth­a­sone (41 vs 24 weeks). In gen­eral, no age- or frailty-based

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/46/1852113/46grant.pdf by guest on 13 December 2021


sessment can fur­ther define next treat­ment deci­sions. Most stud­ dose atten­u­a­tions are recommended for mono­clo­nal anti­body
ies sug­gest that health-related qual­ity of life tracks with dis­ease ther­apy.48 In terms of under­ly­ing health sta­tus, par­tic­u­larly rel­e­
con­ trol; there­ fore, addressing next steps for relapsed dis­ ease vant are periph­eral neu­rop­a­thy, car­dio­vas­cu­lar health, throm­bo­
would also improve health-related qual­ity of life. For relapsed MM, sis, recur­rent infec­tions, bone mar­row reserve, and pul­mo­nary
most phase 3 ran­dom­ized con­trolled tri­als have com­pared the health. Cardiovascular events are of sig­nif­i­cant inter­est in aging
effec­tive­ness and safety of 3-drug reg­i­mens to 2-drug reg­i­mens, adults, par­ tic­u­larly as this applies to carfilizomib-based strat­e­
gen­er­ally depicting PFS ben­e­fits of 3-drug com­bi­na­tions over 2- gies. In a post hoc anal­y­sis,49 eval­u­at­ing 3 carfilizomib-based tri­als
drug com­bi­na­tions. For fit patients, a 3-drug reg­i­men has become (ASPIRE,50 ENDEAVOR,51 ARROW52), 25% to 35% of patients across
the stan­dard of care for relapsed dis­ease. For patients who are frail stud­ies were cat­e­go­rized as frail using IMWG cri­te­ria. OS was
according to stan­dard­ized cri­te­ria (eg, IMWG), data are quite lim­ reduced, and treat­ ment-emer­gent adverse events were more
ited on tol­er­ance of relapsed ther­a­pies. Age-strat­i­fied sub­group com­mon among frail patients (Table 3), includ­ing increased car­
anal­y­sis is not an indi­ca­tor of health sta­tus and should not be used diac fail­ure events (Table 4).49 In a pro­spec­tive study eval­u­at­ing
as a sur­ro­gate of tol­er­ance. In this patient sce­nario, a thought­ful 95 patients receiv­ing a PI (bortezomib or carfilizomib) in relapsed
explo­ra­tion of options to per­son­al­ize ther­apy is indi­cated. dis­ease, more than half of patients expe­ri­enced grade 3 car­diac
Treatment options for relapsed dis­ease can be tai­lored based events, and most events occurred within the first 3 months of
on dis­ease biol­ogy, tempo of dis­ease relapse, tol­er­ance of prior ther­apy.54 Moreover, patients who expe­ri­enced a car­dio­vas­cu­lar
ther­a­pies, health sta­tus, and shared deci­sion mak­ing.42 Drug clas­ event had infe­rior PFS and worse OS.54 A global assess­ment of
ses include immunomodulatory drugs (lenalidomide, pomalido- health and reassessment of frailty can fur­ther define next treat­
mide), proteasome inhibitors (bortezomib, carfilizomib, ixazomib), ment options.
mono­clo­nal antibodies (daratumumab, isatuximab, elotuzumab),
B cell maturation antigen tar­gets (balantamab-mafodotin), novel
ther­apy (histone deacetylase inhib­i­tors, selec­tive inhib­i­tors of CLINICAL CASE (con­clu­sion)
nuclear export), or alkylator ther­ apy (cyclo­ phos­pha­mide, mel­
pha­lan, mel­pha­lan flufenamide [pep­tide con­ju­gate]) with ste­roids The patient recov­ered from her hos­pi­tal admis­sion. Recalcula-
(dexa­meth­a­sone, pred­ni­sone) in var­i­ous com­bi­na­tions (Table 2). tion of the IMWG score remained inter­me­di­ate-fit. The patient
Class switching from first-line treat­ment to sec­ond-line ther­apy was started on a daratumumab, bortezomib, and dose-reduced
is recommended using a dif­fer­ent mech­an ­ ism of action not pre­vi­ dexa­meth­a­sone for relapsed MM. Special atten­tion to neu­ro-
ously used. Among frail patients, if 2-drug ther­apy was offered at p­a­thy was tracked closely. The patient benefited from phys­i­cal
induc­tion, such as lenalidomide and dexa­meth­a­sone or bortezo- ther­apy to assist with recov­ery from back pain and tol­er­ated
mib and dexa­meth­a­sone, if frailty remains or intol­er­ance devel­ops, ther­apy with­out dif­fi­culty.
the patients could read­ily switch to the alter­nate 2-drug class.
Consistently across stud­ies, dexa­meth­a­sone is dose atten­u­ated The mul­ti­ply­ing ther­a­pies of mye­loma have resulted in chal­
by advanc­ing age and is recommended for bet­ter tol­er­ance. Ste- leng­ing clin­i­cal deci­sions. Moreover, the future of MM treat­ment
roids are a back­bone of mye­loma ther­apy but are asso­ci­ated with deci­sions will soon include not only trans­plant deci­sion mak­ing
many multiorgan side effects that can affect qual­ity of life and but also cel­lu­lar ther­apy can­di­dacy. How to best pre­scribe these
adher­ence, par­tic­u­larly for older adults.44 Steroid dose atten­u­a­ ther­a­pies for aging adults has yet to be deter­mined. To rec­on­
tions are well char­ac­ter­ized in MM, and low-dose dexa­meth­a­sone cile dif­fer­ences in health with aging, par­tic­u­larly for com­plex MM
is recommended due to infe­rior sur­vival with higher-dose dexa­ deci­sions, a com­pre­hen­sive eval­u­a­tion of health is required. The
meth­a­sone.45 More recent lit­er­a­ture exam­ined lower dos­ages of MM com­ mu­nity has embraced sev­ eral frailty scor­ ing sys­
tems,
lenalidomide with early dis­con­tin­u­a­tion of dexa­meth­a­sone that each with lim­i­ta­tions, yet impor­tantly, these impor­tant first steps
was proven safe and resulted in sim­i­lar out­comes to patients on of embed­ding assess­ments in clin­ic ­ al tri­als are a start. In sum­
con­tin­u­ous lenalidomide-dexa­meth­a­sone.46 Unlike many front­line mary, mye­loma treat­ments can be mod­i­fied or informed by frailty
stud­ies with ste­roid atten­u­a­tions, lim­ited pro­spec­tive data are scor­ing sys­tems, and these tools dem­on­strate how geri­at­ric prin­
avail­­able for dose atten­u­a­tions of stan­dard relapsed ther­a­pies for ci­ples can be incor­po­rated into risk strat­i­fi­ca­tion to improve out­
frail patients. comes.

52  |  Hematology 2021  |  ASH Education Program


Conflict-of-inter­est dis­clo­sure ment algo­ rithms in can­ cer patients at advanced age. Haematologica.
2020;105(5):1183-1188.
Shakira J. Grant: no conflicts to disclose.
18. Larocca A, Dold SM, Zweegman S, et  al. Patient-cen­ tered prac­tice in
Ciara L. Freeman received research funding from Lundbeck. elderly mye­loma patients: an over­view and con­sen­sus from the Euro­pean
Ashley E. Rosko: no conflicts to disclose. Myeloma Network (EMN). Leukemia. 2018;32(8):1697-1712.
19. Grant SJ, Mian HS, Giri S, et al. Transplant-inel­i­gi­ble newly diag­nosed mul­
Off-label drug use ti­ple mye­loma: cur­rent and future approaches to clin­i­cal care: a Young
Shakira J. Grant: nothing to disclose. International Society of Geriatric Oncology review paper. J Geriatr Oncol.
2021;12(4):499-507.
Ciara L. Freeman: nothing to disclose.
20. O’Donnell EK, Laubach JP, Yee AJ, et al. A phase 2 study of mod­i­fied lena-
Ashley E. Rosko: nothing to disclose. lidomide, bortezomib and dexa­meth­a­sone in trans­plant-inel­i­gi­ble mul­ti­ple
mye­loma. Br J Haematol. 2018;182(2):222-230.
Correspondence 21. Facon T, Kumar S, Plesner T, et al; MAIA Trial Investigators. Daratumumab
Ashley E. Rosko, Division of Hematology, The Ohio State Univer- plus lenalidomide and dexa­ meth­ a­
sone for untreated mye­ loma. N Engl
J Med. 2019;380(22):2104-2115.
sity, 1150C Lin­coln Tower, 1800 Cannon Dr, Colum­bus, OH 43210;
22. Kumar SK, Facon T, Usmani SZ, et al. Updated anal­y­sis of daratumumab plus
e-mail: ashley​­.rosko@osumc​­.edu. lenalidomide and dexa­meth­a­sone (D-Rd) ver­sus lenalidomide and dexa­

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/46/1852113/46grant.pdf by guest on 13 December 2021


meth­a­sone (Rd) in patients with trans­plant-inel­i­gi­ble newly diag­nosed
References mul­ti­ple mye­loma (NDMM): the phase 3 MAIA study. Blood. 2020;136:24-
1. Wildes TM, Tuchman SA, Klepin HD, et  al. Geriatric assess­ment in older 26.
adults with mul­ti­ple mye­loma. J Am Geriatr Soc. 2019;67(5):987-991. 23. Moreau P, Touzeau C, Vij R, Goldsmith SR, Rosko AE. Newly diag­nosed
2. Hshieh TT, Driver JA, Abel GA. Cognitive impair­ment among older patients mye­loma in 2020. Am Soc Clin Oncol Educ Book. 2020;40:1-15.
with hema­to­logic can­cers—reply. JAMA Oncol. 2018;4(12):1784. 24. Berbari HE, Kumar SK. Initial ther­a­peu­tic approaches to patients with mul­ti­ple
3. Hamaker ME, Prins MC, Stauder R. The rel­e­vance of a geri­at­ric assess­ mye­loma. Adv Ther. 2021;38(7):3694-3711.
ment for elderly patients with a haematological malig­nancy—a sys­tem­atic 25. Lee HC, Ailawadhi S, Gasparetto C, et al. Treatment pat­terns and out­comes
review. Leuk Res. 2014;38(3):275-283. in elderly patients with newly diag­nosed mul­ti­ple mye­loma: results from
4. Kuroda J, Shimura Y, Ohta K, et al; Kansai Myeloma Forum Investigators. the Connect® MM Registry. Blood Cancer J. 2021;11(7):134.
Limited value of the inter­na­tional stag­ing sys­tem for predicting long-term 26. Cairns D, Pawlyn C, Royle K-L, et al. Frailty-adjusted ther­apy in trans­plant
out­come of trans­plant-inel­i­gi­ble, newly diag­nosed, symp­tom­atic mul­ti­ple non-eli­gi­ble patients with newly diag­nosed mul­ti­ple mye­loma (FiTNEss): a
mye­loma in the era of novel agents. Int J Hematol. 2014;99(4):441-449. UK mye­loma research alli­ance study, mye­loma XIV. Blood. 2019;134(suppl
5. Pawlyn C, Cairns D, Kaiser M, et  al. The rel­a­tive impor­tance of fac­tors pre- 1):3153.
dicting out­come for mye­loma patients at dif­fer­ent ages: results from 3894 27. PETHEMA Foundation. Treatment for elderly fit newly diag­nosed mul­ti­
patients in the Myeloma XI trial. Leukemia. 2020;34(2):604-612. ple mye­loma patients aged between 65 and 80 years. Accessed 1 Apr
6. Palumbo A, Bringhen S, Mateos M-V, et  al. Geriatric assess­ ment pre­ 2021. https:​­//­ClinicalTrials​­.gov​­/show​­/NCT03742297
dicts sur­vival and toxicities in elderly mye­loma patients: an International 28. Onlus FEI. Study to deter­mine the effi­cacy and safety of stan­dard sched­
Myeloma Working Group report. Blood. 2015;125:2068-2074. ule ver­sus a new algo­rithm of dose reduc­tions in elderly and unfit newly
7. Engelhardt M, Domm A-S, Dold S-M, et  al. A con­cise revised Myeloma diag­nosed mul­ti­ple mye­loma patients receiv­ing lenalidomide plus ste­
Comorbidity Index as a valid prog­nos­tic instru­ment in a large cohort of 801 roids. Accessed 1 Apr 2021. https:​­/​­/ClinicalTrials​­.gov​­/show​­/NCT02215980
mul­ti­ple mye­loma patients. Haematologica. 2017;102(5):910-921. 29. Leeds Uo, UK CR, Takeda, et al. Myeloma XIV: frailty-adjusted ther­apy in
8. Facon T, Dimopoulos MA, Meuleman N, et  al. A sim­ pli­fied frailty scale trans­plant non-eli­gi­ble patients with newly diag­nosed mul­ti­ple mye­loma.
pre­dicts out­comes in trans­plant-inel­i­gi­ble patients with newly diag­ Accessed 1 Apr 2021. https:​­/​­/ClinicalTrials​­.gov​­/show​­/NCT03720041
nosed mul­ti­ple mye­loma treated in the FIRST (MM-020) trial. Leukemia. 30. Stege CAM, Nasserinejad K, van der Spek E, et al. Efficacy and tol­er­a­bil­ity
2020;34(1):224-233. of ixazomib, daratumumab and low dose dexa­meth­a­sone (Ixa Dara dex) in
9. Oken MM, Creech RH, Tormey DC, et al. Toxicity and response cri­te­ria of unfit and frail newly diag­nosed mul­ti­ple mye­loma (NDMM) patients; results
the Eastern Cooperative Oncology Group. Am J Clin Oncol. 1982;5(6):649- of the interim effi­cacy anal­y­sis of the phase II HOVON 143 study. Blood.
655. 2019;134(suppl 1):695.
10. Cook G, Royle K-L, Pawlyn C, et  al. A clin­i­cal pre­dic­tion model for out­ 31. Al Hamed R, Bazarbachi AH, Malard F, Harousseau J-L, Mohty M. Current
come and ther­apy deliv­ery in trans­plant-inel­i­gi­ble patients with mye­loma sta­tus of autol­o­gous stem cell trans­plan­ta­tion for mul­ti­ple mye­loma. Blood
(UK mye­loma research alli­ance risk pro­file): a devel­op­ment and val­i­da­tion Cancer J. 2019;9(4):44.
study. Lancet Haematol. 2019;6(3):e154-e166. 32. Mina R, Bringhen S, Wildes TM, Zweegman S, Rosko AE. Approach to
11. Milani P, Vincent Rajkumar S, Merlini G, et  al. N-ter­mi­nal frag­ment of the the older adult with mul­ti­ple mye­loma. Am Soc Clin Oncol Educ Book.
type-B natri­uretic pep­tide (NT-proBNP) con­trib­utes to a sim­ple new frailty 2019;93(39):500-518.
score in patients with newly diag­nosed mul­ti­ple mye­loma. Am J Hematol. 33. Dimopoulos MA, Moreau P, Terpos E, et al; EHA Guidelines Committee. Mul-
2016;91(11):1129-1134. tiple mye­loma: EHA-ESMO Clinical Practice Guidelines for diag­no­sis, treat­
12. Offidani M, Corvatta L, Polloni C, et  al. Assessment of vul­ner­a­bil­ity mea­ ment and fol­low-up. Ann Oncol. 2021;32(3):309-322.
sures and their effect on sur­vival in a real-life pop­u­la­tion of mul­ti­ple mye­ 34. Munshi PN, Vesole D, Jurczyszyn A, et  al. Age no bar: a CIBMTR anal­y­sis
loma patients reg­is­tered at Marche Region Multiple Myeloma Registry. Clin of elderly patients under­go­ing autol­o­gous hema­to­poi­etic cell trans­plan­ta­
Lymphoma Myeloma Leuk. 2012;12(6):423-432. tion for mul­ti­ple mye­loma. Cancer. 2020;126(23):5077-5087.
13. Fried LP, Tangen CM, Walston J, et al; Cardiovascular Health Study Collab- 35. Mian H, Mian OS, Rochwerg B, Foley R, Wildes TM. Autologous stem
orative Research Group. Frailty in older adults: evi­dence for a phe­no­type. cell trans­plant in older patients (age ≥ 65) with newly diag­nosed mul­ti­
J Gerontol A Biol Sci Med Sci. 2001;56(3):M146-M156. ple mye­ loma: a sys­ tem­atic review and meta-anal­ y­ sis. J Geriatr Oncol.
14. Benboubker L, Dimopoulos MA, Dispenzieri A, et al; FIRST Trial Team. Lena- 2020;11(1):93-99.
lidomide and dexa­meth­a­sone in trans­plant-inel­i­gi­ble patients with mye­ 36. Pawlyn C, Cairns D, Menzies T, et al. Autologous stem cell trans­plan­ta­tion is
loma. N Engl J Med. 2014;371(10):906-917. safe and effec­tive for fit older mye­loma patients: explor­atory results from
15. Facon T, Dimopoulos MA, Dispenzieri A, et al. Final anal­y­sis of sur­vival out­ the Myeloma XI trial [published online 3 Decem­ber 2020]. Haematologica.
comes in the phase 3 FIRST trial of up-front treat­ment for mul­ti­ple mye­ 37. Terpos E, Mikhael J, Hajek R, et  al. Management of patients with mul­ti­
loma. Blood. 2018;131(3):301-310. ple mye­loma beyond the clin­i­cal-trial set­ting: under­stand­ing the bal­ance
16. Stege CAM, Van Der Holt B, Dinmohamed AG, et al. Validation of the FIRST between effi­cacy, safety and tol­er­a­bil­ity, and qual­ity of life. Blood Cancer
sim­pli­fied frailty scale using the ECOG per­ for­
mance sta­ tus instead of J. 2021;11(2):40.
patient-reported activ­i­ties. Leukemia. 2020;34(7):1964-1966. 38. Fifer S, Galinsky J, Richard S. Myeloma patient value map­ping: a dis­crete
17. Engelhardt M, Ihorst G, Duque-Afonso J, et  al. Structured assess­ ment choice exper­i­ment on mye­loma treat­ment pref­er­ences in the UK. Patient
of frailty in mul­ ti­
ple mye­loma as a par­ a­
digm of indi­ vid­ u­
al­
ized treat­ Prefer Adherence. 2020;14:1283-1293.

Prakash Singh Shekhawat


Frailty in mul­ti­ple mye­loma  |  53
39. Saad A, Mahindra A, Zhang M-J, et al. Hematopoietic cell trans­plant comor- 48. Wildes TM, Anderson KC. Approach to the treat­ment of the older, unfit
bidity index is pre­dic­tive of sur­vival after autol­o­gous hema­to­poi­etic cell patient with mye­loma from diag­no­sis to relapse: per­spec­tives of a US
trans­plan­ta­tion in mul­ti­ple mye­loma. Biol Blood Marrow Transplant. hema­tol­o­gist and a geri­at­ric hema­tol­o­gist. Hematology Am Soc Hematol
2014;20(3):402-408.e1408e1. Educ Program. 2018;2018(1):88-96.
40. Shah N, Callander N, Ganguly S, et al; Amer­i­can Society for Blood and Mar- 49. Facon T, Niesvizky R, Mateos M-V, et  al. Efficacy and safety of carfilzo-
row Transplantation. Hematopoietic stem cell trans­plan­ta­tion for mul­ti­ple mib-based reg­i­mens in frail patients with relapsed and/or refrac­tory mul­ti­
mye­loma: guide­lines from the Amer­i­can Society for Blood and Marrow ple mye­loma. Blood Adv. 2020;4(21):5449-5459.
Transplantation. Biol Blood Marrow Transplant. 2015;21(7):1155-1166. 50. Dimopoulos MA, Stewart AK, Masszi T, et al. Carfilzomib, lenalidomide, and
41. Rosko A, Giralt S, Mateos M-V, Dispenzieri A. Myeloma in elderly patients: dexa­meth­a­sone in patients with relapsed mul­ti­ple mye­loma categorised
when less is more and more is more. Am Soc Clin Oncol Educ Book. by age: sec­ond­ary anal­y­sis from the phase 3 ASPIRE study. Br J Haematol.
2017;37:575-585. 2017;177(3):404-413.
42. Bazarbachi AH, Al Hamed R, Malard F, Harousseau J-L, Mohty M. Relapsed 51. Dimopoulos MA, Goldschmidt H, Niesvizky R, et al. Carfilzomib or borte-
refrac­tory mul­ti­ple mye­loma: a com­pre­hen­sive over­view. Leukemia. zomib in relapsed or refrac­tory mul­ti­ple mye­loma (ENDEAVOR): an interim
2019;33(10):2343-2357. over­all sur­vival anal­y­sis of an open-label, randomised, phase 3 trial. Lancet
43. Nathwani N, Bertamini L, Banerjee R, Gay F, Shah N, Krishnan A. When and Oncol. 2017;18(10):1327-1337.
how to treat relapsed mul­ti­ple mye­loma. Am Soc Clin Oncol Educ Book. 52. Mateos MV, Moreau P, Berenson JR, et  al. Once-weekly vs twice-weekly
2021;41:358-375. carfilzomib (K) dos­ing plus dexa­meth­a­sone (d) in patients with relapsed

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/46/1852113/46grant.pdf by guest on 13 December 2021


44. Faiman B, Bilotti E, Mangan PA, Rogers K; IMF Nurse Leadership Board. and refrac­tory mul­ti­ple mye­loma (RRMM): results of the ran­dom­ized phase 3
Steroid-asso­ci­ated side effects in patients with mul­ti­ple mye­loma: con­ study ARROW. J Clin Oncol. 2018;19(7):953-964.
sen­sus state­ment of the IMF Nurse Leadership Board. Clin J Oncol Nurs. 53. Moreau P, Stewart KA, Dimopoulos M, et al. Once-weekly (70 mg/m2) vs
2008;12(3, suppl):53-63. twice-weekly (56 mg/m2) dos­ing of carfilzomib in patients with relapsed
45. Rajkumar SV, Jaco­bus S, Callander NS, et al; Eastern Cooperative Oncology or refrac­tory mul­ti­ple mye­loma: a post hoc anal­y­sis of the ENDEAVOR,
Group. Lenalidomide plus high-dose dexa­meth­a­sone ver­sus lenalidomide ARROW, and CHAMPION-1 tri­als. Cancer Med. 2020;9(9):2989-2996.
plus low-dose dexa­meth­a­sone as ini­tial ther­apy for newly diag­nosed mul­ 54. Cornell RF, Ky B, Weiss BM, et al. Prospective study of car­diac events dur­
ti­ple mye­loma: an open-label randomised con­trolled trial. Lancet Oncol. ing proteasome inhib­i­tor ther­apy for relapsed mul­ti­ple mye­loma. J Clin
2010;11(1):29-37. Oncol. 2019;37(22):1946-1955.
46. Larocca A, Bonello F, Gaidano G, et al. Dose/sched­ule-adjusted Rd-R vs con­
tin­uo
­ us Rd for elderly, inter­me­di­ate-fit patients with newly diag­nosed mul­ti­
ple mye­loma. Blood. 2021;137(22):3027-3036.
47. Attal M, Richardson PG, Rajkumar SV, et al; ICARIA-MM study group. Isat-
uximab plus pomalidomide and low-dose dexa­meth­a­sone ver­sus poma-
lidomide and low-dose dexa­ meth­ as­one in patients with relapsed and
refrac­ tory mul­ ti­
ple mye­ loma (ICARIA-MM): a randomised, multicentre, © 2021 by The Amer­i­can Society of Hematology
open-label, phase 3 study. Lancet. 2019;394(10214):2096-2107. DOI 10.1182/hema­tol­ogy.2021000231

54  |  Hematology 2021  |  ASH Education Program


CLL: EXTENDING SURVIVAL

Upfront therapy: the case for continuous


treatment

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/55/1851744/55tam.pdf by guest on 13 December 2021


Constantine S. Tam
Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; University of Melbourne, Parkville, Victoria, Australia; and The Royal Melbourne
Hospital, Parkville, Victoria, Australia

Both BTKi and BCL2i are regarded as standards of care for frontline treatment of CLL. In this paper, I present the argu-
ments for favoring BTKi as initial therapy. Venetoclax-based regimens have the advantage of being fixed in duration, but
patients with select high-risk features may experience inferior PFS relative to those without high-risk features.

LEARNING OBJECTIVES
• Understand that both BTK inhibitors (BTKi) and the BCL2 antagonist venetoclax are effective treatments in
frontline CLL
• Understand that BTKis are active across the breadth of prognostic subgroups but are associated with long­term
toxicities and the need for indefnite therapy

median progression­free survival (PFS) of 52 months and


CLINICAL CASE 7­year overall survival (OS) of 55%,3 compared with historic
A 71­year­old man was observed for chronic lymphocytic median OS expectations of less than 1 year for similar pop­
leukemia (CLL) for 2 years and progressed with doubling ulations.4,5 Since then, new generations of BTKis, includ­
time of 6 months and bulky lymphadenopathy. He has ing more specifc covalent inhibitors with the potential for
stable hypertension, atrial fbrillation, and osteoarthri­ reduced side effects (eg, acalabrutinib,6 zanubrutinib7) and
tis. Prognostic workup showed del(17p) by fluorescence even newer “reversible” agents (eg, pirtobrutinib8), have
in situ hybridization, unmutated immunoglobulin heavy emerged. Multiple phase 3 studies have confrmed the
chain (IgHV), and TP53 mutation with complex karyotype. superiority of BTKi­based therapies over established com­
He lives in a rural location and travels 1.5 hours by car to parators in CLL, in both the frontline (1L) (RESONATE­2,9
see his treating oncologist. Should he receive upfront con­ ELEVATE­TN,10 ILLUMINATE,11 E1912,12 ALLIANCE13) and R/R
tinuous BTK inhibitor (BTKi) therapy or limited­duration settings (RESONATE,14 ASCEND15).
venetoclax­obinutuzumab? Although broadly effective and well tolerated as a class,
the paradigm for BTKi therapy is indefnite therapy due to
incomplete eradication of the CLL population, as manifest
BTKis and the paradigm for continuous therapy by low complete remission rates of approximately 25% to
Therapy for CLL in the chemotherapy era is traditionally 35% and approximately 10% in the 1L and R/R settings,3,11,16,17
limited in duration due to cumulative myelosuppression respectively, and largely absent chances of attaining unde­
and other side effects.1 This paradigm is now challenged by tectable minimal residual disease status (uMRD; defned as
the advent of targeted therapies with improved tolerance <1 CLL cell per 10,000 leukocytes). The addition of the anti­
profles. In this article, we discuss the advantages of contin­ CD20 rituximab to ibrutinib failed to improve PFS in 2 ran­
uous BTKi therapy relative to limited­duration alternatives. domized studies in CLL,13,18 although a slight PFS advantage
Eight years ago, the phase 1 report of the frst­in­class to the combination of acalabrutinib and obinutuzumab to
BTKi ibrutinib was published in the New England Journal acalabrutinib alone was seen in the ELEVATE­TN study.10,19
of Medicine.2 Ibrutinib was groundbreaking for patients Thus, the nature of BTKi­based therapy remains continuous
with relapsed/refractory (R/R) CLL, achieving prolonged and indefnite.

Prakash Singh Shekhawat


Upfront therapy: the case for continuous treatment | 55
The alter­na­tive: BCL2 inhib­i­tors and lim­ited-dura­tion Results of ibrutinib in 1L CLL with adverse geno­mics were even
ther­apy more favor­able. In the long-term fol­low-up of the National Insti­
Over the sim­i­lar period, the BCL2 inhib­i­tor venetoclax was devel­ tutes of Health phase 2 study of 34 patients with TP53 muta­tions
oped.20 Similar to ibrutinib, venetoclax showed remark­able sin­ receiv­ing 1L ibrutinib ther­apy, 6-year PFS was 61%.33 When pooled
gle-agent activ­ity in patients with heavily pretreated CLL, with with the results of 3 other 1L stud­ies (RESONATE-2, ECOG 1912,
median PFS of 30 months and 3-year OS of 71%.21 Different from and ILLUMINATE), 89 patients in total with TP53 aber­rant CLL were
BTKi, venetoclax is capa­ble of attaining uMRD as monotherapy in treated 1L on ibrutinib-based reg­i­mens; in aggre­gate, a 4-year
R/R CLL (27%–30% in blood, 16% in mar­row21,22). PFS of 79% was reported.34 Balancing these favor­able reports, the
The obser­va­tion that some patients in uMRD remis­sions were ALLIANCE 1L study of BR vs ibrutinib vs ibrutinib-rituximab found
­able to main­tain dura­ble responses off ther­apy23,24 led to the design a less favor­able PFS for patients with del(17p), with a 2-year PFS
of time-lim­ited reg­i­mens such as the 24-month venetoclax-rituximab of approx­i­ma­tely 75% for ibrutinib reg­i­mens; the cor­re­spond­ing
(Ven-R) reg­ i­
men in the MURANO study of R/R CLL25,26 and the 2-year PFS for patients with CK was approx­i­ma­tely 90%.13
12-month venetoclax-obinutuzumab (Ven-O) reg­im ­ en in the CLL14 Next-gen­ er­

tion cova­ lent BTKis such as acalabrutinib and
study of 1L CLL.27 These reg­i­mens have the appeal of pro­vid­ing a zanubrutinib show improved tol­er­ance com­pared with ibrutinib in

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/55/1851744/55tam.pdf by guest on 13 December 2021


rel­a­tively short expo­sure to ther­a­peu­tic agents and their atten­dant head-to-head stud­ies.35-37 In one of the larg­est pro­spec­tive series of
side effects. patients with del(17p) reported to date (SEQUOIA Arm C, n = 109),
However, emerg­ ing data sug­ gest that lim­ ited-dura­ tion 1L treat­ment with zanubrutinib resulted in and over­all response
venetoclax reg­i­mens may not be suit­able in all­types of CLL, with rate of 95% and an 18-month PFS of 91%.38 Importantly, for those
par­tic­ul­ar con­cern for patients with geno­mic high-risk dis­ease. In patients with the highest risk cat­e­gory of del(17p) and con­com­i­
the MURANO study, com­pared with the whole-study uMRD rate tant CK, a favor­able 18-month PFS of 94% was maintained.38 Simi­
of 84% fol­low­ing Ven-R, uMRD rates were 42% for patients with larly favor­able 1L results for TP53 aber­rant CLL were reported in the
del(17p), 55% for com­plex kar­yo­type (CK), 48% for TP53 muta­ ELEVATE-TN study of acalabrutinib vs acalabrutinib-obinutuzumab
tion, and 40% to 43% for muta­tions in NOTCH1, XPO1, or BRAF.26 vs chlorambucil-obinutuzumab, with an 18-month PFS of more
Among patients who responded to Ven-R, 25 patients have than 80% for acalabrutinib arms in patients with del(17p) and/or
been rechallenged after ini­tial remis­sions of 12 or more months.28 TP53 muta­tion.10
Genomic ana­ly­ses com­par­ing serial sam­ples taken at base­line Finally, all­BTKi stud­ies to date showed no dis­ad­van­tage for
(before Ven-R) and at retreatment showed selec­tion for del(17p) patients with IgHV-unmutated status, with con­sis­tently overlap­
and CK, and patients car­ry­ing these clones were highly unlikely ping PFS curves irrespective of set­ting (1L or R/R), TP53 sta­tus,
to reach uMRD from venetoclax reexposure.28 or agent stud­ied.10-13,15,17,31,33,38
Similarly, in updated data sets of CLL14, patients with IgHV- In aggre­gate, avail­­able data across the cova­lent BTKi expe­ri­
unmutated sta­tus, del(17p), or TP53 muta­tions expe­ri­enced shorter ence suggested that the impact of adverse geno­mics (par­tic­u­
PFS fol­low­ing Ven-O (haz­ard ratio [HR], 2.14, 3.19, and 2.42, respec­ larly TP53 aber­ra­tions) is lim­ited mainly to patients with heavily
tively), and del(17p) was addi­tion­ally asso­ci­ated with reduced OS pretreated CLL, with largely favor­able out­comes observed in
fol­low­ing Ven-O (HR, 3.52).29 Interestingly, clonal regrowth rates phase 2 to 3 stud­ies of patients treated in the 1L.10,13,33,34,38 Con­
were faster in patients with IgHV-unmutated CLL,30 suggesting ceptually, one can argue the­o­ret­i­cally that the patients with the
that con­tin­u­ous “sup­pres­sive” BTKi ther­apy may be pref­er­a­ble highest geno­mic risks (ie, del(17p) with CK) may be best served
in these patients. by indef­i­nite sup­pres­sion of the CLL clone at a time early in the
treat­ment sequence rather than being chal­lenged by inter­mit­
tent ther­a­pies that may give rise to subclonal com­plex­ity later in
BTKis are broadly effec­tive in high geno­mic risk CLL
the dis­ease course.39
The ear­li­est expe­ri­ence with ibrutinib in the PCYC-1102 phase
1b/2 trial suggested that del(17p) and/or CK may asso­ci­ated
with infe­rior out­comes: com­pared with the whole-study R/R Other advan­tages of con­tin­u­ous BTKi ther­apy
CLL median PFS (mPFS) of 52 months, mPFS was 26 months for In gen­eral, BTKi ther­apy is straight­for­ward to com­mence and
del(17p) and 31 months for CK.3 However, the adverse risk in the broadly appli­ca­ble across a range of patients, includ­ing those
CK sub­group was entirely attrib­ut­­able to over­lap with del(17p), with mild to mod­er­ate organ impair­ments, with a low risk of
as patients with CK alone (with­out del(17p)) had a favor­able mPFS tumor lysis syn­drome.10-13,15,17,31,33,38 Therefore, BTKis may be par­tic­
of 88 months. In com­par­ing the results of this study with those u­larly suit­able in patients with high tumor bur­den dis­ease, those
reported in sub­se­quent BTKi stud­ies, it is impor­tant to note that with renal impair­ment, or those sit­ua ­ ­tions (eg, patients resid­ing
the patients in PCYC-1102 were very heavily pretreated (59% had in rural com­mu­ni­ties) where real-time mon­i­tor­ing for tumor lysis
≥4 prior lines of ther­apy3) and may be less rep­re­sen­ta­tive of pa­ syndrome for venetoclax step-up may be logis­ti­cally dif­fi­cult.
tients seen in clinic today. Indeed, a clear rela­tion­ship between Their rel­a­tively broader “cov­er­age” against geno­mic high-risk
increas­ing prior lines of ther­apy and infe­rior PFS has been clearly CLL also makes them eas­ier agents to admin­is­ter in com­mu­nity-
established across mul­ti­ple ibrutinib stud­ies.3,31,32 based clin­ics where advanced test­ing for CLL geno­mics may be
The phase 3 RESONATE study com­pared ibrutinib and ofatu­ less avail­­able. Indeed, emerg­ing data from real-world reg­is­tries
mumab in a less heavily pretreated R/R CLL pop­u­la­tion.14 Del(17p), show low rates of test­ing for TP53 and other high-risk fea­tures in
TP53 muta­tion, and CK were pres­ent in 32%, 51%, and 25% of patients with CLL man­aged in the United States.40
patients on the ibrutinib arm, respec­tively.31 Patients with del(17p)
and/or TP53 muta­tion con­tin­ued to show a slight dis­ad­van­tage Drawbacks of con­tin­u­ous BTKi ther­apy
(mPFS of 41 months) com­pared with other sub­groups, but no PFS Continuous BTKi ther­a­pies do have their price in terms of poten­
dif­fer­ences were observed in patients with and with­out CK.31 tial for emer­gence of resis­tance, side effect bur­den on patient

56  |  Hematology 2021  |  ASH Education Program


qual­ity of life, and eco­nomic cost. Addressing the poten­tial of 6. Byrd JC, Harrington B, O’Brien S, et al. Acalabrutinib (ACP-196) in relapsed
resis­tance emer­gence, it is the­o­ret­i­cally pos­si­ble that inter­mit­ chronic lym­pho­cytic leu­ke­mia. N Engl J Med. 2016;374(4):323-332.
7. Tam CS, Trotman J, Opat S, et al. Phase 1 study of the selec­tive BTK inhib­
tent expo­sure to a drug may have a lower risk of induc­ing treat­ i­tor zanubrutinib in B-cell malig­nan­cies and safety and effi­cacy eval­u­a­tion
ment resis­tance com­pared with long-term expo­sure. However, in CLL. Blood. 2019;134(11):851-859.
as we have seen in the early MURANO retreatment expe­ri­ence,28 8. Mato AR, Shah NN, Jurczak W, et al. Pirtobrutinib in relapsed or refrac­tory
even fixed-dura­tion venetoclax expo­sure appears to induce the B-cell malig­nan­cies (BRUIN): a phase ½ study. Lancet. 2021;397(10277):892-
901.
emer­gence of geno­mic high-risk, treat­ment-resis­tant clones. To
9. Burger JA, Tedeschi A, Barr PM, et al; RESONATE-2 Investigators. Ibrutinib
date, there are no data on whether lim­ited-dura­tion BTKi expo­ as ini­tial ther­apy for patients with chronic lym­pho­cytic leu­ke­mia. N Engl
sure (eg, in tri­als com­bin­ing ibrutinib and venetoclax)41 may have J Med. 2015;373(25):2425-2437.
less impact on the sub­ se­
quent emer­ gence of BTKi-resis­ tant 10. Sharman JP, Egyed M, Jurczak W, et al. Acalabrutinib with or with­out obinu­
muta­tions. tuzumab ver­sus chlorambucil and obinutuzmab for treat­ment-naive chronic
lym­pho­cytic leu­kae­mia (ELEVATE TN): a randomised, con­trolled, phase 3
Many patients on con­tin­u­ous BTKi expe­ri­ence side effects that trial. Lancet. 2020;395(10232):1278-1291.
may be tol­er­a­ble in the short term but cumu­la­tively rep­re­sent a 11. Moreno C, Greil R, Demirkan F, et al. Ibrutinib plus obinutuzumab ver­sus
sub­stan­tial impact on their qual­ity of life in the long term.42 Real- chlorambucil plus obinutuzumab in first-line treat­ ment of chronic lym­

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/55/1851744/55tam.pdf by guest on 13 December 2021


world stud­ies suggested that up to 41% of patients discontinue pho­cytic leu­kae­mia (iLLUMINATE): a multicentre, randomised, open-label,
phase 3 trial. Lancet Oncol. 2019;20(1):43-56.
ibrutinib after a median of 17 months of fol­low-up, largely due
12. Shanafelt TD, Wang XV, Kay NE, et al. Ibrutinib-rituximab or chemoimmuno­
to side effects.43 Balanced against these chronic tox­ic­ity con­sid­ therapy for chronic lym­pho­cytic leu­ke­mia. N Engl J Med. 2019;381(5):432-
er­ations, it is note­wor­thy that all 3 recently reported head-to- 443.
head com­par­i­sons of ibrutinib vs acalabrutinib or zanubrutinib 13. Woyach JA, Ruppert AS, Heerema NA, et  al. Ibrutinib reg­i­mens ver­sus
were con­cor­dant in reporting reduced car­dio­vas­cu­lar and mus­ chemoimmunotherapy in older patients with untreated CLL. N Engl J Med.
2018;379(26):2517-2528.
cu­lo­skel­e­tal com­pli­ca­tions in favor of the sec­ond-gen­er­a­tion 14. Byrd JC, Brown JR, O’Brien S, et al. Ibrutinib ver­sus ofatumumab in pre­vi­
agent.35-37 Therefore, the tol­er­ance pro­file of indef­i­nite BTK inhi­bi­ ously treated chronic lym­phoid leu­ke­mia. N Engl J Med. 2014;371(3):213-
tion in the sec­ond-gen­er­a­tion BTKi era is likely to be sub­stan­tially 223.
improved com­pared with that of the ibrutinib era. 15. Ghia P, Pluta A, Wach M, et al. ASCEND: phase III, ran­dom­ized trial of aca­
labrutinib ver­sus idelalisib plus rituximab or bendamustine plus rituximab
in relapsed or refrac­tory chronic lym­pho­cytic leu­ke­mia. J Clin Oncol. 2020;
38(25):2849-2861.
16. O’Brien SM, Jaglowski S, Byrd JC, et  al. Prognostic fac­tors for com­plete
response to ibrutinib in patients with chronic lym­pho­cytic leu­ke­mia: a
CLINICAL CASE (Con­t in­u ed) pooled anal­y­sis of 2 clin­i­cal tri­als. JAMA Oncol. 2018;4(5):712-716.
After dis­cus­sions between the phy­si­cian and patient, our 71-year- 17. Burger JA, Barr PM, Robak T, et al. Long-term effi­cacy and safety of first-line
old patient was started on front­line BTKi due to his rural loca­tion ibrutinib treat­ment for patients with CLL/SLL: 5 years of fol­low-up from the
phase 3 RESONATE-2 study. Leukemia. 2020;34(3):787-798.
and poor-risk geno­mic fea­tures. Acalabrutinib was cho­sen over 18. Burger JA, Sivina M, Jain N, et  al. Randomized trial of ibrutinib vs ibruti­
ibrutinib due to a lower risk of car­dio­vas­cu­lar toxicities. nib plus rituximab in patients with chronic lym­pho­cytic leu­ke­mia. Blood.
2019;133(10):1011-1019.
19. Sharman JP, Egyed M, Jurczak W, et al. Acalabrutinib±obinutuzumab ver­sus
obinutuzumab  +  chlorambucil in treat­ment-naïve chronic lym­pho­cytic leu­
Conflict-of-inter­est dis­clo­sure ke­mia: ele­vate-TN four-year fol­low up. J Clin Oncol. 2021;39(15, suppl):7509.
Constantine S. Tam: Research funding from Janssen, AbbVie, and 20. Roberts AW, Davids MS, Pagel JM, et al. Targeting BCL2 with venetoclax
Beigene; hon­o­raria from Janssen, Pharmacyclics, Beigene, and in relapsed chronic lym­pho­cytic leu­ke­mia. N Engl J Med. 2016;374(4):311-
AbbVie. 322.
21. Roberts AW, Ma S, Kipps TJ, et al. Efficacy of venetoclax in relapsed chronic
lym­pho­cytic leu­ke­mia is influ­enced by dis­ease and response var­i­ables.
Off-label drug use Blood. 2019;134(2):111-122.
Constantine S. Tam: none discussed. 22. Stilgenbauer S, Eichhorst B, Schetelig J, et al. Venetoclax for patients with
chronic lym­pho­cytic leu­ke­mia with 17p dele­tion: results from the full pop­
u­la­tion of a phase II piv­otal trial. J Clin Oncol. 2018;36(19):1973-1980.
Correspondence 23. Seymour JF, Ma S, Brander DM, et al. Venetoclax plus rituximab in relapsed
Constantine S. Tam, Department of Haematology, Peter MacCal­ or refrac­tory chronic lym­pho­cytic leu­kae­mia: a phase 1b study. Lancet
lum Cancer Centre, 305 Grattan Street, Melbourne 3050, Victo­ Oncol. 2017;18(2):230-240.
ria, Australia; e-mail: constantine​­.tam@petermac​­.org. 24. Ma S, Seymour JF, Brander DM, et al. Efficacy of venetoclax plus rituximab
for relapsed CLL: 5-year fol­low-up of con­tin­u­ous or lim­ited-dura­tion ther­
apy. Blood. 2021;138(10):836-846.
References 25. Seymour JF, Kipps TJ, Eichhorst B, et al. Venetoclax-rituximab in relapsed or
1. Tam CS, O’Brien S, Wierda W, et al. Long-term results of the fludarabine, refrac­tory chronic lym­pho­cytic leu­ke­mia. N Engl J Med. 2018;378(12):1107-
cyclo­phos­pha­mide, and rituximab reg­i­men as ini­tial ther­apy of chronic 1120.
lym­pho­cytic leu­ke­mia. Blood. 2008;112(4):975-980. 26. Kater AP, Wu JQ , Kipps T, et  al. Venetoclax plus rituximab in relapsed
2. Byrd JC, O’Brien S, James DF. Ibrutinib in relapsed chronic lym­pho­cytic leu­ chronic lym­ pho­cytic leu­ ke­
mia: 4-year results and eval­ u­a­
tion of impact
ke­mia. N Engl J Med. 2013;369(13):1278-1279. of geno­mic com­plex­ity and gene muta­tions from the MURANO phase III
3. Byrd JC, Furman RR, Coutre SE, et al. Ibrutinib treat­ment for first-line and study. J Clin Oncol. 2020;38(34):4042-4054.
relapsed/refrac­tory chronic lym­pho­cytic leu­ke­mia: final anal­y­sis of the piv­ 27. Fischer K, Al-Sawaf O, Bahlo J, et al. Venetoclax and obinutuzumab in patients
otal phase Ib/II PCYC-1102 study. Clin Cancer Res. 2020;26(15):3918-3927. with CLL and coexisting con­di­tions. N Engl J Med. 2019;380(23):2225-2236.
4. Keating MJ, O’Brien S, Kontoyiannis D, et al. Results of first sal­vage ther­apy 28. Wu JQ , Seymour JF, Eichhorst B, et al. Impact of unfa­vor­able genet­ics on
for patients refrac­tory to a fludarabine reg­i­men in chronic lym­pho­cytic leu­ min­i­mal resid­ual dis­ease (MRD) response to venetoclax+rituximab retreat­
ke­mia. Leuk Lymphoma. 2002;43(9):1755-1762. ment in relapsed or refrac­tory CLL: phase 3 MURANO substudy. Presented
5. Tam CS, O’Brien S, Lerner S, et  al. The nat­ u­ral his­
tory of fludarabine- at: EHA2021 Virtual Congress; June 9-12, 2021. Poster EP599.
refrac­tory chronic lym­pho­cytic leu­ke­mia patients who fail alemtuzumab or 29. Tausch E, Schneider C, Yosifov D, Robrecht S, Zhang C. Genetic mark­ers
Prakash Singh Shekhawat
have bulky lymph­ade­nop­a­thy. Leuk Lymphoma. 2007;48(10):1931-1939. and out­come with front line obinutuzumab plus either chlorambucil or

Upfront ther­apy: the case for con­tin­u­ous treat­ment  |  57


venetoclax—updated anal­y­sis of the CLL14 trial. Presented at: EHA2021 vs ibrutinib in patients with relapsed/refrac­tory CLL/SLL. Presented at:
Virtual Congress; June 9-12, 2021. Poster S144. EHA2021 Virtual Congress; June 9-12, 2021. Poster LB1900.
30. Al-Sawaf O, Zhang C, Robrecht S, et  al. Clonal dynam­ics after veneto­ 38. Tam CS, Robak T, Ghia P, et al. Zanubrutinib monotherapy for patients with
clax-obinutuzumab ther­apy: novel insights from the ran­dom­ized, phase 3 treat­ment naïve chronic lym­pho­cytic leu­ke­mia and 17p dele­tion. Haema-
CLL14 trial. Blood. 2020;136(suppl 1):22-23. tologica. 2020;106(9):2354-2363.
31. Munir T, Brown JR, O’Brien S, et al. Final anal­y­sis from RESONATE: up to six 39. Landau DA, Tausch E, Taylor-Weiner AN, et al. Mutations driv­ing CLL and
years of fol­low-up on ibrutinib in patients with pre­vi­ously treated chronic their evo­ lu­
tion in pro­
gres­ sion and relapse. Nature. 2015;526(7574):525-
lym­pho­cytic leu­ke­mia or small lym­pho­cytic lym­phoma. Am J Hematol. 530.
2019;94(12):1353-1363. 40. Mato A, Nabhan C, Kay NE, et al. prog­nos­tic test­ing pat­terns and out­comes
32. Barr PM, Tedeschi A, Munir T, et al. Using ibrutinib in ear­lier lines of treat­ment of chronic lym­pho­cytic leu­ke­mia patients strat­i­fied by fluo­res­cence in situ
results in bet­ter out­comes for patients with chronic lym­pho­cytic leu­ke­mia/ hybrid­iza­tion/cyto­ge­net­ics: a real-world clin­i­cal expe­ri­ence in the con­nect
small lym­pho­cytic lym­phoma. Blood. 2019;134(suppl 1):3054. CLL reg­is­try. Clin Lymphoma Myeloma Leuk. 2018;18(2):114-124.e2124e2.
33. Ahn IE, Tian X, Wiestner A. Ibrutinib for chronic lym­pho­cytic leu­ke­mia with 41. Wierda WG, Tam CS, Allan JN, et  al. Ibrutinib (Ibr) plus venetoclax (Ven)
TP53 alter­ations. N Engl J Med. 2020;383(5):498-500. for first-line treat­ment of chronic lym­pho­cytic leu­ke­mia (CLL)/small lym­
34. Allan JN, Shanafelt T, Wiestner A, et al. long-term effi­cacy of first-line ibru­ pho­cytic lym­phoma (SLL): 1-year dis­ease-free sur­vival (DFS) results from
tinib treat­ment for chronic lym­pho­cytic leu­ke­mia (CLL) with 4 years of the MRD cohort of the phase 2 CAPTIVATE study. Blood. 2020;136(suppl 1):
fol­low-up in patients with TP53 aber­ra­tions (del(17p) or TP53 muta­tion): a 16-17.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/55/1851744/55tam.pdf by guest on 13 December 2021


pooled anal­y­sis from 4 clin­i­cal tri­als. Blood. 2020;136(suppl 1):23-24. 42. Lasica M, Tam CS. Management of ibrutinib toxicities: a prac­ti­cal guide.
35. Tam CS, Opat S, D’Sa S, et al. A ran­dom­ized phase 3 trial of zanubrutinib Curr Hematol Malig Rep. 2020;15(3):177-186.
vs ibrutinib in symp­tom­atic Waldenström mac­ro­glob­u­li­ne­mia: the ASPEN 43. Mato AR, Nabhan C, Thompson MC, et al. Toxicities and out­comes of 621
study. Blood. 2020;136(18):2038-2050. ibrutinib-treated chronic lym­ pho­cytic leu­ke­ mia patients in the United
36. Byrd JC, Hillmen P, Ghia P, et al. First results of a head-to-head trial of aca­ States: a real-world anal­y­sis. Haematologica. 2018;103(5):874-879.
labrutinib vs ibrutinib in pre­vi­ously treated CLL. J Clin Oncol. 2021;39(15,
suppl):7500.
37. Hillmen P, Eichhort B, Brown JR, Lamanna N, O’Brien S. First interim anal­y­ © 2021 by The Amer­i­can Society of Hematology
sis of ALPINE study: results of a phase 3 ran­dom­ized study of zanubrutinib DOI 10.1182/hema­tol­ogy.2021000232

58  |  Hematology 2021  |  ASH Education Program


CLL: EXTENDING SURVIVAL

Frontline treatment in CLL: the case


for time-limited treatment

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/59/1851402/59levy.pdf by guest on 13 December 2021


Vincent Lévy,1 Alain Delmer,2 and Florence Cymbalista3
Département de Recherche Clinique, Hôpital Avicenne AP­HP, Université Sorbonne Paris Nord, Bobigny, France, and INSERM CRESS­UMR 1153,
1

Hôpital Saint Louis, Paris, France; 2Hematology Department, Reims University Hospital and Reims Champagne Ardenne University, Reims, France;
and 3Hematology Biology, Hôpital Avicenne, AP­HP, Université Sorbonne Paris Nord, Bobigny, France, and INSERM UMR 978, Bobigny, France

Over the last decade, the advent of Bruton tyrosine kinase inhibitors (BTKi) has profoundly modified the therapeutic
strategy in chronic lymphocytic leukemia (CLL), introducing the concept of treatment until progression. Initially, the
bcl-2 inhibitor venetoclax (VEN) was used as a single agent and then was rapidly combined in VEN-based regimens asso-
ciated with either anti-CD20 or with BTKi. These regimens yielded a high rate of complete remission, leading to their use
as a fixed duration treatment. The decision between continuous treatment with BTKi and VEN-based combinations relies
mostly on comorbidities, comedications, and patient/physician preferences. Notably, with BTKi, cardiovascular comor-
bidities, hypertension, and potential pharmacological interactions should be carefully evaluated. On the other hand, the
risk of tumor lysis syndrome with VEN should be monitored at treatment initiation. TP53 alteration and IGHV mutational
status should also be assessed, as they remain important for therapeutic decisions. Fit patients with a TP53 wild type
and IGHV-mutated CLL may still benefit from fludarabine-cyclophosphamide-rituximab chemoimmunotherapy (CIT), as
it may result in a very long remission duration. VEN-based treatments are well tolerated, and no additional toxicity has
been observed when combined with anti-CD20 or BTKi. The 1-year fixed-duration association of VEN plus obinutuzumab
was evaluated in frontline for older adult patients. Nonetheless, considering the favorable outcome, an extension of
indication for fit younger patients is expected. The association of VEN and BTKi is promising, even if the follow-up is still
short. It is currently being tested against CIT, BTKi continuous treatment, and VEN plus anti-CD20.

LEARNING OBJECTIVES
• Recognize the important biological pretherapeutic parameters for allocating CLL patients to optimal frontline
treatment
• Review the indications of time­limited treatment for the various categories of treatment­naive CLL patients
• Review the various fixed­duration combination therapies and the major trials testing the time­limited strategies
• Be aware of future developments that will allow a more personalized therapeutic approach

Introduction plete response and undetectable minimal residual disease


The concept of time­limited treatment is obviously not (uMRD), and it can be discontinued safely after optimal
new in chronic lymphocytic leukemia (CLL) since up until response is achieved.3
the arrival of BCR inhibitors, treatment relied mainly on
chemotherapy and was therefore of limited duration. In
recent years, Bruton tyrosine kinase inhibitors (BTKi) have
been shown to be superior overall to chemoimmunother­
apy (CIT) in the first­line treatment of CLL, but they must CLINICAL CASE 1
be given continuously until progression as they have only Mr. V., who is 58, is fit and works as an engineer. He has a history
a suspensive effect on the disease.1,2 This has led to new of coronary thrombosis dating from 5 years ago, is on acetyl­
concerns about compliance, quality of life, and cost. The salicylic acid, and takes amlodipine for high blood pressure.
BCL2 inhibitor venetoclax (VEN), either alone or combined At diagnosis of CLL, his blood count showed 23×109/L lym­
with other agents, is likely to yield high rates of both com­ phocytes and no cytopenia. An immunophenotype revealed
Prakash Singh Shekhawat
Up­front therapy: time­limited treatment | 59
Table 1. Recommended mark­ers for the diag­no­sis of CLL (Euro­pean Research Initiative on CLL rec­om­men­da­tions)

Minimally recommended Other impor­tant mark­ers


CD19 Intracellular Ig light chains kappa and lambda (if absence
CD5 of mem­brane staining)
Ig light chains kappa and lambda (mem­brane staining) CD81
CD23 CD43
CD79b and/or CD22 ROR1
CD20 CD38
FMC7 CD10
CD200
Ig, immu­no­glob­u­lin.

clonal B cells with low CD20 expres­sion har­bor­ing pos­i­tive CD5 55 mL/min, and the direct anti­body test was neg­a­tive. He was

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/59/1851402/59levy.pdf by guest on 13 December 2021


expression, CD23 and CD200hi, low kappa light chain, and CD79b. started in 2010 on 6 courses of bendamustine-rituximab (BR).
CD38 was not expressed on CLL cells. Clinical exam­i­na­tion found
centimetric cer­vi­cal and axil­lary lymph nodes and no spleno­meg­
aly. Five years later in 2010, Mr. V. was still fit, but his CLL had pro­ What if we had to do it again today?
gressed with lymph­ade­nop­a­thy from 3 to 5 cm (cer­vi­cal, axil­lary, Indication for treat­ment
and ingui­nal) and a spleno­meg­aly 6 cm below the cos­tal mar­gin. It is impor­tant to keep in mind that the ther­a­peu­tic options dis­
He had lost 3 kg and expe­ri­enced night sweats. His blood counts cussed here are intended for bona fide CLL and that ade­quate
were as fol­ lows: lym­ pho­ cytes 120 × 
109/L; hemo­glo­bin 135 g/L; phenotyping (Table 1) should be performed at diag­no­sis.
plate­lets 176 × 10 /L. Biological param­e­ters were as fol­lows: esti­
9 Indications for treat­ment ini­ti­a­tion according to the Interna­
mated glo­mer­u­lar fil­tra­tion rate (eGFR), 70 mL/min; del13q dele­tion tional Workshop on Chronic Lymphocytic Leukemia have not
and no 17p dele­tion; mutated IGHV sta­tus with VH 3-30 with 95% dra­mat­i­cally changed over the years. As exem­pli­fied in these 2
homol­ogy; and hypogammaglobulinemia at 5 g/L. In 2010 he was cases, spe­cific treat­ment is indi­cated when patients show signs
started on 6 courses of an oral fludarabine-cyclo­phos­pha­mide- of CLL pro­gres­sion. The pro­gres­sion cri­te­ria have been very
rituximab (FCR) reg­i­men. clearly defined by the International Workshop on Chronic Lym­
phocytic Leukemia and are sum­ma­rized in Table 2.4

Pretreatment assess­ment and pre­ven­tive mea­sures


International and national soci­e­ties have published com­mon rec­
om­men­da­tions for pre­treat­ment assess­ments, includ­ing his­tory,
CLINICAL CASE 2 phys­i­cal exam­i­na­tion, and stan­dard bio­log­i­cal tests (Table 3).4-6
Mr. F. is 72 years old, retired, and a pre­vi­ous bank employee. He The cur­rent pretherapeutic workup includes the pre­vi­ous rec­
is a heavy smoker with type 2 dia­be­tes and dyslipidemia and om­men­da­tions of the CIT era, but some new assess­ments have
takes biguanide and atorvastatin. He was diag­nosed with CLL been added.
in 2009 with a char­ac­ter­is­tic phe­no­type. He rap­idly progressed Whenever targeted agents are con­ sid­ered, nota­ bly BTKi,
into active dis­ease, with mul­ti­ple lymph­ade­nop­a­thy, spleno­ car­dio­vas­cu­lar comorbidities, hyper­ten­sion, and comedications
meg­aly, and night sweats. His lym­pho­cyte dou­bling time was should be care­fully assessed.7 The pre­ven­tion of tumor lysis syn­
6 months, and blood counts showed hemo­glo­bin 110 × 109/L; drome is well established and should be applied to any VEN-
lym­pho­cytes: 130  ×  109/L; and plate­lets: 90 × 109/L. He had VH1- based strat­egy.8
69 IGHV with 100% homol­ ogy and a nor­ mal kar­yo­ type and Patients with CLL are at risk of infec­tion with targeted agents as
fluo­res­cence in situ hybrid­iza­tion (FISH) anal­y­sis. His eGFR was well, and all­pos­si­ble pre­ven­tive mea­sures should be under­taken

Table 2. Criteria for treat­ment ini­ti­a­tion

•  Progressive BM fail­ure with the devel­op­ment or aggra­va­tion of ane­mia and/or throm­bo­cy­to­pe­nia.


•  Massive or pro­gres­sive or symp­tom­atic spleno­meg­aly.
•  Significantly enlarged or symp­tom­atic or pro­gres­sive lymph­ade­nop­a­thies.
• Progressive lym­pho­cy­to­sis, with an increase of more than 50% over a period of 2 months or an LDT of less than 6 months. In patients with an ini­tial
lym­pho­cyte count <30 × 109/L, LDT alone should not be used as a sin­gle param­e­ter to decide treat­ment ini­ti­a­tion and should be interpreted in the
over­all clin­i­cal con­text.
•  Autoimmune ane­mia and/or throm­bo­cy­to­pe­nia that is poorly respon­sive to cor­ti­co­ste­roids or other stan­dard ther­a­pies.
•  The pres­ence of con­sti­tu­tive symp­toms as defined by 1 or more of the fol­low­ing signs or symp­toms related to the dis­ease:
  Unintentional weight loss of 10% or more in the pre­vi­ous 6 months,
sig­nif­i­cant fatigue (ECOG PS 2 or worse; inabil­ity to per­form usual activ­i­ties),
fever over 38.0 °C for 2 weeks or more with­out signs of infec­tion, and
night sweats last­ing more than a month with no sign of infec­tion.
At least one of the cri­te­ria must be fulfilled.

60  |  Hematology 2021  |  ASH Education Program


Table 3. Pretreatment eval­u­a­tion

Routine prac­tice Clinical tri­als spe­cific


Clinical eval­u­a­tion
  ECOG PS Always
  Constitutive symp­toms Always
  Clinical exam­i­na­tion Always
Cardiovascular comorbidities Always
Comedications Always
Biological stan­dard tests
 Complete blood count and retic­u­lo­cyte Always
count

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/59/1851402/59levy.pdf by guest on 13 December 2021


  LDH, hap­to­glo­bin and direct anti­glob­u­lin test Always
 Serum cre­at­i­nine level and glo­mer­u­lar fil­tra­ Always
tion rate
  Transaminases, bil­i­ru­bin, GGT Always
  LDH and beta-2 microglobulin Always
  Serum pro­tein elec­tro­pho­re­sis Always
  HIV, B and C hep­a­ti­tis serol­ogy Always
 Cryopreservation of blood cells (tumor Desirable Always
library)
Genetics
 Karyotype Desirable Always
 FISH
  17p dele­
tion Always
  11q dele­
tion Desirable Always
  13q dele­
tion Desirable Always
   Trisomy 12 Desirable Always
IGHV muta­tional sta­tus Always
TP53 muta­tions by NGS Always
NGS of recur­rent muta­tions NGI Desirable
Imaging
CT scan of chest, abdo­men, and pel­vis Desirable Recommended
Preventive mea­sures
  Vaccination against pneu­mo­coc­cus Always
  Vaccination against influ­enza (yearly) Always
  Vaccination against SARS-CoV-2 Always
CT, com­puted tomog­ra­phy; GGT, gamma-glutamyl trans­fer­ase; LDH, lac­tate dehy­dro­ge­nase; NGI, not generally indicated.

ated with resis­tance to CIT and remain an unfa­vor­able prog­nos­


before starting ther­apy, nota­bly vac­ci­na­tion (pneu­mo­coc­cus and
annu­ally for influ­enza).9 SARS-CoV-2 vac­ci­na­tion should be per­tic fac­tor with targeted agents.2,3,11,12 It is there­fore man­da­tory to
formed, pref­er­a­bly before starting ther­apy, par­tic­u­larly if the patient
test for both these alter­ations.
is to receive anti-CD20 or BTKi.10 In the 2 cases illus­trated here, in 2010 only 17p FISH was avail­­
In gen­eral, a body com­puted tomog­ra­phy scan may be help­ able. It should be stressed that FISH alone detects only half of
ful to assess the tumor load and risk of tumor lysis syn­drome, TP53 alter­ations, as in approx­i­ma­tely 50% of cases only a TP53
mainly before treat­ment with the BCL2 inhib­i­tor VEN.5 muta­tion is pres­ent and there­fore not iden­ti­fied by FISH anal­y­sis.13
The fol­low­ ing tests have become impor­ tant for deci­Nowadays, sequenc­ing anal­y­sis of TP53 is required before the
sion-
mak­ing. ini­ti­a­tion of ther­apy.4,14 Next-gen­er­a­tion sequenc­ing (NGS) is rap­
idly replacing Sanger sequenc­ing, as it allows the detec­tion of
TP53  Alteration of TP53 func­tion can be medi­ated by either 17p small subclones. In our obser­va­tions, both patients were tested
Prakash
dele­tion, TP53 muta­tion, or both. These Singh
alter­ations are Shekhawat
asso­ci­ ret­ro­spec­tively for TP53 muta­tions and were neg­a­tive.

Up-front ther­apy: time-lim­ited treat­ment  |  61


IGHV  The var­i­able region of immu­no­glob­u­lin heavy chain gene ATM,21 and some of them have been asso­ci­ated with expo­nen­tial
(IGHV) muta­tional sta­tus, which until recently was only indi­cated growth.22 This expanding knowl­edge about growth dynam­ics in
in clin­ i­
cal tri­als, has become an impor­ tant deci­ sion-mak­ing indi­vid­ual patients will be help­ful for predicting the course of the
param­ e­ ter and must be performed before front­ line ther­
apy.4 dis­ease and informing ther­apy in the future but is not yet incor­
IGHV muta­tional sta­tus has been con­sid­ered in the last 20 years po­rated into a treat­ment deci­sion algo­rithm.
to be the most pow­er­ful pre­dic­tive indi­ca­tor. Patients with un­
mutated IGHV genes (UM-IGHV CLL, ie, ≥98% homol­ogy with Treatment options today
germ line sequence) have infe­rior out­comes after any CIT reg­ Outside of clin­i­cal tri­als, as of today, 2 dif­fer­ent strat­e­gies may
i­men.11,15,16 Targeted agents have dem­on­strated a clear ben­e­fit be con­sid­ered: either con­tin­u­ous ther­apy until pro­gres­sion with
over CIT for these patients.17 BTKi alone or com­bined with obinutuzumab (O), or time-lim­ited
Even for FCR inel­i­gi­ble patients, deter­mi­na­tion of IGHV muta­ treat­ment with either CIT or VEN-based reg­i­mens. In this sec­tion,
tional sta­tus is of inter­est. The genetic com­plex­ity appears dif­ we will con­sider only the time-lim­ited ther­a­pies.
fer­ent: UM-IGHV patients accu­mu­late sig­nif­i­cantly more genetic CIT is the pro­to­type of time-lim­ited treat­ment and may still
alter­ations than M-IGHV CLL.18 The neg­ a­
tive impact of this be of ben­e­fit for some patients. The ther­a­peu­tic reg­i­mens have

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/59/1851402/59levy.pdf by guest on 13 December 2021


increased muta­tion bur­den has been reduced in UM-IGHV CLL been mod­u­lated based on fit­ness and age, from chlorambucil
by targeted agents, but there are dif­fer­ent dom­i­nant muta­genic (CLB) and anti-CD20 to BR/FCR.11,15,16 Targeted-agent reg­i­mens
mech­a­nisms in M-IGHV and UM-IGHV, under­ly­ing a very likely dif­ have been com­pared to CIT in front­line (Table 4; Figure 1).
fer­ent clonal evo­lu­tion at relapse.19 Therefore, deter­mi­na­tion of Ibrutinib-rituximab (I-R) until pro­gres­sion has been com­pared
IGHV muta­tional sta­tus has been recommended for all­patients.4,5 to FCR in a large ran­dom­ized (2:1) phase 3 trial includ­ing 529
Whether a kar­yo­type should be performed in rou­tine prac­ patients youn­ger than 70 with no 17p dele­tion.17 I-R is clearly
tice remains open. Complex karyotype is a known det­ri­men­tal supe­rior in terms of pro­gres­sion-free sur­vival (PFS) and even OS.
fac­tor in clin­i­cal tri­als, both for CIT and VEN.20 It is of inter­est This is related to the large ben­e­fit of I-R over FCR for UM-IGHV
when it can be eas­ily obtained, but the results still do not clearly patients. It is note­wor­thy that in M-IGHV cases, PFS after I-R and
influ­ence treat­ment deci­sion. Similarly, sev­eral recur­rent gene FCR remains sim­i­lar at a fol­low-up of 33.6 months despite the
muta­tions are observed in CLL, such as NOTCH1, SF3B1, and fact that FCR is a 6-month treat­ment. The inci­dence of grade >3

Table 4. Summary of time-lim­ited front­line tri­als

Reference Trial Patients Primary end point Results


Randomized phase 3 tri­als
Shanafelt et al17, FCR vs I-R Age <70 PFS I - R > FCR (89.4% vs 72.9% at 3 y;
ECOG1912 no del 17p FU: 33.6 mo HR: 0.35; 95% CI, 0.22-0.56; P < .001)
N = 529 OS: I - R > FCR (98.8% vs 91.5% at 3 y)
HR: 0.17; 95% CI, 0.05-0.54; P < .001
Al-Sawaf et al3, CLB-O vs VEN-O CIRS >6 and/or Cl. creat PFS VEN-O > CLB-O (NE vs 35.5 mo)
CLL14 30-69 mL/min FU: 39.6 mo HR: 0.31, 95% CI, 0.22–0.44; P < .0001
N = 432
Woyach et al1, ALLIANCE BR/I-R/I Age >65 PFS I-R > BR. HR: 0.38; 95% CI, 0.25-0.59;
N = 447 FU: 38 mo P < .001
I > BR. HR: 0.39; 95% CI, 0.26-0.58;
P < .001
Kater et al23, VEN-I/CLB-O Age >65 or CIRS >6 or Cl. PFS VEN-I > CLB-O
GLOW creat <70 mL/min FU: 27.7 mo HR: 0.216 (95% CI, 0.131-0.357);
N = 211 p < .0001)

Phase 2 tri­als
Michallet et al24, ICLL07 I-O+/−4 courses FCO-I Fit patients, no del 17p or p53 CR + BM MRD <0.01% CR + MRD <0.01%: 62% (95% CI 55%
N = 135 FU: 26.3 to 69%)
Davids et al25 FCR 6 courses-I (stop 2 y if Age <65 CR + BM uMRD 2 mo CR + BM uMRD: 33%, (95% CI 23-
BM uMRD) N = 85 post FCR 44%)
FU: 16.5 mo
Jain et al27 VEN-I Del17P and/or TP53 mut CR + CRi CR + CRi: 88%, CR + uMRD: 61%
and/or del11q and/or FU: 14.8 mo
U-IGHV and/or age >65
N = 80
Wierda et al28, CAPTIVATE VEN-I Age <70 1 y DFS I vs pla­cebo: NS (95% vs 100%)
uMRD : I vs placebo N = 164 FU: NA 30-mo PFS ∼95% in all­arms
MRD+ : I vs V-I
A, acalabrutinib; B, bendamustine; C, Cyclophosphamide; Cl. creat, clear­ing of cre­a­tine; CR, complete remission; DFS, dis­ease-free sur­vival;
F, Fludarabine; FU, median fol­low-up; I, ibrutinib; NA, not appli­ca­ble; NE, not evaluable; NS, not significant; O, obinutuzumab; OS, overall survival;
PFS, progression-free survival; R, rituximab; VEN, venetoclax.

62  |  Hematology 2021  |  ASH Education Program


Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/59/1851402/59levy.pdf by guest on 13 December 2021

Figure 1. Outline of phase 3 and phase 2 time-lim­ited front­line tri­als.

Prakash Singh Shekhawat


Up-front ther­apy: time-lim­ited treat­ment  |  63
adverse events (AEs) is sim­i­lar in both arms, with fewer grade 3 17p or 11q dele­tion, TP53 muta­tion, UM-IGHV, or an age of 65
or more infec­tious com­pli­ca­tions in the I-R arm than in the FCR years or older.27 In this het­ero­ge­neous pop­ul­a­tion, the CR + CRi
(10.5% vs 20.3%). (com­plete remis­sion with incom­plete recov­ery) rate was 88%,
The ALLIANCE trial com­pared 183 patients receiv­ing benda­ and CR with uMRD was 61%. There was no addi­tional tox­ic­ity in
mustine plus rituximab BR to 182 patients receiv­ing ibrutinib plus the com­bi­na­tion of the 2 agents. Considering the short fol­low-
rituximab I-R and 182 receiv­ing ibrutinib (I) alone in a phase 3 up (14.8 months), no sur­vival data are avail­­able.
ran­dom­ized (1:1:1) trial includ­ing older adult patients (≥65 years Finally, in the CAPTIVATE MRD trial,28 VEN-I was also tested
of age).1 The median fol­low-up was 38 months. PFS was supe­ in 164 “high-risk” patients defined by at least one of the fol­low­
rior with the ibrutinib reg­i­men vs the addi­tion of B + R, with no ing: del(17p), del(11q) TP53 muta­tion, com­plex kar­yo­type, or UM-
dif­fer­ence between I and I-R. There was no dif­fer­ence in over­ IGHV. There was an MRD-guided ran­dom­i­za­tion after 12 cycles
all sur­vival (OS). Patients receiv­ing BR had a higher num­ber of of com­ bined treat­ment, and uMRD patients were ran­ domly
grade ≥3 hema­to­logic AEs com­pared to ibrutinib-based treat­ assigned to pla­cebo or ibrutinib, the oth­ers being ran­dom­ized to
ments (61% vs 41% and 29%) and a lower num­ber of grade ≥3 receive ibrutinib or con­tin­ued VEN-I. PFS rates were >95% across
nonhematologic AEs (63% vs 74% each). all­ran­dom­ized arms at 30 months.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/59/1851402/59levy.pdf by guest on 13 December 2021


The CLL14 phase 3 trial com­par­ing the venetoclax-obinutu­ In sum­mary, all­phase 3 tri­als have shown the supe­ri­or­ity of
zumab (VEN-O) com­bi­na­tion with chlorambucil-obinutuzumab targeted agents over CIT when con­sid­er­ing the whole cohort.
(CLB-O) ran­dom­ized (1:1) 432 frail patients as defined by a cumu­ This dif­fer­ence is related to the large ben­e­fit of targeted agents
lative illnesses rating scale (CIRS) >6 and/or eGFR between in UM-IGHV cases. When con­sid­er­ing M-IGHV cases, only CLB-O
30 and 69 mL/min.3 With a fol­low-up of 39.6 months, PFS and time was infe­rior to a targeted-agent reg­i­men whether com­pared to
to next treat­ment were sig­nif­i­cantly lon­ger with VEN-O in both ibrutinib or I-O (ILLUMINATE) or VEN-O (CLL14).3,12 Conversely, in
M- and UM-IGHV CLL, but no sur­vival ben­efi ­ t was observed. Seri­ the ECOG trial and ALLIANCE trial no ben­e­fit of BTKi over CIT
ous AEs occurred in 54% and 44% of the patients, respec­tively. treat­ment (FCR or BR) was observed for M-IGHV cases whether
The most com­mon grade ≥3 was neutropenia in both groups alone or in com­bi­na­tion with rituximab.1,17
(48% and 54%, respec­tively). Neutropenia occurred mainly dur­ Phase 2 tri­als have explored the addi­tion of ibrutinib to an FC-
ing the course of obinutuzumab treat­ment. anti-CD20 back­bone with impres­sive results, but unfor­tu­nately
The GLOW trial is a ran­dom­ized (1:1) phase 3 trial strat­i­fied this strat­egy was not com­pared to chemo-free ther­a­pies. Recent
on IGHV muta­tional sta­tus and del(11q) com­par­ing ibrutinib and tri­als have tested time-lim­ited targeted-agent com­bi­na­tions with
VEN to CLB-O in patients older than 65 or youn­ger with a CIRS very good results. The VEN-I com­bi­na­tion MRD data are very
>6 or with a cre­at­i­nine clear­ance <70 mL/min.23 Patients with a encour­ag­ing, but fol­low-up is still short for sur­vival anal­y­sis.27,28
del(17p) or TP53 muta­tion were not eli­gi­ble. Two hun­dred and It is note­wor­thy that targeted agents show a sig­nif­i­cant activ­
eleven patients were ran­dom­ized (median age, 71 years). After a ity in TP53-altered cases. Any chemo-free reg­i­men was supe­rior
median fol­low-up of 27.7 months, VEN-I was sig­nif­i­cantly supe­rior to what was pre­vi­ously observed with CIT, as TP53-altered cases
to CLB + O in terms of PFS (haz­ard ratio [HR], 0.216), com­plete are resis­tant to CIT. Available data are the result of unplanned
remis­sion (CR) rate, uMRD in bone mar­row (BM), and time to sub­group anal­y­sis in tri­als and tend to favor the use of BTKi or
next treat­ment. The most fre­quent grade >3 events for patients VEN-I over VEN +/− anti-CD20, despite the absence of a cur­rent
treated by I + V were neutropenia (34.9%), diar­rhea (10.4%), and head-to-head com­par­i­son.
hyper­ten­sion (7.5%). In the absence of comorbidities pre­vent­ing the use a cer­tain
In addi­tion to these phase 3 tri­als, sev­eral phase 2 tri­als have targeted agent, usu­ally BTKi, the deci­sion of which targeted
explored the com­bi­na­tion of ibrutinib with either CIT or VEN in agent is pref­er­a­ble is always a mat­ter of debate among phy­
front­line (Table 4; Figure 1). si­cians and of dis­cus­sion with the patient. Besides the case of
In the ICLL07FILO trial,24 135 fit TP53 wild-type unmutated IGHV TP53 alter­ations, some clin­i­cal sit­u­a­tions may tip the bal­ance in
CLL patients received the com­ bi­
na­
tion of obinutuzumab and favor of one or the other targeted agent. For exam­ple, in the
15-month fixed-dura­tion ibrutinib. At month 9, the 120 patients case of a pro­gres­sive CLL with a very short dou­bling time but
(92%) who were not in BM CR with MRD <0.01% received no bulky lymph nodes we would tend to favor the use of VEN +
4 courses of FC-O. At the final eval­u­a­tion at month 15, a CR with anti-CD20, whereas in the case of a bulky dis­ease, BTKi might
BM uMRD rate of 62% was obtained with an accept­able tox­ic­ity. be pre­ferred.
At a median fol­low-up at 36.7 months, OS and PFS at 3 years Several tri­als are also ongo­ing that are test­ing mainly time-
were 95.7% and 98%, respec­tively. lim­ited strat­e­gies (Table 6). They explore com­bi­na­tions with
In a phase 2 trial,25 85 patients aged 65 or youn­ger received other BTKi and/or the tri­ple com­bi­na­tion of VEN with BTKi and
I + FCR (6 cycles). Responders con­tin­ued on ibrutinib main­ anti-CD20. The results will be avail­­able in the near future.
te­nance for up to 2 years, and patients with BM uMRD were
­able to ­discontinue treat­ment. Thirty-three per­cent of patients
achieved a CR with BM uMRD 2 months after the last course
of FCR. The most com­mon all­-grade toxic effects were hema­
to­log­i­cal. At a median fol­low-up of 16.5 months, 1 death was CLINICAL CASE 1 (Con­tin­ued)
observed but no pro­gres­sion. The M. D. Anderson group tested How would you treat the first patient today? In 2021, 11 years
3 cycles of the I-FC-O com­bi­na­tion in 45 M-IGHV fit patients, after receiv­ing FCR, this patient is still in clin­i­cal CR. These very
with sim­i­lar excel­lent results.26 long remis­sions were high­lighted by the M. D. Anderson group,
The venetoclax-ibrutinib com­bi­na­tion (VEN-I) was tested in show­ing that a sig­nif­i­cant per­cent­age of M-IGHV patients expe­
80 “high-risk” patients defined by at least one of the fol­low­ing: ri­ence very prolonged remis­sions (>10 years) after FCR.29

64  |  Hematology 2021  |  ASH Education Program


Table 6. Major ongo­ing tri­als eval­u­at­ing fixed-dura­tion targeted ther­a­pies against CIT or con­tin­u­ous targeted ther­a­pies

Trial N pts Trial pop­u­la­tion Compared arms Primary end point


CLL13- GAIA 926 ≥18 y CIT: FCR/BR × 6 cycles MRD at mo 15 (CIT vs GVe)
GCLLSG fit/TP53 abn excluded vs RVe: VEN-R (12 mo) PFS (CIT vs GIVe)
NCT02950051 vs GVe: VEN-O (12 mo)
vs GIVe: VEN-O + I (up to 36 mo;
VEN 12 mo)
ACE-CL-311 780 ≥18 y CIT: FCR/BR × 6 cycles PFS (CIT vs AV)
AstraZeneca fit/TP53 abn excluded vs AV: VEN-O + acalabrutinib
NCT03836261 (15 mo; VEN 12 mo)
vs AVG: VEN-O + acalabrutinib
(15 mo; VEN 12 mo)
CRISTALLO 165 ≥18 y CIT: FCR or BR (if ≥65 y) × 6 BM MRD at mo 15
Hoffmann La Roche fit/TP53 abn excluded cycles

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/59/1851402/59levy.pdf by guest on 13 December 2021


NCT04285567 vs VG: VEN-O (12 mo)
FILO ERADIC 120 ≥18 y CIT: FCR × 6 cycles BM MRD at mo 27
NCT04010668 fit/TP53 abn excluded vs VI: VEN-I (15 or 27 mo
according to MRD)
NCRI UK CLL10 FLAIR 1516 ≤75 y CIT: FCR × 6 cycles PFS
CRUK/12/037 fit/eGFR >30 mL/min vs IR → I*: I + ritux → I (until
del(17p) <20% pro­gres­sion)
vs VI: VEN-I (flex­i­ble dura­tion
according to MRD)
* arm IR replaced by
I monotherapy in 2018
ALLIANCE A041702 454 ≥70 y IO: I + O (until pro­gres­sion) PFS
NCT03737981 vs IVO: VEN-O + I (15 mo; VEN
12 mo)
ECOG-ACRIN EA9161 720 18-69 y IO: I + O (until pro­gres­sion) PFS
NCT03701282 del(17p) excluded vs IVO: VEN-O + I (19 mo; VEN
12 mo)
GCLLSG CLL17 897 ≥18 y I: I (until pro­gres­sion) PFS
NCT04608318 fit and unfit vs VG: VEN-O (12 mo)
w/or w/o TP53 abn vs VI: VEN-I (15 mo; VEN 12 mo)
abn, abnor­mal­i­ties; A, acalabrutinib; I, ibrutinib; O, obinutuzumab; R, rituximab; VEN, venetoclax.

Ten years later, this patient would still be eli­gi­ble for treat­ with an anti-CD20 or BTKi. Data from the CLL14 trial show the
ment with FCR since he has M-IGHV CLL and no TP53 alter­ation. clear supe­ri­or­ity of a 1-year ­treat­ment of VEN-O over CLB-O.
In 2021, con­tin­u­ous BTKi can also be con­sid­ered but con­tin­ued Moreover, in the CLL14 trial it was recently reported that the
until pro­gres­sion. His comorbidities (high blood pres­ sure, a VEN-O com­bi­na­tion could reduce the growth dynam­ics, as a
his­tory of cor­o­nary throm­bo­sis on ace­tyl­sal­i­cylic acid) do not lower growth rate was observed at r­ eprogression after VEN-O.30
restrain FCR but on the con­trary, ibrutinib should be used with Approval of the targeted agents was obtained through
cau­tion. Concerning the 1-year VEN-O com­bi­na­tion, the results randomiza­tion against a ref­er­enced ­ther­a­peu­tic ­reg­i­men, ie,
from CLL14 might be extrap­o­lated to youn­ger patients, in which age-adapted CIT. Data from a head-to-head com­par­i­son of chemo-
the data show the supe­ri­or­ity of VEN-O over CLB-O even in free reg­i­mens are not avail­­able yet, and for patients eli­gi­ble for
M-IGHV patients. As of yet, how­ever, no data are avail­­able esti­ either con­tin­u­ous BTKi or time-lim­ited VEN-based com­bi­na­tions,
mat­ing the mag­ni­tude of the effect—if any—com­pared to FCR. a treat­ment deci­sion relies mostly on comorbidities, comedica­
tions, and patient pref­er­ences. It is most likely that the life expec­
tancy of this patient would have been increased with first-line
targeted agents. This patient today would receive a targeted
agent, at least in relapse.
CLINICAL CASE 2 (Con­t in­u ed)
How would you treat the sec­ond patient today? The out­come
was, unfor­tu­nately, poorer in this patient, who progressed What would be help­ful for adjusting the dura­tion of time-
30 months after BR with altered performance status and rap­idly lim­ited treat­ment?
grow­ing super­fi­cial polyadenopathy and spleno­meg­aly. Despite Most of the data avail­­able to date in front­line have been gen­er­
ade­quate sup­port­ive care with infec­tion pro­phy­laxis, he died of ated by treat­ment strat­e­gies of an arbi­trarily fixed dura­tion, such
sep­sis dur­ing the reintroduction of BR. as the CLL14 trial with a 1-year fixed dura­tion of VEN-O.
All tri­als show the supe­ri­or­ity of targeted agents in UM-IGHV The value of MRD sta­tus as a sur­ro­gate of PFS has been widely
CLL. Time-lim­ited treat­ments are VEN-based com­bi­na­tions, either proven in the con­text of CIT.31 The dem­on­stra­tion of the MRD
Prakash Singh Shekhawat
Up-front ther­apy: time-lim­ited treat­ment  |  65
value through large tri­als led to approval by the Euro­pean Medi­ 5. Eichhorst B, Robak T, Montserrat E, et al; Euro­pean Society for Medical
cines Agency of the use of uMRD as an inter­me­di­ate end point in Oncology Guidelines Committee. Chronic lym­pho­cytic leu­kae­mia: ESMO
Clinical Practice Guidelines for diag­no­sis, treat­ment and fol­low-up. Ann
ran­dom­ized clin­i­cal tri­als for drug approval.32 It appeared to be a Oncol. 2021;32(1):23-33.
wor­thy pre­dic­tor in VEN-based treat­ment.3 6. Quinquenel A, Aurran-Schleinitz T, Clavert A, et al. Diagnosis and treat­ment
Through the anal­ y­sis of a large num­ ber of patients who of chronic lym­pho­cytic leu­ke­mia: rec­om­men­da­tions of the French CLL
received FCR in 3 clin­i­cal tri­als, it was dem­on­strated that high- Study Group (FILO). Hemasphere. 2020;4(5):e473.
7. Gribben JG, Bosch F, Cymbalista F, et al. Optimising out­comes for patients
sen­si­tiv­ity (0.0007%) MRD assess­ment in blood yielded addi­
with chronic lym­pho­cytic leu­kae­mia on ibrutinib ther­apy: Euro­pean rec­
tional prog­nos­tic infor­ma­tion beyond the cur­rent stan­dard om­men­da­tions for clin­i­cal prac­tice. Br J Haematol. 2018;180(5):666-679.
sen­si­tiv­ity (0.01%).33 Currently, the dura­tion of time-lim­ited treat­ 8. Gribben JG. Practical man­age­ment of tumour lysis syn­drome in veneto­
ment is not guided by MRD sta­tus at the end of treat­ment. In clax-treated patients with chronic lym­pho­cytic leu­kae­mia. Br J Haematol.
the future, the use of high-sen­si­tiv­ity MRD assess­ment could be 2020;188(6):844-851.
9. Mauro FR, Giannarelli D, Galluzzo CM, et  al. Response to the con­ju­gate
use­ful for deter­min­ing response at a deep level in poten­tially pneu­mo­coc­cal vac­cine (PCV13) in patients with chronic lym­pho­cytic leu­
cura­tive time-lim­ited reg­im ­ ens. Moreover, MRD kinet­ics will be ke­mia (CLL). Leukemia. 2021;35(3):737-746.
needed to bet­ter under­stand the impact of the var­i­ous novel 10. Herishanu Y, Avivi I, Aharon A, et  al. Efficacy of the BNT162b2 mRNA

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/59/1851402/59levy.pdf by guest on 13 December 2021


agents and to be ­able to decide when to discontinue ther­apy in COVID-19 vac­cine in patients with chronic lym­pho­cytic leu­ke­mia. Blood.
2021;137(23):3165-3173.
patients who do or do not achieve unde­tect­able dis­ease.34
11. Fischer K, Bahlo J, Fink AM, et al. Long-term remis­sions after FCR chemoim­
munotherapy in pre­vi­ously untreated patients with CLL: updated results of
Conclusion the CLL8 trial. Blood. 2016;127(2):208-215.
Numerous ongo­ing tri­als dem­on­strate both phy­si­cian and 12. Moreno C, Greil R, Demirkan F, et al. Ibrutinib plus obinutuzumab ver­sus
patient aspi­ ra­
tions for time-lim­ ited ther­
a­pies. Combining tar­ chlorambucil plus obinutuzumab in first-line treat­ ment of chronic lym­
geted agents is very prom­is­ing, but it is impor­tant to keep in pho­cytic leu­kae­mia (iLLUMINATE): a multicentre, randomised, open-label,
phase 3 trial. Lancet Oncol. 2019;20(1):43-56.
mind that fol­low-up is still short and that infor­ma­tion on the effi­ 13. Zenz T, Eichhorst B, Busch R, et al. TP53 muta­tion and sur­vival in chronic
cacy of sub­se­quent lines of ther­apy is lacking. Sensitive MRD lym­pho­cytic leu­ke­mia. J Clin Oncol. 2010;28(29):4473-4479.
should help tai­lor the opti­mal dura­tion of the var­i­ous strat­eg
­ ies. 14. Campo E, Cymbalista F, Ghia P, et al. TP53 aber­ra­tions in chronic lym­pho­
Today, a deci­sion between con­tin­u­ous treat­ment with BCR cytic leu­ke­mia: an over­view of the clin­ic
­ al impli­ca­tions of improved diag­nos­
tics. Haematologica. 2018;103(12):1956-1968.
inhib­i­tors and VEN-based com­bi­na­tions relies mostly on comor­
15. Eichhorst B, Fink AM, Bahlo J, et al; inter­na­tional group of inves­ti­ga­tors;
bidities, comedications, and patient/phy­si­cian pref­er­ences. The Ger­man CLL Study Group. First-line chemoimmunotherapy with benda­
rap­idly increas­ ing under­ stand­ ing of genetic back­ ground and mustine and rituximab ver­sus fludarabine, cyclo­phos­pha­mide, and ritux­
clonal evo­lu­tion is not yet inte­grated into ther­a­peu­tic deci­sions, imab in patients with advanced chronic lym­pho­cytic leu­kae­mia (CLL10): an
but in the com­ing years it could sup­plant the “one-size-fits-all­” inter­na­tional, open-label, randomised, phase 3, non-infe­ri­or­ity trial. Lancet
Oncol. 2016;17(7):928-942.
ten­dency and allow more per­son­al­ized approaches. 16. Goede V, Fischer K, Busch R, et  al. Obinutuzumab plus chlorambucil in
patients with CLL and coexisting con­di­tions. N Engl J Med. 2014;370(12):1101-
Conflict-of-inter­est dis­clo­sure 1110.
Vincent Lévy: hon­ o­raria: Abbvie, Astra-Zeneca, CSL Behring, 17. Shanafelt TD, Wang XV, Kay NE, et al. Ibrutinib-rituximab or chemoimmuno­
Janssen. therapy for chronic lym­pho­cytic leu­ke­mia. N Engl J Med. 2019;381(5):432-
443.
Alain Delmer: hon­o­raria: Janssen, Abbvie, AstraZeneca, Roche.
18. Landau DA, Tausch E, Taylor-Weiner AN, et al. Mutations driv­ing CLL and
Florence Cymbalista: hon­o­raria: Janssen, Abbvie, AstraZeneca, their evo­lu­tion in pro­gres­sion and relapse. Nature. 2015;526(7574):525-530.
Roche. 19. Burns A, Alsolami R, Becq J, et al. Whole-genome sequenc­ing of chronic
lym­pho­cytic leu­kae­mia reveals dis­tinct dif­fer­ences in the muta­tional land­
Off-label drug use scape between IgHVmut and IgHVunmut sub­groups. Leukemia. 2018;32(2):
Vincent Lévy: nothing to disclose. 332-342.
20. Baliakas P, Jeromin S, Iskas M, et al; Euro­pean Research Initiative on CLL.
Alain Delmer: nothing to disclose. Cytogenetic com­plex­ity in chronic lym­pho­cytic leu­ke­mia: def­i­ni­tions,
Florence Cymbalista: nothing to disclose. asso­ci­a­tions, and clin­i­cal impact. Blood. 2019;133(11):1205-1216.
21. Lazarian G, Guièze R, Wu CJ. Clinical impli­ca­tions of novel geno­mic dis­cov­
Correspondence er­ies in chronic lym­pho­cytic leu­ke­mia. J Clin Oncol. 2017;35(9):984-993.
Florence Cymbalista, Hematology Biology, CHU Paris Seine- 22. Gruber M, Bozic I, Leshchiner I, et  al. Growth dynam­ics in nat­u­rally pro­
gressing chronic lym­pho­cytic leu­kae­mia. Nature. 2019;570(7762):474-479.
Saint-Denis - Hôpital Avicenne, 125 rue de Stalingrad, Bobigny,
23. Arnon Kater A, Carolyn Owen C, Carol Moreno C, et al. Fixed-dura­tion ibru­
France 93009; e-mail: florence.cymbalista@aphp.fr. tinib and venetoclax (I + V) ver­sus chlorambucil plus obinutuzumab (CLB+O)
for first line (1L) chronic lym­pho­cytic leu­ke­mia (CLL): pri­mary anal­y­sis of the
References phase 3 GLOW study. EHA Library. 6 June 2021. Abstract LB1902.
1. Woyach JA, Ruppert AS, Heerema NA, et  al. Ibrutinib reg­i­mens ver­sus 24. Michallet AS, Dilhuydy MS, Subtil F, et  al. Obinutuzumab and ibrutinib
chemoimmunotherapy in older patients with untreated CLL. N Engl J Med. induc­tion ther­apy followed by a min­im ­ al resid­ual dis­ease-driven strat­egy
2018;379(26):2517-2528. in patients with chronic lym­pho­cytic leu­kae­mia (ICLL07 FILO): a sin­gle-arm,
2. Burger JA, Barr PM, Robak T, et al. Long-term effi­cacy and safety of first-line multicentre, phase 2 trial. Lancet Haematol. 2019;6(9):e470-e479.
ibrutinib treat­ment for patients with CLL/SLL: 5 years of fol­low-up from the 25. Davids MS, Brander DM, Kim HT, et al; Blood Cancer Research Partnership
phase 3 RESONATE-2 study. Leukemia. 2020;34(3):787-798. of the Leukemia and Lymphoma Society. Ibrutinib plus fludarabine, cyclo­
3. Al-Sawaf O, Zhang C, Tandon M, et al. Venetoclax plus obinutuzumab ver­sus phos­pha­mide, and rituximab as ini­tial treat­ment for youn­ger patients with
chlorambucil plus obinutuzumab for pre­vi­ously untreated chronic lym­pho­ chronic lym­pho­cytic leu­kae­mia: a sin­gle-arm, multicentre, phase 2 trial.
cytic leu­kae­mia (CLL14): fol­low-up results from a multicentre, open-label, Lancet Haematol. 2019;6(8):e419-e428.
randomised, phase 3 trial. Lancet Oncol. 2020;21(9):1188-1200. 26. Jain N, Thompson PA, Burger JA, et  al. Ibrutinib, fludarabine, cyclo­phos­
4. Hallek M, Cheson BD, Catovsky D, et  al. iwCLL guide­lines for diag­no­sis, pha­mide, and obinutuzumab (iFCG) reg­i­men for chronic lym­pho­cytic leu­
indi­ca­tions for treat­ment, response assess­ment, and sup­port­ive man­age­ ke­mia (CLL) with mutated IGHV and with­out TP53 aber­ra­tions. Leukemia.
ment of CLL. Blood. 2018;131(25):2745-2760. Published online 18 May 2021.

66  |  Hematology 2021  |  ASH Education Program


27. Jain N, Keating M, Thompson P, et al. Ibrutinib and venetoclax for first-line 32. Euro­pean Medicines Agency Committee for Medicinal Products for Human
treat­ment of CLL. N Engl J Med. 2019;380(22):2095-2103. Use. Appendix 4 to the guide­line on the eval­u­a­tion of anti­can­cer medic­i­nal
28. Wierda WG, Tam CS, Allan JN, et  al. Ibrutinib (Ibr) plus venetoclax (Ven) prod­ucts in man [updated 15 Feb­ru­ary 2016]. Accessed 10 Jan­u­ary 2018.
for first-line treat­ment of chronic lym­pho­cytic leu­ke­mia (CLL)/small lym­ http:​­/​­/www​­.ema​­.europa​­.eu​­/docs​­/en_GB​­/document_library​­/Scientific​
pho­cytic lym­phoma (SLL): 1-year dis­ease-free sur­vival (DFS) results from ­_guideline​­/2016​­/02​­/WC500201945​­.pdf.
the MRD cohort of the phase 2 CAPTIVATE study. Blood. 2020;136(suppl 1): 33. Letestu R, Dahmani A, Boubaya M, et al; French Innovative Leukemia Orga­
16-17, abstract 123. nization. Prognostic value of high-sen­si­tiv­ity mea­sur­able resid­ual dis­ease
29. Thompson PA, Tam CS, O’Brien SM, et  al. Fludarabine, cyclo­ phos­pha­ assess­ment after front-line chemoimmunotherapy in chronic lym­pho­cytic
mide, and rituximab treat­ment achieves long-term dis­ease-free sur­vival in leu­ke­mia. Leukemia. 2021;35(6):1597-1609.
IGHV-mutated chronic lym­pho­cytic leu­ke­mia. Blood. 2016;127(3):303-309. 34. Fürstenau M, De Silva N, Eichhorst B, Hallek M. Minimal resid­ual dis­ease
30. Al-Sawaf O, Zhang C, Robrecht S, et  al. Clonal dynam­ics after veneto­ assess­ment in CLL: ready for use in clin­i­cal rou­tine? Hemasphere. 2019;3(5):
clax-obinutuzumab ther­apy: novel insights from the ran­dom­ized, phase 3 e287.
CLL14 trial. Blood. 2020;136(suppl 1):22-23.
31. Kovacs G, Robrecht S, Fink AM, et al. Minimal resid­ual dis­ease assess­ment
improves pre­dic­tion of out­come in patients with chronic lym­pho­cytic leu­
ke­mia (CLL) who achieve par­tial response: com­pre­hen­sive anal­y­sis of two
phase III stud­ies of the Ger­man CLL study group. J Clin Oncol. 2016;34(31): © 2021 by The Amer­i­can Society of Hematology

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/59/1851402/59levy.pdf by guest on 13 December 2021


3758-3765. DOI 10.1182/hema­tol­ogy.2021000233

Prakash Singh Shekhawat


Up-front ther­apy: time-lim­ited treat­ment  |  67
CLL: EXTENDING SURVIVAL

Is there a role for anti-CD20 antibodies in CLL?


Harsh R. Shah and Deborah M. Stephens

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/68/1851825/68shah.pdf by guest on 13 December 2021


University of Utah, Salt Lake City, UT

Anti-CD20 monoclonal antibodies (mAbs) have revolutionized the treatment of chronic lymphocytic leukemia (CLL) by
improving survival of patients with CLL in conjunction with chemotherapy. However, the novel targeted agents such
as Bruton tyrosine kinase inhibitors (BTKis) and venetoclax have now mostly replaced chemotherapy in frontline treat-
ment of CLL. Several clinical trials have been conducted to examine the role of anti-CD20 mAbs in combination with
BTK inhibitors and venetoclax. Addition of rituximab to ibrutinib does not improve progression-free survival (PFS) of
treatment-naive patients with CLL, possibly related to ibrutinib’s antagonistic effect on anti-CD20 antibodies. Alterna-
tively, addition of a glycoengineered anti-CD20 mAb obinutuzumab to a more selective BTKi acalabrutinib may improve
PFS but does not improve overall survival of patients with CLL in the frontline setting, pending long-term follow-up. Thus,
we suggest that the addition of an anti-CD20 mAb to a BTKi is of most benefit to patients with autoimmune cytopenia
or rapidly progressive disease. In contrast to BTKis, combination of fixed-duration venetoclax and anti-CD20 mAb can
induce deep remission with high rates of undetectable minimal residual disease, correlating with improved survival of
patients with CLL in both frontline and relapsed/refractory settings. In this review, we discuss clinical trials of BTKis and
venetoclax that have investigated the role of anti-CD20 mAbs in frontline and relapsed settings of CLL treatment. We
also provide an algorithm suggesting how anti-CD20 mAbs may be incorporated in the treatment of patients with CLL,
including specific scenarios.

LEARNING OBJECTIVES
• Anti-CD20 monoclonal antibody plus BTK inhibitor may provide clinical benefit in patients with active autoimmune
cytopenia or rapidly progressive disease.
• Adding anti-CD20 antibody to venetoclax induces deep remissions, allowing for fixed-duration treatment.

I antibody that eradicates CLL cells primarily by means of


CLINICAL CASE complement-dependent cytotoxicity and antibody-depen-
Mr. Smith, a 67-year-old man, was diagnosed with chronic dent cellular cytotoxicity (ADCC) after binding to CD20
lymphocytic leukemia (CLL) 3 years ago. He was asymp- (Figure 1).1 Addition of rituximab to the standard chemo-
tomatic, without cytopenias, and was observed. Three therapy agents fludarabine and cyclophosphamide (FCR)
months ago, he had purpura and thrombocytopenia. improved overall survival (OS) and progression-free survival
Evaluation revealed immune thrombocytopenic purpura (PFS) compared with fludarabine and cyclophophamide in
(ITP). Over the course of 2 months, his ITP was refractory the frontline treatment of patients with CLL. This combina-
to treatment with steroids and intravenous immunoglob- tion became a standard option for fit patients.2 Although
ulin. Today, Mr. Smith reports drenching night sweats, bendamustine (another standard chemotherapy agent)
fatigue, and early satiety. Laboratory tests show anemia with or without rituximab was not directly compared in
and thrombocytopenia. He has refractory ITP and meets a head-to-head clinical trial, bendamustine plus rituximab
criteria for CLL treatment. (BR) showed favorable overall response rates (ORRs) and
PFS compared with bendamustine alone in treatment-
naive patients with CLL in an imprecise cross-trial com-
parison.3,4 The direct comparison of FCR to BR for treat-
Introduction ment-naive fit patients with CLL concluded, though, that
Anti-CD20 monoclonal antibodies (mAbs) have been inte- BR was better tolerated in patients older than 65 years and
grated in treatment of CLL in combination with chemother- is considered an option for this population.5 Obinutuzumab
apy for longer than a decade. Rituximab is a chimeric type is a potent humanized, glycoengineered type II antibody

68 | Hematology 2021 | ASH Education Program


Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/68/1851825/68shah.pdf by guest on 13 December 2021
Figure 1. Mechanism of action of select anti-CD20 monoclonal antibodies used to treat CLL. More “+” sign indicates stronger
mechanism of action. ADCP, antibody-dependent cellular phagocytosis; CDC, complement dependent cytotoxicity; MAC, membrane
attack complex.

that also tar­gets CD20 on the cell sur­face but with enhanced of anti-CD20 mAbs has a firmly established role in the front­line
ADCC, anti­body-depen­dent phago­cy­to­sis, and direct cell death treat­ment of CLL when used in com­bi­na­tion with stan­dard che­
effects com­pared with rituximab (Figure 1).6,7 In treat­ment-naive mo­ther­apy agents. Over the past few years, the stan­dard of
patients with CLL with preexisting con­ di­
tions, rituximab or care has shifted from stan­dard che­mo­ther­apy agents to using
obinutuzumab in com­bi­na­tion with chlorambucil was com­pared targeted kinase inhib­i­tors, such as Bruton tyro­sine kinase inhib­i­
head-to-head.8 The com­bi­na­tion of chlorambucil and obinutu- tors (BTKis) or B-cell lym­phoma 2 (BCL2) inhib­i­tors. It is not clear
zumab (Chl-O) resulted in higher com­plete remis­sion (CR) rates, whether anti-CD20 mAbs are required for effi­cacy when used in
PFS, and OS com­pared with the rituximab com­bi­na­tion, pro­vid­ com­bi­na­tion with these targeted kinase inhib­i­tors. In this study,
ing evi­dence to sup­port the supe­ri­or­ity of obinutuzumab com­ we review the avail­­able data surrounding anti-CD20 mAb com­
pared with rituximab.8,9 Based on these piv­otal stud­ies, the use bi­na­tions in CLL treat­ment.
Prakash Singh Shekhawat
Anti-CD20 antibodies in CLL | 69
BTK Inhibitors Acalabrutinib ± obinutuzumab
BTK is a kinase within the TEC fam­ily that leads to down­stream Acalabrutinib is a highly selec­tive BTKi with less off-tar­get inhi­
acti­va­tion of AKT, extra­cel­lu­lar sig­nal–reg­u­lated kinase, and nu- bi­tion, includ­ing induc­ible tyro­sine kinase, com­pared with
clear fac­tor κ light-chain enhancer of acti­vated B cells, path­ways ibrutinib.22,23 In pre­clin­i­
cal stud­ ies, acalabrutinib did not inhibit
impor­tant for malig­nant B cells’ growth and sur­vival.10-12 Ibrutinib anti-CD20 mAb-depen­dent NK-cell medi­ated cyto­tox­ic­ity.22 The
is an irre­vers­ible BTKi that cova­lently binds to the cys­te­ine 481 phase 3 ELEVATE treatment-naive (TN) study was designed to an-
amino acid of the BTK enzyme. It has shown sig­nif­i­cant activ­ity in swer whether acalabrutinib is supe­rior to chemoimmunotherapy
patients with CLL in both front­line and relapsed/refrac­tory (R/R) (Chl-O) in terms of PFS in the front­line treat­ment of CLL. It also
set­tings.13,14 Because of the dura­bil­ity of responses with BTKis as eval­u­ated whether there is a ben­e­fit of adding a more potent an-
monotherapy, there is a ques­tion of whether there is a need for ti-CD20 mAb obinutuzumab to acalabrutinib (AO), although the
an anti-CD20 mAb in con­junc­tion to improve clin­i­cal effi­cacy. study was not powered to detect this dif­fer­ence.19 As expected,
patients who received acalabrutinib ±​­ obinutuzumab had improved
Ibrutinib ± rituximab or obinutuzumab PFS com­pared with Chl-O. After 28 months of fol­low-up, the 2-year
The E1912 trial com­pared FCR to ibrutinib plus rituximab (IR) in PFS rates were not clin­ic ­ ally or sta­tis­ti­cally dif­fer­ent between the

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/68/1851825/68shah.pdf by guest on 13 December 2021


treat­ment-naive patients youn­ger than 70 years (Table 1).15 This AO and acalabrutinib (93% vs 87%) groups. More patients with
study dem­on­strated prolonged PFS and OS of patients receiv­ing AO had CR (13% vs 1%) and uMRD (56% vs 7% of those eval­u­ated)
IR com­pared with FCR. Results of this trial led to approval of the com­pared with acalabrutinib alone. AO resulted in higher rates of
IR com­bi­na­tion in front­line treat­ment of CLL, but this study did grade 3 or higher neutropenia, infec­tions, and infu­sion reac­tions
not answer whether addi­tion of rituximab to ibrutinib is nec­es­ com­pared with acalabrutinib alone. Findings from this study ulti­
sary to obtain sim­i­lar clin­i­cal ben­e­fit. mately led to FDA approval for mar­ket­ing of acalabrutinib ± obinu-
The A041202 study eval­ua ­ ted front­line ther­apy with ibrutinib tuzumab for front­line treat­ment of patients with CLL.
vs IR vs BR in patients with CLL 65 years or older.16 This study As we await the long-term results of the ELEVATE TN study,
dem­on­strated prolonged PFS with ibrutinib-based treat­ments the mature data from the orig­i­nal phase 2 study of sin­gle-agent
com­pared with BR. However, there was no dif­fer­ence in PFS acalabrutinib in treat­ment-naive CLL are encour­ag­ing.24 After a
and OS ben­e­fits between ibrutinib alone and the IR com­bi­na­ median fol­low-up of 53 months, median event-free sur­vival and
tion. Rates of CR (12% vs 7%) and unde­tect­able min­i­mal resid­ dura­tion of response are not reached with a very high ORR of 97%
ual dis­ease (uMRD) (4% vs 1%) were only mar­gin­ally higher with (7% CR, 90% partial response). In addi­tion, 86% of patients are
the IR com­bi­na­tion. The safety pro­file of both ibrutinib alone and still receiv­ing treat­ment, and only 6% have discontinued due to
the IR com­bi­na­tion was sim­il­ar except for slightly higher grade adverse events. The first data describ­ing a head-to-head com­par­
3 or more neutropenia with IR. Another study (NCT02007044) i­son of acalabrutinib and ibrutinib in patients with R/R CLL were
com­par­ing ibrutinib vs IR in treat­ment-naive (13% of total) and presented at the Amer­ic ­ an Society of Clinical Oncology meet­ing
R/R (87% of total) set­tings also failed to detect improve­ment in 2021 (ELEVATE Relapsed/Refractory (RR) Study).25 This study
in PFS with IR but con­cluded that time to CR was faster (11.5 vs found that acalabrutinib was noninferior to ibrutinib in terms of
22.2 months), and patients had lower lev­els of resid­ual dis­ease PFS and had a bet­ter tox­ic­ity pro­file—impor­tantly, a lower inci­
as assessed by flow cytom­e­try with the IR com­bi­na­tion.17 Con- dence of atrial fibril­ la­tion and hyper­ ten­sion.25 This improved
sidering the existing data and con­tin­u­ous appli­ca­tion of BTKis, tox­ic­ity pro­file makes acalabrutinib a very appeal­ing treat­ment
the sig­nif­i­cance of uMRD and its rela­tion to long-term dis­ease option for treat­ment-naive patients with CLL, espe­cially those
erad­i­ca­tion has not been well established. Based on these 2 who are elderly or have mul­ti­ple med­ic ­ al comorbidities.
stud­ies, addi­tion of rituximab to ibrutinib does not seem to pro­
vide any clin­i­cally mean­ing­ful sur­vival ben­e­fit to most patients Summary
with treat­ment-naive CLL. In a pre­clin­i­cal study, ibrutinib’s off- Of the described tri­als that com­pared a BTKi plus anti-CD20 mAb
tar­get inhi­bi­tion of inter­leu­kin 2 induc­ible tyro­sine kinase led to vs BTKi alone, none dem­on­strated a clin­i­cally mean­ing­ful supe­
downregulation of nat­u­ral killer (NK) cell-medi­ated ADCC and ri­or­ity of the anti-CD20 mAb com­bi­na­tion in terms of improved
impaired anti­body-depen­dent phago­cy­to­sis, both of which are PFS or OS. The depth of response (CR and uMRD rates) and the
known anti-CD20 mAb mech­a­nisms of action (Figure 1).20,21 This speed of response (time to CR) are mar­gin­ally improved with the
find­ing has been hypoth­e­sized as one of the expla­na­tions for the addi­tion of an anti-CD20 mAb. As such, there are 2 sce­nar­ios in
lack of sig­nif­i­cant sur­vival ben­e­fit with the addi­tion of rituximab which addi­tion of an anti-CD20 mAb to a BTKi may be clin­i­cally
to ibrutinib. mean­ing­ful. The first sce­nario is in the pres­ence of an active and
The iLLUMINATE study com­ pared front­ line CLL ther­apy of ste­roid-refrac­tory auto­im­mune cytopenia such as ITP or auto­
ibrutinib plus obinutuzumab vs Chl-O.18 This study dem­on­strated im­mune hemo­lytic ane­mia. Both rituximab and BTKi have been
prolonged PFS with the ibrutinib-based reg­i­men in treat­ment- effec­tive as monotherapy in con­trol or res­ol­u­tion of auto­im­mune
naive patients with CLL. Adding obinutuzumab to ibrutinib deep­ cytopenia related to CLL.26-31 Thus, a com­bi­na­tion might lead to
ened responses (35% with uMRD), but seri­ous adverse events deeper and more rapid con­trol of auto­im­mune cytopenia, espe­
were more com­mon in this arm. This study led to Food and Drug cially in the ste­roid-refrac­tory patients. The sec­ond sce­nario, in
Administration (FDA) approval for mar­ket­ing of the ibrutinib and which the upfront addi­tion of a mAb to BTKi could be help­ful, is
obinutuzumab com­bi­na­tion in the front­line set­ting. However, no in patients need­ing rapid con­trol of dis­ease (eg, lymph­ade­nop­
pro­spec­tive head-to-head com­par­i­sons of ibrutinib vs ibrutinib a­thy endan­ger­ing a crit­i­cal organ). From a prac­ti­cal stand­point,
plus obinutuzumab have been done, leav­ ing the ques­ tion of BTKis can take time to be autho­rized by insur­ance and are not
the clin­i­cal sig­nif­i­cance of obinutuzumab’s addi­tion to ibrutinib often included on inpa­tient hos­pi­tal for­mu­lar­ies sec­ond­ary to the
unanswered. high cost of these ther­ap ­ ies. Anti-CD20 mAbs are often read­ily

70  |  Hematology 2021  |  ASH Education Program


Table 1.  Frontline CLL stud­ies with BTKi ± anti-CD20 mono­clo­nal anti­body

Median
Targeted N (targeted fol­low-up,
Study reg­i­men Comparator reg­i­men) ORR, % CRR, % uMRD, % PFS OS Notable AEs mo
E191215 IR FCR IR = 354 95 17 8 (*n = 276) NR (3-y 89%) NR (3-y 98%) Grade ≥3 infec­tions: FCR = 20.3% 33.6
vs IR = 10.5%; grade ≥3 HTN:
FCR = 8.2% vs IR = 18.8%
A04120216 I ± R BR I = 182 I = 93 I = 7 I = 1 I = NR (2-y 87%) I = NR (2-y 90%) Grade ≥3 neutropenia: 38
IR = 182 IR = 94 IR = 12 IR = 4 IR = NR (2-y 88%) IR = NR (2-y 94%) IR = 21% vs I = 15%
NCT0200704417 IR Ibrutinib I = 104 I = 92.3 I = 20.2 NA I = NR (3-y 86%) I = NR (3-y 89%) No sig­nif­i­cant dif­fer­ence in 36
IR = 104 IR = 92.3 IR = 26 IR = NR (3-y 86.9%) IR = NR (3-y 92%) tox­ic­ity pro­file
TN = 27
RR = 181

Prakash Singh Shekhawat


iLLUMINATE18 IO Chl-O IO = 113 88 19 35 (*n = 113) NR (30-mo 79%) NR (30-mo 86%) SAEs in 58% of IO and 35% of 31.3
Chl-O; obinutuzumab-related
SAEs (febrile neutropenia,
throm­bo­cy­to­pe­nia) in 15% of
the IO arm
ELEVATE TN19 A ± O Chl-O A = 179 A = 86 A = 1 A = 7% of those in A = NR (2-y 87%) A = NR (2-y 95%) Grade ≥3 neutropenia: AO = 30% 28.3
AO = 179 AO = 94 AO = 13 CR/CRi (*n = 14) AO = NR (2-y 93%) AO = NR (2-y 95%) vs A = 10%; grade ≥3 infec­tions:
AO = 56% of those in AO = 21% vs A = 14%
CR/CRi (*n = 43)
*Of those evaluable for uMRD in the bone mar­row/periph­eral blood.
A, acalabrutinib; AE, adverse event; CRi, com­plete response with incom­plete count recov­ery; CRR, com­plete response rate; HTN, hypertension; I, ibrutinib; IO, ibrutinib and obinutuzumab;
NA, not avail­­able; NR, not reached; RR, relapsed/refractory; SAE, seri­ous adverse event; TN, treatment-naive.

Anti-CD20 antibodies in CLL | 71


Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/68/1851825/68shah.pdf by guest on 13 December 2021
avail­­able and can expe­dite treat­ment for patients who need a supe­rior PFS in both groups. Based on the results of the MURANO
rapid response. For all­other patients, we rec­om­mend the use study, the com­bi­na­tion of fixed-dura­tion V-R has been approved
of ibrutinib or acalabrutinib alone in front­line treat­ment of CLL. for treat­ment in patients with R/R CLL. No ran­dom­ized study
com­ par­
ing venetoclax vs venetoclax plus rituximab has been
reported. Due to the time-lim­ited nature of this reg­i­men, the deep
responses are more desir­able, and as such, the V-R com­bi­na­tion
CLINICAL CASE (Con­t in­u ed) has become stan­dard of care when used in the R/R set­ting.

Mr. Smith received 6 cycles of obinutuzumab and con­tin­u­ous Venetoclax + obinutuzumab


oral acalabrutinib and achieved clin­ic
­ al remis­sion. Three years Alternatively, in the front­line treat­ment of CLL, venetoclax has
later, he sought treat­ment from the clinic with a pro­gres­sive been stan­dardly paired with obinutuzumab (V-O). The phase 3
return of drenching night sweats and decline of hemo­glo­bin. CLL14 study com­pared front­line CLL ther­apy with a fixed 1-year
No evi­dence of hemo­ly­sis was detected. A next-gen­er­a­tion dura­tion of V-O vs Chl-O.40 Patients who received V-O had sig­nif­
sequenc­ing panel showed a muta­tion of cys­te­ine to ser­ine at i­cantly lon­ger PFS than patients receiv­ing Chl-O (not reached vs

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/68/1851825/68shah.pdf by guest on 13 December 2021


the 481 moi­ety of BTK (C481S). 35.6 months).41 Of the patients who received V-O, 76% achieved
uMRD com­pared with 35% for the Chl-O group 3 months after
com­ple­tion of ther­apy. Based on this study, V-O has been ap-
Venetoclax + rituximab proved by the FDA for mar­ket­ing as a fixed-dura­tion ther­apy for
Venetoclax is an oral, highly selec­tive BCL2 homol­ogy domain treat­ment-naive patients with CLL.
3-mimetic that binds to BCL2, resulting in ini­ti­a­tion of apo­pto­sis
medi­ated by BCL2-asso­ci­ated pro­tein X and BCL2-antag­o­nist/ Summary
killer in primed cells.32 Single-agent venetoclax showed prom­is­ing Although the evi­dence from a direct ran­dom­ized study is lack-
activ­ity in patients with R/R CLL who had pre­vi­ously progressed ing, com­bi­na­tion of an anti-CD20 mAb and venetoclax in CLL
on ibrutinib or idelalisib or had 17p dele­tion.33-35 However, the allows for time-lim­ited ther­apy by achiev­ing clin­i­cally rel­e­vant
com­bi­na­tion of venetoclax with rituximab was found to be a ­ ble improve­ment in PFS, OS, and uMRD rates. Thus, we rec­om­mend
to over­come micro­en­vi­ron­ment-induced resis­tance to veneto- a fixed course of V-R for 2 years in the R/R set­ting and a fixed
clax in pre­clin­i­cal stud­ies, lead­ing to its fur­ther inves­ti­ga­tion in course of V-O in the front­line set­ting.
patients with R/R CLL.36 In the phase 1 study of venetoclax plus
rituximab (V-R) in R/R CLL, no sig­nif­i­cant addi­tional toxicities Umbralisib + ublituximab
were found with addi­tion of rituximab. The com­bi­na­tion resulted Ublituximab is a type I glycoengineered anti-CD20 mAb with en-
in a 2-year PFS of 82% with a 51% CR rate and 57% of the patients hanced ADCC that tar­gets a unique epi­tope on CD20 (Figure 1).
achiev­ing uMRD.37 The depth and dura­ bil­
ity of the response Ublituximab has been recently com­bined with umbralisib, a dual
observed with V-R pop­u­lar­ized the con­cept of a time-lim­ited inhib­i­tor of phosphoinositide 3-kinase δ (PI3Kδ) and casein ki-
course in the treat­ment of patients with CLL, a treat­ment strat­ nase 1ε. In the phase 3 UNITY CLL trial, this com­bi­na­tion (U2)
egy to avoid costs and tox­ic­ity from con­tin­uo ­ us drug expo­sure. yielded improved 2-year PFS of 76.6% com­pared with 52.1% with
The phase 3 ran­dom­ized MURANO trial com­pared the time- Chl-O in treat­ment-naive patients and 2-year PFS of 41.3% with
lim­ited 2-year course of V-R with BR in patients with R/R CLL U2 com­pared with 24.8% with Chl-O in patients with R/R CLL.42
(Table 2).38 Rates of 4-year PFS (57% vs 5%) and OS (85% vs 67%) The safety pro­file of umbralisib was favor­able com­pared with
were higher with V-R com­pared with BR.39 At the end of com­bi­na­ his­tor­i­cal data with other PI3K inhib­i­tors. As such, the U2 com­
tion ther­apy, 62.4% of patients with V-R had uMRD com­pared with bi­na­tion will likely be sub­mit­ted for eval­u­at­ion of FDA approval
13.3% of those with BR, and this end point was asso­ci­ated with within the next few years.

Table 2.  Select piv­otal CLL stud­ies with venetoclax + anti-CD20 mono­clo­nal anti­body

N Median
Targeted (targeted fol­low-up,
Study Population reg­i­men Comparator reg­i­men) ORR, % CRR, % uMRD, % PFS OS Notable AEs mo
MURANO 37-43
R/R V-R BR 194 92 27 62 (*n = 194) 53.6 mo NR (5-y More grade ≥3 59.2
Peripheral 82.1%) neutropenia with
blood V-R; more grade ≥3
febrile neutropenia
and infec­tions with
BR
CLL1439-44 Treatment V-O Chl-O 216 85 50 76 (*n = 216) NR (4-y NR Grade ≥3 neutropenia 52.4
naive Peripheral 74%) (4-y (53%) and throm­
blood 85%) bo­cy­to­pe­nia (13%)
of V-O
*Of those evaluable for uMRD in the bone mar­row/periph­eral blood.
CRR, com­plete response rate; NR, not reached.

72  |  Hematology 2021  |  ASH Education Program


Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/68/1851825/68shah.pdf by guest on 13 December 2021
Figure 2.  How to incorporate anti-CD20 monoclonal antibody into frontline treatment for patients with CLL. *All eligible patients
should be considered for participation in clinical trials if available. **Anti-C20 monoclonal antibody. +Pending Food and Drug Adminis-
tration approval for marketing for CLL at time of submission. GERD, gastroesophageal reflux disease; IGHV, immunoglobulin variable
heavy chain; TLS, tumor lysis syndrome.

CLINICAL CASE (Con­t in­u ed) incor­po­ra­tion of anti-CD20 mAbs for both front­line and R/R CLL
Mr. Smith received V-R ther­ apy, which led to another clin­ i­
cal is sum­ma­rized in Figures 2 and 3. Given the mul­ti­ple choices for
remis­sion. There is some ques­tion whether main­te­nance with ther­apy, a clin­i­cal trial is always pref­er­a­ble for patients who qual­
anti-CD20 mAbs can improve clin­ i­
cal out­comes. Ofatumumab, ify. However, we have lim­ited our algo­rithms to cur­rently avail­­able
a type I human­ized anti-CD20 mAb with a bet­ter com­ple­ment- ther­a­pies. If using che­mo­ther­apy for a young patient (aged <65
depen­dent cyto­tox­ic­ity pro­file com­pared with rituximab (Figure 1), years) with good prog­nos­tic fac­tors, FCR can be con­sid­ered. If a
is approved for main­te­nance ther­apy after chemoimmunother- patient has ste­roid-refrac­tory auto­im­mune cytopenia or a need
apy in patients with R/R CLL but has not been widely accepted for an urgent response, a com­bi­na­tion of a BTKi and an anti-CD20
into clin­i­cal prac­tice due to the emer­gence of novel agents; mAb can be con­sid­ered. Due to ibrutinib’s suspected antag­o­nism
we do not rou­tinely rec­om­mend its uti­li­za­tion.43 Our algo­rithm on of an anti-CD20 mAb mech­a­nism, we would favor an acalabrutinib

Figure 3.  How to incorporate anti-CD20 monoclonal antibody into subsequent lines of treatment for patients with CLL. *All eligible
patients should be considered for participation in clinical trials if available. **Anti-C20 monoclonal antibody. +Pending Food and Drug
Administration approval for marketing for CLL at time of submission. GERD, gastroesophageal reflux disease; IGHV, immunoglobulin
variable heavy chain; TLS, tumor lysis syndrome.
Prakash Singh Shekhawat
Anti-CD20 antibodies in CLL | 73
back­bone with an anti-CD20 mAb. A fixed 1-year course of V-O References
would also be accept­able in this sce­nario. In all­other patients, 1. Maloney DG. Mechanism of action of rituximab. Anticancer Drugs. 2001;​
12(suppl 2):​S1-S4.
we rec­om­mend monotherapy with a BTKi based on the patient’s
2. Fischer K, Bahlo J, Fink AM, et al. Long-term remis­sions after FCR chemoim-
comorbidities (eg, acalabrutinib is pre­ ferred for frail/elderly munotherapy in pre­vi­ously untreated patients with CLL: updated results of
patients due to a favor­able tox­ic­ity pro­file and should be avoided the CLL8 trial. Blood. 2016;127(2):208-215.
in patients with sig­nif­i­cant gas­tro­esoph­a­geal reflux dis­ease requir­ 3. Knauf WU, Lissichkov T, Aldaoud A, et al. Phase III ran­dom­ized study of ben-
ing pro­ton pump inhib­i­tors due to drug inter­ac­tion) or V-O com­ damustine com­pared with chlorambucil in pre­vi­ously untreated patients
with chronic lym­pho­cytic leu­ke­mia. J Clin Oncol. 2009;27(26):4378-4384.
bi­na­tion in front­line treat­ment. In the R/R set­ting for patients with
4. Fischer K, Cramer P, Busch R, et  al. Bendamustine in com­bi­na­tion with
CLL with pre­vi­ous intol­er­ance or pro­gres­sion on a BTKi, we would rituximab for pre­vi­ously untreated patients with chronic lym­pho­cytic leu­
favor 2-year fixed treat­ment with a V-R com­bi­na­tion. For those ke­mia: a mul­ti­cen­ter phase II trial of the Ger­man Chronic Lymphocytic Leu-
patients with venetoclax-refrac­tory dis­ease, we would use mono- kemia Study Group. J Clin Oncol. 2012;30(26):3209-3216.
5. Eichhorst B, Fink A-M, Bahlo J, et al; International Group of Investigators;
therapy with a BTKi. Idelalisib plus rituximab is also approved in
Ger­ man CLL Study Group (GCLLSG). First-line chemoimmunotherapy
the R/R CLL set­ting, but uti­li­za­tion of this PI3K inhib­i­tor requires with bendamustine and rituximab ver­sus fludarabine, cyclo­phos­pha­mide,
care­ful man­age­ment of toxicities. The com­bi­na­tion of umbralisib and rituximab in patients with advanced chronic lym­pho­cytic leu­kae­mia

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/68/1851825/68shah.pdf by guest on 13 December 2021


and ublituximab is an emerg­ing option in patients who are not (CLL10): an inter­na­tional, open-label, randomised, phase 3, non-infe­ri­or­ity
­able to use BTKis or venetoclax in both front­line and R/R set­tings, trial. Lancet Oncol. 2016;17(7):928-942.
6. Freeman CL, Sehn LH. A tale of two antibodies: obinutuzumab ver­sus ritux-
but FDA approval for mar­ket­ing is pend­ing.
imab. Br J Haematol. 2018;182(1):29-45.
7. Tobinai K, Klein C, Oya N, Fingerle-Rowson G. A review of obinutuzumab
(GA101), a novel type II anti-CD20 mono­clo­nal anti­body, for the treat­ment
of patients with B-cell malig­nan­cies. Adv Ther. 2017;34(2):324-356.
Conclusion 8. Goede V, Fischer K, Busch R, et  al. Obinutuzumab plus chlorambucil in
Addition of an anti-CD20 mAb to che­mo­ther­apy is con­sid­ered patients with CLL and coexisting con­di­tions. N Engl J Med. 2014;370(12):1101-
stan­dard in the treat­ment of CLL. However, with the emer­gence 1110.
of potent targeted agents such as BTKis and venetoclax, the ques­ 9. Goede V, Fischer K, Bosch F, et al. Updated sur­vival anal­y­sis from the CLL11
study: obinutuzumab ver­sus rituximab in chemoimmunotherapy-treated
tion of the clin­i­cal impact of mAbs has been raised. Several phase
patients with chronic lym­pho­cytic leu­ke­mia. Blood. 2015;126(23):1733-
3 tri­als have shown that addi­tion of an anti-CD20 mAb to ibrutinib 1733.
does not improve the PFS or OS of patients with CLL, pos­si­bly 10. Craxton A, Jiang A, Kurosaki T, Clark EA. Syk and Bruton’s tyro­sine kinase
related to ibrutinib’s off-tar­get effects. Long-term fol­low-up of are required for B cell anti­gen recep­tor-medi­ated acti­va­tion of the kinase
the acalabrutinib plus obinutuzumab com­bi­na­tion is awaited. On Akt. J Biol Chem. 1999;274(43):30644-30650.
11. Petro JB, Rahman SM, Ballard DW, Khan WN. Bruton’s tyro­sine kinase is
the con­trary, com­bi­na­tion of venetoclax and an anti-CD20 mAb
required for acti­va­tion of IkappaB kinase and nuclear fac­tor kappaB in
leads to deep remis­sions with uMRD, allowing for fixed-dura­tion response to B cell recep­tor engage­ment. J Exp Med. 2000;191(10):1745-
ther­apy and ulti­mately cor­re­lat­ing with improved sur­vival. The 1754.
role of anti-CD20 mAbs remains impor­tant for patients with CLL 12. Tomlinson MG, Woods DB, McMahon M, et al. A con­di­tional form of Bru-
ton’s tyro­sine kinase is suf­fi­cient to acti­vate mul­ti­ple down­stream sig­nal­ing
who have active auto­im­mune cytopenia or need urgent response
path­ways via PLC Gamma 2 in B cells. BMC Immunol. 2001;2:4.
regard­less of a com­bi­na­tion targeted ther­apy being used. Nov- 13. Byrd JC, Furman RR, Coutre SE, et al. Ibrutinib treat­ment for first-line and
el anti-CD20 mAbs such as ublituximab are fur­ther being inves­ relapsed/refrac­tory chronic lym­pho­cytic leu­ke­mia: final anal­y­sis of the
ti­gated in com­bi­na­tion with novel agents. As a whole, treating piv­otal phase Ib/II PCYC-1102 study. Clin Cancer Res. 2020;26(15):3918-
phy­si­cians have a strong arma­men­tar­ium to com­bat CLL. 3927.
14. Burger JA, Tedeschi A, Barr PM, et al; RESONATE-2 Investigators. Ibrutinib
as ini­tial ther­apy for patients with chronic lym­pho­cytic leu­ke­mia. N Engl
Acknowledgment  J Med. 2015;373(25):2425-2437.
D.S. is supported by NIH/NCI Grant K23 CA212271. 15. Shanafelt TD, Wang XV, Kay NE, et al. Ibrutinib-rituximab or chemoimmuno-
therapy for chronic lym­pho­cytic leu­ke­mia. N Engl J Med. 2019;381(5):432-
443.
Conflict-of-inter­est dis­clo­sure 16. Woyach JA, Ruppert AS, Heerema NA, et  al. Ibrutinib reg­i­mens ver­sus
Harsh R. Shah has received research funding from Epizyme. chemoimmunotherapy in older patients with untreated CLL. N Engl J Med.
Deb­or­ ah M. Stephens has received research funding from Acerta, 2018;379(26):2517-2528.
17. Burger JA, Sivina M, Jain N, et  al. Randomized trial of ibrutinib vs ibruti-
Gilead, Karyopharm, Verastem, JUNO, Arqule, and Mingsight.
nib plus rituximab in patients with chronic lym­pho­cytic leu­ke­mia. Blood.
She has served on advi­sory boards for Innate, Jannsen/Pharma- 2019;133(10):1011-1019.
cyclics, Epizyme, Karyopharm, Beigene, Adaptive, AstraZeneca, 18. Moreno C, Greil R, Demirkan F, et al. Ibrutinib plus obinutuzumab ver­sus
and TG Therapeutics. chlorambucil plus obinutuzumab in first-line treat­ ment of chronic lym­
pho­cytic leu­kae­mia (iLLUMINATE): a multicentre, randomised, open-label,
phase 3 trial. Lancet Oncol. 2019;20(1):43-56.
Off-label drug use 19. Sharman JP, Egyed M, Jurczak W, et al. Acalabrutinib with or with­out obinu-
Harsh R. Shah: Ublituximab and umbralisib are discussed and nei- tuzumab ver­sus chlorambucil and obinutuzmab for treat­ment-naive chronic
ther drug is yet FDA approved. lym­pho­cytic leu­kae­mia (ELEVATE TN): a randomised, con­trolled, phase 3 trial.
Lancet. 2020;395(10232):1278-1291.
Deb­or­ ah M. Stephens: Ublituximab and umbralisib are discussed
20. Kohrt HE, Sagiv-Barfi I, Rafiq S, et  al. Ibrutinib antag­ o­nizes rituximab-
and neither drug is yet FDA approved. depen­dent NK cell-medi­ated cyto­tox­ic­ity. Blood. 2014;123(12):1957-1960.
21. Mercedes B, María Belén A, Enrique P, et al. Ibrutinib impairs the phago­cy­
Correspondence to­sis of rituximab-coated leu­ke­mic cells from chronic lym­pho­cytic leu­ke­mia
patients by human mac­ro­phages. Haematologica. 2015;100(4):e140-e142.
Deb­or­ah M. Stephens, Huntsman Cancer Institute, University of 22. Rajasekaran N, Sadaram M, Hebb J, et  al. Three BTK-spe­cific inhib­i­tors,
Utah, 2000 Circle of Hope, Research South 5509, Salt Lake City, in con­trast to ibrutinib, do not antag­o­nize rituximab-depen­dent NK-cell
UT 84112; e-mail: Deb­o­rah​­.stephens@hci​­.utah​­.edu medi­ated cyto­tox­ic­ity. Blood. 2014;124(21):3118-3118.

74  |  Hematology 2021  |  ASH Education Program


23. Barf T, Covey T, Izumi R, et  al. Acalabrutinib (ACP-196): a cova­lent Bruton 35. Jones JA, Mato AR, Wierda WG, et al. Venetoclax for chronic lym­pho­cytic
tyro­sine kinase inhib­i­tor with a dif­fer­en­ti­ated selec­tiv­ity and in vivo potency leu­kae­mia progressing after ibrutinib: an interim anal­y­sis of a multicentre,
pro­file. J Pharmacol Exp Ther. 2017;363(2):240-252. open-label, phase 2 trial. Lancet Oncol. 2018;19(1):65-75.
24. Byrd JC, Woyach JA, Furman RR, et  al. Acalabrutinib in treat­ment-naive 36. Rachel T, Erik S, Katinka W, et  al. Resistance to ABT-199 induced by
chronic lym­pho­cytic leu­ke­mia. Blood. 2021;137(24):3327-3338. micro­en­vi­ron­men­tal sig­nals in chronic lym­pho­cytic leu­ke­mia can be
25. Byrd JC, Hillmen P, Ghia P, et al. First results of a head-to-head trial of acal- counteracted by CD20 antibodies or kinase inhib­i­tors. Haematologica.
abrutinib ver­sus ibrutinib in pre­vi­ously treated chronic lym­pho­cytic leu­ke­mia. 2015;100(8):e302-e306.
J Clin Oncol. 2021;39(15, suppl):7500. 37. Seymour JF, Ma S, Brander DM, et al. Venetoclax plus rituximab in relapsed
26. Garcia-Chavez J, Majluf-Cruz A, Montiel-Cervantes L, Esparza M-G, Vela- or refrac­tory chronic lym­pho­cytic leu­kae­mia: a phase 1b study. Lancet
Ojeda J; Mex­i­can Hematology Study Group. Rituximab ther­apy for chronic Oncol. 2017;18(2):230-240.
and refrac­tory immune throm­bo­cy­to­pe­nic pur­pura: a long-term fol­low-up 38. Seymour JF, Kipps TJ, Eichhorst B, et al. Venetoclax-rituximab in relapsed or
anal­y­sis. Ann Hematol. 2007;86(12):871-877. refrac­tory chronic lym­pho­cytic leu­ke­mia. N Engl J Med. 2018;378(12):1107-
27. Hegde UP, Wilson WH, White T, Cheson BD. Rituximab treat­ment of refrac­ 1120.
tory fludarabine-asso­ci­ated immune throm­bo­cy­to­pe­nia in chronic lym­pho­ 39. Kater AP, Wu JQ , Kipps T, et  al. Venetoclax plus rituximab in relapsed
cytic leu­ke­mia. Blood. 2002;100(6):2260-2262. chronic lym­ pho­cytic leu­ke­
mia: 4-year results and eval­ u­
a­tion of impact
28. Visco C, Barcellini W, Maura F, Neri A, Cortelezzi A, Rodeghiero F. Autoim- of geno­mic com­plex­ity and gene muta­tions from the MURANO phase III
mune cytopenias in chronic lym­pho­cytic leu­ke­mia. Am J Hematol. 2014;​ study. J Clin Oncol. 2020;38(34):4042-4054.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/68/1851825/68shah.pdf by guest on 13 December 2021


89(11):1055-1062. 40. Fischer K, Al-Sawaf O, Bahlo J, et al. Venetoclax and obinutuzumab in patients
29. Gupta N, Kavuru S, Patel D, et  al. Rituximab-based che­ mo­ ther­
apy for with CLL and coexisting con­di­tions. N Engl J Med. 2019;380(23):2225-2236.
ste­roid-refrac­tory auto­im­mune hemo­lytic ane­mia of chronic lym­pho­cytic 41. Al-Sawaf O, Zhang C, Robrecht S, et  al. Clonal dynam­ics after veneto-
leu­ke­mia. Leukemia. 2002;16(10):2092-2095. clax-obinutuzumab ther­apy: novel insights from the ran­dom­ized, phase 3
30. Vitale C, Salvetti C, Griggio V, et al. Pre-existing and treat­ment-emer­gent CLL14 trial. Blood. 2020;136(suppl 1):22-23.
auto­im­mune cytopenias in patients with CLL treated with targeted drugs. 42. Gribben JG, Jurczak W, Jacobs RW, et al. Umbralisib plus ublituximab (U2)
Blood. 2021;137(25):3507-3517. is supe­rior to obinutuzumab plus chlorambucil (O+Chl) in patients with
31. Manda S, Dunbar N, Marx-Wood CR, Danilov AV. Ibrutinib is an effec­tive treat­ment naïve (TN) and relapsed/refrac­tory (R/R) chronic lym­pho­cytic
treat­ment of auto­im­mune haemolytic anae­mia in chronic lym­pho­cytic leu­ leu­ke­mia (CLL): results from the phase 3 unity-CLL study. 2020;136(suppl
kae­mia. Br J Haematol. 2015;170(5):734-736. 1):37-39.
32. Souers AJ, Leverson JD, Boghaert ER, et al. ABT-199, a potent and selec­tive 43. van Oers M, Smolej L, Petrini M, et al. Ofatumumab main­te­nance pro­longs
BCL-2 inhib­i­tor, achieves anti­tu­mor activ­ity while spar­ing plate­lets. Nat pro­gres­sion-free sur­vival in relapsed chronic lym­pho­cytic leu­ke­mia: final
Med. 2013;19(2):202-208. anal­y­sis of the PROLONG study. Blood Cancer J. 2019;9(12):98.
33. Coutre S, Choi M, Furman RR, et al. Venetoclax for patients with chronic
lym­pho­cytic leu­ke­mia who progressed dur­ing or after idelalisib ther­apy.
Blood. 2018;131(15):1704-1711.
34. Stilgenbauer S, Eichhorst B, Schetelig J, et al. Venetoclax for patients with
chronic lym­pho­cytic leu­ke­mia with 17p dele­tion: results from the full pop­ © 2021 by The Amer­i­can Society of Hematology
u­la­tion of a phase II piv­otal trial. J Clin Oncol. 2018;36(19):1973-1980. DOI 10.1182/hema­tol­ogy.2021000234

Prakash Singh Shekhawat


Anti-CD20 antibodies in CLL | 75
CLOTTING AND BLEEDING CONUNDRUMS

Unexplained arterial thrombosis:


approach to diagnosis and treatment

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/76/1851903/76may.pdf by guest on 13 December 2021


Jori E. May1 and Stephan Moll2
Department of Medicine, Division of Hematology/Oncology, University of Alabama at Birmingham, Birmingham, AL; and 2Department of Medicine,
1

Division of Hematology, University of North Carolina School of Medicine, Chapel Hill, NC

Arterial thrombotic events in younger patients without a readily apparent etiology present significant diagnostic and
management challenges. We present a structured approach to diagnosis with consideration of common causes, includ-
ing atherosclerosis and embolism, as well as uncommon causes, including medications and substances, vascular and
anatomic abnormalities, systemic disorders, and thrombophilias. We highlight areas of management that have evolved
within the past 5 years, including the use of dual-pathway inhibition in atherosclerotic disease, antithrombotic therapy
selection in embolic stroke of undetermined source and left ventricular thrombus, the role of closure of patent foramen
ovale for secondary stroke prevention, and the thrombotic potential of coronavirus disease 2019 infection and vaccina-
tion. We conclude with a representative case to illustrate the application of the diagnostic framework and discuss the
importance of consideration of bleeding risk and patient preference in determining the appropriate management plan.

LEARNING OBJECTIVES
• Perform a structured evaluation of patients with unexplained arterial thrombosis, including consideration of 6
categories of causes
• Select appropriate antithrombotic therapy and other necessary interventions to prevent recurrent arterial
thrombotic events

CLINICAL CASE ularly in the younger patient, hematologists may be called


A 36-year-old woman with no medical history sought upon to assist in evaluation and management.
treatment for acute-onset left facial droop and arm weak- Herein, we present a systematic approach to the diagnos-
ness. Computed tomography angiography (CTA) revealed tic evaluation and management of patients with unexplained
occlusion of the right middle cerebral artery at the M2 arterial thrombosis while also highlighting areas where the
segment. She underwent endovascular thrombectomy, literature guiding diagnosis and/or treatment of specific
resulting in complete reperfusion. Given the patient’s sites of arterial thrombosis has evolved in the past 5 years.
young age and no apparent risk factors for stroke, hema-
tology was consulted to assist in the workup of stroke Defining the clot
etiology and to determine a management plan for sec- An overview of our approach to patients with unexplained
ondary stroke prevention. arterial clot is presented in Figure 1. The first step when trying
to determine etiology and subsequent treatment of a throm-
botic event is to confirm the anatomic vascular location and
Introduction understand the resultant end-organ damage. Although arte-
Arterial thrombosis is a leading cause of morbidity and rial thrombosis may be clearly evident in some vascular beds,
mortality worldwide.1 The most common forms of arterial others, such as those supplying visceral organs or central ret-
thrombosis, ischemic heart disease (including acute myo- inal vessels, may be mischaracterized as arterial events when
cardial infarction), and ischemic stroke are managed by they are, in fact, venous. A dedicated review of the patient’s
cardiology and neurology, respectively. However, when presenting symptoms, as well as the imaging along with a
arterial events in these sites or in uncommon anatomic discussion with an expert radiologist or ophthalmologist, can
locations occur without a readily apparent etiology, partic- be helpful.

76 | Hematology 2021 | ASH Education Program


Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/76/1851903/76may.pdf by guest on 13 December 2021
Figure 1.  A structured approach to the diagnosis and management of unexplained arterial thrombosis.2-5 *Includes factor V Leiden,
prothrombin G20210A mutation, protein C deficiency, protein S deficiency, and antithrombin deficiency. aCL, anticardiolipin; aβ2GPI,
anti-β2 glycoprotein 1; CBC, complete blood count; CRP, C-reactive protein; DM, diabetes; ESR, erythrocyte sedimentation rate;
HbA1c, hemoglobin A1c; HLD, hyperlipidemia; HTN, hypertension; LA, lupus anticoagulant; PCR, polymerase chain reaction; PNH,
paroxysmal nocturnal hemoglobinuria; TEE, transesophageal echocardiogram; TTE, transthoracic echocardiogram; UA, urinalysis.

Once an arte­rial throm­botic event is con­firmed, a lit­er­a­ture fac­tors, as well as pathol­ogy spec­i­mens if avail­­able. In imag­ing
search for up-to-date com­ pre­
hen­sive reviews, as well as new eval­u­a­tions, iden­ti­fi­ca­tion of lumi­nal narrowing is often used to
devel­op­ments, rel­e­vant to the site of throm­bo­sis may be a good deter­mine ath­ero­scle­rotic eti­­ol­ogy of a throm­botic event despite
next step, as the approach to arte­rial events varies sig­nif­i­cantly by the fact that vis­i­ble narrowing of an artery is a poor pre­dic­tor of
ana­tomic loca­tion and may rap­idly evolve. Helpful recent reviews plaque vul­ner­a­bil­ity.16 This is par­tic­u­larly rel­e­vant in the eval­u­a­tion
for spe­cific sites of arte­rial throm­bo­sis are presented in Figure 2.6-14 of ische­mic stroke—tra­di­tional cri­te­ria require >50% ste­no­sis to
des­ig­nate large-artery ath­ero­scle­ro­sis, but stud­ies sup­port that
Diagnosis and man­age­ment nonstenotic plaques may also rup­ture and cause stroke.17 There-
Next, the approach to eval­u­a­tion and man­age­ment can be guid- fore, eval­u­a­tion of images for the pres­ence of nonstenotic plaque
ed by con­sid­er­ation of 6 categories that can pre­cip­i­tate arte­rial and cal­ci­fi­ca­tion may also be use­ful.
throm­bo­sis: (1) ath­ero­scle­ro­sis, (2) embolism, (3) sys­temic dis­or­ Given the lim­i­ta­tions of imag­ing, thor­ough eval­u­a­tion of ath­
ders, (4) med­i­ca­tions/sub­stances, (5) vas­cu­lar/ana­tomic abnor­ ero­scle­rotic risk fac­tors should be pur­sued and, if iden­ti­fied, man­
mal­i­ties, and (6) thrombophilias (Figure 1). Most arte­rial events aged aggres­sively. Lipoprotein(a) is an emerg­ing bio­marker of
occur due to ath­ero­scle­ro­sis and embolism, so focused eval­u­a­ ath­ero­scle­rotic car­dio­vas­cu­lar dis­ease (ASCVD) risk, and guide­
tion for these causes is recommended prior to pro­ceed­ing with lines now rec­om­mend con­sid­er­ation of test­ing in patients with
eval­u­a­tion for less com­mon causes. pre­ma­ture ASCVD or ische­mic stroke (<55 years of age), fam­ily
Importantly, in all­patients in whom antithrombotic ther­apy is his­tory of ASCVD, or recur­rent/pro­gres­sive ASCVD despite opti­
being con­sid­ered, risk of recur­rent throm­bo­sis must be bal­anced mal lipid-low­er­ing ther­apy.2 New treat­ments, includ­ing propro-
against risk of bleed­ing, so we rec­om­mend a directed his­tory for tein convertase sub­til­i­sin/kexin type 9 inhib­i­tors, show prom­ise
bleed­ing risk assess­ment. Risk cal­cu­la­tors15 may be used to guide in lipo­pro­tein(a) low­er­ing and decreas­ing the risk of recur­rent
questioning and incor­po­rated into the risk con­ver­sa­tion with rec­ ische­mic events.
og­ni­tion that they are not val­i­dated in this rare pop­u­la­tion. Management: Antiplatelet ther­apy has long been the foun­
da­tion of treat­ment and pre­ven­tion of ath­ero­scle­rotic events.
Atherosclerosis However, recent stud­ies in sta­ble cor­o­nary artery dis­ease18
Diagnosis: The diag­no­sis of ath­ero­scle­rotic dis­ease is often based and periph­eral artery dis­ease after revas­cu­lar­i­za­tion19 have
on a com­bi­na­tion of imag­ing find­ings and the pres­ence of risk dem­on­strated the effi­cacy of dual-path­way inhi­bi­tion (DPI;
Prakash Singh Shekhawat
Unexplained arte­rial throm­bo­sis  | 77
Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/76/1851903/76may.pdf by guest on 13 December 2021
Figure 2. Anatomic sites of unexplained acute arterial thrombosis with citations for relevant comprehensive reviews.6-14 *Organ
infarctions can occur due to arterial thrombosis but also venous thrombosis, low flow states, and other causes. “Defining the clot”
(ie, determining if the end-organ infarction is caused by arterial thrombosis) is a first step in determining further workup and best
management.

not to be con­ fused with dual antiplatelet ther­ apy), incor­ la­ture, the find­ing of mul­ti­ple infarcts in dif­fer­ent vas­cu­lar ter­
po­rat­ing very low-dose anticoagulation (rivaroxaban 2.5 mg ri­to­ries has clas­si­cally been con­sid­ered an indi­ca­tor of embo-
twice daily) with antiplatelet ther­apy.20 Because of sim­i­lar­ lism, but a recent inves­ti­ga­tion revealed that the pres­ence of
ity in nomen­cla­ture between DPI and dual antiplatelet ther­ this pat­tern is unable to con­sis­tently pre­dict, and its absence
apy, as well as sim­i­lar­ity between very low-dose rivaroxaban is unable to con­sis­tently exclude, an embolic mech­a­nism.21 In
2.5 mg and low-dose apixaban 2.5 mg, care­ful use of ter­mi­nol­ con­trast, a lacu­nar infarct (defined as a sin­gle, small [<15 mm in
ogy and care­ful selec­tion of direct oral anti­co­ag­u­lant dose diam­e­ter], sub­cor­ti­cal infarct in the dis­tri­bu­tion of a per­fo­rat­ing
are essen­tial to pre­vent med­i­ca­tion error (Table 1). The role of artery)22 may sug­gest small-ves­sel dis­ease and can be help­ful to
DPI beyond cur­rently approved indi­ca­tions (sta­ble cor­o­nary exclude embolic causes. Also, infarcts in cer­tain ana­tomic sites
artery dis­ease, periph­eral artery dis­ease after revas­cu­lar­i­za­ are more com­monly caused by embolic phe­nom­e­non (eg, renal
tion) has not been stud­ied, but we expect it may be increas­ and splenic infarc­tions occur due to embolism in 68% and 43%,
ingly con­sid­ered for events in uncom­mon ana­tomic loca­tions respec­tively13). Again, a dis­cus­sion with an expert radi­ol­o­gist
where lim­ited evi­dence exists to guide antithrombotic ther­ and a lit­er­a­ture review may be help­ful.
apy selec­tion. Cardiac imag­ing with trans­tho­racic echo­car­dio­gram is often
the first step in the eval­u­a­tion for an embolic event, to iden­tify
Embolism increased atrial size, val­vu­lar dis­ease, and intra­car­diac mate­rial.
Diagnosis: Embolic events can arise from the heart, where Initial eval­u­a­tion for pat­ent fora­men ovale (PFO), through which
throm­bus forms due to struc­tural or func­tional abnor­mal­i­ties, venous mate­rial can enter arte­rial cir­cu­la­tion, is performed with
but emboli can orig­in ­ ate any­where in the vas­cu­la­ture and be the use of agi­tated saline while the patient is coughing and/or
com­posed of many mate­ri­als (eg, thromboemboli, atheroem- performing a Valsalva maneu­ver (a “bub­ble study”). Transesopha-
boli, fat emboli, sep­tic emboli). Certain imag­ing char­ac­ter­is­tics geal echo­car­di­og­ra­phy is the gold stan­dard for PFO mor­pho­logic
(eg, dif­fer­ent ages of ische­mic changes in the same vas­cu­lar char­ac­ter­iza­tion and valve eval­u­a­tion and should be con­sid­ered
ter­ri­tory) can sug­gest embolic eti­­ol­ogy. In the cere­bral vas­cu­ if ini­tial stud­ies are unreavealing.23 Imaging of surrounding arteries

78  |  Hematology 2021  |  ASH Education Program


Table 1.  Terminology for anticoagulation and antiplatelet ther­apy in arte­rial throm­bo­sis

Term Abbreviation Meaning


Combination ther­apy Dual antiplatelet ther­apy DAPT Aspirin 81 mg + P2Y12 inhib­i­tor
Dual antithrombotic ther­apy DAT Anticoagulation + one
antiplatelet agent (com­monly
in patients with AF and CAD)
Dual path­way inhi­bi­tion DPI Very low-dose anticoagulation +
antiplatelet agent(s)
Triple antithrombotic ther­apy TAT Anticoagulation + 2 antiplatelet
Triple ther­apy TT agents
DOAC dos­ing Standard-dose DOAC — Apixaban 5 mg BID
Dabigatran 150 mg BID
Dabigatran 220 mg daily

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/76/1851903/76may.pdf by guest on 13 December 2021


Edoxaban 60 mg daily
Rivaroxaban 20 mg daily
Reduced-dose DOAC* — Apixaban 2.5 mg BID†
Rivaroxaban 15 mg daily‡
Low dose DOAC — Apixaban 2.5 mg BID
Prophylactic dose DOAC Rivaroxaban 10 mg daily
Very low-dose DOAC — Rivaroxaban 2.5 mg BID
*Dosing reg­i­mens for patients with renal impair­ment, low body weight, and/or advanced age. See apixaban and rivaroxaban‡ details.


Dosing for patients with AF who meet 2 of the fol­low­ing 3 cri­te­ria: cre­at­i­nine ≥1.5 mg/dL, age ≥80 years, and weight ≤60 kg.

Dosing used in patients with AF with cre­at­i­nine clear­ance ≤50 mL/min.
AF, atrial fibril­la­tion; CAD, cor­o­nary artery dis­ease; DAPT, dual antiplatelet ther­apy; BID, twice daily.

(ie, carotid arteries, aorta) is indi­cated if there is con­cern for an recommending war­fa­rin,27-29 off-label use of direct oral anti­co­ag­
atheroembolic eti­­ol­ogy. Ambulatory rhythm mon­i­tor­ing is also u­lants (DOACs) is com­mon. A recent ret­ro­spec­tive cohort study
recommended, although opti­mal dura­tion and pre­ferred devices (RED VELVT, Retrospective Evaluation of DOACs and Vascular
are debated. One group of experts favors at least 30 days in Endpoints of Left Ventricular Thrombi) intro­duced fur­ther uncer­
ische­mic stroke, with con­sid­er­ation of implant­able loop record­ tainty, as patients tak­ing DOACs had an increased risk of stroke
ers in high-risk patients.24 However, existing data have not yet or sys­temic embolism com­pared with war­fa­rin.30 There are many
dem­on­strated that increased arrhyth­mia detec­tion via extended lim­i­ta­tions to these data, includ­ing ret­ro­spec­tive and obser­va­
mon­ i­
tor­
ing reduces risk of recur­ rent stroke, and the opti­ mal tional design, unknown DOAC dos­ing reg­i­mens, and fre­quent
treat­ment for device-detected brief runs of atrial fibril­la­tion is the cross­over, and a sub­se­quent small, unblinded ran­dom­ized trial
sub­ject of ongo­ing clin­i­cal tri­als (NCT01938248, NCT02618577). (Comparative Study of Oral Anticoagulation in Left Ventricular
Management: When car­diac throm­bo­em­bo­lism asso­ci­ated Thrombi, No-LVT) dem­on­strated rivaroxaban as being noninferior
with atrial fibril­la­tion is iden­ti­fied, man­age­ment is straight­for­ to war­fa­rin.31 Pending addi­tional data, we favor war­fa­rin for left
ward, and evi­ dence-based guide­ lines exist. The man­ age­ment ven­tric­u­lar throm­bus but con­sider patient-spe­cific fac­tors and
of patients with pre­sumed cardioembolism with­out iden­ti­fi­able have a detailed dis­cus­sion of data lim­i­ta­tions with all­patients.
arrhyth­mia and with­out a documented intra­car­diac throm­bo­em­ The role of PFO clo­sure in the pre­ven­tion of recur­rent embolic
bolic source has been explored in ische­mic stroke with the cre­ events is also debated and has been exten­sively reviewed else­
a­tion of the cat­e­gory of embolic stroke of unde­ter­mined source where.23 PFO is pres­ent in 25% of the adult pop­u­la­tion,32 so the
(ESUS).25 It was hypoth­e­sized that ESUS was com­monly caused by chal­lenge for cli­ni­cians is to deter­mine when a PFO is coin­ci­
covert atrial fibril­la­tion, and there­fore, anticoagulation would be den­tal, there­fore not warranting clo­sure, vs caus­a­tive, there­
supe­rior to antiplatelet ther­apy for sec­ond­ary pre­ven­tion. How- fore warranting clo­sure. Updated guide­lines from the Amer­i­can
ever, 2 large ran­dom­ized tri­als in patients with ESUS revealed that Academy of Neurology out­line fac­tors to con­sider when deter­
the use of anticoagulation (rivaroxaban 15 mg daily, dabigatran min­ing the value of PFO clo­sure.22 An algo­rithm based on these
150 mg or 100 mg twice daily) com­pared with aspi­rin 100 mg daily guide­lines is presented in Figure 3. Importantly, no sin­gle fac­tor
did not decrease recur­rence yet was asso­ci­ated with increased is suf­fi­cient to jus­tify inter­ven­tion, so a com­pre­hen­sive, patient-
bleed­ing. Therefore, antiplatelet agents remain the pre­ferred anti- spe­cific eval­u­a­tion in close col­lab­o­ra­tion with neu­rol­ogy and
thrombotic agent in patients with ESUS.26 Consequently, out­side car­di­­ol­ogy is essen­tial, with sig­nif­i­cant con­sid­er­ation of patient
of clinic tri­als, there is no cur­rent clin­i­cal use­ful­ness of defin­ing a pref­er­ence.
stroke as ESUS. The appli­ca­bil­ity of these find­ings to other sites of
unex­plained arte­rial throm­bo­sis is unknown, and the deci­sion to Medications and sub­stances
use antiplatelet ther­apy vs anticoagulation is often empiric. After exten­sive eval­u­a­tion for ath­ero­scle­ro­sis and embolism, we
Management of left ven­tric­u­lar throm­bus is an area of con­ then performed a struc­tured inves­ti­ga­tion of less com­mon causes
tro­versy and care evo­lu­tion, where despite existing guide­lines (Figure 1). Certain med­i­ca­tions are known to increase throm­botic
Prakash Singh Shekhawat
Unexplained arte­rial throm­bo­sis  | 79
Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/76/1851903/76may.pdf by guest on 13 December 2021
Figure 3.  Algorithm for consideration of PFO closure in secondary stroke prevention based on the 2020 Practice Advisory Update
Summary from the American Academy of Neurology.22 *Shunt descriptor refers to degree of right-to-left shunting, not anatomic
size. ^Includes hypertension, diabetes, hyperlipidemia, or smoking. °Single, small (<15 mm), subcortical, in the distribution of single
penetrating artery.

risk, although asso­ci­a­tions with venous events are more c ­ om­mon on age (eg, colonoscopy, mam­mo­gram) and risk fac­tors (eg, low-
than with arte­rial ones. Furthermore, risk is more con­sis­tently dose com­puted tomog­ra­phy of the chest in patients with tobacco
dem­on­strated with car­dio­vas­cu­lar events and ische­mic stroke, use). The ben­e­fit of more exten­sive imag­ing in patients with arte­
as throm­bo­ses in other sites are rare, and it is there­fore dif­fi­cult to rial throm­bo­em­bo­lism has not been inves­ti­gated. However, given
deter­mine cau­sal­ity. Example med­i­ca­tions and sub­stances, along the rar­ity of arte­rial events due to can­cer, supported by the lack
with their pro­posed path­o­phys­i­o­logic mech­a­nism, are presented of ben­e­fit even in patients with venous throm­bo­em­bo­lism,43 it is
in Table 2. unlikely to be ben­e­fi­cial, and we there­fore do not rec­om­mend it.
Additional categories of sys­temic dis­or­ders to con­sider include
Systemic dis­eases auto­im­mune dis­or­ders (eg, rheu­ma­toid arthri­tis, lupus, vas­cu­li­tis)44
Patients with malig­nancy are at increased risk for arte­rial throm­ and hema­to­logic dis­or­ders (eg, mye­lo­pro­lif­er­a­tive neo­plasms,
bo­sis in the 5 months prior to diag­no­sis, with highest risk 1 month par­ox­ys­mal noc­tur­nal hemoglobulinuria [PNH], sickle cell dis­ease,
prior.42 A thor­ough his­tory and phys­i­cal exam­i­na­tion for signs of plasma cell dis­or­ders, cryoglobulinemia), with pro­posed eval­u­a­
malig­nancy should be performed, with can­cer screen­ing based tion outlined in Figure 1.

80  |  Hematology 2021  |  ASH Education Program


Table 2.  Medications asso­ci­ated with increased risk of arte­rial throm­bo­sis33-41

Medication Sites Proposed mech­a­nism(s) Notes


Estrogens33 Stroke, MI Increased plate­let acti­va­tion and Variable risk reported, likely low
procoagulant fac­tors, decreased abso­lute risk with com­monly
anti­co­ag­u­lant and fibri­no­ly­sis used lower estro­gen doses.
fac­tors Higher risk in patients with HTN,
smok­ing
Androgenic ana­bolic ste­roids34 Stroke, MI Accelerated ath­ero­scle­ro­sis, General term for per­for­mance-
coag­u­la­tion abnor­mal­i­ties, enhanc­ing drugs with
ele­vated hemat­o­crit promyogenic and andro­genic
effects
Heparin35 PA > stroke, MI Autoimmune response to hep­a­rin- Well-documented arte­rial
PF4 com­plex caus­ing hep­a­rin- throm­botic risk, but venous
induced throm­bo­cy­to­pe­nia throm­bo­sis more com­mon

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/76/1851903/76may.pdf by guest on 13 December 2021


Intravenous immu­no­glob­u­lin36 Stroke, MI Many pro­posed: hyper­vis­cos­ity, Risk fac­tors: older age, hyper­ten­sion,
plate­let acti­va­tion, vaso­spasm, hyper­cho­les­ter­­ol­emia. Consider
fac­tor XI con­tent lower dose, slower infu­sion rate
Cocaine37 Stroke, MI, PA Platelet acti­va­tion, Contributes to acute throm­bo­sis
vaso­con­stric­tion, accel­er­ated risk and chronic arte­rial
ath­ero­scle­ro­sis remodeling
Tobacco, can­na­bis38 PA Thromboangiitis obliterans Inflammatory vas­cu­lar dis­ease of
(Buerger dis­ease) the small- and medium-sized
ves­sels of the extrem­i­ties
Erythropoiesis-stim­u­lat­ing Stroke, MI Platelet acti­va­tion, hyper­vis­cos­ity, Risk most clearly documented
agents39 hyper­ten­sion, increased in patients with renal dis­ease
procoagulant fac­tors treated to higher hemo­glo­bin
tar­gets
Anticancer ther­a­pies40
  VEGF inhib­i­tors* Stroke, MI Endothelial dys­func­tion, increased Also asso­ci­ated with hem­or­rhage
procoagulant fac­tors,
accel­er­ated ath­ero­scle­ro­sis,
HTN
 Tamoxifen Stroke Unknown Initial tri­als suggested increased
stroke risk but decreased car­diac
mor­tal­ity, but var­i­able in later
tri­als
 Fluorouracil MI Vasospasm, endo­the­lial dam­age, Cardiotoxicity inci­dence 4% to 19%,
increased procoagulant fac­tors but role of throm­bo­sis vs other
mech­a­nisms unclear
  Immune check­point Stroke, MI, PA Increased procoagulant fac­tors, Venous throm­bo­sis more com­mon
inhib­i­tors41 plate­lets acti­va­tion, impaired than arte­rial (reported 13% vs 2%)
fibri­no­ly­sis, accel­er­ated
ath­ero­scle­ro­sis
*Includes bevacizumab, tyro­sine kinase inhib­i­tors (eg, sorafenib), aflibercept.
HTN, hyper­ten­sion; MI, myo­car­dial infarc­tion; PA, periph­eral artery; PF4, plate­let fac­tor 4; VEGF, vas­cu­lar endo­the­lial growth fac­tor.

Emerging data indi­ cate infec­ tion with severe acute respi­ Vascular and ana­tomic dis­or­ders
ra­tory syn­drome coronavirus 2 is asso­ci­ated with an increased Many vas­cu­lar abnor­mal­i­ties with poten­tial to pre­cip­i­tate throm­
throm­botic risk, with reported occur­rences in mul­ti­ple arte­rial bus for­ma­tion have been reviewed in detail.47 Such dis­or­ders may
sites.45 The COVID-19 vac­ cines manufactured by AstraZeneca affect the artery wall (eg, dis­sec­tion, aneu­rysm, fibromuscular
and Johnson & Johnson have also been shown to cause a pro- dys­pla­sia, vas­cu­li­tis) and/or cal­i­ber (eg, exter­nal com­pres­sion,
thrombotic syn­drome that mim­ics hep­a­rin-induced throm­bo­cy­ vaso­spasm). Therefore, existing imag­ ing should be reviewed
to­pe­nia, named throm­bo­sis with throm­bo­cy­to­pe­nia syn­drome closely for ves­sel abnor­mal­i­ties. Additional imag­ing with CTA,
(TTS) or vac­cine-induced immune throm­botic throm­bo­cy­to­pe­nia mag­netic res­o­nance angi­og­ra­phy, or cath­e­ter-directed angio­
(VITT). TTS/VITT pres­ents with throm­bo­cy­to­pe­nia and pri­mar­ily gram may be required to bet­ter visu­al­ize areas of con­cern, with
cen­tral venous sinus throm­bo­sis but has also been reported to unique con­sid­er­ations for each vas­cu­lar bed.48
cause arte­rial throm­bo­sis.46 A care­ful his­tory of poten­tial COVID-
19 expo­sures, vac­ci­na­tion tim­ing and man­u­fac­turer, COVID-19 Thrombophilias
test­ing, and plate­let count (if con­cern for TTS/VITT) should be The asso­ci­a­tion between inherited throm­botic dis­or­ders and
con­sid­ered in all­patients dur­ing the pan­demic. arte­rial throm­bo­sis is not well char­ac­ter­ized. We have pre­vi­ously
Prakash Singh Shekhawat
Unexplained arte­rial throm­bo­sis  | 81
­reviewed the existing lit­er­a­ture,49 which is lim­ited to case series 1.  Atherosclerosis: The patient con­ firmed no diag­ no­sis of
and ret­ro­spec­tive ana­ly­ses. Given the rar­ity of the stron­ger inherit- hyper­ten­sion or dia­be­tes, and she did not use tobacco prod­
ed thrombophilias and their ques­tion­able role in arte­rial throm­bo­ ucts. She was not obese and had no known fam­ily his­tory of
sis for­ma­tion, we only con­sider test­ing after exten­sive eval­u­a­tion pre­ma­ture stroke or car­dio­vas­cu­lar dis­ease. Intracranial vas­cu­lar
for other causes in youn­ger patients (youn­ger than 50-55 years), imag­ing was reviewed with radi­ol­ogy and did not dem­on­strate
par­tic­u­larly if there is a fam­ily his­tory of venous or arte­rial throm­ find­ings concerning for ath­ero­scle­ro­sis. Bilateral carotid ultra­so­
bo­sis. If test­ing is pur­sued, we con­sider eval­u­a­tion for pro­tein C nog­ra­phy was performed and there was no sig­nif­i­cant ste­no­sis.
defi­ciency, pro­tein S defi­ciency, anti­throm­bin defi­ciency, fac­tor V Lipid panel, hemo­glo­bin A1c, and lipo­pro­tein(a) were sent and
Leiden muta­tions, and pro­throm­bin 20210 muta­tions. Testing for with­out abnor­mal­ity.
fac­tor V Leiden and pro­throm­bin 20210 is performed to iden­tify 2.  Embolism: Telemetry dur­ing hos­pi­tal­i­za­tion did not dem­
homo­zy­gous or com­pound het­ero­zy­gous patients only, as iso­ on­strate arrhyth­mia, so arrange­ments were made at dis­charge
lated het­ero­zy­gos­ity does not have a clin­i­cally sig­nif­i­cant asso­ for 30-day ambu­la­tory car­diac mon­i­tor­ing. Trans­tho­racic echo­
ci­
a­tion with arte­ rial throm­botic risk. Importantly, some assays car­dio­gram with nor­mal saline bub­ble study was with­out val­vu­
are inac­cu­rate dur­ing an acute throm­botic epi­sode and while on lar abnor­mal­ity, intra­car­diac mate­rial, and increased atrial size

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/76/1851903/76may.pdf by guest on 13 December 2021


anticoagulation, so it is essen­tial to under­stand assay lim­i­ta­tions but did dem­ on­
strate an intra­ car­diac shunt. Transesophageal
to ensure appro­pri­ate tim­ing of col­lec­tion and cor­rect inter­pre­ta­ echo­car­di­og­ra­phy revealed a PFO with a mild bidi­rec­tional
tion. Antithrombotic agent selec­tion in patients with an iden­ti­fied shunt. Upper and lower extrem­ity ultra­sounds were neg­a­tive for
inherited thrombophilia is again with­out data for guid­ance. We deep vein throm­bo­sis. Per the algo­rithm in Figure 3, con­tin­ued
con­sider the use of anticoagulation, with or with­out aspi­rin 81 mg eval­u­a­tion for alter­na­tive eti­­ol­o­gies was pur­sued before deter­
daily, in patients with low bleed­ing risk but with reg­u­lar eval­ua ­­ min­ing the role of PFO clo­sure.
tion of risk/ben­efi ­ t and con­sid­er­ation of newly published data. 3.  Medications and sub­stances: The patient was tak­ing no
Antiphospholipid syn­ drome (APS) is an acquired thrombo- med­i­ca­tions. She had a pro­ges­ter­one intra­uter­ine device for
philia with a well-documented risk of arte­rial throm­bo­sis, so we con­ tra­cep­tion. She denied use of nonprescribed sub­ stances,
con­sider test­ing based on the updated Sapporo (also known as and a urine drug screen was neg­at­ ive.
Sydney) cri­te­ria5 in all­ patients with­out an iden­ti­fi­able eti­­ol­ogy. 4.  Vascular and ana­tomic: CTA of the head and neck was
Patients with arte­rial throm­bo­sis and antiphospholipid antibodies obtained and did not reveal evi­dence of ste­no­sis or com­pres­
(APLAs) who do not ful­fill Sydney cri­te­ria for APS (eg, low titer sion, dis­sec­tion, or ves­sel wall thick­en­ing.
APLA, pos­i­tive assay on only 1 occa­sion) should be man­aged in 5.  Systemic dis­or­ders: The patient denied recent weight loss,
the same man­ner as APLA-neg­at­ ive patients with a sim­i­lar throm­ fever, rash, joint pain, or sys­temic symp­toms. Physical exam­i­na­tion
botic event.50 The opti­mal antithrombotic strat­egy for patients was with­out adenopathy, organomegaly, or breast masses. She
with con­firmed APS is debated; anticoagulation, antiplatelet ther­ had a nor­mal Pap smear 1 year prior and denied any fam­ily his­tory
apy, or a com­bi­na­tion of both can be con­sid­ered.50 We favor com­ of malig­nancy. A com­plete blood count was with­out abnor­mal­ity,
bi­na­tion ther­apy in patients with a low bleed­ing risk based on a and there was no evi­dence of hemo­ly­sis. JAK2V617F muta­tion test­
small ran­dom­ized trial dem­on­strat­ing lower inci­dence of stroke ing and flow for PNH were con­sid­ered but not sent pend­ing addi­
recur­rence and sim­i­lar bleed­ing inci­dence with war­fa­rin (tar­get tional eval­u­a­tion given low clin­i­cal sus­pi­cion. In the absence of
inter­na­tional nor­mal­ized ratio [INR] 2-3) plus aspi­rin 100 mg daily sys­temic symp­toms concerning for auto­im­mune dis­or­ders, sero­
vs aspi­rin alone.51 When anticoagulation is used, war­fa­rin is pre­ logic test­ing was also deferred. She had received the COVID-19
ferred, given 2 recent tri­als dem­on­strat­ing an increased risk of vac­cine manufactured by Pfizer-BioNTech 2 months prior.
arte­rial throm­bo­sis with rivaroxaban vs war­fa­rin.52,53 However, 6.  Thrombophilias: The patient denied a fam­ ily his­
tory of
debate remains if war­fa­rin is required for all­patients with APS venous and arte­rial throm­bo­sis in first- or sec­ond-degree rel­a­
vs only those with high throm­bo­sis risk fea­tures (tri­ple pos­i­tive), tives. She had had 2 preg­nan­cies with­out com­pli­ca­tions and no
given the 2 afore­men­tioned tri­als pri­mar­ily included tri­ple-pos­i­tive mis­car­riages. In the absence of a fam­ily or per­sonal his­tory, the
patients and had impor­tant methodologic lim­i­ta­tions.54 Although deci­sion was first made to test for APS, an acquired dis­or­der, prior
we gen­er­ally favor war­fa­rin for anticoagulation in patients with to con­sid­er­ation of test­ing for inherited dis­or­ders. Lupus anti­co­ag­
APS, we con­sider DOACs in patients who are sin­gle or dou­ble u­lant returned clearly pos­it­ ive, IgG anticardiolipin was ele­vated at
pos­i­tive and who (1) have tol­er­ated a DOAC for more than a year, 85 microgram of IgG antibody (GPL) (nor­mal, 0-14 GPL), and IgG
(2) strug­gle with the lim­i­ta­tions of war­fa­rin (eg, fluc­tu­a­tion in INR, anti-β2 gly­co­pro­tein I was ele­vated at 95 GPL (nor­mal, 0-20 GPL).
inabil­ity to com­ply with reg­u­lar mon­i­tor­ing), or (3) have an invalid In sum­mary, the diag­nos­tic workup iden­ti­fied a PFO and clearly
INR due to the pres­ence of a lupus anti­co­ag­ul­ant. However, given pos­i­tive APLA (“tri­ple pos­i­tive”), likely representing APS pend­ing
the lim­i­ta­tions of existing data, an informed dis­cus­sion with the repeat test­ing at 12 weeks. After a mul­ti­dis­ci­plin­ary dis­cus­sion
patient and the incor­po­ra­tion of patient pref­er­ence are impor­tant between hema­tol­ogy, car­di­­ol­ogy, and neu­rol­ogy and dis­cus­sion
com­po­nents of anticoagulation selec­tion. with the patient, the deci­sion was made to ini­ti­ate anticoagula-
tion for sec­ond­ary stroke pre­ven­tion for likely APS. Warfarin was
recommended given con­cern for increased arte­rial throm­botic
risk with rivaroxaban. The patient had no sig­nif­i­cant bleed­ing risk
CLINICAL CASE (Con­t in­u ed) fac­tors and was very concerned about the poten­tial for repeat
Additional eval­u­a­tion was performed based on con­sid­er­ation of stroke, so the joint deci­sion was made to add aspi­rin 81 mg daily
the 6 poten­tial causes of unex­plained arte­rial throm­bo­sis out- with close fol­low-up for reg­u­lar reassessment of bleed­ing risk.
lined in Figure 1. PFO clo­sure was not recommended given the small degree of

82  |  Hematology 2021  |  ASH Education Program


right-to-left shunting observed and the fact that the patient would 12. Brett AS, Azizzadeh N, Miller EM, Collins RJ, Seegars MB, Marcus MA.
be con­tin­ued on indef­i­nite anticoagulation for APS regard­less of Assessment of clin­i­cal con­di­tions asso­ci­ated with splenic infarc­tion in
adult patients. JAMA Intern Med. 2020;180(8):1125-1128.
plan for PFO clo­sure. 13. Weber E, Grangeon F, Reynaud Q , et  al. Acute renal and splenic infarc­
tions: a review. QJM. 2020;113(3):186-193.
14. Brandt LJ, Boley SJ. AGA tech­ni­cal review on intes­ti­nal ische­mia. Gastroen-
Conclusion terology. 2000;118(5):954-968.
The diag­no­sis and man­age­ment of unex­plained arte­rial throm­ 15. Klok FA, Huisman MV. How I assess and man­age the risk of bleed­ing in
patients treated for venous throm­bo­em­bo­lism. Blood. 2020;135(10):724–734.
bo­sis are a com­plex, mul­ti­dis­ci­plin­ary effort in which hema­tol­ogy 16. Ambrose JA, Tannenbaum MA, Alexopoulos D, et al. Angiographic pro­gres­
plays an inte­gral role. A sys­tem­atic approach to eval­u­a­tion is war- sion of cor­o­nary artery dis­ease and the devel­op­ment of myo­car­dial infarc­
ranted and includes con­sid­er­ation of com­mon eti­­ol­o­gies (ath­ero­ tion. J Am Coll Cardiol. 1988;12(1):56-62.
scle­ro­sis and throm­bo­em­bo­lism) as well as uncom­mon eti­­ol­o­gies 17. Coutinho JM, Derkatch S, Potvin AR, et  al. Nonstenotic carotid plaque
on CT angi­ og­ ra­
phy in patients with cryp­ to­
genic stroke. Neurology.
(med­i­ca­tions and sub­stances, vas­cu­lar and ana­tomic abnor­mal­
2016;87(7):665-672.
i­ties, sys­temic dis­or­ders, and thrombophilias). Management re- 18. Eikelboom JW, Connolly SJ, Bosch J, et al; COMPASS Investigators. Rivarox-
quires review of avail­­able lit­er­a­ture for spe­cific vas­cu­lar ter­ri­to­ries aban with or with­out aspi­rin in sta­ble car­dio­vas­cu­lar dis­ease. N Engl J Med.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/76/1851903/76may.pdf by guest on 13 December 2021


or con­di­tions and relies heavily on patient-spe­cific throm­botic 2017;377(14):1319-1330.
and bleed­ing risk and patient pref­er­ence. Continued close fol­ 19. Bonaca MP, Bauersachs RM, Anand SS, et al. Rivaroxaban in periph­eral artery
dis­ease after revas­cu­lar­i­za­tion. N Engl J Med. 2020;382(21):1994–2004​.
low-up with reg­u­lar reassessment of the man­age­ment plan is also 20. Capodanno D, Bhatt DL, Eikelboom JW, et al. Dual-path­way inhi­bi­tion for
essen­tial, as new data or new symp­toms may pres­ent over time, sec­ond­ary and ter­tiary antithrombotic pre­ven­tion in car­dio­vas­cu­lar dis­
and new bleed­ing and throm­botic risk fac­tors may develop. ease. Nat Rev Cardiol. 2020;17(4):242-257.
21. Powers WJ, Kam CH, Ritter VS, Fine JP. Diagnostic accu­ racy of acute
infarcts in mul­ti­ple cere­bral cir­cu­la­tions for cardioembolic stroke: lit­er­a­ture
Acknowledgment 
review and meta-anal­y­sis. J Stroke Cerebrovasc Dis. 2020;29(7):104849.
Figures were cre­ated with BioRender​­.com. 22. Messé SR, Gronseth GS, Kent DM, et  al. Practice advi­sory update sum­
mary: pat­ent fora­men ovale and sec­ond­ary stroke pre­ven­tion: report of
Conflict-of-inter­est dis­clo­sure the Guideline Subcommittee of the Amer­i­can Academy of Neurology. Neu-
Jori E. May: no com­pet­ing finan­cial inter­ests to declare. rology. 2020;94(20):876-885.
23. Collado FMS, Poulin M-F, Mur­phy J-J, Jneid H, Kavinsky C-J. Patent fora­men
Stephan Moll: no com­pet­ing finan­cial inter­ests to declare.
ovale clo­sure for stroke pre­ven­tion and other dis­or­ders. J Am Heart Assoc.
2018;7(12).
Off-label drug use 24. Albers GW, Bernstein RA, Brachmann J, et  al. Heart rhythm mon­i­tor­ing
Jori E. May: off-label use of DOACs for left ventricular thrombus strat­e­gies for cryp­to­genic stroke: 2015 diag­nos­tics and mon­i­tor­ing stroke
is discussed. focus group report. J Am Heart Assoc. 2016;5(3):e002944.
25. Hart RG, Diener H-C, Coutts S-B, et al; Cryptogenic stroke/ESUS Interna-
Stephan Moll: off-label use of DOACs for left ventricular thrombus
tional Working Group. Embolic strokes of unde­ter­mined source: the case
is discussed. for a new clin­i­cal con­struct. Lancet Neurol. 2014;13(4):429-438.
26. Ntaios G. Embolic stroke of unde­ter­mined source: JACC review topic of the
Correspondence week. J Am Coll Cardiol. 2020;75(3):333-340.
Jori E. May, Department of Medicine, Division of Hematology/On- 27. Guyatt GH, Akl EA, Crowther M, Gutterman DD, Schuünemann HJ. Anti-
thrombotic ther­apy and pre­ven­tion of throm­bo­sis, 9th ed: Amer­i­can
cology, University of Alabama at Birmingham, 1720 2nd Ave South,
College of Chest Physicians Evidence-Based Clinical Practice Guidelines.
NP 2503, Birmingham, AL 35294; e-mail: jemay@uabmc​­.edu. Chest. 2012;141(2):7S–47S.
28. Kernan WN, Ovbiagele B, Black HR, et  al. Guidelines for the pre­ven­tion
References of stroke in patients with stroke and tran­sient ische­mic attack: a guide­­­
1. Wendelboe AM, Raskob GE. Global bur­den of throm­bo­sis: epi­de­mi­o­logic line for healthcare pro­fes­sion­als from the Amer­i­can Heart Association/
aspects. Circ Res. 2016;118(9):1340-1347. Amer­i­can Stroke Association. Stroke. 2014;45(7):2160-2236.
2. Wilson DP, Jacobson TA, Jones PH, et  al. Use of lipo­pro­tein(a) in clin­i­cal 29. O’Gara PT, Kushner FG, Ascheim DD, et al. 2013 ACCF/AHA guide­line for the
prac­tice: a bio­marker whose time has come. A sci­en­tific state­ment from man­age­ment of ST-ele­va­tion myo­car­dial infarc­tion: a report of the Amer­
the National Lipid Association. J Clin Lipidol. 2019;13(3):374-392. i­can College of Cardiology Foundation/Amer­i­can Heart Association task
3. Roane DW, Griger DR. An approach to diag­no­sis and ini­tial man­age­ment of force on prac­tice guide­lines. Circulation. 2013;127(4):e362–e425.
sys­temic vas­cu­li­tis. Am Fam Physician. 1999;60(5):1421-1430. PMID: 10524486. 30. Robinson AA, Trankle CR, Eubanks G, et al. Off-label use of direct oral anti­
4. Stinton LM, Fritzler MJ. A clin­i­cal approach to auto­an­ti­body test­ing in sys­ co­ag­u­lants com­pared with war­fa­rin for left ven­tric­u­lar thrombi. JAMA Car-
temic auto­im­mune rheu­matic dis­or­ders. Autoimmun Rev. 2007;7(1):77-84. diol. 2020;5(6):685.
5. Miyakis S, Lockshin MD, Atsumi T, et al. International con­sen­sus state­ment 31. Abdelnabi M, Saleh Y, Fareed A, et  al. Comparative study of oral anti-
on an update of the clas­ si­
fi­
ca­
tion cri­
te­
ria for def­i­
nite antiphospholipid coagulation in left ven­tric­u­lar thrombi (no-LVT trial). J Am Coll Cardiol.
syn­drome (APS). J Thromb Haemost. 2006;4(2):295-306. 2021;77(12):1590-1592.
6. Saver JL. Cryptogenic stroke. N Engl J Med. 2016;374(21):2065-2074. 32. Hagen P, Scholz D, Edwards W. Incidence and size of pat­ent fora­men ovale
7. Gulati R, Behfar A, Narula J, et  al. Acute myo­car­dial infarc­tion in young dur­ing the first 10 decades of life: an autopsy study of 965 nor­mal hearts.
indi­vid­u­als. Mayo Clin Proc. 2020;95(1):136-156. Mayo Clin Proc. 1984;59(1):17-20.
8. Weinberg I, Jaff MR. Nonatherosclerotic arte­rial dis­or­ders of the lower 33. Petitti DB. Hormonal con­tra­cep­tives and arte­rial throm­bo­sis—not risk-free
extrem­i­ties. Circulation. 2012;126(2):213-222. but safe enough. N Engl J Med. 2012;366(24):2316-2318.
9. Mart D, Shatzel J, DeLoughery T. Cryptogenic acute limb ische­mia: a ret­ 34. Pope HG, Wood RI, Rogol A, Nyberg F, Bowers L, Bhasin S. Adverse health
ro­spec­tive cohort study defin­ing a pre­vi­ously undescribed clin­i­cal entity. con­se­quences of per­for­mance-enhanc­ing drugs: an Endocrine Society sci­
J Thromb Thrombolysis. 2018;45(3):397-402. en­tific state­ment. Endocr Rev. 2014;35(3):341-375.
10. Hayreh SS. Acute ret­i­nal arte­rial occlu­sive dis­or­ders. Prog Retin Eye Res. 35. Warkentin TE. Heparin-induced throm­bo­cy­to­pe­nia: path­o­gen­e­sis and man­
2011;30(5):359-394. age­ment. Br J Haematol. 2003;121(4):535-555.
11. Massussi M, Scotti A, Lip GYH, Proietti R. Left ven­tric­u­lar throm­bo­sis: new 36. Marie I, Maurey G, Hervé F, Hellot M, Levesque H. Intravenous immu­no­
per­spec­tives on an old prob­lem. Eur Heart J Cardiovasc Pharmacother. glob­u­lin-asso­ci­ated arte­rial and venous throm­bo­sis; report of a series and
2021;7(2):158-167. review of the lit­er­a­ture. Br J Dermatol. 2006;155(4):714-721.

Prakash Singh Shekhawat


Unexplained arte­rial throm­bo­sis  | 83
37. Bachi K, Mani V, Jeyachandran D, Fayad ZA, Goldstein RZ, Alia-Klein N. Vas- 48. Mur­phy DJ, Aghayev A, Steigner ML. Vascular CT and MRI: a prac­ti­cal guide
cular dis­ease in cocaine addic­tion. Atherosclerosis. 2017;262:154-162. to imag­ing pro­to­cols. Insights Imaging. 2018;9(2):215-236.
38. Olin JW. Thromboangiitis obliterans: 110 years old and lit­tle prog­ress made. 49. May JE, Moll S. How I treat unex­ plained arte­ rial throm­
bo­sis. Blood.
J Am Heart Assoc. 2018;7(23):e011214. 2020;136(13):1487-1498.
39. Lippi G, Franchini M, Favaloro E. Thrombotic com­pli­ca­tions of eryth­ro­poi­e­sis- 50. Ruiz-Irastorza G, Cuadrado MJ, Ruiz-Arruza I, et al. Evidence-based rec­om­
stim­u­lat­ing agents. Semin Thromb Hemost. 2010;36(5):537-549. men­da­tions for the pre­ven­tion and long-term man­age­ment of throm­bo­
40. Jiang D, Lee AI. Thrombotic risk from che­mo­ther­apy and other can­cer ther­ sis in antiphospholipid anti­body-pos­i­tive patients: report of a task force
a­pies. Cancer Treat Res. 2019;179:87-101. at the 13th International Congress on Antiphospholipid Antibodies. Lupus.
41. Moik F, Chan W-SE, Wiedemann S, et al. Incidence, risk fac­tors, and out­ 2011;20(2):206-218.
comes of venous and arte­rial throm­bo­em­bo­lism in immune check­point 51. Okuma H, Kitagawa Y, Yasuda T, Tokuoka K, Takagi S. Comparison between
inhib­i­tor ther­apy. Blood. 2021;137(12):1669-1678. sin­gle antiplatelet ther­apy and com­bi­na­tion of antiplatelet and anticoag-
42. Navi BB, Reiner AS, Kamel H, et al. Arterial throm­bo­em­bolic events pre­ced­ ulation ther­apy for sec­ond­ary pre­ven­tion in ische­mic stroke patients with
ing the diag­no­sis of can­cer in older per­sons. Blood. 2019;133(8):781-789. antiphospholipid syn­drome. Int J Med Sci. 2010;7(1):15-18.
43. Carrier M, Lazo-Langner A, Shivakumar S. Screening for occult can­cer in 52. Pengo V, Denas G, Zoppellaro G, et al. Rivaroxaban vs war­fa­rin in high-risk
unpro­voked venous throm­bo­em­bo­lism. J Vasc Surg. 2015;62(6):1679. patients with antiphospholipid syn­drome. Blood. 2018;132(13):1365-1371.
44. Scalera A, Emmi G, Squatrito D, et  al. Thrombosis in auto­im­mune dis­ 53. Ordi-Ros J, Sáez-Comet L, Pérez-Conesa M, et al. Rivaroxaban ver­sus vita­min
eases: a role for immu­no­sup­pres­sive treat­ments? Semin Thromb Hemost. K antag­o­nist in antiphospholipid syn­drome. Ann Int Med. 2019;171(10):685–

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/76/1851903/76may.pdf by guest on 13 December 2021


2016;42(6):650-661. 694.
45. Cheruiyot I, Kipkorir V, Ngure B, Misiani M, Munguti J, Ogeng’O J. Arterial 54. Moll S. Rivaroxaban ver­sus vita­min K antag­o­nists in antiphospholipid syn­
throm­bo­sis in coronavirus dis­ease 2019 patients: a rapid sys­tem­atic review. drome. Ann Int Med. 2020;172(7):510.
Ann Vasc Surg. 2021;70:273-281.
46. Cines DB, Bussel JB. SARS-CoV-2 vac­ cine-induced immune throm­ botic
throm­bo­cy­to­pe­nia. N Engl J Med. 2021;384(23):2254-2256.
47. Moll S. Nonarteriosclerotic dis­or­ders of the arte­rial sys­tem. In: Kitchens C,
Konkle B, Kessler C, eds. Consultative Hemostasis and Thrombosis. 4th ed. © 2021 by The Amer­i­can Society of Hematology
Elsevier; 2019:415-429. DOI 10.1182/hema­tol­ogy.2021000235

84  |  Hematology 2021  |  ASH Education Program


CLOTTING AND BLEEDING CONUNDRUMS

Cryptogenic oozers and bruisers


Kristi J. Smock1,2 and Karen A. Moser1,2

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/85/1851788/85smock.pdf by guest on 13 December 2021


Department of Pathology, University of Utah, Salt Lake City, UT, and 2ARUP Laboratories, Salt Lake City, UT
1

Bleeding disorders with normal, borderline, or nondiagnostic coagulation tests represent a diagnostic challenge. Disor-
ders of primary hemostasis can be further evaluated by additional platelet function testing modalities, platelet electron
microscopy, repeat von Willebrand disease testing, and specialized von Willebrand factor testing beyond the usual
initial panel. Secondary hemostasis is further evaluated by coagulation factor assays, and factor XIII assays are used to
diagnose disorders of fibrin clot stabilization. Fibrinolytic disorders are particularly difficult to diagnose with current
testing options. A significant number of patients remain unclassified after thorough testing; most unclassified patients
have a clinically mild bleeding phenotype, and many may have undiagnosed platelet function disorders. High-through-
put genetic testing using large gene panels for bleeding disorders may allow diagnosis of a larger number of these
patients in the future, but more study is needed. A logical laboratory workup in the context of the clinical setting and
with a high level of expertise regarding test interpretation and limitations facilitates a diagnosis for as many patients
as possible.

LEARNING OBJECTIVES
• Diagnose bleeding disorders that present with normal conventional coagulation tests
• Identify disorders of primary hemostasis that present diagnostic challenges and recommend specialized testing that
may be diagnostically informative
• Explain a multistep approach for evaluating bleeding disorders of secondary hemostasis, clot stabilization, and fibri-
nolysis

Introduction stabilized by factor XIII (FXIII). Fibrinolysis is the normal


Laboratory coagulation testing plays an important role breakdown of fibrin clot that occurs during healing.2
in the diagnosis and management of bleeding disorders.
However, patients with a suspected bleeding disorder who
have normal or nondiagnostic initial coagulation tests pres-
ent a significant challenge. Both inherited and acquired CASE 1
abnormalities should be carefully considered, with close A 15-year-old girl was evaluated for easy bruising, frequent
attention to bleeding assessment tool results, medical his- nosebleeds, and heavy menstrual bleeding. There was a
tory, family history, medications, liver and renal function, vague family history of an uncharacterized mucocutane-
and physical examination findings.1 In this review, we dis- ous bleeding disorder. Her platelet count, blood smear
cuss how to best approach the laboratory workup beyond morphology, basic clotting times, and fibrinogen were
the common tests, including limitations and interpretation normal. Prior von Willebrand testing had been performed
of laboratory testing. The article is organized by hemostasis and showed similar values for VWF antigen (VWF:Ag) and
component: primary hemostasis, secondary hemostasis, ristocetin cofactor activity (VWF:RCo) that were either
and fibrinolysis. Primary hemostasis refers to platelet plug normal or slightly below the reference interval, not defin-
formation that results from platelet adhesion to damaged itive for von Willebrand disease (VWD) and considered
vasculature via von Willebrand factor (VWF) and direct possible “low VWF.” Prior VWF multimer analysis was nor-
platelet binding, secretion of granules, and platelet aggre- mal. She was referred to our laboratory for a repeat von
gation, whereas secondary hemostasis refers to fibrin for- Willebrand panel and platelet aggregation studies.
mation via the coagulation cascade that is subsequently

Prakash Singh Shekhawat


Workup of bleeding disorders | 85
ing domain) and redun­dant plate­let ago­nists (includ­ing col­la­gen
CASE 2 that binds plate­let col­la­gen recep­tors and shear-acti­vated VWF)
A 12-year-old boy sought eval­u­a­tion from the hema­tol­ogy clinic tends to mask mild abnor­mal­i­ties, and a nor­mal clo­sure time re-
after hav­ing mul­ti­ple epi­sodes of intra­mus­cu­lar bleed­ing that sult only excludes more severe VWD (such as types 2 and 3) or
began after he joined his junior high school foot­ball team. He severe plate­let dys­func­tion (such as Glanzmann thrombasthenia
reported a prior epi­sode of prolonged bleed­ing after a tooth and Bernard-Soulier syn­drome).12,13 The PFA-100 and other micro-
extrac­tion. Initial test­ing, includ­ing plate­let count and periph­ fluidics-based assays are ini­ti­ated by the adhe­sion func­tions of
eral smear review, basic clot­ting times, and a von Willebrand VWF and plate­lets and are thus more sen­si­tive to dis­or­ders of
panel (VWF:Ag, VWF:RCo, and fac­tor VIII [FVIII] activ­ity), was adhe­sion. PFA-100 sen­si­tiv­ity to VWD is esti­mated at approx­i­
nor­mal. Due to the high clin­i­cal sus­pi­cion for a bleed­ing dis­ ma­tely 60% to 70% over­all, with milder type 1 phe­no­types and
or­der, there were ques­tions about what addi­tional test­ing to those with “low VWF” as a bleed­ing risk fac­tor more com­monly
per­form. missed.5,12 Although stor­age pool deficiencies and secre­tion de-
fects rep­re­sent the most com­mon her­i­ta­ble plate­let func­tion
abnor­mal­i­ties, stud­ies sug­gest that up to half of these cases are

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/85/1851788/85smock.pdf by guest on 13 December 2021


Initial test­ing for a bleed­ing dis­or­der missed by PFA-100 test­ing.5,12 If there is strong sus­pi­cion for a
Coagulation test­ing is most use­ful in patients with an abnor­mal bleed­ing dis­or­der of pri­mary hemo­sta­sis, spe­cific VWD test­ing
bleed­ing his­tory, and thus a higher like­li­hood of a bleed­ing dis­ pan­els and plate­let aggregometry should be used despite the
or­der, and is less use­ful for screen­ing unse­lected patients (such increased tech­ni­cal and inter­pre­tive com­plex­ity of aggregom-
as in the pre­op­er­a­tive set­ting).3,4 Initial test­ing typ­i­cally includes etry.1,12 Collagen dis­ or­ders, such as Ehlers-Danlos syn­ drome,
plate­let count and mor­phol­ogy review, pro­throm­bin time (PT), should also be con­sid­ered and are eval­u­ated by joint mobil­ity
acti­vated par­tial throm­bo­plas­tin time (aPTT), and fibrin­o­gen stud­ies and genetic test­ing in some cases. Because a sig­nif­i­cant
activ­ity.3,5-7 The most com­mon fibrin­o­gen assay avail­­able in hos­ num­ber of patients with Ehlers-Danlos syn­drome who have an
pi­tal lab­o­ra­to­ries is the Clauss fibrin­o­gen activ­ity assay, which ele­vated bleed­ing score have been shown to also have plate­let
is a calibrated test based on the throm­bin time. The test will be dys­func­tion, plate­let func­tion test­ing is con­sid­ered use­ful in this
abnor­mal in hypofibrinogenemia and most cases of dysfibrino- set­ting.14
genemia. Additional workup of dysfibrinogenemia may include Comprehensive inves­ti­ga­tion for VWD is beyond the scope of
fibrin­o­gen anti­gen, throm­bin time (TT), reptilase time, and ge- this review, but test­ing should include VWF anti­gen and activ­ity
netic test­ing. Testing for VWD and plate­let func­tion dis­or­ders is (such as VWF:RCo or newer assays), FVIII, and addi­tional spe­cial­
some­times included in the ini­tial eval­u­a­tion and, if not, should ized test­ing as needed to con­firm the diag­no­sis and sub­type.15
be the next step due to the fre­quency of these dis­or­ders and Moderate/severe VWD cases are detected most read­ily due to
inabil­ity of rou­tine tests to detect them.4,5 The aPTT is prolonged their more pro­nounced bleed­ing and lab­o­ra­tory abnor­mal­i­ties.
in VWD if the FVIII is suf­fi­ciently low, but the aPTT is nor­mal in Mild type 1 cases can be chal­leng­ing to diag­nose, some­times
most cases, as is the plate­let count.8 Platelet func­tion dis­or­ders presenting with a nor­mal panel due to the acute phase nature
fre­quently have nor­mal plate­let count and mor­phol­ogy. of VWF.8,15,16 If there is high clin­i­cal sus­pi­cion for VWD (his­tory of
In a study designed to eval­ua ­ te the util­ity of com­mon coag­ imme­di­ate muco­cu­ta­ne­ous bleed­ing), repeated mea­sure­ments
u­la­tion screen­ing tests, results from 800 patients referred for may be nec­es­sary to uncover the true base­line val­ues.8,15,17 Due to
bleed­ing dis­or­der eval­u­a­tion were ana­lyzed, and only 11% had 1 estro­gen’s effects on VWF lev­els, women with heavy men­strual
or more abnor­mal­i­ties of PT, aPTT, fibrin­o­gen, or TT.3 The sen­si­ bleed­ing are often tested at the onset of men­ses when estro­
tiv­ity of this group of tests to a clin­i­cally sig­nif­i­cant abnor­mal­ity gen is low­est, and VWD screen­ing should be avoided in women
was only 3.7%, but this increased to 8.5% when a basic VWD on estro­gen ther­apy or dur­ing or imme­di­ately after preg­nancy.
panel was added and to 30% when both a VWD panel and plate­ A recent study of ini­tial and fol­low-up VWF test­ing in women
let func­tion test­ing (by light trans­mis­sion aggregometry) were seen in an emer­gency depart­ment for heavy men­strual bleed­ing
added, suggesting that these should be included in the ini­ showed higher median VWF and FVIII lev­els dur­ing acute bleed­
tial test­ing.3 Early plate­let aggregometry is recommended in a ing as opposed to fol­low-up, high­light­ing the lim­ited diag­nos­tic
recent guide­line from the International Society on Thrombosis util­ity dur­ing acute bleed­ing.15 The authors also noted that if the
and Haemostasis.9 ini­tial FVIII value was near the nor­mal range, the asso­ci­ated VWF
lev­els were closer to base­line, suggesting that because FVIII also
Challenges in diag­no­sis of dis­or­ders of pri­mary has acute phases, close atten­tion to the FVIII value may help
hemo­sta­sis deter­mine if an acute phase response is pres­ent, but find­ings
Patients with dis­or­ders of pri­mary hemo­sta­sis dem­on­strate a should be interpreted with cau­tion.18 Another recent ret­ro­spec­
muco­cu­ta­ne­ous pat­tern of imme­di­ate bleed­ing (prolonged time tive study of 811 pedi­at­ric patients with suspected VWD indi­
to achieve ini­tial hemo­sta­sis).2,10 Although the skin bleed­ing time cated that 70% of these patients had von Willebrand (VW) panel
is an out­dated method for assessing pri­mary hemo­sta­sis, it has results diag­nos­tic of VWD in the first test­ing epi­sode. Patients
been supplanted in many cen­ters by devices such as the Platelet with VW panel val­ues between 30 and 50 IU/dL are likely most
Function Analyzer (PFA)–100 (or the PFA-200 in some loca­tions at risk for repeat test­ing prior to diag­no­sis; higher VW panel val­
out­side of the United States) as an ini­tial screen of pri­mary hemo­ ues are more likely to exclude VWD (neg­at­ive pre­dic­tive value
sta­sis, pri­mar­ily due to acces­si­bil­ity and ease of use.11 Although for VWF anti­gen and activ­ity >75 IU/dL was 94.1% and 97.4%,
the PFA-100 con­cep­tu­ally rep­re­sents a global test for eval­u­at­ing respec­tively).15,19
pri­mary hemo­sta­sis, the pres­ence of high shear (which results in Other set­tings in which an ini­tial VWD panel may be unre-
a VWF con­for­ma­tional change that exposes the plate­let bind­ vealing include rare sub­types of type 2M VWD due to col­la­gen

86  |  Hematology 2021  |  ASH Education Program


bind­ing defects because rou­tine pan­els do not typ­i­cally assess plate­let func­tion dis­or­ders, high­light­ing genetic com­plex­ity, but
this func­tion.20 A col­la­gen bind­ing assay could be performed is not com­monly performed.32
(VWF:CB) and would dem­on­strate decreased col­la­gen bind­ing Laboratory con­sid­er­ations for patients suspected to have a
out of pro­por­tion to the VWF pro­tein level.5,21 A sub­set of type bleed­ing dis­or­der of pri­mary hemo­sta­sis are shown in Table 1.
2B cases can also appear nor­mal in basic VWD pan­els and have
nor­mal plate­let count and VWF multimers as opposed to the Challenges in diag­no­sis of dis­or­ders of sec­ond­ary
clas­sic type 2B pro­file of throm­bo­cy­to­pe­nia, miss­ing large VWF hemo­sta­sis
multimers, and decreased VWF activ­ity with a low activ­ity/anti­ Patients with dis­or­ders of sec­ond­ary hemo­sta­sis dem­on­strate var­
gen ratio.22 Low-dose ristocetin-induced plate­let aggre­ga­tion i­able bleed­ing pat­terns that can include mus­cle and soft tis­sue
can uncover the diag­no­sis by dem­on­strat­ing the abnor­mally bleed­ing and delayed bleed­ing after achiev­ing ini­tial hemo­sta­
high affin­ity of mutant VWF for plate­lets. Cases with nor­mal sis.4,6 Conventional clot­ting times such as PT and aPTT are pro-
multimers have been shown to have dif­fer­ent type 2B muta­ longed by mod­er­ate or severe fac­tor deficiencies but fre­quently
tions and milder bleed­ing as opposed to type 2B patients with lack sen­si­tiv­ity to milder deficiencies that may be clin­i­cally sig­nif­i­
multimeric defects.3,22 Genetic test­ing for VWD is also avail­­able cant with bleed­ing chal­lenges.4,34,35 Coagulation fac­tor assays can

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/85/1851788/85smock.pdf by guest on 13 December 2021


and has proven most use­ful for type 2 sub­types with chal­leng­ be ordered in a step­wise approach focus­ing on more com­mon
ing lab­o­ra­tory phe­no­types or where phe­no­typic test­ing is not dis­or­ders first with assess­ment of fac­tors VIII, IX, and XI via clot-
avail­­able.20 based activ­ity assays. Because some cases of nonsevere hemo­
Acquired von Willebrand syn­drome (AVWS), which is a het­ philia A (HA) dem­on­strate discrepancies between clot-based and
ero­ge­neous dis­or­der due to anti­body-medi­ated clear­ance, chro­mo­genic FVIII assays based on how spe­cific muta­tions affect
adsorp­tion to plate­lets or neo­plas­tic cells, or pro­te­­ol­y­sis due to the FVIII pro­tein, it is use­ful to per­form both types of FVIII assays.36
increased shear stress, can be diag­nos­ti­cally chal­leng­ing. Spe- It is esti­mated that 40% of mild HA cases have sig­nif­i­cant assay dis-
cific clin­i­cal cir­cum­stances fre­quently asso­ci­ated with AVWS crepancies and that 5% to 10% of mild HA cases have nor­mal FVIII
include patients treated with extra­cor­po­real mem­brane oxy­gen­ activ­ity by either assay, which can mask the diag­no­sis.5,36 Clinical
a­tion, heart pump (such as Abiomed’s Impella), and left ven­tric­ phe­no­types have cor­re­lated most closely with the low­est results
u­lar assist devices.23,24 VWF val­ues are often in the nor­mal range observed. A sim­i­lar phe­nom­e­non has been seen in patients with
due to ade­quate VWF pro­duc­tion and acute phas­ing. Even if the nonsevere hemophilia B (HB) when clot-based and chro­mo­genic
abso­lute val­ues are nor­mal, a decreased activ­ity/anti­gen ratio fac­tor IX activ­ity assays are com­pared, which sup­ports a sim­i­lar
(such as VWF:RCo/Ag or VWF:CB/Ag) is help­ful and may indi­ approach.37 Chromogenic fac­tor IX assays are less widely avail­­able
cate clear­ance of large multimers, which occurs in many, but not than chro­mo­genic FVIII assays. Because fac­tor activ­i­ties may be
all­, cases of AVWS.25 Subtle loss of large multimers by VWF elec­ bor­der­line in mild hemo­philia, genetic test­ing (gene sequenc­ing)
tro­pho­re­sis may be the only abnor­mal­ity observed, and upfront to con­firm an HA or HB path­o­genic muta­tion is often help­ful to
multimer anal­y­sis is rea­son­able if AVWS is suspected.25,26 diag­nose mild cases or female car­ri­ers.38,39
Although plate­ let aggre­ga­tion is the pre­ ferred ini­ tial test Additional assays for uncom­mon mild deficiencies of fac­tors
for assessing plate­let func­tion, it is not sen­si­tive to all­dis­or­ II, V, VII, and X could be con­sid­ered if the ini­tial fac­tor assays are
ders and suf­fers in prac­tice from nonstandardization despite nor­mal. Alternatively, a large bat­tery of fac­tor assays under­taken
lit­er­a­ture supporting stan­dard­i­za­tion and har­mo­ni­za­tion.4,9,10,27,28 at once can save time and min­i­mize blood draws. Some lab­o­
Platelet dense gran­ule defi­ciency is thought to be as com­mon ra­to­ries may offer reflex­ive bleed­ing dis­or­der pan­els to assist
as VWD, may dem­on­strate nor­mal aggre­ga­tion results, and is with timely eval­u­a­tion. Acquired fac­tor deficiencies are usu­ally
best assessed by dense gran­ ule counts using whole-mount mul­ti­ple (due to vita­min K defi­ciency, liver dis­ease, con­sump­tive
plate­let elec­tron micros­copy.4,5,13,29 Although elec­tron micros­ coagulopathies, etc.), which has a more pro­nounced effect on
copy test­ing has lim­ited avail­abil­ity, it dem­on­strates supe­rior the PT and aPTT, mak­ing them more likely to be iden­ti­fied in the
per­for­mance char­ac­ter­is­tics com­pared with the more widely ini­tial test­ing. It should also be remem­bered that clot­ting time
avail­­able lumiaggregometry meth­ods that mea­sure aden­o­ pro­lon­ga­tions due to a lupus anti­co­ag­u­lant may mask or con­
sine tri­phos­phate release from plate­let dense gran­ules.4,13,29-31 found iden­ti­fi­ca­tion of a fac­tor defi­ciency or inhib­i­tor. Current
However, aden­o­sine tri­phos­phate secre­tion anal­y­sis may iden­ lab­o­ra­tory test­ing guide­lines for lupus anti­co­ag­u­lant iden­ti­fi­ca­
tify secre­tion defects that are not due to gran­ule defi­ciency.32 tion indi­cate that fac­tor assays should be performed if there is
Scott syn­drome (plate­let fac­tor 3 defi­ciency) is a rare reces­sive sus­pi­cion for a fac­tor defi­ciency (such as a pos­i­tive bleed­ing his­
bleed­ing dis­or­der caused by abnor­mal­i­ties in plate­let procoag- tory or clin­i­cal con­di­tion asso­ci­ated with fac­tor deficiencies).40
ulant activ­ity due to lack of phosphatidylserine (PS) expo­sure Table 2 dem­on­strates an exam­ple of aPTT reagent sen­si­tiv­
after plate­let acti­va­tion. Calcium-depen­dent coag­u­la­tion fac­ ity to pro­gres­sive sin­gle FVIII defi­ciency or pan-defi­ciency of
tors require bind­ing to neg­a­tively charged phos­pho­lip­ids, such all­coag­u­la­tion fac­tors. The upper limit of nor­mal for this aPTT
as PS, pro­mot­ing for­ma­tion of the tenase and prothrombinase reagent is 35 sec­onds, and the aPTT is more sen­si­tive to mul­ti­ple
complexes of coag­u­la­tion and resulting in nor­mal throm­bin and deficiencies as opposed to a sin­gle defi­ciency. Mild HA with FVIII
fibrin gen­er­a­tion. Scott syn­drome is not iden­ti­fied by rou­tine activ­ity in the 20% to 40% range may dem­on­strate a nor­mal or
coag­u­la­tion test­ing or plate­let aggre­ga­tion and requires either min­i­mally prolonged aPTT. Different aPTT reagents may dif­fer in
flow cytometric test­ing of plate­let PS expo­sure or genetic test­ these pat­terns.
ing for ANO6 muta­tions.33 More than 30 inherited plate­let dis­
or­ders, due to abnor­mal­i­ties in more than 50 genes, have been Challenges in diag­no­sis of FXIII defi­ciency
described, although not all­affect plate­let func­tion.7 Next-gen­ FXIII defi­ciency is a rare reces­sive bleed­ing dis­or­der char­ac­ter­
er­a­tion sequenc­ing has been used to inter­ro­gate patients with ized by severe bleed­ing with a delayed bleed­ing pat­tern and
Prakash Singh Shekhawat
Workup of bleed­ing dis­or­ders  |  87
Table 1. Laboratory con­ sid­er­
ations for patients with a suspected bleed­ ing dis­
or­
der of pri­
mary hemo­ sta­sis and nor­ mal
or nondiagnostic ini­tial tests (nor­mal plate­let count/mor­phol­ogy, rou­tine clot­ting times, VWD panel, and plate­let func­tion test­ing)

Problem Laboratory con­sid­er­ations


VWD
Normal vs low VWF vs VWD masked PFA-100 lacks sen­si­tiv­ity and spec­i­fic­ity for VWD; ini­tial panel should include VWF anti­gen, activ­ity, and
by acute phase response FVIII activ­ity.
Avoid test­ing dur­ing acute bleed­ing epi­sodes, ill­ness, and stress.
Use care to avoid trau­matic phle­bot­omy.
Repeated mea­sure­ments may be nec­es­sary due to nor­mal bio­logic var­i­abil­ity of VWF, par­tic­u­larly with
VWF anti­gen and activ­ity val­ues between 30 IU/dL and 50 IU/dL.
Consider val­ues of other acute phase reac­tants drawn con­cur­rently (FVIII, fibrin­o­gen).
Less com­mon VWD sub­types/ Consider type 2M due to a col­la­gen bind­ing defect. First-line activ­ity tests assess plate­let bind­ing
phe­no­types func­tion rather than col­la­gen bind­ing func­tion. Order VWF:CB to assess for decreased VWF:CB/anti­

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/85/1851788/85smock.pdf by guest on 13 December 2021


gen ratio.
Consider type 2B due to a muta­tion that results in a nonclassic lab­o­ra­tory phe­no­type (rel­a­tively nor­
mal activ­ity and anti­gen with­out decreased activ­ity/anti­gen ratio, nor­mal multimers and plate­let
count). Perform LD-RIPA to assess for abnor­mally high affin­ity for plate­lets.
LD-RIPA mixing stud­ies (if avail­­able) can be used to dif­fer­en­ti­ate type 2B from plate­let-type VWD.
VWD genetic test­ing can be performed and is most use­ful for type 2 cases with chal­leng­ing lab­o­ra­tory
phe­no­types or when phe­no­typic test­ing is not avail­­able (LD-RIPA not pos­si­ble if local lab­o­ra­to­ries do
not per­form aggre­ga­tion test­ing and LD-RIPA mixing stud­ies are not widely performed). Also use­ful
to dif­fer­en­ti­ate type 2B from plate­let-type VWD and type 2N from HA.
Acquired VWS Consideration of and appro­pri­ate test­ing for asso­ci­ated dis­or­ders such as auto­im­mune,
lymphoproliferative or mye­lo­pro­lif­er­a­tive neo­plasms, plasma cell dys­cra­sia, and so on.
Absolute VWF activ­ity and anti­gen val­ues often in the nor­mal range; look for decreased activ­ity/anti­
gen ratio.
VWF multimer anal­y­sis recommended as a first-line test since sub­tle defects of large multimers may be
the only abnor­mal­ity observed.
Platelet func­tion
Platelet func­tion tests are PFA-100 lacks sen­si­tiv­ity and spec­i­fic­ity for the most com­mon plate­let func­tion dis­or­ders. Platelet
nonstandardized and variably sen­si­ aggregometry is con­sid­ered the gold-stan­dard test but has more lim­ited avail­abil­ity. Some lab­or­ a­to­
tive to the most com­mon dis­or­ders ries offer aggregometry with an extended ago­nist panel.
Aggregometry is often nor­mal in com­mon dis­or­ders such as dense gran­ule defi­ciency and secre­tion
defects. Consider plate­let EM and lumiaggregometry.
Mild PFDs remain dif­fi­cult to diag­nose; genetic test­ing has some clin­i­cal util­ity but is not in wide­spread
clin­i­cal use. Consider treating undi­ag­nosed dis­or­ders with tranexamic acid and desmopressin.
Abbreviations: EM, elec­tron micros­copy; LD-RIPA, low-dose ristocetin-induced plate­let acti­va­tion; PFD, platelet function disorder.

nor­mal stan­dard coag­u­la­tion tests. FXIII cross-links fibrin and Challenges in diag­no­sis of fibri­no­lytic dis­or­ders
incor­po­rates the fibri­no­lytic inhib­i­tor α2-antiplasmin (A2AP) Rare patients have inherited reces­sive defects in the fibri­no­lytic
to pre­vent pre­ma­ture deg­ra­da­tion.41 Qualitative clot sol­u­bil­ity inhib­i­tors A2AP and plas­min­o­gen acti­va­tor inhib­i­tor 1 (PAI-1),
tests are fre­quently used for screen­ing but have sig­nif­i­cant lim­i­ resulting in a bleed­ing dis­or­der with delayed bleed­ing due to
ta­tions. Solubility tests are nonstandardized between lab­o­ra­to­ increased lysis of fibrin clots.1,43 Routine clot­ting times are nor­
ries and sen­si­tive to only the most severe deficiencies (abnor­ mal.1,44 Testing for A2AP and PAI-1 should be reserved until abnor­
mal with <1% activ­ity in our lab­o­ra­tory).41 They also dem­on­strate mal­i­ties that are more com­mon have been excluded. Testing for
nonspecificity because abnor­mal results occur with hypo- or fibri­no­lytic dis­or­ders can be dif­fic
­ ult to inter­pret due to com­plex
dysfibrinogenemia, although these con­di­tions would likely be inter­ac­tions between fibri­no­lytic acti­va­tors and inhib­i­tors and ef-
iden­ti­fied prior to FXIII test­ing by fibrin­o­gen test­ing.41 Activity fects of preanalytical var­i­ables related to blood draw qual­ity and
assays are pre­ferred because they detect both quan­ti­ta­tive and tim­ing. Clinically avail­­able PAI-1 assays have tech­ni­cal lim­i­ta­tions
qual­it­a­tive abnor­mal­it­ies and are more spe­cific, but they may in the low range and may not be ­able to defin­i­tively iden­tify defi-
have sub­op­ti­mal lower lim­its of quan­ti­ta­tion (our assay can reli­ ciencies.44 PAI-1 is an acute phase reac­tant, which may mask mild
ably quan­tify down to 5% activ­ity) or use meth­ods that may defi­ciency. PAI-1–defi­cient patients have reduced tis­sue plas­min­
over­es­ti­mate FXIII activ­ity.41,42 Some lab­o­ra­to­ries use FXIII anti­ o­gen acti­va­tor anti­gen due to faster clear­ance, and tis­sue plas­
gen tests for ini­tial eval­u­at­ion. FXIII defi­ciency should be sub- min­o­gen acti­va­tor anti­gen should be tested con­cur­rently.
classified as either A or B sub­unit sub­types due to treat­ment Dominantly inherited profibrinolytic plate­lets due to increased
impli­ca­tions (a com­monly used recom­bi­nant ther­apy con­tains uro­ki­nase-type plas­min­o­gen acti­va­tor in plate­let alpha gran­ules
only the A sub­units). Subtyping can be performed using anti­gen result in a bleed­ing dis­or­der known as Que­bec plate­let dis­or­der.
assays or genetic test­ing. Ultimately, accu­rate diag­no­sis and Bleeding is due to intraplatelet plas­min gen­er­a­tion and pro­te­­
clas­si­fi­ca­tion of FXIII defi­ciency typ­i­cally requires a com­bi­na­tion ol­y­sis of plate­let pro­teins and coag­u­la­tion fac­tors in the alpha
of tests to be used.42 gran­ules.44 Coagulation and plate­let aggre­ga­tion test­ing is nor­

88  |  Hematology 2021  |  ASH Education Program


Table 2.  aPTT reagent sen­si­tiv­ity to pro­gres­sive sin­gle FVIII defi­ciency or pan-defi­ciency of all­fac­tors

FVIII activ­ity, % aPTT, s* All fac­tor activ­ity, % aPTT, s*


100 28.6 100 29.4
90 28.6 90 30.1
80 29.6 80 31.9
70 30.7 70 33.9**
60 31.5 60 37.4**
50 32.5 50 43.0
40 34.3** 40 51.9
30 36.1** 30 69.9

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/85/1851788/85smock.pdf by guest on 13 December 2021


20 39.1 20 108.0
15 40.5 15 No clot
10 44.5 10 No clot
5 49.6 5 No clot
1 61.1 1 No clot
0.001 79.6 0.001 No clot
*Reference inter­val for this aPTT reagent (24-35 sec­onds).
**The bold font indicates when the aPTT seconds begin to exceed the reference interval.

mal in this con­di­tion (with the excep­tion of absent epi­neph­rine due to mul­ti­ple gene inter­ac­tions or inter­ac­tion with acquired
response), and genetic test­ing for PLAU gene dupli­ca­tion should fac­tors.32 These pan­els may increase diag­no­sis and under­stand­
be performed for diag­no­sis.44 ing of rare dis­ or­
ders, such as fibri­
no­
lytic dis­
or­
ders, in which
Because vis­co­elas­tic test­ing meth­ods have become more cur­rently avail­­able test­ing is not widely avail­­able, com­plex to
com­mon­place, it is use­ful under­stand the expected pat­terns in per­form or inter­pret, or lim­ited by poor assay per­for­mance char­
patients with fibri­no­lytic dis­or­ders. These assays are often insen­ ac­ter­is­tics.16,42,44 When large pan­els were stud­ied in the set­ting
si­tive to lysis caused by small changes in plas­min­o­gen acti­va­tor of UBD/BUC, the over­all diag­nos­tic yield was low (3.2% for the
lev­els but can show evi­dence of lysis when there are acti­va­tor ThromboGenomics panel in a recent study of 2396 patients), and
lev­els sig­nif­i­cant enough to over­come PAI-1, includ­ing severe more study of high-through­put genetic modal­i­ties is needed.43,46
trauma, car­dio­pul­mo­nary bypass, and the anhepatic phase of Currently, the Amer­i­can Thrombosis and Hemostasis Network, a
liver trans­plant. Viscoelastic test­ing meth­ods have been used to net­work of 140 hemo­sta­sis cen­ters across all­50 states, is offer­
opti­mize trans­fu­sion reg­i­mens or other ther­a­pies in these set­ ing genetic test­ing (whole-gene sequenc­ing) for 30 rare clot­ting
tings, but there is also con­tro­versy around the degree of ben­e­fit and plate­let dis­or­der genes, includ­ing FXIII defi­ciency. This rep­
and around sen­si­tiv­ity and spec­i­fic­ity in other types of patients.45 re­sents a true net­work of diag­nos­tic and ther­a­peu­tic options for
Laboratory con­sid­er­ations for patients suspected to have a patients and fam­i­lies with rare and ultra-rare bleed­ing dis­or­ders,
bleed­ing dis­or­der of sec­ond­ary hemo­sta­sis, clot sta­bi­li­za­tion and the test­ing is cur­rently pro­vided free of charge. Without a
(FXIII), or fibri­no­ly­sis are shown in Table 3. spe­cific diag­no­sis to tai­lor treat­ment, UBC/BUC is often treated
with tranexamic acid and desmopressin with reported suc­cess.1
Commentary on other test­ing modal­i­ties
Optimized lab­o­ra­tory test­ing approaches can max­i­mize the like­li­ Case res­o­lu­tion and con­clu­sion
hood of a defin­i­tive diag­no­sis in patients with a bleed­ing dis­or­der. Case 1
Although some patients are diag­nosed after thor­ough lab­o­ra­ In this female patient with a muco­cu­ta­ne­ous bleed­ing his­tory,
tory inves­ti­ga­tion, oth­ers remain undi­ag­nosed after an exten­sive the repeat VWD panel results again showed bor­der­line low activ­
search and fall into the cat­e­gory of unclas­si­fied bleed­ing dis­or­ ity and anti­gen results with­out clear dis­cor­dance and a nor­mal
ders (UBDs), also known as bleed­ing of unknown cause (BUC).16 multimer pat­tern (VWF:Ag, 55%; VWF:RCo, 48%; activ­ity/anti­
Mild bleed­ing dis­or­ders are over­rep­re­sented in this group, and gen ratio, 0.87). Her plate­lets responded normally to most plate­
mild plate­let func­tion dis­or­ders are underdiagnosed and remain let ago­nists in the aggre­ga­tion study, although the low-dose
diag­nos­ti­cally chal­leng­ing.43 Testing such as vis­co­elas­tic stud­ies, ristocetin-induced plate­let aggre­ga­tion showed an abnor­mally
throm­bin gen­er­a­tion, or other cur­rently research-based test­ing increased response to low-dose ristocetin (0.5  mg/mL) with
do not yet have a clear role in this set­ting due to var­i­able per­for­ com­plete plate­let aggre­ga­tion. Type 2B VWD was con­firmed by
mance in the lit­er­a­ture or lack of clin­i­cal avail­abil­ity.1,5 iden­ti­fi­ca­tion of a muta­tion in VWF exon 28 (p.R1379C).22
Genetic test­ing using large gene pan­els has the poten­tial to
iden­tify rare abnor­mal­i­ties not ade­quately assessed by clin­i­cally Case 2
avail­­able tests and may be use­ful for cer­tain patients, although In this male patient with recent onset of pro­nounced bleed­
var­i­ant inter­pre­ta­tion may be dif­fi­cult.7,43,46,47 Genetic abnor­mal­ ing after mild to mod­er­ate trauma, addi­tional test­ing included
i­ties may cor­re­late poorly with thePrakashclin­i­cal bleed­ ing ten­
d ency
Singh Shekhawat plate­let aggre­ga­tion stud­ies (nor­mal), a com­pre­hen­sive pan-

Workup of bleed­ing dis­or­ders  |  89


Table 3. Laboratory con­sid­er­ations for patients with a suspected bleed­ing dis­or­der of sec­ond­ary hemo­sta­sis, clot sta­bi­li­za­tion, or
fibri­no­ly­sis and nor­mal or nondiagnostic ini­tial tests (rou­tine clot­ting times, fibrin­og
­ en)

Problem Laboratory con­sid­er­ations


Factor deficiencies
Mild fac­tor deficiencies may not - Perform fac­tor activ­ity assays with a step­wise approach focus­ing on more com­mon deficiencies first
pro­long clot­ting times (fac­tors VIII, IX, and XI), followed by less com­mon deficiencies (fac­tors II, V, VII, and X).
- Consider chro­mo­genic fac­tor VIII and IX assays due to poten­tial for clin­i­cally sig­nif­i­cant dis­crep­ant
results in nonsevere hemo­philia; chro­mo­genic fac­tor IX assays are less widely avail­­able.
- In dis­crep­ant nonsevere HA, a VWD panel may pro­vide a clue in the form of a decreased FVIII
activ­ity/VWF:Ag ratio, even if the abso­lute FVIII result is nor­mal; this is also seen in type 2N VWD.
- Factor activ­i­ties may be bor­der­line, confirm mild cases and female car­ri­ers of HA and HB
with genetic test­ing.
Prolongation of clot­ting times by a - Perform fac­tor assays if there is a his­tory of bleed­ing or a clin­i­cal con­di­tion strongly asso­ci­ated with

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/85/1851788/85smock.pdf by guest on 13 December 2021


lupus anti­co­ag­u­lant may mask a fac­tor deficiencies. Lupus anticoagulants often interfere with quantification of factor activity in clot-
fac­tor defi­ciency or inhib­i­tor based factor assays; exercise caution in interpretation.
  FXIII defi­ciency does not pro­long - Clot sol­u­bil­ity tests are most widely avail­­able world­wide but lack sen­si­tiv­ity (pick up only the most
clot­ting times severe deficiencies) and spec­i­fic­ity (abnor­mal with fibrin­o­gen and other abnor­mal­i­ties).
- Activity assays are the pre­ferred first-line tests but may not accu­rately quan­tify the most severe
deficiencies or may over­es­ti­mate FXIII activ­ity.
- Consider FXIII anti­gen test­ing (sen­si­tive to severe defi­ciency at the cost of miss­ing qual­i­ta­tive abnor­
mal­i­ties) and use of test­ing com­bi­na­tions for diag­no­sis.
- FXIII defi­ciency subtyping should be performed by anti­gen test­ing or genetic test­ing due to impor­tant
treat­ment impli­ca­tions.
Fibrinolytic dis­or­ders are poorly - Conditions are rare, and test­ing has lim­ited avail­abil­ity.
defined and do not pro­long - Results are sig­nif­i­cantly affected by blood draw tim­ing and qual­ity.
clot­ting times - Results are dif­fi­cult to inter­pret due to acute phase responses, com­plex inter­ac­tions between fibri­no­
lytic sys­tem com­po­nents, and tech­ni­cal lim­i­ta­tions of assays (deficiencies are dif­fi­cult to iden­tify).
- Test PAI-1 (activ­ity and/or anti­gen) and tis­sue plas­min­o­gen acti­va­tor anti­gen con­cur­rently to look for
expected pat­terns.
- Platelet aggre­ga­tion is mostly nor­mal in Que­bec plate­let syn­drome; per­form genetic test­ing for PLAU
dupli­ca­tion.
- Viscoelastic stud­ies are sen­si­tive to severe defects but may lack sen­si­tiv­ity for mild or mod­er­ate
abnor­mal­i­ties.
- Fibrinolytic dis­or­ders remain dif­fi­cult to diag­nose; genetic test­ing has some clin­i­cal util­ity but is
not in wide­spread clin­i­cal use. Consider treating undi­ag­nosed dis­or­ders with tranexamic acid and
desmopressin

el of clot-based fac­tor assays (nor­mal), and FXIII activ­ity test­ Correspondence


ing (nor­mal). A chro­mo­genic FVIII assay showed a decreased Kristi J. Smock, Department of Pathology, University of Utah,
FVIII activ­ity of 30% (com­pared with the nor­mal clot-based ARUP Laboratories, 500 Chipeta Way, Mail Stop 115-G04, Salt
[1-stage] result of 65%). Mild HA was con­firmed by iden­ti­fi­ca­ Lake City, UT 84108; e-mail: kristi​­.smock@aruplab​­.com.
tion of an F8 gene muta­tion (p.R546W).48 On rereview, his FVIII
activ­ity/VWF:Ag ratio was decreased on the ini­tial VWD panel References
(65%/115%, ratio 0.57), which may have pro­vided a clue to the 1. Thomas W, Downes K, Desborough MJR. Bleeding of unknown cause and
unclas­si­fied bleed­ing dis­or­ders: diag­no­sis, path­o­phys­i­­ol­ogy and man­age­
diag­no­sis. A dis­cor­dant FVIII activ­ity/VWF:Ag ratio is also seen
ment. Haemophilia. 2020;26(6):946-957.
in type 2N VWD.49 2. Lippi G, Favaloro EJ. Laboratory hemo­sta­sis: from biol­ogy to the bench.
In con­clu­sion, in-depth knowl­edge of the var­i­abil­ity of bleed­ Clin Chem Lab Med. 2018;56(7):1035-1045.
ing dis­or­der phe­no­types and lab­o­ra­tory test­ing per­for­mance 3. Hayward CP, Moffat KA, Liu Y. Laboratory inves­ti­ga­tions for bleed­ing dis­or­
ders. Semin Thromb Hemost. 2012;38(7):742-752.
char­ac­ter­is­tics and lim­i­ta­tions resulted in a con­clu­sive diag­no­sis
4. Hayward CPM. How I inves­ti­gate for bleed­ing dis­or­ders. Int J Lab Hematol.
for these patients. 2018;40(suppl 1):6-14.
5. Boender J, Kruip MJ, Leebeek FW. A diag­nos­tic approach to mild bleed­
ing dis­or­ders. J Thromb Haemost. 2016;14(8):1507-1516.
Conflict-of-inter­est dis­clo­sure 6. Lippi G, Franchini M, Favaloro EJ. Diagnostics of inherited bleed­ ing
Kristi J. Smock: no com­pet­ing finan­cial inter­ests to declare. dis­or­ders of sec­ond­ary hemo­sta­sis: an easy guide for rou­tine clin­i­cal lab­o­
Karen A. Moser: no com­pet­ing finan­cial inter­ests to declare. ra­to­ries. Semin Thromb Hemost. 2016;42(5):471-477.
7. Zaninetti C, Greinacher A. Diagnosis of inherited plate­let dis­or­ders on a
blood smear. J Clin Med. 2020;9(2):539.
Off-label drug use 8. Bowman ML, James PD. Controversies in the diag­no­sis of type 1 von Wille-
Kristi J. Smock: Off-label use of tranexamic acid and DDAVP for brand dis­ease. Int J Lab Hematol. 2017;39(suppl 1):61-68.
9. Gresele P; Subcommittee on Platelet Physiology of the International Soci-
bleeding of unknown cause is discussed.
ety on Thrombosis and Hemostasis. Diagnosis of inherited plate­let func­
Karen A. Moser: Off-label use of tranexamic acid and DDAVP for tion dis­or­ders: guid­ance from the SSC of the ISTH. J Thromb Haemost.
bleeding of unknown cause is discussed. 2015;13(2):314-322.

90  |  Hematology 2021  |  ASH Education Program


10. Mezzano D, Quiroga T, Pereira J. The level of lab­o­ra­tory test­ing required for 31. Dupuis A, Bordet J-C, Eckly A, Gachet C. Platelet δ-stor­age pool dis­ease: an
diag­no­sis or exclu­sion of a plate­let func­tion dis­or­der using plate­let aggre­ update. J Clin Med. 2020;9(8):2508.
ga­tion and secre­tion assays. Semin Thromb Hemost. 2009;35(2):242-254. 32. Leo VC, Mor­gan NV, Bem D, et al; UK GAPP Study Group. Use of next-gen­
11. Favaloro EJ, Bonar R. An update on qual­ity con­trol for the PFA-100/PFA-200. er­a­tion sequenc­ing and can­di­date gene anal­y­sis to iden­tify under­ly­ing
Platelets. 2018;29(6):622-627. defects in patients with inherited plate­let func­tion dis­or­ders. J Thromb
12. Favaloro EJ. Clinical util­ity of clo­sure times using the plate­let func­tion ana­ Haemost. 2015;13(4):643-650.
lyzer-100/200. Am J Hematol. 2017;92(4):398-404. 33. Millington-Burgess SL, Harper MT. Gene of the issue: ANO6 and Scott syn­
13. Rand ML, Reddy EC, Isra­els SJ. Laboratory diag­no­sis of inherited plate­let drome. Platelets. 2020;31(7):964-967.
func­tion dis­or­ders. Transfus Apher Sci. 2018;57(4):485-493. 34. Toulon P, Eloit Y, Smahi M, et al. In vitro sen­si­tiv­ity of dif­fer­ent acti­vated
14. Artoni A, Bassotti A, Abbattista M, et al. Hemostatic abnor­mal­i­ties in patients par­tial throm­bo­plas­tin time reagents to mild clot­ting fac­tor deficiencies.
with Ehlers-Danlos syn­drome. J Thromb Haemost. 2018;16(12):2425-2431. Int J Lab Hematol. 2016;38(4):389-396.
15. James PD, Connell NT, Ameer B, et al. ASH ISTH NHF WFH 2021 guide­lines 35. Favaloro EJ, Kershaw G, Mohammed S, Lippi G. How to opti­mize acti­vated
on the diag­no­sis of von Willebrand dis­ease. Blood Adv. 2021;5(1):280-300. par­tial throm­bo­plas­tin time (APTT) test­ing: solu­tions to establishing and
16. Mezzano D, Quiroga T. Diagnostic chal­lenges of inherited mild bleed­ing ver­i­fy­ing nor­mal ref­er­ence inter­vals and assessing APTT reagents for sen­si­
dis­or­ders: a bait for poorly explored clin­i­cal and basic research. J Thromb tiv­ity to hep­a­rin, lupus anti­co­ag­u­lant, and clot­ting fac­tors. Semin Thromb
Haemost. 2019;17(2):257-270. Hemost. 2019;45(1):22-35.
17. Baronciani L, Peyvandi F. How we make an accu­rate diag­no­sis of von Will- 36. Al-Huniti A, Sharathkumar A, Krantz M, Watkinson K, Bhagavathi S. Dis-

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/85/1851788/85smock.pdf by guest on 13 December 2021


ebrand dis­ease. Thromb Res. 2020;196:579-589. crepant hemo­philia A: an underdiagnosed dis­ease entity. Am J Clin Pathol.
18. Brown MC, White MH, Friedberg R, et al. Elevated von Willebrand fac­tor 2020;154(1):78-87.
lev­els dur­ing heavy men­strual bleed­ing epi­sodes limit the diag­nos­tic util­ity 37. Kihlberg K, Strandberg K, Rosén S, Ljung R, Astermark J. Discrepancies
for von Willebrand dis­ease. Res Pract Thromb Haemost. 2021;5(4):e12513. between the one-stage clot­ting assay and the chro­mo­genic assay in hae-
19. Doshi BS, Rogers RS, Whitworth HB, et al. Utility of repeat test­ing in the mophilia B. Haemophilia. 2017;23(4):620-627.
eval­u­a­tion for von Willebrand dis­ease in pedi­at­ric patients. J Thromb Hae- 38. Konkle BA, Huston H, Nakaya Fletcher S. In: Adam MP, Ardinger HH, Pagon
most. 2019;17(11):1838-1847. RA, et al, eds. Hemophilia A. GeneReviews((R)). University of Washington;
20. Sharma R, Flood VH. Advances in the diag­no­sis and treat­ment of von Will- 2017.
ebrand dis­ease. Blood. 2017;130(22):2386-2391. 39. Konkle BA, Huston H, Nakaya Fletcher S. In: Adam MP, Ardinger HH, Pagon
21. Keesler DA, Flood VH. Current issues in diag­no­sis and treat­ment of von RA, et al, eds. Hemophilia B. GeneReviews((R)). University of Washington;
Willebrand dis­ease. Res Pract Thromb Haemost. 2018;2(1):34-41. 2017.
22. Casonato A, Daidone V, Galletta E, Bertomoro A. Type 2B von Willebrand 40. Moffat KA, Ledford-Kraemer MR, Plumhoff EA, et al. Are lab­o­ra­to­ries fol­low­
dis­ease with or with­out large multimers: a dis­tinc­tion of the two sides of ing published rec­om­men­da­tions for lupus anti­co­ag­u­lant test­ing? An inter­
the dis­or­der is long over­due. PLoS One. 2017;12(6):e0179566. na­tional eval­u­a­tion of prac­tices. Thromb Haemost. 2009;101(1):178-184.
23. Goldfarb M, Czer LS, Lam LD, Moriguchi J, Arabia FA, Volod O. High molec­ 41. Karimi M, Peyvandi F, Naderi M, Shapiro A. Factor XIII defi­ciency diag­no­sis:
u­lar weight von Willebrand fac­tor multimer loss and bleed­ing in patients chal­lenges and tools. Int J Lab Hematol. 2018;40(1):3-11.
with short-term mechan­i­cal cir­cu­la­tory sup­port devices: a case series. J 42. Durda MA, Wolberg AS, Kerlin BA. State of the art in fac­tor XIII lab­o­ra­tory
Extra Corpor Technol. 2018;50(2):77-82. assess­ment. Transfus Apher Sci. 2018;57(6):700-704.
24. Geisen U, Heilmann C, Beyersdorf F, et al. Non-sur­gi­cal bleed­ing in patients 43. Sivapalaratnam S, Collins J, Gomez K. Diagnosis of inherited bleed­ing dis­
with ven­tric­u­lar assist devices could be explained by acquired von Wille- or­ders in the geno­mic era. Br J Haematol. 2017;179(3):363-376.
brand dis­ease. Eur J Cardiothorac Surg. 2008;33(4):679-684. 44. Jain S, Acharya SS. Inherited dis­or­ders of the fibri­no­lytic path­way. Transfus
25. Tiede A, Rand JH, Budde U, Ganser A, Federici AB. How I treat the acquired Apher Sci. 2019;58(5):572-577.
von Willebrand syn­drome. Blood. 2011;117(25):6777-6785. 45. Longstaff C. Measuring fibri­no­ly­sis: from research to rou­tine diag­nos­tic
26. Blackshear JL, McRee CW, Safford RE, et al. von Willebrand fac­tor abnor­ assays. J Thromb Haemost. 2018;16(4):652-662.
mal­i­ties and Heyde syn­drome in dys­func­tional heart valve pros­the­ses. 46. Downes K, Megy K, Duarte D, et al; NIHR BioResource. Diagnostic high-
JAMA Cardiol. 2016;1(2):198-204. through­put sequenc­ing of 2396 patients with bleed­ing, throm­botic, and
27. Munnix ICA, Van Oerle R, Verhezen P, et al. Harmonizing light trans­mis­ plate­let dis­or­ders. Blood. 2019;134(23):2082-2091.
sion aggregometry in the Netherlands by implementation of the SSC- 47. Simeoni I, Stephens JC, Hu F, et al. A high-through­put sequenc­ing test for
ISTH guide­line. Platelets. 2021;32(4):516-523. diag­nos­ing inherited bleed­ing, throm­botic, and plate­let dis­or­ders. Blood.
28. Cattaneo M, Cerletti C, Harrison P, et al. Recommendations for the stan­ 2016;127(23):2791-2803.
dard­ i­
za­
tion of light trans­ mis­sion aggregometry: a con­ sen­sus of the 48. Johnsen JM, Fletcher SN, Huston H, et al. Novel approach to genetic anal­y­sis
Working Party from the Platelet Physiology Subcommittee of SSC/ISTH and results in 3000 hemo­philia patients enrolled in the My Life, Our Future
[published online 10 April 2013]. J Thromb Haemost. ini­tia­tive. Blood Adv. 2017;1(13):824-834.
29. Brunet JG, Iyer JK, Badin MS, et  al. Electron micros­copy exam­i­na­tion of 49. Seidizadeh O, Peyvandi F, Mannucci PM. Von Willebrand dis­ease type 2N:
plate­let whole mount prep­a­ra­tions to quan­ti­tate plate­let dense gran­ule an update. J Thromb Haemost. 2021;19(4):909-916.
num­bers: impli­ca­tions for diag­nos­ing suspected plate­let func­tion dis­or­
ders due to dense gran­ule defi­ciency. Int J Lab Hematol. 2018;40(4):400-
407.
30. Badin MS, Graf L, Iyer JK, Moffat KA, Seecharan JL, Hayward CP. Variability
in plate­let dense gran­ule aden­o­sine tri­phos­phate release find­ings amongst
patients tested mul­ti­ple times as part of an assess­ment for a bleed­ing dis­ © 2021 by The Amer­i­can Society of Hematology
or­der. Int J Lab Hematol. 2016;38(6):648-657. DOI 10.1182/hema­tol­ogy.2021000236

Prakash Singh Shekhawat


Workup of bleed­ing dis­or­ders  |  91
CLOTTING AND BLEEDING CONUNDRUMS

Clots in unusual places: lots of stress, limited


data, critical decisions

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/92/1851781/92mathew.pdf by guest on 13 December 2021


Carol Mathew and Marc Zumberg
Internal Medicine, Division of Hematology/Oncology, University of Florida, Gainesville, FL

Although much less common than deep vein thrombosis of the lower extremities or lungs, clots in unusual locations,
including the splanchnic, cerebral, retinal, upper-extremity, and renal locations, present with significant morbidity and
mortality. In the last 2 decades, treatment of clots in these unusual locations is primarily managed medically, with inter-
ventional and surgical approaches reserved for more severe or refractory cases. The hematologist is well positioned to
provide consultation to organ-specific specialties (ie, neurosurgery, hepatology, ophthalmology), especially because
acquired and congenital hypercoagulability plays a major role, and anticoagulation is often the primary treatment. His-
torically, treatment has been based on expert opinion, but systematic reviews and meta-analyses have recently been
published. Various societies have produced guidelines for the treatment of clots in unusual locations; however, random-
ized clinical trial data remain scarce. In the last few years, increasing data have emerged concerning the efficacy of the
direct oral anticoagulants in treating clots in unusual locations. Cases have recently been described highlighting atypical
thrombosis associated with COVID-19 infection as well as with the ChAdOx1 nCoV-19 (AstraZeneca) vaccine and John-
son and Johnson’s Janssen Ad26.COV2.S vaccine. This article reviews clots in unusual locations with an emphasis on the
splanchnic (mesenteric, portal, splenic, hepatic) and cerebral circulation. Through a case-based approach, key questions
are posed, and data are presented to help guide diagnosis and treatment.

LEARNING OBJECTIVES
• Diagnose and treat clots in the splanchnic and cerebral veins despite (1) heterogeneous clinical presentations,
(2) limited high­level evidence, and (3) concomitant bleeding and clotting risks
• Work within a team consisting of organ­specialized physicians as well as hematologists

Introduction (eg, myeloproliferative diseases in splanchnic vein throm­


Although different definitions exist, clots in unusual sites typ­ boses [SVT] and estrogens in cerebral vein thrombosis
ically refer to any location outside of the lower extremities [CVT]) possibly due to complex interactions with the local
and pulmonary arteries.1,2 Unlike the treatment of deep vein vessel wall, altering hemostatic balance and predisposing
thrombosis (DVT) and pulmonary embolism, thrombosis at to thrombosis.4 Familiarity with patients with hypercoagu­
unusual sites is much less common, and treatment is typi­ lable disorders allows the hematologist to maintain a high
cally guided by expert opinion based on observational stud­ index of suspicion of these rare conditions and remain an
ies or small randomized trials.1,3 Despite the rarity of these integral part of diagnosis and treatment.
conditions, they are detected with increasing frequency due During the prior year, clots in unusual locations such as
to modern radiographic imaging, leading to earlier diagno­ the splanchnic, cerebral, and retinal circulation have been
sis and allowing the earlier institution of effective therapy.1 described in patients infected with COVID­19 (Table 1).5,6 In
Often, these unique thromboses are provoked by a addition, a subset of vaccine­related splanchnic vein and
pathologic condition in the organ supplied by the spe­ cerebral vein clots (termed vaccine­induced thrombocy­
cific venous segments (eg, splenic vein thrombosis in a topenia with thrombosis or thrombotic thrombocytopenic
patient with pancreatitis; portal vein thrombosis (PVT) syndrome) that resemble autoimmune heparin­induced
in a patient with cirrhosis).2 Frequently, these events are thrombocytopenia with thrombosis has been described by
associated with acquired and/or congenital hypercoagu­ Greinacher.7 In this case series, 9 patients developed CVT,
lable disorders.2,4 Different thrombotic manifestations can and 3 patients developed SVT after vaccination with the
result from the various acquired and inherited conditions ChAdOx1 nCoV­19 (AstraZeneca) vaccine.7 Thrombosis in

92 | Hematology 2021 | ASH Education Program


Table 1.  COVID-19 vac­ci­na­tion and clots in unusual loca­tions: Thrombosis in the splanch­nic cir­cu­la­tion is often due to a local
key points risk fac­tor such as liver cir­rho­sis, solid tumor, or pan­cre­a­ti­tis, typ­
i­cally in com­bi­na­tion with tran­sient risk fac­tors such as sur­gery or
• The major­ity of throm­botic events asso­ci­ated with COVID-19 are DVT local inflam­ma­tion.13 Myeloproliferative neo­plasms are the most
and pul­mo­nary embolism com­mon sys­temic risk fac­tor, occur­ring in 40% of HVT and 31%
•  A
 n increas­ing num­ber of reports of splanch­nic and CVT have been of non­ma­lig­nant, noncirrhotic PVT.14 Next-gen­er­a­tion sequenc­ing
reported in asso­ci­a­tion with COVID-19 infec­tion has also been recently used and has iden­ti­fied a novel JAK-2-exon
•  A
 lthough the inci­dence of post­vac­ci­na­tion atyp­i­cal throm­bo­sis 12 muta­tion in one-third of tri­ple-neg­a­tive patients with idi­o­
remains quite low, 9 cases of CVT and 3 cases of SVT have been pathic or exclu­sively noncirrhotic SVT.15 Inherited thrombophilic
reported after vac­ci­na­tion with the ChAdOx1 nCoV-19 (AstraZeneca) dis­or­ders are rec­og­nized risk fac­tors reported in mul­ti­ple sys­
vac­cine as well as 6 cases of CVT and 2 of con­com­i­tant SVT after temic reviews, but the inci­dence and pen­e­trance dif­fer between
Johnson & Johnson’s Janssen Ad26.COV2.S vac­cine
the var­i­ous muta­tions and the var­i­ous sites in the splanch­nic cir­
    T
○  he lit­er­a­ture surrounding the inci­dence, risk, and path­o­phys­i­­ol­ogy cu­la­tion.3 SVT is con­sid­ered unpro­voked in 15% to 27% of cases
con­tin­ues to evolve even as this arti­cle was writ­ten
and diag­nosed inci­den­tally in up to one-third of patients.3

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/92/1851781/92mathew.pdf by guest on 13 December 2021


    T
○  hese cases have been asso­ci­ated with throm­bo­cy­to­pe­nia resem­
bling auto­im­mune hep­a­rin-induced throm­bo­cy­to­pe­nia
    T
○  he ISTH has devel­oped treat­ment guide­lines that include
nonheparin anti­co­ag­u­lants as well as IVIG in con­firmed cases
CLINICAL CASE 1
•  M
 ost patients had no obvi­ous under­ly­ing risk fac­tors or known
thrombophilia A 45-year-old man with alco­holic cir­rho­sis noted wors­en­ing
abdom­i­nal dis­ten­tion. Ultrasound revealed a cir­rhotic liver with
•  Most patients were treated with anti­co­ag­u­lant ther­apy
new throm­bo­sis of the PV as well as new asci­tes. The plate­let
•  Mortality was high count was sta­ble at 49 000/µL, and pro­throm­bin time (PT) was
•  C
 OVID-19 test­ing should be con­sid­ered as well as ascer­tain­ment of 19 sec­onds.
vac­cine sta­tus, prod­uct, and tim­ing in patients who pres­ent with
unex­plained splanch­nic or CVT
IVIG, intravenous immunoglobulin. Question
What is the role of anticoagulation in PVT?
the cere­bral and splanch­nic cir­cu­la­tion has also been described
after Johnson and Johnson’s Janssen Ad26.COV2.S vac­cine.8 At Data
the time of this writ­ing, it is recommended that hep­a­rin anti­ When decid­ing on proper treat­ment for a patient with PVT, it is
coagulation in these patients be avoided until an antiheparin impor­tant to con­sider whether the throm­bo­sis is acute or chronic
plate­let fac­tor 4 anti­body or func­tional assay returns neg­a­tive.7 and whether the liver is cir­rhotic or not.16 The goals of anticoagu­
In addi­tion, the International Society on Thrombosis and Haemo­ lation in PVT are to increase the chance of recan­a­li­za­tion in order
stasis (ISTH) recently published guide­lines suggesting the use of to improve hepatic blood flow and decrease variceal for­ma­tion
intravenous immunoglobulin in addi­tion to nonheparin anticoag­ and sub­se­quent bleed­ing.13,17 A recent meta-anal­y­sis by Valeriani
ulation in con­firmed cases.9 and col­leagues consisted of 7969 SVT patients in 97 stud­ies (54%
The treat­ment of clots in unusual loca­tions typ­i­cally con­sists of whom had PVT) and documented a recan­a­li­za­tion rate of 58%
of anticoagulation and less com­monly fibri­no­lytic agents, with vs 22%, a pro­gres­sion rate of 11% vs 15%, and major bleed­ing in
interventional and sur­gi­cal tech­niques playing a sec­ond­ary role 9% vs 16% in those receiv­ing vs not receiv­ing anticoagulation
and often reserved for pro­gres­sive organ dys­func­tion or infarc­ (Figure 1).13 Compared to no treat­ment, anticoagulation was asso­
tion (eg, mes­en­teric vein throm­bo­sis).1,10,11 Data on the effi­cacy of ci­ated with higher recan­a­li­za­tion (rel­a­tive risk [RR], 2.29; 95% CI,
the direct oral anti­co­ag­u­lants (DOACs) com­pared to war­fa­rin will 1.66-3.44), lower pro­gres­sion (RR, 0.24; 95% CI, 0.13-0.42), major
be cov­ered in greater detail in the evi­dence-based minireview bleed­ing (RR, 0.73; 95% CI, 0.58-0.92), and over­all mor­tal­ity (RR,
Should Warfarin or a DOAC Be Used in Patients Presenting With 0.45; 95% CI, 0.33-0.60). The con­clu­sion was that anticoagulation
Thrombosis in the Splanchnic or Cerebral Veins? for SVT throm­bo­sis reduced the risk of throm­bus pro­gres­sion and
In the remain­der of this arti­cle, we focus on a case-based improved the rate of recan­a­li­za­tion with­out increas­ing bleed­ing
review of throm­bo­sis in the splanch­nic and cere­bral veins. Further risk.13 Two sys­temic reviews and meta-ana­ly­ses showed the ben­e­
com­pre­hen­sive review of these top­ics as well as clots in other fit of anticoagulation in cir­rhotic patients with SVT.17,18 The first by
unusual loca­tions can be found in sev­eral excel­lent reviews.1-4 Valeriani reported decreased RRs of bleed­ing (RR, 0.52; 95% CI,
1.42-7.17) and mor­tal­ity (RR, 0.42; 95% CI, 0.24-0.73) and higher
SVT recan­a­li­za­tion rates (RR, 3.19; 95% CI, 1.42-7.17) in the anticoagu­
SVT refers to venous throm­bo­em­bo­lism of the por­tal vein (PV), lated group.17 The sec­ond by Ghazaleh also showed that antico­
mes­en­teric, splenic, or hepatic veins. Hepatic vein throm­bo­sis agulation increased the rate of PV recan­a­li­za­tion and decreased
(HVT) is typ­i­cally referred to as Budd-Chiari syn­drome (BCS) and the rate of variceal bleed­ing in cir­rhotic patients with non­ma­lig­
includes venous out­flow obstruc­tion any­where from the level of nant PV throm­bo­sis.18 Prophylactic banding should be insti­tuted
the hepatic venules prox­i­mally to the junc­tion of the inferior vena in cir­rhotic patients to decrease variceal bleed­ing.3
cava and right atrium.12 Although the inci­dence of SVT is at least An ISTH sub­com­mit­tee intro­duced recent guide­lines recom­
25 times less than lower-extrem­ity DVT, the pre­sen­ta­tion is often mending early anticoagulation in cir­rhotic and noncirrhotic patients
much more dra­matic.1-4 with acute SVT with no active bleed­ing or con­tra­in­di­ca­tions for at
Prakash Singh Shekhawat
Clots in unusual places | 93
Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/92/1851781/92mathew.pdf by guest on 13 December 2021
Figure 1.  Outcomes of anticoagulant therapy for splanchnic vein thrombosis, as listed on the top.13

least 3 to 6 months.11 In patients with chronic throm­bo­sis, an indi­ and based pri­mar­ily on expert opin­ion, some experts rec­om­mend
vid­u­al­ized care plan is recommended, while treat­ment sim­i­lar to decreas­ ing the inten­ sity of anticoagulation to half-ther­ a­ peu­
tic
symp­tom­atic acute throm­bo­sis is recommended for inci­den­tally doses in cir­rhotic patients if the plate­let counts are between 50 000
detected SVT.11 The 2017 guide­ lines by the Euro­ pean Associa­ and 100 000/µL to pro­phy­lac­tic doses between 30 000 and
tion for the Study of the Liver rec­om­mend anticoagulation for all­ 50 000/µL and with­hold­ing anticoagulation if the plate­let count is
patients with non­ma­lig­nant, noncirrhotic PVT for at least 6 months, below 30 000/µL.2 In patients with under­ly­ing coagulopathy, low-
while the 2012 American College of Chest Physicians guide­lines molec­u­lar-weight hep­a­rin (LMWH) may be pre­ferred to war­fa­rin,
rec­om­mend only treating symp­tom­atic SVT and not anticoagulat­ given the inabil­ity to ade­quately fol­low the effects of war­fa­rin with
ing those inci­den­tally detected unless the SVT is exten­sive or asso­ PT mon­i­tor­ing. A recent sys­tem­atic review suggested that throm­
ci­ated with malig­nancy.2,19,20 However, more recent obser­va­tional bolysis is effec­tive and safe for PVT in noncirrhotic patients who
stud­ies and guide­lines have chal­lenged the American College of have failed anticoagulation.21 Figure 2 depicts gen­eral treat­ment
Chest Physicians rec­om­men­da­tions.2,19 While still con­tro­ver­sial rec­om­men­da­tions for SVT.2

Figure 2.  Recommendations for treatment of SVT.2

94  |  Hematology 2021  |  ASH Education Program


prothrombotic state is iden­ti­fied in 88% of BCS patients.13 A
CLINICAL CASE 1 (Con­t in­u ed) mye­lo­pro­lif­er­a­tive neo­plasm (MPN) has been reported in up to
Consistent with the ISTH guide­lines ref­er­enced above, the deci­ 62% of cases of idi­o­pathic BCS, with poly­cy­the­mia vera being
sion was made to start anticoagulation given the new, symp­tom­ the most com­mon sub­type (18%-43%). A JAK2 V617F muta­tion
atic PV clot. We discussed the options of LWMH, war­fa­rin, and is found in 26% to 52% of patients.2 The diag­no­sis of BCS is
DOACs with the patient. He opted for daily enoxaparin (Lovenox) the presenting symp­tom of an MPN in 74% of cases, with many
injec­tions. Given the plate­let count of 49 000/µL and a PT of 19 patients hav­ing nor­mal blood counts at the time.2 Calreticulin
sec­onds, the deci­sion was made in con­junc­tion with the patient muta­tions have been reported in 2.9% of BCS patients, in the
and hepatology team to decrease the enoxaparin dose to 1 mg/ absence of the JAK2 V617F muta­tion.12 The MPL muta­tion has
kg/d. He under­went suc­cess­ful liver trans­plan­ta­tion 4 months later, been reported in only a few cases.12 Any patient who pres­ents
and anticoagulation was stopped after his post­op­er­a­tive recov­ery. with idi­o­pathic BCS should be eval­u­ated with an MPN genetic
panel consisting of at least the above muta­tions.12 Antiphos­
pholipid anti­body syn­drome and par­ox­ys­mal noc­tur­nal hemo­
glo­bin­uria are also over­rep­re­sented in patients with BCS and

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/92/1851781/92mathew.pdf by guest on 13 December 2021


CLINICAL CASE 2
should be con­sid­ered if an MPN panel is neg­a­tive.12
A 43-year-old woman who has no prior med­i­cal his­tory pre­ The man­age­ment of BCS, com­pared to other SVTs, is unique
sented with 2 weeks of pro­gres­sive jaun­dice, abdom­i­nal pain, as treat­ment gen­er­ally con­sists of both short- and long-term
and a distended abdo­men. She took no med­i­ca­tions and did anticoagulation as well as decom­pres­sion of the hepatic venous
not smoke or con­sume alco­hol. Her fam­ily his­tory was unknown. out­flow track (Figure 3). Lifelong anticoagulation is typ­i­cally rec­
Physical exam­i­na­tion was sig­nif­i­cant for scleral icterus and jaun­ ommended for BCS, although based mostly on expert opin­ion
dice. The abdo­men was distended, the liver edge was pal­pated with no ran­dom­ized con­trolled tri­als hav­ing been reported.12,19
3 cm below the cos­tal mar­gin, and a fluid wave was detected. LMWH or unfractionated hep­a­rin is typ­i­cally begun, and endos­
There was mild ankle edema, but the rest of the exam­i­na­tion copy is performed to eval­u­ate for and treat varices.12 Historically,
was unre­mark­able. Hemoglobin was 16.7 g/dL with an MCV of long-term anticoagulation has been with war­fa­rin.
76 fL and a plate­let count of 608 000/µL. Imaging con­firmed Endovascular inter­ven­tion con­sists of angio­plasty, stenting,
occlu­sive throm­bo­sis of the main hepatic vein. and thrombolysis aimed at open­ing up the hepatic out­flow
tract and pre­ serv­ing hepatic func­ tion. Transvenous angio­
plasty is cur­rently performed in a major­ity of BCS patients.12,22
Questions Stent place­ment should be pur­sued if there is inad­e­quate pres­
How com­mon is the JAK2 V617F muta­tion in BCS? Is anti­co­ag­u­ sure reduc­tion with angio­plasty, based on supe­rior results in
lant ther­apy, local inter­ven­tion, or both pre­ferred in newly diag­ a recent ran­dom­ized clin­i­cal trial.12,23 If this is not effec­tive or
nosed BCS? fea­si­ble, transjugular intrahepatic portosystemic shunt is usu­
ally performed to reduce por­tal pres­sure.3 Surgical shunting,
Data while the his­tor­i­cal treat­ment of choice, has mostly given way
BCS occurs due to obstruc­tion of the hepatic venous out­flow to endovascular inter­ven­tion and anticoagulation.1,3 Using the
any­where between the liver and the heart.12 An under­ly­ing step­wise approach, 1-year and 5-year sur­vival have improved

Figure 3.  Stepwise treatment algorithm for BCS.


Prakash Singh Shekhawat
Clots in unusual places | 95
to 96% and 89%, respec­tively.11 Liver trans­plan­ta­tion has been lev­els increased over the next 12 hours. The deci­sion was made
performed in patients with acute or pro­gres­sive liver fail­ure.12 to pro­ceed with explor­atory lap­a­rot­omy due to con­cern for
peri­to­ni­tis and necrotic bowel.

CLINICAL CASE 2 (Con­t in­u ed)


Endovascular ther­apy (EVT) should be lim­ited to cen­ters with
The patient was imme­di­ately placed on ther­a­peu­tic unfraction­ exper­tise in the pro­ce­dure.24 In the afore­men­tioned sys­tem­atic
ated hep­a­rin, and angio­plasty with stenting of the main hepatic review, EVT was only used at 2 cen­ters, and the bowel resec­tion
vein was performed within 48 hours with excel­lent results. JAK-2 rate remained high at 65% and 78%, respec­tively.24-26 However,
V617F test­ing sub­se­quently returned pos­i­tive, and she started in an addi­tional study 5 out of 8 patients who under­went EVT
ther­a­peu­tic phle­bot­omy as well as hydroxy­urea. After a dis­cus­ avoided sur­gi­cal resec­tion.27
sion about oral anti­co­ag­u­lant options, the patient decided to Surgical explo­ra­tion is often required when bowel ische­mia
be treated with apixaban 5 mg twice a day. Six months later is suspected. However, the dis­tinc­tion between revers­ible and
her hemo­glo­bin and plate­let count remained nor­mal, and there irre­vers­ible bowel ische­mia is dif­fi­cult.24 Often a pri­mary lap­a­

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/92/1851781/92mathew.pdf by guest on 13 December 2021


was no recur­rent throm­bo­sis on a reg­i­men of peri­odic phle­bot­ rot­omy is performed, but only frankly necrotic bowel should be
omy, hydroxy­urea, and apixaban. resected.24 The abdo­men is typ­i­cally left open, and a sec­ond-
look oper­a­tion is performed at a later date.24

CLINICAL CASE 3
A 50-year-old woman with a prior his­tory of a left pop­li­teal DVT
devel­oped severe midabdominal pain, cramping, and fever. CLINICAL CASE 3 (Con­tin­ued)
The pain progressed over the next few days, and she presented During the ini­ tial sur­
gi­
cal explo­
ra­
tion, 5 cm of necrotic jeju­
to her local emer­gency room. A com­plete blood count showed num was resected. Upon tran­sec­tion of the bowel wall, mul­
a mild leu­ko­cy­to­sis with a left shift. Lactic acid was mod­er­ately ti­
ple “worm­ like” clots were extruded. Anticoagulation was
ele­
vated. A CT venogram (CTV) showed throm­ bo­sis of the restarted and she clin­i­cally improved and did not require addi­
supe­rior mes­en­teric vein with bowel wall edema through­out tional resec­tion. A thrombophilia workup includ­ing JAK-2 V617F
the jeju­num and prox­i­mal ileum. test­ing was under­taken and returned sig­nif­i­cant for high-titer
anticardiolipin and beta-2-gly­co­pro­tein antibodies. Long-term
anticoagulation with war­fa­rin was ini­ti­ated.
Question
Should MVT be man­aged med­i­cally, sur­gi­cally, or both?
MVT is a rare cause of mes­en­teric ische­mia, representing
CVT
only 5% to 20% of cases.1,4,24 With improved imag­ing tech­
CVT refers to throm­bo­ses of the cere­bral veins or dural venous
niques, includ­ ing contrasted CTV or mag­ netic res­

nance
sinuses. CVT is rare, with an approx­i­mate inci­dence of 13.2 to 15.7
imag­ ing (MRI), the diag­ no­ sis can typ­ i­
cally be made radio­
per mil­lion per­sons per year. CVT is more com­mon in women
graph­i­cally instead of sur­gi­cally.1 Guidelines published by the
than men (3:1) and in youn­ger age groups (mean age of 40).3
Euro­pean Association for the Study of the Liver rec­om­mend
Clinical man­i­fes­ta­tions of CVT include neu­ro­log­i­cal symp­
treat­ment with LMWH or, pref­er­a­bly, intra­ve­nous hep­a­rin, as
toms rang­ing from head­aches, papilledema, focal def­i­cits, sei­
early lap­ ar­ot­
omy may be required.3,4,24 A recent sys­tem­atic
zures, enceph­a­lop­a­thy, and coma.28 As with other atyp­i­cal sites
review of 11 published stud­ies recommended early and imme­
of throm­bo­sis, risk fac­tors for CVT can be divided into local,
di­ate anticoagulation.24
mechan­i­cal, and sys­temic fac­tors (Table 2).
CVT is con­firmed by neuroimaging. Most patients are ini­
tially eval­u­ated with a noncontrast CT to quickly eval­u­ate for
CLINICAL CASE 3 (Con­t in­u ed) intra­cra­nial hem­or­rhage as well as other pathol­o­gies. How­
Despite ther­a­peu­tic anticoagulation, intra­ve­nous flu­ids, bowel ever, unenhanced CT scans lack the sen­si­tiv­ity to detect the
rest, and anti­bi­ot­ics, she became hypo­ten­sive and lac­tic acid major­ity of CVT. A con­trast-enhanced CT increases sen­si­tiv­ity

Table 2.  Risk fac­tors for CVT

Local risk fac­tors (8%-12%) Mechanical causes (2%-5%) CNS dis­or­ders (2%) Systemic risk fac­tors
Infections involv­ing the ears, Trauma, lum­bar punc­ture, Arteriovenous malformation, dural Pregnancy/puer­pe­rium (10%-17% women),
sinuses, mouth, face, neck, CNS jug­u­lar vein cath­e­ter­i­za­tion, fis­tu­lae, CNS malig­nan­cies hor­monal treat­ment (50%-53% women),
neu­ro­sur­gi­cal inter­ven­tions thrombophilia (approx. 33%), mye­lo­pro­lif­
er­a­tive neo­plasms (3%-4%),
COVID-19 infec­tion, COVID-19 vac­cines
(AstraZeneca, J&J)
Adapted from Riva et al.4
CNS, cen­tral ner­vous sys­tem.

96  |  Hematology 2021  |  ASH Education Program


to 88% to 99%, and MRI remains the gold stan­dard for diag­no­ agulated group (12.7% vs 18.3%; haz­ard ratio, 0.73; CI, 0.44-1.21)
sis.29 CTVs may be use­ful in patients who have a con­tra­in­di­ca­ was seen.32 A sub­se­quent sys­tem­atic review and meta-anal­y­sis
tion to MRI.3 showed a trend toward decreased mor­tal­ity and neu­ro­log­i­cal
out­comes with LMWH com­pared to UFH.33 Bleeding events were
sim­i­lar in both groups.33 Based on these stud­ies, the most recent
guide­lines from the Euro­pean Stroke Organization (2017), the
CLINICAL CASE 4 Amer­i­can Heart Association/Amer­i­can Stroke Association (2014),
A 23-year-old woman presented to the emer­gency room with and the Amer­i­can Academy of Chest Physicians (2012) rec­om­
wors­en­ing head­ache over the past week. She had no sig­nif­i­cant mend either UFH or LMWH for treat­ment of acute CVT even in
past med­i­cal his­tory. Her only med­i­ca­tion was a com­bined oral cases with asso­ci­ated cere­bral hem­or­rhage.27,34,35
con­tra­cep­tive pill. A com­puted tomog­ra­phy (CT) scan of the
head with­out con­trast showed a “dense tri­an­gle sign.” While in
the emer­gency room, she suf­fered a tonic-clonic sei­zure. MRI
of the brain showed throm­bo­sis involv­ing the supe­rior sag­it­tal

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/92/1851781/92mathew.pdf by guest on 13 December 2021


CLINICAL CASE 4 (Con­tin­ued)
sinus with an asso­ci­ated small area of intra­cra­nial hem­or­rhage.
The patient was started on an anti­ep­i­lep­tic and intra­ve­nous
hep­a­rin; how­ever, she devel­oped wors­en­ing men­tal sta­tus
requir­ing mechan­i­cal intu­ba­tion. Repeat imag­ing did not show
Question
new intra­cra­nial hem­or­rhage. The neurointerventional radi­ol­
What is the ini­tial treat­ment of acute-phase CVT?
ogy team was consulted to eval­u­ate for thrombectomy.
Many cases of CVT are accom­pa­nied by intra­ce­re­bral hem­
or­rhage, mak­ing treat­ment com­pli­cated given the con­cern for
wors­en­ing hem­or­rhage. Two ran­dom­ized tri­als exam­ined the
ben­e­fit of par­en­teral anticoagulation com­pared to pla­cebo in Question
the treat­ment of acute-phase CVT. In the first trial, published in What is the role of endovascular inter­ven­tions in the treat­ment
1991, patients were ran­dom­ized to intra­ve­nous hep­a­rin treat­ment of CVT?
(n = 10) vs pla­cebo (n = 10). The hep­a­rin group had improved recov­ The evi­dence thus far sug­gests no added ben­e­fit from endo­
ery and bet­ter long-term out­comes.30 The sec­ond trial consisted vascular thrombolysis or thrombectomy. Data pri­mar­ily come
of 60 patients ran­dom­ized to nadroparin vs pla­cebo. Better out­ from sys­tem­atic reviews that show higher fatal­ity rates, more
comes in the nadroparin arm were noted, although they did not bleed­ing (mostly intra­cra­nial), and poor neu­ro­log­i­cal out­comes
reach sta­tis­ti­cal sig­nif­i­cance.31 Although sam­ple sizes were small, with thrombolysis (Table 3). A ran­dom­ized con­trolled trial (TO-
these tri­als dem­on­strated that anticoagulation with hep­a­rin or ACT) exam­ined the ben­e­fit of thrombolysis in addi­tion to antico­
LMWH is bet­ter than no treat­ment in acute CVT. Approximately agulation and was stopped pre­ma­turely for futil­ity.36 Given the
43% of these patients had intra­cra­nial hem­or­rhage at diag­no­sis, lack of high-qual­ity evi­dence suggesting ben­e­fit, the con­sen­sus
and none of the patients ran­dom­ized to anticoagulation devel­ is to avoid up-front EVT except in select cases with high pre­
oped a new intra­cra­nial hem­or­rhage, whereas 3 patients in the treat­ment risk of poor out­comes.
pla­cebo group expe­ri­enced a new hem­or­rhage.30,31 In the Inter­ CVT can lead to other com­ pli­
ca­
tions, includ­ ing sei­
zures,
national Study on Cerebral Vein and Dural Sinus Thrombosis, a ele­vated intra­cra­nial pres­sure, brain her­ni­a­tion, and infec­tions,
trend toward lower rates of death or depen­dency in the antico­ requir­ing addi­tional sup­port­ive mea­sures (Figure 4).

Table 3.  Endovascular ther­apy in acute CVT

No of
Trial Study design patients Intervention Results Limitations
Coutinho et al 36
Randomized, 67 Endovascular treat­ment in patients Stopped pre­ma­turely for Small sam­ple size
(TO-ACT trial) con­trolled Inclusion cri­te­ria: ≥1 Risk fac­tor for futil­ity
clin­i­cal dete­ri­o­ra­tion (coma, men­
tal sta­tus changes, CVT in deep
venous sys­tem, intra­ce­re­bral
hem­or­rhage)
Dentali et al37 Systematic review of 156 Mechanical Death rate 9%
15 stud­ies thrombectomy/thrombolysis Major bleed­ing 10% with local
thrombolysis
8% intra­cra­nial hem­or­rhage
58% fatal
Siddiqui et al40 Systematic review of 185 Mechanical 84% good out­come Concern for bias as
42 stud­ies thrombectomy/thrombolysis (71%) 12% mor­tal­ity the stud­ies were
60% pre­treat­ment intra­cra­nial 10% new/wors­ened intra­ce­re­ not blinded
hem­or­rhage bral hem­or­rhage
47% stu­por, coma 95% recan­a­li­za­tion rate

Prakash Singh Shekhawat


Clots in unusual places | 97
Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/92/1851781/92mathew.pdf by guest on 13 December 2021
Figure 4.  Recommendations for treatment of CVT.2

6 to 12 months. In cases in which per­sis­tent hyper­co­ag­u­la­bil­ity


CLINICAL CASE 4 (Con­t in­u ed) and a risk for throm­bo­sis have been iden­ti­fied, long-term anti­
In the fol­low­ing days, she slowly recov­ered and was extubated, coagulation should be con­sid­ered. Similarly, a lon­ger dura­tion of
with a plan to dis­charge her to a reha­bil­it­ a­tion facil­ity. anticoagulation is suggested for the treat­ment of recur­rent CVT.
The EXCOA-CVT study is an ongo­ing pro­spec­tive study that aims
to com­pare the out­comes of short-dura­tion (3-6 months) vs long-
term (12 months) anticoagulation treat­ment in CVT patients.39
Question
What is the best long-term treat­ment for CVT? Conflict-of-inter­est dis­clo­sure
The Cerebral Vein Thrombosis International Study reported Carol Mathew: no com­pet­ing finan­cial inter­ests to declare.
on 706 patients with CVT. Of these, 85% were treated with hep­a­ Marc Zumberg: mem­ber: American Board of Internal Medicine
rin in the acute phase followed by 84% with war­fa­rin for a median Hematology Board; Amer­i­can Society of Hematology speaker
dura­tion of 12 months. At a median fol­low-up of 40 months, 89.1% reim­burse­ment, law case review.
of patients were found to have com­pletely recov­ered.37 In the
VENOST study, 67% of 1144 CVT patients were treated with war­ Off-label drug use
fa­rin after ini­tial treat­ment with hep­ar­ in. At 1-year fol­low-up, 93.1% Carol Mathew: direct oral anticoagulants for splanchnic and
of 691 patients for whom data were avail­­able showed a com­plete cerebral vein thrombosis are discussed.
recov­ery.38 Most soci­ety guide­lines rec­om­mend the use of war­fa­ Marc Zumberg: direct oral anticoagulants for splanchnic and
rin, although emerg­ing data show that DOACs may have similar cerebral vein thrombosis are discussed.
efficacy and safety as compared to war­fa­rin. This topic will be
cov­ered in greater detail in the accom­pa­ny­ing evi­dence-based Correspondence
minireview Should Warfarin or a DOAC Be Used in Patients Pre­ Marc Zumberg, University of Florida Health, 1600 SW Archer Rd,
senting With Thrombosis in the Splanchnic or Cerebral Veins? Gainesville, FL 32610; e-mail: zumbems@med­i­cine​­.ufl​­.edu.
There is no defin­i­tive evi­dence regard­ing the opti­mal dura­tion
of anticoagulation in the treat­ment of CVT. Extrapolating from References
1. Rajasekhar A, Zumberg M. Venous throm­bo­ses at unusual sites. In: Kitchens
evi­dence from lower-extrem­ity DVT, most experts rec­om­mend 3
CS, Alving BM, Kessler CM, eds. Consultative Hemostasis and Thrombosis.
to 12 months of anticoagulation. If CVT occurred in the set­ting of 3rd ed. W. B. Saunders Co.; 2013:262-290.
a tran­sient risk fac­tor, 3 to 6 months of anticoagulation may be 2. Abbattista M, Capecchi M, Martinelli I. Treatment of unusual throm­botic
ade­quate. If there were no pro­vok­ing fac­tors, we treat for at least man­i­fes­ta­tions. Blood. 2020;135(5):326-334.

98  |  Hematology 2021  |  ASH Education Program


3. Riva N, Ageno W. Cerebral and splanch­nic vein throm­bo­sis: advances, 24. Acosta S, Salim S. Management of acute mes­en­teric venous throm­bo­sis: a
chal­lenges, and unanswered ques­tions. J Clin Med. 2020;9(3):743. sys­tem­atic review of con­tem­po­rary stud­ies. Scand J Surg. 2020;110(2):123-
4. Shatzel JJ, O’Donnell M, Olson SR, et al. Venous throm­bo­sis in unusual sites: 129.
a prac­ti­cal review for the hema­tol­o­gist. Eur J Haematol. 2019;102(1):53-62. 25. Liu K, Liu S, Li L, et al. Evaluation of endovascular ther­apy com­bined with
5. Mowla A, Shakibajahromi B, Shahjouei S, et al. Cerebral venous sinus throm­ bowel resec­ tion treat­ment on patients with acute mes­ en­teric venous
bo­sis asso­ci­ated with SARS-CoV-2: a mul­ti­na­tional case series. J Neurol Sci. throm­bo­sis. Ann Vasc Surg. 2020;65(May):72-81.
2020;419(15 Decem­ber):117183. 26. Yang S, Zhang L, Liu K, et al. Postoperative cath­e­ter-directed thromboly­
6. Singh B, Kaur P, Maroules M. Splanchnic vein throm­bo­sis in COVID-19: a sis ver­sus sys­temic anticoagulation for acute supe­rior mes­en­teric venous
review of lit­er­a­ture. Dig Liver Dis. 2020;52(12):1407-1409. throm­bo­sis. Ann Vasc Surg. 2016;35(August):88-97.
7. Greinacher A, Thiele T, Warkentin TE, Weisser K, Kyrle PA, Eichinger S. 27. Salim S, Zarrouk M, Elf J, Gottsäter A, Ekberg O, Acosta S. Improved prog­
Thrombotic throm­bo­cy­to­pe­nia after ChAdOx1 nCov-19 vac­ci­na­tion. N Engl no­sis and low fail­ure rate with anticoagulation as first-line ther­apy in mes­
J Med. 2021;384(22):2092-2101. en­teric venous throm­bo­sis. World J Surg. 2018;42(11):3803-3811.
8. Schultz NH, Sørvoll IH, Michelsen AE, et al. Thrombosis and throm­bo­cy­to­ 28. Ferro JM, Bousser MG, Canhão P, et al; Euro­ pean Stroke Organization.
pe­nia after ChAdOx1 nCoV-19 vac­ci­na­tion. N Engl J Med. 2021;384(22):2124- Euro­pean Stroke Organization guide­line for the diag­no­sis and treat­ment
2130. of cere­bral venous throm­bo­sis—endorsed by the Euro­pean Academy of
9. Nazy I, Sachs UJ, Arnold DM, et al. Recommendations for the clin­i­cal and Neurology. Eur J Neurol. 2017;24(10):1203-1213.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/92/1851781/92mathew.pdf by guest on 13 December 2021


lab­o­ra­tory diag­no­sis of VITT against COVID-19: com­mu­ni­ca­tion from the 29. Capecchi M, Abbattista M, Martinelli I. Cerebral venous sinus throm­bo­sis.
ISTH SSC sub­com­mit­tee on plate­let immu­nol­ogy. J Thromb Haemost. J Thromb Haemost. 2018;16(10):1918-1931.
2021;19(6):1585-1588. 30. Einhäupl KM, Villringer A, Meister W, et  al. Heparin treat­ ment in sinus
10. Riva N, Carrier M, Gatt A, Ageno W. Anticoagulation in splanch­nic and cere­ venous throm­bo­sis. Lancet. 1991;338(8767):597-600.
bral vein throm­bo­sis: an inter­na­tional vignette-based sur­vey. Res Pract 31. de Bruijn SF, Stam J. Randomized, pla­cebo-con­trolled trial of anti­co­ag­u­lant
Thromb Haemost. 2020;4(7):1192-1202. treat­ment with low-molec­u­lar-weight hep­a­rin for cere­bral sinus throm­bo­
11. Di Nisio M, Valeriani E, Riva N, Schulman S, Beyer-Westendorf J, Ageno W. sis. Stroke. 1999;30(3):484-488.
Anticoagulant ther­apy for splanch­nic vein throm­bo­sis: ISTH SSC sub­com­ 32. Ferro JM, Canhão P, Stam J, Bousser MG, Barinagarrementeria F; ISCVT
mit­tee con­ trol of anticoagulation. J Thromb Haemost. 2020;18(7):1562- Investigators. Prognosis of cere­bral vein and dural sinus throm­bo­sis: results
1568. of the International Study on Cerebral Vein and Dural Sinus Thrombosis
12. Sharma A, Keshava SN, Eapen A, Elias E, Eapen CE. An update on the man­ (ISCVT). Stroke. 2004;35(3):664-670.
age­ment of Budd-Chiari syn­drome. Dig Dis Sci. 2021;66(6):1780-1790. 33. Qureshi A, Perera A. Low molec­u­lar weight hep­a­rin ver­sus unfractionated
13. Valeriani E, Di Nisio M, Riva N, et  al. Anticoagulant ther­apy for splanch­ hep­a­rin in the man­age­ment of cere­bral venous throm­bo­sis: a sys­tem­atic
nic vein throm­bo­sis: a sys­tem­atic review and meta-anal­y­sis. Blood. review and meta-anal­y­sis. Ann Med Surg (Lond). 2017;17(May):22-26.
2021;137(9):1233-1240. 34. Kernan WN, Ovbiagele B, Black HR, et al; Amer­ i­
can Heart Association
14. De Stefano V, Qi X, Betti S, Rossi E. Splanchnic vein throm­bo­sis and mye­ Stroke Council, Council on Cardiovascular and Stroke Nursing, Council on
lo­pro­lif­er­a­tive neo­plasms: molec­u­lar-driven diag­no­sis and long-term treat­ Clinical Cardiology, and Council on Peripheral Vascular Disease. Guidelines
ment. Thromb Haemost. 2016;115(2):240-249. for the pre­ven­tion of stroke in patients with stroke and tran­sient ische­mic
15. Magaz M, Alvarez-Larrán A, Colomer D, et al. Next-gen­er­a­tion sequenc­ing attack: a guide­line for healthcare pro­fes­sion­als from the Amer­i­can Heart
in the diag­no­sis of non-cir­rhotic splanch­nic vein throm­bo­sis. J Hepatol. Association/Amer­i­can Stroke Association. Stroke. 2014;45(7):2160-2236.
2021;74(1):89-95. 35. Lansberg MG, O’Donnell MJ, Khatri P, et al. Antithrombotic and throm­bo­
16. Intagliata NM, Caldwell SH, Tripodi A. Diagnosis, devel­op­ment, and treat­ lytic ther­apy for ische­mic stroke: Antithrombotic Therapy and Prevention of
ment of por­tal vein throm­bo­sis in patients with and with­out cir­rho­sis. Gas- Thrombosis, 9th ed: Amer­i­can College of Chest Physicians Evidence-Based
troenterology. 2019;156(6):1582-1599.e11599e1. Clinical Practice Guidelines. Chest. 2012;141(suppl 2):e601S-e636S.
17. Valeriani E, Di Nisio M, Riva N, et al. Anticoagulant treat­ment for splanch­nic 36. Coutinho JM, Ferro JM, Zuurbier SM, et al. Thrombolysis or anticoagulation
vein throm­bo­sis in liver cir­rho­sis: a sys­tem­atic review and meta-anal­y­sis. for cere­bral venous throm­bo­sis: ratio­nale and design of the TO-ACT trial.
Thromb Haemost. 2021;121(7):867-876. Int J Stroke. 2013;8(2):135-140.
18. Ghazaleh S, Beran A, Aburayyan K, et al. Efficacy and safety of anticoagula­ 37. Dentali F, Poli D, Scoditti U, et al; Cerebral Vein Thrombosis International
tion in non-malig­nant por­tal vein throm­bo­sis in patients with liver cir­rho­sis: Study Investigators. Long-term out­comes of patients with cere­bral vein
a sys­tem­atic review and meta-anal­y­sis. Ann Gastroenterol. 2021;34(1):104- throm­bo­sis: a mul­ti­cen­ter study. J Thromb Haemost. 2012;10(7):1297-1302.
110. 38. Duman T, Uluduz D, Midi I, et al; VENOST Study Group. A mul­ti­cen­ter study
19. Euro­pean Association for the Study of the Liver. EASL Clinical Practice of 1144 patients with cere­ bral venous throm­bo­ sis: the VENOST study.
Guidelines: vas­cu­lar dis­eases of the liver. J Hepatol. 2016;64(1):179-202. J Stroke Cerebrovasc Dis. 2017;26(8):1848-1857.
20. Kearon C, Akl EA, Comerota AJ, et  al. Antithrombotic ther­apy for VTE 39. Miranda B, Aaron S, Arauz A, et al. The ben­e­fit of EXtending oral antiCO­
dis­ease: Antithrombotic Therapy and Prevention of Thrombosis, 9th ed: Agulation treat­ment (EXCOA) after acute cere­bral vein throm­bo­sis (CVT):
Amer­i­can College of Chest Physicians Evidence-Based Clinical Practice EXCOA-CVT clus­ter ran­dom­ized trial pro­to­col. Int J Stroke. 2018;13(7):771-
Guidelines. Chest. 2012;141(suppl 2):e419S-e496S. 774.
21. Cheng Q , Tree K. Systematic review of thrombolysis ther­apy in the man­ 40. Siddiqui FM, Dandapat S, Banerjee C, et  al. Mechanical thrombectomy
age­ment of non-cir­rho­sis-related por­tal vein throm­bo­sis. J Gastrointest in cere­bral venous throm­bo­sis: sys­tem­atic review of 185 cases. Stroke.
Surg. 2021;25(6):1579-1590. 2015;46(5):1263-1268.
22. Tripathi D, Sunderraj L, Vemala V, et al. Long-term out­comes fol­low­ing per­
cu­ta­ne­ous hepatic vein recan­a­li­za­tion for Budd-Chiari syn­drome. Liver Int.
2017;37(1):111-120.
23. Wang Q , Li K, He C, et al. Angioplasty with ver­sus with­out rou­tine stent
place­ment for Budd-Chiari syn­drome: a randomised con­trolled trial. Lan- © 2021 by The Amer­i­can Society of Hematology
cet Gastroenterol Hepatol. 2019;4(9):686-697. DOI 10.1182/hema­tol­ogy.2021000237

Prakash Singh Shekhawat


Clots in unusual places | 99
CLOTTING AND BLEEDING CONUNDRUMS

EVIDENCE-BASED MINIREVIEW

Should warfarin or a direct oral anticoagulant


be used in patients presenting with thrombosis

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/100/1851729/100mathew.pdf by guest on 13 December 2021


in the splanchnic or cerebral veins?
Carol Mathew and Marc Zumberg
University of Florida, Internal Medicine, Division of Hematology/Oncology, Gainesville, FL

Case 1: A 23-year-old female third-year medical student who has no medical history seeks treatment for abdominal dis-
tention. She takes an estrogen-containing birth control pill and does not smoke or consume alcohol. Family history is
unremarkable. Physical examination is significant for abdominal distention, and an abdominal fluid wave is detected.
Complete blood count is normal. Imaging confirms occlusive thrombosis of the main portal vein. On endoscopy, grade 1
to 2 esophageal varices are noted and banded. Unfractionated heparin is begun. Subsequent workup reveals a homozy-
gous factor V Leiden mutation. Long-term anticoagulation is planned, and she asks if warfarin can be avoided given her
hectic ward rotations, erratic diet, and need for monitoring. Case 2: A 35-year-old woman who has no medical history
seeks treatment for progressively worsening posterior headaches for 1 week. Magnetic resonance imaging of the brain
shows dural sinus thrombosis with associated small areas of petechial cerebral hemorrhage. She is started on a contin-
uous unfractionated heparin infusion and admitted to the hospital for further observation. Her grandmother is on warfa-
rin for atrial fibrillation, and the patient would prefer to avoid warfarin because she does not think she can comply with
the frequent monitoring that will be required. She inquires about other oral anticoagulant options for her condition.

LEARNING OBJECTIVES
• Discuss the relative risks, benefits, and unique characteristics of the DOACs vs warfarin in patients with thrombosis
in the splanchnic or cerebral veins
• Choose an oral anticoagulant in a patient with thrombosis in the splanchnic or cerebral veins

Introduction warfarin has been the only oral anticoagulant available for
When not otherwise contraindicated, most experts and use in patients with chronic splanchnic or CVT.
societal guidelines recommend early anticoagulation in pa- Since 2010, 4 direct oral anticoagulants (DOACs) have
tients with splanchnic or cerebral vein thrombosis (CVT).1–6 been approved by the US Food and Drug Administration for
However, many of these recommendations are based pri- the treatment of venous thromboembolism (VTE) Based on
marily on small case series, as only a few randomized trials large, randomized trials, both the CHEST and ASH guidelines
exist.4–8 Whether anticoagulation is recommended for a lim- recommend DOACs as the treatment of choice for oral anti-
ited time period or indefinitely depends on whether the clot coagulation in patients with deep vein thrombosis or pul-
was provoked and if the provocation was transient or will be monary embolism.9–11 Patients with atypical site thrombosis
longstanding.1–6 Most of the studies referenced in this arti- were generally excluded from these trials, and therefore
cle do not clearly identify whether outcomes differed be- recommendations concerning the use of DOACs in splanch-
tween provoked and unprovoked splanchnic or CVT. Most nic vein thrombosis (SVT) or CVT remain challenging. The
experts recommend 3 to 12 months of anticoagulation for DOACs have several advantages over warfarin, including
a provoked splanchnic or CVT. Long-term anticoagulation fewer drug-drug interactions, fixed dosing, shorter half-life,
is often recommended for persistent (severe thrombophilia, lack of need for monitoring, quick onset and offset, and less
cancer, myeloproliferative disorder, etc) risk factors, recur- major bleeding in large randomized clinical trials. In the past
rent thrombosis, or hepatic vein thrombosis.9 Historically, several years, small case series and a few randomized trials

100 | Hematology 2021 | ASH Education Program


Table 1. DOAC use in SVT and CVT

•  Evidence is lim­ited but grow­ing. Few ran­dom­ized tri­als have been com­pleted.
•  Most ret­ro­spec­tive and pro­spec­tive data show the DOACs are at least as effec­tive as VKAs, and bleed­ing risk is not higher.
•  Multiple tri­als are cur­rently accru­ing patients.
•  Despite being off label, in a recent sur­vey, DOACs were used in 28% of low bleed­ing risk patients.
•  For patients with SVT, DOACs are contraindicated in Child-Pugh class C liver dis­ease and for rivaroxaban in class B and C liver dis­ease.
• For patients with CVT, con­sider inter­ac­tions with anti­ep­i­lep­tic drugs, and if con­com­i­tant intracranial hemorrhage, con­sider ini­tial use of a
short-act­ing anti­co­ag­u­lant.

have been published reporting on the effi­cacy and bleed­ing risk vein treated with apixaban (n = 20), rivaroxaban (n = 65), and dab-

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/100/1851729/100mathew.pdf by guest on 13 December 2021


of the DOACs in splanch­nic and CVT.7,8,12 Table 1 sum­ma­rizes our igatran (n = 8) com­pared with low molec­u­lar weight hep­a­rin
inter­pre­ta­tion of this lit­er­a­ture. (LMWH) (n = 70) and war­fa­rin (n = 108).14 Seventy-three per­cent of
patients had predisposing fac­tors for PVT. Complete res­o­lu­tion
of throm­bo­sis was seen in 66% of DOAC-treated patients com­
pared with 57% with LMWH and 31% with war­fa­rin (haz­ard ratio,
CASE 1 2.91; 95% CI, 1.87-4.52 for DOAC vs war­fa­rin).14 There was less
A 23-year-old female third-year med­i­cal stu­dent who has no med­ major bleed­ing with the DOACs com­pared with war­fa­rin (haz­ard
i­cal his­tory seeks treat­ment for abdom­i­nal dis­ten­tion. She takes ratio for DOACs: war­fa­rin, 0.20; 95% CI, 0.05-0.86; P = .0307).14
an estro­gen-containing birth con­trol pill and does not smoke or A recent sys­tem­atic review and meta-anal­y­sis published in
con­sume alco­hol. Family his­tory is unre­mark­able. Physical exam­ Blood by Valeriani et al4 reported on 79 patients with SVT in 5
i­na­tion is sig­nif­i­cant for abdom­i­nal dis­ten­tion, and an abdom­i­nal stud­ies who were treated with DOACs. SVT was unpro­voked in
fluid wave is detected. Complete blood count is nor­mal. Imaging 25% of cases. The rates of SVT recan­a­li­za­tion, throm­bo­sis pro­
con­firms occlu­sive throm­bo­sis of the main por­tal vein. On endos­ gres­sion, major bleed­ing, and over­all mor­tal­ity did not sig­nif­i­
copy, grade 1 to 2 esoph­a­geal varices are noted and banded. cantly dif­fer between type of anti­co­ag­u­lant used.4 A 2021 study
Unfractionated hep­a­rin is begun. Subsequent workup reveals a by Kawata et al15 also showed no dif­fer­ence in clot recan­a­li­za­tion
homo­zy­gous fac­tor V Leiden muta­tion. Long-term anticoagula- between patients with SVT treated with LMWH/war­fa­rin or with
tion is planned, and she asks if war­fa­rin can be avoided given any of the DOACs. Local risk fac­tors were pres­ent in 118 (76.1%)
her hec­tic ward rota­tions, erratic diet, and need for mon­i­tor­ing. patients (sec­ond­ary SVT), whereas 7 (4.5%) patients had no risk
fac­tors and 30 (19.4%) patients had only sys­temic or thrombo-
philia (pri­mary SVT).15
Only a sin­gle ran­dom­ized clin­i­cal trial com­par­ing a DOAC to Limited data exist concerning the use of DOACs in Budd-Chiari
war­fa­rin has been reported in patients with SVT.12 Hanafy et al12 syn­drome and mes­en­teric vein throm­bo­sis (MVT). In a ret­ro­spec­
stud­ied 80 patients with acute non­ma­lig­nant por­tal vein throm­ tive anal­y­sis, patients with Budd-Chiari syn­drome treated after
bo­sis (PVT) with hep­a­ti­tis C–related com­pen­sated liver dis­ease. endovascular inter­ ven­ tion with dabigatran (n = 36) were com­
After ini­tial treat­ment with enoxaparin at ther­a­peu­tic doses for pared with those tak­ing war­fa­rin (n = 62).16 Baseline data on hyper­
3 days, patients at 2 Egyp­tian cen­ters were ran­dom­ized between co­ag­u­la­ble states were avail­­able in 45 (45.9%) of 98 patients.16
war­fa­rin with a tar­get inter­na­tional nor­mal­ized ratio (INR) of 2 to At 12 months, stent patency rates (91% vs 93%), major bleed­ing
2.5 and rivaroxaban 10 mg bid. Recanalization of the por­tal vein (3.5% vs 6.5%), and the com­pos­ite end point of mor­tal­ity and
was found in 100% of patients in the rivaroxaban group com­ major bleed­ing (4% vs 8%) did not dif­fer between the dabiga-
pared with 45% in the war­fa­rin group (P = .001). Major gas­tro­in­ tran- and war­fa­rin-treated groups.16 In a ret­ro­spec­tive study of
tes­ti­nal (GI) bleed­ing was not seen in the rivaroxaban group but 102 patients with MVT, no dif­fer­ence was seen between patients
occurred in 43% of patients in the war­fa­rin group (P = .001).12 No treated with a DOAC or war­fa­rin in terms of recan­a­li­za­tion (69%
deaths occurred in the rivaroxaban group com­pared with 36% in vs 71%, P = .88) or major bleed­ing (9.1% vs 14.3%, P = .54).16 No
the war­fa­rin group (P = .001).12 recur­rences of MVT were seen in either group.17
Riva and Ageno9 sum­ma­rize most of the remaining evi­dence ADAM VTE, com­pared with the HOKUSAI VTE, SELECT-D, and
reporting on the use DOACs in SVT, which con­sists mostly of CARAVAGGIO stud­ies, is the only large, ran­dom­ized treat­ment trial
small obser­va­tional tri­als, the major­ity ret­ro­spec­tive in nature. of DOACs in patients with can­cer to include patients with SVT.18-21
Serrao et al13 describe a recent pro­spec­tive study of 28 patients Of the enrolled patients, 8% of those receiv­ing apixaban had a SVT
with SVT and ongo­ing risk fac­tors maintained on chronic treat­ as the qual­i­fy­ing event, as opposed to 18% in the dalteparin group.
ment with war­fa­rin who were switched to DOACs and com­pared In the group treated with apixaban, there was a sin­gle recur­rent
out­comes to 42 patients remaining on war­fa­rin. There was no event (lower extrem­ity throm­bo­sis) and no major bleed­ing.18
dif­fer­ence in throm­botic events between the groups. No major
bleed­ing events occurred in either group, while minor bleed­ing
did not occur in the DOAC group but was documented in 26% of
war­fa­rin-treated patients (P = .09).13 CASE 2
In the larg­est of the ret­ro­spec­tive tri­als, Naymagon et al14 in A 35-year-old woman who has no med­i­cal his­tory seeks treat­
Blood Advances reported on 93 patients with noncirrhotic
Prakash Singh Shekhawat portal ment for pro­gres­sively wors­en­ing pos­te­rior head­aches for 1 week.

Warfarin vs DOAC in SVT or CVT  |  101


Magnetic res­ o­
nance imag­ ing of the brain shows dural sinus nial hem­or­rhage (0.7%) and recur­rent CVT (1.5%) were very low
throm­bo­sis with asso­ci­ated small areas of pete­chial cere­bral in patients treated with DOACs.25 Excellent func­tional out­comes
hem­or­rhage. She is started on a con­tin­uo­ us unfractionated hep­ were observed in 94% of patients in the DOAC arm with an RR of
a­rin infu­sion and admit­ted to the hos­pi­tal for fur­ther obser­va­ 1.13 (95% CI, 1.02-1.25).25
tion. Her grand­mother is tak­ing war­fa­rin for atrial fibril­la­tion, and The larg­est pro­spec­tive obser­va­tional study was performed
the patient would pre­fer to avoid war­fa­rin because she does not by Wasay et al26 and published in the Journal of Stroke. This study
think she can com­ply with the fre­quent mon­i­tor­ing that will be reported on patients from Saudi Arabia, Pakistan, Egypt, and
required. She inquires about other oral anti­co­ag­ul­ant options. United Arab­ Emirates.26 They included 111 patients—45 on DOACs
and 66 on war­fa­rin. The most com­mon DOAC used was rivarox-
aban (n = 36), followed by dabigatran (n = 9). The war­fa­rin dose was
Data supporting the use of DOACs in CVT are emerg­ing. High- adjusted to main­tain an INR range of 2 to 3.26 Initiation of war­fa­
qual­ity evi­dence is still lim­ited, as there has been only 1 ran­dom­ rin or a DOAC hap­pened at a median time of 1 week from the
ized con­trolled trial reported.22 The RE-SPECT CVT trial, published time of CVT diag­no­sis, and the median dura­tion of fol­low-up was

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/100/1851729/100mathew.pdf by guest on 13 December 2021


in 2019, is a mul­ti­cen­ter, open-label study that ran­dom­ized 120 8 months (range, 6-13 months).26 Most patients had pro­vok­ing
patients with CVT to either dabigatran 150 mg twice daily or war­ fac­tors, includ­ing preg­nancy/puer­pe­rium (23.1%), sys­temic infec­
fa­rin with a goal INR of 2 to 3 for 24 weeks. Approximately 50% tions (32.4%), drugs/oral con­tra­cep­tive pills (9.9%), and other
of the patients in each group had iden­ti­fi­able pro­vok­ing fac­tors. med­i­cal con­di­tions (6.3%).26 No recur­rent VTE events were seen
All patients received ini­tial treat­ment with par­en­teral hep­a­rin or in either group. One patient in each group had major bleed­ing
LMWH (5-15 days). The pri­mary end point of the study was new as clas­si­fied by ISTH cri­te­ria. Four patients (6.1%) and 2 patients
venous throm­bo­em­bo­lism—new CVT, pul­mo­nary embolism, limb (4.4%) in the war­fa­rin and DOAC group, respec­tively, had any
DVT, SVT, or major bleed­ing.22 Major bleed­ing was defined accord- bleed­ing.26 Rusin et al27 pro­spec­tively recruited 36 patients with
ing to the International Society of Thrombosis and Haemostasis CVT who were treated with DOACs, includ­ing dabigatran (n = 18),
(ISTH) cri­te­ria.23 Mean treat­ment dura­tion was 5.1 months in the rivaroxaban (n = 10), and apixaban (n = 8), for a median dura­tion of
dabigatran arm and 5.3 months in the war­fa­rin group, with a mean 8.5 months. Of the patients, 94.4% (n = 34) were noted to have
time in ther­a­peu­tic range of 66.1% for war­fa­rin-treated patients.22 com­plete or par­tial recan­a­li­za­tion of the cere­bral veins.23 CVT
There were no recur­rent venous throm­bo­em­bolic events in either recur­rence was observed in 5.6% (n = 2) after anticoagulation was
treat­ment group, and the inci­dence of major bleed­ing was low discontinued.27 Major bleed­ing occurred in 8.3% (n = 3; 2 rivarox-
in this study. Two patients treated with war­ fa­
rin expe­ri­
enced aban [men­or­rha­gia], 1 dabigatran [GI bleed­ing]), which was rel­a­
major intra­cra­nial bleed­ing (3.3%; 95% CI, 0.4%-11.5%), 1 patient tively high com­pared with other stud­ies.27 DOACs as the upfront
had major GI bleed­ing (1.7%; 95% CI, 0.0%-8.9%), and another treat­ment of acute phase CVT with­out hep­a­rin were exam­ined in
patient expe­ri­enced clin­i­cally rel­e­vant non­ma­jor gen­i­to­uri­nary a pro­spec­tive study by Shankar Iyer et al.28 The study was small
bleed­ing.22 The sec­ond­ary end point of cere­bral vein recan­a­li­za­ (n = 20), and crit­ i­
cally ill patients were excluded.28 All patients
tion occurred in 33 patients (60.0%; 95% CI, 45.9%-73.0%) in the were treated with rivaroxaban only, with­out hep­a­rin or LMWH
dabigatran group and 35 patients (67.3%; 95% CI, 52.9%-79.7%) in ther­apy.28 Cerebral vein recan­a­li­za­tion was seen in all­patients
the war­fa­rin group.18 An excel­lent func­tional out­come (mod­i­fied (60% com­plete and 40% par­tial), and excel­lent func­tional out­
Rankin scale 0-1 points) was seen in 91.5% and 91.4% in the war­fa­ come was noted in 95% (n = 19) of the patients.28 Patients were
rin and dabigatran groups, respec­tively.22 Based on the results of treated for a median of 6 months with no major bleed­ing noted.28
the above trial, dabigatran and war­fa­rin appear to have a sim­i­lar At least 5 ret­ro­spec­tive obser­va­tional stud­ies were published
effi­cacy and safety pro­file in the treat­ment of patients with CVT between 2014 and 2019.29-33 Sample sizes were small (range n = 6-
after brief, ini­tial ther­apy with unfractionated heparin or LMWH. 15 patients). Dabigatran was the most com­ monly pre­ scribed
A sys­tem­atic review and meta-anal­y­sis of 6 stud­ies (5 obser­ DOAC, followed by rivaroxaban and apixaban, respec­ tively.
va­tional and 1 ran­dom­ized) exam­in­ing the safety and effi­cacy Treatment dura­tion ranged from 6 to 12 months. Recurrent VTE
of DOACs vs vita­ min K antag­o­
nists (VKAs) was published in was not observed in any of the patients reported in these ret­ro­
2020.24 There were 412 patients in total, and 151 were treated spec­tive stud­ies. Cerebral vein recan­a­li­za­tion rates ranged from
with DOACs and 261 with VKAs.20 Provoking fac­tors that were 55.6% to 100%.29-33 Bleeding was min­i­mal, with no major bleed­
reported in the stud­ies included oral con­tra­cep­tive use (24.2%, ing observed. Minor bleed­ing ranged from 0% to 28.6%.29-33
n = 100/412) and preg­ nancy (6.3%, n  = 
26/412). DOAC effi­ cacy The afore­men­tioned ADAM VTE trial of apixaban in patients
was com­pa­ra­ble to VKA for cere­bral vein recan­a­li­za­tion (rel­a­tive with can­cer included only a sin­gle patient with CVT.18 In a ret­ro­
risk [RR], 1.02; 95% CI, 0.89-1.16) and excel­lent func­tional out­ spec­tive MD Anderson study of 45 patients with can­cer and CVT,
comes as mea­sured by a mod­i­fied Rankin scale less than 2 (RR, 33 were treated with anti­co­ag­u­lants (LMWH, 23 cases; war­fa­rin,
1.02; 95% CI, 0.93-1.13).24 There was a trend toward less major 10 cases).34 The spe­cific recur­rent rates of not anticoagulated,
bleed­ing in the DOAC treat­ment group, but this did not reach LMWH-treated, and war­fa­rin-treated patients were 25.0%, 13.0%,
sta­tis­ti­cal sig­nif­i­cance (RR, 0.44; 95% CI, 0.12-1.59).24 Another and 10.0%, respec­tively, which did not reach sta­tis­ti­cal sig­nif­i­
sys­tem­atic review that included 33 stud­ies (1 ran­dom­ized con­ cance. The inci­dence of intracranial hemorrhage after CVT diag­
trolled trial, 5 obser­va­tional, 27 case series/stud­ies) reported no­sis was higher in the group receiv­ing anticoagulation (30.3%;
on patients with CVT who were treated with DOACs (n = 279) vs 95% CI, 16.8%-47.1%) vs no anticoagulation (25.0%; 95% CI, 7.6%-
VKAs (n = 315).25 The most com­mon DOAC was rivaroxaban (47%), 52.9%), with no sig­nif­i­cant sta­tis­ti­cal dif­fer­ence (P = 1.000). The
followed by dabigatran (41%), apixaban (10%), and edoxaban choice of anti­co­ag­u­lant was asso­ci­ated with bleed­ing events
(2%).25 Results showed a sim­i­lar risk of death in the DOAC and (LMWH, 34.8%; war­fa­rin, 80.0%; odds ratio, 7.50; 95% CI, 1.05-54.3;
VKA arms (RR, 2.12; 95% CI, 0.29-15.59).25 Rates of new intra­cra­ P = .048). No patients were treated with DOACs in this can­cer trial.

102  |  Hematology 2021  |  ASH Education Program


Table 2. Ongoing tri­als eval­u­at­ing DOACs in the treat­ment of throm­bo­sis in unusual sites

Site of Primary No. of


Title Identifier Study design Intervention throm­bo­sis out­come patients Sponsor
The effi­cacy and NCT03217448 Phase 3 Dabigatran vs CVT Recanalization 80 Capital Medical
safety of dabigatran (interventional), war­fa­rin for after 6 University,
etexilate com­pared ran­dom­ized, 6 months months Beijing, China
with war­fa­rin for par­al­lel assign­
the anticoagulation ment, open label,
treat­ment of cere­bral sin­gle (out­comes
venous throm­bo­sis: a asses­sor)
pilot study
Multicenter, pro­spec­ NCT03178864 Interventional, Rivaroxaban vs CVT Safety of 100 University of
tive ran­dom­ized open ran­dom­ized, par­ stan­dard of rivaroxaban Brit­ish Colum­

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/100/1851729/100mathew.pdf by guest on 13 December 2021


label, blinded end­point al­lel assign­ment, care bia, Canada
(PROBE) con­trolled trial sin­gle (out­comes
of early anticoagulation asses­sor)
with rivaroxaban ver­sus
stan­dard of care in
deter­min­ing safety at
365 days in symp­tom­
atic cere­bral venous
throm­bo­sis (SECRET)
Comparing treat­ment NCT03747081 Interventional, ran­ Rivaroxaban vs CVT Efficacy of 50 Isfahan
out­comes in CVT dom­ized, par­al­lel war­fa­rin rivaroxaban University
patients who were assign­ment vs war­fa­rin of Medical
treated with war­fa­ Sciences, Iran
rin and rivaroxaban in
Isfahan, Iran
Rivaroxaban com­pared to NCT04569279 Interventional, par­al­ Rivaroxaban vs CVT Change of 71 Damascus
war­fa­rin for treat­ment lel assign­ment, war­fa­rin 6 sinus venous University,
of cere­bral venous open label months throm­bo­sis Syria
throm­bo­sis: a ran­dom­ sever­ity scale
ized con­trolled trial
Comparison of the effi­ NCT03191305 Interventional, Rivaroxaban vs CVT Efficacy of 50 Foundation
cacy of rivaroxaban to nonrandomized, coumadin rivaroxaban University
coumadin (war­fa­rin) in par­al­lel assign­ and com­ Islamabad,
cere­bral venous throm­ ment par­i­son to Pakistan
bo­sis war­fa­rin
Treatment of por­tal, mes­ NCT02627053 Interventional, Rivaroxaban PVT, MVT, Safety and 100 Università
en­teric, and splenic sin­gle group SVT effi­cacy of degli Studi
vein throm­bo­sis with assign­ment, pro­ rivaroxaban dell’Insubria,
rivaroxaban. A pilot, spec­tive, cohort at 3 months Italy
pro­spec­tive cohort study
study
Direct oral anti­co­ag­u­lants NCT04660747 Observational, com­ 3:2 ratio CVT Composite 500 Academisch
for the treat­ment of par­a­tive cohort DOAC/VKA of major Medisch
cere­bral venous throm­ study, pro­spec­ bleed­ing and Centrum—
bo­sis: an inter­na­tional tive symp­tom­atic Universiteit
phase IV study recur­rent van
venous Amsterdam
throm­bo­ (AMC-
sis after 6 UvA), The
months Netherlands
International reg­is­try on NCT03778502 Observational, Any DOAC All sites Evaluate 100 University of
the use of the direct patient reg­is­ the use of Malta, Malta
oral anti­co­ag­u­lants try, pro­spec­tive, DOACs in
for the treat­ment of cohort study unusual-site
unusual site venous VTE and to
throm­bo­em­bo­lism assess safety
and effec­
tive­ness

Prakash Singh Shekhawat


Warfarin vs DOAC in SVT or CVT  |  103
The authors con­cluded that a nar­row ther­a­peu­tic index of antico- base­line pro­throm­bin time, we rec­om­mend use of LMWH and
agulation may exist in can­cer-asso­ci­ated CVT.34 con­sid­er­ation of dose reduc­tion based on the degree of throm­
Despite being off label, the DOACs are being pre­ scribed bo­cy­to­pe­nia and coagulopathy. In SVT, we rec­om­mend treat­
in an increas­ing num­ber of patients with SVT and CVT. A 2020 ment of at least 3 to 6 months. Extended-dura­tion anticoagula-
cross-sec­tional sur­vey of mem­bers of 3 dif­fer­ent hemo­sta­sis and tion should be con­sid­ered in patients with ongo­ing risk fac­tors
throm­bo­sis soci­e­ties explored choices of anti­co­ag­u­lant ther­apy for throm­bo­sis.
in 4 dif­fer­ent clin­i­cal vignettes.35 Across the 4 vignettes, great CVT: Based on the avail­­ able data, we rec­ om­mend use of
var­i­
abil­
ity existed, but VKAs were the most com­ mon choice either war­fa­rin or dabigatran for treat­ment of an ini­tial epi­sode
(44%-63%), followed by the DOACs (23%-27%) in low bleed­ing of CVT for a dura­tion of at least 3 to 6 months. Extended-dura­tion
risk patients.35 In high bleed­ing risk sce­nar­ios, par­en­teral agents anticoagulation should be con­sid­ered in patients with ongo­ing
were the sec­ond most com­mon choice.35 risk fac­tors for throm­bo­sis. In patients who can­not receive war­
Most of the stud­ies presented in this arti­cle have many lim­i­ fa­rin or dabigatran, use of rivaroxaban and apixaban can be con­
ta­tions, mak­ing defin­i­tive rec­om­men­da­tions dif­fi­cult; few were sid­ered based on pro­spec­tive and ret­ro­spec­tive stud­ies.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/100/1851729/100mathew.pdf by guest on 13 December 2021


ran­dom­ized, sites of throm­bo­sis dif­fered, type of DOAC use dif­
fered, and length of ther­apy was het­ero­ge­neous. Future stud­ies Conflict-of-inter­est dis­clo­sure
should be ran­dom­ized, include well-defined sites of throm­bo­sis Carol Mathew: no rel­e­vant con­flicts to dis­close.
(por­tal, mes­en­teric, hepatic, or cere­bral), choose a sin­gle DOAC Marc Zumberg: mem­ber, ABIM Hematology Board, ASH speaker
to com­pare with war­fa­rin, and treat for a con­sis­tent length of reim­burse­ment, law case review.
time. Multiple tri­als test­ing DOACs in clots in unusual places are
ongo­ing, as sum­ma­rized by Riva et al.36 Off-label drug use
Carol Mathew: DOAC use in splanchnic and cerebral vein throm-
Current soci­ety guide­lines/expert opin­ion bosis is discussed.
Riva and Ageno,9 in a state-of-the-art ISTH 2020 report, con­ Marc Zumberg: DOAC use in splanchnic and cerebral vein throm-
clude that the cur­rent evi­dence sug­gests that DOACs can be bosis is discussed.
used in select patients with unusual-site throm­bo­sis given com­
pa­ra­ble effi­cacy and a trend toward lower bleed­ing com­pared Correspondence
with VKAs. The authors stress that cau­tion should be taken in Marc Zumberg, University of Florida Health, 1600 S.W. Archer Rd,
high-risk pop­u­la­tions such as those with cir­rho­sis, varices, cen­ Gainesville, FL 32610; e-mail: zumbems@med­i­cine​­.ufl​­.edu.
tral ner­vous sys­tem infec­tion, or trauma. In addi­tion, they note
that indi­vid­u­al­ized deci­sions need to be con­sid­ered in patients
References
with Child-Pugh class B to C liver dis­ease, mod­er­ate to severe 1. Di Nisio M, Valeriani E, Riva N, Schulman S, Beyer-Westendorf J, Ageno W.
renal dys­func­tion, lumi­nal GI malig­nancy (SVT), and those tak­ing Anticoagulant ther­apy for splanch­nic vein throm­bo­sis: ISTH SSC sub­com­
anti­sei­zure med­i­ca­tion (CVT).9 mit­tee con­ trol of anticoagulation. J Thromb Haemost. 2020;18(7):1562-
In 2016, rec­om­men­da­tions by the European Association for 1568.
2. Euro­pean Association for the Study of the Liver. EASL clin­ i­cal prac­tice
the Study of the Liver included LMWH followed by VKAs for non­
guide­lines: vas­cu­lar dis­eases of the liver. J Hepatol. 2016;64(1):179-202.
ma­lig­nant, noncirrhotic PVT.2 3. Kearon C, Akl EA, Comerota AJ, et  al. Antithrombotic ther­apy for VTE
The Euro­ pean Society for Vascular Surgery recommended dis­ease: antithrombotic ther­apy and pre­ven­tion of throm­bo­sis, 9th ed:
LMWH or unfractionated heparin followed by either VKA or DOAC Amer­i­can College of Chest Physicians Evidence-Based Clinical Practice
Guidelines. Chest. 2012;141(2, suppl):e419S-e496S.
in patients with MVT.36,37
4. Valeriani E, Di Nisio M, Riva N, et  al. Anticoagulant ther­apy for splanch­
The most recent guide­lines for CVT treat­ment from the Euro­ nic vein throm­bo­sis: a sys­tem­atic review and meta-anal­y­sis. Blood.
pean Stroke Organization and endorsed by the Euro­pean Acad- 2021;137(9):1233-1240.
emy of Neurology do not rec­om­mend DOAC use in the acute 5. Valeriani E, Di Nisio M, Riva N, et al. Anticoagulant treat­ment for splanch­nic
phase of CVT treat­ment but sug­gest a pref­er­ence for LMWH.38 vein throm­bo­sis in liver cir­rho­sis: a sys­tem­atic review and meta-anal­y­sis.
Thromb Haemost. 2021;121(7):867-876.
These guide­lines were published before the results of the RE- 6. Ghazaleh S, Beran A, Aburayyan K, et al. Efficacy and safety of anticoagula-
SPECT CVT trial and most of the pro­spec­tive trial data men­tioned tion in non-malig­nant por­tal vein throm­bo­sis in patients with liver cir­rho­sis:
above. With the new infor­ma­tion, upcom­ing guide­lines would a sys­tem­atic review and meta-anal­y­sis. Ann Gastroenterol. 2021;34(1):104-
likely include use of DOACs in CVT as sec­ond­ary pro­phy­laxis. 110.
7. de Bruijn SF, Stam J. Randomized, pla­cebo-con­trolled trial of anti­co­ag­ul­ant
Ongoing tri­als should allow fur­ther rec­om­men­da­tions in the
treat­ment with low-molec­u­lar-weight hep­a­rin for cere­bral sinus throm­bo­
near future (Table 2). sis. Stroke. 1999;30(3):484-488.
8. Ferro JM, Canhão P, Stam J, Bousser M-G, Barinagarrementeria F; ISCVT
Author rec­om­men­da­tions Investigators. Prognosis of cere­bral vein and dural sinus throm­bo­sis: results
SVT: Based on the avail­­able data, we rec­om­mend use of either of the International Study on Cerebral Vein and Dural Sinus Thrombosis
(ISCVT). Stroke. 2004;35(3):664-670.
war­fa­rin or a DOAC in patients with SVT. In patients with Child- 9. Riva N, Ageno W. Direct oral anti­co­ag­ul­ants for unusual-site venous throm­
Pugh B liver dis­ ease, rivaroxaban should be avoided, and in bo­em­bo­lism. Res Pract Thromb Haemost. 2021;5(2):265-277.
Child-Pugh C liver dis­ease, all­of the DOACs are contraindicated. 10. Kearon C, Akl EA, Ornelas J. et al. Antithrombotic ther­apy for VTE dis­ease:
We rec­om­mend treat­ment for inci­den­tally detected SVT, unless CHEST guide­line and expert panel report. Chest. 2016;149(2):315-352.
11. Ortel TL, Neumann I, Ageno W, et  al. Amer­i­can Society of Hematology
it is con­firmed to be chronic com­pared with prior scans. In pa- 2020 guide­lines for man­age­ment of venous throm­bo­em­bo­lism: treat­
tients with advanced liver dis­ease in whom the DOACs are con- ment of deep vein throm­ bo­sis and pul­ mo­
nary embolism. Blood Adv.
traindicated and war­fa­rin would be dif­fi­cult, given the ele­vated 2020;4(19):4693-4738.

104  |  Hematology 2021  |  ASH Education Program


12. Hanafy AS, Abd-Elsalam S, Dawoud MM. Randomized con­trolled trial of 26. Wasay M, Khan M, Raj­put HM, et al. New oral anti­co­ag­u­lants ver­sus war­
rivaroxaban ver­sus war­fa­rin in the man­age­ment of acute non-neo­plas­tic fa­rin for cere­bral venous throm­bo­sis: a multi-cen­ter, obser­va­tional study. J
por­tal vein throm­bo­sis. Vascul Pharmacol. 2019;113:86-91. Stroke. 2019;21(2):220-223.
13. Serrao A, Merli M, Lucani B, et  al. Outcomes of long-term anti­co­ag­u­lant 27. Rusin G, Wypasek E, Papuga-Szela E, Żuk J, Undas A. Direct oral anti­co­ag­u­
treat­ment for the sec­ond­ary pro­phy­laxis of splanch­nic venous throm­bo­sis. lants in the treat­ment of cere­bral venous sinus throm­bo­sis: a sin­gle insti­tu­
Eur J Clin Invest. 2021;51(1):e13356. tion’s expe­ri­ence. Neurol Neurochir Pol. 2019;53(5):384-387.
14. Naymagon L, Tremblay D, Zubizarreta N, et al. The effi­cacy and safety of 28. Shankar Iyer R, Tcr R, Akhtar S, Muthukalathi K, Kumar P, Muthukumar K.
direct oral anti­co­ag­u­lants in noncirrhotic por­tal vein throm­bo­sis. Blood Is it safe to treat cere­bral venous throm­bo­sis with oral rivaroxaban with­
Adv. 2020;4(4):655-666. out hep­a­rin? A pre­lim­i­nary study from 20 patients. Clin Neurol Neurosurg.
15. Kawata E, Siew D-A, Payne J-G, Louzada M, Kovacs M-J, Lazo-Langner A. 2018;175:108-111.
Splanchnic vein throm­bo­sis: clin­i­cal man­i­fes­ta­tions, risk fac­tors, man­age­ 29. Geisbüsch C, Richter D, Herweh C, Ringleb PA, Nagel S. Novel fac­tor Xa
ment, and out­comes. Thromb Res. 2021;202:90-95. inhib­i­tor for the treat­ment of cere­bral venous and sinus throm­bo­sis: first
16. Sharma S, Kumar R, Rout G, Gamanagatti SR, Shalimar. Dabigatran as an expe­ri­ence in 7 patients. Stroke. 2014;45(8):2469-2471.
oral anti­co­ag­u­lant in patients with Budd-Chiari syn­drome post-per­cu­ta­ne­ 30. Mendonça MD, Barbosa R, Cruz-e-Silva V, Calado S, Viana-Baptista M. Oral
ous endovascular inter­ven­tion. J Gastroenterol Hepatol. 2020;35(4):654- direct throm­bin inhib­i­tor as an alter­na­tive in the man­age­ment of cere­bral
662. venous throm­bo­sis: a series of 15 patients. Int J Stroke. 2015;10(7):1115-1118.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/100/1851729/100mathew.pdf by guest on 13 December 2021


17. Salim S, Ekberg O, Elf J, Zarrouk M, Gottsäter A, Acosta S. Evaluation of 31. Anticoli S, Pezzella FR, Scifoni G, Ferrari C, Pozzessere C. Treatment of cere­
direct oral anti­co­ag­u­lants and vita­min K antag­o­nists in mes­en­teric venous bral venous throm­bo­sis with rivaroxaban. J Biomed Sci. 2016;5(3):3.
throm­bo­sis. Phlebology. 2019;34(3):171-178. 32. Herweh C, Griebe M, Geisbüsch C, et  al. Frequency and tem­po­ral pro­
18. McBan RD II, Wysokinski WE, Le-Rademacher JG, et  al. Apixaban and file of recan­a­li­za­tion after cere­bral vein and sinus throm­bo­sis. Eur J Neurol.
dalteparin in active malig­nancy-asso­ci­ated venous throm­bo­em­bo­lism: the 2016;23(4):681-687.
ADAM VTE trial. J Thromb Haemost. 2020;18(2):411-421. 33. Covut F, Kewan T, Perez O, Flores M, Haddad A, Daw H. Apixaban and
19. Raskob GE, van Es N, Verhamme P, et al; Hokusai VTE can­cer inves­ti­ga­tors. rivaroxaban in patients with cere­ bral venous throm­ bo­sis. Thromb Res.
Edoxaban for the treat­ment of can­cer-asso­ci­ated venous throm­bo­em­bo­ 2019;173:77-78.
lism. N Engl J Med. 2018;378(7):615-624. 34. Abelhad NI, Qiao W, Garg N, Rojas-Hernandez CM. Thrombosis and bleed­
20. Young AM, Marshall A, Thirlwall J, et  al. Comparison of an oral fac­tor Xa ing out­comes in the treat­ment of cere­bral venous throm­bo­sis in can­cer.
inhib­i­tor with low molec­u­lar weight hep­a­rin in patients with can­cer with Thromb J. 2021;19(1):49.
venous throm­bo­em­bo­lism: results of a ran­dom­ized trial (SELECT-D). J Clin 35. Riva N, Carrier M, Gatt A, Ageno W. Anticoagulation in splanch­nic and cere­
Oncol. 2018;36(20):2017-2023. bral vein throm­bo­sis: an inter­na­tional vignette-based sur­vey. Res Pract
21. Agnelli G, Becattini C, Meyer G, et al; Caravaggio Investigators. Apixaban Thromb Haemost. 2020;4(7):1192-1202.
for the treat­ment of venous throm­bo­em­bo­lism asso­ci­ated with can­cer. N 36. Riva N, Ageno W. Cerebral and splanch­nic vein throm­bo­sis: advances,
Engl J Med. 2020;382(17):1599-1607. chal­lenges, and unanswered ques­tions. J Clin Med. 2020;9(3):743.
22. Ferro JM, Coutinho JM, Dentali F, et al; RE-SPECT CVT Study Group. Safety 37. Björck M, Koelemay M, Acosta S, et al. Editor’s choice—man­age­ment of the
and effi­cacy of dabigatran etexilate vs dose-adjusted war­fa­rin in patients dis­eases of mes­en­teric arteries and veins: clin­i­cal Practice Guidelines of the
with cere­bral venous throm­bo­sis: a ran­dom­ized clin­i­cal trial. JAMA Neurol. Euro­pean Society of Vascular Surgery (ESVS). Eur J Vasc Endovasc Surg.
2019;76(12):1457-1465. 2017;53(4):460-510.
23. Schulman S, Kearon C; Subcommittee on Control of Anticoagulation of the 38. Ferro JM, Bousser M-G, Canhão P, et al; Euro­pean Stroke Organization.
Scientific and Standardization Committee of the International Society on Euro­pean Stroke Organization guide­line for the diag­no­sis and treat­ment
Thrombosis and Haemostasis. Definition of major bleed­ing in clin­i­cal inves­ of cere­bral venous throm­bo­sis—endorsed by the Euro­pean Academy of
ti­ga­tions of antihemostatic medic­i­nal prod­ucts in non-sur­gi­cal patients. J Neurology. Eur J Neurol. 2017;24(10):1203-1213.
Thromb Haemost. 2005;3(4):692-694.
24. Lee GKH, Chen VH, Tan C-H, et al. Comparing the effi­cacy and safety of
direct oral anti­co­ag­u­lants with vita­min K antag­o­nist in cere­bral venous
throm­bo­sis. J Thromb Thrombolysis. 2020;50(3):724-731.
25. Bose G, Graveline J, Yogendrakumar V, et al. Direct oral anti­co­ag­u­lants in
treat­ment of cere­bral venous throm­bo­sis: a sys­tem­atic review. BMJ Open. © 2021 by The Amer­i­can Society of Hematology
2021;11(2):e040212. DOI 10.1182/hema­tol­ogy.2021000319

Prakash Singh Shekhawat


Warfarin vs DOAC in SVT or CVT  |  105
CML: SUCCESS BREEDS MORE SUCCESS

TKI discontinuation in CML: how do we make


more patients eligible? How do we increase the

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/106/1851817/106hochhaus.pdf by guest on 13 December 2021


chances of a successful treatment-free remission?
Andreas Hochhaus and Thomas Ernst
Klinik für Innere Medizin II, Universitätsklinikum Jena, Jena, Germany

Treatment-free remission (TFR) is a new and significant goal of chronic myeloid leukemia management. TFR should be
considered for patients in stable deep molecular response (DMR) after careful discussion in the shared decision-making
process. Second-generation tyrosine kinase inhibitors (TKIs) improve the speed of response and the incidence of DMR.
Treatment may be changed to a more active TKI to improve the depth of response in selected patients who have not
reached DMR. Stem cell persistence is associated with active immune surveillance and activation of BCR-ABL1-inde-
pendent pathways, eg, STAT3, JAK1/2, and BCL2. Ongoing studies aim to prove the efficacy of maintenance therapies
targeting these pathways after TKI discontinuation.

LEARNING OBJECTIVES
• Review the options for achieving deep molecular remission in CML patients, with the possibility of discontinuing
therapy and staying in long-term treatment-free remission, which is equivalent to an operational cure
• Discuss the biology of persisting leukemic stem cells as the origin of relapse
• Defne the mechanism of action of experimental therapies to eradicate or silence residual leukemia

Introduction harmonize recommendations for treatment discontinuation


A signifcant proportion of chronic myeloid leukemia (CML) and increase the safety of stopping procedures.4,5
patients achieve a deep molecular response (DMR) with As a multicenter academic approach, the European
tyrosine kinase inhibitor (TKI) therapy defned as residual Stop Kinase Inhibitor (EURO-SKI, NCT01596114) study ana-
BCR-ABL1 transcript levels of at least MR4 on the interna- lyzed 755 patients with similar overall results. The minimum
tional scale (IS). The speed of response is faster, and the requirements for stopping in most studies were a duration
incidence of DMR at specifc time points is higher under of TKI therapy of at least 3 years and a sustained DMR of at
treatment with second-generation inhibitors (2G-TKIs) least 1 year.6
than with imatinib.1 In general, about 50% of patients will relapse, regardless
An attempt to discontinue therapy can be considered of the TKI used. Most molecular recurrences occur within
after the achievement and long-term maintenance of DMR. the frst 6 months after TKI discontinuation. However, treat-
The concept was established with the Stop Imatinib 1 (STIM1) ment discontinuation is safe if eligible patients are care-
trial. After a median follow-up of 77 months after stopping fully selected, and high-quality standardized qRT-PCR is
TKI therapy, 38% of patients maintained a molecular remis- employed. The available data and procedures required
sion. In this trial, patients were eligible for treatment dis- should be discussed with the patient. Some patients eli-
continuation with negative results of a sensitive quantitative gible for treatment-free remission (TFR) may prefer to
reverse transcriptase polymerase chain reaction (qRT-PCR) remain on their current treatment. A loss of major molecular
sustained for 2 years before stopping.2,3 After this innova- response (MMR; BCR-ABL1>0.1% IS) has been established as
tive pilot study, multiple trials were conducted, each with the trigger for restarting therapy.5,7 However, considering
different entry criteria and defnitions of relapse, prompt- that the rate of DMR in a newly diagnosed patient on cur-
ing a restart of TKIs. Concurrently, PCR methodology and rent treatment options is about 40% and the chance for sta-
defnitions of PCR sensitivity were standardized in order to ble TFR is about 50%, the overall chance of long-term TFR is

106 | Hematology 2021 | ASH Education Program


only 20% for a CML patient newly diag­nosed in 2021. In addi­tion, Confirmation of loss of MMR with a sec­ond PCR is not recom-
the cost of TKI ther­apy is a sig­nif­i­cant prob­lem for patients and mended and could delay restarting ther­apy. Almost all­patients
the health sys­tem in gen­eral. Hence, it is cru­cial to increase the with molec­u­lar recur­rence regain their ini­tial MR level after a TKI
rate of dura­ble DMR and to con­vert DMR into func­tional cures, restart. The choice of TKI for the restart depends on the pre­vi­ous
defined as nonrecurrence of active CML after stop­ping TKIs.8 tol­er­a­bil­ity, comorbidities, risk of long-term tox­ic­ity, and expec­ta­
tions toward a sec­ond TFR attempt. So far, very few of the thou­
sands of patients in TFR tri­als have had unfa­vor­able out­comes.
There are occa­sional reports of pro­gres­sion to blast phase, but
CLINICAL CASE it is uncer­tain whether TKI dis­con­tin­u­a­tion con­trib­uted to the
under­ly­ing biol­ogy of pro­gres­sion. Irrespective of higher eli­gi­bil­ity
Our patient is a 27-year-old woman, a teacher, diag­nosed with rates for TFR stud­ies with sec­ond-gen­er­a­tion TKI, the prob­a­bil­ity
chronic-phase CML in May 2017. Calculation of the EUTOS long- of maintaining TFR has been about 50% after nilotinib or dasatinib
term sur­vival score revealed low-risk dis­ease. Cytogenetics dem­ ther­a­pies, sim­i­lar to the results after stop­ping imatinib.9-13

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/106/1851817/106hochhaus.pdf by guest on 13 December 2021


on­strated the involve­ ment of 3 chro­ mo­somes, 46,XX,t(1;9;22) The French RE-STIM trial inves­ti­gated the out­come of sec­ond
(q25;q34;q11), but mul­ti­plex PCR revealed a typ­i­cal e14a2 BCR- or sub­se­quent TFR attempts. Seventy patients who regained
ABL1 tran­script. Using next-gen­er­a­tion sequenc­ing, no BCR-ABL1- DMR after molec­u­lar recur­rence after the first TKI dis­con­tin­u­a­tion
inde­pen­dent muta­tions were detected.Spleen size was nor­mal, stopped treat­ment for a sec­ond time. The pro­por­tion of patients
and no med­i­ca­tion or con­com­i­tant dis­eases were reported. There with­out molec­u­lar recur­rence was 35% at 36 months after the
was a strong desire to estab­lish a fam­ily in the com­ing years. We sec­ond TKI dis­con­tin­u­a­tion. RE-STIM data actu­ally sug­gest that
thor­oughly discussed with the patient the diag­no­sis, treat­ment patients in TFR los­ing MMR have a lower chance for a suc­cess­
options, goals of ther­apy, and risks and ben­e­fits of each treat­ ful sec­ond TFR attempt than patients with molec­u­lar recur­rence
ment option. Shared deci­sion-mak­ing led to the con­clu­sion to according to the orig­i­nal STIM def­i­ni­tion (pos­i­tive PCR).14
start nilotinib 300 mg twice daily in June 2017. Treatment was well
tol­er­ated; the only side effect was a grade 1 facial exan­thema. Patients’ per­spec­tive
Blood counts nor­mal­ized within 4 weeks. BCR-ABL1 tran­script lev­ In gen­eral, rec­om­men­da­tions for safe dis­con­tin­u­a­tion and treat­
els in periph­eral blood were mea­sured in a spe­cial­ized lab stan­ ment restart are well established and published. However, the
dard­ized for sen­si­tive quan­ti­fi­ca­tion according to the IS: expe­ri­ence of patients with regard to their deci­sion about TFR
Sep­tem­ber 2017: 3.2% and their needs dur­ing TFR are widely unknown. Using a sur­vey,
Decem­ber 2017: 0.41% inter­na­tional patient advo­cates sought to check the patient per­
March 2018: 0.23% spec­tive, to iden­tify areas of unmet needs, and to cre­ate rec­om­
June 2018: 0.072% men­da­tions for improve­ments. Fear or anx­i­ety dur­ing treat­ment
Decem­ber 2018: 0.015% dis­con­tin­u­a­tion were reported by 56% of patients, but only 7%
June 2019: 0.0030% of patients were asked about their needs for psy­cho­log­i­cal sup­
Sep­tem­ber 2019: 0.0015% port. After treat­ment restart, 59% of patients felt scared or anx­
Decem­ber 2019: 0.0023% ious, and 56% felt depressed. Twenty-six per­cent of reinitiated
March 2020: 0.0010% patients received psy­cho­log­i­cal and/or emo­tional sup­port. Six-
June 2020: 0% with 96 000 ABL1 tran­scripts (MR4.5). ty per­cent of patients expe­ri­enced with­drawal symp­toms after
treat­ment dis­con­tin­u­a­tion. However, 40% of patients with with­
At this stage, 3 years after the start of ther­apy, the patient drawal symp­toms reported a lack of sup­port from their phy­si­cian
requested that ther­apy be discontinued. in symp­tom man­age­ment. Hence, the mon­i­tor­ing of psy­cho­log­
i­cal con­se­quences before, dur­ing, and after TFR should be con­
sid­ered.15
Clinical expe­ri­ence Musculoskeletal and/or joint pain starting a few weeks or
The pro­spec­tive, nonrandomized EURO-SKI was designed to de- months after TKI dis­con­tin­u­a­tion has been observed in about
fine opti­mal con­di­tions for treat­ment dis­con­tin­u­a­tion. Sixty-one 25% of patients. This TKI with­drawal syn­drome is likely based on
sites in 11 Euro­ pean countries par­ tic­i­
pated. Eligibility cri­te­
ria a release of off-tar­get effects of the TKI. In most cases symp­toms
included chronic-phase CML, TKI treat­ment for at least 3 years are mild and tran­sient, but some patients may require tem­po­rary
(with­out treat­ment fail­ure according to Euro­pean LeukemiaNet anti-inflam­ma­tory treat­ment.16 Surveillance and symp­ tom­atic
rec­om­men­da­tions), and con­firmed MR4 or bet­ter for at least 1 treat­ment of with­drawal symp­toms should be a pri­or­ity dur­ing
year. The pri­mary end point was molec­u­lar relapse-free sur­vival, treat­ment dis­con­tin­u­a­tion.15
defined as sur­vival with­out a loss of MMR. Secondary end points
were the anal­y­sis of prog­nos­tic fac­tors affect­ing MMR main­te­ Predictors of suc­cess
nance and the eco­nomic impact of stop­ping ther­apy. Molecu- Various prog­nos­tic fac­tors for a suc­cess­ful TFR have been re-
lar relapse-free sur­vival was 61% and 50% at 6 and 24 months, ported. Longer dura­tion of TKI ther­apy and DMR and prior treat­
respec­tively. In the prog­nos­tic anal­y­sis of patients who received ment with inter­feron alpha (IFNA) were iden­ti­fied as impor­tant
first-line imatinib ther­apy, lon­ger treat­ment dura­tion and lon­ger pre­dic­tors for suc­cess­ful TFR in EURO-SKI; of these, dura­tion of
DMR dura­tion were advan­ta­geous for main­te­nance of MMR over DMR was shown to be the most impor­tant fac­tor, cor­rob­o­rat­ing
a period of six months. Treatment dis­con­tin­u­a­tion did not result data from a study in Japan.6,17 A Cana­dian TFR study revealed
in seri­ous adverse events. For study par­tic­i­pants, the over­all cost 6 years of imatinib ther­apy and 4.5 years MR4 dura­tion as opti­
sav­ings were esti­mated at about €22 mil­lion.6 mal time frames for a suc­cess­ful TFR.18
Prakash Singh Shekhawat
TKI dis­con­tin­u­a­tion in CML  |  107
Table 1. Predictive data of eli­gi­bil­ity and sta­bil­ity of TFR19,39

Parameter Impact on eli­gi­bil­ity for TFR Impact on sta­bil­ity of TFR Study exam­ples
Prognostic score DMR more fre­quent in low-risk TFR more sta­ble in low-risk STIM, STIM2, ENESTfreedom,
patients patients; risk of relapse high in TWISTER
high-risk patients
TKI type DMR more fre­quent with 2G-TKIs No major dif­fer­ence
vs imatinib
Initial slope of BCR-ABL1 Probability of DMR bet­ter in No major dif­fer­ence
tran­script decline or patients with EMR
“halv­ing time”
MMR at 12 months Probability of DMR bet­ter in No major dif­fer­ence
patients with MMR

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/106/1851817/106hochhaus.pdf by guest on 13 December 2021


Type of BCR-ABL1 tran­script DMR more likely in patients with TFR more sta­ble with e14a2
e14a2 vs e13a2 (incon­sis­tent) (incon­sis­tent)
Mutations out­side BCR-ABL1 MMR and DMR more likely in Data still imma­ture
patients lacking BCR-ABL1-
inde­pen­dent muta­tions
Duration of DMR Longer DMR pre­dic­tive EURO-SKI
Level of MR before stop Probably yes; unde­tect­able EURO-SKI, ENESTfreedom
BCR-ABL1 pre­dic­tive
Duration of TKI ther­apy Inconsistent, overlapping with STIM
dura­tion of DMR
Proportion of nat­u­ral killer cells Reduced risk of relapse after
imatinib and dasatinib but not
after nilotinib
EMR, early molec­u­lar response.

Age and sex have been reported to be asso­ci­ated with During fol­low-up, qRT-PCR revealed an increase of BCR-ABL1
TFR in early stud­ies.6 The Sokal score was reported to pre­dict tran­scripts:
TFR out­come, eg, in the STIM study.2 However, the EURO-SKI
Sep­tem­ber 2020: 0.06% (IS)
study failed to dem­on­strate the pre­dic­tive value of age, sex,
Decem­ber 2020: 0.15%.
and Sokal score for TFR in patients after first-line imatinib. A
selec­tion bias of patients on IFNA ther­apy prior to imatinib Due to loss of MMR, treat­ment reintroduction was discussed.
might play a role. Data published recently indi­cate that the Good response and good tol­er­a­bil­ity of nilotinib was bal­anced
BCR-ABL1 tran­script type might influ­ence TFR out­come, with against a sec­ond chance with an alter­na­tive TKI. The deci­sion
a lower inci­dence of relapse in patients and e14a2 com­pared was made to com­mence dasatinib 100 mg/d, lead­ing to a rapid
to e13a2.19 decline of BCR-ABL1 tran­scripts:
So far, the dura­tion of first-line nilotinib or dasatinib ther­apy
March 2021: 0.0020%.
has not been found to affect the prob­a­bil­ity of TFR suc­cess. The
June 2021: 0% with 290 000 ABL1 tran­scripts (MR5).
dura­tion of DMR on sec­ond-line nilotinib was asso­ci­ated with
suc­cess­ful TFR in ENESTop (Table 1).9,10,19 A bone mar­row aspi­ra­tion was performed to quan­tify per-
Differences in trial designs must be con­sid­ered when results sisting stem cells on a geno­mic level and to char­ac­ter­ize their
from var­i­ous stud­ies are com­pared: the pro­por­tion of suc­cess­ful envi­ron­ment. The opti­mal dura­tion of TKI treat­ment for the sec­
TFR attempts is higher in stud­ies that define dis­ease relapse as ond attempt of TFR is unknown. The inhi­bi­tion of BCR-ABL1-inde­
loss of MMR than in those that spec­ify loss of unde­tect­able BCR- pen­dent path­ways respon­si­ble for stem cell per­sis­tence and for
ABL1 tran­scripts as the cri­te­rion. In addi­tion, a deeper MR prior acti­va­tion of immune sur­veil­lance is a lead­ing topic in cur­rent
to treat­ment dis­con­tin­u­a­tion may be asso­ci­ated with a bet­ter research.
chance of suc­cess­ful TFR.17

Analyzing per­sis­tent stem cells


In sev­
eral stud­ ies, highly sen­si­
tive DNA-based BCR-ABL1 PCR
dem­on­strated per­sis­tence of the CML clone in most patients
CLINICAL CASE (Con­t in­u ed) with neg­a­tive qRT-PCR, indi­cat­ing low BCR-ABL1 expres­sion in
In June 2020, 3 years after starting ther­apy and 1 year in MR4.5, persisting CML stem cells. The detec­tion of the patient-spe­cific
nilotinib ther­apy was discontinued. Grade 2 mus­cle and joint geno­mic BCR-ABL1 break point requires a DNA sam­ple col­lected
pain, respon­sive to acet­amin­op
­ hen and last­ing about 4 weeks, at diag­no­sis.20-22 The anal­y­
sis of bone mar­ row vs periph­
eral
were signs of a mild TKI with­drawal syn­drome. blood for this pur­pose is ongo­ing.23

108  |  Hematology 2021  |  ASH Education Program


In a Euro­pean study, DNA and mRNA BCR-ABL1 lev­els by quan­ Table 2. Clinical tri­als to main­tain TFR in addi­tion to TKI
ti­ta­tive PCR and by dig­i­tal PCR were com­pared. A good cor­re­
la­tion was found at BCR-ABL1 tran­script lev­els above MR4; the TIGER nilotinib ± peg-IFNA2b (phase 3) NCT01657604
cor­re­la­tion was poor for sam­ples taken in DMR. A com­bi­na­tion PETALs nilotinib ± peg-IFNα2a (phase 3) NCT02201459
of geno­mic and RT-PCR was ­able to bet­ter pre­dict molec­u­lar IFNA main­te­nance ther­apy
relapse-free sur­vival after TKI stop.24   TIGER peg-IFNA2b (phase 3) NCT01657604
Leukemic stem cells are CD34+/CD38−, but this phe­no­type   ENDURE pegylated-pro­line-IFNA2b (phase 3) NCT03117816
is not exclu­ sive to CML. Dipeptidyl pep­ ti­
dase-4 (CD26) may Checkpoint inhib­i­tors
rep­ re­sent a spe­ cific marker for CML stem cells.25 Using the   ACTIW pioglitazone (phase 1), avelumab (phase 2) NCT02767063
CD34+CD38−CD26+ phe­no­type, a study iden­ti­fied CML stem cells   Dasatinib nivolumab (phase 1B) NCT02011945
cir­cu­lat­ing in the periph­eral blood in the major­ity of patients in
JAK inhi­bi­tion
TFR after TKI dis­con­tin­u­a­tion,26 which is, how­ever, still conflict-   Preclinical data
ing. Another study iden­ti­fied persisting leu­ke­mic stem cells in   Nilotinib + ruxolitinib phase 1/2 NCT01914484

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/106/1851817/106hochhaus.pdf by guest on 13 December 2021


the bone mar­row of patients in DMR, in con­trast to the periph­   Nilotinib + ruxolitinib phase 1 NCT02253277
eral blood.23 Further, CD93 expres­ sion has been con­ sis­
tently   Nilotinib + ruxolitinib phase 1 NCT01702064
shown in a lin-CD34+CD38−CD90+ pop­u­la­tion with stem cell char­   ABL-TKI + ruxolitinib phase 2 NCT03610971
ac­ter­is­tics.27   ABL-TKI + ruxolitinib phase 2 NCT03654768
Using fluo­res­cence-acti­vated cell sorting and geno­mic PCR, BCL-2 inhi­bi­tion
resid­ual dis­ease in TFR dif­fered according to the cell lin­e­age.   Preclinical data
BCR-ABL1 was iden­ti­fied pre­dom­i­nantly in the lym­phoid com­   Dasatinib ± venetoclax phase 3 NCT02689440
part­ment and never in granulocytes. B cells were more often BTK inhi­bi­tion
BCR-ABL1+ than T cells. Lineage-spe­cific assess­ment of persist-   Preclinical data
ing dis­ease prior to treat­ment dis­con­tin­u­a­tion could improve the
pre­dic­tion of suc­cess­ful TFR.28

Innovative approaches BCL2


JAK, BTK BCL2-related antiapoptotic pro­teins are expressed at higher lev­els
In TKI-resis­tant CML, STAT3 inhi­ bi­
tion was ­ able to reduce sur­ in CML stem cells com­pared to nor­mal stem cells. Inhibiting BCL2
vival of CML cells. Kuepper et al iden­ti­fied Jak1 but not Jak2 as the increases kill­ing of CML stem cells by TKIs in mod­els. Venetoclax
STAT3-acti­vat­ing kinase. The com­bi­na­tion of BCR-ABL1 and JAK1 com­bined with a TKI reduces the serial re-engraft­ment capac­ity
inhib­i­tors fur­ther reduced col­ony-forming units from murine CML of CML stem cells in mice bet­ter than a TKI alone. Thus, BCL2 inhi­
bone mar­row and human CML mono­nu­clear cells as well as CD34+ bi­tion may be another option for targeting persisting CML stem
CML cells. Combination ther­apy induced apo­pto­sis even in qui­es­ cells unre­spon­sive to BCR-ABL1 TKIs.31,32 A ran­dom­ized first-line
cent leu­ke­mic stem cells, suggesting JAK1 as a poten­tial ther­a­peu­ study test­ing the impact of the addi­tion of venetoclax to dasati-
tic tar­get for cura­tive CML ther­a­pies.29 Ruxolitinib, a dual JAK1/2 nib in newly diag­nosed CML patients is ongo­ing (NCT02689440).
inhib­i­tor, com­bined with nilotinib may selec­tively decrease CML However, the impact of venetoclax on per­sis­tent stem cells should
stem cells.25 A few early-phase stud­ies test­ing the com­bi­na­tion of be tested in a main­te­nance for­mat after achieve­ment of DMR.
BCR-ABL1 targeting TKIs with ruxolitinib were pre­ma­turely closed
due to tox­ic­ity; oth­ers are still recruiting (Table 2). Fc gamma Immunotherapy
recep­tor IIb (CD32b) was shown to be crit­i­cal in stem cell per­sis­ Numerous stud­ies have revealed an asso­ci­a­tion between cel­
tence. Hence, targeting FcγRIIb down­stream sig­nal­ing, eg, with a lu­lar immune param­e­ters and TFR, includ­ing higher num­bers of
BTK inhib­i­tor, pro­vi­des a prom­is­ing ther­a­peu­tic approach.30 ­nat­u­ral killer cells and lower num­bers of both T reg­u­la­tory cells

Figure 1. Innovative strategies to improve chances


Prakash of treatment-free
Singh Shekhawat remission.

TKI dis­con­tin­u­a­tion in CML  |  109


Table 3. Recommendations for TFR

National Comprehensive Cancer French Chronic Myeloid Leukemia


Parameter Euro­pean LeukemiaNet 20205 Network 202138 Study Group 201839
Mandatory
  Age, phase of the dis­ease, Adult patients, CML in first Age ≥18 years. Chronic-phase CML. Age ≥18 years. Chronic-phase CML.
dis­ease his­tory chronic phase only. No prior No prior his­tory of accel­er­ated No prior his­tory of allo­ge­neic
treat­ment fail­ure. or blast-phase CML. stem cell trans­plan­ta­tion, pro­
gres­sion, resis­tance, sub­op­ti­mal
response, or warn­ing.
Motivation, com­mu­ni­ca­tion, Motivated patient with struc­tured Consultation with a CML spe­cial­ Adherence to mon­i­tor­ing and
infor­ma­tion com­mu­ni­ca­tion. ist to review eli­gi­bil­ity for TKI retreatment rec­om­men­da­tions.
Patient’s accep­tance of more fre­ dis­con­tin­u­a­tion and poten­tial Written infor­ma­tion and instruc­
quent PCR tests after stop­ping risks and ben­e­fits of treat­ tions may be pro­vided.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/106/1851817/106hochhaus.pdf by guest on 13 December 2021


treat­ment. ment dis­con­tin­u­a­tion, includ­
ing TKI with­drawal syn­drome.
Discontinuation of TKI ther­apy
should only be performed in
consenting patients after a thor­
ough dis­cus­sion of the poten­tial
risks and ben­e­fits.
  Molecular-mon­i­tor­ing infra­ Access to high-qual­ity qRT-PCR Access to a reli­able quan­ti­ta­tive Results should be avail­­able within
struc­ture using the IS with rapid turn­ PCR test with a sen­si­tiv­ity of 2-3 weeks. Molecular biol­o­gists
around of PCR test results detec­tion of at least MR4.5 should be aware of TKI dis­con­
(BCR-ABL1 ≤ 0.0032% IS) that tin­u­a­tions in indi­vid­ual patients.
pro­vi­des results within 2 weeks.
  Molecular-mon­i­tor­ing fre­ Monthly molec­u­lar mon­i­tor­ing for the first 6 months, bimonthly dur­ing Monthly dur­ing the first half year,
quency months 7-12, and quar­terly there­af­ter (indef­i­nitely) every 2 months. In the sec­ond
half year, quar­terly dur­ing the
sec­ond year and then every 3 to
6 months.
Minimal (stop allowed)
  Line of treat­ment First-line or sec­ond-line ther­apy
if intol­er­ance was the only rea­
son for chang­ing TKI
  BCR-ABL1 tran­script type Typical e13a2 or e14a2 BCR-ABL1 Prior evi­dence of a quan­ti­fi­able BCR-ABL1 tran­scripts e13a2, e14a2,
tran­scripts BCR-ABL1 tran­script or e13a2/e14a2
  Duration of TKI ther­apy Duration of TKI ther­apy >5 years On approved TKI ther­apy for at TKI treat­ment dura­tion ≥5 years.
(>4 years for sec­ond-gen­er­a­tion least 3 years
TKI)
  Duration of DMR DMR (MR4 or bet­ter) for >2 years Stable MR (MR4; BCR-ABL1 ≤ 0.01% DMR at least MR4.5.
IS) for ≥2 years, as documented DMR dura­tion ≥2 years.
on at least 4 tests, performed at MR4.5 is con­firmed in ≥4 con­sec­
least 3 mo apart. u­tive tests. At most 1 of the 4
assess­ments show­ing an MR4 is
accepted.
Optimal (stop recommended for con­sid­er­ation)
Duration of TKI ther­apy >5 years
Duration of DMR >3 years if MR4
Duration of DMR >2 years if MR4.5

and CD86+ plasmacytoid den­dritic cells. Cumulative results sug­ most patients after prior imatinib/IFNA com­bi­na­tion ther­apy and
gest an immu­no­log­i­cal con­trol of persisting CML stem cells in may result in improved MR. Induction of a pro­tein­ase 3-spe­cific
patients with suc­cess­ful TFR.19,33 cyto­toxic T-lym­pho­cyte response by IFNA may con­trib­ute to this
According to pre­clin­i­cal and clin­i­cal stud­ies, IFNA was ­able effect.35 Studies inves­ti­gat­ing the com­bi­na­tion of IFNA with nilo-
to induce cyto­ge­netic remis­sions in a minor­ity of CML patients. tinib or dasatinib are ongo­ing.
Moreover, a small per­cent­age of patients treated with IFNA sus- In a phase 1 study, the com­bi­na­tion of imatinib and ropegin-
tained dura­ble remis­sions after discontinuing ther­apy. The mech­ terferon α2b was shown to be safe and resulted in a DMR in
a­nisms behind this clin­ic­ al obser­va­tion are not well under­stood; patients with chronic-phase CML who did not achieve a DMR with
acti­va­tion of leu­ke­mia-spe­cific immu­nity may be a key fac­tor.34 imatinib alone.36 Clinical tri­als explor­ing the impact of check­
Treatment with IFNA per­mits the dis­con­tin­u­a­tion of imatinib in point inhi­bi­tion include nivolumab as a programmed cell death

110  |  Hematology 2021  |  ASH Education Program


1 pro­tein inhib­i­tor in com­bi­na­tion with dasatinib (NCT02011945) Correspondence
and avelumab as a programmed cell death 1 ligand 1 inhib­i­ Andreas Hochhaus, Klinik für Innere Medizin II, Universitätsklini-
tor com­bined with TKIs (ACTIW trial, NCT02767063; Table 2, kum Jena, Am Klinikum 1, 07740 Jena, Germany; e-mail: andreas​
Figure 1).37 ­.hochhaus@med​­.uni-jena​­.de.
In any attempt to enhance the sta­bil­ity of TFR, the antic­i­pated
effects should be bal­anced against the risk of addi­tional side References
1. Kantarjian HM, Hughes TP, Larson RA, et al. Long-term out­comes with front­
effects caused by the immune acti­va­tion of the block of BCR-
line nilotinib ver­sus imatinib in newly diag­nosed chronic mye­loid leu­ke­mia
ABL1-inde­pen­dent path­ways. in chronic phase: ENESTnd 10-year anal­y­sis. Leukemia. 2021;35(2):440-453.
2. Mahon FX, Réa D, Guilhot J, et al; Intergroupe Français des Leucémies
Myéloïdes Chroniques. Discontinuation of imatinib in patients with chronic
Recommendations
mye­loid leu­kae­mia who have maintained com­plete molec­u­lar remis­sion for
The rec­om­men­da­tions of the Euro­pean LeukemiaNet panel, the at least 2 years: the pro­spec­tive, multicentre Stop Imatinib (STIM) trial.
National Comprehensive Cancer Network, and the French CML Lancet Oncol. 2010;11(11):1029-1035.
Study Group are sum­ma­rized in Table 3.5,38,39 3. Etienne G, Guilhot J, Rea D, et al. Long-term fol­low-up of the French Stop

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/106/1851817/106hochhaus.pdf by guest on 13 December 2021


Certain require­ments are cru­cial: the access to a reli­able, sen­si­ Imatinib (STIM1) study in patients with chronic mye­loid leu­ke­mia. J Clin
Oncol. 2017;35(3):298-305.
tive, stan­dard­ized qRT-PCR with rapid turn­around and a moti­vated
4. Cross NC, White HE, Colomer D, et al. Laboratory rec­om­men­da­tions for
patient who accepts more fre­ quent PCR ana­ ly­
ses. In gen­
eral, scor­ing deep molec­u­lar responses fol­low­ing treat­ment for chronic mye­
chronic-phase patients in first-line ther­apy or in sec­ond-line after loid leu­ke­mia. Leukemia. 2015;29(5):999-1003.
TKI intol­er­ance should be con­sid­ered. Data for patients after TKI 5. Hochhaus A, Baccarani M, Silver RT, et al. Euro­pean LeukemiaNet 2020 rec­
om­men­da­tions for treating chronic mye­loid leu­ke­mia. Leukemia. 2020;34(4):
resis­tance are still lacking. Standardized qRT-PCR requires spec­i­fi­
966-984.
ca­tion of the BCR-ABL1 tran­script type. For patients with atyp­i­cal 6. Saussele S, Richter J, Guilhot J, et al; EURO-SKI Investigators. Discontinuation
BCR-ABL1 tran­scripts, the term “indi­vid­ual molec­u­lar response” of tyro­sine kinase inhib­i­tor ther­apy in chronic mye­loid leu­kae­mia (EURO-SKI):
has been coined. Such patients could start TFR if DMR can be reli­ a prespecified interim anal­y­sis of a pro­spec­tive, multicentre, non-randomised,
ably quan­ti­fied for this spe­cific tran­script (Table 3).40 trial. Lancet Oncol. 2018;19(6):747-757.
7. Rousselot P, Charbonnier A, Cony-Makhoul P, et al. Loss of major molec­
u­lar response as a trig­ ger for restarting tyro­ sine kinase inhib­ i­
tor ther­
apy in patients with chronic-phase chronic mye­log­e­nous leu­ke­mia who
have stopped imatinib after dura­ble unde­tect­able dis­ease. J Clin Oncol.
CLINICAL CASE (Con­t in­u ed) 2014;32(5):424-430.
8. Zhao H, Deininger MW. Declaration of Bcr-Abl1 inde­pen­dence. Leukemia.
Our patient decided to con­tinue dasatinib as a monotherapy. 2020;34(11):2827-2836.
The treat­ ment dura­ tion and the dura­ tion of DMR had been 9. Radich JP, Hochhaus A, Masszi T, et al. Treatment-free remis­sion fol­low­ing
rather short prior to the attempted treat­ment dis­con­tin­u­a­tion. front­line nilotinib in patients with chronic phase chronic mye­loid leu­ke­mia:
5-year update of the ENESTfreedom trial. Leukemia. 2021;35(5):1344-1355.
Current guide­lines rec­om­mend a treat­ment dura­tion of at least
10. Hughes TP, Clementino NCD, Fominykh M, et al. Long-term treat­ment-free
4 years with a 2G-TKI and a DMR dura­tion of at least 2 years in remis­sion in patients with chronic mye­loid leu­ke­mia after sec­ond-line nilo-
MR4.5 (Table 3).5 However, as a low-risk patient with a rapid tinib: ENESTop 5-year update. Leukemia. 2021;35(6):1631-1642.
MR to the first-line treat­ment, she has a good chance of main- 11. Takahashi N, Nishiwaki K, Nakaseko C, et al. Treatment-free remis­sion after
taining DMR and thus discontinuing ther­apy in about 3 addi­ two-year con­sol­i­da­tion ther­apy with nilotinib in patients with chronic mye­
loid leu­ke­mia: STAT2 trial in Japan. Haematologica. 2018;103(11):1835-1842.
tional treat­ment years. The var­i­ant cyto­ge­netic trans­lo­ca­tion
12. Yamaguchi H, Takezako N, Ohashi K, et al. Treatment-free remis­sion after
does not have an impact on prog­no­sis. The pre­dic­tive impact first-line dasatinib treat­ment in patients with chronic mye­loid leu­ke­mia in
of muta­tions out­side BCR-ABL1 is the sub­ject of ongo­ing coop­ the chronic phase: the D-NewS Study of the Kanto CML Study Group. Int J
er­a­tive stud­ies. Hematol. 2020;111(3):401-408.
13. Imagawa J, Tanaka H, Okada M, et al; DADI Trial Group. Discontinuation of
In our clin­i­cal prac­tice, we dis­cuss the con­cept of TFR with
dasatinib in patients with chronic mye­loid leu­kae­mia who have maintained
chronic-phase CML patients prior to starting treat­ment. The dis­ deep molec­u­lar response for lon­ger than 1 year (DADI trial): a multicentre
cus­sion of the goals of CML treat­ment and the selec­tion of first- phase 2 trial. Lancet Haematol. 2015;2(12):e528-e535.
line ther­apy should include the infor­ma­tion that an opti­mal MR 14. Legros L, Nicolini FE, Etienne G, et al; French Intergroup for Chronic Myeloid
min­i­mizes the risk of pro­gres­sion and max­i­mizes the oppor­tu­nity Leukemias. Second tyro­sine kinase inhib­i­tor dis­con­tin­u­a­tion attempt in
patients with chronic mye­loid leu­ke­mia. Cancer. 2017;123(22):4403-4410.
for discontinuing ther­apy. The pros­pect of TFR can serve as an
15. Sharf G, Marin C, Bradley JA, et al. Treatment-free remis­sion in chronic mye­
addi­tional moti­va­tor to opti­mize adher­ence to TKI ther­apy.19 loid leu­ke­mia: the patient per­spec­tive and areas of unmet needs. Leuke-
mia. 2020;34(8):2102-2112.
16. Richter J, Söderlund S, Lübking A, et  al. Musculoskeletal pain in patients
with chronic mye­loid leu­ke­mia after dis­con­tin­u­a­tion of imatinib: a tyro­
Conflict-of-inter­est dis­clo­sure
sine kinase inhib­i­tor with­drawal syn­drome? J Clin Oncol. 2014;32(25):2821-
Andreas Hochhaus: con­sul­tancy: Novartis, Pfizer, BMS, Incyte; re- 2823.
search funding: Novartis, Pfizer, BMS, Incyte. 17. Takahashi N, Tauchi T, Kitamura K, et al; Japan Adult Leukemia Study Group.
Thomas Ernst: hon­o­raria: Novartis, BMS, Incyte; research fund- Deeper molec­u­lar response is a pre­dic­tive fac­tor for treat­ment-free remis­
ing: Novartis. sion after imatinib dis­con­tin­u­a­tion in patients with chronic phase chronic
mye­loid leu­ke­mia: the JALSGSTIM213 study. Int J Hematol. 2018;107(2):185-
193.
Off-label drug use 18. Kim DDH, Novitzky-Basso I, Kim TS, et al. Optimal dura­tion of imatinib treat­
Andreas Hochhaus: experimental design (study proposals): BCL-2, ment/deep molec­u­lar response for treat­ment-free remis­sion after imatinib
dis­con­tin­u­a­tion from a Cana­dian tyro­sine kinase inhib­i­tor dis­con­tin­u­a­tion
BTK, JAK inhibitors are discussed. trial. Br J Haematol. 2021;193(4):779-791.
Thomas Ernst: experimental design (study proposals): BCL-2, 19. Ross DM, Hughes TP. Treatment-free remis­sion in patients with chronic
BTK, JAK inhibitors are discussed. mye­loid leu­kae­mia. Nat Rev Clin Oncol. 2020;17(8):493-503.
Prakash Singh Shekhawat
TKI dis­con­tin­u­a­tion in CML  |  111
20. Ross DM, Branford S, Seymour JF, et al. Safety and effi­cacy of imatinib ces­ 32. Houshmand M, Garello F, Stefania R, et al. Targeting chronic mye­loid leu­
sa­tion for CML patients with sta­ble unde­tect­able min­i­mal resid­ual dis­ease: ke­mia stem/pro­gen­i­tor cells using venetoclax-loaded immunoliposome.
results from the TWISTER study. Blood. 2013;122(4):515-522. Cancers (Basel). 2021;13(6):1311.
21. Ross DM, Branford S, Seymour JF, et al. Patients with chronic mye­loid leu­ke­ 33. Schütz C, Inselmann S, Saussele S, et  al. Expression of the CTLA-4 ligand
mia who main­tain a com­plete molec­u­lar response after stop­ping imatinib CD86 on plasmacytoid den­dritic cells (pDC) pre­dicts risk of dis­ease recur­
treat­ment have evi­dence of per­sis­tent leu­ke­mia by DNA PCR. Leukemia. rence after treat­ment dis­con­tin­u­a­tion in CML. Leukemia. 2017;31(4):829-836.
2010;24(10):1719-1724. 34. Talpaz M, Hehlmann R, Quintás-Cardama A, Mercer J, Cortes J. Re-emer­gence
22. Ross DM, Pagani IS, Shanmuganathan N, et al; Australasian Leukaemia of inter­feron-α in the treat­ ment of chronic mye­ loid leu­ke­ mia. Leukemia.
and Lymphoma Group. Long-term treat­ ment-free remis­ sion of chronic 2013;27(4):803-812.
mye­loid leu­ke­mia with fall­ing lev­els of resid­ual leu­ke­mic cells. Leukemia. 35. Burchert A, Saussele S, Eigendorff E, et al. Interferon alpha 2 main­te­nance
2018;32(12):2572-2579. ther­apy may enable high rates of treat­ment dis­con­tin­ua ­ ­tion in chronic
23. Ilhan O, Narli Ozdemir Z, Dalva K, et  al. Leukemic stem cells shall be mye­loid leu­ke­mia. Leukemia. 2015;29(6):1331-1335.
searched in the bone mar­row before “tyro­sine kinase inhib­i­tor-dis­con­tin­ 36. Heibl S, Buxhofer-Ausch V, Schmidt S, et  al. A phase 1 study to eval­ua ­ te
u­a­tion” in chronic mye­loid leu­ke­mia. Int J Lab Hematol. Published online the fea­si­bil­ity and effi­cacy of the addi­tion of ropeginterferon alpha-2b to
9 April 2021. imatinib treat­ment in patients with chronic phase chronic mye­loid leu­ke­
24. Machova Polakova K, Zizkova H, Zuna J, et al. Analysis of chronic mye­loid mia (CML) not achiev­ing a deep molec­ul­ar response (molec­u­lar remis­sion

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/106/1851817/106hochhaus.pdf by guest on 13 December 2021


leu­kae­mia dur­ing deep molec­u­lar response by geno­mic PCR: a traf­fic light 4.5)-AGMT_CML 1. Hematol Oncol. 2020;38(5):792-798.
strat­i­fi­ca­tion model with impact on treat­ment-free remis­sion. Leukemia. 37. Hsieh YC, Kirschner K, Copland M. Improving out­comes in chronic mye­
2020;34(8):2113-2124. loid leu­ke­mia through harnessing the immu­no­log­i­cal land­scape. Leukemia.
25. Houshmand M, Simonetti G, Circosta P, et  al. Chronic mye­loid leu­ke­mia 2021;35(5):1229-1242.
stem cells. Leukemia. 2019;33(7):1543-1556. 38. National Comprehensive Cancer Network. Chronic mye­ loid leu­
ke­
mia.
26. Bocchia M, Sicuranza A, Abruzzese E, et al. Residual periph­eral blood CD26+ NCCN Clinical Practice Guidelines in Oncology Version 3.2021. J Natl
leu­ke­mic stem cells in chronic mye­loid leu­ke­mia patients dur­ing TKI ther­apy Compr Canc Netw. 2020;18(10):1385-1415.
and dur­ing treat­ment-free remis­sion. Front Oncol. 2018;8(30 May):194. 39. Rea D, Ame S, Berger M, et al; French Chronic Myeloid Leukemia Study
27. Kinstrie R, Horne GA, Morrison H, et  al. CD93 is expressed on chronic Group. Discontinuation of tyro­ sine kinase inhib­ it­ors in chronic mye­ loid
mye­loid leu­ke­mia stem cells and identifies a qui­es­cent pop­u­la­tion which leu­ke­mia: rec­om­men­da­tions for clin­i­cal prac­tice from the French Chronic
per­sists after tyro­sine kinase inhib­i­tor ther­apy. Leukemia. 2020;34(6):1613- Myeloid Leukemia Study Group. Cancer. 2018;124(14):2956-2963.
1625. 40. Schäfer V, White HE, Gerrard G, Möbius S, Saussele S, Franke GN, et  al.
28. Pagani IS, Dang P, Saunders VA, et al. Lineage of mea­sur­able resid­ual dis­ Assessment of indi­vid­ual molec­u­lar response in chronic mye­loid leu­ke­mia
ease in patients with chronic mye­loid leu­ke­mia in treat­ment-free remis­ patients with atyp­i­cal BCR-ABL1 fusion tran­scripts: rec­om­men­da­tions by
sion. Leukemia. 2020;34(4):1052-1061. the EUTOS coop­er­a­tive net­work. J Cancer Res Clin Oncol. 2021;147(3):3081-
29. Kuepper MK, Bütow M, Herrmann O, et  al. Stem cell per­sis­tence in CML 3089.
is medi­ated by extrin­si­cally acti­vated JAK1-STAT3 sig­nal­ing. Leukemia.
2019;33(8):1964-1977.
30. Parting O, Langer S, Kuepper MK, et al. Therapeutic inhi­bi­tion of FcγRIIb
sig­nal­ing tar­gets leu­ke­mic stem cells in chronic mye­loid leu­ke­mia. Leuke-
mia. 2020;34(10):2635-2647.
31. Carter BZ, Mak PY, Mu H, et al. Combined targeting of BCL-2 and BCR-ABL
tyro­sine kinase erad­i­cates chronic mye­loid leu­ke­mia stem cells. Sci Transl © 2021 by The Amer­i­can Society of Hematology
Med. 2016;8(355):355ra117. DOI 10.1182/hema­tol­ogy.2021000238

112  |  Hematology 2021  |  ASH Education Program


CML: SUCCESS BREEDS MORE SUCCESS

Lifelong TKI therapy: how to manage


cardiovascular and other risks

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/113/1851432/113mauro.pdf by guest on 13 December 2021


Michael J. Mauro
Myeloproliferative Neoplasms Program, Leukemia Service, Memorial Sloan Kettering Cancer Center, New York, NY

Beginning with imatinib and now spanning 6 oral, highly active, and mostly safe agents, the development of specific tar-
geted therapy for patients with chronic myeloid leukemia (CML) has created a new world featuring chronic maintenance
chemotherapy for all treated as such, treatment-free remission, and functional cure after prolonged deep remission in a
subset. As a result comes a necessary shift in focus from acute to chronic toxicity, increasing attention to patient comor-
bidities, and critical thinking around specific adverse events such as metabolic, cardiovascular, and cardiopulmonary
effects, which vary from agent to agent. This review aims to pull together the state of the art of managing the “C” in
CML—a chronic myeloproliferative neoplasm treated at present over many years with oral BCR-ABL-targeted agents in a
population whose overall health can be complex and potentially affected by disease and therapy—and determine how
we can better manage a highly treatable and increasingly curable cancer.

LEARNING OBJECTIVES
• Understand the risks of a CML diagnosis and the importance of comorbidity assessment, especially cardiovascular
• Understand the specifc risks of CML tyrosine kinase inhibitors (approved and forthcoming) and how to screen for
and mitigate adverse events

Introduction events are rare to absent.2 The duration of treatment and


As has been stated often, the treatment of chronic mye- duration of deep remission remain strong predictors of TFR
loid leukemia (CML) was and remains revolutionized, with success,3 while factors such as minimal residual disease
lower-risk small-molecule-specifc targeted therapy with stability,4 immune factors,5 and myeloid mutations beyond
oral ABL tyrosine kinase inhibitors (TKIs) as the overwhelm- BCR-ABL1 continue to be investigated as to their role.6
ing mainstay approach. Palliative cytoreductive agents Increasingly, the number of patients potentially eligi-
(hydroxyurea, busulfan) are rarely used except for initial ble or who have engaged in cessation of TKI therapy (TFR)
temporization of blood counts; interferon-based therapy grows. A number of unmet needs remain for the optimal
remains active in other myeloproliferative neoplasms but treatment of CML, ranging from maximizing initial response
is only used rarely and in special circumstances in which while minimizing toxicity and progression risk, to safer
TKI toxicity may be prohibitive (including pregnancy). and more effcient salvage approaches for heavily treated
Through advances in conditioning, graft-versus-host pre- multi-TKI resistant or multi-TKI resistant/intolerant or even
vention, posttransplant TKI integration, and alternative multi-TKI intolerant scenarios, to advanced phase or trans-
donor sources, allogeneic stem cell transplant continues to formed disease; added to this list now is improving the
be optimized, yet its use greatly diminished.1 Allografting outcomes of TFR—namely, by increasing the fraction of
was previously viewed as the only “cure” for CML. Pres- successful cessation. Many, or perhaps most, view the cur-
ently, patients treated with TKI therapy may avail them- rent goal of therapy to be successful treatment cessation
selves of the possibility of a “functional cure”—suffcient subsequent to sustained deep remission—what has been
treatment with TKI therapy into a sustained deep remission termed as a functional cure. While this path is streamlined
followed by carefully observed TKI cessation—treatment- and being made possible for a higher fraction of patients,
free remission (TFR). Long-term results from the earliest assuming the potential for lifelong TKI therapy remains a
study of TFR demonstrate, importantly, that durable remis- pragmatic approach and perhaps the best way to balance
sion without TKI therapy can be sustained for many years, the risks and benefts of treatment strategies for those liv-
retreatment for failed TFR attempts is effective, and late ing with CML.
Prakash Singh Shekhawat
Risks with lifelong TKI therapy in CML | 113
Survival in CML and bone mar­row stud­ies sup­port chronic-phase dis­ease mor­
The long-term fol­low-up of the piv­otal trial in CML, the IRIS pho­log­i­cally, with cyto­ge­net­ics reveal­ing 100% sole Philadel-
(International Randomized Study of Interferon and STI571) trial, phia chro­mo­some trans­lo­ca­tion in 20 meta­phases and base­line
dem­on­strated the vastly improved out­look for patients treated BCR-ABL tran­scripts of the e14a2 var­i­ant of p210 fusion mea­sur­
with TKI ther­apy com­pared to the prior stan­dard, inter­feron- ing 90% on the International Scale. She wishes to hear from you
based ther­apy,7 heralding the “TKI era.” A com­pre­hen­sive anal­ on risks of dis­ease and med­i­ca­tion choices and under­stands
y­sis of out­comes of cur­rent CML patients’ sur­vival rel­a­tive to that treat­ment “may not need to be for­ever.”
age-matched peers, irrespective of decade of age, con­cluded
no sig­nif­i­cant dif­fer­ence—lead­ing to the wel­come con­clu­sion of
a “nor­mal life span” in the TKI era.8 While the inci­dence of CML Risks at pre­sen­ta­tion with a diag­no­sis of CML
remains steady—Euro­pean CML reg­is­try data cite an annual The diag­no­sis of CML encompasses a num­ber of signs and symp­
inci­dence rate of 0.7 to 1.0/100 000, a median age at diag­no­ toms, includ­ing fatigue, weight loss, loss of energy, decreased
sis of 57 to 60 years, and a male/female ratio of 1.2 to 1.7—the exer­cise tol­er­ance, low-grade fevers, sweats resulting from hyper­
me­tab­o­lism, bone pain, and early sati­ety resulting from encroach­

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/113/1851432/113mauro.pdf by guest on 13 December 2021


prev­a­lence of CML steadily increases world­wide due to mark­
edly improved sur­vival.9 Estimates in the US pro­ject increase ment by an enlarged spleen on the stom­ach, as well as left upper
from a pre–TKI era prev­a­lence of roughly 25 000 indi­vid­u­als quad­rant full­ness and pain. Infarction of the spleen can be observed,
liv­ing with CML to 200 000 plus such indi­vid­u­als over the next espe­cially with marked enlarge­ment. Blood count changes include
3 decades. Such remark­able suc­cess focuses atten­tion on many leu­ko­cy­to­sis and var­i­able plate­let count ele­va­tion or, with advanc­
fronts, includ­ing global dis­par­ity, addressed by groups such as ing-phase dis­ease, reduc­tion; ane­mia is gen­er­ally pro­por­tional to
the International CML Foundation, regard­ ing edu­ca­
tion and dis­ease bur­den. Bleeding, pete­chiae, and ecchy­mo­ses are more
treat­ment, and the Max Foundation, regard­ing patient advo­ likely with advanced dis­ease and throm­bo­cy­to­pe­nia.
cacy and med­i­ca­tion access; cost of ther­apy, given the mul­ The pre­sen­ta­tion of CML with marked thrombocytosis (plate­
ti­plier effect of patient and patient-years on TKI ther­apy; and lets >1 mil­lion) may occur, and vas­cu­lar injury and throm­botic
last and per­haps most impor­tantly, increased atten­tion to the events appear more com­mon in such cases, as well as a female
qual­ity of life and long-term out­come of CML “sur­vi­vors.” pre­dom­i­nance and youn­ger age.12 In chil­dren, throm­bo­sis may be
less likely, but as is seen in other mye­lo­pro­lif­er­a­tive neo­plasms,
New fron­tier: CML sur­vi­vor­ship acquired von Willebrand syn­drome and mild clin­i­cal bleed­ing
A nat­u­ral exten­sion of highly effec­tive ther­apy, the high frac­tion may develop due to the bind­ing of von Willebrand fac­tor multi-
of patients ush­ered into deep and sta­ble remis­sions, and the mers, espe­cially large multimers, to plate­lets.13 Not sur­pris­ingly,
prom­ise of a grow­ing poten­tial frac­tion of patients deemed func­ a grow­ing num­ber of case reports are emerg­ing iden­ti­fy­ing the
tion­ally cured begs the ques­tion of “CML sur­vi­vor­ship.” Distinct coex­is­tence of mye­lo­pro­lif­er­a­tive “driver” muta­tions, such as
from the past in which CML sur­vi­vors would be a mix of those fol- JAK2 v617F and calreticulin along with BCR-ABL fusion, as deep
lowed after allografting in the pro­to­typ­i­cal posttransplant sur­vi­ sequenc­ing becomes more uti­lized, suggesting that bet­ter char­
vor­ship clinic and a rare long-term responder to inter­feron-based ac­ter­iza­tion of mye­loid muta­tions in CML may be of clin­i­cal use.
ther­apy, the pres­ent era calls for the orga­nized care of those Often iso­lated to set­tings where pre­sen­ta­tion and diag­no­sis
suc­cess­fully treated with TKIs, whether in TFR or not. Organized may be delayed, com­pli­ca­tions related to leukostasis in tar­get
care of the leu­ke­mia itself, of course, remains high pri­or­ity; for organs at risk for vas­cu­lar injury include priapism and ret­i­nop­a­
those remaining on TKI ther­apy, reg­u­lar blood-based mon­i­tor­ing thy. Leukapheresis can be used for clin­i­cal signs of injury due to
with highly sen­si­tive quan­ti­ta­tive poly­mer­ase chain reac­tion for leukostasis and remains a ther­a­peu­tic “bridg­ing” strat­egy dur­ing
the BCR-ABL1 fusion con­tin­ues indef­i­nitely per cur­rent rec­om­ CML and preg­nancy, when cytoreductive ther­apy and defin­i­tive
men­da­tions.10 For those patients who have under­gone suc­cess­ful ther­apy with TKIs may pose exces­sive fetal risk.14
TFR, guid­ance regard­ing opti­mal ini­tial mon­i­tor­ing con­tin­ues to
be scru­ti­nized; sub­se­quent ques­tions will include the more dif­fi­ Risks asso­ci­ated with the first-line TKIs imatinib, nilotinib,
cult ques­tion of taper­ing of long-term mon­i­tor­ing after suc­cess­ dasatinib, and bosutinib
ful TFR.11 Patients with CML thus con­tinue to have long-stand­ing The ini­ tial treat­
ment of CML increas­ ingly is the direct use of
rela­tion­ships with their CML phy­si­cians, and the devel­op­ment of TKI ther­apy in order to promptly induce hema­to­logic con­trol/
a tai­lored approach to the “life­long” TKI algo­rithm, to tackle the com­plete hema­to­logic response; cytoreduction with hydroxy­
“end” of CML treat­ment for those in TFR, and to delin­eate what urea prior to TKI start, if used, may aug­ment the myelosuppres-
comes next from the per­spec­tive of rel­e­vant fol­low-up and risk sion observed with sub­se­quent TKI ther­apy. Myelosuppression
man­age­ment is a next step ripe for devel­op­ment. remains a ubiq­ui­tous ini­tial effect com­men­su­rate with TKI start
and is some­what var­i­able among the avail­­able ABL kinase inhib­i­
tors; in com­par­a­tive tri­als of first-line ther­apy with imatinib, nilo-
tinib, dasatinib, and bosutinib, dasatinib was asso­ci­ated with the
CLINICAL CASE greatest rates of grade 3 or 4 neutropenia and throm­bo­cy­to­pe­
A 55-year-old woman is referred in (via telehealth) for a recent nia. Myelosuppression may be best viewed as a mix of dis­ease
diag­no­sis of CML. Records indi­cate lower-risk dis­ease by the and ther­apy-related effects given its (early) kinet­ics and pro­
Sokal and EUTOS Long Term Survival score, as her white blood pen­sity to resolve; a sub­set of patients, often with higher-risk
cell count is 32 000 with nor­mal hemo­glo­bin and plate­lets, no fea­tures, may face severe and per­sis­tent myelosuppression pre­
cir­cu­lat­ing blasts, and a mod­est left-shifted dif­fer­en­tial with clud­ing TKI deliv­ery and response to ther­apy, pos­ing a spe­cific
3% each baso­phils and eosin­o­phils. She has no spleno­meg­aly, intol­er­ance and chal­lenge.

114  |  Hematology 2021  |  ASH Education Program


Other adverse events com­ mon to the entire class of ABL and the spec­ter of clonal hema­to­poi­e­sis has been rightly raised
inhib­i­tors used at diag­no­sis are gen­er­ally revers­ible and pose lit­ as a poten­tial con­trib­ut­ing fac­tor in throm­botic events in CML.34
tle known long-term risk. Examples of such lab­o­ra­tory find­ings Of note, the pres­ence and poten­tial impact of other mye­loid
include trans­am­i­nase ele­va­tion, lipase ele­va­tion (more often bio­ muta­tions aside from BCR-ABL1 fusion on sev­eral fronts, includ­
chem­i­cal and lower with pan­creas inflam­ma­tion or pan­cre­a­ti­ ing pri­mary response, the abil­ity to achieve suc­cess­ful TFR in oth­
tis), hyperbilirubinemia (observed more with nilotinib and often er­wise “eli­gi­ble” cases, and adverse events, espe­cially vas­cu­lar,
unmasking Gilbert’s syn­drome), and oth­ers.15 Adverse events more are just begin­ning to be unveiled as broader, deeper sequenc­ing
closely linked to imatinib include edema (periorbital and periph­ is performed in CML.6
eral), mus­cle cramps, mus­cu­lo­skel­e­tal pain, and diar­rhea; those Pulmonary com­pli­ca­tions of TKI ther­apy are also of spe­cific
more closely linked to nilotinib include rash, pru­ri­tus, head­ache, inter­est, warranting aware­ness, early inter­ven­tion, and con­sid­er­
nasopharyngitis, constipation, abdom­i­nal pain, vomiting, pyrexia, ation of spe­cific sur­veil­lance. Most nota­bly, pleu­ral and peri­car­dial
upper uri­ nary tract infec­
tion, back pain, cough, and asthe­ nia; fluid accu­mu­la­tion as well as pul­mo­nary arte­rial hyper­ten­sion have
those more closely related to dasatinib include, as pre­ vi­
ously been observed, most often with dasatinib ther­apy. Initial anal­y­sis

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/113/1851432/113mauro.pdf by guest on 13 December 2021


men­tioned, myelosuppression as well as fluid reten­tion, includ­ing suggested that car­diac dis­ease and sys­temic arte­rial hyper­ten­sion
pleu­ral and peri­car­dial effu­sions and head­ache; and those more were predisposing fac­tors to develop pleu­ral effu­sions on dasat-
closely related to bosutinib include diar­rhea and increases in ala­ inib35; a larger meta-anal­y­sis with lon­ger fol­low-up suggested, in
nine and aspar­tate ami­no­trans­fer­ase. Common, more fre­quent mul­ti­var­i­ate anal­y­sis, that age was an inde­pen­dent pre­dic­tor.36
adverse events across TKIs include fatigue myal­gias and gas­tro­ Distinct from many other effects asso­ci­ated with TKIs, the risk
in­tes­ti­nal symp­toms such as nau­sea and vomiting. Given a broad of devel­op­ing pleu­ral effu­sions may be spread over many years’
inves­ti­ga­tion into the opti­mal ther­a­peu­tic dose, imatinib plasma time or the entirety of treat­ment; a 7-year fol­low-up of sec­ond-line
trough lev­els were noted to cor­re­late with effi­cacy and the cor­ dasatinib noted a cumu­la­tive inci­dence of 28% to 35% (depending
rec­tion of sub­op­ti­mal response16; how­ever, drug lev­els did not cor­ on TKI dose) and a 5% inci­dence of new events in year 7.37 The
re­late with adverse events. Longer fol­low-up of imatinib-treated DASISION study of front­line dasatinib noted a steady 8% per year
patients in the land­mark IRIS trial con­cluded that adverse events inci­dence and a cumu­la­tive inci­dence of 38% at 5 years and also
were not typ­i­cally observed after the first year of ther­apy.7 cited age as a pre­dic­tor.38 Both dose and sched­ule influ­enced the
Particular aware­ ness and pre­ pared­ ness are warranted for like­li­hood of pleu­ral effu­sions in sec­ond-line stud­ies, con­trib­ut­ing
adverse events of spe­cific inter­est (asso­ci­ated with greater mor­ to the change to opti­mized once-daily reduced dos­ing of dasatinib
bid­ity and poten­tial long-term impact) for TKIs used at diag­no­sis. in CML. Continued inquiry into opti­miz­ing the risk/ben­e­fit bal­ance
Imatinib is asso­ci­ated with hypophosphatemia; fur­ther inves­ti­ with dasatinib is evidenced by a com­pel­ling sin­gle-cen­ter, sin­gle-
ga­tion dem­on­strated a poten­tial link to altered bone min­er­al­i­ arm study of 50 mg dos­ing for newly diag­nosed chronic-phase CML
za­tion.17 Renal injury, manifesting as a decline of the glo­mer­u­lar in which pleu­ral effu­sions were observed in only 6% of patients
fil­tra­tion rate, was noted with imatinib ther­apy, with reduc­tions after a median fol­low-up of 24 months.39
steadily over 4 years’ time before pla­teau and iso­lated to those Specific man­age­ment rec­om­men­da­tions in the case of dasat-
with no chronic kid­ney dis­ease prior18; in a sep­a­rate study, nearly inib pleu­ral effu­sions have been devel­oped40; how­ever, the exact
iden­ti­cal changes were observed for bosutinib ther­apy.19 Inter- mech­a­nisms of action and the best mit­i­ga­tion strat­egy remain
estingly, TKI dose did not cor­re­late with injury and revers­ibil­ity elu­sive. Given its pro­file beyond ABL kinase inhi­ bi­tion, as an
was appar­ent. Nilotinib is asso­ci­ated with impaired fasting glu­ inhib­i­tor of the SRC fam­ily of kinases and lym­pho­cyte-spe­cific
cose lev­els,20 whereas imatinib has been suggested to improve pro­tein kinase, a related phe­nom­e­non observed with dasati-
them21; this effect with nilotinib is reported to be revers­ible, to nib ther­apy includes periph­eral blood lym­pho­cy­to­sis,41 with a
be linked to increased body mass index, and not to trig­ger type large gran­u­lar lym­pho­cyte mor­phol­ogy that is directly cor­rel­
2 dia­be­tes.22 Lipid pro­files may show sim­i­lar changes, with ima- a­tive to dasatinib dos­ing.42 While poten­tially asso­ci­ated with
tinib improv­ing lev­els and nilotinib, as early as 3 months into other inflam­ma­tory phe­nom­ena, includ­ing coli­tis and fol­lic­u­lar
treat­ment, trig­ger­ing increases in cho­les­terol (both low-den­sity hyper­pla­sia/lymph node enlarge­ment,43 periph­eral blood lym­
and high-den­sity lipo­pro­teins and in turn total cho­les­terol); any pho­cy­to­sis is observed in approx­i­ma­tely one-third of treated
effect of dasatinib appears more mod­est.23-25 patients and is asso­ci­ated with higher response rates, lon­ger
In con­trast to imatinib, vas­cu­lar adverse events have been response dura­tions, and increased over­all sur­vival, support-
asso­ci­ated with newer-gen­er­a­tion TKIs, most prom­i­nently with ing a poten­tial immu­no­mod­u­la­tory role.44 Additionally, pul­mo­
the later-line ther­apy agent ponatinib as well as with the sec­ond- nary arte­rial hyper­ten­sion has been observed, thank­fully rarely
gen­er­a­tion TKIs dasatinib and nilotinib,26,27 while min­i­mal asso­ci­ (<1%).45 It is most often asso­ci­ated with dasatinib (and occurs
a­tion has been observed with bosutinib.28 Long-term fol­low-up less often but has been noted with bosutinib and ponatinib) yet
of the ENESTnd trial com­pared nilotinib at var­ied doses vs imati- con­founded by how infre­quently right heart cath­e­ter­i­za­tion is
nib for newly diag­nosed CML. At the 10-year median fol­low-up, performed to inves­ti­gate symp­toms prop­erly. Carefully vet­ted
rates were 16.5% for nilotinib at 300 mg twice daily and 23.5% for cases (diag­nosed with right heart cath­e­ter­i­za­tion) warranted TKI
400 mg twice daily vs 3.6% for imatinib, includ­ing in Framingham dis­con­tin­u­a­tion, and while the major­ity were revers­ible, approx­i­
car­dio­vas­cu­lar-assessed low-risk patients.29 Mechanisms remain ma­tely one-third of cases dem­on­strated per­sis­tence,46 val­i­dat­ing
under inves­ti­ga­tion; pos­tu­lated causes include endo­the­lial cell the need for closer atten­tion to this TKI risk.
effects,30 proinflammatory and pro-oxi­ da­tive stress fac­tors,31
and lipid, coag­u­la­tion, and plate­let effects (some par­a­dox­i­cal),32 Risks asso­ci­ated with ponatinib
among oth­ers. Sequencing stud­ies have suggested dif­fer­en­tial Considered inde­pen­dently given its more dis­tinct risk pro­file,
expres­sion pat­terns in tar­get tis­sues (such as endo­the­lial cells),33 ponatinib offers an excel­lent abil­ity to sal­vage poor response in
Prakash Singh Shekhawat
Risks with life­long TKI ther­apy in CML  |  115
chronic-phase CML, shows effi­cacy in the face of select resis­ 20%, while the EAC ver­i­fied 16%. With these data and a sup­
tance (ABL kinase domain muta­tion thre­o­nine for iso­leu­cine at ple­men­tal new drug appli­ca­tion, the FDA approved an updated
posi­ tion 315-T315I), and has a prob­ a­
bly often mis­ un­der­ stood indi­ca­tion for ponatinib in the US—namely, patients with resis­
adverse event pro­file. Ponatinib expe­ri­ence in a phase 1 study tance or intol­er­ance to at least 2 prior kinase inhib­i­tors.
noted prominent adverse effects, includ­ing rash and acnei-
form der­ma­ti­tis, arthral­gias/fatigue, and a clear risk of pan­creas On the hori­zon: asciminib
inflam­ma­tion greater than other TKIs with lipase ele­va­tion, hyper- Asciminib is a novel ABL kinase inhib­i­tor, a so-called STAMP (spe­
triglyceridemia, and pan­cre­a­ti­tis at equally ele­vated rates47; a cif­i­cally targeting the ABL myristoyl pocket) inhib­i­tor. Function-
sig­nal of vas­cu­lar tox­ic­ity had not yet emerged. Commensurate ing as an allo­ste­ric inhib­i­tor of the deregulated BCR-ABL kinase
with pub­li­ca­tion of the PACE trial in late 2013,48 the US Food and by targeting the myristoyl pocket in con­ trast to the active
Drug Administration (FDA) requested and the man­uf­ac­turer (at site (aden­o­sine tri­phos­phate-bind­ing) mech­a­nism of all­ other
that time, Ariad Pharmaceuticals) agreed to sus­pend the mar­ licensed ABL kinase inhib­i­tors, asciminib holds the prom­ise of
ket­ing of ponatinib. Data from PACE showed “sig­nif­i­cant anti­leu­ dis­tinct sin­gle-agent activ­ity in Philadelphia-pos­i­tive leu­ke­mias

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/113/1851432/113mauro.pdf by guest on 13 December 2021


ke­mic activ­ity across categories of dis­ease stage and muta­tion as well as the poten­tial for ratio­nal com­bi­na­tion ther­apy (aden­
sta­tus” but reported that seri­ ous arte­ rial throm­botic events o­sine tri­phos­phate-bind­ing + myristoyl-bind­ing ABL inhib­i­tor
(includ­ing cere­bro­vas­cu­lar, car­dio­vas­cu­lar, and periph­eral vas­cu­ ther­apy). Initial phase 1 study reports in mainly chronic-phase
lar events) were ris­ing, ini­tially num­ber­ing 8.9% (2.9% treat­ment patients with multi-TKI resis­tance/intol­er­ance indi­cated con­
related) dur­ing the study and then going up to 11.8%. The inci­ vinc­ing effi­cacy and min­i­mal resis­tance via novel muta­tions.53
dence of all­arte­rial throm­botic events, seri­ous or not, was 17.1%. A sub­se­quent ran­dom­ized study of asciminib in com­par­i­son to
While the rate was not increas­ing over time, the accu­mu­la­tion bosutinib for chronic-phase CML patients with 2 prior ther­a­
with addi­tional fol­low-up (>24 mo) led to the mar­ket­ing sus­pen­ pies and no resis­tance muta­tions to bosutinib or T315I muta­tion
sion and halt­ing of ponatinib front­line stud­ies. As men­tioned rel­ con­firmed asciminib’s supe­ri­or­ity regard­ing major molec­u­
a­tive to the sec­ond-gen­er­a­tion TKIs nilotinib and bosutinib, the lar response at 6 months, the pri­mary end point of the study,
mech­an ­ isms and basis for such events remain unclear, with sev­ as well as other end points, includ­ing com­plete cyto­ge­netic
eral hypoth­es­es. A focus on ponatinib in par­tic­ul­ar is ongo­ing response.54 Given its nov­elty as a STAMP inhib­i­tor, its potency
and includes such con­cepts as a throm­botic throm­bo­cy­to­pe­nic and spec­i­fic­ity for the myristoyl pocket of ABL1, and its broad
pur­pura-like microangiopathic mech­a­nism.49 inac­tiv­ity against other kinases, includ­ing SRC kinase,55 the tox­
Based on addi­tional data from the phase 1 study and the afore­ ic­ity pro­file of asciminib to date has also been prom­is­ing. The
men­tioned emerg­ing data, the label for ponatinib then included phase 1 study lim­it­ing tox­ic­ity was lim­ited to lipase ele­va­tion
a vas­cu­lar adverse event (arte­rial and venous) rate of 27% and with a sin­gle case of pan­cre­a­ti­tis, arthral­gia/myal­gia, abdom­
a fairly nar­row indi­ca­tion (T315I muta­tion-bear­ing patients and i­nal pain, and bron­cho­spasm and a sin­gle case of acute cor­
those in whom “no other TKI is indi­cated”), and a risk eval­u­a­tion o­nary syn­drome; lipase ele­va­tion/pan­cre­a­ti­tis remains a class
and mit­i­ga­tion strat­egy was requested. In con­trast, ponatinib effect prominent with nilotinib and ponatinib and now asso­ci­
retained Euro­pean Medicines Agency mar­ket­ing autho­ri­za­tion ated with asciminib. In the phase 3 ASCEMBL study com­par­ing
as well as its indi­ca­tion for patients car­ry­ing the T315I muta­tion asciminib to bosutinib, mark­edly lower-gas­tro­in­tes­ti­nal adverse
and those resis­tant or intol­er­ant to dasatinib or nilotinib and for events were observed with asciminib com­pared to bosutinib, as
whom imatinib was not clin­ic ­ ally appro­pri­ate and con­cluded one might expect; how­ever, as with all­TKIs, closer atten­tion is
a some­what lower vas­cu­lar event rate. Data and fur­ther guid­ often paid to vas­cu­lar adverse events, and 2 deaths occurred in
ance for the opti­mal use of ponatinib have con­tin­ued to evolve; the asciminib arm fall­ing into this cat­e­gory (ische­mic stroke and
as such the 2020 updated guide­lines issued by the Euro­pean arte­rial embolism per the report). Dissecting out any asso­ci­a­
LeukemiaNet rec­om­mend ponatinib in patients with resis­tance tion and risk will take more time, and rec­og­ni­tion of the heavily
to a sec­ond-gen­er­a­tion TKI (dasatinib, nilotinib, and bosutinib), pretreated nature of the patient pop­u­la­tion in the ASCEMBL trial
even with­out spe­cific muta­tions, unless its use is pre­cluded by is warranted. At the pres­ent time, asciminib appears to have
the pres­ence of car­dio­vas­cu­lar risk fac­tors.50 Very recent data a nar­row spec­trum of tox­ic­ity, and no con­clu­sive evi­dence of a
from the dose-find­ing OPTIC study,51 enroll­ing patients with CML vas­cu­lar/car­dio­vas­cu­lar sig­nal exists as of yet, nota­bly after a
resis­tant/intol­er­ant to ≥2 TKIs or car­ry­ing the T315I muta­tion to very long-last­ing (>7 years) phase 1 study. It offers great prom­
receive 45, 30, or 15 mg ini­tially followed by 15 mg main­te­nance ise in resis­tant/intol­er­ant patients, will inspire ongo­ing explo­ra­
upon achiev­ing ≤1% BCR-ABL lev­els, sug­gest an opti­mal ben­e­fit/ tion into com­bi­na­tion approaches, and rep­re­sents a sig­nif­i­cant
risk pro­file for the 45 mg starting dose. advance­ment in the field.
What lies at the heart of the mat­ ter—no pun intended—
regard­ing the true risk of vas­cu­lar adverse events is per­haps how
they are quan­ti­fied and attrib­uted. A recent study deployed an
inde­pen­dent end point adju­di­ca­tion com­mit­tee (EAC) includ­
ing car­di­­ol­ogy, hema­tol­ogy, and neu­rol­ogy input to reexamine CLINICAL CASE (Con­tin­ued)
the vas­cu­lar adverse events from the PACE study of ponatinib. After thor­ough dis­cus­sion about risk strat­i­fi­ca­tion and the rel­
It noted that while AOEs iden­ti­fied by the Medical Dictionary a­tive mer­its and risks of avail­­able TKI ther­ap
­ ies spe­cific to her
for Regulatory Activities pre­ferred terms num­bered 25%, the case, you find your­ self not­
ing the safety and likely suc­ cess
blinded EAC, using Amer­i­can College of Cardiology/Amer­i­can of imatinib-based ther­ apy contrasted with the poten­ tial for
Heart Association def­i­ni­tions, noted 17%.52 Serious AOEs by the improved early and deep response with any of the sec­ond-
Medical Dictionary for Regulatory Activities terms num­ bered gen­er­a­tion TKI options and presenting either option as rea­son­able.

116  |  Hematology 2021  |  ASH Education Program


You pro­ceed to take a more detailed back­ground med­i­cal his­ tumor), and AIDS—was exam­ined in the set­ting of imatinib-based
tory and find her to be free of any active car­dio­vas­cu­lar or met­ ther­apy in the Ger­man CML IV study and in the broader set­ting
a­bolic dis­or­ders such as heart dis­ease, hyper­ten­sion, dia­be­tes, of imatinib, nilotinib, and dasatinib.61,62 Both stud­ies found that
renal dis­ease, hyper­lip­id­emia, or other chronic con­di­tions. Her the Charlson Comorbidity Index was con­sis­tently the stron­gest
weight is near ideal, and she does not smoke; how­ever, she pre­dic­tor of sur­vival in CML, includ­ing age-inde­pen­dent anal­y­sis,
can­not offer a fam­ily his­tory as she was adopted as an infant. and in fact did not affect TKI treat­ment suc­cess, lead­ing to the
When asked about recent screen­ing for the above con­di­tions, con­clu­sion that sur­vival of patients with CML is deter­mined more
she can­not recall her last pri­mary care appoint­ment as she is by comorbidities than by CML itself.
“very healthy and has not needed to go to the doc­tor, espe­
cially dur­ing the pan­demic” and has never seen a car­di­­ol­o­gist The who, what, and when of risk assess­ment
or had any car­dio­vas­cu­lar stud­ies. and mit­i­ga­tion for the patient with CML
One of my most mem­o­ra­ble quotes from med­i­cal school, from
a cherished leu­ke­mia attend­ing, was sim­ple and pow­er­ful: “You

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/113/1851432/113mauro.pdf by guest on 13 December 2021


Assessing comorbidity in the CML patient pop­u­la­tion can’t be a good hema­tol­o­gist/oncol­o­gist unless you are a good
Simply stated, to man­age the car­dio­vas­cu­lar and other risks in inter­nist first.” A leu­ke­mia diag­no­sis, even one with such hope,
patients with CML, one must embrace the “whole patient”—the mul­ti­plic­ity of effec­tive options, and guid­ance based on response
full health his­ tory, active and qui­ es­
cent comorbid con­ di­
tions, mile­stones as CML has, often leaves other health issues and fol­low-
vulnerabilities as best one can judge—plac­ing gen­eral med­i­cal up in a blur as leu­ke­mia care comes into focus. Leukemia care
con­sid­er­ations in clear focus in antic­i­pa­tion of leu­ke­mia care. The engulfs the pres­ent, with fre­quent blood work and check­ups; given
aver­age age at CML diag­no­sis varies widely across the globe,56 the inten­sity and nuances of care, oth­ers feel it best to step aside
rang­ing from medi­ans in the 40s in Asia and Africa, to the 60s in and stay at a dis­tance for fear of neg­a­tively affect­ing can­cer treat­
North America, and to >70 in the Oceanic region (Australia, etc). ment suc­cess. Rather than a point of sep­a­ra­tion, in CML, the time
The local/regional spe­cif­ics of poten­tial clin­i­cal chal­lenges vary of diag­no­sis and there­af­ter need to be the oppo­site—a point of
and may not be well char­ac­ter­ized—eg, in a large ter­tiary refer­ alli­ance between leu­ke­mia spe­cial­ists, pri­mary care/inter­nal med­
ral cen­ter in Soweto, Johannesburg, South Africa, 7% of all­CML i­cine, and oth­ers as needed, par­tic­u­larly in car­dio­vas­cu­lar med­i­
patients carry a diag­no­sis of both HIV and CML.57 Access to ther­apy cine. Continued fol­low-up with pri­mary care/inter­nal med­i­cine is
is not equi­ta­ble glob­ally; orga­ni­za­tions such as the non­profit MAX essen­tial for patients with CML. With high rates of remis­sion and
Foundation (www​­.themaxfoundation​­.org) have partnered with key data supporting a nor­mal life expec­tancy, every­thing stays on the
spon­sors to facil­i­tate low- or no-cost TKI ther­apy for CML and other table. Age-appro­pri­ate “other” can­cer screen­ings should pro­ceed,
dis­eases, account­ing for >10 mil­lion doses of ther­apy over a recent as well as man­age­ment of the fre­quently observed and poten­tially
span of 3 years. Underserved regions may have lim­ited access to exac­er­bated comorbidities of hyper­gly­ce­mia/dia­be­tes, hyper­lip­
pro­vid­ers and risk assess­ment, adding to the chal­lenge. id­emia, and hyper­ten­sion and first and fore­most, screen­ing for and
In more eco­nom­i­cally favor­able set­tings, data sug­gest that man­age­ment of car­dio­vas­cu­lar dis­ease. Open dia­logue, divi­sion
comorbidities are com­mon in patients with CML and may influ­ of labor, and free com­mu­ni­ca­tion regard­ing treat­ment plans and
ence treat­ment choice and out­come. A large US series of >2000 inter­ven­tions must be established early after diag­no­sis to pre­vent
CML patients and data from the EUTOS (Euro­pean Treatment missed oppor­tu­nity. Seemingly minor things, such as pre­scrip­tion
and Outcome Study for CML) study with nearly 3000 cases of acid sup­pres­sion med­i­ca­tion such as pro­ton pump inhib­i­tors,
found that comorbidities were pres­ent in >50% of CML cases, may severely impair TKI expo­sure and response.63
and car­dio­vas­cu­lar-related comorbidities such as hyper­ten­sion The sub­spe­cialty field of “cardio-oncol­ogy”—car­di­­ol­o­gists
and dia­be­tes were pres­ent in approx­i­ma­tely 40% of cases.58,59 who focus on can­cer dis­ease and ther­apy com­pli­ca­tions—has
The large obser­va­tional SIMPLICITY study reported that the been a boon to hema­tol­ogy/oncol­ogy pro­vid­ers man­ag­ing CML
pri­mary basis for TKI choice (US/EU data set) was for “per­ as the tri­fecta of (1) remark­ably lon­ger sur­vival/nor­mal life span
ceived effi­cacy” in >50%, while only approx­i­ma­tely 9% pri­mar­ expec­ta­tions with the devel­op­ment of TKIs, (2) the breadth of
ily regarded comorbidities.60 Widely used treat­ment guide­lines TKI choices, includ­ing more potent BCR-ABL1 inhib­i­tors, and,
such as the Euro­pean LeukemiaNet have begun to incor­po­ unfor­tu­nately, (3) the rec­og­ni­tion that vas­cu­lar/car­dio­vas­cu­lar
rate a broader con­sid­er­ation of comorbidities into the ini­tial AEs may be among the most impactful risks hin­der­ing the over­
screen­ing (inclu­sion of lipid pro­file, dia­be­tes screen­ing, etc) all suc­cess of CML ther­apy. Having car­di­­ol­ogy pres­ent as part
and com­ment on patient selec­tion based on comorbid risks of the CML edu­ca­tion ses­sion at the 2017 Amer­i­can Society of
and adverse event pro­files of the var­i­ous TKIs.50 The US National Hematology meet­ing dem­on­strated the need for closer alli­ance,
Comprehensive Cancer Network,10 while clearly stat­ing TKI tox- col­lab­o­ra­tion, and coman­age­ment.64 Provisional guide­lines have
icities and man­age­ment, has yet to sig­nif­i­cantly incor­po­rate been published in reviews over the last sev­eral years focused
comorbidity assess­ment or related risk strat­i­fi­ca­tion. on man­ag­ing car­dio­vas­cu­lar adverse events in CML,65-67 focused
Several stud­ ies have clearly driven home the cen­ tral­
ity of spe­ cif­i­
cally on ponatinib and nilotinib,68-70 as well as posi­ tion
comorbidities in CML out­comes. The Charlson Comorbidity Index, papers suggesting CML clin­ i­
cal trial adverse event reporting
which incor­po­rates age along with key con­di­tions—includ­ing should align with car­dio­vas­cu­lar med­i­cine def­i­ni­tions.71
myo­car­dial infarc­tion, con­ges­tive heart fail­ure, periph­eral vas­cu­ So, who? Practically speak­ing, inven­tory of comorbid con­di­
lar dis­ease, cere­bro­vas­cu­lar acci­dent or tran­sient ische­mic attack, tions and vet­ting of nec­es­sary inter­ven­tions, and spe­cific inven­
demen­tia, chronic obstruc­tive pul­mo­nary dis­ease, con­nec­tive tory of car­dio­vas­cu­lar risk, is pru­dent for all­ patients. How?
tis­sue dis­ease, pep­tic ulcer dis­ease, liver dis­ease, dia­be­tes melli- Several val­i­dated tools are read­ily avail­­able to assess car­dio­
tus, hemi­ple­gia, chronic kid­ney dis­ease, can­cer (blood and solid vas­cu­lar risk and sub­se­quent event (for exam­ple, 10 year) risk,
Prakash Singh Shekhawat
Risks with life­long TKI ther­apy in CML  |  117
includ­ing the Framingham “hard” (non­di­ab ­ etic, no prior clau­ pri­mary care and hema­tol­ogy clin­ics with basic blood pres­sure
di­ca­tion/cor­o­nary heart dis­ease) risk score stem­ming from the and met­a­bolic dis­ease screen­ing, with a return to car­di­­ol­ogy if
long-run­ning Framingham heart study.72,73 The Euro­pean Society symp­toms or evi­dence of vas­cu­lar dis­ease devel­ops.
of Cardiology devel­oped and val­i­dated the SCORE (Systematic
Coronary Risk Evaluation) Risk Charts for use as a car­dio­vas­cu­
lar dis­ease risk assess­ment model for patients in Europe. It is Cardiovascular risk man­age­ment and pre­ven­tion:
divided into higher- and lower-risk countries and can be cali- too much, too lit­tle?
brated for spe­cific countries’ mor­tal­ity data.74 The SCORE charts It is inar­ gu­
able that inter­ven­ tion for comorbidities, such as
were exam­ined in the set­ting of nilotinib ther­apy by sev­eral aspi­rin or lipid-low­er­ing ther­apy as pri­mary or sec­ond­ary pre­
CML groups, includ­ing France and Italy,75,76 dem­on­strat­ing that ven­tion in patients with iden­ti­fied indi­ca­tions based on car­dio­
base­line assess­ment via SCORE could be a valid tool to iden­ vas­cu­lar guide­lines, is essen­tial. Intervention based on risk of
tify patients at high risk of ath­ero­scle­rotic events dur­ing nilo- com­pli­ca­tions dur­ing TKI ther­apy, based solely on the TKI risk,
tinib treat­ment. Long-term data emerg­ing from the ENESTnd is much less clear of a ben­e­fit/risk and ide­ally should be stud­ied

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/113/1851432/113mauro.pdf by guest on 13 December 2021


study of front­line nilotinib, using Framingham risk assess­ment, in con­trolled tri­als. Given the size needed for such tri­als done
cau­tioned that in addi­tion to higher cumu­la­tive rates of car­ in the non-CML set­ting, firm evi­dence-based data for pre­emp­
dio­vas­cu­lar events reported with nilotinib (300 mg twice daily, tive inter­ven­tion may be elu­sive. Smaller directed stud­ies have
16.5%; 400 mg twice daily, 23.5%) vs imatinib (3.6%), said events looked at pre­ven­ta­tive inter­ven­tions; an Ital­ian study exam­in­
were pos­si­ble in Framingham low-risk patients.29 Other region- ing ponatinib-treated patients (n = 85), strat­i­fied based on the
spe­cific risk assess­ment tools can and should be incor­po­rated Euro­pean Society of Cardiology SCORE charts as was done for
into prac­tice with CML patients; in the United Kingdom, the nilotinib, included a pri­mary pre­ven­tion strat­egy using 100 mg
QRISK score also cal­cu­lates the 10-year risk of car­dio­vas­cu­lar of aspi­ rin daily in a sub­
set (n = 19); no sig­ nif­i­
cant dif­
fer­
ences
or cere­bro­vas­cu­lar events spe­cific to the UK pop­u­la­tion and were noted, but trends toward decreased AOEs and improved
incor­po­rates postal codes in addi­tion to an increas­ing num­ber sur­vival were reported.82 Drug-drug inter­ ac­
tions and other
of clin­i­cal pre­dic­tors.77 adverse event risks from adding pre­ven­ta­tive ther­apy are not
The “when” of car­dio­vas­cu­lar risk assess­ment—fre­quency or to be overlooked. While we may feel com­pelled to inter­vene
peri­od­ic­ity of key inter­ven­tions at a gen­eral level at least and ide­ with pre­ven­tion for all­patients treated with higher-risk TKIs, risk
ally patient or patient/TKI risk level—and likely the more pre­cise strat­i­fi­ca­tion may help jus­tify inter­ven­tion in larger num­bers of
details of which diag­nos­tic tools, poten­tial bio­mark­ers, etc, are cases, but an “only where indi­cated” approach may be more
most infor­ma­tive con­tinue to be inves­ti­gated. A unique pro­spec­ pru­dent.
tive study in the US of car­dio­vas­cu­lar and met­a­bolic param­e­ters
at diag­no­sis of CML and sub­se­quently through treat­ment, agnos­ Is TKI ther­apy life­long, and could bet­ter ther­apy help
tic to TKI choice, has reported high rates of base­line risk and con­ mit­i­gate risk?
tin­ues to gather data on changes with time on treat­ment.78 At We are at a cross­roads in the treat­ment of CML, hav­ing first
pres­ent, the use of pro­vi­sional cardio-oncol­ogy-derived guide­ rev­o­lu­tion­ized the prin­ci­pal approach with the devel­op­ment of
lines and leu­ke­mia con­sor­tium-based guide­lines,64,79,80,81 driven TKIs and now mov­ing away from a “trans­plant when pos­si­ble”
by patient comorbidity and TKI choice, adds sim­ple and lower approach and expec­ta­tions of dimin­ished sur­vival in oth­ers to
cost empiric mon­i­tor­ing based on known risks and thee best- a par­a­digm of “trans­plant only when abso­lutely needed” and
avail­­able diag­nos­tics/pre­dic­tors of preexisting or accel­er­at­ing near-nor­mal or nor­mal life expec­tancy for most. Targeted ther­
vas­cu­lar dis­ease. At the most basic level, broad use of a sim­ple apy in CML has fos­tered a new modus ope­randi of chronic main­
ABCDE approach—(a)wareness, (a)nkle-bra­ chial index test­ ing, te­nance che­mo­ther­apy—new in can­cer for the most part—thus
(a)spirin ther­apy, (b)lood pres­sure con­trol, (c)igarette ces­sa­tion, rais­ing rea­son­able fears of mor­bid­ity from con­tin­ual, poten­tially
and (c)holesterol low­er­ing—will raise aware­ness and at a min­i­ life­
long che­ mo­ther­
apy. Twenty years after imatinib’s record-
mum facil­i­tate stan­dard of care inter­ven­tion for comorbid find­ break­ing fast approval by the FDA, we take solace in the rel­a­tive
ings in CML patients. safety of TKIs, led by imatinib, followed by more hes­i­tant con­
fi­dence in the long-term safety of our more potent and sub­se­
quently devel­oped agents. The advent of TFR as an option for
CML has brought the next cross­road: Can we make CML ther­
CLINICAL CASE (Con­t in­u ed) apy as lim­ited as pos­si­ble, and func­tion­ally cure the can­cer, in
While final­iz­ing the last hema­to­logic assess­ments and confirm- more/many/all­ patients?
ing her genetic results and pathol­ogy, you offer the patient TFR stud­ ies have expanded greatly, and data from tri­ als
a con­sul­ta­tion in your cen­ter’s pri­mary care clinic, which she performed after pri­mary or sec­ond­ary treat­ment with sec­ond-
eagerly pur­sues as she is wor­ried about her gen­eral health gen­er­a­tion TKIs are avail­­able. Trials with nilotinib and dasatinib
given the CML diag­no­sis. The result of your TKI choice dis­cus­ as sec­ond-line ther­apy after imatinib resis­tance or intol­er­ance,
sion led you to rec­om­mend a sec­ond-gen­er­a­tion TKI, and in or as a means to opti­mize deep remis­sion, have proven that TFR
dis­cus­sion with the pri­mary care team, a screen­ing eval­u­a­tion is both fea­si­ble and sim­i­larly suc­cess­ful to a more uncom­pli­
with the cen­ter’s car­di­­ol­ogy group is sched­uled. She under- cated TFR after pri­mary imatinib or sec­ond-gen­er­a­tion TKIs.83,84
goes basic met­a­bolic and func­tional imag­ing and is found to While over­all rates of TFR suc­cess may not be higher in stud­ies
have a low risk of events in the next 10 years. Cardiovascular with sec­ond-gen­er­a­tion TKIs used as front­line ther­apy,84,85 the
mon­i­tor­ing is pre­scribed by the car­di­­ol­o­gist to con­tinue in the rapid­ity of deep remis­sion may afford more rapid eli­gi­bil­ity to

118  |  Hematology 2021  |  ASH Education Program


con­sider TFR and thus reduce treat­ment dura­tion/expo­sure and Correspondence
the poten­tial like­li­hood of toxicities. If TFR were more per­va­sive Michael J. Mauro, Memorial Sloan Kettering Cancer Center, 1275
and suc­cess­ful and achiev­able on a more global level, this would York Ave, Box 489, New York, NY 10065; e-mail: maurom@mskcc​
change the per­spec­tive on tox­ic­ity for cer­tain as short-term and ­.org.
life­long risk are strik­ingly dif­fer­ent. The next steps in long-term
tox­ic­ity assess­ment should incor­po­rate crit­i­cal think­ing into References
the rel­a­tive ben­e­fits of more potent ther­a­pies and strat­e­gies 1. Craddock CF. We do still trans­plant CML, don’t we? Hematology Am Soc
cou­pled with the ben­e­fits of lim­it­ing dura­tion of treat­ment for Hematol Educ Program. 2018;2018(1):177-184.
patients with CML. 2. Etienne G, Guilhot J, Rea D, et al. Long-term fol­low-up of the French Stop
Imatinib (STIM1) study in patients with chronic mye­loid leu­ke­mia. J Clin
Oncol. 2017;35(3):298-305.
3. Saussele S, Richter J, Guilhot J, et al; EURO-SKI Investigators. Discontin-
uation of tyro­sine kinase inhib­i­tor ther­apy in chronic mye­loid leu­kae­mia
(EURO-SKI): a prespecified interim anal­y­sis of a pro­spec­tive, multicentre,
CLINICAL CASE (Con­t in­u ed) non-randomised, trial. Lancet Oncol. 2018;19(6):747-757.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/113/1851432/113mauro.pdf by guest on 13 December 2021


Your patient proceeded to ther­apy with a sec­ond-gen­er­a­tion 4. Rousselot P, Loiseau C, Delord M, Cayuela JM, Spentchian M. Late molec­
u­lar recur­rences in patients with chronic mye­loid leu­ke­mia expe­ri­enc­ing
TKI and achieved rapid and deep response. Major molec­u­lar
treat­ment-free remis­sion. Blood Adv. 2020;4(13):3034-3040.
response occurred within 6 months, and deep remis­sion (>MR 5. Hsieh YC, Kirschner K, Copland M. Improving out­comes in chronic mye­
4, <0.01% IS BCR-ABL) was pres­ent from 9 months through a loid leu­ke­mia through harnessing the immu­no­log­i­cal land­scape. Leukemia.
total 3 years of treat­ment. She con­tin­ued with her new pri­ 2021;35(May):1229-1242.
mary phy­si­cian and did not develop any major com­pli­ca­tions. 6. Branford S, Kim DDH, Apperley JF, et al; International CML Foundation
Genomics Alliance. Laying the foun­da­tion for genomically-based risk
She was delighted with her response and eager to pur­sue TFR
assess­ment in chronic mye­loid leu­ke­mia. Leukemia. 2019;33(8):1835-1850.
shortly after her 3 years in remis­sion and 2 ¼ years in deep 7. Hochhaus A, Larson RA, Guilhot F, et al; IRIS Investigators. Long-term out­
molec­u­lar remis­sion. She remains off treat­ment in suc­cess­ful comes of imatinib treat­ment for chronic mye­loid leu­ke­mia. N Engl J Med.
TFR and is eter­nally grate­ful for you man­ag­ing her CML so well 2017;376(10):917-927.
8. Bower H, Björkholm M, Dickman PW, Höglund M, Lambert PC, Anders-
and ensur­ing she had excel­lent pri­mary care and an intro­duc­
son TML. Life expec­tancy of patients with chronic mye­loid leu­ke­mia
tion to car­di­­ol­ogy care. approaches the life expec­tancy of the gen­eral pop­u­la­tion. J Clin Oncol.
2016;34(24):2851-2857.
9. Höglund M, Sandin F, Simonsson B. Epidemiology of chronic mye­loid leu­
kae­mia: an update. Ann Hematol. 2015;94(suppl 2):S241-S247.
Key points
10. National Comprehensive Cancer Network. NCCN guide­lines: chronic mye­
• At diag­no­sis/ini­tial pre­sen­ta­tion of CML, appraisal of the pa- loid leu­ke­mia. Accessed 1 June 2021.
tient’s full comorbidity pro­file is vitally impor­tant and affects 11. Shanmuganathan N, Braley JA, Yong ASM, et al. Modeling the safe min­i­mum
sur­vival more than the CML. fre­quency of molec­u­lar mon­i­tor­ing for CML patients attempting treat­ment-
• CML risk strat­i­fi­ca­tion (Sokal, EUTOS Long Term Survival, etc) free remis­sion. Blood. 2019;134(1):85-89.
12. Findakly D, Arslan W. Clinical fea­ tures and out­ comes of patients with
affects TKI response and the suc­cess of poten­tial TFR out­
chronic mye­loid leu­ke­mia presenting with iso­lated thrombocytosis: a sys­
come and may assist with TKI choice. tem­atic review and a case from our insti­tu­tion. Cureus. 2020;12(6):e8788.
• Cardiovascular risk assess­ment (Framingham, SCORE, oth­ers) 13. Knöfler R, Lange BS, Paul F, Tiebel O, Suttorp M. Bleeding signs due to
at diag­no­sis can also assist with TKI choice and can clar­ify the acquired von Willebrand syn­drome at diag­no­sis of chronic mye­loid leu­kae­
mia in chil­dren. Br J Haematol. 2020;188(5):701-706.
need and mer­its of any pre­ven­ta­tive inter­ven­tions.
14. Abruzzese E, Mauro M, Apperley J, Chelysheva E. Tyrosine kinase inhib­i­tors
• TKI choice dic­tates gen­eral (class) and drug-spe­cific base­ and preg­nancy in chronic mye­loid leu­ke­mia: opin­ion, evi­dence, and rec­om­
line and lon­gi­tu­di­nal mon­i­tor­ing for adverse events; TKIs with men­da­tions. Ther Adv Hematol. 2020;11(31 Octo­ber):2040620720966120.
more sig­nif­i­cant car­dio­vas­cu­lar risk war­rant reg­u­lar car­dio­ 15. Qosa H, Avaritt BR, Hartman NR, Volpe DA. In vitro UGT1A1 inhi­bi­tion by
vas­cu­lar mon­i­tor­ing. tyro­sine kinase inhib­i­tors and asso­ci­a­tion with drug-induced hyperbiliru-
binemia. Cancer Chemother Pharmacol. 2018;82(5):795-802.
• Partnership and coor­di­na­tion with pri­mary care/gen­eral inter­
16. Guilhot F, Hughes TP, Cortes J, et al. Plasma expo­sure of imatinib and its
nal med­i­cine and sub­spe­cialty med­i­cine, espe­cially car­di­­ol­ogy/ cor­re­la­tion with clin­i­cal response in the tyro­sine kinase inhib­i­tor opti­mi­za­
cardio-oncol­ogy, serves CML patients best to iden­tify and tion and selec­tiv­ity trial. Haematologica. 2012;97(5):731-738.
man­age adverse events and con­tinue age-appro­pri­ate health 17. Berman E, Nicolaides M, Maki RG, et al. Altered bone and min­eral metab­o­lism
and other can­cer screen­ings. in patients receiv­ing imatinib mesylate. N Engl J Med. 2006;354(19):2006-
2013.
• TFR as a goal of ther­apy, by lim­it­ing TKI dura­tion, may sig­nif­i­ 18. Yilmaz M, Lahoti A, O’Brien S, et  al. Estimated glo­mer­u­lar fil­tra­tion rate
cantly mod­ify TKI ther­apy risk, but life­long ther­apy remains a changes in patients with chronic mye­loid leu­ke­mia treated with tyro­sine
nec­es­sary par­a­digm and mindset in man­ag­ing the increas­ing kinase inhib­i­tors. Cancer. 2015;121(21):3894-3904.
num­ber of patients sur­viv­ing CML. 19. Cortes JE, Gambacorti-Passerini C, Kim DW, et al. Effects of bosutinib treat­
ment on renal func­tion in patients with Philadelphia chro­mo­some-pos­i­tive
leu­ke­mias. Clin Lymphoma Myeloma Leuk. 2017;17(10):684-695.e6695e6.
20. Breccia M, Muscaritoli M, Gentilini F, et al. Impaired fasting glu­cose level as
Conflict-of-inter­est dis­clo­sure met­a­bolic side effect of nilotinib in non-dia­betic chronic mye­loid leu­ke­mia
Michael J. Mauro: research funding: Novartis, Bristol Myers patients resis­tant to imatinib. Leuk Res. 2007;31(12):1770-1772.
Squibb, Sun Pharma/Sparc, Takeda; con­sul­tancy: Novartis, Bris- 21. Gómez-Sámano MA, Baquerizo-Burgos JE, Coronel MFC, et  al. Effect of
tol Myers Squibb, Takeda, Pfizer. imatinib on plasma glu­cose con­cen­tra­tion in sub­jects with chronic mye­
loid leu­ke­mia and gas­tro­in­tes­ti­nal stro­mal tumor. BMC Endocr Disord.
2018;18(1):77.
Off-label drug use 22. Breccia M, Loglisci G, Salaroli A, Serrao A, Alimena G. Nilotinib-medi­ated
Michael J. Mauro: nothing to disclose. increase in fasting glu­cose level is revers­ible, does not con­vert to type 2
Prakash Singh Shekhawat
Risks with life­long TKI ther­apy in CML  |  119
dia­be­tes and is likely cor­re­lated with increased body mass index. Leuk Res. 43. Ozawa MG, Ewalt MD, Gratzinger D. Dasatinib-related fol­lic­ul­ar hyper­pla­
2012;36(4):e66-e67. sia: an underrecognized entity with char­ac­ter­is­tic mor­phol­ogy. Am J Surg
23. Iurlo A, Orsi E, Cattaneo D, et  al. Effects of first- and sec­ond-gen­er­a­tion Pathol. 2015;39(10):1363-1369.
tyro­sine kinase inhib­ i­
tor ther­apy on glu­ cose and lipid metab­ o­
lism in 44. Schiffer CA, Cortes JE, Hochhaus A, et al. Lymphocytosis after treat­ment
chronic mye­loid leu­ke­mia patients: a real clin­i­cal prob­lem? Oncotarget. with dasatinib in chronic mye­loid leu­ke­mia: effects on response and tox­ic­
2015;6(32):33944-33951. ity. Cancer. 2016;122(9):1398-1407.
24. Rea D, Mirault T, Cluzeau T, et al. Early onset hyper­cho­les­ter­­ol­emia induced 45. Weatherald J, Bondeelle L, Chaumais MC, et al. Pulmonary com­pli­ca­tions
by the 2nd-gen­er­a­tion tyro­sine kinase inhib­i­tor nilotinib in patients with of Bcr-Abl tyro­sine kinase inhib­i­tors. Eur Respir J. 2020;56(4):2000279.
chronic phase-chronic mye­loid leu­ke­mia. Haematologica. 2014;99(7):1197- 46. Weatherald J, Chaumais MC, Savale L, et al. Long-term out­comes of dasat-
1203. inib-induced pul­mo­nary arte­rial hyper­ten­sion: a pop­u­la­tion-based study.
25. Franklin M, Burns L, Perez S, Yerragolam D, Makenbaeva D. Incidence of Eur Respir J. 2017;50(1):1700217.
type 2 dia­be­tes mellitus and hyper­lip­id­emia in patients pre­scribed dasat- 47. Cortes JE, Kantarjian H, Shah NP, et al. Ponatinib in refrac­tory Philadelphia
inib or nilotinib as first- or sec­ond-line ther­apy for chronic mye­log­e­nous chro­mo­some-pos­i­tive leu­ke­mias. N Engl J Med. 2012;367(22):2075-2088.
leu­ke­mia in the US. Curr Med Res Opin. 2018;34(2):353-360. 48. Cortes JE, Kim DW, Pinilla-Ibarz J, et al; PACE Investigators. A phase 2 trial
26. Jain P, Kantarjian H, Boddu PC, et al. Analysis of car­dio­vas­cu­lar and arteri- of ponatinib in Philadelphia chro­mo­some-pos­i­tive leu­ke­mias. N Engl J Med.
othrombotic adverse events in chronic-phase CML patients after front­line 2013;369(19):1783-1796.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/113/1851432/113mauro.pdf by guest on 13 December 2021


TKIs. Blood Adv. 2019;3(6):851-861. 49. Latifi Y, Moccetti F, Wu M, et al. Thrombotic microangiopathy as a cause of
27. Douxfils J, Haguet H, Mullier F, Chatelain C, Graux C, Dogné JM. Association car­dio­vas­cu­lar tox­ic­ity from the BCR-ABL1 tyro­sine kinase inhib­i­tor ponati-
between BCR-ABL tyro­sine kinase inhib­i­tors for chronic mye­loid leu­ke­mia nib. Blood. 2019;133(14):1597-1606.
and car­dio­vas­cu­lar events, major molec­u­lar response, and over­all sur­vival: 50. Hochhaus A, Baccarani M, Silver RT, et  al. Euro­pean LeukemiaNet 2020
a sys­tem­atic review and meta-anal­y­sis. JAMA Oncol. 2016;2(5):625-632. rec­om­men­da­tions for treating chronic mye­loid leu­ke­mia. Leukemia. 2020;
28. Cortes JE, Kantarjian HM, Mauro MJ, et  al. Long-term car­diac, vas­cu­lar, 34(4):966-984.
hyper­ten­sion, and effu­sion safety of bosutinib in patients with Philadelphia 51. Cortes J, Apperley J, Lomaia E, et  al. OPTIC pri­ mary anal­ y­sis: a dose-
chro­mo­some-pos­i­tive leu­ke­mia resis­tant or intol­er­ant to prior ther­apy. Eur opti­mi­za­tion study of 3 starting doses of ponatinib (PON). J Clin Oncol.
J Haematol. 2021;106(6):808-820. 2021;39(suppl 15):7000.
29. Kantarjian HM, Hughes TP, Larson RA, et al. Long-term out­comes with front­ 52. Januzzi JL, Garasic J, Kasner S, et al. An inde­pen­dent review of arte­rial occlu­
line nilotinib ver­sus imatinib in newly diag­nosed chronic mye­loid leu­ke­mia sive events (AOEs) in the ponatinib (PON) phase II PACE trial (NCT01207440)
in chronic phase: ENESTnd 10-year anal­y­sis. Leukemia. 2021;35(2):440-453. in patients (pts) with Ph+ leu­ke­mia. J Clin Oncol. 2020;38(suppl 15):7550.
30. Hadzijusufovic E, Albrecht-Schgoer K, Huber K, et al. Nilotinib-induced vas- 53. Hughes TP, Mauro MJ, Cortes JE, et al. Asciminib in chronic mye­loid leu­ke­
culopathy: iden­ti­fi­ca­tion of vas­cu­lar endo­the­lial cells as a pri­mary tar­get mia after ABL kinase inhib­i­tor fail­ure. N Engl J Med. 2019;381(24):2315-2326.
site. Leukemia. 2017;31(11):2388-2397. 54. Hochhaus A, Boquimpani B, Réa D, et al. Efficacy and safety results from
31. Bocchia M, Galimberti S, Aprile L, et al. Genetic pre­dis­po­si­tion and induced ASCEMBL, a mul­ti­cen­ter, open-label, phase 3 study of asciminib, a first-
pro-inflam­ma­tory/pro-oxi­da­tive sta­tus may play a role in increased athero- in-class STAMP inhib­i­tor, vs bosutinib in patients with chronic mye­loid
thrombotic events in nilotinib treated chronic mye­loid leu­ke­mia patients. leu­ke­mia in chronic phase pre­vi­ously treated with ≥2 tyro­sine kinase
Oncotarget. 2016;7(44):72311-72321. inhib­i­tors. Blood. 2020;136(suppl 2):LBA-4.
32. Pouwer MG, Pieterman EJ, Verschuren L, et  al. The BCR-ABL1 inhib­i­tors 55. Wylie AA, Schoepfer J, Jahnke W, et al. The allo­ste­ric inhib­i­tor ABL001 enables
imatinib and ponatinib decrease plasma cho­les­terol and ath­ero­scle­ro­sis, dual targeting of BCR-ABL1. Nature. 2017;543(7647):733-737.
and nilotinib and ponatinib acti­vate coag­u­la­tion in a trans­la­tional mouse 56. Mendizabal AM, Younes N, Levine PH. Geographic and income var­i­a­tions in
model. Front Cardiovasc Med. 2018;5(12 June):55. age at diag­no­sis and inci­dence of chronic mye­loid leu­ke­mia. Int J Hematol.
33. Gover-Proaktor A, Granot G, Pasmanik-Chor M, et al. Bosutinib, dasatinib, 2016;103(1):70-78.
imatinib, nilotinib, and ponatinib dif­fer­en­tially affect the vas­cu­lar molec­u­lar 57. Patel M, Philip V, Fazel F, et al. Human immu­no­de­fi­ciency virus infec­tion and
path­ways and func­tion­al­ity of human endo­the­lial cells. Leuk Lymphoma. chronic mye­loid leu­ke­mia. Leuk Res. 2012;36(11):1334-1338.
2019;60(1):189-199. 58. Jabbour E, Makenbaeva D, Lingohr-Smith M, Lin J. Use of real-world claim
34. Sant’Antonio E, Camerini C, Rizzo V, Musolino C, Allegra A. Genetic het­ero­ data­bases to assess prev­a­lence of comorbid con­di­tions rel­e­vant to the
ge­ne­ity in chronic mye­loid leu­ke­mia: how clonal hema­to­poi­e­sis and clonal treat­ment of chronic mye­log­e­nous leu­ke­mia based on National Compre-
evo­lu­tion may influ­ence prog­no­sis, treat­ment out­come, and risk of car­dio­ hensive Network Treatment Guidelines. Clin Lymphoma Myeloma Leuk.
vas­cu­lar events. Clin Lymphoma Myeloma Leuk. 2021;21(9):573-579. 2015;15(12):797-802.
35. Quintás-Cardama A, Kantarjian H, O’Brien S, et  al. Pleural effu­ sion in 59. Hoffmann VS, Baccarani M, Hasford J, et al. The EUTOS pop­u­la­tion-based reg­
patients with chronic mye­log­e­nous leu­ke­mia treated with dasatinib after is­try: inci­dence and clin­i­cal char­ac­ter­is­tics of 2904 CML patients in 20 Euro­
imatinib fail­ure. J Clin Oncol. 2007;25(25):3908-3914. pean countries. Leukemia. 2015;29(6):1336-1343.
36. Hughes TP, Laneuville P, Rousselot P, et  al. Incidence, out­comes, and risk 60. Goldberg SL, Cortes JE, Gambacorti-Passerini C, et  al. First-line treat­
fac­tors of pleu­ral effu­sion in patients receiv­ing dasatinib ther­apy for Philadel- ment selec­tion and early mon­i­tor­ing pat­terns in chronic phase-chronic
phia chro­mo­some-pos­i­tive leu­ke­mia. Haematologica. 2019;104(1):93-101. mye­loid leu­ke­mia in rou­tine clin­i­cal prac­tice: SIMPLICITY. Am J Hematol.
37. Shah NP, Rousselot P, Schiffer C, et  al. Dasatinib in imatinib-resis­tant or 2017;92(11):1214-1223.
-intol­er­ant chronic-phase, chronic mye­loid leu­ke­mia patients: 7-year fol­low- 61. Saussele S, Krauss MP, Hehlmann R, et al; Schweizerische Arbeitsgemein-
up of study CA180-034. Am J Hematol. 2016;91(9):869-874. schaft für Klinische Krebsforschung; Ger­man CML Study Group. Impact of
38. Cortes JE, Saglio G, Kantarjian HM, et al. Final 5-year study results of DASI- comorbidities on over­all sur­vival in patients with chronic mye­loid leu­ke­
SION: the dasatinib ver­sus imatinib study in treat­ment-naïve chronic mye­ mia: results of the ran­dom­ized CML study IV. Blood. 2015;126(1):42-49.
loid leu­ke­mia patients trial. J Clin Oncol. 2016;34(20):2333-2340. 62. Ono T, Takahashi N, Kizaki M, et  al. Prognostic effect of comorbidities in
39. Naqvi K, Jabbour E, Skinner J, et al. Long-term fol­low-up of lower dose dasat- patients with chronic mye­ loid leu­ ke­ mia treated with a tyro­ sine kinase
inib (50 mg daily) as front­ line ther­
apy in newly diag­ nosed chronic-phase inhib­i­tor. Cancer Sci. 2020;111(10):3714-3725.
chronic mye­loid leu­ke­mia. Cancer. 2020;126(1):67-75. 63. Takahashi N, Miura M, Niioka T, Sawada K. Influence of H2-recep­tor antag­o­
40. Cortes JE, Jimenez CA, Mauro MJ, Geyer A, Pinilla-Ibarz J, Smith BD. Pleu- nists and pro­ton pump inhib­i­tors on dasatinib phar­ma­co­ki­net­ics in Jap­a­nese
ral effu­sion in dasatinib-treated patients with chronic mye­loid leu­ke­mia in leu­ke­mia patients. Cancer Chemother Pharmacol. 2012;69(4):999-1004.
chronic phase: iden­ti­fi­ca­tion and man­age­ment. Clin Lymphoma Myeloma 64. Barber MC, Mauro MJ, Moslehi J. Cardiovascular care of patients with
Leuk. 2017;17(2):78-82. chronic mye­loid leu­ke­mia (CML) on tyro­sine kinase inhib­i­tor (TKI) ther­apy.
41. Mustjoki S, Ekblom M, Arstila TP, et al. Clonal expan­sion of T/NK-cells dur­ Hematology Am Soc Hematol Educ Program. 2017;2017(1):110-114.
ing tyro­sine kinase inhib­i­tor dasatinib ther­apy. Leukemia. 2009;23(8):1398- 65. Seguro FS, Silva CMPDC, Moura CMB, et al. Recommendations for the man­
1405. age­ment of car­dio­vas­cu­lar risk in patients with chronic mye­loid leu­ke­mia
42. Mustjoki S, Auvinen K, Kreutzman A, et al. Rapid mobi­li­za­tion of cyto­toxic on tyro­sine kinase inhib­i­tors: risk assess­ment, strat­i­fi­ca­tion, treat­ment and
lym­pho­cytes induced by dasatinib ther­apy. Leukemia. 2013;27(4):914-924. mon­i­tor­ing. Hematol Transfus Cell Ther. 2021;43(2):191-200.

120  |  Hematology 2021  |  ASH Education Program


66. Kondapalli L, Worth S, Hawi R, et al. Collaborative car­dio­vas­cu­lar man­age­ 78. Mauro MJ, Oehler VG, Thompson JE, et  al. Cardiovascular and met­a­bolic
ment of patients with chronic mye­loid leu­ke­mia on tyro­sine kinase inhib­i­ risk in patients with chronic mye­loid leu­ke­mia in chronic phase receiv­ing
tors. Vasc Med. 2020;25(3):246-254. first-line BCR-ABL1 tyro­sine kinase inhib­i­tors in the United States: base­line
67. Medeiros BC, Possick J, Fradley M. Cardiovascular, pul­mo­nary, and met­a­ and six-month fol­low-up results from a pro­spec­tive real-world obser­va­
bolic toxicities com­pli­cat­ing tyro­sine kinase inhib­i­tor ther­apy in chronic tional study. Blood. 2020;136(suppl 1):39-40.
mye­loid leu­ke­mia: strat­e­gies for mon­i­tor­ing, detecting, and man­ag­ing. 79. Lyon AR, Dent S, Stanway S, et al. Baseline car­dio­vas­cu­lar risk assess­ment in
Blood Rev. 2018;32(4):289-299. can­cer patients sched­uled to receive cardiotoxic can­cer ther­a­pies: a posi­
68. Casavecchia G, Galderisi M, Novo G, et al. Early diag­no­sis, clin­i­cal man­age­ tion state­ment and new risk assess­ment tools from the Cardio-Oncology
ment, and fol­low-up of car­dio­vas­cu­lar events with ponatinib. Heart Fail Study Group of the Heart Failure Association of the Euro­pean Society of
Rev. 2020;25(3):447-456. Cardiology in col­lab­o­ra­tion with the International Cardio-Oncology Soci-
69. Saussele S, Haverkamp W, Lang F, et  al. Ponatinib in the treat­ment of ety. Eur J Heart Fail. 2020;22(11):1945-1960.
chronic mye­loid leu­ke­mia and Philadelphia chro­mo­some-pos­i­tive acute 80. Manouchehri A, Kanu E, Mauro MJ, Aday AW, Lindner JR, Moslehi J. Tyrosine
leu­ke­mia: rec­om­men­da­tions of a Ger­man expert con­sen­sus panel with kinase inhib­i­tors in leu­ke­mia and car­dio­vas­cu­lar events: from mech­a­nism
focus on car­dio­vas­cu­lar man­age­ment. Acta Haematol. 2020;143(3):217- to patient care. Arterioscler Thromb Vasc Biol. 2020;40(2):301-308.
231. 81. Steegmann JL, Baccarani M, Breccia M, et al. Euro­pean LeukemiaNet rec­
70. Valent P, Hadzijusufovic E, Hoermann G, et al. Risk fac­tors and mech­a­nisms om­men­da­tions for the man­age­ment and avoid­ance of adverse events of

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/113/1851432/113mauro.pdf by guest on 13 December 2021


con­trib­ut­ing to TKI-induced vas­cu­lar events in patients with CML. Leuk treat­ment in chronic mye­loid leu­kae­mia. Leukemia. 2016;30(8):1648-1671.
Res. 2017;59(August):47-54. 82. Caocci G, Mulas O, Abruzzese E, et al. Arterial occlu­sive events in chronic
71. Aghel N, Delgado DH, Lipton JH. Cardiovascular events in chronic mye­loid mye­loid leu­ke­mia patients treated with ponatinib in the real-life prac­tice
leu­ke­mia clin­i­cal tri­als. Is it time to reassess and report the events accord- are predicted by the Systematic Coronary Risk Evaluation (SCORE) chart.
ing to car­di­­ol­ogy guide­lines? Leukemia. 2018;32(10):2095-2104. Hematol Oncol. 2019;37(3):296-302.
72. Framingham Heart Study. Hard cor­o­nary heart dis­ease. Accessed 18 Octo­ber 83. Hughes TP, Clementino NCD, Fominykh M, et al. Long-term treat­ment-free
2021. remis­sion in patients with chronic mye­loid leu­ke­mia after sec­ond-line nilo-
73. D’Agostino RB Sr, Vasan RS, Pencina MJ, et al. General car­dio­vas­cu­lar risk tinib: ENESTop 5-year update. Leukemia. 2021;35(6):1631-1642.
pro­file for use in pri­mary care: the Framingham Heart Study. Circulation. 84. Shah NP, García-Gutiérrez V, Jiménez-Velasco A, et  al. Dasatinib dis­con­
2008;117(6):743-753. tin­u­a­tion in patients with chronic-phase chronic mye­loid leu­ke­mia and
74. Euro­pean Society of Cardiology. Score risk charts: the Euro­pean car­dio­vas­ sta­ble deep molec­u­lar response: the DASFREE study. Leuk Lymphoma.
cu­lar dis­ease risk assess­ment mod­els. Accessed 18 Octo­ber 2021. 2020;61(3):650-659.
75. Rea D, Mirault T, Raffoux E, et  al. Usefulness of the 2012 Euro­pean CVD 85. Radich JP, Hochhaus A, Masszi T, et al. Treatment-free remis­sion fol­low­ing
risk assess­ment model to iden­tify patients at high risk of car­dio­vas­cu­lar front­line nilotinib in patients with chronic phase chronic mye­loid leu­ke­mia:
events dur­ing nilotinib ther­apy in chronic mye­loid leu­ke­mia. Leukemia. 5-year update of the ENESTfreedom trial. Leukemia. 2021;35(5):1344-1355.
2015;29(5):1206-1209.
76. Breccia M, Molica M, Zacheo I, Serrao A, Alimena G. Application of sys­tem­
atic cor­o­nary risk eval­u­a­tion chart to iden­tify chronic mye­loid leu­ke­mia
patients at risk of car­dio­vas­cu­lar dis­eases dur­ing nilotinib treat­ment. Ann
Hematol. 2015;94(3):393-397.
77. Hippisley-Cox J, Coupland C, Brindle P. Development and val­i­da­tion of
QRISK3 risk pre­dic­tion algorithms to estimate future risk of cardiovascular © 2021 by The Amer­i­can Society of Hematology
disease: prospective cohort study. BMJ. 2017;357: j2099. DOI 10.1182/hema­tol­ogy.2021000239

Prakash Singh Shekhawat


Risks with life­long TKI ther­apy in CML  |  121
CML: SUCCESS BREEDS MORE SUCCESS

Blast and accelerated phase CML:


room for improvement

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/122/1851605/122how.pdf by guest on 13 December 2021


Joan How,1–3 Vinayak Venkataraman,1 and Gabriela Soriano Hobbs1
1
Department of Medical Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA; 2Division of Hematology, Department
of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA; and 3Department of Medical Oncology, Dana­Farber Cancer
Institute, Harvard Medical School, Boston, MA

Tyrosine kinase inhibitors (TKIs) revolutionized the treatment of chronic myeloid leukemia (CML). With TKI therapy, the
percentage of patients who progress to accelerated phase (AP) or blast phase (BP) CML has decreased from more than
20% to 1% to 1.5% per year. Although AP- and BP-CML occur in a minority of patients, outcomes in these patients are
significantly worse compared with chronic phase CML, with decreased response rates and duration of response to TKI.
Despite this, TKIs have improved outcomes in advanced phase CML, particularly in de novo AP patients, but are often
inadequate for lasting remissions. The goal of initial therapy in advanced CML is a return to a chronic phase followed by
consideration for bone marrow transplantation. The addition of induction chemotherapy with TKI is often necessary for
achievement of a second chronic phase. Given the small population of patients with advanced CML, development of
novel treatment strategies and investigational agents is challenging, although clinical trial participation is encouraged
in AP and BP patients, whenever possible. We review the overall management approach to advanced CML, including TKI
selection, combination therapy, consideration of transplant, and novel agents.

LEARNING OBJECTIVES
• Understand the treatment approach to accelerated and blast phase CML in de novo disease and occurring in
patients with underlying CP­CML
• Understand response rates and outcomes in patients with advanced CML treated with TKI, TKI and chemotherapy,
and hematopoietic stem cell transplantation

blasts, but M351T and T3151 mutations and KMT2A par­


CLINICAL CASE tial duplication were identified. She was started on pona­
NM is a 54­year­old woman diagnosed with chronic mye­ tinib 45 mg daily. Four months after ponatinib initiation,
loid leukemia (CML) in chronic phase (CP), after present­ polymerase chain reaction for BCR­ABL rose to more than
ing with a white blood cell (WBC) count of 270 × 109/L, 50%, and bone marrow biopsy specimen showed 25%
hemoglobin of 7.5 g/dL, and platelets of 100 × 109/L. She myeloblasts along with rising peripheral WBC count. She
was initiated on imatinib, which she took irregularly and was transferred to our hospital and received cytarabine
eventually discontinued. She established care with a new 200 mg/m2 intravenously daily for 7 days and idarubicin
physician and resumed imatinib when she was found to 12 mg/m2 for 3 days. Ponatinib was resumed at the end
have a WBC count of 68 × 109/L, hemoglobin of 10.4 g/dL, of induction. She went into a second CP and underwent
and platelets of 129 × 109/L. After initial response, periph­ haploidentical stem cell transplant with posttransplant
eral blood polymerase chain reaction for BCR­ABL rose cyclophosphamide. She received fludarabine, melphalan,
to 35% after 6 months. A bone marrow biopsy specimen and total body irradiation for conditioning. She remains
demonstrated CP­CML, and she was switched to dasat­ in complete molecular remission 9 months after trans­
inib 100 mg daily. After 7 months of treatment on dasati­ plant and has discontinued tyrosine kinase inhibitor (TKI)
nib, BCR­ABL rose from less than 1% to 8.013%. A repeat therapy.
bone marrow biopsy specimen did not show increased

122 | Hematology 2021 | ASH Education Program


Introduction trans­for­ma­tion is asso­ci­ated with ini­tial treat­ment with sec­ond-
Chronic mye­loid leu­ke­mia is a mye­lo­pro­lif­er­a­tive neo­plasm gen­er­a­tion TKIs com­pared with imatinib.4
char­ac­ter­ized by the Philadelphia chro­mo­some (Ph) that affects Complicating the epi­de­mi­­ol­ogy and treat­ment of advanced
1 to 2 per 100 000 new patients per year and com­prises 15% of phase CML are the dif­fer­ent clas­si­fi­ca­tion sys­tems used to define
leu­ke­mias in adults.1 The dis­ease is driven by a recip­ro­cal trans­ AP- and BP-CML (Table 1), which include the International Blood
lo­ca­tion of chro­mo­somes 9 and 22, which results in the BCR- and Marrow Transplant Registry (IBMTR),5 M. D. Anderson Can­
ABL fusion pro­tein and dysregulated tyro­sine kinase activ­ity. cer Center (MDACC),6 Euro­pean LeukemiaNet,7 and World Health
Patients most com­monly pres­ent in CP, but with­out treat­ment, Organization (WHO) cri­te­ria.8 One major dif­fer­ence between clas­
CP-CML will prog­ress to accel­er­ated phase (AP-CML) and blast si­fi­ca­tion sys­tems is the thresh­old blast per­cent­age used to dis­tin­
cri­sis (BP-CML) within 3 to 5 years. Tyrosine kinase inhib­i­tors guish CP-, AP-, and BP-CML, with the WHO defin­ing BP as a blast
rev­o­lu­tion­ized the care of CML with the approval of imatinib, per­cent­age of more than 20% and all­other clas­si­fi­ca­tion sys­tems
the only first-gen­er­a­tion TKI. Three sec­ond-gen­er­a­tion TKIs, da­ using a thresh­old of more than 30%. Of note, the acqui­si­tion of
satinib, nilotinib, and bosutinib, are also avail­­able for front­line major-route ACAs on treat­ment is con­sid­ered a hall­mark of AP-CML,
use. Progression to advanced CML is due to con­tin­ued BCR- and the WHO also con­sid­ers the pres­ence of major-route ACAs at

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/122/1851605/122how.pdf by guest on 13 December 2021


ABL activ­ity, which results in not only con­tin­ued pro­lif­er­a­tion of diag­no­sis as diag­nos­tic for AP-CML.8 The WHO has also recently
leu­ke­mic cells but fur­ther genetic insta­bil­ity and DNA dam­age. included pro­vi­sional cri­te­ria based on ini­tial response to TKIs (Table
This invari­ably leads to clonal evo­lu­tion and muta­tions both in­ 1), which will require fur­ther val­i­da­tion in pro­spec­tive tri­als.8
side and out­side the BCR-ABL kinase domain, as well as addi­ The vary­ing clas­si­fi­ca­tion sys­tems and their def­i­ni­tions of
tional chro­mo­somal abnor­mal­i­ties (ACAs). This review focuses advanced CML must be kept in mind when interpreting and apply­
on treat­ment con­sid­er­ations for patients who have or prog­ress ing trial results to indi­vid­ual patients. The MDACC and IBMTR cri­
to AP- and BP-CML, includ­ing TKI selec­tion, con­sid­er­ation, and te­ria are more fre­quently used as eli­gi­bil­ity cri­te­ria in clin­i­cal tri­als,
tim­ing of hema­to­poi­etic stem cell trans­plan­ta­tion (HSCT), and and National Comprehensive Cancer Network (NCCN) guide­lines
emerg­ing ther­a­pies. dis­fa­vor use of the WHO clas­si­fi­ca­tion sys­tem for this rea­son.9
Overall, the lack of uni­for­mity seen in these major clas­si­fi­ca­tion
Definition and epi­de­mi­­ol­ogy of advanced phase CML sys­tems empha­sizes a clin­i­cal spec­trum within each phase of the
The over­all inci­dence of AP- and BP-CML at diag­no­sis is 3.5% dis­ease. Recommendations for treat­ment in AP- or BP-CML are
and 2.2%,2 respec­tively, and with the intro­duc­tion of TKIs, the there­fore rarely one-size-fits-all­.
num­ber has decreased sig­nif­i­cantly. Long-term fol­low-up of the
International Randomized Study of Interferon and STI571 trial Treatment of AP-CML
dem­on­strated 6.9% cumu­la­tive pro­gres­sion to AP- or BP-CML The ini­tial goal of ther­apy in advanced phase CML is to revert to a
after 10 years.3 This num­ber is lower in recent long-term stud­ CP or a remis­sion prior to HSCT. The hema­to­logic and com­plete
ies, likely due to improve­ments in the man­age­ment of patients cyto­ge­netic responses (CCyRs) to the var­i­ous TKIs in advanced
with CP-CML with an inad­e­quate response.4 A lower inci­dence of CML are sum­ma­rized in Table 2.

Table 1.  Major clas­si­fi­ca­tion sys­tems used in chronic mye­loid leu­ke­mia

MDACC IBMTR Euro­pean LeukemiaNet WHO


Accelerated phase
  PB blasts 15%-29% PB or BM blasts 10%-29% PB or BM blasts 15%-29% PB or BM blasts 10%-19%
  PB blasts + promyelocytes ≥30% BP blasts + promyelocytes >20% PB blasts + promyelocytes ≥30%
  PB baso­phils ≥20% PB baso­phils ≥20% PB baso­phils ≥20% PB baso­phils ≥20%
  Platelets ≤100 × 109/L (unre­lated Platelets ≤100 × 109/L (unre­lated Platelets ≤100 × 109/L (unre­lated Platelets ≤100 × 109/L (unre­lated to
to ther­apy) to ther­apy) or >1000 × 109/L to ther­apy) ther­apy) or >1000 × 109/L
  Splenomegaly (unre­spon­sive to (unre­spon­sive to ther­apy) (unre­spon­sive to ther­apy)
ther­apy) Anemia Hb <8 g/dL (unre­spon­sive Splenomegaly (unre­spon­sive to
to ther­apy) ther­apy)
Splenomegaly (unre­spon­sive to
ther­apy)
  Cytogenetic evo­lu­tion on Cytogenetic evo­lu­tion on treat­ Cytogenetic evo­lu­tion on ACA/Ph+ major route, com­plex
treat­ment ment treat­ment kar­yo­type, or 3q26.2 abnor­mal­i­ties,
at diag­no­sis
Cytogenetic evo­lu­tion on treat­ment
Provisional: fail­ure to achieve CHR to
first TKI; any indi­ca­tion of resis­tance
to 2 sequen­tial TKIs; occur­rence of
>2 muta­tions on BCR-ABL dur­ing TKI
Blast phase
  PB or BM blasts ≥30% PB or BM blasts ≥20%
  Extramedullary blast pro­lif­er­a­tion Extramedullary blast pro­lif­er­a­tion

Prakash Singh Shekhawat


BM, bone mar­row; Hb, hemo­glo­bin; PB, periph­eral blood.

Advanced phase CML: room for improve­ment | 123


Table 2.  Summary of hema­to­logic and cyto­ge­netic responses to TKI in advanced phase CML*

CHR CCyR MMR OS


AP BP AP BP AP AP BP
Imatinib 70%- > 90% 11%-35% 16%-60% 0%-10% 19%-63% 50%-60% at 5 years 7-10 months†
Nilotinib (de novo) >90% — 80%-90% — 70% 90% at 3 years
Nilotinib (progressed) 22%-46% 21%-42% ‡
0%-21% 14%-38% §
10% 60%-70% at 2 years 32% at 2 years
Dasatinib (de novo) >90% — 80%-90% — 70% 90% at 3 years
Dasatinib (progressed) 45%-52% 28%-61%‡ 18%-33% 27%-35%§ 10% 60%-70% at 2 years 30% at 2 years
Bosutinib (progressed) 57%‡ 28%‡ 40%§ 50%§ 11% 60% at 4 years 17% at 4 years
Ponatinib (progressed) 55% ‡
32% ‡
24% 18% §
34% 84% at 1 year 29% at 1 year

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/122/1851605/122how.pdf by guest on 13 December 2021


*Adapted from Bonifacio et al.10

Median OS.

Hematologic response.
§
Major cyto­ge­netic response.

In patients presenting with de novo AP-CML, responses to hema­to­logic remis­sion (CHR) rates are 10% to 20% and 20% to
TKIs are robust. Imatinib results in CCyRs of 60% to 80% and 30% for nilotinib,19 30% and 50% for dasatinib,20 and 40% and 57%
major molec­u­lar responses (MMRs) of 40% to 60%,11,12 and these for bosutinib.21 Ponatinib is a third-gen­er­a­tion BCR-ABL inhib­i­tor
responses are fur­ther improved in de novo AP patients whose that over­ comes many muta­ tions that con­fer resis­tance to ear­
only hall­mark of AP is the pres­ence of ACAs. Second-gen­er­a­tion lier-gen­er­a­tion TKIs, includ­ing the gate­keeper T315I muta­tion. The
TKIs have greater potency and BCR-ABL selec­tiv­ity com­pared Ponatinib Ph+ ALL and CML Evaluation trial enrolled 267 heavily pre­
with imatinib. CCyRs and MMRs were 90% and 76% with nilotinib treated patients with CML, includ­ing 83 patients in AP.22 Ponatinib
and dasatinib, respec­tively, in newly diag­nosed AP patients.11 resulted in CCyR and CHR rates of 24% and 55% in AP patients,
Most patients were diag­nosed with AP based on iso­lated clonal respec­ tively, with median dura­ tion of response last­ ing 12.9
evo­lu­tion or baso­philia, again dem­on­strat­ing how het­ero­ge­ne­ months.23 However, arte­rial and venous throm­bo­sis were fre­quent
ity in clas­si­fi­ca­tion sys­tems makes inter­pre­ta­tion of responses adverse events (AEs), lead­ing to dis­con­tin­u­a­tion for severe AEs in
with dif­fer­ent inter­ven­tions chal­leng­ing. Consistent with NCCN 11% of patients. The over­all cumu­la­tive inci­dence of AEs was 25%
guide­lines, we approach ini­tial man­age­ment of patients with de in the 5-year fol­low-up, includ­ing 21% seri­ous AEs.23 The Ponatinib
novo AP-CML sim­i­larly to patients with CP-CML, espe­cially if they in Participants with Resistant Chronic Phase Chronic Myeloid Leu­
have low-risk Sokal scores or iso­lated ACA/Ph+ abnor­mal­i­ties kemia to Characterize the Efficacy and Safety of a Range of Doses
as their only AP fea­ture.9 Of note, cer­tain ACA abnor­mal­i­ties are (OPTIC) trial eval­u­ated lower starting doses of ponatinib and de-
higher risk than oth­ers, which may affect treat­ment deci­sions esca­lated doses once BCR-ABL lev­els reached 1%, and lon­ger-term
(Table 3). Milestones are not well defined in AP-CML, but mon­ fol­low-up of this cohort may sup­port alter­nate dos­ing of ponatinib

tor­ing of BCR-ABL is recommended at 3-month inter­ vals, as to improve tol­er­a­bil­ity and effi­cacy.24 Omecataxine is a pro­tein syn­
in CP. Failure to achieve mile­stones as used in CP-CML should the­sis inhib­i­tor with approval by the US Food and Drug Administra­
prompt con­sid­er­ation change in ther­apy and HSCT. tion (FDA) for AP-CML resis­tant or intol­er­ant to TKIs, includ­ing the
Prognosis, how­ever, is worse in AP patients who prog­ress from T315I muta­tion. In a phase 2 trial of heavily pretreated patients with
CP while on treat­ment. For these patients, CCyRs and com­plete CML, among the 51 AP patients, 29% achieved CHR and 4% CCyR.25
These responses are less than that seen with ponatinib but can be
Table 3.  Prognostic risk fac­tors in advanced phase CML con­sid­ered in this patient pop­u­la­tion with few remaining options.
Ultimately, selec­tion of a TKI is affected by patient comorbid­
Characteristic Poor risk fac­tors ity, costs, prior treat­ment, and BCR-ABL muta­tional sta­tus. Better
Clinical13,14
Blast % (most impor­tant prog­nos­tic indi­ca­tor, def­i­ni­tions of AP-CML are needed with more refined risk strat­i­fic ­ a­
greater impact in AP com­pared with BP) tion and treat­ment indi­ca­tions, as cer­tain patients with de novo
Older age AP-CML behave sim­i­lar to those with CP-CML. Table 3 sum­ma­rizes
Anemia clin­i­cal, chro­mo­somal, and molec­u­lar risk fac­tors that, if pres­ent in
Thrombocytopenia
Basophil % AP patients, may war­rant more aggres­sive approaches. Patients
Prior TKI with AP-CML with excess blasts often need treat­ment such as BP-
Myeloid immunophenotype CML. Given the higher response rates, later-gen­er­a­tion TKIs are
Chromosomal13,15-17 +8, +Ph, i(17q), +17, +19, +21, 3q26.2, 11q23, −7/7q, pre­ferred over imatinib for de novo AP, and a switch in TKI is war­
com­plex, del17p, hyperdiploidy, chro­mo­some 15 ranted for patients progressing from CP; if appli­ca­ble, selec­tion
abnor­mal­i­ties should be made based on ABL1 kinase domain muta­tions. Pona­
Molecular15,18 TP53 tinib is the only FDA-approved TKI with activ­ity against the T315I
ASXL1 muta­tion, although it car­ries a risk of vas­cu­lar occlu­sive events.
Acquisition of new muta­tions dur­ing TKI treat­ment Although our patient did not dem­on­strate overt signs of AP-CML,
(ABL1 kinase muta­tions, TP53, KMT2D, TET2)
pro­gres­sion on imatinib and then dasatinib is concerning. Given

124  |  Hematology 2021  |  ASH Education Program


the pres­ence of the T315I muta­tion while tak­ing dasatinib, she were too small to make a defin­i­tive com­par­i­son, dasatinib led to
was switched to ponatinib for fur­ther man­age­ment. improved out­comes com­pared with imatinib. Rates of CCyR and
MMR were also sig­nif­i­cantly higher with TKI and che­mo­ther­apy
Treatment of BP-CML com­bi­na­tion.13 However, intense ther­apy may not be fea­si­ble in
Chemotherapy in addi­tion to a TKI is gen­er­ally recommended in all­patients. TKIs with hypomethylating agents have dem­on­strated
patients with BP-CML, with the type of induc­tion che­mo­ther­apy some effi­cacy and OS ben­e­fit.32,33
guided by the mye­loid or lym­phoid lin­e­age of the blasts. Induc­ Overall, BP-CML is a rare entity, and defin­ing uni­fied treat­ment
tion che­mo­ther­apy is recommended in con­junc­tion with TKI as re­ guide­lines is chal­leng­ing. However, patients with high blast bur­
sponse rates asso­ci­ated with TKI treat­ments alone are inad­e­quate. den almost always need treat­ment with stan­dard che­mo­ther­apy
Response to sin­gle-agent imatinib is sig­nif­i­cantly lower in BP-CML, in addi­tion to TKIs. For the rare patient with de novo blast cri­sis,
with CCyRs occur­ring in approx­i­ma­tely 10% of patients and median some experts con­sider use of TKI alone with close mon­i­tor­ing,
over­all sur­vival (OS) around 7 to 10 months.26 Responses are slightly although there are lit­tle data to guide this approach.10 Patients
improved with sec­ond gen­er­a­tion TKIs.27,28 In the Ponatinib Ph+ ALL who are unfit for che­mo­ther­apy and with lower blast bur­den may
and CML Evaluation trial, ponatinib dem­on­strated an 18% CCyR in also be can­di­dates for TKI alone with close mon­i­tor­ing. In the

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/122/1851605/122how.pdf by guest on 13 December 2021


62 patients with BP-CML and a median dura­tion of response of 6 case of our patient, she was treated with stan­dard 7 + 3 induc­tion
months.22 Of note, nilotinib is not FDA approved for BP-CML. che­mo­ther­apy and ponatinib given the T315I muta­tion. We also
Multiple che­mo­ther­apy reg­i­mens have been explored in com­bi­ use higher doses of TKI for BP-CML com­pared with CP-CML, con­
na­tion with TKIs in small, ret­ro­spec­tive stud­ies.13 Cytarabine-based sis­tent with FDA label­ing indi­ca­tions.
reg­i­mens, includ­ing 7-day con­tin­u­ous infu­sion of cytarabine
200 mg/m2 and dau­no­ru­bi­cin 60 mg/m2 on days 1 to 3 (7 + 3)29 HSCT in advanced CML
and fludarabine, cytarabine, granulocyte col­ony-stim­u­lat­ing fac­ HSCT is an impor­tant man­age­ment tool in advanced CML given
tor, and idarubicin, are com­monly used for CML in mye­loid blast poorer responses and inad­e­quate response dura­bil­ity to ini­tial
cri­sis.30 Hyperfractionated cyclo­phos­pha­mide, vin­cris­tine, doxo­ ther­apy.34 For BP-CML espe­cially, most long-term sur­vi­vors are
ru­bi­cin, and dexa­meth­a­sone in com­bi­na­tion with dasatinib has trans­plant recip­i­ents. Recent anal­y­sis dem­on­strates 5-year OS
dem­on­strated effi­cacy in lym­phoid blast cri­sis.31 Cytogenetic rates of more than 90% for patients under­go­ing trans­plan­ta­tion
responses of approx­i­ma­tely 30% are seen with mye­loid induc­ in CP.35 This decreases to 60% in patients who undergo trans­
tion reg­i­mens in com­bi­na­tion with imatinib.29 A ret­ro­spec­tive plan­ta­tion in AP/BP.35 Patients who have trans­plan­ta­tion after
anal­y­sis of 477 patients with BP-CML treated with che­mo­ther­apy, achieve­ment of a sec­ond CP do sig­nif­i­cantly worse com­pared
TKI and che­mo­ther­apy, or non-TKI-based ther­apy dem­on­strated with patients who undergo trans­plan­ta­tion in their first CP. The
that patients treated with TKI in com­bi­na­tion with che­mo­ther­apy choice of myeloablative vs reduced-inten­sity con­di­tion­ing is un­
had supe­rior sur­vival com­pared with TKI alone or non-TKI-based clear, although the lat­ter may be a rea­son­able alter­na­tive as it
ther­apy (5-year OS, 30% vs 14% vs 9%).13 Although the num­bers offers sim­i­lar sur­vival rates at the cost of ear­lier posttransplant

Table 4.  Novel ther­a­pies in advanced CML under inves­ti­ga­tion

Drug class Clinical tri­als N Response


Asciminib (BCR-ABL TKI) Hughes et al (phase 1—asciminib in CP/AP CML after TKI fail­ure)
41
N = 9 (AP) CHR 8/9 (AP)
NCT02081378 (phase 1—asciminib ± TKI in CP/AP/BP CML after TKI MMR 1/9 (AP)
fail­ure)
NCT03595917 (phase 1—asciminib + dasatinib + pred­ni­sone in Ph+
ALL/CML lym­phoid BP)
HQP1351(BCR-ABL TKI) Jiang et al43 (phase 2—HQP1351 in T315I-mutated CP/AP CML) N = 23 (AP) MHR 78% (AP)
MCyR 52% (AP)
K0706 (BCR-ABL TKI) NCT02629692 (phase 1/2—K0706 in AP/CP/BP CML after TKI fail­ure)
PF-114 (BCR-ABL TKI) Turkina et al44 (phase 1—PF-114 CP/AP CML after TKI fail­ure or T315I) N = 51 (CP/AP) MHR 42% (CP/AP)
MCyR 29% (CP/AP)
PHA-739358 (aurora kinase inhib­i­tor) Borthakur et al45 (phase 1—PHA-739358 in AP/BP CML) N = 29 (AP/BP) HR 14% (AP/BP)
SCH 6636 (farnesyl trans­fer­ase inhib­i­tor) Cortes et al (phase 1—SCH 6636 CP/AP/BP CML after imatinib
46
N = 15 (AP/BP) CHR 14% (AP/BP)
fail­ure)
Venetoclax (BCL2 inhib­i­tor) Maiti et al42 (ret­ro­spec­tive—venetoclax + TKI) N = 9 (BP) ORR 75% (BP)
BP1001 (lipo­so­mal Grb-2 anti­sense Ohanian et al47 (phase 1/1b—BP1001 ± low-dose cytarabine in N = 5 (BP) ORR 1/5 (BP)
oli­go­nu­cle­o­tide) advanced mye­loid malig­nan­cies)
Nivolumab (anti–PD-1) NCT02011945 (phase 1b—nivolumab + dasatinib in CP/AP CML)
Inotuzumab (anti-CD22) Jain et al48 (phase 1/2—inotuzumab+bosutinib in Ph+ ALL/CML lym­ N = 2 (BP) ORR 1/2 (BP)
phoid BP)
BCL2, B-cell lym­phoma 2; HR, hema­to­logic response; MCyR, major cyto­ge­netic response; MHR, major hema­to­logic response; ORR, over­all
response rate.
Prakash Singh Shekhawat
Advanced phase CML: room for improve­ment | 125
Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/122/1851605/122how.pdf by guest on 13 December 2021
Figure 1.  Algorithm for treatment of (A) de novo or (B) progressed advanced phase CML.

relapse.36 Overall 5-year OS rates were 53% in both myeloabla­ relapse in CML.39 However, a recent IBMTR anal­y­sis of main­te­
tive and reduced-inten­sity con­di­tion­ing groups. nance TKI after trans­plant for CML showed no ben­e­fit at 100 days
For AP patients, how­ever, tim­ing of trans­plant can be dif­fi­ in terms of OS, leu­ke­mia-free sur­vival, relapse rates, trans­plant-
cult. Certain de novo AP patients have out­comes sim­i­lar to CP- related mor­tal­ity, and chronic graft-ver­sus-host dis­ease.40 NCCN
CML, with 1 study suggesting that trans­plant can be deferred guide­lines rec­om­mend 12 months of main­te­nance TKI, although
in AP patients who lack high-risk char­ac­ter­is­tics, includ­ing total data guid­ing this rec­om­men­da­tion are unclear.9 Minimal resid­ual
CML dis­ease dura­tion more than 12 months, hemo­glo­bin less dis­ease mon­i­tor­ing is an essen­tial tool to deter­mine dura­tion and
than 10 g/dL, and periph­eral blood blasts more than 5%.37 For need for TKI main­te­nance, although firm guide­lines in this area
most de novo AP patients, treat­ment with TKI alone is likely suf­ are needed. Our patient was offered trans­plant after reverting to
fi­cient, with sub­se­quent trans­plan­ta­tion deci­sions based on the CP with ponatinib and induc­tion che­mo­ther­apy. She discontin­
patients’ response to ther­apy. However, HSCT is recommended ued ponatinib at 6 months given its side effect pro­file and lack of
in all­eli­gi­ble patients who have progressed to AP from CP and all­ resid­ual dis­ease on fol­low-up.
BP patients, includ­ing those presenting with de novo dis­ease.9
Once a patient has under­gone HSCT, the use and dura­tion of Novel ther­a­peu­tics for advanced CML
TKI main­te­nance after trans­plan­ta­tion are unknown. In Ph+ acute Despite dra­matic improve­ments in out­comes for patients with
lym­pho­blas­tic leu­ke­mia, main­te­nance TKI improves leu­ke­mia- CP-CML, patients with AP- and BP-CML have sig­nif­i­cantly worse
free sur­vival, relapse, and OS.38 Smaller pro­spec­tive stud­ies have prog­no­sis, and thus new ther­a­peu­tic approaches are needed.
dem­ on­ strated safety of main­ te­nance TKI with lower rates of Table 4 sum­ma­rizes novel ther­a­peu­tics cur­rently under inves­ti­ga­

126  |  Hematology 2021  |  ASH Education Program


tion. Asciminib, an allo­ste­ric inhib­i­tor that binds to the myristoyl 4. Cortes JE, Gambacorti-Passerini C, Deininger MW, et al. Bosutinib ver­sus
site of the BCR-ABL tyro­sine kinase, has dem­on­strated effi­cacy imatinib for newly diag­nosed chronic mye­loid leu­ke­mia: results from the
ran­dom­ized BFORE trial. J Clin Oncol. 2018;36(3):231-237.
in heavily pretreated patients with CML.41 In 9 patients with AP- 5. Speck B, Bortin MM, Champlin R, et al. Allogeneic bone-mar­row trans­plan­
CML, 8 had a CHR, and 1 of 9 had an MMR. As asciminib tar­gets a ta­tion for chronic mye­log­e­nous leu­kae­mia. Lancet. 1984;1(8378):665-668.
dis­tinct bind­ing site com­pared with other TKIs, its devel­op­ment 6. Kantarjian HM, Dixon D, Keating MJ, et al. Characteristics of accel­er­ated dis­
and recent approval open the pos­si­bil­ity for future TKI-TKI drug ease in chronic mye­log­e­nous leu­ke­mia. Cancer. 1988;61(7):1441-1446.
7. Baccarani M, Deininger MW, Rosti G, et al. Euro­pean LeukemiaNet rec­om­
com­bi­na­tions, which are being eval­u­ated in phase 1 and 2 tri­als
men­da­tions for the man­age­ment of chronic mye­loid leu­ke­mia: 2013. Blood.
(NCT03595917; NCT03578367). Other novel small-mol­e­cule inhib­ 2013;122(6):872-884.
i­tors, includ­ing newer-gen­er­a­tion TKI inhib­i­tors, are also under 8. Arber DA, Orazi A, Hasserjian R, et al. The 2016 revi­sion to the World Health
eval­u­a­tion (Table 4). Venetoclax, a selec­tive B-cell lym­phoma 2 Organization clas­si­fi­ca­tion of mye­loid neo­plasms and acute leu­ke­mia.
inhib­i­tor, has shown syn­er­gism with TKIs in elim­i­nat­ing leu­ke­mic Blood. 2016;127(20):2391-2405.
9. Network NCC. Myeloproliferative neo­plasms (ver­sion 3.2021). Accessed
stem cells in advanced CML, with encour­ag­ing response rates 31 Jan­u­ary 2021. https:​­/​­/www​­.nccn​­.org​­/professionals​­/physician_gls​­/pdf​­/
for venetoclax and TKI com­bi­na­tions in ret­ro­spec­tive stud­ies for mpn_blocks​­.pdf.
CML in mye­loid BP.42 Given dys­func­tion in immune sur­veil­lance 10. Bonifacio M, Stagno F, Scaffidi L, Krampera M, Di Raimondo F. Management

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/122/1851605/122how.pdf by guest on 13 December 2021


within CML, immune ther­a­pies could poten­tially be lev­er­aged in of chronic mye­loid leu­ke­mia in advanced phase. Front Oncol. 2019;9:1132.
11. Ohanian M, Kantarjian HM, Quintas-Cardama A, et al. Tyrosine kinase inhib­
advanced phase CML, par­tic­u­larly in TKI-resis­tant patients. Over­
i­tors as ini­tial ther­apy for patients with chronic mye­loid leu­ke­mia in accel­
all, treat­ment of advanced phase CML remains a chal­lenge, and er­ated phase. Clin Lymphoma Myeloma Leuk. 2014;14(2):155-162.e1162e1.
pre­ven­tion of dis­ease pro­gres­sion is the most par­a­mount strat­ 12. Rea D, Etienne G, Nicolini F, et al. First-line imatinib mesylate in patients
egy. Clinical trial par­tic­i­pa­tion is encour­aged for all­patients who with newly diag­nosed accel­er­ated phase-chronic mye­loid leu­ke­mia. Leu-
develop or have advanced phase CML. kemia. 2012;26(10):2254-2259.
13. Jain P, Kantarjian HM, Ghorab A, et al. Prognostic fac­tors and sur­vival out­
comes in patients with chronic mye­loid leu­ke­mia in blast phase in the tyro­
Conclusions sine kinase inhib­i­tor era: cohort study of 477 patients. Cancer. 2017;123(22):​
TKI ther­apy rev­o­lu­tion­ized the care of patients with CML. Al­ 4391-4402.
though the inci­dence of AP- and BP-CML has declined, prog­no­sis 14. Lauseker M, Bachl K, Turkina A, et  al. Prognosis of patients with chronic
is sig­nif­i­cantly worse, and TKI ther­apy is not as effec­tive. In addi­ mye­loid leu­ke­mia presenting in advanced phase is defined mainly by blast
count, but also by age, chro­mo­somal aber­ra­tions and hemo­glo­bin. Am J
tion to small num­bers and lim­ited evi­dence to guide treat­ment, Hematol. 2019;94(11):1236-1243.
it is impor­tant to rec­og­nize that there is het­ero­ge­ne­ity in the 15. Ochi Y, Yoshida K, Huang Y-J, et al. Clonal evo­lu­tion and clin­i­cal impli­ca­
def­i­ni­tions of AP and BP, com­pli­cat­ing treat­ment deci­sions. Our tions of genetic abnor­mal­i­ties in blastic trans­for­ma­tion of chronic mye­loid
gen­eral approach to patients with advanced CML is sum­ma­rized leu­kae­mia. Nat Commun. 2021;12(1):2833.
16. Wang W, Cortes JE, Lin P, et  al. Clinical and prog­nos­tic sig­nif­i­cance of
in Figure 1. Given lim­ited options in advanced CML, pre­ven­tion of
3q26.2 and other chro­mo­some 3 abnor­mal­i­ties in CML in the era of tyro­
pro­gres­sion with strict adher­ence to evi­dence-based treat­ment sine kinase inhib­i­tors. Blood. 2015;126(14):1699-1706.
guide­lines for CP-CML remains the best strat­egy; how­ever, novel 17. Hehlmann R, Voskanyan A, Lauseker M, et al; SAKK and the Ger­man CML
ther­a­peu­tics and treat­ment strat­e­gies are being explored for the Study Group. High-risk addi­tional chro­mo­somal abnor­mal­i­ties at low blast
small group of patients who do trans­form. counts her­ald death by CML. Leukemia. 2020;34(8):2074-2086.
18. Kim T, Tyndel MS, Kim HJ, et  al. Spectrum of somatic muta­tion dynam­
ics in chronic mye­loid leu­ke­mia fol­low­ing tyro­sine kinase inhib­i­tor ther­apy.
Conflict-of-inter­est dis­clo­sure Blood. 2017;129(1):38-47.
Joan How: no conflicts of interest. 19. Nicolini FE, Masszi T, Shen Z, et al. Expanding Nilotinib Access in Clinical
Vinayak Venkataraman: no conflicts of interest. Trials (ENACT), an open-label mul­ti­cen­ter study of oral nilotinib in adult
patients with imatinib-resis­tant or -intol­er­ant chronic mye­loid leu­ke­mia in
Gabriela Soriano Hobbs: Scientific Advisory Board (SAB) for No­
accel­er­ated phase or blast cri­sis. Leuk Lymphoma. 2012;53(5):907-914.
vartis, Celgene/BMS, Constellation, AbbVie, Blueprint Medicines, 20. Kantarjian H, Cortes J, Kim DW, et  al. Phase 3 study of dasatinib 140mg
and Incyte. Research sup­port from Constellation and Incyte. once daily ver­sus 70mg twice daily in patients with chronic mye­loid leu­
ke­mia in accel­er­ated phase resis­tant or intol­er­ant to imatinib: 15-month
Off-label drug use median fol­low-up. Blood. 2009;113(25):6322-6329.
21. Gambacorti-Passerini C, Kantarjian HM, Kim D-W, et  al. Long-term effi­
Joan How: nothing to disclose.
cacy and safety of bosutinib in patients with advanced leu­ke­mia fol­low­ing
Vinayak Venkataraman: nothing to disclose. resis­tance/intol­er­ance to imatinib and other tyro­sine kinase inhib­i­tors. Am
Gabriela Soriano Hobbs: nothing to disclose. J Hematol. 2015;90(9):755-768.
22. Cortes JE, Kim D-W, Pinilla-Ibarz J, et al; PACE Investigators. A phase 2 trial
Correspondence of ponatinib in Philadelphia chro­mo­some-pos­i­tive leu­ke­mias. N Engl J Med.
2013;369(19):1783-1796.
Gabriela Soriano Hobbs, Zero Emerson Office 138, Bos­ton, MA
23. Cortes JE, Kim D-W, Pinilla-Ibarz J, et  al. Ponatinib effi­cacy and safety in
02114; e-mail: ghobbs@part­ners​­.org. Philadelphia chro­ mo­some-pos­ i­
tive leu­ ke­
mia: final 5-year results of the
phase 2 PACE trial. Blood. 2018;132(4):393-404.
References 24. Kantarjian H, Deininger M, Abruzzese E, et al. Efficacy and safety of pona­
1. Shallis RM, Wang R, Davidoff A, Ma X, Podoltsev NA, Zeidan AM. Epidemi­ tinib (PON) in patients with chronic-phase chronic mye­loid leu­ke­mia (CP-
ology of the clas­si­cal mye­lo­pro­lif­er­a­tive neo­plasms: the four cor­ners of an CML) who failed one or more sec­ ond-gen­ er­
a­tion (2G) tyro­ sine kinase
expan­sive and com­plex map. Blood Rev. 2020;42:100706. inhib­i­tors (TKIs): ana­ly­ses based on PACE and optic. Blood. 2020;136:43-44.
2. Hoffmann VS, Baccarani M, Hasford J, et al. The EUTOS pop­u­la­tion-based reg­is­ 25. Khoury HJ, Cortes J, Baccarani M, et  al. Omacetaxine mepesuccinate in
try: inci­dence and clin­i­cal char­ac­ter­is­tics of 2904 CML patients in 20 Euro­pean patients with advanced chronic mye­loid leu­ke­mia with resis­tance or intol­
Countries. Leukemia. 2015;29(6):1336-1343. er­ance to tyro­sine kinase inhib­i­tors. Leuk Lymphoma. 2015;56(1):120-127.
3. Hochhaus A, Larson RA, Guilhot F, et al; IRIS Investigators. Long-term out­ 26. Sawyers CL, Hochhaus A, Feldman E, et al. Imatinib induces hema­to­logic and
comes of imatinib treat­ment for chronic mye­loid leu­ke­mia. N Engl J Med. cyto­ge­netic responses in patients with chronic mye­log­e­nous leu­ke­mia in
2017;376(10):917-927. mye­loid blast cri­sis: results of a phase II study. Blood. 2002;99(10):3530-3539.

Prakash Singh Shekhawat


Advanced phase CML: room for improve­ment | 127
27. Nicolini FE, Basak GW, Kim D-W, et  al. Overall sur­ vival with ponatinib 39. Olavarria E, Siddique S, Griffiths MJ, et al. Posttransplantation imatinib as
ver­sus allo­ge­neic stem cell trans­plan­ta­tion in Philadelphia chro­mo­some- a strat­egy to post­pone the require­ment for immu­no­ther­apy in patients
pos­i­tive leu­ke­mias with the T315I muta­tion. Cancer. 2017;123(15):2875- under­go­ing reduced-inten­sity allo­grafts for chronic mye­loid leu­ke­mia.
2880. Blood. 2007;110(13):4614-4617.
28. Saglio G, Hochhaus A, Goh YT, et  al. Dasatinib in imatinib-resis­ tant or 40. DeFilipp Z, Ancheta R, Liu Y, et al. Maintenance tyro­sine kinase inhib­it­ors
imatinib-intol­er­ant chronic mye­loid leu­ke­mia in blast phase after 2 years fol­low­ing allo­ge­neic hema­to­poi­etic stem cell trans­plan­ta­tion for chronic
of fol­low-up in a phase 3 study: effi­cacy and tol­er­a­bil­ity of 140 mil­li­grams mye­log­e­nous leu­ke­mia: a cen­ter for inter­na­tional blood and mar­row trans­
once daily and 70 mil­li­grams twice daily. Cancer. 2010;116(16):3852-3861. plant research study. Biol Blood Marrow Transplant. 2020;26(3):472-479.
29. Deau B, Nicolini FE, Guilhot J, et al. The addi­tion of dau­no­ru­bi­cin to imatinib 41. Hughes TP, Mauro MJ, Cortes JE, et al. Asciminib in chronic mye­loid leu­ke­
mesylate in com­bi­na­tion with cytarabine improves the response rate and mia after ABL kinase inhib­i­tor fail­ure. N Engl J Med. 2019;381(24):2315-2326.
the sur­vival of patients with mye­loid blast cri­sis chronic mye­log­e­nous leu­ 42. Maiti A, Rausch CR, Cortes JE, et al. Outcomes of relapsed or refrac­tory acute
ke­mia (AFR01 study). Leuk Res. 2011;35(6):777-782. mye­loid leu­ke­mia after front­line hypomethylating agent and venetoclax reg­
30. Milojkovic D, Ibrahim A, Reid A, Foroni L, Apperley J, Marin D. Efficacy of i­mens. Haematologica. 2021;106(3):894-898.
com­bin­ing dasatinib and FLAG-IDA for patients with chronic mye­loid leu­ 43. Jiang Q , Huang X, Chen Z, et al. Novel BCR-ABL1 tyro­sine kinase inhib­i­tor
ke­mia in blastic trans­for­ma­tion. Haematologica. 2012;97(3):473-474. (TKI) HQP1351 (olverembatinib) is effi­ca­cious and well tol­er­ated in patients
31. Strati P, Kantarjian H, Thomas D, et al. HCVAD plus imatinib or dasatinib in with T315I-mutated chronic mye­ loid leu­ke­
mia (CML): results of piv­ otal
lym­phoid blastic phase chronic mye­loid leu­ke­mia. Cancer. 2014;120(3):373- (phase II) tri­als. Blood. 2020;136(suppl 1):50-51.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/122/1851605/122how.pdf by guest on 13 December 2021


380. 44. Turkina AG, Vinogradova O, Lomaia E, et al. Phase-1 study of PF-114 mesylate
32. Ghez D, Micol J-B, Pasquier F, et al. Clinical effi­cacy of sec­ond gen­er­a­tion in CML fail­ing prior tyro­sine kinase-inhib­i­tor ther­apy. Blood. 2018;132(suppl
tyro­sine kinase inhib­i­tor and 5-azacytidine com­bi­na­tion in chronic mye­ 1):790.
log­e­nous leu­kae­mia in mye­loid blast cri­sis. Eur J Cancer. 2013;49(17):3666- 45. Borthakur G, Dombret H, Schafhausen P, et al. A phase I study of danusertib
3670. (PHA-739358) in adult patients with accel­er­ated or blastic phase chronic
33. Ruggiu M, Oberkampf F, Ghez D, et al. Azacytidine in com­bi­na­tion with tyro­ mye­loid leu­ke­mia and Philadelphia chro­mo­some-pos­i­tive acute lym­pho­
sine kinase inhib­i­tors induced dura­ble responses in patients with advanced blas­tic leu­ke­mia resis­tant or intol­er­ant to imatinib and/or other sec­ond
phase chronic mye­log­e­nous leu­ke­mia. Leuk Lymphoma. 2018;59(7):1659- gen­er­a­tion c-ABL ther­apy. Haematologica. 2015;100(7):898-904.
1665. 46. Cortes J, Jabbour E, Daley GQ , et al. Phase 1 study of lonafarnib (SCH 66336)
34. Hehlmann R, Lauseker M, Jung-Munkwitz S, et  al. Tolerability-adapted and imatinib mesylate in patients with chronic mye­loid leu­ke­mia who have
imatinib 800 mg/d ver­sus 400 mg/d ver­sus 400 mg/d plus inter­feron-α in failed prior sin­gle-agent ther­apy with imatinib. Cancer. 2007;110(6):1295-
newly diag­nosed chronic mye­loid leu­ke­mia. J Clin Oncol. 2011;29(12):1634- 1302.
1642. 47. Ohanian M, Tari Ashizawa A, Garcia-Manero G, et  al. Liposomal Grb2
35. Saussele S, Lauseker M, Gratwohl A, et al; Ger­man CML Study Group. Allo­ anti­ sense oligodeoxynucleotide (BP1001) in patients with refrac­ tory or
geneic hema­to­poi­etic stem cell trans­plan­ta­tion (allo SCT) for chronic mye­ relapsed haematological malig­nan­cies: a sin­gle-cen­tre, open-label, dose-
loid leu­ke­mia in the imatinib era: eval­u­a­tion of its impact within a sub­group esca­la­tion, phase 1/1b trial. Lancet Haematol. 2018;5(4):e136-e146.
of the ran­dom­ized Ger­man CML Study IV. Blood. 2010;115(10):1880-1885. 48. Jain N, Maiti A, Ravandi F, et al. Inotuzumab ozogamicin with bosutinib for
36. Chhabra S, Ahn KW, Hu Z-H, et al. Myeloablative vs reduced-inten­sity con­ relapsed or refrac­tory Philadelphia chro­mo­some pos­i­tive acute lym­pho­
di­tion­ing allo­ge­neic hema­to­poi­etic cell trans­plan­ta­tion for chronic mye­ blas­tic leu­ke­mia or lym­phoid blast phase of chronic mye­loid leu­ke­mia. Am
loid leu­ke­mia. Blood Adv. 2018;2(21):2922-2936. J Hematol. 2021;96(8):1000-1007.
37. Jiang Q , Xu L-P, Liu D-H, et al. Imatinib mesylate ver­sus allo­ge­neic hema­
to­poi­etic stem cell trans­plan­ta­tion for patients with chronic mye­log­e­nous
leu­ke­mia in the accel­er­ated phase. Blood. 2011;117(11):3032-3040.
38. Brissot E, Labopin M, Beckers MM, et al. Tyrosine kinase inhib­i­tors improve
long-term out­come of allo­ge­neic hema­to­poi­etic stem cell trans­plan­ta­tion
for adult patients with Philadelphia chro­mo­some pos­i­tive acute lym­pho­ © 2021 by The Amer­i­can Society of Hematology
blas­tic leu­ke­mia. Haematologica. 2015;100(3):392-399. DOI 10.1182/hema­tol­ogy.2021000240

128  |  Hematology 2021  |  ASH Education Program


COAGULATION LABORATORY POTPOURRI

Direct oral anticoagulant (DOAC) interference


in hemostasis assays

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/129/1851777/129moser.pdf by guest on 13 December 2021


Karen A. Moser1 and Kristi J. Smock2
Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT; and 2ARUP Institute for Clinical and Experimental Pathology,
1

Salt Lake City, UT

Direct oral anticoagulants (DOACs) are a group of direct coagulation factor inhibitors including both direct thrombin
inhibitors and direct factor Xa inhibitors. These medications may cause hemostasis assay interference by falsely increas-
ing or decreasing measured values, depending on the analyte. Considering the potential for DOAC interference in a vari-
ety of hemostasis assays is essential to avoid erroneous interpretation of results. Preanalytic strategies to avoid DOAC
interference include selecting alternatives to clot-based hemostasis assays in patients taking DOACs when possible and
sample collection timed when the patient is off anticoagulant therapy or at the expected drug trough. Clinical labora-
tories may also provide educational materials that clearly describe possible interferences from DOAC, develop testing
algorithms to aid in detection of DOAC in submitted samples, use DOAC-neutralizing agents to remove DOACs before
continuing with testing, and write interpretive comments that explain the effects of DOAC interference in hemostasis
tests. Using a combination of the described strategies will aid physicians and laboratorians in correctly interpreting
hemostasis and thrombosis laboratory tests in the presence of DOACs.

LEARNING OBJECTIVES
• Describe the patterns of interference in hemostasis assays due to direct thrombin inhibitors and direct Xa inhibi­
tors
• List potential strategies physicians and clinical laboratories can use to decrease DOAC interference in hemostasis
assays

Since the introduction of the direct oral anticoagulants embolism, prompting testing for lupus anticoagulant (LA)
(DOACs) in the 2010s, clinical laboratories have struggled and treatment with rivaroxaban. Laboratory results for
to mitigate the effects of these drugs in clot­based hemo­ both her initial LA profile as well as follow­up testing 12
stasis and thrombosis assays. DOACs are a group of di­ weeks later are included in Table 2.
rect coagulation factor inhibitors that include both direct Case 2: A 37­year­old man sought treatment for an
thrombin inhibitors (dabigatran) and direct Xa inhibitors unprovoked pulmonary embolism and was evaluated for
(rivaroxaban, apixaban, edoxaban).1 The DOACs may cause thrombophilia risk factors, including LA, while taking apix­
assay interference by falsely increasing or decreasing mea­ aban therapy. Laboratory results for his initial LA panel are
sured values, depending on the analyte. Data from both included in Table 2. An anti­Xa activity assay calibrated for
individual laboratory studies and external quality assess­ unfractionated heparin (UFH) showed measurable anti­Xa
ment programs describe expected patterns with a variety activity in the plasma sample submitted for the LA profile.
of reagents (Table 1).1-8

Patterns of DOAC interference in hemostasis assays


For basic hemostasis assays such as prothrombin time (PT)
and activated partial thromboplastin time (aPTT), response
CLINICAL CASES to DOACs varies considerably by drug, drug concentration,
Case 1: A 54­year­old woman was diagnosed with pneu­ and reagent. In general, the aPTT may be prolonged with
monia and was bedridden due to the severity of her ill­ dabigatran but does not tend to be prolonged by direct
ness. A few weeks later, she developed a pulmonary Xa inhibitors.1,2,9 A normal aPTT is insufficient to exclude the
Prakash Singh Shekhawat
DOAC interference | 129
Table 1.  Patterns of DOAC inter­fer­ence in hemo­sta­sis/throm­bo­sis assays

Expected change Assays Notes


Clotting time pro­lon­ga­tion • aPTT (dabigatran > direct Xa inhib­i­tors) Effects on clot­ting times are reagent depen­dent.
• PT (rivaroxaban > edoxaban > apixaban) aPTT and PT mixing tests are expected to show incom­
• Thrombin time (dabigatran) plete cor­rec­tion in the pres­ence of DOACs.
False increase • Clot-based pro­tein C activ­ity False increase in pro­tein C, pro­tein S, and anti­throm­bin
• Clot-based pro­tein S activ­ity activ­i­ties may result in mis­di­ag­no­sis of a patient with
• Antithrombin activ­ity (in fac­tor IIa–based assays with true defi­ciency as nor­mal.
dabigatran, in fac­tor Xa–based assays with direct Xa Falsely ele­vated acti­vated pro­tein C resis­tance ratio may
inhib­i­tors) result in mis­di­ag­no­sis of a patient with fac­tor V Leiden
• Activated pro­tein C resis­tance ratio muta­tion as nor­mal.
False decrease • aPTT-based fac­tor assays (VIII, IX, XI, XII) Dilutions in fac­tor assays may show non­spe­cific inhib­i­tor
• PT-based fac­tor assays (II, V, VII, X) effect.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/129/1851777/129moser.pdf by guest on 13 December 2021


False pos­i­tive (or poten­tially • LA assays Includes aPTT- and DRVVT-based assays, among other
false neg­a­tive) clot­ting time-based LA assays; effects are drug and
reagent depen­dent.
No change • Clauss fibrin­o­gen activ­ity (for most reagents, rare
meth­ods show false decrease in pres­ence of high
con­cen­tra­tions of dabigatran)
• D-dimer
• Chromogenic pro­tein C activ­ity
• Free and total pro­tein S anti­gen
• Anticardiolipin, anti-β2GP1 ELISAs
• von Willebrand activ­ity and anti­gen assays
• DNA-based assays (eg, fac­tor V Leiden muta­tion, pro­
throm­bin G20210A muta­tion)

pres­ence of dabigatran.1 The PT may be prolonged with rivar­ S, and anti­throm­bin may be falsely increased in the pres­ence
oxaban and edoxaban but tends to be rel­a­tively insen­si­tive to of DOACs, which may result in a false-neg­a­tive result.2-6,11 That
the pres­ence of apixaban in all­reagents eval­u­ated to date.2,9 The is, a patient with a true defi­ciency may have a nor­mal result if
dif­fer­ence between the observed PT pro­lon­ga­tion in the clin­i­cal plasma is tested for pro­tein C, pro­tein S, or anti­throm­bin activ­ity
cases high­lights the dif­fer­en­tial effects of rivaroxaban and apix­ using clot-based assays in the pres­ence of DOACs. DOACs do
aban on the PT. Dabigatran does not tend to pro­long the PT.2,9 not affect chro­mo­genic pro­tein C activ­ity assays or total pro­tein
In mixing tests for both aPTT and PT, DOACs will appear as non­ C anti­gen assays.1,11 Protein S activ­ity assays are clot based and
spe­cific inhib­i­tors, with the inhib­it­ or effect most pro­nounced at expe­ri­ence inter­fer­ence by DOACs, whereas free and total pro­
higher drug con­cen­tra­tions.1 Fibrinogen activ­ity tends to be unaf­ tein S anti­gen assays are immu­no­as­says and do not show DOAC
fected by DOACs, par­tic­u­larly the com­monly used Clauss fibrin­ inter­fer­ence.3,4,15 Antithrombin activ­ity assays may be either fac­
o­gen assay, which uses plasma dilu­tion as well as a high con­ tor IIa based or fac­tor Xa based; the design deter­mines which
cen­tra­tion of throm­bin to con­vert patient fibrin­o­gen to fibrin.2-6 DOACs will inter­fere. Factor IIa–based assays show false ele­va­
Factitiously decreased fibrin­ og­en activ­ ity was reported by tion with direct throm­bin inhib­i­tors, whereas fac­tor Xa–based
some lab­o­ra­to­ries at higher dabigatran con­cen­tra­tions (385 and assays show false ele­va­tion with direct Xa inhib­i­tors.3-6 Activated
744 ng/mL) in an exter­nal qual­ity assess­ment pro­gram.3 D-dimer pro­tein C resis­tance assays also have the poten­tial to give false-
assays are typ­i­cally immu­no­as­says, such as enzyme-linked immu­ neg­a­tive results in the pres­ence of DOACs.11,16
no­sor­bent assay (ELISAs) or latex immu­no­as­says; these meth­ods Other clot-based hemo­sta­sis assays, such as fac­tor assays,
are not affected by the pres­ence of DOACs.1 may show decreased activ­ ity in the pres­ ence of DOACs.1,3,4,11
Among spe­cial­ized hemo­sta­sis assays, those eval­u­at­ing for Hemostasis assays based on ELISA or other immu­no­as­say meth­
thrombophilia risk fac­tors are at par­tic­u­lar risk for inter­fer­ence, ods (eg, von Willebrand fac­tor anti­gen, solid-phase antiphospho­
given that many of these assays are clot based and may be mea­ lipid antibodies) will not show inter­fer­ence by DOACs; the results
sured in patients who have throm­bo­sis and are treated with of these assays will not be affected if performed in a sam­ple from
anti­co­ag­u­lant ther­apy.10-12 The clin­i­cal cases illus­trate two typ­ a patient receiv­ing DOAC ther­apy.1 Likewise, DNA-based assays
i­cal exam­ples of how LA assays are affected by DOACs. Dilute (eg, fac­tor V Leiden muta­tion, pro­throm­bin G20210A muta­tion)
Russell viper venom time (DRVVT) and aPTT-based LA assays will be unaf­fected by the pres­ence of DOACs.
(eg, plate­let neu­tral­i­za­tion pro­ce­dure, hex­ag­o­nal phos­pho­lipid One par­tic­u­larly chal­leng­ing area where direct Xa inhib­i­tor
neu­tral­i­za­tion) may show false-pos­i­tive results in the pres­ence inter­fer­ence can limit ther­a­peu­tic mon­i­tor­ing of another drug is
of DOACs.10,12,13 False-pos­it­ive DRVVT-based test­ing appears to the spe­cial case of patients switching from a direct Xa inhib­i­tor
be a par­tic­u­lar risk with rivaroxaban, but there also appears to to ther­apy with UFH or low molec­u­lar weight hep­a­rin (LMWH).17
be risk of DRVVT false neg­a­tives for LA in sam­ples containing Early reports of effi­cacy of DOAC-Stop (D-S; Haematex Research)
apixaban.14 Just as with screen­ing aPTT and PT, the mixing test for remov­ing the mea­sur­able anti-Xa activ­ity effect of rivaroxaban
steps in DRVVT and aPTT-based LA assays may show a non­spe­ and apixaban but not hep­a­rins raise the pos­si­bil­ity that plas­mas
cific inhib­i­tor pat­tern.13 Assays for clot-based pro­tein C, pro­tein in patients transitioning between a direct Xa inhib­i­tor and UFH

130  |  Hematology 2021  |  ASH Education Program


Table 2.  LA panel results for clin­i­cal cases 1 and 2 anti­throm­bin anti­gen) do not detect rare deficiencies due to pro­
tein dys­func­tion and would not rep­re­sent appro­pri­ate sub­sti­tutes
Case 1: for func­tional assays. If a mea­sure­ment must be made, test­ing at
Case 1: LA panel Case 2: expected drug trough con­cen­tra­tion is recommended: either 12
ini­tial 12 weeks ini­tial Reference hours postdose for DOACs dosed twice daily or 24 hours post­
Test LA panel later LA panel inter­val
dose for DOACs admin­is­tered once daily.9,14 LA test sys­tems (aPTT
PT(s) 23.0 12.8 14.3 12.0-15.5 and DRVVT based) have dem­on­strated inter­fer­ence from DOACs
DRVVT screen(s) 57 35 91 33-44 even at low drug con­cen­tra­tions; there­fore, test­ing a sam­ple col­
DRVVT 1:1 mix(s) 52 NA 80 33-44 lected at an expected drug trough con­cen­tra­tion may not com­
pletely elim­i­nate the pos­si­bil­ity of inter­fer­ence.21
DRVVT con­firm(s) Negative NA Positive Negative
aPTT screen(s) 61 38 119 32-48 What can clin­i­cal lab­o­ra­to­ries do to avoid DOAC
TT(s) 15.7 NA 15.1 14.7-19.5 inter­fer­ence in hemo­sta­sis assays?

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/129/1851777/129moser.pdf by guest on 13 December 2021


Clinical lab­o­ra­to­ries like­wise have mul­ti­ple options for miti­gat­
aPTT 1:1 mix(s) 49 NA 85 32-48
ing the effects of DOAC inter­fer­ence in hemo­sta­sis and throm­
PNP Negative NA Positive Negative bo­sis assays.14 A lab­o­ra­tory can pro­vide edu­ca­tional mate­rial to
Hex phos Positive NA NA Negative phy­si­cians and other health care pro­vid­ers using the lab­o­ra­tory’s
neu­tral­i­za­tion ser­vices that include clear descrip­tions of expected pat­terns of
DRVVT con­firm, PNP, and hex phos neutralization are LA con­fir­ma­tory DOAC inter­fer­ence, sim­i­lar to Table 1.22 Laboratories may also de­
reagents containing high phos­pho­lipid con­cen­tra­tions. sign test­ing algo­rithms that allow detec­tion of inter­fer­ing anti­
hex phos neu­tral­i­za­tion, hex­ag­o­nal phase phos­pho­lipid neu­tral­i­za­tion co­ag­u­lants in plasma sam­ples in cases where a patient’s his­tory
test; NA, not applicable; PNP, plate­let neu­tral­i­za­tion pro­ce­dure;
of anti­co­ag­u­lant ther­apy is not avail­­able.11,13 In clin­i­cal case 1, the
TT, throm­bin time.
prolonged PT offered a clue to the pres­ence of an anti­co­ag­u­lant
med­i­ca­tion, either war­fa­rin or a direct Xa inhib­i­tor. Detecting
or LMWH could be treated with D-S prior to mea­sure­ment with a anti-Xa activ­ity in the sam­ple is another, more sen­si­tive way to
UFH- or LMWH-calibrated anti-Xa activ­ity assay or aPTT for ther­ iden­tify a direct Xa inhib­i­tor in a plasma sam­ple; throm­bin time
a­peu­tic mon­i­tor­ing.17-19 It is impor­tant to note that the degree to can be used to iden­tify the pres­ence of direct throm­bin inhib­i­
which acti­vated car­bon adsorbs LMWH is incom­pletely under­ tors and hep­a­rins.11,13 Laboratories should also pro­vide inter­pre­
stood, and fur­ther study in this area is needed before rou­tinely tive result com­ments that explain the poten­tial for DOAC inter­
using DOAC-neu­tral­iz­ing com­pounds in sam­ples also containing fer­ence in the assays performed, par­tic­u­larly for LA pro­files as
LMWH.19 recommended in cur­rent International Society on Thrombosis
and Haemostasis (ISTH) and Clinical and Laboratory Standards
What can phy­si­cians do to avoid DOAC inter­fer­ence Institute (CLSI) guide­lines.13,23 Some lab­o­ra­to­ries may elect not
in hemo­sta­sis assays? to com­plete test­ing for a LA in the event DOAC inter­fer­ence is
Physicians can choose from a few dif­fer­ent prac­ti­cal strat­e­gies to detected and may sim­ply issue a com­ment stat­ing that DOAC
min­i­mize DOAC inter­fer­ence in hemo­sta­sis assays. The sim­plest inter­fer­ence ren­ders results of LA test­ing uninterpretable.23
and best way to avoid DOAC inter­fer­ence is to avoid order­ing Recently, adsorbing agents that can neu­tral­ize DOAC effects
and performing hemo­ sta­
sis assays in patients tak­ ing DOACs. in plasma sam­ples in the hemo­sta­sis lab­o­ra­tory have been devel­
Another option would be to stop DOAC ther­apy for 2 to 3 days oped, although these agents are not yet FDA approved. Clinical
prior to collecting a sam­ple for hemo­sta­sis test­ing; how­ever, giv­ lab­o­ra­to­ries have exten­sive expe­ri­ence with remov­ing hep­a­rin
en the risk of throm­bo­sis with inter­rup­tion or change of treat­ effects from plasma sam­ ples using heparinase or polybrene.24
ment, this option may not be clin­i­cally fea­si­ble in most cases.11,12,20 Using DOAC-neu­tral­iz­ing agents is an attrac­tive pos­si­bil­ity that
Briefly transitioning to an anti­co­ag­u­lant with less assay inter­fer­ could be incor­ po­
rated into lab­ o­ra­
tory workflows that already
ence, such as LMWH, could also be an option but also may not use hep­a­rin neu­tral­iz­ers in a sim­i­lar fash­ion. Currently avail­­able
be prac­ti­cal or fea­si­ble in many cases. In some clin­i­cal sit­u­a­tions, DOAC-neu­tral­iz­ing agents in tab­let form include DOAC-Stop (D-S;
test­ing in patients tak­ing DOACs may be desired (eg, in patients Haematex Research) and DOAC-Remove (D-R; 5-Diagnostics
­
requir­ing indef­i­nite anticoagulation or detecting an LA in the set­ AG).11 These agents are com­posed of acti­vated car­bon-containing
ting of unpro­voked throm­bo­sis to gain infor­ma­tion about recur­ adsor­bent com­pounds. One D-S or D-R tab­let must be added to
rence risk).12 If test­ing is under­taken, phy­si­cians can con­sider both 1 mL patient plasma, with time allowed for adsorp­tion of DOACs
choice of assay and tim­ing of sam­ple col­lec­tion to decrease the and sub­se­quent cen­tri­fu­ga­tion and removal of DOAC-free plasma
pos­si­bil­ity of DOAC inter­fer­ence. For analytes in which there is a for test­ing (Figure 1).10,18,25 DOAC-spiked plas­mas that show false-
choice between a func­tional assay and an assay such as a chro­ pos­i­tive LA results gen­er­ally con­vert to neg­a­tive LA results with
mo­genic, an anti­genic, or a DNA-based assay where inter­fer­ence DRVVT- and aPTT-based test­ ing fol­low­
ing D-S treat­ ment, with
is not expected, phy­si­cians can choose the assay with­out expect­ results com­pa­ra­ble to neu­tral­i­za­tion with idarucizumab or andex­
ed inter­fer­ence. For exam­ple, using this prin­ci­ple would favor anet alfa.10,19,26,27 One tab­ let of D-S in 1  mL plasma can report­
muta­tion anal­y­sis for fac­tor V Leiden over func­tional test­ing with edly neu­tral­ize up to 708 ng/mL apixaban, 1060 ng/mL edoxaban,
acti­vated pro­tein C resis­tance assays in a patient tak­ing DOACs 1020 ng/mL rivaroxaban, and 360 ng/mL dabigatran.10 D-S or D-R
in whom thrombophilia test­ing is desired. Simply substitut­ing an­ has also been reported to decrease DOAC inter­fer­ence in chro­
other test does not work in all­cases in which alter­nate assays are mo­ genic and clot-based fac­ tor VIII activ­ ity,18,26 1-stage fac­ tor
avail­­able. Some tests that lack inter­fer­ence (such as pro­tein C or IX activ­ity,26 throm­bin gen­er­a­tion assays,28 acti­ vated pro­ tein C
Prakash Singh Shekhawat
DOAC inter­fer­ence  | 131
A. DOAC-neutralizing tablets with B. DOAC-neutralizing filter extracting
activated carbon (eg, DOAC-Stop, DOACs through noncovalent binding in
DOAC-Remove) solid phase (eg, DOAC Filter)

Filter
Plasma
Connector
1 tablet

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/129/1851777/129moser.pdf by guest on 13 December 2021


added to
1 mL
patient Filter DOAC-free
Sample Filter placed on apparatus
citrated plasma in
mixed for 5 connecter attached centrifuged
plasma microtainer
minutes on to microtainer with 15 minutes, and ready
rocking DOAC-free 600 µL platelet-poor 300 g, room for testing
mixer and plasma plasma loaded temperature
centrifuged supernatant
5 minutes removed
at 2000 g and ready
for testing

Figure 1.  DOAC-neutralizing processes with tablet-based adsorbing agents (A) and filter systems (B).

resis­tance,25,29 and anti­throm­bin activ­ity.29,30 D-S has been reported both pro­files were performed while the patient was receiv­ing
to cause decreased fac­tor activ­i­ties in treated plas­mas, rais­ing rivaroxaban. Differential diag­nos­tic con­sid­er­ations for this pat­
the pos­si­bil­ity of adsorp­tion of coag­u­la­tion fac­tors in addi­tion to tern include a tran­sient LA, dif­fer­ences in tim­ing of draw (eg,
DOACs; how­ever, D-S does not pro­duce aPTT pro­lon­ga­tion with first pro­file drawn at drug peak and sec­ond pro­file drawn at
all­reagents tested.10,31 Caution in result inter­pre­ta­tion is recom­ drug trough), or nonadherence to the rivaroxaban reg­i­men at
mended when using DOAC-neu­tral­iz­ing agents in the clin­i­cal lab­o­ the time of the sec­ond pro­file. The prolonged PT in the first LA
ra­tory as com­plete neu­tral­i­za­tion was not observed in all­cases.10,18 pro­file sug­gests one of the lat­ter 2 pos­si­bil­i­ties.
One even more recently described option, DOAC Filter (Diagnos­ Case 2: This case dem­on­strates the rel­a­tive insen­si­tiv­ity of
tica Stago), is a car­tridge containing a solid phase designed to the PT to apixaban. The mea­sur­able direct Xa activ­ity sug­gests
extract DOACs through noncovalent bind­ing when 600 µL citrated that the pos­i­tive LA results obtained were caused by apixaban
plasma is added and centri­fuged at 300 × g for 15 min­utes.32 DOAC inter­fer­ence. A repeat sam­ple col­lected when the patient is not
Filter treat­ ment resulted in unde­ tect­able lev­ els of DOACs as tak­ing apixaban, repeat sam­ ple col­ lected at expected drug
assessed by quan­ti­ta­tive assays and no change in selected hemo­ trough, or treating the cur­rent sam­ple with a DOAC-neu­tral­iz­ing
sta­sis assays (PT, aPTT, fibrin­o­gen, anti­throm­bin activ­ity, pro­tein C agent and repeat­ ing the LA pro­ file are pos­ si­ble options to
activ­ity, acti­vated pro­tein C resis­tance, LA tests) in postfiltration decrease apixaban inter­fer­ence.
sam­ples.32 Further study is needed to bet­ter describe the effi­cacy
of DOAC-neu­tral­iz­ing reagents and the risk for inad­ver­tent adsorp­
tion of other coag­u­la­tion fac­tors from plasma in the clin­i­cal lab­o­
ra­tory. It is also impor­tant to remem­ber that performing addi­tional Conclusion
assays to detect DOACs and use of neu­tral­iz­ing agents adds cost Considering the poten­tial for DOAC inter­fer­ence in a vari­ety of
and time to test­ing and requires addi­tional patient plasma. Labo­ hemo­sta­sis assays is essen­tial to avoid erro­ne­ous inter­pre­ta­tion of
ratories will need to care­fully con­sider how these meth­ods would results. Preanalytic strat­e­gies to avoid DOAC inter­fer­ence include
fit into existing test workflows. avoiding clot-based hemo­sta­sis assays in patients tak­ing DOACs
and sam­ple col­lec­tion timed when the patient is not tak­ing anti­
co­ag­u­lant ther­apy or at the expected drug trough (as opposed
to peak or ran­dom sam­ples). Strategies clin­i­cal lab­o­ra­to­ries may
employ to avoid DOAC inter­fer­ence include pro­vid­ing edu­ca­tional
CLINICAL CASE (Con­t in­u ed) mate­ri­als that clearly describe pos­si­ble inter­fer­ences from DOACs,
Case 1: This patient had an ini­tial pos­it­ ive LA pro­file with a fol­ devel­op­ing test­ing algo­rithms to aid in the detec­tion of DOACs
low-up pro­file 12 weeks later in which an LA was not detected; in sub­mit­ted sam­ples, using DOAC-neu­tral­iz­ing agents to remove

132  |  Hematology 2021  |  ASH Education Program


DOACs before con­tinu­ing with test­ing, and writ­ing inter­pre­tive update of the guide­lines for lupus anti­co­ag­u­lant detec­tion and inter­pre­ta­
com­ments that explain the effects of DOAC inter­fer­ence in hemo­ tion. J Thromb Haemost. 2020;18(11):2828-2839.
14. Favaloro EJ, Mohammed S, Curnow J, Pasalic L. Laboratory test­ing for lupus
sta­sis tests. Using a com­bi­na­tion of the described strat­e­gies will anti­co­ag­u­lant (LA) in patients tak­ing direct oral anti­co­ag­u­lants (DOACs):
aid phy­si­cians and laboratorians in cor­rectly interpreting hemo­ poten­tial for false pos­i­tives and false neg­a­tives. Pathology. 2019;51(3):292-
sta­sis and throm­bo­sis lab­o­ra­tory tests in the pres­ence of DOACs. 300.
15. Smock KJ, Plumhoff EA, Meijer P, et al. Protein S test­ing in patients with
pro­tein S defi­ciency, fac­tor V Leiden, and rivaroxaban by North Amer­i­can
Conflict-of-inter­est dis­clo­sures
Specialized Coagulation Laboratories. Thromb Haemost. 2016;116(1):50-57.
Karen A. Moser: dis­close no rel­e­vant con­flicts of inter­est. 16. Moore GW, Van Cott EM, Cutler JA, Mitchell MJ, Adcock DM; Subcommittee
Kristi J. Smock: dis­close no rel­e­vant con­flicts of inter­est. on Plasma Coagulation Inhibitors. Recommendations for clin­i­cal lab­o­ra­
tory test­ing of acti­vated pro­tein C resis­tance; com­mu­ni­ca­tion from the
Off-label drug use SSC of the ISTH. J Thromb Haemost. 2019;17(9):1555-1561.
17. Douxfils J, Adcock DM, Bates SM, et  al. 2021 Update of the International
Karen A. Moser: nothing to disclose. Council for Standardization in Haematology rec­ om­men­ da­ tions for lab­
Kristi J. Smock: nothing to disclose. o­ra­tory mea­sure­ment of direct oral anti­co­ag­u­lants. Thromb Haemost.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/129/1851777/129moser.pdf by guest on 13 December 2021


2021;121(8):1008-1020.
Correspondence 18. Platton S, Hunt C. Influence of DOAC Stop on coag­ u­
la­tion assays in
sam­ples from patients on rivaroxaban or apixaban. Int J Lab Hematol.
Karen A. Moser, ARUP Laboratories, 500 Chipeta Way, Mail Stop
2019;41(2):227-233.
115-G04, Salt Lake City, UT 84108; e-mail: karen​­.moser@hsc​­.utah​ 19. Frans G, Meeus P, Bailleul E. Resolving DOAC inter­fer­ence on aPTT, PT, and
­.edu. lupus anti­co­ag­u­lant test­ing by the use of acti­vated car­bon. J Thromb Hae-
most. 2019;17(8):1354-1362.
20. Godier A, Dincq A-S, Martin A-C, et al. Predictors of pre-pro­ce­dural con­cen­
References tra­tions of direct oral anti­co­ag­u­lants: a pro­spec­tive multicentre study. Eur
1. Gosselin RC, Adcock DM, Bates SM, et al. International Council for Standard­
Heart J. 2017;38(31):2431-2439.
ization in Haematology (ICSH) rec­om­men­da­tions for lab­o­ra­tory mea­sure­
21. Ratzinger F, Lang M, Belik S, et  al. Lupus-anti­co­ag­u­lant test­ing at NOAC
ment of direct oral anti­co­ag­u­lants. Thromb Haemost. 2018;118(3):437-450.
trough lev­els. Thromb Haemost. 2016;116(2):235-240.
2. Gosselin R, Grant RP, Adcock DM. Comparison of the effect of the anti-Xa
22. College of Amer­i­can Pathologists. Hematology and Coagulation Check-
direct oral anti­co­ag­u­lants apixaban, edoxaban, and rivaroxaban on coag­u­
list. College of Amer­i­can Pathologists; 2021.
la­tion assays. Int J Lab Hematol. 2016;38(5):505-513.
23. CLSI. Laboratory Testing for the Lupus Anticoagulant; Approved Guide-
3. Bonar R, Favaloro EJ, Mohammed S, Pasalic L, Sioufi J, Marsden K. The effect
line. CLSI doc­u­ment H60-A. Clinical and Laboratory Standards Institute;
of dabigatran on haemostasis tests: a com­pre­hen­sive assess­ment using in
2014.
vitro and ex vivo sam­ples. Pathology. 2015;47(4):355-364.
24. Harenberg J, Reichel T, Malsch R, Hirsh J, Rustagi P. Multicentric eval­u­a­
4. Bonar R, Favaloro EJ, Mohammed S, et al. The effect of the direct fac­tor Xa
tion of heparinase on aPTT, throm­bin clot­ting time and a new PT reagent
inhib­i­tors apixaban and rivaroxaban on haemostasis tests: a com­pre­hen­sive
based on recom­ bi­
nant human tis­ sue fac­tor. Blood Coagul Fibrinolysis.
assess­ment using in vitro and ex vivo sam­ples. Pathology. 2016;48(1):60-71.
1996;7(4):453-458.
5. Van Blerk M, Bailleul E, Chatelain B, et al. Influence of apixaban on com­
25. Jilma-Stohlawetz P, Lysy K, Sunder-Plassmann R, et al. Limitations of a cali­
monly used coag­u­la­tion assays: results from the Bel­gian national exter­nal
brated, quan­ti­ta­tive APC-R assay under rou­tine con­di­tions. Int J Lab Hema-
qual­ity assess­ment scheme. Int J Lab Hematol. 2017;39(4):402-408.
tol. 2021;43(2):318-323.
6. Van Blerk M, Bailleul E, Chatelain B, et  al. Influence of dabigatran and
26. Favaloro EJ, Gilmore G, Bonar R, et al. Reducing the effect of DOAC inter­
rivaroxaban on rou­tine coag­u­la­tion assays: a nation­wide Bel­gian sur­vey.
fer­ence in lab­o­ra­tory test­ing for fac­tor VIII and fac­tor IX: a com­par­a­tive
Thromb Haemost. 2015;113(1):154-164.
study using DOAC Stop and andexanet alfa to neu­ tral­
ize rivaroxaban
7. Hillarp A, Gustafsson KM, Faxälv L, et al. Effects of the oral, direct fac­tor
effects. Haemophilia. 2020;26(2):354-362.
Xa inhib­i­tor apixaban on rou­tine coag­u­la­tion assays and anti-FXa assays.
27. Baker SA, Jin J, Pfaffroth C, Vu T, Zehnder JL. DOAC–Stop in lupus anti­co­
J Thromb Haemost. 2014;12(9):1545-1553.
ag­u­lant test­ing: direct oral anti­co­ag­u­lant inter­fer­ence removed in most
8. Hillarp A, Baghaei F, Fagerberg Blixter I, et  al. Effects of the oral, direct
sam­ples. Res Pr Thromb Haemost. 2021;5(2):314-325.
fac­ tor Xa inhib­i­
tor rivaroxaban on com­ monly used coag­ u­la­
tion assays.
28. Kopatz WF, Brinkman HJM, Meijers JCM. Use of DOAC Stop for elim­i­na­
J Thromb Haemost. 2011;9(1):133-139.
tion of anti­co­ag­u­lants in the throm­bin gen­er­a­tion assay. Thromb Res.
9. Douxfils J, Ageno W, Samama C-M, et  al. Laboratory test­ing in patients
2018;170:97-101.
treated with direct oral anti­co­ag­u­lants: a prac­ti­cal guide for cli­ni­cians.
29. Favresse J, Lardinois B, Sabor L, et al. Evaluation of the DOAC-Stop® pro­ce­
J Thromb Haemost. 2018;16(2):209-219.
dure to over­come the effect of DOACs on sev­eral thrombophilia screen­ing
10. De Kesel PM, Devreese KMJ. Direct oral anti­co­ag­u­lant adsorp­tion: impact
tests. TH Open. 2018;2(2):e202-e209.
on lupus anti­co­ag­u­lant test­ing—review of the lit­er­a­ture and eval­u­a­tion on
30. Ząbczyk M, Natorska J, Kopytek M, Malinowski KP, Undas A. The effect of
spiked and patient sam­ples. J Thromb Haemost. 2020;18(8):2003-2017.
direct oral anti­co­ag­u­lants on anti­throm­bin activ­ity test­ing is abolished by
11. Siriez R, Dogné J-M, Gosselin R, Laloy J, Mullier F, Douxfils J. Comprehen­
DOAC-stop in venous throm­bo­em­bo­lism patients. Arch Pathol Lab Med.
sive review of the impact of direct oral anti­co­ag­u­lants on thrombophilia
2021;145(1):99-104.
diag­nos­tic tests: prac­ti­cal rec­om­men­da­tions for the lab­o­ra­tory. Int J Lab
31. Riva N, Vella K, Hickey K, et al. The effect of DOAC-Stop(®) on sev­eral oral
Hematol. 2021;43(1):7-20.
and par­en­teral anti­co­ag­u­lants. Int J Lab Hematol. 2021;43(4):171-175.
12. Tripodi A, Cohen H, Devreese KMJ. Lupus anti­co­ag­u­lant detec­tion in anti­
32. Sevenet PO, Cucini V, Hervé T, et al. Evaluation of DOAC Filter, a new device
coagulated patients: guid­ ance from the Scientific and Standardization
to remove direct oral anti­co­ag­u­lants from plasma sam­ples. Int J Lab Hema-
Committee for lupus anti­ co­
ag­u­
lant/antiphospholipid antibodies of the
tol. 2020;42(5):636-642.
International Society on Thrombosis and Haemostasis. J Thromb Haemost.
2020;18(7):1569-1575.
13. Devreese KMJ, de Groot PG, de Laat B, et al. Guidance from the Scientific
and Standardization Committee for lupus anti­co­ag­u­lant/antiphospholipid © 2021 by The Amer­i­can Society of Hematology
antibodies of the International Society on Thrombosis and Haemostasis: DOI 10.1182/hema­tol­ogy.2021000241

Prakash Singh Shekhawat


DOAC inter­fer­ence  | 133
WHAT DO APLASTIC ANEMIA, PNH, AND “ HYPOPLASTIC ” LEUKEMIA HAVE IN COMMON ?

The clinical and laboratory evaluation of patients


with suspected hypocellular marrow failure

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/134/1852140/134geddis.pdf by guest on 13 December 2021


Siobán Keel1 and Amy Geddis2
University of Washington, Seattle, WA; and 2Seattle Children’s Hospital, Seattle, WA
1

The overlap in clinical presentation and bone marrow features of acquired and inherited causes of hypocellular marrow
failure poses a significant diagnostic challenge in real case scenarios, particularly in nonsevere disease. The distinction
between acquired aplastic anemia (aAA), hypocellular myelodysplastic syndrome (MDS), and inherited bone marrow fail-
ure syndromes presenting with marrow hypocellularity is critical to inform appropriate care. Here, we review the workup
of hypocellular marrow failure in adolescents through adults. Given the limitations of relying on clinical stigmata or fam-
ily history to identify patients with inherited etiologies, we outline a diagnostic approach incorporating comprehensive
genetic testing in patients with hypocellular marrow failure that does not require immediate therapy and thus allows time
to complete the evaluation. We also review the clinical utility of marrow array to detect acquired 6p copy number-neutral
loss of heterozygosity to support a diagnosis of aAA, the complexities of telomere length testing in patients with aAA,
short telomere syndromes, and other inherited bone marrow failure syndromes, as well as the limitations of somatic muta-
tion testing for mutations in myeloid malignancy genes for discriminating between the various diagnostic possibilities.

LEARNING OBJECTIVES
• Frame a comprehensive clinical and laboratory evaluation of hypocellular marrow failure based on disease severity
• Recognize that acquired 6p CN-LOH in the context of hypocellular marrow failure favors a diagnosis of aAA
• Recognize that germline mutations in SAMD9, SAMD9L, and GATA2 are enriched among young patients with MDS
with chromosome 7 abnormalities
• Recognize the clinical utility and limitations of telomere length testing by flow-FISH in discriminating between
causes of hypocellular marrow failure

age and results from acquired and inherited causes that can
CLINICAL CASE overlap in clinical presentation and bone marrow features.
A 28-year-old woman seeks treatment for fatigue and easy For the purpose of this manuscript and to avoid ambiguity,
bruising. Complete blood count reveals a hemoglobin of we restrict the term aplastic anemia to the immunolog-
10g/dL, absolute neutrophil count of 1.5×109/L, platelet ically mediated disease entity, acquired aplastic anemia
count of 45×109/L, and an absolute reticulocyte count of (aAA). Our workup centers on classifying patients as hav-
70 000/µL. Marrow aspirate is hypocellular for age with ing hypocellular marrow failure requiring urgent therapy,
no frank morphologic dysplasia. Trephine biopsy specimen including disease defined as severe by modified Camitta’s
shows an estimated marrow cellularity of 30% and no retic- criteria1 (Table 1) or nonsevere hypocellular marrow failure,
ulin fibrosis. Flow cytometry shows normal myeloid matura- which we define as cytopenia(s) and a hypocellular mar-
tion and no abnormal or expanded myeloid blast population. row for age and not meeting criteria for severe disease and
who do not require urgent therapy.
Causes of hypocellular marrow failure are shown in
Defining hypocellular marrow failure, the differential Table 2. aAA is bimodal in its age at presentation, is most
diagnosis, and rationale for workup commonly idiopathic, and accounts for most patients with
Hypocellular marrow failure is characterized by peripheral severe disease.3 Rarely and enriched among pediatric pre-
cytopenia(s) and bone marrow cellularity deemed low for sentations, aAA is due to an underlying inborn error in

134 | Hematology 2021 | ASH Education Program


Table 1.  Severe hypocellular mar­row fail­ure clas­si­fi­ca­tion Table 2.  Differential diag­no­sis of hypocellular mar­row fail­ure
cri­te­ria
Acquired Inherited
Marrow cel­lu­lar­ity aAA Classical IBMFS/MMP
<25% or Fanconi anemia
Anorexia nervosa
25%-50% with <30% resid­ual hema­to­poi­etic cells Short telo­mere syn­dromes
Shwachman-Diamond Syndrome
AND cytopenias (at least 2 of 3) GATA2 defi­ciency
Absolute neu­tro­phil count <500 × 109/L SAMD9/SAMD9L dis­or­ders
Platelets <20 × 109/L Hypocellular myelodysplastic Inborn errors of immu­nity (eg, X-linked
Absolute retic­u­lo­cyte count <60 × 109/L syndrome lymphoproliferative dis­or­der)
The immu­no­log­i­cally medi­ated dis­ease, severe aAA, accounts for the Medications/tox­ins
major­ity of severe hypocellular mar­row fail­ure.
This table is not exhaus­tive and is intended to cap­ture the more com­

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/134/1852140/134geddis.pdf by guest on 13 December 2021


monly encoun­ter enti­ties that can pres­ent as aplastic ane­mia. In our
immu­nity such as X-linked lymphoproliferative dis­or­der.4 Accu- clin­i­cal expe­ri­ence, the IBMFSs/MMPs listed here are not always
char­ac­ter­ized by hypocellular mar­rows. Gelatinous deg­ra­da­tion of the
rate diag­no­sis of patients with nonsevere hypocellular mar­row bone mar­row also may be seen in anorexia nervosa.2
fail­ure is clin­i­cally more chal­leng­ing and requires con­sid­er­ation
of hypocellular myelodysplastic syn­drome (MDS) with or with­
out an under­ly­ing clas­si­cal inherited bone mar­row fail­ure syn­ in Table 3. A tai­lored his­tory and phys­i­cal exam­i­na­tion can be
drome or inherited mye­loid malig­nancy pre­dis­po­si­tion syn­drome help­ful in iden­ti­fy­ing the under­ly­ing cause (Table 4), par­tic­u­larly
(IBMFS/MMP). Among the most com­mon clas­si­cal IBMFSs, Fan- doc­u­men­ta­tion of prior com­plete blood counts as longstanding
coni ane­mia (FA), short telo­mere syn­dromes, and Shwachman-Di- cytopenias or mac­ro­cy­to­sis can indi­cate a her­i­ta­ble cause. As
amond syn­drome (SDS) may pres­ent with hypocellular mar­row IBMFS/MMP can occur with­out clin­i­cal stig­mata or fam­ily his­tory,
fail­ure. While rare and in con­trast to MDS in the elderly, child­hood an unre­mark­able his­tory and exam­i­na­tion do not reli­ably exclude
MDS is most com­monly hypocellular,5 and also, approx­i­ma­tely 15% these dis­or­ders; the pres­ence of select dis­ease fea­tures should
of adults with MDS have a hypocellular mar­row.6 Further com­pli­ raise clin­i­cal sus­pi­cion (Table 5). If an MDS-defin­ing cyto­ge­netic
cat­ing the diag­no­sis, patients with IBMFS/MMP (eg, muta­tions in abnor­mal­ity is absent, the dis­tinc­tion between aAA and hypo-
GATA27) may have cytopenias and mega­kar­yo­cytic atypia with­ cellular MDS is based on mor­pho­logic fea­tures (dys­pla­sia and in-
out overt malig­nant clonal dis­ease that can be mis­taken for MDS. creased blasts).15 Notably, cyto­ge­netic abnor­mal­i­ties can occur
Important diag­nos­tic pos­si­bil­i­ties to con­sider in young per­sons in aAA. Trisomy 8 and del(13q) are seen in aAA, where both carry
with hypocellular MDS char­ac­ter­ized by chro­mo­some 7 abnor­ a favor­able prog­no­sis and are asso­ci­ated with response to IST.16,17
mal­i­ties are GATA2 defi­ciency8,9 and SAMD9/9L dis­or­ders.10 While Hypocellular MDS, par­ tic­
u­larly among young patients, should
IBMFS/MMP may be suspected based on per­ti­nent his­tory and prompt con­sid­er­ation of an under­ly­ing IBMFS/MMP.
phys­i­cal exam find­ings (Table 3), cryp­tic pre­sen­ta­tions of these Diagnostic test­ing in hypocellular mar­row fail­ure requir­ing
dis­or­ders are increas­ingly rec­og­nized and thus war­rant con­sid­ ther­apy (Figure 1A) is done simul­ta­neously, rather than sequen­
er­ation even in the absence of these clues.3 Last, in all­patients, tially, to allow defin­i­tive treat­ment to start expe­di­tiously to limit
the poten­tial of a revers­ible cause—namely, med­i­ca­tion-induced trans­fu­sions and the risk of seri­ous infec­tion. We aim to ini­ti­ate
hypocellular mar­row fail­ure—should be excluded. treat­ment within approx­i­ma­tely 3 weeks of pre­sen­ta­tion, which
The cor­rect diag­no­sis of hypocellular mar­row fail­ure is clin­ allows time to com­plete diag­nos­tic stud­ies and donor eval­u­a­

cally par­ a­mount. Patients with severe aAA are treated with tions. Evaluation of hypocellular mar­ row fail­
ure not requir­ ing
either hema­to­poi­etic stem cell trans­plant (HSCT) or immu­no­sup­ urgent ther­apy can typ­i­cally pro­ceed in a more step­wise fash­ion
pres­sive ther­apy (IST) with equine antithymocyte glob­u­lin and (Figure 1B). There are clin­i­cal set­tings where genetic test­ing for
cyclo­spor­ine with or with­out the addi­tion of a thrombopoietin IBMFS/MMP may not be per­ti­nent (eg, elderly patient with mul­
recep­tor ago­nist.11 Marrow fail­ure due to IBMFS/MMP or MDS is ti­ple comorbidities and no poten­tially affected fam­ily mem­bers
unlikely to respond to IST, and HSCT may be indi­cated. Recog- in whom test­ing would not inform the patient’s or fam­ily’s care).
nition of a her­i­ta­ble dis­or­der has impor­tant impli­ca­tions to the
selec­tion of an appro­pri­ate con­di­tion­ing reg­i­men, eval­u­a­tion of
poten­tial related stem cell donors, and sur­veil­lance for hema­to­
poi­etic and nonhematopoietic com­pli­ca­tions, as well as coun­sel­
ing for fam­ily mem­bers. CLINICAL CASE (Con­tin­ued)
Old records are requested and doc­ u­
ment sev­ eral nor­
mal
Clinical and lab­o­ra­tory eval­u­a­tion of hypocellular past com­ plete blood counts. Medical his­ tory and exam­ i­
na­
mar­row fail­ure tion are unre­mark­able. Chromosomal break­age stud­ies return
A diag­nos­tic approach to hypocellular mar­row fail­ure is shown unre­mark­able. Telomere length test­ing falls in the 30th age-
in Figure 1. Notably, since mar­row cel­lu­lar­ity can be patchy, it adjusted per­cen­tile.
is crit­i­cal that the biopsy spec­i­men be of ade­quate size (aim
for 2 cm) and qual­ity to ensure it is rep­re­sen­ta­tive. Numerous
guide­ lines detailing the recommended and suggested ini­ tial Chromosomal break­age test
lab­o­ra­tory stud­ies of hypocellular mar­row fail­ure exist.12–14 Our Given the impact a diag­no­sis of FA has on treat­ment and sur­veil­
recommended and suggested lab­ o­
ra­tory stud­ ies are listed lance strat­e­gies, a chro­mo­somal break­age test is indi­cated in
Prakash Singh Shekhawat
Evaluation of hypocellular mar­row fail­ure | 135
Table 3.  Suggested ini­tial lab­o­ra­tory eval­u­a­tion of hypocellular mar­row fail­ure

Implications for
Study Source Diagnosis treat­ment
Peripheral blood
Flow cytometry for PNH PB aAA IST
Chromosomal break­age test­ing PB FA HSCT
Modified con­di­tion­ing
Telomere lengths by flow-FISH PB STS HSCT
Modified con­di­tion­ing
Immunoglobulins, lym­pho­cyte sub­sets, natural killer PB Inborn error of immu­nity, GATA2 defi­ciency ­ HSCT
cell func­tion syn­drome, XLP Modified con­di­tion­ing
Infection pro­phy­laxis

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/134/1852140/134geddis.pdf by guest on 13 December 2021


Pancreatic isoamylase PB SDS
HLA typ­ing on patient and full sib­lings PB Severe aAA or MDS HSCT
Donor test­ing
Bone mar­row
Morphologic review of mar­row BM MDS, GATA2 defi­ciency—mega­kar­yo­cytic atypia
Routine kar­yo­type BM MDS, IBMFS/MMP HSCT
FISH BM MDS, IBMFS/MMP HSCT
includ­ing −7/del, −5/del, +8, del(q20)
Chromosome geno­mic array BM 6p CN-LOH in aAA; germline CNAs in
IBMFS/MMP; risk strat­i­fi­ca­tion of NK MDS, 7q
LOH in SAMD9/9L dis­or­ders
Somatic multigene genetic test­ing for muta­tions in BM See text for complexities
mye­loid malig­nancy genes
Cultured skin fibro­blasts
Germline multigene genetic test­ing Fibroblasts* IBMFS/MMP HSCT
Modified con­di­tion­ing
Donor test­ing
Studies recommended in all­patients are bolded.
*When clin­i­cally pos­si­ble, cul­tured skin fibro­blasts are the recommended DNA source for germline test­ing.
BM, bone mar­row aspi­rate; NK, nor­mal kar­yo­type; PB, periph­eral blood; PNH, par­ox­ys­mal noc­tur­nal hemo­glo­bin­uria.

all­patients with hypocellular mar­row fail­ure. Abnormal test re- test­ing performed on lym­pho­cytes shows 2 pop­ul­a­tions of cells,
sults should be followed with genetic test­ing of a germline tis­sue some appearing nor­mal and oth­ers with mul­ti­ple breaks per cell.
source to try to iden­tify the caus­a­tive muta­tion as this pro­vi­des Negative results should be con­firmed in cul­tured fibro­blasts if FA
addi­tional prog­nos­tic infor­ma­tion regard­ing can­cer and other is strongly suspected. Breakage stud­ies on fibro­blasts can also
risks as well as facil­i­tates car­rier test­ing for fam­ily mem­bers.18 be con­sid­ered when stud­ies in lym­pho­cytes fail due to severe
Breakage stud­ies allow diag­no­sis of patients whose muta­tions leu­ko­pe­nia or poor growth in cul­ture.
are missed by stan­dard genetic test­ing and inform the func­tional
con­se­quence of genetic var­i­ants that might oth­er­wise be of un- Complexities of telo­mere length test­ing
clear clin­i­cal sig­nif­i­cance. This test­ing is performed by cul­tur­ing The short telo­mere syn­dromes are a group of genetic dis­or­ders
either periph­eral blood lym­pho­cytes or skin fibro­blasts in the that are caused by muta­tions in com­po­nents of the telomerase
pres­ence of DNA cross-linking agents and com­par­ing the num­ enzyme and other telo­mere main­te­nance genes. Telomere length
ber of chro­mo­somal breaks, includ­ing rearrangements, gaps, en- test­ing by flow cytom­et­ ry and fluo­res­cence in situ hybrid­iza­tion
doreduplications, and exchanges, to con­trols under base­line and (flow-FISH) is the gold stan­dard because of its high repro­duc­ibil­
stim­u­lated con­di­tions. Results are reported as both the num­ber ity. It mea­sures sin­gle-cell telo­mere length using a fluorescently
of cells with abnor­mal breaks and the num­ber of breaks per cell. labeled probe that hybrid­izes to telo­mere DNA. Clinical test­ing
Although periph­eral blood lym­pho­cytes are the most acces­si­ is performed on a periph­eral blood sam­ple, and avail­­able assays
ble source of tis­sue for break­age and genetic stud­ies, lym­pho­ report results in total lym­pho­cytes and granulocytes (so called
cytes are sus­cep­ti­ble to the phe­nom­e­non of somatic mosa­i­cism 2-panel test­ing) or granulocytes and total lym­pho­cytes and lym­
resulting from expan­sion of hema­to­poi­etic stem cells that have pho­cyte sub­sets (naive T cells, mem­ory T cells, B cells, nat­u­ral
under­gone molec­u­lar rever­sion that cor­rects the FA phe­no­type. killer cells), so-called 6-panel test­ing. Two-panel test­ing appears
This is esti­mated to occur in approx­i­ma­tely 10% to 25% of pa- suf­fi­cient for the ini­tial diag­nos­tic workup of a patient with hypo-
tients with FA.19 Somatic rever­sion may be suspected if fra­gil­ity cellular mar­row fail­ure.20

136  |  Hematology 2021  |  ASH Education Program


Initial work-up to include:
Pancytopenia & - History and physical examination (see Table 4)
hypocellular marrow CBC, retic, and marrow cellularity c/w - Cytogenetics (routine karyotype, FISH, and array) on BM
severe aplastic anemia - PNH screen on PB
- Chromosomal breakage test on PB
Laboratory work-up and HLA - Telomere length by flow-FISH
typing underway - Pancreatic isoamylase
- HLA typing on patient and full siblings
1 wk
Normal morphology and cytogenetics? Consider Consider underlying IBMFS/MMP
No hypocellular MDS
Yes

2 wk Negative or Initial work-up


PNH or 6p CN-LOH positive positive

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/134/1852140/134geddis.pdf by guest on 13 December 2021


Severe aAA IBMFS/MMP
-PNH or 6p CN-LOH
further supports diagnosis

3 wk
Rapid panel-based NGS
Initiate therapy testing for IBMFS/MMP
(IST or HSCT) of patient & related
donor on PB

If severe lymphopenia, MDS, or suspicion for IBMF/AL-MDS obtain a skin biopsy to culture
skin fibroblasts in case needed for chromosomal fragility testing or germline genetic testing.
If clinical suspicion of Shwachman-Diamond Syndrome.
Is an MDS-defining finding present?

Pancytopenia & CBC, retic, and marrow cellularity consistent Initial work-up to include:
hypocellular marrow with non-severe hypocellular marrow failure - History and physical examination Table 4)
(not requiring urgent therapy) - Cytogenetics (routine karyotype, FISH, and array) on BM
- PNH screen on PB
- Chromosomal breakage study on PB
Laboratory work-up underway
- Telomere length by flow-FISH
1 wk

Initial work-up
2 wk Negative and Positive or
PNH or 6p CN-LOH positive PNH/6p CN-LOH negative

• Consider targeted capture or WES of myeloid


malignancy genes (include BCOR/BCORL) on BM

3 wk Skin biopsy to culture


Nonsevere aAA
fibroblasts

Panel-based NGS testing for IBMFS/MMP • There are clinical settings where genetic testing
on fibroblast DNA IBMFS/MMP may not be pertinent
6 wk & syndrome specific testing
Monitor

IBMFS/MMP and/or hypocellular MDS or aAA

Does not reliably distinguish among causes of hypocellular marrow failure; may provide some prognostic information.
Serum pancreatic isoamylase for Shwachman-Diamond Syndrome
If MDS-defining finding present.

Figure 1. Diagnostic approach to hypocellular marrow failure based on severity. (A) Diagnostic approach to severe hypocellu-
lar marrow failure requiring therapy—time-limited. (B) Diagnostic approach to nonsevere hypocellular marrow failure not requiring
therapy—not time-limited. Time-limited indicates the urgency to get the workup completed before starting therapy. BM, bone mar-
row aspirate; PB, peripheral blood; PNH, paroxysmal nocturnal hemoglobinuria.

Prakash Singh Shekhawat


Evaluation of hypocellular mar­row fail­ure | 137
Table 4.  Focused med­i­cal his­tory and phys­i­cal exam­i­na­tion

Questions Supportive infor­ma­tion Implications


Presentation Duration of symp­toms Prior CBC/diff Inherited vs acquired
Bleeding Platelet count, coag­u­la­tion stud­ies Transfusion require­ments
Fatigue Hemoglobin, pulse oximetry, CXR Transfusion require­ments
S/Sx of infec­tion Fever, respi­ra­tory symp­toms Identify and treat infec­tions
Medical his­tory Adrenal hypo­pla­sia SAMD9
Ataxia Brain MRI SAMD9L
Atypical infec­tions Immunologic test­ing, vac­ci­na­tion his­tory SDS, GATA2, STS, SAMD9/9L
Birth his­tory Birth weight, ges­ta­tional age, IUGR, ane­mia DBA, FA, SAMD9/9L, SDS, STS

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/134/1852140/134geddis.pdf by guest on 13 December 2021


Cancer or leu­ke­mia Type of che­mo­ther­apy and toxicities IBMFS, sec­ond­ary MDS
Congenital malformations Echo, abdom­i­nal US, brain MRI, x-rays FA
GI (diar­rhea, dif­fi­culty swallowing) LFTs, abdom­i­nal US, endos­copy aAA, SDS, STS, PNH
Growth Growth chart FA
Hearing loss Audiogram FA, GATA2
Nutrition (vegan, weight loss, sup­ple­ments) B12/MMA/HC, ceru­lo­plas­min/cop­per B12, folate, cop­per defi­ciency
Pulmonary dis­ease (TE fis­tula, fibro­sis, CXR, CT, PFTs, bron­chos­copy FA, GATA2, STS, SAMD9/9L
pneu­mo­nia, alve­o­lar proteinosis)
Renal dis­ease (con­gen­i­tal anom­aly, dark urine) Urinalysis, renal US FA, PNH
Skeletal dys­pla­sia, skel­e­tal abnor­mal­i­ties X-rays SDS, FA, DBA
Family his­tory Ancestry Founder muta­tions in cer­tain
pop­u­la­tions
Consanguinity Autosomal reces­sive dis­or­ders
Full sib­lings Potential mar­row donors
Marrow fail­ure, MDS, leu­ke­mia, can­cer IBMFS
Examination find­ings Syndromic facies IBMFS
Microcephaly STS, SAMD9/9L
Leukoplakia STS
Hepatomegaly SDS
Abnormal thumbs, radial abnor­mal­i­ties FA
Nail dys­pla­sia STS
Lymphedema GATA2
Café-au-lait spots FA
Reticulated pig­men­ta­tion on neck STS
Warts GATA2
CBC/diff, com­plete blood count with dif­fer­en­tial; CT, com­put­er­ized tomog­ra­phy; CXR, chest x-ray; GI, gas­tro­in­tes­ti­nal; IUGR, intra­uter­ine growth
retar­da­tion; GATA2, GATA2 defi­ciency; HC, homocysteine; LFT, liver function testing; MMA, methylmalonic acid; mag­netic res­o­nance imag­ing;
PFT, pul­mo­nary func­tion test; S/Sx, signs/symp­toms; SAMD9/9L, SAMD9/SAMD9L dis­or­ders; TE, tracheoesophageal fis­tula; US, ultrasound.

Telomere length (TL) assess­ment is recommended in the ini­tial been reported in patients with other IBMFSs, includ­ ing FA and
diag­nos­tic workup of hypocellular mar­row fail­ure. TL assess­ment SDS,21,23 and so con­sid­er­ation of the clin­i­cal pre­sen­ta­tion and addi­
is use­ful in rul­ing out a diag­no­sis of a con­sti­tu­tional short telo­mere tional genetic test­ing results are nec­es­sary to estab­lish the cor­rect
syn­drome upfront in patients with mar­ row fail­ ure. Lymphocyte diag­no­sis. In a pre­dom­i­nantly pedi­at­ric and youn­ger adult cohort
telo­mere length above the age-adjusted 50th per­ cen­tile has a of patients youn­ger than 40 years (0-31 years; median, 14 years)
100% neg­a­tive pre­dic­tive value in iden­ti­fy­ing a clin­i­cally mean­ing­ presenting with idi­o­pathic mar­row fail­ure, all­of the patients with
ful muta­tion in a short telomere syndrome (STS) gene in pedi­at­ immu­no­ther­apy-respon­sive mar­row fail­ure had lym­pho­cyte telo­
ric and adult patients.21 Among patients youn­ger than 40 years, a mere lengths above the age-adjusted first per­cen­tile.21 Nearly half of
lym­pho­cyte telo­mere length below the age-adjusted first per­cen­ the patients with genet­i­cally con­firmed short telo­mere syn­dromes
tile strongly supports a diagnosis of a STS but lack specificity.21,22 older than 40 years have telo­mere length mea­sure­ments above
Telomere lengths below the age-adjusted first per­ cen­tile have the age-adjusted first per­cen­tile, and so telo­mere lengths above

138  |  Hematology 2021  |  ASH Education Program


Table 5.  Features of select inherited bone mar­row fail­ure and inherited MDS/leu­ke­mia pre­dis­po­si­tion syn­dromes that
may pres­ent as aplastic ane­mia

Syndrome Congenital find­ings Malignancy risk Screening test Genetics Other hints
Fanconi anemia Ear abnor­mal­i­ties, heart defects, short AML, MDS, GYN CA, Increased chro­mo­ FANCA, C, G account Sensitivity to
stat­ure, skin pig­men­ta­tion (café- head and neck some break­age for 95% of cases che­mo­ther­apy
au-lait spots or hypopigmentation), CA, and oth­ers
skel­e­tal anom­a­lies (thumbs, arms),
TE fis­tula, tri­an­gu­lar facies, uro­gen­
i­tal defects
GATA2 defi­ciency Lymphedema, immu­no­de­fi­ciency AML, MDS GATA2 Megakaryocyte
with atyp­i­cal atypia, pedi­at­ric
myco­bac­te­rial infec­tions MDS with mono­
somy 7, del7q, tri­

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/134/1852140/134geddis.pdf by guest on 13 December 2021


somy 8, der(1;7)
SAMD9/SAMD9L MIRAGE (SAMD9): MDS, infec­tion, AML, MDS SAMD9, SAMD9L Pediatric MDS with
dis­or­ders restric­tion of growth, adre­nal hypo­ mono­somy 7,
pla­sia, gen­i­tal phe­no­types, and del7q, or CN-LOH
enter­op­a­thy 7q
Ataxia-pan­cy­to­pe­nia syn­drome
(SAMD9L): cer­e­bel­lar atro­phy and
white mat­ter hyperintensities, gait
dis­tur­bance, nys­tag­mus
Short telo­mere Young adults: infec­tions, nail dys­tro­ AML, MDS Short telo­mere lengths DKC1, RTEL1, TERT,
syn­dromes phy, oral leukoplakia, skin hyper­ Rectal ade­no­car­ (cor­re­lates with TERC, TINF2,
pig­men­ta­tion ci­noma, SCC phe­no­type) RTEL1 account for
Adults: emphy­sema, early hair anus/oral cav­ major­ity of cases
graying, immune defi­ciency, liver ity/tongue
fibro­sis/cir­rho­sis, mac­ro­cy­to­sis,
pul­mo­nary AVMs and HPS
Shwachman- Pancreatic insuf­fi­ciency (can improve AML, MDS Low pan­cre­atic SBDS, SRP54, ELF1, Isolated neutropenia,
Diamond with age), skel­e­tal abnor­mal­i­ties isoamylase (>3 years DNAJC21 somatic muta­tions
Syndrome old) and low fecal in EIF6 and TP53
elas­tase (pediatric
and adult patients)
AML, acute myeloid leukemia; AVM, arteriovenous malformations; CA, cancer; GYN CA, gynecological cancers; HPS, hepatopulmonary syndrome;
SCC, squamous cell carcinoma; TE, tracheoesophageal fis­tula.

this thresh­old do not exclude the diag­no­sis of a short telo­mere array early in the course of their dis­ease.24,25 Clonal hema­to­poi­e­sis
syn­drome in older patients and requires diagnosis here addi­tional does not equate to malig­nancy and reflects an acquired genet-
clin­i­cal and genetic infor­ma­tion.21,22 Telomere length assess­ment in ic change in a hema­to­poi­etic stem cell, allowing for com­pet­i­tive
var­i­ous causes of hypocellular mar­row fail­ure are shown in Figure 2. out­growth of the mutated hema­to­poi­etic stem cell’s prog­eny. The
2 most com­mon somatic muta­tions in aAA are muta­tional inac­ti­
va­tion of PIGA and the loss of HLA class I alleles. The clin­i­cal util­ity
of detecting PIGA loss or the func­tional con­se­quence of a detect­
CLINICAL CASE (Con­t in­u ed) able par­ox­ys­mal noc­tur­nal hemo­glo­bin­uria clone in hypocellular
Marrow kar­ yo­
type returns 46,XX. Microarray on the mar­ row mar­row fail­ure is cov­ered in Education Session entitled “When
aspi­rate sam­ple iden­ti­fied copy number-neutral loss of het­ero­ does a PNH clone have clinical significance?”26 PIGA loss is most
zy­gos­ity of 6p in 10% of the cells. Flow cytometry on periph­eral com­monly diag­nosed by flow-cytometric anal­y­sis to detect cells
blood did not iden­tify any glycosylphosphatidylinositol (GPI)- lacking GPI-linked pro­teins. This study is more sen­si­tive when per-
defi­cient pop­u­la­tions. formed on a periph­eral blood sam­ple com­pared with a bone mar­
row aspi­rate sam­ple as the GPI-linked pro­teins typ­i­cally assessed
are most highly expressed on periph­eral blood cells. Thus, test­ing
Diagnostic util­ity of select genetic abnor­mal­i­ties on periph­eral blood as opposed to a mar­row sam­ple for a par­ox­
in hypocellular mar­row fail­ure ys­mal noc­tur­nal hemo­glo­bin­uria clone is recommended.27
Acquired copy number-neutral loss of het­ ero­zy­
gos­
ity of Loss of HLA class I alleles occurs either through a loss-of-func­tion
chro­mo­some arm 6p and chro­mo­somal microarray test­ing on muta­tion in 1 of the HLA class I genes or, more com­monly, through
plat­forms includ­ing sin­gle-nucle­o­tide poly­mor­phism probes the loss of 1 paren­tal HLA hap­lo­type through the acqui­si­tion of
More than 70% of patients (and over 60% of pedi­at­ric patients) with copy number-neutral loss of het­ero­zy­gos­ity of chro­mo­some arm
aAA have clonal hema­to­poi­e­sis with somatic muta­tions detected 6p (6p CN-LOH) involv­ing the human leu­ko­cyte anti­gen locus. A
by whole-exome sequenc­ing or sin­gle-nucle­o­tide poly­mor­phism prevailing hypoth­e­sis is that the loss of HLA class I alleles dis­rupts
Prakash Singh Shekhawat
Evaluation of hypocellular mar­row fail­ure | 139
Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/134/1852140/134geddis.pdf by guest on 13 December 2021
Figure 2.  Utility of telomere length testing in the diagnosis of hypocellular marrow failure. The red circles denote patients with
bone marrow failure secondary to a genetically confirmed diagnosis of a short telomere syndrome. The blue circles denote patients
with presumed idiopathic acquired aplastic anemia based on response to treatment with IST. The green circles denote patients with
bone marrow failure due to an underlying genetically confirmed inherited bone marrow failure syndrome apart from a short telomere
syndrome (LIG4, RUNX1, GATA2). The black circles denote patients diagnosed with a short telomere syndrome at >40 years of age.
BMF, bone marrow failure. Based on data from Alder et al.21

anti­gen pre­sen­ta­tion, allowing non­dom­i­nant clones to evade the with MDS or in nor­mal aging.30,34,35 The prev­a­lence of clonal 6p
immune sys­tem. An anal­o­gous path­o­phys­i­­ol­ogy may con­trib­ute CN-LOH among pedi­at­ric and adult patients with aAA is approx­
to posttransplant relapse in haploidentical HSCT recip­ i­
ents, in i­ma­tely 10% to 13%.24,33,36 Constitutional 6p homo­zy­gos­ity is not
whom the relapsed dis­ease has acquired loss of the HLA hap­lo­ enriched among patients with aAA.
type that dif­fered from the donor’s hap­lo­type, thus damp­en­ing Clinical test­ing for 6p CN-LOH requires chro­mo­somal microar-
the graft-ver­sus-leu­ke­mia effect.26,28,29 In aAA, the HLA alleles spe­ ray anal­y­sis (CMA). Outside of detecting 6p CN-LOH, addi­tional
cif­i­cally lost through 6p CN-LOH vary across age and eth­nic ori­gin ratio­nale supporting inclu­sion of CMA in the workup of hypocel-
of patients. That said, stud­ies report a bias of the miss­ing HLA lular mar­row fail­ure includes that this test­ing can be performed
allele in 6p CN-LOH to par­tic­u­lar HLA types. This, together with on DNA extracted from resid­ ual cell pel­ lets, aspi­
rate smears,
the find­ing that the HLA allele is com­monly involved across the touch preps, or for­ma­lin-fixed, par­af­fin embed­ded tis­sues. Cells
6p CN-LOH reported in aAA, sug­gests the find­ing is linked to the obtained from patients with hypocellular mar­ row fail­ure may
path­o­gen­e­sis of aAA and not merely a sec­ond­ary event. Although not grow well in cul­ture for kar­yo­typic anal­y­sis. In addi­tion to
addi­tional stud­ies are needed to clar­ify the impact of acquired 6p detecting CN-LOH, CMA can iden­tify microdeletions and micro-
CN-LOH on response to IST, the pres­ence of 6p CN-LOH appears duplications beyond the res­o­lu­tion of con­ven­tional kar­yo­type
to be diag­nos­ti­cally infor­ma­tive in distinguishing patients with and FISH,37 poten­tially pro­vid­ing a ratio­nale for closer clin­i­cal sur­
immune-medi­ated mar­row fail­ure from those with inherited bone veil­lance for malig­nant clonal evo­lu­tion. In addi­tion, CMA may
mar­row fail­ure con­di­tions.30-33 Acquired 6p CN-LOH appears to be detect copy num­ber alter­ations (CNAs) missed by some targeted
rel­a­tively spe­cific for aAA, occur­ring in less than 1% of patients next-gen­er­a­tion sequenc­ing (NGS)–based test­ing. Causative

140  |  Hematology 2021  |  ASH Education Program


The evaluation of hypocellular marrow failure

Inial worku
workup:
History and phy
physical
morphology and cytogene
cs
Marrow morph
- 6p CN-LOH
PNH
PNH Chromosomal b breakage test
TTelomere
Te lomere length
AA HLA typing of ppa
ent and siblings

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/134/1852140/134geddis.pdf by guest on 13 December 2021


6p CN-LOH MDS
Severe disease
- ANC <500 x 109/L
- Plt <20 x 109/L Acquired Inherited
- Abs re
c <60 x 109/L

Nonsevere disease Likely acquired Germline


- Not requiring urgent genec tesng
therapy Consider acquired or inherited

Figure 3. The evaluation of hypocellular marrow failure.

CNAs have been reported in a num­ber of IBMFs/MMPs, includ­ing refrac­tory cytopenia of child­ hood,9,11 adult hypo­plas­tic MDS,44,45
FA and oth­ers.38,39 In 1 cohort study of patients with suspected and aAA, 24,44
so this find­ing is also not infor­ma­tive diag­nos­ti­cally.
inherited bone mar­row fail­ure, CMA iden­ti­fied germline path­o­ When fea­si­ble, this test­ing can be con­sid­ered for its poten­tial
genic CNAs in 16.4% (11/67) of patients tested.40 prog­nos­tic value.46 As exam­ples, the acqui­si­tion of biallelic TP53
muta­tions is asso­ci­ated with devel­op­ment of leu­ke­mia in SDS,47
Somatic muta­tion test­ing for muta­tions in mye­loid and in aAA, BCOR and BCORL1 muta­tions are asso­ci­ated with re-
malig­nancy genes sponse to immu­no­sup­pres­sion and a favor­able prog­no­sis.24
The pro­ file of somatic muta­ tions iden­ ti­
fied by NGS strat­ e­
gies
in aAA, pedi­at­ric MDS, hypocellular MDS in adults, and IBMFS/
MMP dif­fers from one another. An under­stand­ing of this con­text-
depen­dent clonal land­scape and its dynam­ics con­tin­ues to pro­ CLINICAL CASE (Con­tin­ued)
vide impor­tant insights into malig­nant and non­ma­lig­nant clonal Based on her workup, the patient was diag­nosed with nonse-
evo­lu­tion and, in select set­tings, offers prog­nos­tic insights. This vere aAA and is being followed by serial blood counts.
has been recently reviewed.40-42 The cur­rent diag­nos­tic util­ity of
NGS pan­els and whole-exome sequenc­ing for somatic muta­tions
in mye­loid malig­nancy genes to reli­ably dis­tin­guish among these Conclusion
hypocellular mar­row fail­ure syn­dromes in the clinic remains unclear. The eval­u­a­tion of hypocellular mar­row fail­ure (Figure 3) is based
A spe­cific somatic muta­tional land­scape that can reli­ably iden­tify on dis­ease sever­ity and aims to dis­tin­guish among the inherited
bona fide MDS (in the absence of defin­i­tive mor­pho­logic or cyto­ and acquired causes to inform opti­mal patient care.
ge­netic find­ings) for clin­i­cal prac­tice in this set­ting has not been
established. Approximately 25% to 30% of patients with aAA have Acknowledgments 
clonal muta­tions in genes that are asso­ci­ated with both mye­loid We thank patients and fam­i­lies for par­tici­pat­ing in mar­row fail­
malig­nancy and with aging, includ­ing ASXL1, DNMT3A, JAK2, and ure research and Mary Armanios at Johns Hopkins University for
TP53. These muta­tions are pres­ent at low var­i­ant allele fre­quency pro­vid­ing Figure 2.
sim­i­lar to what is seen in age-related clonal hema­to­poi­e­sis, and
these genes can also be mutated in adult hypocellular MDS.24,43,44 Conflict-of-inter­est dis­clo­sures
In addi­tion, although muta­tions in the epi­ge­netic mod­i­fier genes, Amy Geddis: no rel­e­vant con­flicts of inter­est.
BCOR and BCORL1, appear sub­tly enriched in aAA com­pared with Siobán Keel: no rel­e­vant con­flicts of inter­est.
hypocellular MDS, both dis­eases can carry these muta­tions, and
so this find­ing is not suf­fi­cient to dis­tin­guish between these dis­or­ Off-label drug use
ders.44 In con­trast to typ­i­cal adult MDS, muta­tions in the spliceo- Amy Geddis: nothing to disclose.
some com­plex (ie, SF3B1, SRSF2, ZRSR2, U2AF1) are rare across Siobán Keel: nothing to disclose.
Prakash Singh Shekhawat
Evaluation of hypocellular mar­row fail­ure | 141
Correspondence 25. Babushok DV, Perdigones N, Perin JC, et al. Emergence of clonal hema­to­
poi­es­ is in the major­ity of patients with acquired aplastic ane­mia. Cancer
Siobán Keel, University of Washington, Division of Hematology,
Genet. 2015;208(4):115-128.
Seattle, WA 98105; e-mail: sioban@uw​­.edu. 26. Shaw YB. et al. Blood Advances 2021: The predictive value of PNH clones,
6p CN-LOH, and clonal TCR gene rearrangement for aplastic anemia. 5(16).
References 27. Borowitz MJ, Craig FE, Digiuseppe JA, et al; Clinical Cytometry Society.
1. Camitta BM, Rappeport JM, Parkman R, Nathan DG. Selection of patients Guidelines for the diag­no­sis and mon­i­tor­ing of par­ox­ys­mal noc­tur­nal
for bone mar­ row trans­ plan­
ta­
tion in severe aplastic ane­ mia. Blood. hemo­glo­bin­uria and related dis­or­ders by flow cytom­e­try. Cytometry B
1975;45(3):355-363. Clin Cytom. 2010;78(4):211-230.
2. Abella E, Feliu E, Granada I, et al. Bone mar­row changes in anorexia nervosa 28. Villalobos IB, Takahashi Y, Akatsuka Y, et al. Relapse of leu­ke­mia with loss
are cor­re­lated with the amount of weight loss and not with other clin­i­cal of mismatched HLA resulting from uni­pa­ren­tal disomy after haploiden-
find­ings. Am J Clin Pathol. 2002;118(4):582-588. tical hema­to­poi­etic stem cell trans­plan­ta­tion. Blood. 2010;115(15):3158-
3. Keel SB, Scott A, Sanchez-Bonilla M, et  al. Genetic fea­ tures of myelo- 3161.
dysplastic syn­drome and aplastic ane­mia in pedi­at­ric and young adult 29. Vago L, Perna SK, Zanussi M, et  al. Loss of mismatched HLA in leu­ke­mia
patients. Haematologica. 2016;101(11):1343-1350. after stem-cell trans­plan­ta­tion. N Engl J Med. 2009;361(5):478-488.
4. Pachlopnik Schmid J, Canioni D, Moshous D, et al. Clinical sim­i­lar­i­ties and 30. Afable MG II, Wlodarski M, Makishima H, et al. SNP array-based karyotyp-

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/134/1852140/134geddis.pdf by guest on 13 December 2021


dif­fer­ences of patients with X-linked lymphoproliferative syn­drome type ing: dif­fer­ences and sim­i­lar­i­ties between aplastic ane­mia and hypocellular
1 (XLP-1/SAP defi­ciency) ver­sus type 2 (XLP-2/XIAP defi­ciency). Blood. myelodysplastic syn­dromes. Blood. 2011;117(25):6876-6884.
2011;117(5):1522-1529. 31. Babushok DV, Olson TS, Bessler M. Somatic muta­tions and clonal hema­to­
5. Niemeyer CM, Baumann I. Classification of child­hood aplastic ane­mia and poi­e­sis in aplastic ane­mia. N Engl J Med. 2015;373(17):1673.
myelodysplastic syn­drome. Hematol Am Soc Hematol Educ Program. 32. Babushok DV, Xie HM, Roth JJ, et al. Single nucle­o­tide poly­mor­phism array
2011;2011:84-89. anal­y­sis of bone mar­row fail­ure patients reveals char­ac­ter­is­tic pat­terns of
6. Nazha A, Seastone D, Radivoyevitch T, et al. Genomic pat­terns asso­ci­ated genetic changes. Br J Haematol. 2014;164(1):73-82.
with hypo­plas­tic com­pared to hyper­plas­tic myelodysplastic syn­dromes. 33. Katagiri T, Sato-Otsubo A, Kashiwase K, et al; Japan Marrow Donor Pro-
Haematologica. 2015;100(11):e434-e437. gram. Frequent loss of HLA alleles asso­ci­ated with copy num­ber-neu­tral
7. McReynolds LJ, Calvo KR, Holland SM. Germline GATA2 muta­tion and bone 6pLOH in acquired aplastic ane­mia. Blood. 2011;118(25):6601-6609.
mar­row fail­ure. Hematol Oncol Clin North Am. 2018;32(4):713-728. 34. Mohamedali AM, Gäken J, Ahmed M, et al. High con­cor­dance of geno­mic
8. Pastor V, Hirabayashi S, Karow A, et  al. Mutational land­scape in chil­dren and cyto­ge­netic aber­ra­tions between periph­eral blood and bone mar­row
with myelodysplastic syn­dromes is dis­tinct from adults: spe­cific somatic in myelodysplastic syn­drome (MDS). Leukemia. 2015;29(9):1928-1938.
driv­ers and novel germline var­i­ants. Leukemia. 2017;31(3):759-762. 35. Score J, Chase A, Forsberg LA, et  al. Detection of leu­ke­mia-asso­ci­ated
9. Wlodarski MW, Hirabayashi S, Pastor V, et al; EWOG-MDS. Prevalence, clin­ muta­tions in periph­eral blood DNA of hematologically nor­mal elderly indi­
i­cal char­ac­ter­is­tics, and prog­no­sis of GATA2-related myelodysplastic syn­ vid­u­als. Leukemia. 2015;29(7):1600-1602.
dromes in chil­dren and ado­les­cents. Blood. 2016;127(11):1387-1397, quiz 1518. 36. Betensky M, Babushok D, Roth JJ, et al. Clonal evo­lu­tion and clin­i­cal sig­nif­
10. Schwartz JR, Ma J, Lamprecht T, et al. The geno­mic land­scape of pedi­at­ric i­cance of copy num­ber neu­tral loss of het­ero­zy­gos­ity of chro­mo­some arm
myelodysplastic syn­dromes. Nat Commun. 2017;8(1):1557. 6p in acquired aplastic ane­mia. Cancer Genet. 2016;209(1-2):1-10.
11. Townsley DM, Scheinberg P, Winkler T, et  al. Eltrombopag added 37. Kanagal-Shamanna R, Hodge JC, Tucker T, et  al. Assessing copy num­ber
to stan­dard immu­no­sup­pres­sion for aplastic ane­mia. N Engl J Med. aber­ra­tions and copy neu­tral loss of het­ero­zy­gos­ity across the genome
2017;376(16):1540-1550. as best prac­tice: an evi­dence based review of clin­i­cal util­ity from the
12. Killick SB, Bown N, Cavenagh J, et al; Brit­ish Society for Standards in Hae- Cancer Genomics Consortium (CGC) Working Group for Myelodysplas-
matology. Guidelines for the diag­no­sis and man­age­ment of adult aplastic tic Syndrome, Myelodysplastic/Myeloproliferative and Myeloproliferative
anae­mia. Br J Haematol. 2016;172(2):187-207. Neoplasms. Cancer Genet. 2018;228-229:197-217.
13. Peslak SA, Olson T, Babushok DV. Diagnosis and treat­ment of aplastic ane­ 38. Kuramitsu M, Sato-Otsubo A, Morio T, et al. Extensive gene dele­tions in Jap­
mia. Curr Treat Options Oncol. 2017;18(12):70. a­nese patients with Diamond-Blackfan ane­mia. Blood. 2012;119(10):2376-
14. Shimano KA, et al. Am J Hematol. 2021. Online ahead of print. 2384.
15. Arber DA, Orazi A, Hasserjian R, et al. The 2016 revi­sion to the World Health 39. Waespe N, Dhanraj S, Wahala M, et al. The clin­i­cal impact of copy num­
Organization clas­si­fi­ca­tion of mye­loid neo­plasms and acute leu­ke­mia. ber var­i­ants in inherited bone mar­row fail­ure syn­dromes. NPJ Genom Med.
Blood. 2016;127(20):2391-2405. 2017;2:18.
16. Hosokawa K, Sugimori N, Katagiri T, et  al. Increased glycosylphosphati- 40. Sperling AS, Gib­son CJ, Ebert BL. The genet­ics of myelodysplastic syn­
dylinositol-anchored pro­tein-defi­cient granulocytes define a benign sub­ drome: from clonal haematopoiesis to sec­ond­ary leu­kae­mia. Nat Rev Can-
set of bone mar­row fail­ures in patients with tri­somy 8. Eur J Haematol. cer. 2017;17(1):5-19.
2015;95(3):230-238. 41. Sun L, Babushok DV. Secondary myelodysplastic syn­drome and leu­ke­mia
17. Ishiyama K, Karasawa M, Miyawaki S, et al. Aplastic anae­mia with 13q-: a in acquired aplastic ane­mia and par­ox­ys­mal noc­tur­nal hemo­glo­bin­uria.
benign sub­set of bone mar­row fail­ure respon­sive to immu­no­sup­pres­sive Blood. 2020;136(1):36-49.
ther­apy. Br J Haematol. 2002;117(3):747-750. 42. Tsai FD, Lindsley RC. Clonal hema­to­poi­e­sis in the inherited bone mar­row
18. Chandrasekharappa SC, Lach FP, Kimble DC, et al; NISC Comparative fail­ure syn­dromes. Blood. 2020;136(14):1615-1622.
Sequencing Program. Massively par­al­lel sequenc­ing, aCGH, and RNA-seq 43. Lane AA, Odejide O, Kopp N, et al. Low fre­quency clonal muta­tions recov­
tech­nol­o­gies pro­vide a com­pre­hen­sive molec­u­lar diag­no­sis of Fanconi er­able by deep sequenc­ing in patients with aplastic ane­mia. Leukemia.
ane­mia. Blood. 2013;121(22):e138-e148. 2013;27(4):968-971.
19. Nicoletti E, Rao G, Bueren JA, et al. Mosaicism in Fanconi ane­mia: con­cise 44. Negoro E, Nagata Y, Clemente MJ, et al. Origins of myelodysplastic syn­
review and eval­u­a­tion of published cases with focus on clin­i­cal course of dromes after aplastic ane­mia. Blood. 2017;130(17):1953-1957.
blood count nor­mal­i­za­tion. Ann Hematol. 2020;99(5):913-924. 45. Bono E, McLornan D, Travaglino E, et  al. Clinical, his­to­path­o­log­ic ­ al and
20. Alter BP, Rosenberg PS, Giri N, Baerlocher GM, Lansdorp PM, Savage SA. molec­u­lar char­ac­ter­iza­tion of hypo­plas­tic myelodysplastic syn­drome. Leu-
Telomere length is asso­ci­ated with dis­ease sever­ity and declines with age kemia. 2019;33(10):2495-2505.
in dyskeratosis congenita. Haematologica. 2012;97(3):353-359. 46. Kulasekararaj AG, Jiang J, Smith AE, et al. Somatic muta­tions iden­tify a sub­
21. Alder JK, Hanumanthu VS, Strong MA, et  al. Diagnostic util­ ity of telo­ group of aplastic ane­mia patients who prog­ress to myelodysplastic syn­
mere length test­ing in a hos­pi­tal-based set­ting. Proc Natl Acad Sci U S A. drome. Blood. 2014;124(17):2698-2704.
2018;115(10):E2358-E2365. 47. Kennedy AL, Myers KC, Bowman J, et al. Distinct genetic path­ways define
22. Schratz KE, Haley L, Danoff SK, et al. Cancer spec­trum and out­comes in the pre-malig­nant ver­sus com­pen­sa­tory clonal hema­to­poi­e­sis in Shwach-
Men­de­lian short telo­mere syn­dromes. Blood. 2020;135(22):1946-1956. man-Diamond syn­drome. Nat Commun. 2021;12(1):1334.
23. Alter BP, Baerlocher GM, Savage SA, et al. Very short telo­mere length by
flow fluo­res­cence in situ hybrid­iza­tion identifies patients with dyskeratosis
congenita. Blood. 2007;110(5):1439-1447.
24. Yoshizato T, Dumitriu B, Hosokawa K, et al. Somatic muta­tions and clonal © 2021 by The Amer­i­can Society of Hematology
hema­to­poi­e­sis in aplastic ane­mia. N Engl J Med. 2015;373(1):35-47. DOI 10.1182/hema­tol­ogy.2021000244

142  |  Hematology 2021  |  ASH Education Program


WHAT DO APLASTIC ANEMIA, PNH, AND “ HYPOPLASTIC ” LEUKEMIA HAVE IN COMMON ?

When does a PNH clone have clinical signifcance?


Daria V. Babushok

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/143/1852074/143babushok.pdf by guest on 13 December 2021


Division of Hematology-Oncology, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA; and Comprehensive Bone
Marrow Failure Center, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA

Paroxysmal nocturnal hemoglobinuria (PNH) is an acquired blood disease caused by somatic mutations in the phosphati-
dylinositol glycan class A (PIGA) gene required to produce glycophosphatidyl inositol (GPI) anchors. Although PNH cells
are readily identified by flow cytometry due to their deficiency of GPI-anchored proteins, the assessment of the clinical
significance of a PNH clone is more nuanced. The interpretation of results requires an understanding of PNH pathogene-
sis and its relationship to immune-mediated bone marrow failure. Only about one-third of patients with PNH clones have
classical PNH disease with overt hemolysis, its associated symptoms, and the highly prothrombotic state characteristic
of PNH. Patients with classical PNH benefit the most from complement inhibitors. In contrast, two-thirds of PNH clones
occur in patients whose clinical presentation is that of bone marrow failure with few, if any, PNH-related symptoms.
The clinical presentations are closely associated with PNH clone size. Although exceptions occur, bone marrow failure
patients usually have smaller, subclinical PNH clones. This review addresses the common scenarios that arise in evalu-
ating the clinical significance of PNH clones and provides practical guidelines for approaching a patient with a positive
PNH result.

LEARNING OBJECTIVES
• Interpret the validity and significance of PNH flow cytometry results
• Recognize the significance of PNH clones in the diagnostic assessment of patients with bone marrow failure
• Recognize indications for starting complement inhibitor therapy in PNH

Introduction prevalent in patients with immune-mediated bone mar-


Paroxysmal nocturnal hemoglobinuria (PNH) is an acquired row failure.8-10 Nearly half of patients with acquired aplastic
blood disease caused by somatic mutations in the phos- anemia (AA), a T-cell-mediated autoimmune bone marrow
phatidylinositol glycan class A (PIGA) gene.1 PIGA encodes failure disorder, develop PNH clones.8-10 In AA, PNH cells
an enzyme required for the first step of glycophosphatidyl are thought to evade the hematopoietic stem and progen-
inositol (GPI) anchor biosynthesis. PIGA-mutant (PNH) cells itor cell (HSPC)-directed autoimmune attack, and conse-
lack all GPI-anchored proteins, including 2 essential com- quently, PNH cells outgrow wild-type cells forming PNH
plement regulatory proteins, CD55 (complement decay- clones.1 PNH clones can also emerge in patients with myel-
accelerating factor) and CD59 (inhibitor of the complement odysplastic syndrome (MDS) and, more rarely, in patients
membrane attack complex). The deficiency of CD55 and with myeloproliferative neoplasms.11,12 Although immune-
CD59 makes PNH red blood cells (PNH RBCs) susceptible to mediated bone marrow failure is the primary risk factor for
complement-mediated hemolysis. developing PNH clones, patients can be diagnosed with
Although very small polyclonal populations of PNH cells classical PNH without a known diagnosis of bone marrow
of approximately 0.001% to 0.005% (approximately 10 to failure or a history of cytopenias. In such cases, prior sub-
50 per million) can be detected in most healthy individ- clinical marrow failure is frequently presumed; alternatively,
uals,2,3 PNH hematopoietic cells have no intrinsic growth secondary proliferative mutations within a PNH HSPC can
advantage and do not clonally expand under normal con- promote PNH clone expansion.1,13
ditions.4-6 Slightly larger PNH clones of 0.02% to 0.03% can Studies of patients with positive PNH flow cytometry
occur in approximately 1% of healthy individuals; however, results have found that PNH clones have a bimodal size
more significant clonal expansions of PNH cells do not occur distribution closely related to the patients’ clinical pre-
in the absence of pathology.2,3,7 In contrast, PNH clones are sentations12,14 (Figure 1, Table 1). Approximately one-third
Prakash Singh Shekhawat
Evaluation and significance of PNH clones | 143
cor­pus­cu­lar vol­ume of 121.4 fL, an abso­lute retic­u­lo­cyte count
of 45.8 × 103 cells/µL, and plate­lets of 29 × 103 cells/µL. The white
blood cells (WBCs) were low at 2.9 × 103 cells/µL with 58.9%
granulocytes, 6.2% mono­cytes, and 34.9% lym­pho­cytes. Bone
mar­row was hypocellular with­out dys­plas­tic changes. PNH flow
cytom­et­ ry revealed an iso­lated PNH clone of 2.9% in RBCs only,
with no PNH granulocytes. Lactate dehy­dro­ge­nase (LDH) was
nor­mal at 191 U/L. Haptoglobin was bor­der­line low at 32 mg/dL.
Given the absence of granulocyte PNH clone and the lack of
hemo­ly­sis, a false-pos­i­tive PNH clone was suspected. PNH flow
cytom­et­ ry test­ing was repeated, with no evi­dence of PNH cells
on sev­eral sub­se­quent tests. Additional workup showed short
lym­pho­cyte telo­mere lengths for age and a path­o­genic var­i­ant

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/143/1852074/143babushok.pdf by guest on 13 December 2021


in TERC, confirming the diag­no­sis of short telo­mere syn­drome.

Figure 1. The bimodal distribution of PNH clone sizes closely


correlates with the patients’ clinical presentations. The styl- The diverse clin­i­cal pre­sen­ta­tions and var­ied PNH cell type
ized schematic depicts the bimodal distribution of granulocyte involve­ment require sys­tem­atic con­sid­er­ation of sev­eral clin­i­cal
PNH clone sizes (black line) based on published PNH clone size and lab­or­a­tory fac­tors to deter­mine the sig­nif­i­cance of a “pos­
distributions.12,14 Red and blue area plots illustrate the different i­tive” (or a “neg­at­ive”) PNH flow cytom­et­ry result for a given
clinical presentations that correlate with the extremes of PNH patient. At a min­ i­
mum, an eval­ u­at­ion of a PNH result should
clone sizes. Patients with bone marrow failure who have an asso- include (1) PNH flow cytom­e­try, which should be performed on
ciated PNH clone are shown in red. Classical PNH presentations periph­eral blood with quan­ti­fi­ca­tion of PNH neu­tro­phils and PNH
are shown in blue. RBCs; (2) a bone mar­row aspi­rate and biopsy, includ­ing cyto­ge­
net­ics (test­ing for somatic muta­tions asso­ci­ated with hema­to­
logic malig­nan­cies can also be help­ful for prog­nos­tic assess­ment
of patients have “clas­si­cal PNH,” with overt hemo­ly­sis, mul­ti­ple of patients with hypocellular mar­row fail­ure13 and in patients in
PNH symp­toms, and an increased risk of throm­bo­ses. Patients whom PNH arose in asso­ci­a­tion with MDS or mye­lo­pro­lif­er­a­tive
with clas­si­cal PNH have a mean PNH clone size of more than neo­plasms); (3) lab­o­ra­tory stud­ies to eval­u­ate for hemo­ly­sis; and
70%.11,12 In con­trast, two-thirds of PNH clones occur in patients (4) his­tory assessing for prior throm­botic events and cytopenias
who have cytopenias and bone mar­row fail­ure with­out clin­i­cal (Figure 2). Here, I dis­cuss sev­eral com­mon clin­i­cal sce­nar­ios that
hemo­ly­sis. Most bone mar­row fail­ure patients are largely asymp­ arise when eval­ua ­ t­ing patients with a pos­i­tive PNH test to illus­
tom­atic from PNH and have small PNH clones (mean clone size of trate the sig­nif­i­cance of PNH clones in clas­si­cal PNH and bone
approx­i­ma­tely 11%) (Figure 1, Table 1).11,12 mar­row fail­ure.
PIGA muta­tions occur early in hema­to­poi­e­sis in hema­to­poi­
Interpreting the sig­nif­i­cance of a PNH flow etic stem cells or multipotent pro­gen­i­tors.7,12,14,15 In most patients,
cytom­e­try result PNH clones can be detected in all­or most mature hema­to­poi­
etic lin­e­ages, includ­ing granulocytes, mono­cytes, plate­lets,
RBCs, and the B, T, and nat­u­ral killer cells.7,12,15 More lim­ited lin­e­
CLINICAL CASE 1: FALSE-POSITIVE PNH TEST age involve­ment can occur but is more char­ac­ter­is­tic of MDS, in
A 55-year-old man with decades-long throm­bo­cy­to­pe­nia was which PNH clones are also more likely to be tran­sient.7
referred for an eval­u­a­tion of bone mar­row fail­ure. The patient had Next-gen­er­a­tion sequenc­ing of the PIGA gene in patients with
mac­ro­cytic ane­mia with a hemo­glo­bin of 10.6  g/dL and a mean PNH iden­ti­fied 2 or more inde­pen­dent PIGA muta­tions in 85% of

Table 1.  Clinical categories of patients with PNH clones

PNH granulocyte Laboratory mark­ers Risk of Utility of com­ple­ment


PNH cat­e­gory clone Clinical hemo­ly­sis of hemo­ly­sis PNH symp­toms throm­bo­sis inhib­i­tor ther­apy
Bone mar­row fail­ure with an asso­ci­ated PNH clone
  Subclinical Small (usu­ally Absent May be pres­ent, Absent Low Do not ben­e­fit from
(major­ity of <30%) depending on com­ple­ment inhib­i­tor
patients) clone size
 Symptomatic Moderate May be pres­ent May be pres­ent May be pres­ent Intermediate Some patients may
ben­e­fit from
com­ple­ment inhib­i­tor
Classical PNH Large (usu­ally Present Present Present High Complement inhib­i­tor
>50%) improves out­comes

144  |  Hematology 2021  |  ASH Education Program


Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/143/1852074/143babushok.pdf by guest on 13 December 2021
Figure 2. The recommended evaluation algorithm for patients with a newly identified PNH clone. CBC, complete blood count;
NGS, next-generation sequencing.

patients with PNH and at least 3 inde­pen­dent PIGA muta­tions Clinically, most flow cytom­e­try lab­o­ra­to­ries now use high-
in 57% of patients.16 Because mul­ti­ple inde­pen­dent muta­tions sen­si­tiv­ity PNH assay, allowing the detec­tion of PNH clones as
together com­prise PNH clones detected by flow cytom­e­try, flow small as 0.01% in granulocyte, mono­cyte, and RBC lin­e­ages. PNH
cytom­e­try is much more sen­si­tive than next-gen­er­a­tion sequenc­ granulocyte and mono­cyte pro­por­tions are closely cor­re­lated
ing for PNH detec­tion. In con­trast to genetic ana­ly­ses, which are in most patients.11,12 Unlike PNH RBCs, which are sus­cep­ti­ble to
more sen­si­tive when performed on bone mar­row than periph­ com­ple­ment-medi­ated hemo­ly­sis, PNH granulocytes have a nor­
eral blood by cap­tur­ing acquired genetic alter­ations in HSPCs, mal life span in vivo.17 For this rea­son, the PNH clone size is com­
the standard clin­ i­
cal PNH flow cytom­ e­
try eval­ u­
ates only the monly esti­mated using the granulocyte lin­e­age. At a min­i­mum,
mature cell pop­u­la­tions (granulocytes, mono­cytes, and RBCs) that PNH flow cytom­e­try should pro­vide a quan­ti­fi­ca­tion of PNH pro­
are well represented in periph­eral blood. PNH clones can thus be por­tions in granulocytes and RBCs.
sen­si­tively detected in periph­eral blood with­out a require­ment for To ensure that the iden­ti­fied GPI-neg­a­tive cell pop­u­la­tions rep­
a bone mar­row biopsy. Sometimes, sub­mit­ting a hypocellular bone re­sent PNH clones and are not cells with aber­rant expres­sion or an
mar­row spec­i­men for PNH flow cytom­e­try may par­a­dox­i­cally result inherited defi­ciency of a sin­gle GPI-anchored pro­tein,18 at least 2
in lower sen­si­tiv­ity for PNH detec­tion if the sub­mit­ted aspi­rate is of GPI-anchored mark­ers should be used to iden­tify PNH cells. The
an inad­e­quate vol­ume containing insuf­fi­cient granulocytes to detect incor­po­ra­tion of the fluo­ro­chrome-con­ju­gated nonlysing form
very small PNH clones. While PNH flow cytometry of other cell pop- of the proaerolysin toxin, which binds to the gly­can moi­ety of
ulations in the bone marrow can be performed, these have not GPI-anchored pro­teins, has fur­ther improved the sen­si­tiv­ity and
been standardized for high sensitivity PNH cell analysis and can be accu­racy of PNH leu­ko­cyte detec­tion.19 Granulocyte and mono­
affected by variation in cell surface marker expression across various cyte PNH clones are quan­ti­fied based on com­plete GPI-anchored
hematopoietic subsets. pro­tein defi­ciency. In con­trast, both the com­plete (type III) and
Prakash Singh Shekhawat
Evaluation and sig­nif­i­cance of PNH clones | 145
the par­tial (type II) GPI-anchored pro­tein defi­ciency are mea­sured fer­ence between the granulocyte and RBC PNH clone sizes, can
for RBCs. Type II cells have a small frac­tion of nor­mal GPI-anchored help inter­pret the accu­racy of both the pos­i­tive and neg­a­tive
pro­tein lev­els and arise from either hypo­mor­phic (mis­sense) PIGA PNH results in a given patient (Figure 2). Follow-up test­ing with
muta­tions or the pas­sive trans­fer of GPI-anchored pro­teins to PNH high-sen­si­tiv­ity PNH assay, performed according to the Inter-
cells.16,20 Rarely, type II cells may sig­nif­i­cantly out­num­ber type III national Clinical Cytometry Society/Euro­pean Society for Clin-
cells, in which case the under­ly­ing PIGA muta­tion is com­monly a ical Cell Analyses guide­lines, can clar­ify unex­pected results and
mis­sense var­i­ant in PIGA, lead­ing to a hypofunctioning allele and a resolve inconsistencies.
par­tial GPI-anchored pro­tein defi­ciency.16
In patients not receiv­ing com­ple­ment inhib­i­tors, PNH RBCs are PNH clones in immune-medi­ated bone mar­row fail­ure
sen­si­tive to com­ple­ment-medi­ated lysis and have a half-life of only
4 to 8 days.21 The short­ened life span of PNH RBCs causes a sig­nif­
i­cantly smaller appar­ent RBC PNH clone size com­pared with PNH
granulocytes. A cor­­ol­lary to this is that the lack of the expected CLINICAL CASE 2: DIAGNOSTIC SIGNIFICANCE OF PNH

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/143/1852074/143babushok.pdf by guest on 13 December 2021


clone size dif­fer­ence between PNH granulocytes and RBCs in an CLONES IN AA
untreated patient with PNH implies a nor­mal PNH RBC life span A 26-year-old pre­vi­ously healthy woman sought treat­ment from
and a lack of clin­i­cally sig­nif­i­cant hemo­ly­sis; in prac­tice, this can the emer­gency room after expe­ri­enc­ing 1 month of fatigue and
arise due to a pre­dom­i­nant type II clone that is less sus­cep­ti­ble dyspnea on exer­tion. Complete blood count revealed pan­cy­
to hemo­ly­sis. Because RBC trans­fu­sions will dilute the PNH RBC to­pe­nia, with WBCs of 1.07  ×  103 cells/µL, with 24.3% neu­tro­
frac­tion, a trans­fu­sion his­tory should be taken into account when phils and 75.7% lym­pho­cytes, a hemo­glo­bin of 4.3 g/dL with
interpreting PNH RBC clone size. When clin­i­cally fea­si­ble, PNH low abso­lute retic­u­lo­cytes of 23  ×  103 cells/µL, and plate­lets of
flow cytom­e­try should be performed before RBC trans­fu­sions. 12 × 103 cells/µL. Physical exam­i­na­tion was nota­ble for pal­lor and
Most PNH clones per­sist long term, of which some (approx­ scattered pete­chiae. There was no fam­ily his­tory of blood count
i­ma­tely 6% to 18%) will expand, while 17% to 24% of clones will abnor­mal­i­ties or con­di­tions sus­pi­cious for inherited mar­row fail­
become smaller over time.9,10,12,14,22 Thus, serial eval­u­a­tion (eg, ure syn­dromes. Additional lab­o­ra­tory stud­ies showed a mildly
annu­ally and with changes in hemo­lytic param­e­ters) is recom- ele­vated LDH of 239  U/L (1.2 × the upper limit of nor­mal [ULN] in
mended for all­patients with PNH clones to mon­i­tor for PNH the test­ing lab­o­ra­tory), a low hap­to­glo­bin less than 30  mg/dL,
clone expan­ sion. Periodic mon­ i­
tor­
ing can also be help­ ful in and nor­mal indi­rect bil­i­ru­bin of 0.9  mg/dL. The mar­row was
patients with clas­si­cal PNH, up to 15% of whom can undergo mark­edly hypocellular for age with­out dys­plas­tic changes. Cyto-
spon­ta­ne­ous remis­sion over time and may no lon­ger require genetics were nor­mal. Flow cytometry revealed a PNH clone of
ther­apy.13 Because most PNH clones arise in patients with AA, 19.11% in neu­tro­phils, 17.83% in mono­cytes, and 0.32% in RBCs.
annual PNH screen­ing is advised for patients with AA to iden­tify A diag­ no­sis of severe immune-medi­ ated AA with an asso­ ci­
newly emer­gent PNH clones.23 ated PNH clone was made. The patient had no matched sib­ling
Reflecting on case 1, the pat­tern of an iso­lated PNH RBC clone donors and was treated with immu­no­sup­pres­sive ther­apy with
with­out the involve­ment of granulocytes is unex­pected. This pat­ horse antithymocyte glob­u­lin, cyclo­spor­ine, and eltrombopag,
tern raises a pos­si­bil­ity of a false-pos­i­tive PNH clone or, per­haps, achiev­ing com­plete remis­sion. One year after ini­tial diag­no­sis,
a tran­sient sin­gle-lin­e­age PNH clone isolated to the ery­throid lin­ a fol­low-up eval­u­a­tion revealed a hemo­glo­bin of 11.9  g/dL and
e­age. Clinically, the patient has no lab­o­ra­tory or clin­i­cal hemo­ 48 × 103 retic­u­lo­cytes/µL with no clin­i­cal or lab­o­ra­tory evi­dence
ly­sis, which is not nec­es­sar­ily unex­pected given the rel­a­tively of hemo­ly­sis that included a nor­mal LDH of 186  U/L. Follow-up
small RBC clone size. Because PNH cells are iden­ti­fied by their PNH flow cytom­e­try showed a smaller PNH clone of 8.26% in
lack of staining for sur­face mark­ers, spe­cial­ized flow cytom­e­try neu­tro­phils, 7.6% in mono­cytes, and 1.59% in RBCs.
pro­to­cols are required to pre­vent falsely misidentifying debris
and other nonstaining cells as PNH clones.24 Historically, fail­ure
to use opti­mal meth­od­ol­ogy has been asso­ci­ated with false- Because the clonal expan­sion of PNH cells is closely linked to
­pos­i­tive results in 16% to 25% of clin­i­cal PNH tests in mul­ti­cen­ter the HSPC-directed auto­im­mune attack in AA,1 a PNH clone can
com­par­i­sons.11,25 The implementation of the International Clinical pro­vide an impor­tant diag­nos­tic clue about the immune-medi­
Cytometry Society/Euro­pean Society for Clinical Cell Analyses ated path­o­gen­e­sis of mar­row aplasia.8,26 The presenting fea­tures
con­sen­sus guide­lines for detec­tion of GPI-defi­cient cells in PNH of AA—pan­cy­to­pe­nia with hypocellular bone mar­row—can be
has dra­mat­i­cally improved the accu­racy and sen­si­tiv­ity of PNH brought on by dif­fer­ent eti­­ol­o­gies and are not spe­cific for the
test­ing. Compared with rou­tine PNH anal­y­sis suit­able for iden­ diag­no­sis of AA. For this rea­son, the diag­no­sis of AA requires
ti­fy­ing PNH clones more than 1%, which misses nearly 40% of sys­tem­atic exclu­sion of all­ alter­na­tive causes of mar­row fail­ure,
smaller PNH clones,19 the high sen­ si­tiv­
ity PNH assay enables includ­ing nutri­tional deficiencies, infec­tions, or tox­ins. Of par­tic­
accu­ rate iden­ti­
fi­
ca­
tion of clones down to 0.01%.19,24 Because u­lar impor­tance for chil­dren and young and mid­dle-aged adults
rare healthy indi­vid­u­als can have tran­sient expan­sions of PNH is the exclu­sion of inherited bone mar­row fail­ure syn­dromes,
cells reaching the level of detec­tion of the high-sen­si­tiv­ity PNH a costly and time-con­sum­ing pro­cess that can leave lin­ger­ing
assay,2,3,7 repeat­ing PNH flow cytom­e­try can help to ascer­tain diag­nos­tic uncertainties. Consequently, clin­i­cal test­ing with a
per­sis­tence of very rare pop­u­la­tions of PNH cells. high pos­i­tive pre­dic­tive value for immune-medi­ated AA can be
In sum, the tech­ ni­
cal char­ ac­ter­
is­tics of a given PNH flow valu­able by streamlining the diag­nos­tic eval­u­at­ ion and allowing
cytom­e­try assay can sig­nif­i­cantly affect the accu­racy of results. for prompt ini­ti­a­tion of AA-directed ther­a­pies.
A care­ful review of the PNH test­ing meth­od­ol­ogy, with close The pre­ dic­
tive value of PNH clones for the diag­ no­sis of
atten­tion to the assay sen­si­tiv­ity and spec­i­fic­ity param­e­ters, the immune-medi­ated AA and the exclu­sion of inherited bone mar­row
pres­ence of PNH cells in mul­ti­ple lin­e­ages, and the expected dif­ fail­ure was eval­ua ­ ted in 2 insti­tu­tional cohorts of bone mar­row

146  |  Hematology 2021  |  ASH Education Program


fail­ure patients8,26 (Table 2). PNH clones were iden­ti­fied in 46% of anom­a­lies and mod­er­ate cytopenias from birth who devel­oped
patients with AA but in none of the eval­u­ated inherited bone mar­ wors­en­ing cytopenias and new trans­fu­sion require­ments fol­low­
row fail­ure patients. A com­bined anal­y­sis of both cohorts showed ing inter­feron ther­apy for hep­a­ti­tis C. Finding a PNH clone, in this
that PNH granulocyte clones, regard­less of clone size, have an or sim­i­lar sit­u­a­tions of poorly explained cytopenias, can bring AA
approx­i­ma­tely 100% pos­i­tive pre­dic­tive value for AA and have higher on the dif­fer­en­tial diag­no­sis. In a hypo­thet­i­cal case with a
25-fold higher odds of being found in AA com­pared with inher- low pre­test prob­a­bil­ity of 2%, a pos­i­tive PNH clone would raise
ited bone mar­row fail­ure.8 Two addi­tional stud­ies found no PNH the post­test prob­a­bil­ity of AA into a mod­er­ate range of 65%.8
clones or PIGA muta­tions in more than 130 unse­lected patients Because PNH clones can emerge later in the dis­ease course,
with an inherited mar­row fail­ure dis­or­der, Shwachman-Diamond repeat­ing PNH flow cytom­e­try can help to iden­tify newly emer­
syn­drome (SDS),27,28 pro­vid­ing addi­tional sup­port to the spec­i­fic­ gent clones in patients who may have tested neg­a­tive at diag­
ity of PNH clones for immune-medi­ated AA. no­sis.23 PNH clones found later in the dis­ease course may be
In clin­i­
cal prac­tice, a Bayes the­ ory nomo­ gram tool, which par­tic­u­larly help­ful by affirming the immune-medi­ated eti­­ol­ogy
incor­po­rates the pre­test prob­a­bil­ity of AA, can be used to esti­ of AA in patients with refrac­tory AA, in whom the diag­no­sis of

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/143/1852074/143babushok.pdf by guest on 13 December 2021


mate the post­test prob­a­bil­ity of AA after a pos­i­tive PNH result immune-medi­ated AA may be in ques­tion.
(Figure 3).8 In Case 2, a pre­vi­ously healthy patient devel­oped In most stud­ies of AA out­comes, PNH clones, regard­less of clone
pan­cy­to­pe­nia as a young adult, with­out a fam­ily his­tory or phys­ size, were asso­ci­ated with improved response to immu­no­sup­pres­
i­cal exam­i­na­tion find­ings sug­ges­tive of a con­gen­i­tal mar­row fail­ sive ther­apy and bet­ter over­all prog­no­sis.9,13,29-34 The rea­sons for
ure dis­or­der. Based on this pre­sen­ta­tion, the clin­i­cal sus­pi­cion of improved responses to immu­no­sup­pres­sion in patients with PNH
immune-medi­ated acquired AA is mod­er­ate to high. Using a con­ clones are incom­pletely under­stood but may be par­tially due to a
ser­va­tive esti­mate of a mod­er­ate pre­test prob­a­bil­ity of AA (eg, more accu­rate diag­no­sis of immune-medi­ated AA. PNH clones are
60%), the pres­ence of a PNH clone would raise the post­test prob­ also asso­ci­ated with a lower rate of MDS pro­gres­sion.9,31,34
a­bil­ity of AA to more than 99%, allowing to con­fi­dently estab­lish
the diag­no­sis of immune-medi­ated AA and ini­ti­ate appro­pri­ate When does a PNH clone require therapy?
ther­ apy. Notably, a neg­ a­
tive PNH test does not sig­ nif­i­
cantly
change the pre­test prob­a­bil­ity of AA, and the absence of a PNH
clone should not be used to exclude AA. PNH test­ing can sim­i­larly
help in sit­u­a­tions where a pri­ori clin­i­cal sus­pi­cion of AA is low. CLINICAL CASE 3 (CLASSICAL PNH)
For exam­ple, a diag­no­sis of AA may be discounted in a young A 20-year-old male col­lege ath­lete sought treat­ment from the
adult with a known syndromic dis­or­der with mul­ti­ple con­gen­i­tal emer­gency room 3 times over the past 6 months with acute

Table 2.  Frequency of PNH in AA com­pared with inherited bone mar­row fail­ure dis­or­ders

Disease: num­ PNH test­ing Frequency of PNH


Study Year Population Study design ber of patients method clones Comparison
DeZern et al 26
2014 JHU cohort Retrospective AA: n = 132 Flow cytometry on AA: 61 of 132 (46%) • PPV for AA: 100%
IBMF: n = 20 periph­eral blood IBMF: 0 of 20 (0%) • NPV for AA: 54%
(sen­si­tiv­ity range
>0.01%-0.1%)
Shah et al8 2021 Penn/CHOP Retrospective AA: n = 126 Flow cytometry on AA: 58 of 126 (46%) • AA vs IBMF, OR
cohort Inherited: n = 9 periph­eral blood Inherited and other: 11.10, P < .05
Other: n = 13 (sen­si­tiv­ity range 0 of 22 (0%) • AA vs all­
>0.01%-1%) inherited dis­or­
ders includ­ing
IBMF, OR 16.23,
P < .05
• AA vs all­non-AA,
non-PNH, OR
38.43, P < .05
• PPV for AA vs
IBMF: 100%
• NPV for AA vs
IBMF: 48.5%
Keller et al27 2002 Camp Sunshine Retrospective n = 28 Flow cytometry on 0 of 28 patients (0%) NA
SDS cohort periph­eral blood
(PNH clones >1%)
Kennedy et al28 2014 SDS cohort Retrospective SDS: n = 99 Targeted, error- 0 of 110 patients NA
SDS-like: n = 11 corrected with PIGA muta­
sequenc­ing of tions (0%)
PIGA in bone
mar­row (>0.1%
VAF)
IBMF, inherited bone mar­row fail­ure; Inherited, inherited hema­to­logic dis­or­ders; JHU, Johns Hopkins University; NA, not applicable; NPV, neg­a­tive pre­dic­tive
Prakash Singh Shekhawat
value; OR, odds ratio; Penn/CHOP, University of Pennsylvania/Children’s Hospital of Philadelphia; PPV, pos­i­tive pre­dic­tive value; VAF, var­i­ant allele frac­tion.

Evaluation and sig­nif­i­cance of PNH clones | 147


lets of 109 × 103 cells/µL, which downtrended to WBCs of 4 × 103
cells/µL, hemo­ glo­bin of 10.3  g/dL, and plate­ lets of 95 × 103
cells/µL after intra­ ve­
nous hydra­ tion. LDH was ele­ vated at
596 IU/L (3.10 × ULN), hap­to­glo­bin was unde­tect­able, and abso­
lute retic­u­lo­cytes were 112  ×  103 cells/µL. The Coombs test
was neg­a­tive, and the D-dimer was ele­vated to 3.87  µg/mL.
Upper extrem­ity super­fi­cial throm­bo­phle­bi­tis was noted at an
old intra­ve­nous line place­ment site. Flow cytometry revealed
a PNH clone with 78.6% PNH granulocytes, 87.3% PNH mono­
cytes, and 0.8% type II and 11.2% type III PNH RBCs. The patient
recalled mul­ti­ple epi­sodes of darker-col­ored urine. A bone mar­
row biopsy, performed to assess for under­ly­ing bone mar­row
fail­ure, showed a 40% cel­lu­lar mar­row with­out dys­plas­tic fea­

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/143/1852074/143babushok.pdf by guest on 13 December 2021


tures, a nor­mal kar­yo­type, and a ­dis­ease-­asso­ci­ated somatic
BCOR muta­tion. The patient was diag­nosed with clas­si­cal
PNH, which likely arose from sub­clin­i­cal nonsevere AA. He was
started on folic acid and ini­ti­ated low-dose aspi­rin for man­age­
ment of throm­bo­phle­bi­tis and gen­eral thromboprophylaxis.
Following vac­ci­na­tion for Neisseria men­in­git­i­des, he started
eculizumab ther­apy with full symp­tom res­o­lu­tion. Once rav-
ulizumab became avail­­able, he was switched to ravulizumab
for patient con­ve­nience. Three years after the ini­tial diag­no­sis,
PNH flow cytom­e­try showed an expan­sion of his PNH clone to
92.8% in granulocytes, 94.1% in mono­cytes, and 36.5% in RBCs
(3.4% type II and 33.1% type III). Laboratory stud­ies performed
before the main­te­nance ravulizumab infu­sion showed WBCs
Figure 3. The Bayesian (Fagan’s) nomogram demonstrating of 3.9 × 103 cells/µL, hemo­glo­bin of 13.0  g/dL, and plate­lets of
the pretest and posttest probability of having a diagnosis of 162 × 103 cells/µL, with retic­u­lo­cytes of 217 × 103 cells/µL and LDH
­immune-mediated AA based on PNH flow cytometry results. of 282  U/L (1.47 × ULN).
PNH clones have a near 100% positive predictive value for the
diagnosis of immune-mediated AA, with a positive LR of approx-
imately 92 for immune-mediated AA.8 Shown is an example cor- The deci­sion to ini­ti­ate com­ple­ment inhib­i­tor ther­apy in a pa-
responding to Case 2 in the text. To apply the nomogram tool, tient with a PNH clone depends on mul­ti­ple fac­tors, includ­ing
one first has to estimate the pretest probability of AA based on clone size, clin­i­cal hemo­ly­sis, and symp­tom bur­den (Table 1).
the clinical presentation. This previously healthy patient devel- Patients whose pri­mary clin­i­cal pic­ture is that of bone mar­row
oped pancytopenia in young adulthood without any physical fail­ure on aver­age have smaller PNH clones, have lit­tle in the way
examination findings or family history suggestive of a congenital of PNH symp­toms, and rarely ben­efi ­ t from PNH-directed ther­
marrow failure disorder. Based on this presentation, her pretest apy. The ane­mia, fre­quently seen in patients with AA, most com­
probability of immune-mediated acquired AA is estimated as monly stems from the under­ly­ing bone mar­row fail­ure and is not
moderate to high. The green line shows the results for a con- caused by hemo­ly­sis. Thus, a care­ful eval­u­a­tion of the eti­­ol­ogy
servative estimate of a moderate (approximately 60%) pretest of ane­mia with par­tic­u­lar atten­tion to ade­quate retic­u­lo­cyte re-
probability of AA, crossing at the positive LR of 92. Detection of sponse is essen­tial when decid­ing on the util­ity of adding com­
a PNH clone increases the posttest probability of AA to 99.3%, ple­ment inhi­bi­tion to stan­dard AA ther­apy.
allowing to confidently establish the diagnosis of immune-medi- In con­trast, com­ple­ment inhib­i­tors improve out­comes in
ated AA and initiate appropriate therapy. Importantly, a negative patients with clas­si­cal PNH,35 who have overt hemo­ly­sis, are fre­
PNH test would not significantly change the pretest probability. quently highly symp­tom­atic, and invari­ably have large (gen­er­ally
Starting with a pretest probability of 60%, a negative PNH test >50%12,14) PNH clones. Nearly one-third of patients with clas­si­
(shown by the red line crossing at the negative LR of 0.54) will cal PNH have abdom­i­nal pain.36 Other clas­si­cal PNH symp­toms
result in a posttest probability of 44.9%. The calculations for the include fatigue, dyspnea, esoph­a­geal spasm, erec­tile dys­func­
Bayesian nomogram plot are based on Shah et al.8 tion, neu­ro­logic dys­func­tion, and gall­stone dis­ease. Before the
avail­abil­ity of com­ple­ment inhib­i­tors, 37% to 39% of patients
with clas­si­cal PNH had throm­botic events, fre­quently involv­ing
onset of nau­ sea, vomiting, and abdom­ i­
nal pain, each time unusual vas­cu­lar beds (eg, mes­en­teric and cere­bro­vas­cu­lar),
resolv­
ing with hydra­ tion and sup­ port­ive care. Contrast-en- which were the lead­ing cause of mor­tal­ity.36-38 Less rec­og­nized
hanced abdom­i­nal com­puted tomog­ra­phy revealed an edem­ seri­ous com­pli­ca­tions of PNH include sub­clin­i­cal pul­mo­nary
a­tous mes­en­tery with prominent mes­en­teric lymph nodes emboli and pul­mo­nary hyper­ten­sion,39 cere­bro­vas­cu­lar ische­
with­out other pathol­ogy. Liver func­tion stud­ies showed indi­ mia,40 and renal dys­func­tion.41
rect hyperbilirubinemia of 2.7  mg/dL and an ele­vated aspar­tate Risk fac­ tors for throm­ bo­sis in patients with PNH clones
ami­no­trans­fer­ase of 81  U/mL. A com­plete blood count showed include granulocyte clone size more than 50%, LDH 1.5 or more
WBCs of 12 × 103 cells/µL, hemo­glo­bin of 12.0  g/dL, and plate­ times the ULN, high PNH symp­tom bur­den, and pre­vi­ous throm­

148  |  Hematology 2021  |  ASH Education Program


Table 3.  Risk fac­tors for throm­botic com­pli­ca­tions in PNH

PNH test­ing Effect of PNH clone


Study Year Population Study design No. of patients method Risk fac­tor for throm­bo­sis size
de Latour 2008 Patients treated in 58 Retrospective 460 Ham test or flow • Age >55 years: HR 1.8 >50% clones asso­ci­
et al36 hema­to­logic cen­ cytom­e­try (PNH • Transfusions: HR 1.7 ated with higher
ters in France clones >5%) • Thrombosis at diag­no­sis: HR 3.7 risk of TE: HR 3.2
• Warfarin as pri­mary pro­phy­laxis: HR 5.2
Lee et al44 2013 South Korean Retrospective 301 Ham test, sucrose- • LDH ≥1.5 × ULN: OR 7.0 No sig­nif­i­cant effect
National PNH lysis test, and Higher pre­dic­tive value in com­bi­na­tion with of clone size
Registry flow cytom­e­ symp­toms: PNH clone <20%:
try (no min­i­mal • LDH ≥1.5 × ULN plus abdom­i­nal pain: OR 17.8 16% TE
cut­off) • LDH ≥1.5 × ULN plus chest pain: OR 19.0 PNH clone 20%-
• LDH ≥1.5 × ULN plus dyspnea: OR 10.3 50%: 19% TE
• LDH ≥1.5 × ULN plus hemo­glo­bin­uria: OR 10.3 PNH clone >50%:
20% TE
Schrezenmeier 2014 International PNH Retrospective 900 Clinical diag­no­ • LDH ≥1.5 × ULN: 15.6% (vs 8.4% <1.5 × ULN) PNH clone <10%:
et al45 Registry sis and/or flow • PNH clone ≥50% 5.3% TE
cytom­e­try (no PNH clone 10%-
min­i­mal cut­off) 49%: 7.7% TE
PNH clone ≥50%:
15.4% TE
Long et al46 2017 Peking Union Medical Retrospective 104 Flow cytometry • PNH clone ≥50%: OR 9.78 PNH clone ≥50%:
College Hospital (PNH clones >1%) • ABO gene rs495828 GT+TT: OR 5.63 OR 9.78
• ABO gene rs2519093 TC+TT: 5.95
Griffin et al. 2019 Leeds UK PNH Retrospective study 25 of 429 patients Flow cytometry • Group 1: PNH white cells >30%, PNH red cells In patients with
201947 Database of throm­bo­sis in had PNH clone (PNH clones <10%, LDH <2 × ULN: 6 of 11 (54%) had TE TEs, PNH clone

Prakash Singh Shekhawat


patients with PNH >10% and LDH >10%) • Group 2: PNH white cells >30%, PNH red ranged from 49%
with PNH clone <2 × ULN cells >10% with higher pro­por­tion of type to 100%; median
>10% and LDH II red cells than type III red cells, LDH clone sizes were
<2 × ULN) <2 × ULN: 2 of 11 (18%) had TE 93% (group 1) and
• Group 3: PNH white cells 10%-30%, PNH 89% (group 2)
red cells <10%, LDH <2 × ULN (0 of 3 had
throm­bo­sis)
Huang et al.48 2019 Peking Union Medical Retrospective 99 Flow cytometry • PNH clone ≥80%: OR 1.056 Only eval­u­ated cut­
College Hospital (PNH clones >1%) • Hemoglobin ≤7.5 g/dL: OR 4.202 off of PNH clone
• Platelets >100 × 109/L: OR 6.547 ≥80%: OR 1.056
• ABO gene rs495828 = G: OR 5.243
Hoechsmann 2020 International PNH Retrospective case 57 TE cases, 189 non- NA • Recent HDA: OR 2.65 PNH clone ≥50%
et al43 [ab­ Registry con­trol TE con­trols • LDH ≥1.5 × ULN plus 2-3 HDA symp­toms:
stract] OR 8.61
• LDH ≥1.5 × ULN plus ≥4 HDA symp­toms: OR
14.5
• History of TE: OR 3.6
• History of MAVE: OR 2.17
• Recent pro­phy­lac­tic anticoagulation: OR
4.35
Füreder et al49 2020 Austrian PNH net­ Retrospective 59 Flow cytometry 14 of 59 patients had TE; 5 of 14 patients had Larger clone size in
work clone size recorded, which ranged from patients with TE
80% to 100%

Evaluation and sig­nif­i­cance of PNH clones | 149


HDA, recent high dis­ease activ­ity, defined as occur­ring within 6 months of TE event, LDH ≥1.5 × ULN, and hemo­glo­bin <10 g/dL or at least 1 of the fol­low­ing symp­toms: abdom­i­nal pain, dyspnea,
dys­pha­gia, fatigue, hemo­glo­bin­uria, and male erec­tile dys­func­tion; HR, haz­ard ratio; MAVE, major adverse vas­cu­lar event; OR, odds ratio; TE, throm­bo­em­bolic event.
Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/143/1852074/143babushok.pdf by guest on 13 December 2021
botic events (Table 3).36-38,42-44 When eval­ua ­ t­ing LDH, it is impor­ com­ple­ment cas­cade, was found to be supe­rior to eculizumab
tant to com­pare a patient’s LDH result with the ref­er­ence range in improv­ing hemo­glo­bin and nor­mal­iz­ing hemo­lytic mark­ers.57
from the test­ing lab­o­ra­tory, as nor­mal LDH val­ues vary between Approved for both front­line or sec­ond-line ther­apy of PNH, the
test­ing sites due to var­i­a­tions in test­ing meth­od­ol­o­gies and plat­ twice-weekly, sub­cu­ta­ne­ously admin­is­tered agent pegcetaco-
forms. RBC clones more than 3% to 5% and granulocyte clones plan will be par­tic­u­larly use­ful in patients who remain mod­er­ately
over approx­i­ma­tely 23% are pre­dic­tive of ele­vated LDH, with or severely ane­mic despite C5 inhi­bi­tion because of extra­vas­cu­lar
each 10% rise in granulocyte clone size asso­ci­ated with approx­i­ hemo­ly­sis. Several other prox­i­mal com­ple­ment inhib­i­tors, some
ma­tely 1.6 higher odds of throm­bo­sis.37 Although PNH clone size of which are orally admin­is­tered, are in clin­ic­ al tri­als.58
cor­re­lates closely with hemo­ly­sis and LDH,45 the rela­tion­ship
between clone size, hemo­ly­sis, and throm­botic risk is not always Summary
lin­ear. Rare patients with more than 50% PNH granulocytes have PNH remains a very rare, orphan dis­ease with a widely avail­­able
devel­oped throm­botic events despite a near-nor­mal or nor­mal and highly sen­si­tive screen­ing test, the results of which can have
LDH.47 Conversely, patients with PNH clones smaller than 50% nuanced impli­ca­tions for diag­no­sis, fol­low-up, and com­ple­ment

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/143/1852074/143babushok.pdf by guest on 13 December 2021


can be at increased risk of throm­bo­sis.44,45 Although hypo­mor­ inhib­i­tor ther­apy. PNH clones are patho­gno­monic for immune-
phic PIGA muta­tions cause only par­tial GPI-anchored pro­tein medi­ated bone mar­row fail­ure. Patients with AA, who com­prise
defi­ ciency (type II cells), which could be less sus­ cep­ ti­
ble to most indi­vid­u­als with PNH clones, have smaller PNH clones and
hemo­ly­sis, throm­bo­ses in patients with type II ­PNH-pre­dom­i­nant are rarely symp­tom­atic from PNH. In the absence of hemo­ly­sis
clones have been reported.47 or PNH symp­toms, patients with AA with sub­clin­i­cal PNH clones
In Case 3, the patient has near-nor­mal hemo­glo­bin and nota­ do not ben­e­fit from com­ple­ment inhib­i­tors but should be mon­
bly does not require ther­apy due to ane­mia. However, he is at i­tored pro­spec­tively as most clas­si­cal PNH arises out of AA. In
high risk of throm­botic com­pli­ca­tions. His risk fac­tors for throm­ con­trast, com­ple­ment inhib­i­tors improve out­comes of patients
bo­sis include a large PNH clone more than 50%, overt hemo­ly­sis with clas­si­cal PNH, who invari­ably have large PNH clones, clin­i­
with LDH more than 3 times the ULN, high symp­tom bur­den, and cal hemo­ly­sis, and a high bur­den of PNH-asso­ci­ated symp­toms.
pre­vi­ous super­fi­cial throm­bo­phle­bi­tis. As evidenced by mes­en­ An indi­vid­u­al­ized approach is required when eval­u­at­ing patients
teric edema and ele­vated D-dimer, his abdom­i­nal pain is likely between these extremes, and refer­ral to a ter­tiary care cen­ter
caused by micro­vas­cu­lar ische­mia in the mes­en­teric vas­cu­la­ture. with exper­tise in man­ag­ing PNH and AA is recommended.
He requires prompt ini­ti­a­tion of a com­ple­ment inhib­i­tor to pre­
vent life-threat­en­ing throm­botic com­pli­ca­tions. Acknowledgments
The prothrombotic state in PNH is man­aged most effec­tively The author would like to thank the cur­rent and for­mer mem­
by com­ple­ment inhi­bi­tion, which reduces throm­botic events from bers of the Penn/CHOP Bone Marrow Failure Center for help­ful
approx­i­ma­tely 7.4% to 1% per patient-year.50 The mech­a­nism of dis­cus­sions as well as patients and their fam­i­lies for par­tici­pat­
throm­bo­sis in PNH involves abnor­mal plate­let acti­va­tion by both ing in research stud­ies of bone mar­row fail­ure. This work was
com­ple­ment and hemo­lytic pro­cesses as well as abnor­mal clot ­supported by NHLBI grant K08 HL132101.
com­po­si­tion.37 Untreated patients with PNH have higher fibrin­
o­gen and throm­bin gen­er­a­tion lev­els, lead­ing to faster-forming Conflict-of-inter­est dis­clo­sure
fibrin clots that are harder to break down; these abnor­mal­i­ties Daria V. Babushok: declares no com­pet­ing finan­cial inter­ests.
are improved by eculizumab.51 Primary anticoagulation pro­phy­
laxis has been pro­posed for patients with more than 50% PNH Off-label drug use
granulocytes who have no con­tra­in­di­ca­tions to anticoagulation.38 Daria V. Babushok: no off-label drug use discussed.
However, pri­ mary anticoagulation pro­ phy­
laxis in PNH remains
con­tro­ver­sial. Patients with PNH who have con­com­i­tant throm­ Correspondence
bo­cy­to­pe­nia have increased risks of bleed­ing with anticoagula- Daria V. Babushok, Room 808 Biomedical Research Build-
tion, which, before the advent of com­ple­ment inhib­i­tors, have ing II/III, 421 Curie Blvd, Philadelphia, PA 19104; e-mail: daria​
con­trib­uted to PNH mor­tal­ity. In addi­tion, unlike com­ple­ment ­.babushok@pennmedicineupenn​­.edu.
inhib­i­tors, ther­a­peu­tic anticoagulation alone does not entirely
pre­vent throm­bo­sis in patients with PNH.37,38,50 In sit­u­a­tions where References
patients with clas­si­cal PNH do not have access to com­ple­ment 1. Hill A, DeZern AE, Kinoshita T, Brodsky RA. Paroxysmal noc­tur­nal haemo-
inhib­i­tors, pri­mary anticoagulation pro­phy­laxis should be con­sid­ globinuria. Nat Rev Dis Primers. 2017;3:17028.
ered. After com­ple­ment inhib­i­tor ther­apy has been ini­ti­ated, pri­ 2. Hu R, Mukhina GL, Piantadosi S, Barber JP, Jones RJ, Brodsky RA. PIG-A
muta­tions in nor­mal hema­to­poi­e­sis. Blood. 2005;105(10):3848-3854.
mary anticoagulation pro­phy­laxis can be discontinued. 3. Araten DJ, Nafa K, Pakdeesuwan K, Luzzatto L. Clonal pop­u­la­tions of hema­
As of 2021, the US Food and Drug Administration has now to­poi­etic cells with par­ox­ys­mal noc­tur­nal hemo­glo­bin­uria geno­type and
approved 3 com­ple­ment inhib­it­ ors for the treat­ment of PNH. Ecu- phe­no­type are pres­ent in nor­mal indi­vid­u­als. Proc Natl Acad Sci U S A.
lizumab and the lon­ger-act­ing ravulizumab both tar­get ter­mi­nal 1999;96(9):5209-5214.
4. Shin T-H, Baek E-J, Corat M-A-F, et al. CRISPR/Cas9 PIG -A gene editing in
com­ple­ment C5 and effec­tively abro­gate intra­vas­cu­lar hemo­ly­sis,
non­hu­man pri­mate model dem­on­strates no intrin­sic clonal expan­sion of
reduce throm­botic events, and improve qual­ity of life of patients PNH HSPCs. Blood. 2019;133(23):2542-2545.
with PNH.52-55 However, approx­ im
­ a­tely 4% to 27% of patients 5. Murakami Y, Kinoshita T, Maeda Y, Nakano T, Kosaka H, Takeda J. Differ-
treated with C5 inhib­i­tors con­tinue to expe­ri­ence ane­mia due ent roles of glycosylphosphatidylinositol in var­i­ous hema­to­poi­etic cells
to extra­vas­cu­lar hemo­ly­sis from the accu­mu­la­tion of unop­posed as revealed by a mouse model of par­ox­ys­mal noc­tur­nal hemo­glo­bin­uria.
Blood. 1999;94(9):2963-2970.
C3b on PNH RBCs.56 Although C5 inhi­bi­tion does not address 6. Keller P, Tremml G, Rosti V, Bessler M. X inac­ti­va­tion and somatic cell selec­
extra­vas­cu­lar hemo­ly­sis, the recently approved, first-in-class tion res­cue female mice car­ry­ing a Piga-null muta­tion. Proc Natl Acad Sci
C3 inhib­i­tor pegcetacoplan, which acts more prox­i­mally in the U S A. 1999;96(13):7479-7483.

150  |  Hematology 2021  |  ASH Education Program


7. Katagiri T, Kawamoto H, Nakakuki T, et al. Individual hema­to­poi­etic stem 29. Zhao X, Zhang L, Jing L, et al. The role of par­ox­ys­mal noc­tur­nal hemo­glo­
cells in human bone mar­row of patients with aplastic ane­mia or myelo- bin­uria clones in response to immu­no­sup­pres­sive ther­apy of patients with
dysplastic syn­drome sta­bly give rise to lim­ited cell lin­e­ages. Stem Cells. severe aplastic ane­mia. Ann Hematol. 2015;94(7):1105-1110.
2013;31(3):536-546. 30. Sugimori C, Chuhjo T, Feng X, et  al. Minor pop­u­la­tion of CD55–CD59–
8. Shah YB, Priore SF, Li Y, et al. The pre­dic­tive value of PNH clones, 6p CN- blood cells pre­dicts response to immu­no­sup­pres­sive ther­apy and prog­no­
LOH, and clonal TCR gene rearrangement for aplastic ane­mia diag­no­sis. sis in patients with aplastic ane­mia. Blood. 2006;107(4):1308-1314.
Blood Adv. 2021;5(16):3216-3226. 31. Wang H, Chuhjo T, Yasue S, Omine M, Nakao S. Clinical sig­nif­i­cance of a
9. Fattizzo B, Ireland R, Dun­lop A, et al. Clinical and prog­nos­tic sig­nif­i­cance minor pop­u­la­tion of par­ox­ys­mal noc­tur­nal hemo­glo­bin­uria-type cells in
of small par­ox­ys­mal noc­tur­nal hemo­glo­bin­uria clones in myelodysplastic bone mar­row fail­ure syn­drome. Blood. 2002;100(12):3897-3902.
syn­drome and aplastic ane­mia [published online 4 March 2021]. Leukemia. 32. Ishiyama K, Chuhjo T, Wang H, Yachie A, Omine M, Nakao S. Polyclonal
10. Sugimori C, Mochizuki K, Qi Z, et al. Origin and fate of blood cells defi­cient hema­to­poi­e­sis maintained in patients with bone mar­row fail­ure har­bor­
in glycosylphosphatidylinositol-anchored pro­ tein among patients with ing a minor pop­u­la­tion of par­ox­ys­mal noc­tur­nal hemo­glo­bin­uria-type cells.
bone mar­row fail­ure. Br J Haematol. 2009;147(1):102-112. Blood. 2003;102(4):1211-1216.
11. Richards SJ, Whitby L, Cullen MJ, et  al. Development and eval­u­a­tion of 33. Wang B, He B, Zhu Y-D, Wu W. The pre­dic­tive value of pre-treat­ment
a sta­bi­lized whole-blood prep­a­ra­tion as a pro­cess con­trol mate­rial for par­ox­ys­mal noc­tur­nal hemo­glo­bin­uria clone on response to immu­no­sup­
screen­ing of par­ox­ys­mal noc­tur­nal hemo­glo­bin­uria by flow cytom­e­try. pres­sive ther­apy in patients with aplastic ane­mia: a meta-anal­y­sis. Hema-

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/143/1852074/143babushok.pdf by guest on 13 December 2021


Cytometry B Clin Cytom. 2009;76(1):47-55. tology. 2020;25(1):464-472.
12. Cannizzo E, Raia M, De Propris MS, et al. Features, rea­son for test­ing, and 34. Yoshizato T, Dumitriu B, Hosokawa K, et al. Somatic muta­tions and clonal
changes with time of 583 par­ox­ys­mal noc­tur­nal hemo­glo­bin­uria clones from hema­to­poi­e­sis in aplastic ane­mia. N Engl J Med. 2015;373(1):35-47.
529 patients: a mul­ti­cen­ter Ital­ian study. Ann Hematol. 2019;98(5):1083- 35. Kelly RJ, Hill A, Arnold LM, et al. Long-term treat­ment with eculizumab in
1093. par­ox­ys­mal noc­tur­nal hemo­glo­bin­uria: sustained effi­cacy and improved
13. Sun L, Babushok DV. Secondary myelodysplastic syn­drome and leu­ke­mia sur­vival. Blood. 2011;117(25):6786-6792.
in acquired aplastic ane­mia and par­ox­ys­mal noc­tur­nal hemo­glo­bin­uria. 36. de Latour RP, Mary JY, Salanoubat C, et al; French Society of Hematol-
Blood. 2020;136(1):36-49. ogy; French Association of Young Hematologists. Paroxysmal noc­ tur­
14. Richards SJ, Dickinson AJ, Cullen MJ, et al. Presentation clin­i­cal, haemato- nal hemo­glo­bin­uria: nat­u­ral his­tory of dis­ease subcategories. Blood.
logical and immunophenotypic fea­tures of 1081 patients with GPI-defi­cient 2008;112(8):3099-3106.
(par­ox­ys­mal noc­tur­nal haemoglobinuria) cells detected by flow cytom­e­try. 37. Hill A, Kelly RJ, Hillmen P. Thrombosis in par­ox­ys­mal noc­tur­nal hemo­glo­bin­
Br J Haematol. 2020;189(5):954-966. uria. Blood. 2013;121(25):4985-4996, quiz 5105.
15. Yoroidaka T, Hosokawa K, Imi T, et al. Hematopoietic stem pro­gen­i­tor cells 38. Hall C, Richards S, Hillmen P. Primary pro­ phy­ laxis with war­ fa­
rin pre­
lacking HLA dif­fer from those lacking GPI-anchored pro­teins in the hier­ vents throm­bo­sis in par­ox­ys­mal noc­tur­nal hemo­glo­bin­uria (PNH). Blood.
ar­chi­cal stage and sen­si­tiv­ity to immune attack in patients with acquired 2003;102(10):3587-3591.
aplastic ane­mia [published online 6 April 2021]. Leukemia. 39. Hill A, Sapsford RJ, Scally A, et  al. Under-rec­ og­ nized com­ pli­
ca­tions in
16. Gurnari C, Pagliuca S, Patel BJ, et al. Implication of PIGA geno­type on eryth­ patients with par­ox­ys­mal noc­tur­nal haemoglobinuria: raised pul­mo­
ro­cytes phe­no­type in par­ox­ys­mal noc­tur­nal hemo­glo­bin­uria. Leukemia. nary pres­sure and reduced right ven­tric­u­lar func­tion. Br J Haematol.
2021;35(8):2431-2434. 2012;158(3):409-414.
17. Brubaker LH, Essig LJ, Mengel CE. Neutrophil life span in par­ox­ys­mal noc­ 40. Barcellini W, Scola E, Lanfranconi S, et al. Paroxysmal noc­tur­nal hemo­glo­
tur­nal hemo­glo­bin­uria. Blood. 1977;50(4):657-662. bin­uria (PNH): brain MRI ische­mic lesions in neu­ro­log­i­cally asymp­tom­atic
18. Reid ME. Cromer-related blood group anti­gens and the gly­co­syl phospha- patients. Sci Rep. 2018;8(1):476.
tidylinositol-linked pro­tein, decay-accel­er­at­ing fac­tor DAF (CD55). Immu- 41. Kokoris SI, Gavriilaki E, Miari A, et al. Renal involve­ment in par­ox­ys­mal noc­
nohematology. 1990;6(2):27-29. tur­nal hemo­glo­bin­uria: an update on clin­i­cal fea­tures, path­o­phys­i­­ol­ogy
19. Borowitz MJ, Craig FE, Digiuseppe JA, et al; Clinical Cytometry Society. and treat­ment. Hematology. 2018;23(8):558-566.
Guidelines for the diag­no­sis and mon­i­tor­ing of par­ox­ys­mal noc­tur­nal 42. Ge M, Li X, Shi J, Shao Y, Zheng Y. Clinical fea­tures and prog­nos­tic fac­tors
hemo­glo­bin­uria and related dis­or­ders by flow cytom­e­try. Cytometry B of Asian patients with par­ox­ys­mal noc­tur­nal hemo­glo­bin­uria: results from a
Clin Cytom. 2010;78(4):211-230. sin­gle cen­ter in China. Ann Hematol. 2012;91(7):1121-1128.
20. Sloand EM, Mainwaring L, Keyvanfar K, et al. Transfer of glycosylphosphati- 43. Hoechsmann B, Peffault De Latour R, Hill A, et al. Risk fac­tors for throm­
dylinositol-anchored pro­teins to defi­cient cells after eryth­ro­cyte trans­fu­sion botic events in patients with PNH: a nested case-con­trol study in the inter­
in par­ox­ys­mal noc­tur­nal hemo­glo­bin­uria. Blood. 2004;104(12):3782-3788. na­tional PNH reg­is­try. Blood. 2020;136(suppl 1):6-8.
21. Rosse WF. The life-span of com­ple­ment-sen­si­tive and -insen­si­tive red cells 44. Lee JW, Jang JH, Kim JS, et al. Clinical signs and symp­toms asso­ci­ated with
in par­ox­ys­mal noc­tur­nal hemo­glo­bin­uria. Blood. 1971;37(5):556-562. increased risk for throm­bo­sis in patients with par­ox­ys­mal noc­tur­nal hemo­
22. Le Garff-Tavernier M, Debliquis A, Boyer T, et al. Persistence of PNH clones glo­bin­uria from a Korean Registry. Int J Hematol. 2013;97(6):749-757.
over time: insights from the mid-term anal­y­sis of the French nation­wide 45. Schrezenmeier H, Muus P, Socié G, et al. Baseline char­ac­ter­is­tics and dis­
mul­ti­cen­ter pro­spec­tive obser­va­tional study. Amer­i­can Society of Hema- ease bur­den in patients in the International Paroxysmal Nocturnal Hemo-
tology. Blood. 2019;134(suppl 1):1218. globinuria Registry. Haematologica. 2014;99(5):922-929.
23. Killick SB, Bown N, Cavenagh J, et al; Brit­ish Society for Standards in Hae- 46. Long Z, Du Y, Li H, Han B. Polymorphism of the ABO gene asso­ci­ate with
matology. Guidelines for the diag­no­sis and man­age­ment of adult aplastic throm­bo­sis risk in patients with par­ox­ys­mal noc­tur­nal hemo­glo­bin­uria.
anae­mia. Br J Haematol. 2016;172(2):187-207. Oncotarget. 2017;8(54):92411-92419.
24. Illingworth AJ, Marinov I, Sutherland DR. Sensitive and accu­rate iden­ti­fi­ca­ 47. Griffin M, Hillmen P, Munir T, et  al. Significant hemo­ly­sis is not required
tion of PNH clones based on ICCS/ESCCA PNH Consensus Guidelines—a for throm­bo­sis in par­ox­ys­mal noc­tur­nal hemo­glo­bin­uria. Haematologica.
sum­mary. Int J Lab Hematol. 2019;41 (suppl 1):73-81. 2019;104(3):e94-e96.
25. Debliquis A, Wagner-Ballon O, Le Garff-Tavernier M, et al; HPN-AFC Group. 48. Huang Y, Liu X, Chen F, et al. Prediction of throm­bo­sis risk in patients with
Evaluation of par­ox­ys­mal noc­tur­nal hemo­glo­bin­uria screen­ing by flow par­ox­ys­mal noc­tur­nal hemo­glo­bin­uria. Ann Hematol. 2019;98(10):2283-
cytom­e­try through multicentric interlaboratory com­par­i­son in four countries. 2291.
Am J Clin Pathol. 2015;144(6):858-868. 49. Füreder W, Sperr WR, Heibl S, et  al. Prognostic fac­tors and fol­low-up
26. DeZern AE, Symons HJ, Resar LS, Borowitz MJ, Armanios MY, Brodsky RA. param­e­ters in patients with par­ox­ys­mal noc­tur­nal hemo­glo­bin­uria (PNH):
Detection of par­ox­ys­mal noc­tur­nal hemo­glo­bin­uria clones to exclude inher- expe­ri­ence of the Austrian PNH net­work. Ann Hematol. 2020;99(10):2303-
ited bone mar­row fail­ure syn­dromes. Eur J Haematol. 2014;92(6):467-470. 2313.
27. Keller P, Debaun MR, Rothbaum RJ, Bessler M. Bone mar­row fail­ure in Shwa- 50. Hillmen P, Muus P, Dührsen U, et  al. Effect of the com­ple­ment inhib­i­tor
chman-Diamond syn­drome does not select for clonal haematopoiesis of eculizumab on throm­bo­em­bo­lism in patients with par­ox­ys­mal noc­tur­nal
the par­ox­ys­mal noc­tur­nal haemoglobinuria phe­no­type. Br J Haematol. hemo­glo­bin­uria. Blood. 2007;110(12):4123-4128.
2002;119(3):830-832. 51. Macrae FL, Peacock-Young B, Bowman P, et al. Patients with par­ox­ys­mal
28. Kennedy AL, Myers KC, Bowman J, et al. Distinct genetic path­ways define noc­tur­nal hemo­glo­bin­uria dem­on­strate a prothrombotic clot­ting phe­no­
pre-malig­nant ver­sus com­pen­sa­tory clonal hema­to­poi­e­sis in Shwachman- type which is improved by com­ple­ment inhi­bi­tion with eculizumab. Am J
Diamond syn­drome. Nat Commun. 2021;12(1):1334. Hematol. 2020;95(8):944-952.
Prakash Singh Shekhawat
Evaluation and sig­nif­i­cance of PNH clones | 151
52. Hillmen P, Young NS, Schubert J, et al. The com­ple­ment inhib­i­tor eculizumab ies of ravulizumab ver­sus eculizumab in adults with par­ox­ys­mal noc­tur­nal
in par­ox­ys­mal noc­tur­nal hemo­glo­bin­uria. N Engl J Med. 2006;355(12):1233- hemo­glo­bin­uria. Haematologica. 2021;106(1):230-237.
1243. 57. Hillmen P, Szer J, Weitz I, et al. Pegcetacoplan ver­sus eculizumab in par­ox­
53. Brodsky RA, Young NS, Antonioli E, et al. Multicenter phase 3 study of the ys­mal noc­tur­nal hemo­glo­bin­uria. N Engl J Med. 2021;384(11):1028-1037.
com­ple­ment inhib­i­tor eculizumab for the treat­ment of patients with par­ox­ 58. Risitano AM, Marotta S. Toward com­ple­ment inhi­bi­tion 2.0: next gen­er­a­
ys­mal noc­tur­nal hemo­glo­bin­uria. Blood. 2008;111(4):1840-1847. tion anticomplement agents for par­ox­ys­mal noc­tur­nal hemo­glo­bin­uria. Am
54. Lee JW, Sicre de Fontbrune F, Wong Lee Lee L, et  al. Ravulizumab J Hematol. 2018;93(4):564-577.
(ALXN1210) vs eculizumab in adult patients with PNH naive to com­ple­ment
inhib­i­tors: the 301 study. Blood. 2019;133(6):530-539.
55. Kulasekararaj AG, Hill A, Rottinghaus ST, et al. Ravulizumab (ALXN1210) vs
eculizumab in C5-inhib­i­tor-expe­ri­enced adult patients with PNH: the 302
study. Blood. 2019;133(6):540-549.
56. Brodsky RA, Peffault de Latour R, Rottinghaus ST, et al. Characterization of © 2021 by The Amer­i­can Society of Hematology
break­through hemo­ly­sis events observed in the phase 3 ran­dom­ized stud­ DOI 10.1182/hema­tol­ogy.2021000245

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/143/1852074/143babushok.pdf by guest on 13 December 2021

152  |  Hematology 2021  |  ASH Education Program


WHAT DO APLASTIC ANEMIA, PNH, AND “ HYPOPLASTIC ” LEUKEMIA HAVE IN COMMON ?

When to worry about inherited bone marrow failure


and myeloid malignancy predisposition syndromes
in the setting of a hypocellular marrow

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/153/1851462/153narla.pdf by guest on 13 December 2021


Anupama Narla
Stanford University School of Medicine, Stanford, CA

With our increasing understanding of inherited marrow failure and myeloid malignancy predisposition syndromes, it has
become clear that there is a wide phenotypic spectrum and that these diseases must be considered in the differential
diagnosis of both children and adults with unexplained defects in hematopoiesis. Moreover, these conditions are not as
rare as previously believed and may present as aplastic anemia, myelodysplastic syndrome, or malignancy over a range
of ages. Establishing the correct diagnosis is essential because it has implications for treatment, medical management,
cancer screening, and family planning. Our goal is to highlight insights into the pathophysiology of these diseases,
review cryptic presentations of these syndromes, and provide useful references for the practicing hematologist.

LEARNING OBJECTIVES
• Review the basic clinical features of the inherited BM failure and myeloid malignancy predisposition syndromes
that can lead to aplastic anemia, myelodysplasia, and malignancy
• Emphasize the role of a thorough physical exam and detailed family history to identify these disorders and deter­
mine the need for genetic testing for the patient and family
• Discuss the initial diagnostic workup of these disorders and potential treatment modifications if a specific condi­
tion is identified

Introduction: Inherited bone marrow failure syndromes presents in infancy and progresses to pancytopenia and
Inherited bone marrow failure syndromes (iBMFs) encom­ bone marrow (BM) failure in later childhood. Clinical man­
pass a diverse collection of diseases. While they are rare ifestations can include petechiae at birth and intracranial
causes of hematologic disorders, it is essential for the hemorrhage, but unlike other iBMFs, congenital anomalies
practicing hematologist to be aware of iBMFs. These dis­ are rare. CAMT is most commonly due to mutations in MPL,
orders have a wide phenotypic spectrum and may pres­ which encodes for the thrombopoietin receptor. In contrast
ent cryptically in adult patients with cytopenias in one or to the activating MPL mutations seen in myeloproliferative
more lineages. Furthermore, our evolving understanding neoplasms, CAMT mutations always lead to diminished or
of the pathophysiology of these disorders is critical to absent signaling of the thrombopoietin receptor. In gen­
our general knowledge of the hematopoietic system. An eral, loss­of­function mutations are associated with more
overview of these syndromes is summarized in Table 1. We severe thrombocytopenia and early­onset pancytopenia
have highlighted the disorders that have a predisposition (CAMT type 1). In cases with MPL gene missense mutations,
to aplastic anemia, myelodysplastic syndrome (MDS), and patients have a transient increase in platelet counts dur­
malignancy. A brief overview of each of these disorders ing the first year of life (CAMT type 2). Heterozygous muta­
follows. tions in MECOM (MDS1 and EVI1 complex locus) have been
reported to be causative of a rare association of CAMT and
Congenital amegakaryocytic thrombocytopenia radioulnar synostosis. Mutations in MECOM can present
Congenital amegakaryocytic thrombocytopenia (CAMT) is with a wide range of phenotypes ranging from thrombo­
a rare autosomal recessive disease characterized by an iso­ cytopenia to full­blown hypocellular marrow failure/aplastic
lated, severe hypomegakaryocytic thrombocytopenia that anemia early in life, again demonstrating the importance of
Prakash Singh Shekhawat
Bone marrow failure syndromes and aplastic anemia | 153
Table 1. Features of inherited bone nar­row and mye­loid malig­nancy pre­dis­po­si­tion syn­dromes that may pres­ent as aplastic
ane­mia/hypocellular mar­row

Disease Presentation Useful clin­i­cal resources


Amegakaryocytic thromobocytopenia Nonsyndromic throm­bo­cy­to­pe­nia, pan­cy­to­pe­nia, https:​­/​­/www​­.pdsa​­.org​­/inherited​
BM fail­ure ­-thrombocytopenia​­.html
Diamond-Blackfan ane­mia Macrocytic ane­mia with reticulocytopenia, con­gen­i­tal https:​­/​­/dbafoundation​­.org​­/
anom­a­lies/short stat­ure https:​­/​­/www​­.dbar​­.org​­/
Dyskeratosis congenita Triad of abnor­mal skin pig­men­ta­tion, oral leukoplakia, https:​­/​­/teamtelomere​­.org​­/
and nail dys­tro­phy; aplastic ane­mia, pul­mo­nary fibro­sis
Fanconi ane­mia Cytopenias, con­gen­i­tal malformations, can­cer at a young https:​­/​­/www​­.fanconi​­.org​­/
age, sen­si­tiv­ity to toxic effects of che­mo­ther­apy or
radi­a­tion, MDS/AML

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/153/1851462/153narla.pdf by guest on 13 December 2021


RUNX1 Thrombocytopenia, MDS/AML https:​­/​­/www​­.runx1​­-fpd​­.org​­/
GATA-2 defi­ciency Recurrent infec­tions, lymphedema, warts, pul­mo­nary https:​­/​­/rarediseases​­.org​­/gard​­-rare​­-disease​­
alve­o­lar proteinosis, MDS/AML /gata2​­-deficiency​­/
SAMD9/9L dis­or­ders MIRAGE (myelodysplasia, infec­tion, restric­tion of growth, https:​­/​­/www​­.stjude​­.org​­/samd9
adre­nal hypo­pla­sia, gen­i­tal phe­no­types, enter­op­a­thy),
MDS/AML
Severe con­gen­i­tal neutropenia Neutropenia, recur­rent bac­te­rial infec­tions https:​­/​­/neutropenianet​­.org​­/
Shwachman-Diamond syn­drome Neutropenia, exo­crine pan­cre­atic insuf­fi­ciency, MDS/AML http:​­/​­/sdsregistry​­.org​­/

screen­ing for these rare dis­eases and the need for ongo­ing sci­en­ loss, short stat­ure, devel­op­men­tal delay, blepharitis, peri­odon­tal
tific dis­cov­ery. dis­ease, pul­mo­nary fibro­sis, esoph­a­geal ste­no­sis, ure­thral ste­no­
sis, liver dis­ease, and avas­cu­lar necro­sis of the hips or shoul­ders.
Diamond-Blackfan ane­mia Almost 90% of patients with DC will even­tu­ally develop a cyto­
Diamond-Blackfan ane­ mia (DBA) was orig­ i­
nally described by penia of one or more periph­eral blood lin­e­ages, and BM fail­ure is
Josephs in 1936 and fur­ ther char­ ac­ter­
ized by Diamond and the major cause of death. Often BM fail­ure devel­ops in the sec­
Blackfan in 1938 as a con­gen­i­tal hypo­plas­tic ane­mia.1 In addi­tion ond or third decade of life, but it can occur at birth or as late as
to hypo­plas­tic ane­mia, the dis­or­der is char­ac­ter­ized by mac­ro­ the sev­enth decade of life. The BM abnor­mal­i­ties can evolve into
cy­to­sis, reticulocytopenia, and ele­vated lev­els of eryth­ro­cyte MDS or acute mye­loid leu­ke­mia (AML).6
aden­ o­
sine deam­ i­
nase. Additionally, half of patients with DBA The clin­i­cal diag­no­sis of DC can be chal­leng­ing given its phe­
also pres­ent with phys­i­cal abnor­mal­i­ties, includ­ing short stat­ure, no­typic het­ero­ge­ne­ity, dif­fer­ent modes of inher­i­tance (X-linked,
thumb abnor­ mal­i­
ties (clas­
si­
cally, a triphalangeal thumb), cra­ auto­so­mal reces­sive, and auto­so­mal dom­i­nant), and var­i­able age
nio­fa­cial defects, and cleft lip/pal­ate. DBA was the first dis­ease of onset. However, despite the wide spec­trum of the dis­ease,
to be linked to impaired ribo­some func­tion and is the founding rang­ing from clas­sic DC to aplastic ane­mia, it is clear that the
mem­ber of a group of dis­or­ders now known as ribosomopa­ under­ly­ing pathol­ogy is due to defec­tive telo­mere main­te­nance.
thies.2 Several other iBMFs as well as the 5q− MDS and can­cers The term “DC” is now used inter­change­ably with telo­mere biol­
have sub­se­quently been linked to muta­tions in genes encoding ogy dis­ or­
ders and short telo­ mere syn­ dromes. The func­ tional
for ribo­somal pro­teins or pro­teins required for nor­mal ribo­some assess­ment of telo­mere length with the find­ing of a telo­mere
func­tion.3,4 While the pre­dis­po­si­tion to MDS is lower than other length less than the first per­cen­tile for age in leu­ko­cyte sub­sets
iBMFs, there is a risk of other tumors, includ­ing colo­rec­tal can­ is highly sen­si­tive but not spe­cific to the diag­no­sis of these dis­
cer. In addi­tion, DBA is known to have incom­plete pen­e­trance, or­ders and has been seen in other inherited dis­or­ders.7,8 Regard­
as dem­on­strated both by the obser­va­tion of sib­lings who carry less, the eval­ua ­ ­tion of telo­mere length is crit­i­cal for any patient
the same muta­tion dis­cor­dant for the pres­ence of ane­mia and by with pan­cy­to­pe­nia and abnor­mal mar­row find­ings given the
the occur­rence of spon­ta­ne­ous remis­sions in affected patients, impli­ca­tions for screen­ing and mod­i­fi­ca­tions in stem cell trans­
which is why cli­ni­cians need to have a high index of sus­pi­cion plant con­di­tion­ing.
in patients with unex­plained ane­mia and mar­row abnor­mal­i­ties.
Fanconi ane­mia
Dyskeratosis congenita Fanconi ane­mia (FA) was first described by Guido Fanconi in 1927 in
In 1910 a case report was the first to define a syn­drome, ulti­mately 3 broth­ers with pan­cy­to­pe­nia and con­gen­i­tal abnor­mal­i­ties.9 The
named dyskeratosis congenita (DC), char­ac­ter­ized by a triad of dis­ease typ­i­cally progresses through sev­eral clin­i­cal stages, orga­
abnor­mal skin pig­men­ta­tion, oral leukoplakia, and nail dys­tro­ nized by age. In the first stage, in infancy and early child­hood, con­
phy.5 The spec­trum of DC has expanded con­sid­er­ably since then gen­i­tal anom­a­lies may be pres­ent, although they are not required
to include effects on every organ sys­tem, par­tic­u­larly the BM. for the diag­no­sis of FA and range from mild to severe. The most
Other clin­i­cal find­ings may include early graying of hair or hair com­ mon malformations include short stat­ ure, hypopigmented

154  |  Hematology 2021  |  ASH Education Program


or café au lait spots, thumb or radial ray abnor­mal­i­ties, micro- or Myeloid malig­nancy pre­dis­po­si­tion syn­dromes
hydro­ceph­aly, struc­tural renal anom­a­lies, hypogonadism, and GATA-2 defi­ciency
devel­op­men­tal delay. The con­stel­la­tion of dis­sem­i­nated nontuberculous myco­bac­te­
Within the first decade, patients may pres­ent with throm­bo­cy­ rial infec­tions; sus­cep­ti­bil­ity to viral infec­tions, espe­cially human
to­pe­nia and mac­ro­cy­to­sis before progressing to BM fail­ure, when pap­il­lo­ma­vi­rus and Epstein-Barr virus; and fun­gal infec­tions, cou­
the diag­no­sis is often first made. During ado­les­cence and adult­ pled with pro­found monocytopenia and B-cell and nat­u­ral killer
hood, the risk of AML and MDS becomes very high. In older adults, cell lymphopenias, was syndromically described as MonoMAC
a range of solid tumors, par­tic­u­larly squa­mous cell car­ci­no­mas of and den­dritic cell, mono­cyte, B, and nat­u­ral killer lym­phoid defi­
the head/neck and gen­i­to­uri­nary track, can be seen.10 Throughout ciency. Mutations in GATA-2 were iden­ti­fied in 2011 as the cause
life, the hema­to­poi­etic phe­no­type can change because of genetic of this group of dis­or­ders, which are also asso­ci­ated with mar­
rever­sion or clonal evo­lu­tion, so a high index of sus­pi­cion and a row fail­ure, MDS, and AML.14 The dis­ease fol­lows an auto­so­mal
care­ful his­tory, includ­ing a fam­ily his­tory of can­cer pre­dis­po­si­tion, dom­i­nant mode of inher­i­tance as well as a large num­ber of spo­
is essen­tial. These patients are highly sen­si­tive to che­mo­ther­apy radic cases. Presentations vary widely among affected mem­bers
and radi­a­tion, which is why it is crit­i­cal to rule out this diag­no­sis even within the same fam­ily, but the BM is often ini­tially hypocel­

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/153/1851462/153narla.pdf by guest on 13 December 2021


prior to trans­plant for any patient with unex­plained pan­cy­to­pe­nia. lular and may have strik­ing multilineage dys­pla­sia.

Severe con­gen­i­tal neutropenia RUNX1


Severe con­gen­i­tal neutropenia (SCN) is char­ac­ter­ized by abso­ RUNX1 is a mem­ber of the core-bind­ing fac­tor fam­ily of tran­scrip­
lute neu­tro­phil counts con­sis­tently less than 200/uL with recur­ tion fac­tors and is indis­pens­able for defin­i­tive hema­to­poi­e­sis;
rent severe infec­tions, which often develop within the first few it is one of the most fre­quently mutated genes in a vari­ety of
months of life. Evaluation of the BM dem­on­strates a mye­loid mat­ hema­to­log­i­cal malig­nan­cies. Germ line muta­tions in RUNX1 have
u­ra­tion arrest. The dis­ease can be auto­so­mal reces­sive (Kostmann been shown to cause a famil­ial plate­let dis­or­der with asso­ci­ated
syn­drome), X-linked, or auto­so­mal dom­i­nant. In Kostmann syn­ malig­nan­cies, includ­ing pedi­at­ric MDS. Patients may pres­ent
drome, muta­tions have been iden­ti­fied in the HAX1, G6PC3, and with iso­lated throm­bo­cy­to­pe­nia and mega­kar­yo­cytic dysmor­
GFI1 genes. In the X-linked dis­ease, muta­tions have been found in phia or atypia on base­line BM eval­u­a­tion before progressing to
the WAS gene. Finally, muta­tions in the ELANE gene are the most pan­cy­to­pe­nia, multilineage dys­pla­sia, increased blasts, and the
com­mon cause of SCN and cause auto­so­mal dom­i­nant dis­ease. As acqui­si­tion of addi­tional somatic muta­tions. A sub­set of patients
with Shwachman-Diamond syn­drome, below), addi­tional somatic may pres­ent with MDS/AML at a young age. Early rec­og­ni­tion of
muta­tions are often acquired over time, which serve as impor­tant germ line muta­tion and pre­dis­po­si­tion to mye­loid malig­nancy
mark­ers for dis­ease risk and may affect treat­ment deci­sions. per­mits appro­pri­ate treat­ment, ade­quate mon­i­tor­ing for dis­ease
pro­gres­sion, and proper donor selec­tion for hema­to­poi­etic stem
Shwachman-Diamond syn­drome cell trans­plan­ta­tion, as well as genetic coun­sel­ing of affected
Shwachman-Diamond syn­drome (SDS, also known as Shwachman- patients and their fam­ily mem­bers.
Diamond-Oski syn­drome) was ini­tially described in the early 1960s
as a dis­or­der of exo­crine pan­cre­atic dys­func­tion and BM fail­ure.11 SAMD9/9L dis­or­ders
Patients gen­er­ally pres­ent with ste­at­or­rhea, growth fail­ure, and Germ line SAMD9 and SAMD9L muta­tions have now been iden­ti­
recur­rent infec­tions. The hema­to­logic abnor­mal­i­ties are pre­ fied as the cause of a spec­trum of mul­ti­sys­tem dis­or­ders that carry
dom­i­nantly neutropenia, although ane­mia, throm­bo­cy­to­pe­nia, a mark­edly increased risk of devel­op­ing mye­loid malig­nan­cies with
and pan­cy­to­pe­nia can occur. Skeletal abnor­mal­i­ties (short stat­ chro­mo­some 7 abnor­mal­i­ties, includ­ing mono­somy 7, del7q, and
ure, delayed appear­ ance of normally shaped epiph­ y­
ses, and CN-loh of 7q. Affected indi­vid­u­als dis­play a highly var­i­able clin­i­cal
pro­gres­sive metaphyseal thick­en­ing/dys­pla­sia) as well as poor course that ranges from mild and tran­sient dyspoietic changes in
growth are com­mon in SDS patients. Patients with­out severe the BM to a rapid pro­gres­sion of MDS or AML with mono­somy 7.
pan­cre­atic dis­ease may pres­ent later in child­hood or as adults, These data have impli­ca­tions for under­stand­ing how SAMD9 and
and some patients may pres­ent with aplastic ane­mia or AML as SAMD9L muta­tions con­trib­ute to mye­loid trans­for­ma­tion and for
the ini­tial man­i­fes­ta­tion of the dis­ease.12 rec­og­niz­ing, coun­sel­ing, and treating affected fam­i­lies.
The diag­no­sis of SDS relies on clin­i­cal find­ings. Exocrine pan­
cre­atic dys­func­tion, which can be sub­clin­i­cal, can be established The role of genetic test­ing
by low serum tryp­sin­o­gen in patients youn­ger than 3 years or low Many of the dis­or­ders discussed have spe­cific, iden­ti­fied gene
pan­cre­atic isoamylase in patients older than 3 years. In addi­tion, muta­tions, but there is increas­ing inter­est in under­stand­ing what
patients may have low fecal elas­tase or a fatty pan­creas that can other genes may be involved within these dis­eases as well as
be dem­on­strated by imag­ing. Genetic test­ing can be help­ful to in dis­or­ders in which no genes have been iden­ti­fied. Increasing
con­firm the diag­no­sis, but a neg­a­tive test does not exclude the num­bers of pan­els are being conducted both in com­mer­cial labs
diag­no­sis. In 90% of affected patients, this auto­so­mal reces­sive and uni­ver­si­ties that cover BMF only vs whole exome sequenc­
dis­or­der is due to muta­tions in the Shwachman-Bodian-Diamond ing. The deci­sion about spe­cific tests to be performed should be
syn­drome (SBDS) gene.13 It is essen­tial to dis­tin­guish SDS from done in con­sul­ta­tion with a genet­i­cist, which will also help guide
cys­tic fibro­sis (the most com­mon cause of exo­crine pan­cre­atic deci­sions about fam­ily test­ing. This infor­ma­tion is crit­i­cal for sur­
insuf­fi­ciency in chil­dren), con­gen­i­tal neutropenia, and Pearson veil­lance and may help steer treat­ment options regard­ing stem
syn­drome. cell donor trans­plan­ta­tion.

Prakash Singh Shekhawat


Bone mar­row fail­ure syn­dromes and aplastic ane­mia  |  155
Acquired BM fail­ure syn­dromes that can pres­ent References
with signs of mar­row aplasia 1. Diamond LK, Blackfan KD. Hypoplastic ane­mia. Am J Dis Child. 1938;15:307.
2. Narla A, Ebert BL. Ribosomopathies: human dis­or­ders of ribo­some dys­
Although beyond the scope of this chap­ter, it is worth not­ing
func­tion. Blood. 2010;115(16):3196-3205.
that sev­eral genetic causes of acquired BM fail­ure should be con­ 3. Ebert BL, Pretz J, Bosco J, et al. Identification of RPS14 as a 5q- syn­drome
sid­ered in the eval­u­a­tion of patients with cytopenias and hypo­ gene by RNA inter­fer­ence screen. Nature. 2008;451(7176):335-339.
plas­tic mar­row. Again, a detailed phys­i­cal exam and a care­ful 4. Pelletier J, Thomas G, Volarević S. Ribosome bio­gen­e­sis in can­cer: new
fam­ily his­tory will help to deter­mine the appro­pri­ate workup and play­ers and ther­a­peu­tic ave­nues. Nat Rev Cancer. 2018;18(1):51-63.
5. Zinsser F. Atrophia cutis reticularis cum pig­men­ta­tions, dystrophia unguium
genetic test­ing. For exam­ple, VEXAS syn­drome is a mono­genic at leukoplakis oris (poikioodermia atrophicans vascularis Jacobi). Ikono­
dis­ease of adult­hood caused by somatic muta­tions in UBA1 in graphia Dermatologica. 1910;5:219-233.
hema­to­poi­etic pro­gen­i­tor cells that can pres­ent with a range of 6. Savage SA, Alter BP. Dyskeratosis congenita. Hematol Oncol Clin North
immu­no­logic and hema­to­logic symp­toms.15 Am. 2009;23(2):215-231.
7. Alder JK, Hanamanthu VS, Strong MA, et al. Diagnostic util­ity of telo­mere
length test­ing in a hos­pi­tal-based set­ting. PNAS. 2018;115(10):E2358-E2365.
Conclusions 8. Schratz KE, Haley L, Danoff SK, et al. Cancer spec­trum and out­comes in the

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/153/1851462/153narla.pdf by guest on 13 December 2021


Maintaining a high sus­pi­cion for rare con­gen­i­tal and acquired Men­de­lian short telo­mere syn­dromes. Blood. 2020;135(22):1946-1956.
9. Lobitz S, Velleuer E. Guido Fanconi (1892-1979): a jack of all­trades. Nat Rev
causes of BM fail­ure is crit­ic ­ al when eval­ua
­ t­ing patients of all­
Cancer. 2006;6(11):893-898.
ages with unusual cytopenias. A detailed phys­i­cal exam­i­na­tion 10. Bagby GC, Alter BP. Fanconi ane­mia. Semin Hematol. 2006;43(3):147-156.
and fam­ily his­tory are also crit­i­cal. These guide the sub­se­quent 11. Shwachman H, Diamond LK, Oski FA, Khaw KT. The syn­drome of pan­cre­atic
lab­o­ra­tory workup, sur­veil­lance sched­ule for malig­nancy, and insuf­fi­ciency and bone mar­row dys­func­tion. J Pediatr. 1964;65(5):645-663.
poten­tial ther­a­peu­tic options. 12. Myers KC, Davies SM, Shimamura A. Clinical and molec­ul­ar path­o­phys­i­­ol­
ogy of Shwachman-Diamond syn­drome: an update. Hematol Oncol Clin
North Am. 2013;27(1):117-128, ix.
Conflict-of-inter­est dis­clo­sure 13. Boocock GR, Morrison JA, Popovic M, et al. Mutations in SBDS are asso­ci­
Anupama Narla: no com­pet­ing finan­cial inter­ests to declare. ated with Shwachman-Diamond syn­drome. Nat Genet. 2003;33(1):97-101.
14. Hsu AP, Sampaio EP, Khan J, et al. Mutations in GATA2 are asso­ci­ated with
the auto­so­mal dom­i­nant and spo­radic monocytopenia and myco­bac­te­rial
Off-label drug use infec­tion (MonoMAC) syn­drome. Blood. 2011;118(10):2653-2655.
Anupama Narla: nothing to disclose. 15. Grayson PC, Patel BA, Young NS. VEXAS syn­drome. Blood. 2021;137(26):3591-
3594.

Correspondence
Anupama Narla, Stanford University School of Medicine, CCSR
South 1215b, 269 Campus Dr, Stanford, CA 94305-5162; e-mail: © 2021 by The Amer­i­can Society of Hematology
anunarla@stanford​­.edu. DOI 10.1182/hema­tol­ogy.2021000246

156  |  Hematology 2021  |  ASH Education Program


DEFEATING DIFFUSE, DOUBLE - HIT, AND DOGGED NON - HODGKIN LYMPHOMA

Double-hit lymphoma: optimizing therapy


Kieron Dunleavy

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/157/1852000/157dunleavy.pdf by guest on 13 December 2021


Division of Hematology and Oncology, Georgetown Lombardi Comprehensive Cancer Center, Georgetown University Hospital, Washington, DC

Aggressive B-cell lymphoma is a heterogeneous entity with disparate outcomes based on clinical and pathological char-
acteristics. While most tumors in this category are diffuse large B-cell lymphoma (DLBCL), the recognition that some
cases have high-grade morphology and frequently harbor MYC and BCL2 and/or BCL6 translocations has led to their
separate categorization. These cases are now considered distinct from DLBCL and are named “high-grade B-cell lym-
phoma” (HGBL). Most are characterized by distinct rearrangements, but others have high-grade morphological features
without these and are called HGBL-not otherwise specified. Studies have demonstrated that this group of diseases leads
to poor outcomes following standard rituximab, cyclophosphamide, hydroxydaunorubicin, vincristine, and prednisone
therapy; retrospective and recent single-arm, multicenter studies suggest they should be approached with dose-intense
treatment platforms. As yet, this has not been validated in randomized trial settings due to the rarity of these diseases. In
the relapsed and refractory setting, novel approaches such as anti-CD19 chimeric antigen receptor T cells and antibodies
against CD19 have demonstrated high efficacy in this subgroup. Recently, genomic studies have made much progress in
investigating some of the molecular underpinnings that drive their lymphomagenesis and have paved the way for testing
additional novel approaches.

LEARNING OBJECTIVES
• Understand recent developments in the elucidation of MYC and BCL2 aberrations that are helpful in the categori-
zation of these lymphomas and the implications for therapeutic approaches
• Review recent clinical outcomes using various strategies for HGBL, both in the up-front and relapsed/refractory
setting, and understand which novel therapies may be useful in managing these diseases

B-cell lymphoma (HGBL) with MYC and BCL2 transloca-


CLINICAL CASE tions. He received treatment with 6 cycles of rituximab,
A 46-year-old previously well man presented with a 4-week cyclophosphamide, hydroxydaunorubicin, vincristine, and
history of bilateral neck swelling, fevers, night sweats, prednisone (R-CHOP) that included intrathecal prophy-
and moderate weight loss. Fluorodeoxyglucose-positron laxis with methotrexate and achieved a complete remis-
emission tomographic (FDG-PET) imaging demonstrated sion by end-of-treatment PET scan. Eight weeks following
hypermetabolic diffuse lymphadenopathy (node diameters the PET scan, he developed recurrent symptoms and neck
up to 4 cm: maximum standardized uptake values >25) on lymphadenopathy. Reimaging and subsequent biopsy
both sides of the diaphragm in addition to bone marrow confirmed recurrent disease.
involvement. His lactate dehydrogenase was elevated; his
Eastern Cooperative Oncology Group performance status
was 1. HIV testing was negative. Following an inconclusive Introduction
fine-needle aspiration, he underwent an excisional biopsy Diffuse large B-cell lymphoma (DLBCL) is now recognized
of a right 3-cm cervical lymph node. This demonstrated as and continues to evolve as a clinically and molecu-
aggres sive CD20+ B -cell lymphoma; tumor cells were larly heterogeneous disease.1-5 While the addition of rit-
CD10+, BCL2+ (>60%), and MYC+ (>90%), and Ki67 was uximab to anthracycline-based treatment has resulted in
>90%. Fluorescence in situ hybridization (FISH) studies an overall survival (OS) benefit, standard R-CHOP ther-
demonstrated a rearrangement of MYC and BCL2 with no apy remains noncurative for a substantial proportion of
BCL6 rearrangement. Hence, his final diagnosis was stage people. Among the challenges faced in managing newly
IVB (International Prognostic Index [IPI] 3), high-grade diagnosed patients are accurately predicting who will not
Prakash Singh Shekhawat
New developments in aggressive B-cell lymphoma | 157
Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/157/1852000/157dunleavy.pdf by guest on 13 December 2021
Figure 1. Category of aggressive B-cell lymphomas “HGBLs with MYC and BCL2 and/or BCL6 rearrangements” described in the
2016 revision to the World Health Organization classification of tumors of hematopoietic and lymphoid tumors. Most cases with
MYC and BCL2 rearrangements are of GCB origin, whereas most cases with BCL6 rearrangements are of ABC origin. This category
includes DH lymphomas, which involve MYC and BCL2 or MYC and BCL6, as well as THLs that involve MYC, BCL2, and BCL6. When
translocated, MYC may have an IG or non-IG partner gene, with the former associated with an inferior outcome. In a large study, 7.9%
of tumors with DLBCL morphology were assigned to HGBL-DHL/THL, composing 13.3% of GCB and 1.7% of ABC DLBCL.30

be cured with R-CHOP and decid­ing if alter­na­tive approaches double-hit is not as well established in prospective experiences,
should be used in cer­tain patient sub­sets. In sev­eral stud­ies over and the opti­mal man­age­ment of cases such as these remains
a long time span, the IPI has been a reli­able pre­dic­tor of out­ con­tro­ver­sial. The goal of this review is to explore this ques­tion
come; the prog­nos­tic value of base­line tumor bio­log­i­cal fac­tors in the con­text of emerg­ing bio­log­i­cal insights and novel clin­i­cal
is much less well established and remains very con­tro­ver­sial.6 data for this sub­set of patients.
Different groups have reported out­comes and asso­ci­a­tions with HGBL cases are now con­sid­ered a dis­tinct entity in the 2016
tumor bio­log­i­cal char­ac­ter­is­tics that vary con­sid­er­ably, par­tic­u­ World Health Organization Lymphoid Tumor Classification.10 They
larly when com­par­ing pro­spec­tive and ret­ro­spec­tive data. It is encom­pass a sub­set of aggres­sive cases that, despite overlap-
well established that most DLBCL cases are of ger­mi­nal cen­ter ping clin­i­cal and path­o­log­i­cal char­ac­ter­is­tics, are dif­fer­ent from
B-cell (GCB) or acti­vated B-cell (ABC) ori­gin, and many stud­ies the par­ ent enti­ ties of DLBCL and BL (Figure 1). The cat­ e­gory
have dem­on­strated an infe­rior out­come for the lat­ter group. In includes lym­pho­mas that were pre­vi­ously named “Burkitt-like”
attempts to improve out­come in this sub­set, efforts have been and “high grade” as well as “dou­ble-hit” or “tri­ple-hit.” In addi­
under­way to add agents to R-CHOP that have selec­tive activ­ tion to the major cat­e­gory “HGBL with MYC and BCL2 and/or
ity in ABC-DLBCL. However, 2 recently reported large ran­dom­ BCL6 trans­lo­ca­tions,” there is a sub­set called “HGBL-not oth­er­
ized stud­ies did not show a ben­e­fit to adding lenalidomide or wise spec­ifi­ ed” (NOS). Most cases with a sin­gle MYC rearrange-
ibrutinib in ABC- or non-GCB-DLBCL.7-9 Since the con­ cep­ tion ment fall under DLBCL, as do most cases with high MYC and BCL2
of these tri­als, the cat­e­go­ri­za­tion of DLBCL has under­gone fur­ expres­sion but with­out rearrangements. The updated cat­e­go­ri­
ther refine­ment and is focused on more accu­rately iden­ti­fy­ing za­tion is help­ful in the clinic, as HGBL behaves more aggres­sively
prognostically mean­ing­ful genetic driv­ers of tumor­i­gen­e­sis, than DLBCL and likely requires dis­tinct ther­a­peu­tic approaches.11
which paves the way for novel and more pre­cise therapeutic Though dou­ble-hit/tri­ple-hit lym­phoma (DHL/THL) and double
approaches.1-3 As well as cell-of-ori­gin-focused stud­ies, the rec­ protein expresser (DPE) cases have MYC and BCL2 aber­ra­tions
og­ni­tion that aber­rant MYC and BCL2 expres­sion is asso­ci­ated in com­mon, they are dis­tinct in terms of their lymphomagenesis
with infe­rior out­comes has led to inves­ti­ga­tions aimed at uncov- because DLH/THL is mostly GCB derived, while DPE cases are
ering the mech­an ­ is­tic basis for MYC and BCL2 overexpression. prin­ci­pally of ABC ori­gin (Figure 2).
In par­al­lel with this, fol­low­ing improved out­comes with dose-
intense approaches in ret­ro­spec­tive com­par­i­sons, alter­na­tives Recent advances in under­stand­ing HGBL cases
to R-CHOP are being inves­ti­gated in these sub­sets. Coming back Recent stud­ies have set out to bet­ter under­stand the bio­log­i­
to our patient, based on his high IPI score and “dou­ble-hit” sta­ cal under­pin­nings of HGBL cases and elu­ci­date the spe­cific
tus, his predicted cur­abil­ity rate with R-CHOP is low. While this is char­ac­ter­is­tics asso­ci­ated with infe­rior out­comes. While FISH test­
clear from ret­ro­spec­tive data, the negative prognostic impact of ing is cur­rently the gold stan­dard to iden­tify HGBL with MYC and

158  |  Hematology 2021  |  ASH Education Program


Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/157/1852000/157dunleavy.pdf by guest on 13 December 2021
Figure 2. Categories of double-expresser lymphomas. These are typically cases that have a high-protein expression of MYC and
BCL2. Most DPE cases that are associated with rearrangements of MYC and BCL2 are of GCB origin, whereas most cases that do not
harbor these rearrangements are of ABC origin. The proportion of DLBCL cases that are double expressers has been calculated at
between 21% and 44% across various studies.31

BCL2 and/or BCL6 rearrangements, emerg­ing data from more an IG gene in almost all­cases. In con­trast, approx­i­ma­tely 50%
com­plex geno­mic stud­ies sug­gest that FISH has sen­si­tiv­ity lim­i­ of HGBL-DHL/THLs have a non-IG part­ner. A recent study dem­
ta­tions com­pared to tech­niques such as whole-exome sequenc­ on­strated infe­rior out­comes for MYC DHL/THL cases with MYC
ing.12 Therefore, an impor­tant clin­i­cally appli­ca­ble ques­tion arises: translocated to an IG part­ ner vs a non-IG part­ner fol­low­ing
Does FISH ade­quately iden­tify—within GCB-DLBCL and HGBL— treat­ment with R-CHOP.14 In interpreting the results of stud­ies
cases with aber­ra­tions of MYC/BCL2 and/or BCL6 that por­ looking at the prog­nos­tic impact of DHL/THL and sin­gle-hit lym­
tend an infe­rior out­come? Probably not, and many cases with phoma (SHL) MYC aber­ra­tions, it is impor­tant to con­sider recent
crit­i­cal DHL/THL aber­ra­tions are not iden­ti­fied by FISH alone. geno­mic stud­ies that have elu­ci­dated novel genetic sub­types of
A dou­ble-hit gene expres­sion sig­na­ture (DHIT-sig) was recently DLBCL.1-4 Within GCB tumors, newly char­ac­ter­ized sub­sets with
pro­posed based on the anal­y­sis of RNA sequenc­ing data from dis­tinct clin­i­cal out­comes exhibit the co-occur­rence of spe­cific
157 cases of GCB-DLBCL (includ­ing HGBL) treated with R-CHOP.4 genetic alter­ations. There is likely sig­nif­i­cant over­lap between
This was a 104 gene sig­na­ture that represented/dis­tin­guished aber­ra­tions in these sub­sets and DHIT-sig+ cases that require
27% of GCB-DLBCLs—and while the major­ity of GCB-DLBCLs future inves­ti­ga­tion.
with high-grade mor­ phol­
ogy had this sig­ na­ture, only half of
the DHIT-sig+ cases har­bored con­cur­rent MYC and BCL2 rear- Approach to HGBL with MYC and BCL2 and/or BCL6
rangements.4 DHIT-sig+ cases had a sig­nif­i­cantly infe­rior time to rearrangements
dis­ease pro­gres­sion com­pared to DHIT− cases. Another recent Currently, no widely accepted stan­dard approach exists to the
study dem­on­strated that DHIT-sig+ cases with con­com­i­tant ini­tial man­age­ment of these cases. Clinical pre­sen­ta­tion dif­fer­
TP53 abnor­mal­i­ties had a par­tic­u­larly poor out­come.13 It is inter­ ences between DLBCL and HGBL have not been well defined,
est­ing to con­sider HGBL cases in the con­text of newer molec­ but early stud­ies dem­on­strated fre­quent extranodal involve­ment
u­lar clas­si­fi­ca­tions of DLBCL and pro­pose where they may lie and a higher rate of cen­tral ner­vous sys­tem (CNS) dis­ease in the
within newly defined, more poten­tially tar­get­able, sub­groups.1-3 lat­ter entity. Multiple ret­ro­spec­tive and obser­va­tional stud­ies
In 1 recent study that com­pre­hen­sively genet­i­cally ana­lyzed 304 have dem­on­strated that fol­low­ing R-CHOP treat­ment, sur­vival
DLBCL cases and defined 5 dis­tinct sub­sets (clus­ters 1-5), tumors is sig­nif­i­cantly infe­rior com­pared to patients who do not har­bor
with co-occur­ring BCL2 and MYC struc­tural var­i­ants were sig­nif­i­ these aber­ra­tions. Early ret­ro­spec­tive stud­ies dem­on­strated a
cantly more fre­quent in clus­ter 3, a GCB-defined sig­na­ture.1 par­tic­u­larly adverse out­come for this sub­set that is less strik­ing
The influ­ence of a rearrangement of MYC is likely affected in some recent stud­ies. This may be partly explained by a his­
by the MYC part­ner gene, whether it is an immu­no­glob­u­lin (IG) tor­i­cal selec­tion bias in apply­ing FISH test­ing to select patients
or a non-IG gene.14 In Bur­kitt lym­phoma, the part­ner of MYC is with more aggres­sive clin­i­cal pre­sen­ta­tions vs the more recent

Prakash Singh Shekhawat


New devel­op­ments in aggres­sive B-cell lym­phoma  |  159
stan­dard of apply­ing it to the major­ity of new cases. A recent mul­ti­cen­ter study of 53 patients with MYC rearrangement
large-scale ret­ro­spec­tive anal­y­sis of DLBCL patient out­comes fol­ (MYC-R) aggres­ sive B-cell lym­phoma.15 More cases were DHL
low­ing R-CHOP—from pro­spec­tive tri­als and patient reg­is­tries— than SHL-MYC-R. The major­ity of patients had advanced-stage
reported inter­est­ing find­ings.14 While the anal­y­sis showed that dis­ease (81%), and 48-month event-free sur­vival (EFS) and OS
patients with a MYC rearrangement had a sig­nif­i­cantly shorter were 71% and 77%, respec­tively. A phase 2 HOVON trial added
pro­gres­sion-free sur­vival (PFS) and OS, the adverse impact of the lenalidomide to R-CHOP in 82 patients, hypoth­e­siz­ing that
rearrangement was con­fined to those har­bor­ing a con­cur­rent because lenalidomide downregulates MYC and its tar­get genes
BCL2 and/or BCL6 rearrangement and an IG vs non-IG part­ner it may be ther­a­peu­ti­cally advan­ta­geous in MYC-R lym­phoma.15
with MYC. It is impor­tant to note that this study only included Sixty-five per­cent of cases had a DHL or THL, and 2-year EFS and
cases with DLBCL mor­phol­ogy, and this may have con­trib­uted OS were 63% and 73%, respec­tively. A recent Brit­ish study eval­
to bet­ter out­comes com­pared to his­tor­i­cal expe­ri­ences in which u­ated patients with high-risk DLBCL using cyclo­phos­pha­mide,
blastoid and Burkitt morphologies were included. vin­cris­tine, doxo­ru­bi­cin, high-dose meth­o­trex­ate, ifosfamide,
Should approaches beyond R-CHOP be con­ sid­ered for etoposide, and high-dose cytarabine (R-CODOX-M/R-IVAC).16 All

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/157/1852000/157dunleavy.pdf by guest on 13 December 2021


HGBCL? Two recent pro­spec­tive stud­ies of DLBCL patients patients had an IPI score of 3 or higher. Most were GCB, and
har­bor­ing a MYC rearrangement (with a high pro­por­tion hav­ over 10% had DHL, suggesting that a high pro­por­tion may have
ing DHL) were reported. Based on ret­ro­spec­tive com­par­i­sons had HGBL. Two-year PFS was 67.9%, and 2-year OS was 76%, and
show­ing that DHL cases did bet­ter with more dose-inten­sive while not directly com­pared to R-CHOP, the results were supe­rior
approaches com­pared to R-CHOP, dose-adjusted etoposide, to his­tor­i­cal expe­ri­ences using R-CHOP in a sim­i­lar pop­u­la­tion.
pred­ni­sone, vin­cris­tine, cyclo­phos­pha­mide, hydroxydaunoru- Leppa and col­leagues inves­ti­gated dose-dense immunochemo-
bicin, and rituximab (DA-EPOCH-R) was tested in a pro­spec­tive, therapy in 139 patients with high-risk DLBCL and dem­on­strated a

Figure 3. Outline for the workup and management of aggressive B-cell lymphomas. Typically, morphological, immunohistochemis-
try, and FISH analysis are performed to differentiate DLBCL from HGBL. HGBL cases are divided into those that are DHL/THL or NOS.
For DLBCL cases that have a high-protein expression of MYC and BCL2, which are usually of ABC origin, dose-intensive therapy or
enrollment in a clinical trial should be considered, particularly for patients with a high IPI score.

160  |  Hematology 2021  |  ASH Education Program


5-year OS rate of 83%.17 The out­come of patients with BCL2/MYC stem cell trans­plan­ta­tion. Unfortunately, 6 weeks fol­low­ing
DHL was sim­i­lar to patients with­out rearrangements, suggesting the trans­ plant, he had fur­ther pro­ gres­
sive dis­ease. He then
a ben­e­fit from dose-intense ther­apy in the DHL group. Also, a went on to receive anti-CD19 chi­me­ric anti­gen recep­tor (CAR)
large, recently published French ret­ro­spec­tive study eval­u­ated T-cell ther­apy (axicabtagene ciloleucel) and had a com­plete
160 patients with HGBL (with MYC and BCL2 and/or BCL6) and response. He was still in remis­sion 6 months fol­low­ing the com­
dem­on­strated a sig­nif­i­cantly lon­ger PFS for inten­sive ther­apy vs ple­tion of CAR T cells.
R-CHOP.18 HGBL-NOS cases are much rarer, and hence, there are
a pau­city of data to inform on opti­mal treat­ment.
In selecting ther­apy for patients with HGBL, the stage of dis­ Approach to relapsed/refrac­tory HGBL
ease and IPI char­ac­ter­is­tics may be impor­tant (Figure 3). Retro- Given the rar­ity of these tumors, it is unknown if relapsed/
spective expe­ri­ences have dem­on­strated good out­comes for refrac­tory HGBL should be approached dif­fer­ently to DLBCL with­
patients with lim­ited-stage aggres­sive B-cell lym­phoma despite out high-risk cyto­ge­net­ics. Some ret­ro­spec­tive stud­ies have
high-risk cyto­ge­net­ics.19 It may be rea­son­able to approach shown that patients with these dis­ eases (com­pared to other

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/157/1852000/157dunleavy.pdf by guest on 13 December 2021


these early-stage patients with stan­dard R-CHOP. For higher- aggres­sive B-cell lym­pho­mas) fare more poorly fol­low­ing autol­o­
stage and high-IPI patients, more inten­sive immunochemother- gous stem cell trans­plan­ta­tion.22 This may be partly explained by
apy approaches are rea­son­able to con­sider, under­stand­ing that the fact that HGBL cases are more likely to receive more inten­
there are a pau­city of robust com­par­i­son data (Table 1). HGBL sive immunochemotherapy than R-CHOP in the up-front set­ting.
tumors are not uncom­monly encoun­tered in the set­ting of HIV Recently published and ongo­ing stud­ies looking at approaches
infec­tion, and based on equiv­ a­lent out­
comes to HIV− cases such as targeting CD19 in the relapsed/refrac­tory set­ting have,
in recent pro­ spec­tive stud­ ies (where up to 25% of patients inter­est­ingly, shown that HGBL biol­ogy is not asso­ci­ated with a
accrued were HIV+), they should not be approached dif­fer­ently. worse out­come.23,24 Considering that this has not been the expe­
ri­ence with autol­o­gous stem cell trans­plan­ta­tion, it sug­gests a
Approach to DPE lym­pho­mas poten­tial ben­e­fit to mov­ing these new treat­ment modal­i­ties to the
MYC, BCL2, and BCL6 are overexpressed by sev­ eral mech­
a­ front­line set­ting.
nisms other than gene rearrangements, and a high pro­por­tion of
aggres­sive B-cell lym­phoma cases have high-pro­tein expres­sion
Promising new approaches
of MYC and BCL2 and/or BCL6. These DPE cases are asso­ci­ated
Many prom­is­ing approaches are in devel­op­ment for this group
with an infe­rior out­come fol­low­ing stan­dard ther­apy—when lack-
of dis­eases. First, strat­e­gies that incor­po­rate targeting BCL2 and
ing rearrangements, they are typ­i­cally of ABC ori­gin. How best to
MYC are under inves­ti­ga­tion in tri­als.25,26 Undoubtedly, the con­
approach them ther­a­peu­ti­cally is unclear. There is no evi­dence
cur­rent high expres­sion of MYC and BCL2, irrespective of path­o­
from ret­ro­spec­tive stud­ies that intensive therapy approaches
gen­e­sis, is asso­ci­ated with a higher risk of treat­ment fail­ure, but
are superior. These patients should there­fore be con­sid­ered for
the indi­vid­ual con­tri­bu­tions of these path­ways, in terms of con­
enroll­ment in clin­i­cal tri­als, where agents and approaches that
fer­ring resis­tance, are not well under­stood. Venetoclax, a highly
tar­get the key path­o­ge­netic mech­a­nisms underpinning MYC and
selec­tive inhib­i­tor of BCL2, has activ­ity in sev­eral lym­pho­mas and
BCL2 acti­va­tion are being investigated.
was recently com­bined with R-CHOP che­mo­ther­apy in a front­line
phase 2 study for patients with DLBCL (phase 2 CAVALI study).27
Role of CNS pro­phy­laxis in HGBL
While this com­ bi­
na­tion dem­ on­strated good activ­ ity and the
As is the case in DLBCL, the role of CNS pro­phy­laxis in HGBL is
poten­tial to improve out­come in a BCL2 immunohistochemistry
con­tro­ver­sial, and unfor­tu­nately, pro­spec­tive stud­ies that could
sub­group, it was asso­ci­ated with increased myelotoxicity com­
poten­tially clar­ify this ques­tion are lacking.20,21 From ret­ro­spec­
pared to R-CHOP alone. Currently, a ran­ dom­ ized pro­ spec­tive
tive data, the inci­dence of CNS relapse in early-stage or low-IPI
study of immunochemotherapy with or with­ out venetoclax is
cases is very low, suggesting that CNS pro­phy­laxis may have very
ongo­ing (NCT03984448). Inhibitors of MYC are also in devel­op­
min­i­mal poten­tial ben­e­fit.19 However, sev­eral series have now
ment; small-mol­e­cule inhib­i­tors of the bromodomain and extra-
dem­on­strated high CNS relapse rates in advanced-stage and
terminal domain pro­teins are also inter­est­ing with respect to MYC.
high-IPI cases, and some ret­ro­spec­tive sin­gle-arm com­par­i­son
Epignentic inhibitors such as those targeting histone deacetylase
expe­ri­ences sug­gest that pro­phy­laxis may dimin­ish CNS relapse
(HDAC)3 and EZH2 inhibitors are under evaluation in pre-clinical
inci­dence.17 It is very chal­leng­ing to con­duct a large-scale pro­
studies. Recently, up-front stud­ies have started to incor­ po­rate
spec­tive trial to prop­erly address this ques­tion and reli­ably guide
novel strat­e­gies such as anti-CD19 CAR T cells in treat­ments for
clin­i­cal prac­tice. Until that is done, how to approach CNS pro­
high-risk DLBCL and HGBCL patients who do not have early com­
phy­laxis will remain con­tro­ver­sial, with a lack of con­sen­sus. Given
plete responses.28
our patient’s stage IV dis­ease with bone mar­row infil­tra­tion and
DHL sta­tus and the high poten­tial for CNS spread, we decided to
insti­tute intra­the­cal CNS pro­phy­laxis. Conclusions
HGBLs are a huge ther­a­peu­tic chal­lenge, and their opti­mal man­
age­ment remains unde­fined at this time. It is crit­i­cal to con­
tinue to make inroads in under­stand­ing their biol­ogy and how
that inter­acts and over­laps with other key genetic and func­
CLINICAL CASE (Con­t in­u ed) tional driv­ers of lymphomagenesis. In that regard, recent work
Following con­f ir­m a­t ion of refrac­tory/relapsed dis­ease, the such as the iden­ti­fi­ca­tion of new prog­nos­tic sig­na­tures such as
patient went on to receive rituximab, ifosfamide, carboplatin, the DHIT-sig is wel­comed; addi­tion­ally, the def­i­ni­tion of novel,
and etoposide (R-ICE) che­mo­ther­aPrakash
py followedSingh Shekhawat
by autol­
o­gous poten­tially more action­able, DLBCL sub­groups is help­ful in the

New devel­op­ments in aggres­sive B-cell lym­phoma  |  161


Table 1. Select recent stud­ies in aggres­sive B-cell lym­phoma looking at (dif­fer­en­tial) out­comes of patients with HGBL
and DHL/THL

Study N Patient pop­u­la­tion/study DHL/THL % Treatment Outcome


Rosenwald 2383: (MYC-R DLBCL and HGBL/ 5.8% R-CHOP MYC-R was asso­ci­ated with
et al14 in 11%) ret­ro­spec­tive anal­y­sis of shorted PFS and OS; neg.
pro­spec­tive and patient prog­nos­tic impact of MYC-R
reg­is­try stud­ies only with BCL2 and/or
BCL6 and an IG part­ner.
Dunleavy 53 MYC-R and aggres­sive B-cell Approx 44%* had MYC-R DA-EPOCH-R 4-year EFS and OS were 71%
et al15 lym­phoma/pro­spec­tive, (SH); 56% had DHL/THL and 77%. No dif­fer­ence for
sin­gle-arm, mul­ti­cen­ter SH vs DHL/THL.
trial

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/157/1852000/157dunleavy.pdf by guest on 13 December 2021


Chamuleau 82 MYC-R DLBCL/pro­spec­tive, Approx 27%* had MYC-R R-CHOP + 2-year EFS and OS were 63%
et al29 sin­gle-arm mul­ti­cen­ter trial (SH); 73% had DHL/THL lenalidomide and 73%.
Leppä 139 DLBCL and high-IPI 12% had DHL Dose-dense chemo 5-year FFS and OS were 74%
et al17 score/high-risk cohort/ (MTX/R-CHOEP-14, and 83%. No sig­nif­i­cant
pro­spec­tive, sin­gle-arm, ARA C worse out­come for DHL
mul­ti­cen­ter trial group.
McMillan 111 DLBCL and IPI 3-5; 12% had 12% had DHL; FISH R-CODOX-M/R-IVAC 2-year PFS and OS were 68%
et al16 HGBL/pro­spec­tive study. performed in approx. 50% and 76%. No worse out­
come for DHL.
Laude 160 All patients had HGBL/ret­ro­ 81% had DHL; 19% had THL R-CHOP vs inten­sive At 32 months, 2 and 4-year
et al18 spec­tive study che­mo­ther­apy PFS were 40% and 28% for
R-CHOP; 57% and 52% for
inten­sive ther­apy.
Of cases tested.
ARA C, cytarabine; CHOEP, CHOP with etoposide; FFS, fail­ure-free sur­vival; MTX, meth­o­trex­ate.

quest to bet­ ter under­ stand the molec­ u­lar basis of MYC and References
BCL2 dysregulation. It is clear that stan­dard R-CHOP treat­ment 1. Chapuy B, Stewart C, Dunford AJ, et al. Molecular sub­types of dif­fuse large
B cell lym­phoma are asso­ci­ated with dis­tinct path­o­genic mech­a­nisms and
remains inad­e­quate for an unac­cept­ably high pro­por­tion of
out­comes. Nat Med. 2018;24(5):679-690.
cases; a sub­set of these cases may ben­e­fit from dose-inten­sive 2. Reddy A, Zhang J, Davis NS, et al. Genetic and func­tional driv­ers of dif­fuse
approaches, but this needs bet­ter inves­ti­ga­tion in, ide­ally, ran­ large B cell lym­phoma. Cell. 2017;171(2):481-494.e15494e15.
dom­ized pro­spec­tive tri­als. The chal­lenge is that these dis­eases 3. Schmitz R, Wright GW, Huang DW, et al. Genetics and path­o­gen­e­sis of dif­
fuse large B-cell lym­phoma. N Engl J Med. 2018;378(15):1396-1407.
are rare, and until these trial results are avail­­able, stan­dards can 4. Ennishi D, Jiang A, Boyle M, et  al. Double-hit gene expres­sion sig­na­ture
only be based on ret­ro­spec­tive/obser­va­tional expe­ri­ences and defi­nes a dis­ tinct sub­group of ger­ mi­
nal cen­
ter B-cell-like dif­
fuse large
sin­gle-arm pro­spec­tive stud­ies. In the relapsed and refrac­tory B-cell lym­phoma.J Clin Oncol. 2019;37(3):190-201.
5. Lap CJ, Nassereddine S, Dunleavy K. Novel bio­log­i­cal insights and new
set­ting, HGBL cases do not fare worse than DLBCL cases fol­low­
devel­op­ments in man­age­ment of Bur­kitt lym­phoma and high-grade B-cell
ing novel strat­e­gies, as discussed ear­lier. This sug­gests a role lym­phoma. Curr Treat Options Oncol. 2021;22(7):60.
for these approaches in ear­lier lines of ther­apy for HGBL. Addi- 6. Frontzek F, Ziepert M, Nickelsen M, et  al. Rituximab plus high-dose che­
tionally, many prom­is­ing novel agents are under inves­ti­ga­tion for mo­ther­apy (MegaCHOEP) or con­ven­tional che­mo­ther­apy (CHOEP-14) in
young, high-risk patients with aggres­sive B-cell lym­phoma: 10-year fol­low-
these dis­eases. Until we aug­ment cure rates sig­nif­i­cantly for this up of a randomised, open-label, phase 3 trial. Lancet Haematol. 2021;8(4):
patient pop­u­la­tion with a widely avail­­able strat­egy, it is a pri­or­ity e267-e277.
to con­sider refer­ral of HGBL patients to prom­is­ing clin­i­cal tri­als. 7. Younes A, Sehn LH, Johnson P, et al; PHOENIX Investigators. Randomized
phase III trial of ibrutinib and rituximab plus cyclo­phos­pha­mide, doxo­ru­bi­
cin, vin­cris­tine, and pred­ni­sone in non-ger­mi­nal cen­ter B-cell dif­fuse large
Conflict-of-inter­est dis­clo­sure B-cell lym­phoma. J Clin Oncol. 2019;37(15):1285-1295.
Kieron Dunleavy: advi­sory board mem­ber: Genmab, Morphosys, 8. Nowakowski GS, Chiappella A, Gascoyne RD, et  al. ROBUST: a phase III
Beigene, Daiichi Sankyo, Abbvie, ADC Therapeutics, Incyte, Astra study of lenalidomide plus R-CHOP ver­sus pla­cebo plus R-CHOP in pre­vi­
ously untreated patients with ABC-type dif­fuse large B-cell lym­phoma. J Clin
Zeneca, Genetech, Janssen.
Oncol. 2021;39(12):1317-1328.
9. Nowakowski GS, Hong F, Scott DW, et  al. Addition of lenalidomide to
Off-label drug use R-CHOP improves out­comes in newly diag­nosed dif­fuse large B-cell lym­
Kieron Dunleavy: nothing to disclose. phoma in a ran­dom­ized phase II US inter­group study ECOG-ACRIN E1412.
J Clin Oncol. 2021;39(12):1329-1338.
10. Ott G. Aggressive B-cell lym­pho­mas in the update of the 4th edi­tion of the
Correspondence World Health Organization clas­si­fi­ca­tion of haematopoietic and lym­phatic
Kieron Dunleavy, Division of Hematology and Oncology, George- tis­sues: refine­ments of the clas­si­fi­ca­tion, new enti­ties and genetic find­ings.
Br J Haematol. 2017;178(6):871-887.
town Lombardi Comprehensive Cancer Center, Georgetown
11. Alsharif R, Dunleavy K. Bur­kitt lym­phoma and other high-grade B-cell lym­
University Hospital, 3800 Reservoir Rd NW, Washington, DC pho­mas with or with­out MYC, BCL2, and/or BCL6 rearrangements. Hema-
20057; e-mail: kmd322@georgetown.edu. tol Oncol Clin North Am. 2019;33(4):587-596.

162  |  Hematology 2021  |  ASH Education Program


12. Hilton LK, Tang J, Ben-Neriah S, et al. The dou­ble-hit sig­na­ture identifies 23. Abramson JS, Palomba ML, Gordon LI, et al. Lisocabtagene maraleucel for
dou­ble-hit dif­fuse large B-cell lym­phoma with genetic events cryp­tic to patients with relapsed or refrac­tory large B-cell lym­pho­mas (TRANSCEND
FISH. Blood. 2019;134(18):1528-1532. NHL 001): a multicentre seam­less design study. Lancet. 2020;396(10254):
13. Song JY, Perry AM, Herrera AF, et al. Double-hit sig­na­ture with TP53 abnor­ 839-852.
mal­i­ties pre­dicts poor sur­vival in patients with ger­mi­nal cen­ter type dif­ 24. Caimi PF, Ai W, Alderuccio JP, et  al. Loncastuximab tesirine in relapsed or
fuse large B-cell lym­phoma treated with R-CHOP. Clin Cancer Res. 2021; refrac­tory dif­fuse large B-cell lym­phoma (LOTIS-2): a multicentre, open-label,
27(6):1671-1680. sin­gle-arm, phase 2 trial. Lancet Oncol. 2021;22(6):790-800.
14. Rosenwald A, Bens S, Advani R, et al. Prognostic sig­nif­i­cance of MYC rear- 25. Deng M, Xu-Monette ZY, Pham LV, et al. Aggressive B-cell lym­phoma with
rangement and trans­lo­ca­tion part­ner in dif­fuse large B-cell lym­phoma: a study MYC/TP53 dual alter­ations dis­plays dis­tinct clinicopathobiological fea­tures
by the Lunenburg Lymphoma Biomarker Consortium. J Clin Oncol. 2019;37 and response to novel targeted agents. Mol Cancer Res. 2021;19(2):249-
(35):3359-3368. 260.
15. Dunleavy K, Fanale MA, Abramson JS, et  al. Dose-adjusted EPOCH-R 26. Fiskus W, Mill CP, Perera D, et al. BET pro­te­­ol­y­sis targeted chi­mera-based
(etoposide, pred­ni­sone, vin­cris­tine, cyclo­phos­pha­mide, doxo­ru­bi­cin, and ther­apy of novel mod­els of Richter trans­for­ma­tion-dif­fuse large B-cell lym­
rituximab)in untreated aggres­sive dif­fuse large B-cell lym­phoma with MYC phoma. Leukemia. 2021;35(9):2621-2634.
rearrangement: a pro­spec­tive, multicentre, sin­gle-arm phase 2 study. 27. Morschhauser F, Feugier P, Flinn IW, et al. A phase 2 study of venetoclax
Lancet Haematol. 2018;5(12):e609-e617. plus R-CHOP as first-line treat­ment for patients with dif­fuse large B-cell

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/157/1852000/157dunleavy.pdf by guest on 13 December 2021


16. McMillan AK, Phil­lips EH, Kirkwood AA, et al. Favourable out­comes for high- lym­phoma. Blood. 2021;137(5):600-609.
risk dif­fuse large B-cell lym­phoma (IPI 3-5) treated with front-line R-CODOX- 28. Neelapu S, Dickinson M, Ulrickson M, et  al. Interim anal­y­sis of ZUMA-12:
M/R-IVAC che­mo­ther­apy: results of a phase 2 UK NCRI trial. Ann Oncol. a phase 2 study of axicabtagene ciloleucel (Axi-Cel) as first-line ther­apy
2020;31(9):1251-1259. in patients with high risk large B-cell lym­phoma. Abstract presented at:
17. Leppä S, Jørgensen J, Tierens A, et al. Patients with high-risk DLBCL ben­e­ 62nd Amer­i­can Society of Hematology Annual Meeting and Exposition; 5-8
fit from dose-dense immunochemotherapy com­bined with early sys­temic Decem­ber 2020; Atlanta, GA. Abstract 405.
CNS pro­phy­laxis. Blood Adv. 2020;4(9):1906-1915. 29. Chamuleau MED, Burggraaff CN, Nijland M, et al. Treatment of patients with
18. Laude MC, Lebras L, Sesques P, et al. First-line treat­ment of dou­ble-hit and MYC rearrangement pos­ i­
tive large B-cell lym­phoma with R-CHOP plus
tri­ple-hit lym­pho­mas: sur­vival and tol­er­ance data from a ret­ro­spec­tive lenalidomide: results of a mul­ti­cen­ter HOVON phase II trial. Haematolog-
mul­ti­cen­ter French study. Am J Hematol. 2021;96(3):302-311. ica. 2020;105(12):2805-2812.
19. Torka P, Kothari SK, Sundaram S, et al. Outcomes of patients with lim­ited- 30. Scott DW, King RL, Staiger AM, et  al. High-grade B-cell lym­phoma with
stage aggres­sive large B-cell lym­phoma with high-risk cyto­ge­net­ics. Blood MYC and BCL2 and/or BCL6 rearrangements with dif­fuse large B-cell lym­
Adv. 2020;4(2):253-262. phoma mor­phol­ogy. Blood. 2018;131(18):2060-2064.
20. Wilson MR, Eyre TA, Martinez-Calle N, et  al. Timing of high-dose meth­o­ 31. Dunleavy K. Double-hit lym­pho­mas: cur­rent par­a­digms and novel treat­ment
trex­ate CNS pro­phy­laxis in DLBCL: an anal­y­sis of tox­ic­ity and impact on approaches. Hematology Am Soc Hematol Educ Program. 2014;2014(1):
R-CHOP deliv­ery. Blood Adv. 2020;4(15):3586-3593. 107-112.
21. Chin CK, Cheah CY. How I treat patients with aggres­sive lym­phoma at high
risk of CNS relapse. Blood. 2017;130(7):867-874.
22. Herrera AF, Mei M, Low L, et al. Relapsed or refrac­tory dou­ble-expressor
and dou­ble-hit lym­pho­mas have infe­rior pro­gres­sion-free sur­vival after © 2021 by The Amer­i­can Society of Hematology
autol­o­gous stem-cell trans­plan­ta­tion. J Clin Oncol. 2017;35(1):24-31. DOI 10.1182/hema­tol­ogy.2021000247

Prakash Singh Shekhawat


New devel­op­ments in aggres­sive B-cell lym­phoma  |  163
DEFEATING DIFFUSE, DOUBLE - HIT, AND DOGGED NON - HODGKIN LYMPHOMA

Relapsed disease: off-the-shelf immunotherapies


vs customized engineered products

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/164/1851658/164karmali.pdf by guest on 13 December 2021


Reem Karmali
Northwestern University Feinberg School of Medicine, Chicago, IL

Innovations in immuno-oncology for lymphomas have outpaced therapeutic developments in any other cancer histology.
In the 1990s, rituximab, a CD20 monoclonal antibody, drastically changed treatment paradigms for B-cell non-Hodgkin
lymphomas (B-NHLs). In parallel, the concept that T cells could be genetically reprogrammed and regulated to address
tumor cell evasion was developed. Twenty years later, this concept has materialized—3 customized engineered CD19 chi-
meric antigen receptor T-cell (CART) constructs have been embraced as third-line therapies and beyond for aggressive
B-NHL. Responses with CARTs are durable in 30% to 40% of patients, with consistent results in older patients, primary
refractory disease, high-grade B-cell lymphoma, and patients with concurrent secondary central nervous system dis-
ease, all features historically associated with poorer outcomes. Challenges associated with the administration of CARTs
include cumbersome and time-consuming manufacturing processes, toxicities, and cost, not to mention a substantial
risk of relapse. Fortunately, as our understanding of how to manipulate the immune system to achieve full antitumor
potential has grown, so has the rapid development of off-the-shelf immunotherapies, with CD20/CD3 bispecific antibod-
ies standing out above all others. These agents have shown promising activity in aggressive B-NHL and have the potential
to circumvent some of the challenges encountered with customized engineered products. However, toxicities remain
substantial, dosing schedules intensive, and experience limited with these agents. Novel customized and off-the-shelf
therapeutics as well as rational combinations of these agents are underway. Ultimately, growing experience with both
customized engineered and off-the-shelf immunotherapies will provide guidance on optimal methods of delivery and
sequencing.

LEARNING OBJECTIVES
• Understand the strengths and limitations of CD19 CARTs as a customized engineered product vs off-the-shelf
immunotherapies such as bispecifc CD20/CD3 antibodies for the treatment of aggressive B-NHL
• Review clinical effcacy and safety data for CD19 CARTs and bispecifc CD20/CD3 antibodies
• Optimize a therapeutic algorithm for relapsed/refractory aggressive B-cell lymphoma with the inclusion of CARTs
and off-the-shelf immunotherapies

CLINICAL CASE: PART 1 months later, the patient relapsed. He was treated with
A 65-year-old male presented with lower back and flank two cycles of R-ICE with complete response (CR) and
pain, fevers, and weight loss. Magnetic resonance imag- consolidated with an autologous stem cell transplanta-
ing of the lumbar spine showed a paraspinal mass. Posi- tion (ASCT). Unfortunately, scans 3 months post-ASCT
tron emission tomography-computed tomography scans demonstrated disease recurrence. He was referred to our
showed diffuse lymphadenopathy with bone marrow institution to discuss treatment options for his second
involvement and highest uptake in bulky retroperitoneal relapse.
lymph nodes and the paraspinal mass. A core biopsy of
the paraspinal mass confrmed high-grade B-cell lym-
phoma with dual rearrangements of MYC and BCL2 (also Introduction
known as double-hit lymphoma). The patient was treated To date, salvage high-dose chemotherapy with ASCT re-
with six cycles of DA-EPOCH-R and achieved a complete mains the standard second-line treatment for relapsed or
metabolic response at the completion of therapy. Twelve refractory (R/R) diffuse large B-cell lymphoma (DLBCL)

164 | Hematology 2021 | ASH Education Program


regard­less of under­ly­ing high-risk bio­logic fea­tures.1 Howev- syn­drome (ICANS). Restaging scans 30 days and 90 days post-
er, few patients are cured with this inten­sive approach, and CART dem­on­strated a com­plete met­a­bolic response.
appli­ca­bil­ity is lim­ited by comorbidities, advanced age, and/or
che­mo­ther­apy-insen­si­tive dis­ease.2,3 In the era predating the
use of immunotherapies, patients with refrac­tory dis­ease or
relapse within 12 months of ASCT had dis­mal out­comes. In the Limitations of CARTs and the emer­gence of off-the-shelf
SCHOLAR-1 mul­ti­cen­ter ret­ro­spec­tive study, the objec­tive re- immunotherapies
sponse rate (ORR) to the next line of ther­apy was 26% (CR, 7%), Although CARTs have changed the treat­ment par­a­digm for R/R
with a median over­all sur­vival (OS) rate of 6.3 months in such aggres­sive B-cell lym­pho­mas, the ther­a­peu­tic has its lim­i­ta­tions.
patients.2 First, CARTs have to be engineered for each indi­vid­ual patient,
Fortunately, the treat­ment land­scape has rap­idly evolved for with a poten­tial for logis­ti­cal delays from the time of patient
R/R DLBCL, with cus­tom­ized engineered immunotherapies—more iden­ti­fi­ca­tion to CART infu­sion as well as a risk of manufactur­ing
spe­cif­i­cally, CD19 chi­me­ric anti­gen recep­tor T cells (CARTs)—and fail­ure. Second, sig­nif­i­cant toxicities are asso­ci­ated with CART
ther­apy that include CRS and ICANS. Such toxicities may pre­

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/164/1851658/164karmali.pdf by guest on 13 December 2021


off-the-shelf immunotherapies tak­ing cen­ter stage.
clude patients with cer­tain comorbid con­di­tions. Most impor­
CARTs ther­apy tantly, although CARTs offer dura­ble responses in some, 60% to
CARTs are autol­o­gous T cells that have been genet­i­cally reengi- 70% of patients will still relapse.4,7,8
neered using viral trans­duc­tion to express a CAR that tar­gets a Since the approval of CARTs for aggres­sive B-cell ther­apy, the
spe­cific tumor anti­gen. For B-cell lym­pho­mas, the CAR includes FDA has approved a wave of immunotherapies (with or with­out
an extra­cel­lu­lar moi­ety derived from an anti-CD19 sin­gle-chain var­ che­mo­ther­apy) and targeted approaches for R/R DLBCL. These
i­able frag­ment for anti­gen rec­og­ni­tion and intra­cel­lu­lar domains include the com­bi­na­tions of anti-CD19 mono­clo­nal anti­body
includ­ing a costimulatory domain, CD28 or 41BB, in tan­dem with (mAb) tafasitamab and lenalidomide,9 anti-CD79b anti­body-drug
a CD3ζ-acti­vat­ing domain. The US Food and Drug Administration con­ju­gate (ADC) polatuzumab vedotin with bendamustine and
(FDA) has approved three con­structs for the treat­ment of R/R rituximab,10 and monotherapy with anti-CD19 ADC loncastuximab
aggres­sive B-cell lym­pho­mas, includ­ing DLBCL, high-grade B-cell tesirine,11 or selinexor, an orally avail­­able selec­tive inhib­i­tor of
lym­phoma, transformed fol­lic­u­lar lym­phoma, and pri­mary medi­as­ nuclear export.12 Each of these options should be con­sid­ered
ti­nal B-cell lym­phoma, after 2 prior lines of sys­temic ther­apy, and for patients who are either poor can­di­dates for CART or who
they show high response rates with dura­ble remis­sions. The first relapse after CART, although data supporting these appli­ca­tions
con­struct, approved in 2017, for this pop­u­la­tion was axicabtagene are lim­ited.
ciloleucel (axi-cel), containing a CD28 costimulatory domain. The In addi­tion to the mAbs and ADCs listed above, a num­ber
mul­ti­cen­ter phase 1/2 ZUMA-1 trial eval­u­ated axi-cel and had the of other off-the-shelf immunotherapies have been eval­u­ated in
lon­gest fol­low-up of all­CART tri­als of greater than 4 years (n = 101); aggres­sive lym­pho­mas (Figure 1).13-20 Bispecific T-cell-engag­ing
responses were dura­ble, with a median OS of 25.8 months and antibodies (BsAbs) have emerged as a novel class of off-the-shelf
a 4-year OS rate of 44%.4,5 The phase 2 JULIET study of tisagen- immunotherapies with clear effi­ cacy in R/R aggres­ sive B-cell
lecleucel dem­on­strated that the effi­cacy is com­pa­ra­ble for this lym­ pho­ mas, includ­ing for those patients relaps­ ing after CART
41BB-containing CART, with a more favor­able tox­ic­ity pro­file.6,7 The ther­apy. BsAbs are designed to simul­ta­neously bind to CD3 epsi­
TRANSCEND study, the larg­est CART trial, eval­u­ated the 41BB con­ lon, a com­po­nent of the T-cell recep­tor com­plex, and CD20 on
struct lisocabtagene maraleucel (liso-cel), manufactured uniquely the cell sur­face of malig­nant B cells, cre­at­ing an “immune syn­
through the sep­a­rate trans­duc­tion, expan­sion, and admin­is­tra­tion apse” that redi­rects T-cell cyto­toxic activ­ity against malig­nant B
of equal tar­get doses of CD4+ and CD8+ CARTs. This trial estab- cells. Four agents eval­u­ated in R/R aggres­sive B-cell lym­pho­mas
lished the appli­ca­bil­ity of CARTs to a broader pop­u­la­tion, includ­ include mosunetuzumab, epcoritamab, glofitamab, and odronex-
ing patients with prior allo­ge­neic stem cell trans­plan­ta­tion and tamab.15-19 Unlike their pre­de­ces­sor blinatumomab, a CD19/CD3
those with sec­ond­ary cen­tral ner­vous sys­tem involve­ment.8 bispecific T-cell engager, CD20/CD3 BsAbs have a lon­ger half-life,
allowing for greater ease of admin­is­tra­tion, and appear to offer
higher response rates in R/R aggres­sive B-cell lym­pho­mas.20 Fur-
thermore, CD20/CD3 BsAbs have the poten­tial to cir­cum­vent the
short­com­ings of CARTs while pro­vid­ing high rates of response.20
CLINICAL CASE: PART 2 Herein, the focus is on com­par­ing and contrasting fea­tures
The patient was enrolled in the TRANSCEND trial with liso-cel. The of CARTs and BsAbs as pro­to­types of cus­tom engineered vs off-
patient under­went leukapheresis for T cells followed by bridg­ing the-shelf immunotherapies, respec­tively, with the strengths and
ther­apy with rituximab and high-dose ste­roids for rap­idly pro­ lim­i­ta­tions of each modal­ity outlined (Table 1).
gres­sive dis­ease. Four weeks after leukapheresis, manufactur­ing
was com­plete. Lymphodepleting che­mo­ther­apy with fludara- Off-the-shelf immunotherapies vs CARTs: ease
bine at 30 mg/m2/d intra­ve­nously (IV) and cyclo­phos­pha­mide at of admin­is­tra­tion
300 mg/m2/d IV for 3 days was admin­is­tered, followed by infu-­ CARTs are cus­tom­ized prod­ucts engineered for each indi­vid­ual
sion of liso-cel at a dose of 100 × 106 cells. He expe­ri­enced grade patient. The manufactur­ing pro­cess has been refined to ensure
2 cyto­kine release syn­drome (CRS) with fever, mild hypo­ten­sion that the end prod­ucts meet spec­i­fi­ca­tions for via­bil­ity and com­
requir­ing intra­ve­nous flu­ids, and mild hyp­oxia 6 days after the po­si­tion.21 However, despite every pre­cau­tion taken, suc­cess­ful
admin­is­tra­tion of CART and was man­aged effec­tively with the IL- man­u­fac­ture is not guaranteed—the final prod­uct may not meet
6 recep­tor antag­o­nist tocilizumab. This resolved within 5 days. spec­i­fi­ca­tions or may entirely fail to gen­er­ate. Furthermore, the
He had no signs of immune effec­torPrakash Singh Shekhawatcess can be lengthy.
cell-asso­ci­ated neu­
ro­
logic pro­

Off-the-shelf vs cus­tom engineered immu­no­ther­apy | 165


Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/164/1851658/164karmali.pdf by guest on 13 December 2021
Figure 1.  Evolving landscape for customized engineered and off-the-shelf immunotherapies in aggressive B-NHL. ADCC: antibody-
dependent cell cytotoxicity; ADCP, antibody-dependent cellular phagocytosis; BiTE, bispecific T-cell engager; PD-1, programmed cell
death 1; PD-L1, programmed cell death ligand 1.

For both the ZUMA-1 and JULIET tri­als, a min­i­mum abso­lute malig­nant cells with IL-6 as a pri­mary driver.4,23 Rates of ICANS
lym­pho­cyte count was required, which may be pro­hib­i­tive range from 21% to 64%, with grade ≥3 events described in 10%
in heavily pretreated patients. Rates of manufactur­ ing fail­
ure to 28%.4,7,8 The mech­a­nism of ICANS is elu­sive but has been asso­
ranged from 1% to 7%. Conversely, in the TRANSCEND trial, ci­ated with high cyto­kine lev­els as well.4
despite lax­ity in eli­gi­bil­ity with no require­ment for min­i­mum Variability in the pre­sen­ta­tion, prev­al­ence, and inten­sity of
abso­lute lym­pho­cyte count, manufactur­ing fail­ure occurred in CRS and ICANS across CART con­structs has been attrib­uted to
only 2 patients.4,7,8 patient-related, dis­ease-related, and prod­uct-spe­cific fac­tors.
Factoring in the time from patient iden­ti­fi­ca­tion, leukaphere- For prod­uct-spe­cific fac­tors, dif­fer­ences in T-cell expan­sion and
sis, and manufactur­ing to even­tual admin­is­tra­tion, with expected pro­lif­er­a­tion kinet­ics con­ferred by the CD28 vs 41BB costimula-
logis­ti­cal delays along the way, the turn­around time for CARTs is tion domains may explain the higher CRS and ICANS rates asso­
unpre­dict­able and typ­i­cally greater than 3 to 4 weeks. Accord- ci­ated with axi-cel.4,7,8
ingly, CARTs may not be fea­si­ble for patients with rap­idly pro­ Although the prev­ a­
lence of CRS and ICANS is high with
gres­sive dis­ease. Bridging ther­apy is often needed dur­ing the CARTs, these toxicities are effec­ tively man­ aged. Early mit­ i­
manufactur­ing period, with lim­ited guid­ance as to which ther­a­ ga­tion strat­e­gies with anti-IL-6 ther­apy and/or ste­roids have
pies are most effec­tive for this pur­pose. Allogeneic off-the- shelf improved the safety pro­file of CARTs with­out hav­ing an impact
CARTs would solve this par­tic­u­lar issue, lead­ing to timely acces­ on CART func­tion, effi­cacy, or per­sis­tence.24,25 Real-world data
si­bil­ity, but their devel­op­ment remains in infancy.22 with CARTs and the move­ment toward out­pa­tient CART admin­
Bispecific antibodies as an off-the-shelf option can be used is­tra­tion are tes­ta­ments to suc­cess­ful tox­ic­ity mit­i­ga­tion.26,27 For
with­out ex vivo T-cell prep­a­ra­tion, allowing imme­di­ate treat­ example, data cap­tured from real-world expe­ri­ence with axi-cel
ment. However, unlike CARTs, treat­ment dura­tion with BsAbs is showed that 43% of patients would not have met eli­gi­bil­ity cri­
prolonged, cre­at­ing issues for acces­si­bil­ity and ease of admin­is­ te­ria for the registrational ZUMA-1 trial because of comorbidi-
tra­tion. These agents are admin­is­tered every 1 to 4 weeks either ties. Despite the inclu­sion of older and sicker patients, toxicities
IV or sub­cu­ta­ne­ously, with shorter inter­vals early in the treat­ment and clin­i­cal out­comes were sim­i­lar for these patients com­pared
course, and may be pur­sued for 12 cycles and beyond, depend- to out­comes in the piv­otal trial.26 Additionally, the fea­si­bil­ity of
ing on the agent used and the dura­bil­ity of response.15-17,19 CART admin­is­tra­tion has been dem­on­strated more for­mally in
the older adult and unfit pop­u­la­tion. The phase 2 PILOT trial
Off-the-shelf immunotherapies vs CARTs: toxicities was the first to assess the safety and effi­cacy of liso-cel as a
and appli­ca­tion in vul­ner­a­ble pop­u­la­tions sec­ond-line ther­apy for trans­plant-inel­i­gi­ble patients with R/R
Side effects asso­ci­ated with CARTs include CRS and ICANS, pro- aggres­sive lym­phoma. This included patients ≥70 years of age or
longed cytopenia, and impair­ment of humoral immu­nity with with impaired organ func­tion includ­ing mod­er­ate car­dio­my­op­a­
increased risk of infec­tion (Figure 2). CRS is the most com­mon thy (left ventricular ejection fraction ≥40%-50%) and/or  pul­mo­
and has been described in 42% to 93% of patients, with grade nary  impair­ment  (DLCO ≤60% but blood-oxygen saturation ≥ 92%).
≥3 events occur­ring in 2% to 22% of patients.4,7,8 The path­o­phys­i­­ Rates of CRS, ICANS, and response were com­pa­ra­ble to those
ol­ogy of CRS has been attrib­uted to an upsurge in cyto­kines and of the TRANSCEND study with liso-cel in third line ther­apy and
chemokines upon acti­va­tion of CARTs after engage­ment with beyond.28

166  |  Hematology 2021  |  ASH Education Program


Table 1.  Summary of clin­i­cal tri­als for CD19 CARTs and bispecific antibodies

Axi-cel4,34 Tisa-cel6,7 Liso-cel8 Mosunetuzumab15 Epcoritamab16 Glofitamab17,18 Odronextamab19


Structure CD19/CD3z/CD28 CD19/CD3z/41BB CD19/CD3z/41BB Full-length human­ CD20/CD3 BsAb CD20/CD3 Hinge-sta­bi­lized,
ized, IgG1 BsAb with fully human IgG4
CD20/CD3 BsAb 2:1 molec­u­lar CD20/CD3 BsAb
con­fig­u­ra­tion
with biva­lent
bind­ing to
CD20 and
mono­va­lent
bind­ing to
CD3
Route of IV IV IV IV or SC SC IV IV
admin­is­tra­tion
Trial/NCT ZUMA-1 JULIET TRANSCEND NCT02500407 NCT03625037 NCT03075696 NCT02290951
N = treated 101 111 269 141 45 127 71
(aggres­sive
his­tol­ogy)
Median lines 3 (2-4) Median not 3 (2-4) 3 (1-14); 23 pts with 3 (1-6); 6 pts with 3 (1-13) 3 (1-11); 29 pts with
prior ther­apy reported; prior CART prior CART prior CART
(range) range 1-6
Dosing 2 × 106/kg × 1 dose 0.6-6.0 × 108 × 1 0.5-1.0 × 108 × 1 dose Step-up dos­ing on Flat dose in 28-day Step-up dos­ing Step-up dose
dose days 1, 8, and 15 of cycles (q1w: on cycle (C) consisting of ini­tial
cycle 1, followed cycles 1–2; q2w: 1, day (D) 1 dose at week (W)
by fixed doses on cycles 3–6; q4w and 8 and 1, an inter­me­di­ate

Prakash Singh Shekhawat


day 1 of each 21- there­af­ter) until then at the dose at W2, and
day cycle for up to dis­ease pro­gres­ tar­get dose there­af­ter a fixed
17 cycles sion or unac­cept­ from C2D1, q weekly dose until
able tox­ic­ity 3 weeks for up W12 followed by
to 12 cycles main­te­nance q2w
Median time to Leukapheresis → Enrollment → Leukapheresis → infu­
man­u­fac­ture infu­sion: 17 days infu­sion: 21- sion: 24-day tar­get
day tar­get Immediate off-the-shelf immu­no­ther­apy
Median 4 years 40.3 months 18.8 months Not reported 8.3 months 13.5 months 3.9 months
fol­low-up
ORR (%) 82 52 73 35 67 71 • Without prior CART
(n = 11): ORR 55%
• Post-CART (n = 24):
ORR 33%
CR (%) 54 40 53 19 33 64 • Without prior CART
(n = 11): CR 55%
• Post-CART (n = 24):
CR 21%

Off-the-shelf vs cus­tom engineered immu­no­ther­apy | 167


Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/164/1851658/164karmali.pdf by guest on 13 December 2021
Table 1.  Summary of clin­i­cal tri­als for CD19 CARTs and bispecific antibodies (Continued)

Axi-cel4,34 Tisa-cel6,7 Liso-cel8 Mosunetuzumab15 Epcoritamab16 Glofitamab17,18 Odronextamab19


Median DOR 8.1 Not reached Not reached Median time from Not yet mature 5.5 • Without prior CART:
(mo) first CR: 8.8 10.3
• Post-CART: 2.8
Median PFS (mo) 5.8 <6 months; not 6.8 Not yet mature Not yet mature 2.9 Not yet mature

168  |  Hematology 2021  |  ASH Education Program


reached in
CRs
Median OS (mo) 25.8 11.1 21.1 Not yet mature Not yet mature Not yet mature Not yet mature
CRS (%) All grades: 93 All grades: 58 All grades: 42 All grades: 28 All grades: 58 All grades: 50 All grades: 62
≥G3: 13 ≥G3: 22 ≥G3: 2 ≥G3: 1.4 ≥G3: 0 ≥G3: 3.5 ≥G3: 7
ICANS (%) All grades: 64 All grades: 21 All grades: 30 All grades: 1.4 All grades: 6 All grades: 5.3 All grades: not
≥G3: 28 ≥G3: 12 ≥G3:10 ≥G3: 0 ≥G3: 3 ≥G3: 0 reported
≥G3: 2.3
Population appli­ DLBCL/PMBCL/TFL DLBCL/TFL DLBCL/PMBCL/trans­ Aggressive B-cell Aggressive B-cell Aggressive B-cell Aggressive B-cell lym­
ca­bil­ity/con­ for­ma­tion from lym­phoma prior to lym­phoma prior to lym­phoma phoma prior to and
sid­er­ations indo­lent lym­phoma; and post-CART and post-CART post-CART
FL grade 3B; pre­
ferred for sys­temic
+ con­cur­rent sec­
ond­ary CNS dis­ease
or post-alloSCT
alloSCT, allo­ge­neic stem cell trans­plan­ta­tion; DOR, dura­tion of response; FL: follicular lymphoma; IgG: immu­no­glob­u­lin G; PMBCL, pri­mary medi­as­ti­nal B-cell lym­phoma; TFL, transformed
fol­lic­u­lar lym­phoma; tisa-cel, tisagenlecleucel.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/164/1851658/164karmali.pdf by guest on 13 December 2021


Figure 2.  CART or BsAb treatment-related adverse effects of interest with an incidence of ≥10% and ≥5%, respectively.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/164/1851658/164karmali.pdf by guest on 13 December 2021


Taken col­lec­tively, such toxicities should not pre­clude the use Off-the-shelf immunotherapies vs CARTs: effi­cacy
of CARTs in older patients. Based on clin­i­cal trial and real-world and sequenc­ing
expe­ri­ence, both CD28 and 41BB con­structs appear to be rea­ For all 3 FDA-approved CD19 CARTs, the pat­terns and dura­bil­ity of
son­able options for older adult and/or frail patients or those who response are sim­i­lar (Table 1). Response rates range from 52% to
have comorbid con­di­tions, rec­og­niz­ing the dif­fer­ences in tox­ic­ 82%, with CR rates of 40% to 54%.4,7,8 Long-term fol­low-up data
ity pro­files for these con­structs. Practically, how­ever, one might for CARTs sug­gest that these responses are dura­ble, par­tic­u­larly
favor the use of 41BB con­structs in patients with under­ly­ing neu­ for patients in CR. For the ZUMA-1 study, the median fol­low-up
ro­logic comorbidities, given the lower rates of ICANS asso­ci­ is now greater than 4 years, the median OS rate is 25.8 months,
ated with these con­structs. Additionally, the grow­ing prac­tice and the 4-year OS rate is 44% (n = 101).5 In the JULIET study, the
of out­pa­tient admin­is­tra­tion of CARTs with pref­er­ence for 41BB median fol­low-up is 40.3 months (n = 115). Although the median
con­structs in this con­text is likely to lead to wider access and OS was 11.1 months, the pro­gres­sion-free sur­vival (PFS) rates at
uti­li­za­tion of this ther­a­peu­tic. 24 and 36 months were 33% and 31%, suggesting that few pa-
Bispecific antibodies can also pro­ duce CRS and neu­ ro­
logic tients who achieve a CR will relapse beyond 24 months.6 For the
toxicities, seem­ingly at lower fre­quen­cies and sever­ity, although TRANSCEND study, with a shorter fol­low-up, median PFS and OS
tox­ic­ity data are emerg­ing and not yet mature. Additional toxici- were 6.8 and 21.1 months, respectively.8
ties described for BsAbs with an inci­dence of ≥10% include pyrexia, For BsAbs, response rates in aggres­sive lym­pho­mas range
reac­tion at the injec­tion site, head­aches, and cytopenia (Figure 2). from 33% to 71%, with CRs of 19% to 64%, and may depend on
Rates of CRS for BsAbs range from 28% to 62%, with grade ≥3 prior CART expo­sure. However, expe­ri­ence with BsAbs is still
events in 0% to 7% of patients, and tend to dis­si­pate after 1 to lim­ited; fol­low-up is short and data on dura­bil­ity of response
2 cycles of admin­is­tra­tion.15-17,19 CRS appears to be driven by IL-6 are lacking. Similarly, the impact of these agents on sur­vival
with BsAbs as well and is man­aged effec­tively with tocilizumab com­pared to CART is not clear. What is clear is that these
if needed.15-17,19 Rates of ICANS for BsAbs are not clearly reported; agents do main­tain their effects in patients with relapse after
rates of grade ≥3 events range from 0% to 3%.15-17,19 Clinical and bio­ CART (Table 1).
logic pre­dic­tors of CRS with BsAbs remain unclear. For mosunetu- For instance, results for mosunetuzumab in 30 patients who
zumab, aggres­ sive dis­ ease his­
tol­
ogy and a base­ line ele­vated had received prior CART ther­apy were high­lighted, and 18 patients
C-reac­tive pro­tein appear to pre­dict greater neu­ro­logic tox­ic­ity.29 were eval­u­ated for response. In this sub­group, mosunetuzumab
Like CARTs, BsAbs have dem­ on­
strated fea­si­
bil­ity in older led to CART expan­sion and gen­er­ated an ORR of 39% and a CR
patients and patients with comorbid con­ di­
tions. As a sin­ gle rate of 22% with long-last­ing responses and tol­er­a­ble safety.15
agent, mosunetuzumab was eval­u­ated as front­line ther­apy in 19 Similarly, odronextamab was eval­u­ated in patients post-CARTs
patients aged ≥80 years or 60 to 79 years with func­tional impair­ (n = 24) and dem­on­strated encour­ag­ing activ­ity with an ORR of
ments or comorbid con­di­tions pre­clud­ing the use of full-dose 33% and a CR rate of 21%.19
chemo-immu­no­ther­apy and dem­on­strated effi­cacy with remark­ With clin­i­cal expe­ri­ence of sequenc­ing strat­e­gies in R/R
able tol­er­a­bil­ity.30 DLBCL essen­tially lim­ited to CARTs as a third-line ther­apy fol-
Furthermore, a num­ber of strat­e­gies are being employed to lowed by BsAbs, this sequence remains favored (Figure 3).
opti­mize the dos­ing and tol­er­a­bil­ity of BsAbs. For exam­ple, step- One could con­sider CD20/CD3 BsAbs as a bridge to CARTs in
up dos­ing for BsAbs is rou­tine. The sub­cu­ta­ne­ous for­mu­la­tion of patients with rap­idly pro­gres­sive dis­ease or even as a bridge
mosunetuzumab was shown to reduce the sever­ity of CRS; CRS to allo­ge­neic stem cell trans­plan­ta­tion. Given the poten­tial for
events were mild, tran­sient, and delayed in onset and required T-cell exhaus­tion with programmed cell death ligand 1 upreg-
min­i­mal inter­ven­tion with no grade ≥3 events reported.31 With ulation in tar­get cells seen with BsAbs, whether uti­liz­ing BsAbs
glofitamab, the use of a cytoreductive anti-CD20 mAb and step- prior to leukapheresis could have an impact on the qual­ ity
up dos­ing have been shown to mit­i­gate CRS.17,18 Although it is of harvested T cells for CART man­u­fac­ture is ques­tion­able.32
unclear which strat­egy is most effec­tive in decreas­ing tox­ic­ity, col­ As both CARTs and BsAbs make their way to ear­lier lines of
lec­tively, these strat­e­gies may allow for higher-dose drug admin­ ther­apy, how best to sequence these agents will con­tinue to
is­tra­tion and foreseeably improved response rates with BsAbs. evolve.30,33
Prakash Singh Shekhawat
Off-the-shelf vs cus­tom engineered immu­no­ther­apy | 169
Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/164/1851658/164karmali.pdf by guest on 13 December 2021
Figure 3.  Algorithm for preferred and alternative treatment options for R/R DLBCL that includes customized engineered and off-
the-shelf immunotherapies.

Off-the-shelf immunotherapies vs CARTs: effi­cacy biopsy of a ret­ro­per­i­to­neal node iden­ti­fied CD19− relapse of
in high-risk pop­u­la­tions dis­ease. The patient was sub­se­quently offered a clin­i­cal trial
Ahead of off-the-shelf immunotherapies, CARTs are being eval­ with a novel CD20/CD3 bispecific anti­body.
u­ated in sev­eral patient sub­sets with poor prog­no­ses and high
unmet needs. First, patients with dou­ble-expressor or high-grade
B-cell lym­pho­mas with rearrangements of MYC and BCL2 and/or
BCL6, also known as dou­ble- and tri­ple-hit lym­pho­mas, were in- Off-the-shelf immunotherapies vs CARTs: mech­a­nisms
cluded in piv­otal tri­als for all 3 FDA-approved CARTs. Response of resis­tance and future direc­tions
rates in these sub­sets were sim­i­lar to those seen for all­patients.7,8,34 Predictors of response and relapse with regard to both engi-
Trials are also under­way for CARTs in patients with intrin­sic neered prod­ucts and off-the-shelf ther­ap ­ ies remain elu­sive. For
che­mo­ther­apy resis­tance. Both the ZUMA-7 (NCT03391466) and CARTs, it is clear that relapses can occur despite the per­sis­
TRANSFORM (NCT03575351) tri­als have com­pared axi-cel or liso- tence of reengineered T cells. CART exhaus­tion stem­ming from
cel, respec­tively, vs ASCT as sec­ond-line ther­apy for patients with an immu­no­sup­pres­sive tumor micro­en­vi­ron­ment (TME) and host
pri­mary refrac­tory dis­ease or relapse within 12 months of front­line sys­temic inflam­ma­tion along with intrin­sic T-cell dys­func­tion
ther­apy, with mature results eagerly awaited. The ZUMA-12 study may explain this phe­nom­e­non.6,36 These find­ings sug­gest oppor­
is eval­u­at­ing axi-cel in patients with large B-cell lym­phoma who tu­ni­ties for com­bi­na­tions with immuno-onco­log­i­cal agents such
had either high-grade lym­phoma or an international prognostic as check­point inhib­i­tors, tyro­sine kinase inhib­i­tors, and immu­
index score ≥3 and a pos­i­tive interim positron emission tomog- no­mod­u­la­tory agents that may reinvigorate per­sis­tent CARTs,
raphy after 2 cycles of R-CHOP/R-CHOP-like ther­apy.35 Thus far, although this runs the risk of increased tox­ic­ity.37
of 12 response-evaluable patients the ORR is 92%, with a CR rate Given that BsAbs also rely on the patient’s own T cells, one
of 75%. Longer-term fol­low-up of these tri­als will pro­vide greater expects T-cell exhaus­tion and dys­func­tion to be rel­e­vant mech­a­
insight into the ben­e­fit of CARTs in pri­mary refrac­tory patients. nisms of resis­tance to said ther­a­peu­tics as well.
Experience with off-the-shelf immunotherapies is lim­ited in Allogeneic CARTs afford the oppor­tu­nity to min­i­mize the con­
these high-risk pop­u­la­tions. In fact, for the L-MIND study, which tri­bu­tion of T-cell dys­func­tion to relapse risk but may not be a ­ ble
eval­u­ated the CD19 human­ized anti­body tafasitamab with lena- to over­come the immu­no­sup­pres­sive effects of the TME (Table 2).
lidomide, patients with pri­mary refrac­tory dis­ease and/or high- Limitations asso­ci­ated with this modal­ity also include a risk of
grade B-cell lym­pho­mas with rearrangements of MYC and BCL2 increased immune toxicities, graft-ver­sus-host dis­ease, and pos­
and/or BCL6 were excluded.9 Data for BsAbs remain imma­ture, si­ble rejec­tion.21 Allogeneic nat­u­ral killer (NK) CARs rep­re­sent
with inad­e­quate ana­ly­ses of sub­set pop­u­la­tions with high-grade another immunocellular plat­form with sev­eral advan­tages over
B-cell lym­phoma and/or refrac­tory dis­ease. It is antic­i­pated that allo­ge­neic CARTs—they can be selected from non-HLA related
this infor­ma­tion will become more read­ily avail­­able with ongo­ing healthy donors, will not cause graft-ver­ sus-host dis­
ease, and
fol­low-up. are less prone to the inhib­i­tory effects of the TME (Table 2).38
Similarly, bispecific dual-affinity retargeting (DART) pro­ teins
designed to tar­get LAG3 and programmed cell death 1 may bet­
ter over­come the neg­a­tive effects of the TME and have dem­
CLINICAL CASE: PART 3 on­strated responses in CART-treated and naive patients.39 As an
Surveillance scans in our patient were conducted 180 days added ben­e­fit, all­ afore­men­tioned prod­ucts rep­re­sent off-the-
post-CART with con­cern for relapse in the retroperitoneum. A shelf options.

170  |  Hematology 2021  |  ASH Education Program


Table 2.  Novel dual-targeted autol­o­gous CARTs, allo­ge­neic CARTs, and NK CARs cur­rently in clin­i­cal trial

Clinicaltrials​­.gov iden­ti­fier Sponsor Phase Target Class


Dual-targeted autol­o­gous CARTs
NCT04186520 Medical College of Wisconsin 1/2 CD19/CD20 Autologous dual-tar­get CART
NCT03287817 Autolus Limited 1/2 CD19/CD22 Autologous dual-tar­get CART
followed by lim­ited dura­
tion of anti-PD-1 anti­body
pembrolizumab
Allogeneic CARTs
NCT03939026 Allogene Therapeutics 1/2 CD19 Single-tar­get allo­ge­neic CART
NCT03398967 Chi­nese PLA General Hospital 1/2 CD19/CD20 or CD19/CD22 Dual-tar­get allo­ge­neic CART

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/164/1851658/164karmali.pdf by guest on 13 December 2021


NK CARs
NCT03056339 M. D. Anderson Cancer Center 1/2 CD19 Cord blood CD19 NK CAR
NCT03774654 Baylor College of Medicine 1 CD19 NK CAR

In rare cir­cum­stances, relapses after CD19 CARTs have been these toxicities are milder and may not require such close atten­
attrib­uted to CD19 anti­gen escape. Mechanisms of anti­gen escape tion. However, the fre­quency and dura­tion of admin­is­tra­tion can
include altered CD19 mem­brane traf­fick­ing and/or inter­nal­i­za­tion, be cum­ber­some. Ultimately, a clearer under­stand­ing of the cost-
expres­sion of CD19 splice var­i­ants that lack the tar­get epi­tope, effec­tive­ness of off-the-shelf and cus­tom­ized engineered immu-
or muta­tions in the CD19 gene that lead to disrupted mem­brane notherapies is needed.
anchor­age.40-43 Of note, concern for antigen escape/loss with use
of CD19-mAb tafasitamab or CD19 ADC loncastuximab tesirine Conclusions
drives apprehension to utilize these agents prior to CARTs. CARTs have changed the treat­ment land­scape for R/R aggres­
CD20 anti­gen loss has also been described in approx­i­ma­ sive B-cell lym­pho­mas, pro­vid­ing dura­ble responses in patients
tely 25% of patients treated with anti-CD 20 mAbs.44 Possible with his­tor­i­cally poor out­comes. CD20/CD3 BsAbs rep­re­sent a
expla­na­tions include loss through clonal selec­tion, epi­ge­netic prom­is­ing new class of off-the-shelf immu­no­ther­apy that is high-
downregulation, inter­nal­i­za­tion of CD20, or arti­fact due to ritux- ly active and offers the oppor­tu­nity to cir­cum­vent some of the
imab-bound CD20.44,45 It has yet to be deter­mined whether this chal­ lenges faced with the admin­ is­
tra­
tion of CARTs. Although
is a rel­e­vant mech­a­nism of resis­tance to CD20/CD3 BsAbs. expe­ri­ence favors the use of CARTs over other immunotherapies
Several dual-targeting CARTs that con­cur­rently tar­get 2 anti­gens at pres­ent, fur­ther stud­ies and lon­ger-term fol­low-up are needed
and would effec­tively address the chal­lenge of anti­gen escape are to elu­ci­date opti­mal sequenc­ing. Along with ratio­nal com­bi­na­
now in devel­op­ment (Table 2). This includes a CD19/CD20 CART tions, a num­ber of other off-the-shelf immunotherapies, includ­
that has dem­on­strated a high response rate of 82% (CR, 64%) with­ ing novel CARs, are being explored to opti­mize ease of admin­
out added tox­ic­ity.46 Rational com­bi­na­tions of immunotherapies is­tra­tion, safety, and effi­cacy, and they will undoubt­edly lead to
directed at mul­ti­ple anti­gens are also a con­sid­er­ation. mea­sur­able impacts on patient out­comes.

Off-the-shelf immunotherapies vs CARTs: at what cost?


Conflict-of-inter­est dis­clo­sure
Both CARTs and BsAbs have been asso­ci­ated with a high finan­cial
Reem Karmali: speak­ers’ bureau: AstraZeneca, BeiGene, Kite/Gil-
bur­den. The cost of FDA-approved CARTs ranges from approx­i­
ead, Morphosys; con­ sul­
tant: Kite/Gilead, BMS/Celgene/Juno,
ma­tely $373,000 to $410,000 and is even higher when fac­tor­ing
Karyopharm, Janssen/Pharmacyclics, Morphosys, Epizyme; re-
in the price asso­ci­ated with the logis­tics of CART admin­is­tra­tion
search funding: Kite/Gilead, BMS/Celgene/Juno, Takeda.
and the man­age­ment of toxicities. In the TRANSCEND study, rel­
e­vant trial-observed health care resource uti­li­za­tion and costs
were sig­nif­i­cantly greater among patients with grade ≥3 CRS Off-label drug use
and/or ICANS (22.8%).47 These data favor the use of 41BB CARTs, Reem Karmali: mosunetuzumab, epcoritamab, glofitamab, and
which are asso­ci­ated with a low inci­dence of severe CRS/ICANS odronextamab are discussed.
and sup­port the devel­op­ment of safer CART options.
With BsAbs, some of these costs are circumvented, but many Correspondence
over­lap, given the tox­ic­ity pro­file. The price tag for CD20/CD3 Reem Karmali, Northwestern University Feinberg School of Med-
BsAbs has yet to be established. However, if one is to learn any­ icine, 676 N St Clair St, Ste 850, Chicago, IL 60611; e-mail: reem​
thing from the blinatumomab story, these may not be a cheaper ­.karmali@north­west­ern​­.edu.
alter­na­tive.
One also needs to con­sider the social bur­den asso­ci­ated with
References
the admin­is­tra­tion of both CARTs and BsAbs. With CARTs, CRS 1. Zelenetz AD, Gordon LI, Abramson JS, et  al. NCCN guide­lines insights:
and ICANs can have a delayed onset and require care­giver sup­ B-cell lym­pho­mas, ver­sion 3.2019. J Natl Compr Canc Netw. 2019;17(6):
port for the first 1 to 2 months after admin­is­tra­tion.4,7,8 For BsAbs, 650-661.
Prakash Singh Shekhawat
Off-the-shelf vs cus­tom engineered immu­no­ther­apy | 171
2. Crump M, Neelapu SS, Farooq U, et al. Outcomes in refrac­tory dif­fuse large ther­apy for acute lym­pho­blas­tic leu­ke­mia. Cancer Discov. 2016;6(6):664-
B-cell lym­phoma: results from the inter­na­tional SCHOLAR-1 study. Blood. 679.
2017;130(16):1800-1808. 24. Molina JC, Shah NN. CAR T cells bet­ter than BiTEs. Blood Adv. 2021;5(2):602-
3. Gisselbrecht C, Glass B, Mounier N, et al. Salvage reg­i­mens with autol­o­ 606.
gous trans­plan­ta­tion for relapsed large B-cell lym­phoma in the rituximab 25. Topp M, Van Meerten T, Houot R, et al. Earlier ste­roid use with axicabta-
era. J Clin Oncol. 2010;28(27):4184-4190. gene ciloleucel (Axi-Cel) in patients with relapsed/refrac­tory large B cell
4. Neelapu SS, Locke FL, Bartlett NL, et  al. Axicabtagene ciloleucel CAR lym­phoma. Blood. 2019;134(suppl 1):243.
T-cell ther­ apy in refrac­ tory large B-cell lym­ phoma. N Engl J Med. 26. Nastoupil LJ, Jain MD, Feng L, et al. Standard-of-care axicabtagene ciloleu-
2017;377(26):2531-2544. cel for relapsed or refrac­tory large B-cell lym­phoma: results from the US
5. Jacobson C, Locke FL, Ghobadi A, et  al. Long-term sur­vival and grad­ual lym­phoma CAR T con­sor­tium. J Clin Oncol. 2020;38(27):3119-3128.
recov­ery of B cells in patients with refrac­tory large B cell lym­phoma treated 27. Pasquini MC, Hu ZH, Curran K, et  al. Real-world evi­dence of tisagenlec-
with axicabtagene ciloleucel (Axi-Cel). Blood. 2020;136(suppl 1):40-42. leucel for pedi­at­ric acute lym­pho­blas­tic leu­ke­mia and non-Hodgkin lym­
6. Jaeger U, Bishop MR, Salles G, et al. Myc expres­sion and tumor-infil­trat­ing phoma. Blood Adv. 2020;4(21):5414-5424.
T cells are asso­ci­ated with response in patients (Pts) with relapsed/refrac­ 28. Sehgal AR, Hildebrandt G, Ghosh N, et  al. Lisocabtagene maraleucel
tory dif­fuse large B-cell lym­phoma (r/r DLBCL) treated with tisagenlecleu- (liso-cel) for treat­ ment of sec­ ond-line (2L) trans­
plant noneligible (TNE)
cel in the JULIET trial. Blood. 2020;136(suppl 1):48-49. relapsed/refrac­tory (R/R) aggres­sive large B-cell non-Hodgkin lym­phoma

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/164/1851658/164karmali.pdf by guest on 13 December 2021


7. Schuster SJ, Bishop MR, Tam CS, et al; JULIET Investigators. Tisagenlecleu- (NHL): updated results from the PILOT study. J Clin Oncol. 2020;38(suppl
cel in adult relapsed or refrac­tory dif­fuse large B-cell lym­phoma. N Engl J 15):8040.
Med. 2019;380(1):45-56. 29. Diefenbach C, Assouline S, Bosch F, et al. An indi­vid­u­al­ized risk mit­i­ga­tion
8. Abramson JS, Palomba ML, Gordon LI, et  al. Lisocabtagene maraleu- approach for safety: expe­ ri­
ence from the mosunetuzumab (CD20/CD3
cel for patients with relapsed or refrac­ tory large B-cell lym­ pho­mas bispecific anti­body) devel­op­ment pro­gram in rela­tion to neu­ro­tox­ic­ity risk.
(TRANSCEND NHL 001): a multicentre seam­ less design study. Lancet. Blood. 2019;134(suppl 1):4728.
2020;396(10254):839-852. 30. Olszewski AJ, Avigdor A, Babu S, et al. Single-agent mosunetuzumab is a
9. Salles G, Duell J, González Barca E, et al. Tafasitamab plus lenalidomide in prom­is­ing safe and effi­ca­cious che­mo­ther­apy-free reg­i­men for elderly/un-
relapsed or refrac­tory dif­fuse large B-cell lym­phoma (L-MIND): a multicentre, fit patients with pre­vi­ously untreated dif­fuse large B-cell lym­phoma. Blood.
pro­spec­tive, sin­gle-arm, phase 2 study. Lancet Oncol. 2020;21(7):978-988. 2020;136(suppl 1):43-45.
10. Sehn LH, Herrera AF, Flowers CR, et al. Polatuzumab vedotin in relapsed or
31. Matasar MJ, Cheah CY, Yoon DH, et al. Subcutaneous mosunetuzumab in
refrac­tory dif­fuse large B-cell lym­phoma. J Clin Oncol. 2020;38(2):155-165.
relapsed or refrac­tory B-cell lym­phoma: prom­is­ing safety and encour­ag­ing
11. Hamadani M, Radford J, Carlo-Stella C, et al. Final results of a phase 1 study
effi­cacy in dose esca­la­tion cohorts. Blood. 2020;136(suppl 1):45-46.
of loncastuximab tesirine in relapsed/refrac­tory B-cell non-Hodgkin lym­
32. Feucht J, Kayser S, Gorodezki D, et al. T-cell responses against CD19+ pedi­
phoma. Blood. 2020;137(19):2634-2645.
at­ric acute lym­pho­blas­tic leu­ke­mia medi­ated by bispecific T-cell engager
12. Kalakonda N, Maerevoet M, Cavallo F, et  al. Selinexor in patients with
(BiTE) are reg­ u­ lated con­
trarily by PD-L1 and CD80/CD86 on leu­ ke­
mic
relapsed or refrac­tory dif­fuse large B-cell lym­phoma (SADAL): a sin­gle-arm,
blasts. Oncotarget. 2016;7(47):76902-76919.
mul­ ti­
na­tional, multicentre, open-label, phase 2 trial. Lancet Haematol.
33. Phil­lips TJ, Olszewski AJ, Munoz J, et al. Mosunetuzumab, a novel CD20/CD3
2020;7(7):e511-e522.
bispecific anti­body, in com­bi­na­tion with CHOP con­fers high response rates
13. Ansell SM, Minnema MC, Johnson P, et al. Nivolumab for relapsed/refrac­
in patients with dif­fuse large B-cell lym­phoma. Blood. 2020;136(suppl 1):37-
tory dif­fuse large B-cell lym­phoma in patients inel­i­gi­ble for or hav­ing failed
38.
autol­o­gous trans­plan­ta­tion: a sin­gle-arm, phase II study. J Clin Oncol.
34. Locke FL, Ghobadi A, Jacobson CA, et al. Long-term safety and activ­ity of
2019;37(6):481-489.
axicabtagene ciloleucel in refrac­tory large B-cell lym­phoma (ZUMA-1): a
14. Advani R, Flinn I, Popplewell L, et al. CD47 block­ade by Hu5F9-G4 and rit-
sin­gle-arm, multicentre, phase 1-2 trial. Lancet Oncol. 2019;20(1):31-42.
uximab in non-Hodgkin’s lym­phoma. N Engl J Med. 2018;379(18):1711-1721.
35. Neelapu SS, Dickinson M, Ulrickson ML, et  al. Interim anal­y­sis of ZUMA-
15. Schuster SJ, Bartlett NL, Assouline S, et al. Mosunetuzumab induces com­
12: a phase 2 study of axicabtagene ciloleucel (Axi-Cel) as first-line ther­
plete remis­ sions in poor prog­ no­sis non-Hodgkin lym­ phoma patients,
apy in patients (Pts) with high-risk large B cell lym­phoma (LBCL). Blood.
includ­ing those who are resis­tant to or relaps­ing after chi­me­ric anti­gen
2020;136(suppl 1):49.
recep­tor T-cell (CAR-T) ther­a­pies, and is active in treat­ment through mul­ti­
ple lines. Blood. 2019;134(suppl 1):6. 36. Locke FL, Rossi JM, Neelapu SS, et  al. Tumor bur­den, inflam­ma­tion, and
16. Hutchings M, Mous R, Clausen MR, et  al. Subcutaneous epcoritamab prod­uct attri­butes deter­mine out­comes of axicabtagene ciloleucel in large
induces com­plete responses with an encour­ag­ing safety pro­file across B-cell lym­phoma. Blood Adv. 2020;4(19):4898-4911.
relapsed/refrac­ tory B-cell non-Hodgkin lym­ phoma sub­ types, includ­ ing 37. Siddiqi T, Abramson JS, Palomba ML, et al. Correlation of patient char­ac­ter­
patients with prior CAR-T ther­apy: updated dose esca­la­tion data. Blood. is­tics and bio­mark­ers with clin­i­cal out­comes of JCAR017 in R/R aggres­sive
2020;136(suppl 1):45-46. B-NHL (TRANSCEND NHL 001 study). J Clin Oncol. 2018;36(suppl 5):122.
17. Hutchings M, Carlo-Stella C, Bachy E, et  al. Glofitamab step-up dos­ing 38. Basar R, Daher M, Rezvani K. Next-gen­er­a­tion cell ther­a­pies: the emerg­ing
induces high response rates in patients with hard-to-treat refrac­tory or role of CAR-NK cells. Blood Adv. 2020;4(22):5868-5876.
relapsed non-Hodgkin lym­phoma. Blood. 2020;136(suppl 1):46-48. 39. Wang J, Asch AS, Hamad N, et al. A phase 1, open-label study of MGD013, a
18. Hutchings M, Morschhauser F, Iacoboni G, et al. Glofitamab, a novel, biva­ bispecific DART® mol­e­cule bind­ing PD-1 and LAG-3 in patients with relapsed
lent CD20-targeting T-cell-engag­ing bispecific anti­body, induces dura­ble or refrac­tory dif­fuse large B-cell lym­phoma. Blood. 2020;136(suppl 1):21-22.
com­plete remis­sions in relapsed or refrac­tory B-cell lym­phoma: a phase I 40. Abramson JS, Lunning M, Palomba ML. Chimeric anti­gen recep­tor T-cell
trial. J Clin Oncol. 2021;39(18):JCO.20.03175. ther­a­pies for aggres­sive B-cell lym­pho­mas: cur­rent and future state of the
19. Bannerji R, Allan JN, Arnason JE, et al. Odronextamab (REGN1979), a human art. Am Soc Clin Oncol Educ Book. 2019;39:446-453.
CD20 × CD3 bispecific anti­body, induces dura­ble, com­plete responses in 41. Sotillo E, Barrett DM, Black KL, et al. Convergence of acquired muta­tions and
patients with highly refrac­tory B-cell non-Hodgkin lym­phoma, includ­ing alter­na­tive splic­ing of CD19 enables resis­tance to CART-19 immu­no­ther­apy.
patients refrac­tory to CAR T ther­apy. Blood. 2020;136(suppl 1):42-43. Cancer Discov. 2015;5(12):1282-1295.
20. Coyle L, Morley NJ, Rambaldi A, et  al. Open-label, phase 2 study of bli- 42. Braig F, Brandt A, Goebeler M, et al. Resistance to anti-CD19/CD3 BiTE in
natumomab as sec­ond sal­vage ther­apy in adults with relapsed/refrac­ acute lym­pho­blas­tic leu­ke­mia may be medi­ated by disrupted CD19 mem­
tory aggres­ sive B-cell non-Hodgkin lym­ phoma. Leuk Lymphoma. brane traf­fick­ing. Blood. 2017;129(1):100-104.
2020;61(9):2103-2112. 43. Orlando EJ, Han X, Tribouley C, et al. Genetic mech­a­nisms of tar­get anti­
21. Subklewe M. BiTEs bet­ter than CAR T cells. Blood Adv. 2021;5(2):607-612. gen loss in CAR19 ther­apy of acute lym­pho­blas­tic leu­ke­mia. Nat Med.
22. Depil S, Duchateau P, Grupp SA, Mufti G, Poirot L. “Off-the-shelf” allo­ 2018;24(10):1504-1506.
ge­neic CAR T cells: devel­op­ment and chal­lenges. Nat Rev Drug Discov. 44. Hiraga J, Tomita A, Sugimoto T, et al. Down-reg­u­la­tion of CD20 expres­sion
2020;19(3):185-199. in B-cell lym­phoma cells after treat­ment with rituximab-containing com­
23. Teachey DT, Lacey SF, Shaw PA, et al. Identification of pre­dic­tive bio­mark­ bi­na­tion chemotherapies: its prev­a­lence and clin­i­cal sig­nif­i­cance. Blood.
ers for cyto­kine release syn­drome after chi­me­ric anti­gen recep­tor T-cell 2009;113(20):4885-4893.

172  |  Hematology 2021  |  ASH Education Program


45. Schmitz K, Brugger W, Weiss B, Kaiserling E, Kanz L. Clonal selec­tion of 47. Abramson JS, Siddiqi T, Garcia J, et al. Cytokine release syn­drome and neu­
CD20-neg­a­tive non-Hodgkin’s lym­phoma cells after treat­ment with anti- ro­log­i­cal event costs in lisocabtagene maraleucel-treated patients in the
CD20 anti­body rituximab. Br J Haematol. 1999;106(2):571-572. TRANSCEND NHL 001 trial. Blood Adv. 2021;5(6):1695-1705.
46. Shah NN, Johnson BD, Schneider D, et al. Bispecific anti-CD20, anti-CD19
CAR T cells for relapsed B cell malig­nan­cies: a phase 1 dose esca­la­tion and © 2021 by The Amer­i­can Society of Hematology
expan­sion trial. Nat Med. 2020;26(10):1569-1575. DOI 10.1182/hema­tol­ogy.2021000249

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/164/1851658/164karmali.pdf by guest on 13 December 2021

Prakash Singh Shekhawat


Off-the-shelf vs cus­tom engineered immu­no­ther­apy | 173
EMERGING THERAPIES AND CONSIDERATIONS FOR SICKLE CELL DISEASE

Gene therapy for sickle cell disease:


where we are now?

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/174/1851456/174kanter.pdf by guest on 13 December 2021


Julie Kanter1 and Corey Falcon2
1
Department of Medicine, University of Alabama at Birmingham, Birmingham, AL; and 2Department of Pediatrics, Ochsner Hospital for Children,
Jefferson, LA

The landscape of sickle cell disease (SCD) treatment continues to evolve rapidly, with new disease-modifying therapies in
development and potentially curative options on the horizon. Until recently, allogeneic stem cell transplant has been the
only proven cure for SCD. Gene therapy is rising to the forefront of the discussion as a potentially curative or highly disease-
modifying option for abating the complications of the disease. Understanding the different types of gene therapy in use,
the differences in their end points, and their potential risks and benefits will be key to optimizing the long-term use of
this therapy.

LEARNING OBJECTIVES
• Have an improved understanding of the different types of gene therapy
• Be able to have a more thoughtful conversation with patients regarding gene therapy

mostly individuals living in low­resource settings or minor­


CLINICAL CASE ity populations living in high­resource nations. Further, the
A 22­year­old woman with sickle cell anemia (HbSS disease) development of disease­modifying therapies has also been
presents to you, a sickle cell specialist (as a referral from her sluggish, with only 1 medication available until 2017.
community hematologist­oncologist), to discuss the possi­ SCD results from the inheritance of at least 1 copy of
bilities of curative treatment. She has a history of frequent sickle hemoglobin (HbS) and a second copy of a gene
vaso­occlusive (VOC) crises and goes to the emergency encoding HbS or another abnormal hemoglobin. HbS is
department about 5 times per year, where she is admitted produced when the β­globin gene (HBB) contains a single
most of those times. She has never had a stroke (that she E6V missense mutation resulting in the replacement of β6
is aware of). She has been on hydroxyurea (HU), 1000mg glutamic acid by valine.2 On deoxygenation, HbS polymer­
per day, since she was 9, pretty reliably. She takes 30mg izes, leading to abnormally shaped red cells and multiple
of morphine sulfate extended release tablets (MS Contin, downstream clinical sequelae, including hemolysis, vaso­
Purdue) taken by mouth twice a day, 10mg of oxycodone occlusion and subsequent progressive and irreversible
every 4 hours as needed, and folic acid (in addition to HU). organ damage, decreased quality of life, and early death.3
She is on no other medications. She has no full siblings and Although HbS polymerization, vaso­occlusion, and hemo­
does not know her father. She lives with her mother and lytic anemia are central to the pathophysiology of SCD, the
2 half siblings approximately 2 hours from your clinic. resultant pathological events are more consistent with sec­
On exam, she weighs 60 kg, with a normal body mass ondary vascular­endothelial dysfunction and widespread
index, a heart rate of 75, a respiration rate of 20, a blood oxy­ inflammation such that the complications of SCD are better
gen level at 93%, and a blood pressure reading of 118/70. understood as inflammatory vascular disorders.
The source of SCD pathology remains a single­point
mutation resulting in the production of abnormal pro­
Sickle cell disease (SCD) has been well characterized for tein (HbS) and subsequent hemoglobin polymerization
over 100 years, with the first clinical report published in 1910 within the red blood cell. The rate of HbS polymerization
describing it as the “first molecular disease.”1 Despite this is highly variable and depends on multiple factors, includ­
long scientific history, progress toward identifying a cure ing the amount of HbS per erythrocyte, the amount of other
has been slow, likely due in part to the fact that SCD affects nonsickling hemoglobin per erythrocyte, and the overall

174 | Hematology 2021 | ASH Education Program


eryth­ro­cyte hemo­glo­bin level. Thus, suf­fi­cient healthy, nor­mal cess.11,12 Multiple stud­ies have dem­on­strated the effi­cacy of HSCT
adult hemo­glo­bin (HbA) in each cell, as in indi­vid­u­als who carry for SCD, includ­ing donor HSC with HbAS.13
the sickle cell trait (HbAS), will pre­vent poly­mer­i­za­tion and reduce While highly effi­ca­cious, HSCT is not an option for all­indi­vid­
the symp­toms and sequelae of the dis­ease except under rare u­als liv­ing with SCD. The best out­comes reported are in youn­ger
instances of extreme phys­i­o­log­i­cal stress.4,5 Similarly, fetal hemo­ indi­vid­u­als with SCD who have a matched sib­ling donor; unfor­
glo­bin (HbF) has an antisickling effect and can reduce or pos­si­ tu­nately, rel­a­tively few indi­vid­u­als with SCD have such a donor.14
bly elim­i­nate hemo­glo­bin poly­mer­i­za­tion if in suf­fi­cient quan­tity Improvements in haploidentical HSCT for SCD are rap­idly evolv­ing
within the ­eryth­ro­cyte.6 and will sig­nif­i­cantly increase the avail­­able donor pool. However,
Four dif­fer­ent SCD-spe­cific med­i­ca­tions have now been there are still poten­tial draw­backs that can include graft fail­ure,
approved to treat the effects of SCD. HU, the first approved, has delayed immune recon­sti­tu­tion, infer­til­ity, sec­ond­ary malig­nancy,
been proven to decrease the fre­quency of VOC, reduce stroke and graft-ver­sus-host dis­ease (GVHD).15 Further, it is unlikely any
risk in some affected patients, and improve the qual­ity and length allo­ge­neic trans­plant could com­pletely remove the risk of GVHD
of life in many affected indi­vid­u­als.7 However, HU has not been dis­ease or the need for long-term immune sup­pres­sion ther­apy,

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/174/1851456/174kanter.pdf by guest on 13 December 2021


proven as a uni­ver­sally accept­able dis­ease-mod­i­fy­ing ther­apy both of which carry their own risks of sub­se­quent com­pli­ca­tions.
because many indi­vid­u­als con­tinue to develop sig­nif­i­cant dis­ease- Thus, there remains a dem­on­strated need for other means of pro­
related com­pli­ca­tions or are unwill­ing to main­tain drug adher­ vid­ing gene trans­fer into HSCs with­out the same immu­no­logic
ence due to con­cerns with side effects, fer­til­ity, or tol­er­ance. risks. In this sce­nario, trans­plan­ta­tion of genet­i­cally mod­i­fied
L-glu­ta­mine, approved in 2017 to treat SCD, has anti­ox­i­dant prop­ autol­o­gous HSCs pro­vi­des a poten­tial alter­na­tive ther­apy.
er­ties that have been shown to ame­lio­rate SCD and decrease the
fre­quency of VOC.8 Long-term dura­bil­ity and util­ity have yet to be
dem­on­strated. The newest med­i­ca­tions, crizanlizumab and vox­
elotor, have proven effi­ca­cious in their own right by decreas­ing CLINICAL CASE (Con­tin­ued)
VOCs and decreas­ing hemo­ly­sis, respec­tively, but nei­ther med­ You dis­cuss poten­tially cura­tive options with your patient that
i­ca­tion is likely to have a demon­stra­ble cura­tive effect, and both include haploidentical HSCT and gene ther­ apy, as you have
require ongo­ing, life­long treat­ment.9,10 Further, these med­i­ca­tions already deter­mined that she does not have any full sib­lings. Her
are not yet proven to reduce, delay, or sta­bi­lize organ-spe­cific mother works full time and has a his­tory of lupus that is on active
com­pli­ca­tions such as renal dis­ease, avas­cu­lar necro­sis, or pul­mo­ treat­ment (and so can­not donate HSCs). She is inter­ested in con­
nary hyper­ten­sion. sid­er­ing haploidentical trans­plant from a sib­ling but is wor­ried
about the dis­tance from home (weekly appoint­ments to mod­u­
late her immune sup­pres­sion, the need to stay in close prox­im­ity
CLINICAL CASE (Con­t in­u ed) after trans­plant, a con­cern for GVHD and sub­se­quent rehospital­
ization). Using shared deci­sion-mak­ing, it seems she may be best
You review her cur­rent dis­ease sta­tus, com­pli­ca­tions, and con­
suited for gene ther­apy, as there is no risk of GVHD and no need
cerns and spe­ cif­i­
cally dis­
cuss the impor­ tance of doing a full
to mon­i­tor immune sup­pres­sion med­i­ca­tion. She asks about the
screen­ing assess­ment of all­organ sys­tems to eval­u­ate the SCD-
dif­fer­ent types of gene ther­a­pies avail­­able.
related com­pli­ca­tions. You per­form a full set of labs, an eval­u­a­tion
for iron over­load (fer­ri­tin, liver mag­netic res­o­nance imag­ing), an
echo­car­dio­gram (due to low oxy­gen at base­line), a workup for
Broadly speak­ing, 4 main types of gene ther­apy are avail­­able for
alloimmunization, and a kid­ney screen (albu­min/cre­at­i­nine ratio).
the treat­ment of SCD. These include gene addi­tion ther­apy, gene
Her lab results are con­sis­tent with HU adher­ence, and the echo­
editing, gene silenc­ing, and gene cor­rec­tion ther­apy (Figure 1).
car­dio­gram shows triv­ial tri­cus­pid regur­gi­ta­tion and mild left
Each type of ther­apy dif­fers in the means by which it induces the
ven­tric­u­lar hyper­tro­phy but is oth­er­wise nor­mal. Labs show that
replace­ment of HbS with nonsickling hemo­glo­bin. The nonsick­
fer­ri­tin is 1200 µg/L, liver iron con­cen­tra­tion is 5.1 mg/g, and there
ling hemo­glo­bin is the tar­get pro­tein in each ther­apy and will
is no con­cern for sig­nif­i­cant alloimmunization. Unfortunately, her
need to be eval­u­ated using mul­ti­ple novel meth­ods and terms.
renal assess­ment shows marked pro­tein­uria.
You dis­ cuss treat­ ment options with the patient, includ­ ing 1. Gene addi­tion ther­apy is the addi­tion of a new gene using a
poten­ tially increas­ ing HU to the max­ i­
mum tol­ er­
ated dose or viral vec­tor (usu­ally) to deliver a nonsickling glo­bin gene to
adding addi­tional med­i­cal ther­a­pies. In frus­tra­tion, how­ever, she the stem cells. In this pro­ce­dure the native HbS gene is not
says, “I want to be cured of this wretched dis­ease!” altered, resulting in the pro­duc­tion of both the new hemo­
glo­bin and the native HbS. Several exam­ples of gene addi­tion
ther­apy are ongo­ing that use a lentiviral vec­tor (LVV) to house
One method by which to cor­rect the path­o­phys­i­o­log­i­cal and deliver a new gene.16
abnor­mal­ity in SCD is to replace the abnor­mal HbS with more 2. Gene editing is most often used to describe a pro­cess of gene
func­tional HbA. The clin­i­cal ben­e­fit of this molec­u­lar replace­ dis­rup­tion in the con­text of SCD and can be used to tar­get sup­
ment has been clearly dem­on­strated by the suc­cess of hema­ pres­sors of HbF as a way to both increase HbF and decrease
to­poi­etic stem cell trans­plant (HSCT) for SCD. In this sce­nario, HbS. Elements of DNA within a gene can be targeted using a
new hema­to­poi­etic stem cells (HSCs) are used as the vehi­cle guide that can iden­tify and tightly bind to the tar­get with high
to deliver healthy hemo­glo­bin and there­fore elim­i­nate eryth­ro­ spec­i­fic­ity cou­pled with an enzyme to cut the DNA, induc­ing
cyte sick­ling and the sec­ond­ary effects of this path­o­log­i­cal pro­ dou­ble-stranded breaks. The spe­cific DNA cut allows one to

Prakash Singh Shekhawat


Gene ther­apy for sickle cell dis­ease  |  175
change the sequence with high pre­ci­sion, usu­ally resulting in an
inser­tion and dele­tion. This type of gene ther­apy most often tar­
gets a dif­fer­ent part of DNA (sep­a­rate from the HbS muta­tion)
to pro­duce an increase in HbF pro­duc­tion while recip­ro­cally
suppressing HbS pro­duc­tion.16 Specifically, many of the cur­rent
ther­a­pies tar­get the BCL11A gene, a neg­a­tive reg­u­la­tor of HbF.
In this exam­ple, the gene editing is used to turn off the reg­u­la­
tion of HbF in order to increase HbF pro­duc­tion.
3. Gene silenc­ing uses the reg­u­la­tion of gene expres­sion in a
cell to pre­vent the expres­sion and resul­tant pro­duc­tion of
cer­tain pro­teins. Similar to gene editing, this type of ther­apy
is being used to sup­press the BC11A gene, resulting in an in­
crease in HbF while recip­ro­cally suppressing HbS pro­duc­tion.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/174/1851456/174kanter.pdf by guest on 13 December 2021


In con­trast to gene editing, this type of gene ther­apy has thus
far relied upon viral vec­tor deliv­ery (sim­i­lar to gene addi­tion)
to deliver an anti­sense to mes­sen­ger RNA to sup­press the
gene prod­uct instead of cut­ting the gene.17
4. Gene cor­rec­tion can be performed in sev­eral dif­fer­ent ways.
In most cases, how­ever, a guide RNA is used to iden­tify the
tar­get muta­tion for cut­ting, and then editing occurs with the
simul­ ta­
neous deliv­ ery of tem­ plate DNA of the cor­ rect se­
quence, directing homol­ogy-directed repair (HDR; Figure 2).
Though this is cur­rently the least effi­cient method, efforts are
under­way to improve gene cor­rec­tion, includ­ing through the Figure 1. Cartoon rendering of different gene therapies. (A) Gene
inser­tion of DNA, direct base editing, and prime editing. This editing, (B) gene correction, (C) gene silencing, and (D) gene
is the only type of gene ther­apy that cur­rently aims to elim­ addition via viral vector.
i­nate HbS pro­duc­tion and intro­duce a nonsickling hemo­glo­
bin simul­ta­neously.18 Therapeutics and Vertex Pharmaceuticals and inves­ ti­
ga­
tors
At pres­ent, all­types of gene ther­apy use the same over­all from Bos­ton C ­ hildren’s Hospital have also presented data on
pro­ce­dure. For each study listed, patients first undergo inten­sive their CLIMB and short hair­ pin RNA (shRNA) stud­ ies, respec­
screen­ing. It is impor­tant to note that the cri­te­ria for each study tively, show­ing that the potent antisickling prop­er­ties of HbF
dif­fer slightly, but all­require the indi­vid­ual to have had sig­nif­i­cant com­bined with the lower lev­els of HbS result in the ­res­o­lu­tion
SCD-related com­pli­ca­tions (as a rea­son for under­go­ing a study of VOCs as well. These gene editing and silenc­ing stud­ies, how­
pro­ce­dure) and to have suf­fic ­ ient organ func­tion to undergo ever, are even ear­lier in their reporting and long-term out­comes.
the che­mo­ther­apy prep­a­ra­tion required. Once fully screened, Gene cor­rec­tion ther­apy will be the next type of gene ther­
patients need to undergo stem cell col­lec­tion using plerixafor apy to enter the clin­i­cal space. Gene cor­rec­tion ther­apy includes
mobi­li­za­tion and apher­e­sis. This pro­cess may be under­taken a com­bi­na­tion of gene editing and gene addi­tion. The CEDAR
more than once to ensure suf­fi­cient stem cells are col­lected study, which received US Food and Drug Administration clear­
for manip­u­la­tion as well as for backup. Once the stem cells are ance to move for­ward to a phase 1 clin­i­cal trial in early 2021, will
appro­pri­ately altered, all­types of gene ther­apy uti­lize che­mo­ con­cur­rently use a high-fidelity clus­tered reg­u­larly interspaced
ther­apy (to make room for altered/manip­u­lated stem cells). In all­ short pal­in­dromic repeats (CRISPR)-Cas9 ribo­nu­cleo­pro­tein (RNP)
but 1 trial below (MOMENTUM), these stud­ies use busul­fan che­ to induce a dou­ble-strand DNA break­age and HDR with a nonin­
mo­ther­apy to pro­vide myeloablation to ensure opti­mal stem cell tegrating adeno-asso­ci­ated virus-6 donor DNA repair tem­plate
engraft­ment. The MOMENTUM trial uses a “reduced-inten­sity” to pro­duce a new gene prod­uct. Unlike the gene-editing ther­
che­mo­ther­apy reg­i­men using mel­pha­lan. a­pies that rely on non­ho­mol­o­gous end join­ing and sub­se­quent
Current and upcom­ing clin­ic ­ al tri­als using gene ther­apy are inser­tion and dele­tion for­ma­tion in the edited space, gene cor­
detailed in Table 1. Note that some of the tri­als listed tar­get rec­tion ther­a­pies rely on the more com­pli­cated and his­tor­i­cally
the BCL11A gene (its ery­throid enhancer of mes­sen­ger RNA), a less effi­cient HDR.21
repres­sor of γ-glo­bin expres­sion, in order to induce HbF pro­duc­
tion in adult eryth­ro­cytes, while oth­ers use ran­dom viral vec­tor
inser­tion to result in HbA pro­duc­tion, and 1 study, nota­bly, tar­
CLINICAL CASE (Con­tin­ued)
gets the sickle cell gene muta­tion itself.17-20 You and your res­i­dent leave the room to allow the patient and
Current clin­i­cal tri­als of lentiviral gene ther­apy based on the her fam­ily time to dis­cuss and digest all­of this infor­ma­tion. Your
addi­tion of a mod­i­fied β-glo­bin gene (HbAT87Q ) have accu­ res­id
­ ent astutely asks how it will be pos­si­ble to com­pare out­
mu­lated the most data so far and have dem­on­strated a ben­ comes and mea­sure effi­cacy in tri­als that use dif­fer­ent meth­
e­fit in the reduc­tion of sig­nif­i­cant VOCs in SCD. However, the ods of hemo­glo­bin induc­tion and dif­fer­ent types of hemo­glo­bin
data remain early, and results regard­ing improve­ments in long- pro­duc­tion.
term dura­bil­ity and organ func­tion are forth­com­ing. CRISPR

176  |  Hematology 2021  |  ASH Education Program


Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/174/1851456/174kanter.pdf by guest on 13 December 2021
Figure 2. CRISPR-Cas-9-induced double-stranded break and its sequential repair pathways. Left: nonhomologous end joining. Right:
HDR, which requires the insertion of a homologous DNA strand used as a template for a high-fidelity double-stranded DNA break.
PAM, protospacer adjacent motif; sgRNA, single-guide RNA.

Table 1. Current and upcom­ing stud­ies of gene ther­apy in SCD

Genetic silenc­ing
Study name LentiGlobin DREPAGLOBE CLIMB PRECIZN-1 of BCL11A MOMENTUM CEDAR
Type of gene ther­apy Gene addi­tion Gene addi­tion Gene editing Gene editing Gene silenc­ing Gene addi­tion Gene cor­rec­tion
Editing tool NA NA CRISPR-Cas9 RNP Zinc fin­ger ShRNA NA HiFi CRISPR-
Cas9 RNP
Type of stem cell Transduction Transduction Electroporation Transfection Transduction Transduction Electroporation
manip­u­la­tion with zinc fin­
ger nucle­ase
mRNA
Vector (y/n) BB305 LVV DROBE 1 LVV None None BCH-BB694 LVV γG16D LVV Nonintegrating
that encodes AAV6 donor
a microRNA- DNA repair
adapted shRNA tem­plate
Genetic tar­get (y/n) NA NA Erythroid lin­e­age- 11A (BCL11A) BCL11A mRNA N/a Sickle muta­tion
spe­cific locus (aden­o­sine—
enhancer of the (ery­throid > thy­mine
BCL11A gene enhancer) [A— > T]
Drug prod­uct LentiGlobin DREPAGLOBE CTX001 BIVV003 BCH-BB694 ARU-180126 GPH101
BB305
Protein prod­uct HbAT87Q βAS3, an antisickling HbF HbF HbF HbFG16D HbA
β-glo­bin pro­tein
(AS3) containing
3 amino acid
sub­sti­tu­tions in
the wild-type HBB
Prakash Singh Shekhawat
Gene ther­apy for sickle cell dis­ease  |  177
Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/174/1851456/174kanter.pdf by guest on 13 December 2021
Figure 3. Gene therapy process and evaluation measures. DP, drug product; VCN, vector copy number.

There are sev­eral ways to eval­ua ­ te effi­cacy in gene ther­apy has read about a patient who got leu­ke­mia after early-stage
for SCD. At the end of the pro­cess, the most impor­tant ques­ gene ther­apy and wants to know the risks of gene ther­apy.
tion is how much of the new hemo­glo­bin (pro­tein prod­uct of the
gene ther­apy) is being pro­duced over time. However, through­
out the pro­cess are many steps in which interim eval­u­a­tions of There are real and poten­tial risks involved with gene ther­apy
effi­cacy are use­ful. These steps and their eval­u­a­tion tech­niques of all­types. The che­mo­ther­apy used in myeloablation car­ries a
are detailed in Figure 3. high risk of infer­til­ity (nearly 100%) and also results in mucositis,
As noted, it is most impor­tant to deter­mine how much non­ nau­sea, loss of appe­tite, alo­pe­cia, and other usu­ally revers­ible
sickling hemo­glo­bin is in each red blood cell and how much of com­pli­ca­tions. Infertility is a major source of con­cern that must
it is a prod­uct of the gene ther­apy vs the myeloablation (which be addressed. While fer­til­ity pres­er­va­tion is pos­si­ble for some
can result in stress eryth­ ro­
poi­e­
sis that causes HbF pro­ duc­ indi­vid­u­als, it is nei­ther uni­ver­sally avail­­able nor effi­ca­cious. It
tion). This can be eval­u­ated by the trans­duc­tion effi­ciency, or is impor­tant to have patients meet with fer­til­ity spe­cial­ists to
the per­cent­age of blood stem cells hav­ing incor­po­rated the review the avail­­able options and their asso­ci­ated risks and ben­
desired genetic mate­rial. Longitudinal stud­ies are needed to e­fits. Secondary malig­nancy is another major risk of gene ther­
deter­mine the dura­bil­ity of gene ther­apy. Last, it will be impor­ apy. Chemotherapy such as busul­ fan car­ ries an inde­ pen­dent
tant to learn over time which symp­toms and/or com­pli­ca­tions long-term risk of sec­ ond­ ary malig­ nancy in patients under­ go­
of SCD improve with gene ther­apy and if the out­comes dif­fer ing both allo­ge­neic and autol­o­gous trans­plant. Another poten­
depending on the type and amount of hemo­glo­bin prod­uct. tial cause of a sec­ond­ary malig­nancy is the trans­plan­ta­tion of
At this time it is not clear what per­cent­age of stem cells must poten­tially dam­aged HSCs. Individuals with SCD are affected by
receive genetic cor­rec­tions to result in suf­fic ­ ient nonsickling chronic inflam­ma­tion and endo­the­lial dam­age as well as hyp­
hemo­glo­bin. Additional fol­low-up assess­ments need to include oxic bone infarc­tion and con­stant eryth­ro­poi­etic stress.22 These
both lab­ o­ra­tory and rheologic mea­ sures of hemo­ ly­
sis and man­i­fes­ta­tions of SCD likely dam­age the HSCs and may result
adhe­sion in addi­tion to in-depth patient-reported out­comes. in a pre­dis­po­si­tion to malig­nant trans­for­ma­tion. At this point it
Finally, it is most impor­tant to assess whether gene ther­apy is unclear how high this risk is or if it can be suit­ably miti­gated
can pre­vent vaso-occlu­sion (which types and to what extent) with changes already in use in gene ther­apy pro­to­cols or future
and can either sta­bi­lize or resolve organ com­pli­ca­tions due to changes to come. Two patients in the ini­tial LentiGlobin HGB-
SCD. These find­ings remain unclear at this time but are highly 206 trial devel­oped acute mye­log­e­nous leu­ke­mia at 3 and
nec­es­sary in advanc­ing out­comes in SCD. For exam­ple, up to 5 years post autol­o­gous gene ther­apy.23 Currently, the workup
10% of per­sons with sickle cell ane­mia may develop end-stage sug­gests that the vec­tor is not asso­ci­ated with the malig­nancy
renal dis­ease. At this time it is unclear whether even allo­ge­neic and that per­haps there is an inher­ent increased risk in those with
trans­ plant can pre­ vent the devel­ op­ ment of end-stage renal SCD wors­ened by low cell dose, low vec­tor copy num­ber, and
dis­ease once some­one has devel­oped chronic kid­ney dis­ease; a return to the SCD phe­no­type of high eryth­ro­poi­etic stress.
gene ther­apy results are fur­ther behind. While data regard­ing These risks may have been miti­gated by the use of a plerixafor-
out­comes for VOC appear clearer, the long-term organ-spe­cific medi­ated stem cell har­vest (in place of bone mar­row col­lec­tion)
response to gene ther­apy will truly mea­sure its effi­cacy. and precollection trans­fu­sion ther­apy; how­ever, the degree to
which this will reduce the long-term risk is unknown.
Regarding gene addi­tion, the major con­cern is the poten­tial
risk of an inser­tion at a pro­moter site that causes unwanted cel­lu­
CLINICAL CASE (Con­t in­u ed) lar pro­lif­er­a­tion or malig­nant trans­for­ma­tion. This issue occurred
You and the res­i­dent return to the patient and her fam­ily. She recently when a patient with cere­bral adre­nal leu­ko­dys­tro­phy
does not have chil­dren but does want to have a fam­ily later in devel­oped myelodysplastic syn­drome after receiv­ing gene ther­
life. She is wor­ried about the poten­tial for infer­til­ity. Further, she apy. The viral vec­tor, Lenti-D (Bluebird Bio), dif­fers from those

178  |  Hematology 2021  |  ASH Education Program


used in SCD-related gene ther­apy but is sim­i­larly designed to add funding: National Institutes of Health, National Heart, Lung, and
func­tional cop­ies of a gene into a stem cell. However, based on Blood Institute, Health Resources and Services Administration,
the loca­tion of the LVV inser­tion, there is con­cern that it caused Centers for Disease Control and Prevention.
the myelodysplastic syn­ drome (press report, Bluebird Bio Corey Falcon: no com­pet­ing finan­cial inter­ests to declare.
Aug 13, 2021). This has not yet been observed (in any LVV-based
gene ther­apy for SCD) but remains a poten­tial risk. Gene editing Off-label drug use
can also result in unin­tended mod­i­fi­ca­tions at other points along Julie Kanter: nothing to disclose.
the genome out­side of the targeted DNA sequence (off-tar­get Corey Falcon: nothing to disclose.
effects). Current tech­nol­ogy allows us to iden­tify off-tar­get muta­
tions that occur at a high fre­quency, but it is pos­si­ble that the
Correspondence
more rare ones could (the­o­ret­i­cally) escape detec­tion and give
Julie Kanter, University of Alabama at Birmingham, 1720 2nd St
cells growth or sur­vival advan­tages that pro­mote can­cer. Further,
South, Birmingham, AL 35294; e-mail: jkanter@uabmc​­.edu.
it is clear that when electroporation is used there is decreased

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/174/1851456/174kanter.pdf by guest on 13 December 2021


stem cell sur­vival24; it is unclear if this will result in any addi­tional
References
neg­a­tive out­comes. 1. Pauling L. On the sta­bil­ity of the S(8) mol­e­cule and the struc­ture of fibrous
sul­fur. Proc Natl Acad Sci USA. 1949;35(9):495-499.
2. Noguchi CT, Torchia DA, Schechter AN. Determination of deoxyhemoglo­
bin S poly­mer in sickle eryth­ro­cytes upon deox­y­gen­ation. Proc Natl Acad
Sci USA. 1980;77(9):5487-5491.
CLINICAL CASE (Con­t in­u ed) 3. Bunn HF. Pathogenesis and treat­ment of sickle cell dis­ease. N Engl J Med.
1997;337(11):762-769.
Your patient returns the fol­low­ing week with her fam­ily. After 4. Heller P, Best WR, Nelson RB, Becktel J. Clinical impli­ca­tions of sickle-cell
sev­eral con­ver­sa­tions with you (her phy­si­cian), her fam­ily, and trait and glu­cose-6-phos­phate dehy­dro­ge­nase defi­ciency in hos­pi­tal­ized
her pas­tor, she decides she under­stands the risks and ben­e­fits black male patients. N Engl J Med. 1979;300(18):1001-1005.
5. Platt OS, Thorington BD, Brambilla DJ, et al. Pain in sickle cell dis­ease: rates
of gene ther­apy and wants to move for­ward. Your res­i­dent asks and risk fac­tors. N Engl J Med. 1991;325(1):11-16.
why it took 3 weeks for her to reach this con­clu­sion. 6. Ware RE, Davis BR, Schultz WH, et al. Hydroxycarbamide ver­sus chronic
trans­fu­sion for main­te­nance of transcranial doppler flow veloc­i­ties in chil­dren
with sickle cell anae­mia-TCD with trans­fu­sions chang­ing to hydroxy­urea
(TWiTCH): a multicentre, open-label, phase 3, non-infe­ri­or­ity trial. Lancet.
A sickle cell expert and team famil­iar with all­of the ther­a­ 2016;387(10019):661-670.
7. Niihara Y, Miller ST, Kanter J, et al; Investigators of the Phase 3 Trial of
peu­tic options for the treat­ment of SCD should be required to l-Glutamine in Sickle Cell Disease. A phase 3 trial of l-glu­ta­mine in sickle cell
help an indi­vid­ual with SCD make a sig­nif­i­cant treat­ment deci­ dis­ease. N Engl J Med. 2018;379(3):226-235.
sion such as gene ther­apy. The patient should be made aware 8. Ataga KI, Kutlar A, Kanter J. Crizanlizumab in sickle cell dis­ease. N Engl
of the real­is­tic expec­ta­tions and poten­tial risks of gene ther­ J Med. 2017;376(18):1796.
9. Vichinsky E, Hoppe CC, Ataga KI, et al; HOPE Trial Investigators. A phase
apy, with an empha­sis on the impor­tance of shared deci­sion-
3 ran­dom­ized trial of voxelotor in sickle cell dis­ease. N Engl J Med. 2019;​
mak­ing between the patient, fam­ily, and phy­si­cian.25 Ample 381(6):​509-519.
time detailing the known and unknown aspects of gene ther­ 10. Bernaudin F, Souillet G, Vannier JP, et al. Report of the French expe­ri­ence
apy with the patient and their fam­ily will facil­i­tate mean­ing­ concerning 26 chil­dren transplanted for severe sickle cell dis­ease. Bone
ful dis­cus­sion and dia­logue. It is also impor­tant to real­ize that Marrow Transplant. 1997;19(suppl 2):112-115.
11. Walters MC, Patience M, Leisenring W, et al. Bone mar­row trans­plan­ta­tion
both ben­e­fits and risks are inher­ent in new pro­ce­dures, but the for sickle cell dis­ease. N Engl J Med. 1996;335(6):369-376.
poten­tial for cure is also avail­­able. Finally, gene ther­apy still 12. Al-Khabori M, Al-Ghafri F, Al-Kindi S, et al. Safety of stem cell mobi­li­za­tion in
requires myeloablation, which may limit uptake and par­tic­i­pa­ donors with sickle cell trait. Bone Marrow Transplant. 2015;50(2):310-311.
tion based on both the risks of infer­til­ity as well as any base­line 13. Eapen M, Brazauskas R, Walters MC, et al. Effect of donor type and con­
di­tion­ing reg­i­men inten­sity on allo­ge­neic trans­plan­ta­tion out­comes in
organ dys­func­tion that would make a poten­tial patient inel­i­gi­
patients with sickle cell dis­ease: a ret­ro­spec­tive multicentre, cohort study.
ble. Thus, thor­ough delib­er­a­tion should be under­taken before Lancet Haematol. 2019;6(11):e585-e596.
screen­ing is ini­ti­ated. 14. Tanhehco YC, Bhatia M. Hematopoietic stem cell trans­plan­ta­tion and cel­
Recent data from gene ther­a­pies in prog­ress dem­on­strate lu­lar ther­apy in sickle cell dis­ease: where are we now? Curr Opin Hematol.
the need for the long-term fol­low-up and con­sis­tent col­lec­tion of 2019;26(6):448-452.
15. Magrin E, Miccio A, Cavazzana M. Lentiviral and genome-editing strat­e­gies
data using com­mon data ele­ments to ensure that out­comes can for the treat­ment of β-hemo­glo­bin­op­a­thies. Blood. 2019;134(15):1203-1213.
be com­pared across tri­als as well as to the nat­u­ral his­tory of SCD. 16. Esrick EB, Lehmann LE, Biffi A, et al. Post-tran­scrip­tional genetic silenc­ing
Additional data are needed regard­ing the use of gene ther­apy of BCL11A to treat sickle cell dis­ease. N Engl J Med. 2021;384(3):205-215.
for the spe­cific reme­di­a­tion of organ-related com­pli­ca­tions in 17. Wilkinson AC, Dever DP, Baik R, et al. Cas9-AAV6 gene cor­rec­tion of beta-
glo­bin in autol­o­gous HSCs improves sickle cell dis­ease eryth­ro­poi­e­sis in
SCD as well as in patients whose pri­mary presenting symp­tom is
mice. Nat Commun. 2021;12(1):686.
severe, chronic pain. Finally, enhanced SCD sur­veil­lance and lon­ 18. Frangoul H, Altshuler D, Cappellini MD, et al. CRISPR-Cas9 gene editing for
gi­tu­di­nal stud­ies are needed to bet­ter under­stand, quan­ti­tate, sickle cell dis­ease and β-thal­as­se­mia. N Engl J Med. 2021;384(3):252-260.
and com­pare the poten­tial for malig­nancy in this pop­u­la­tion. 19. Kanter J, Thompson AA, Mapara MY, et al. Updated results from the Hgb-
206 study in patients with severe sickle cell dis­ ease treated under a
revised pro­to­col with lentiglobin gene ther­apy using plerixafor-mobilised
Conflict-of-inter­est dis­clo­sure haematopoietic stem cells. HemaSphere. 2019;3(suppl 1):754-755.
Julie Kanter: hon­o­raria: Novartis, Graphite Bio, Forma Therapeu­ 20. Li Y, Maule J, Neff JL, et al. Myeloid neo­plasms in the set­ting of sickle cell
tics, Agios, Beam Therapeutics, Guidepoint Global, GLG; research dis­ease: an intrin­sic asso­ci­a­tion with the under­ly­ing con­di­tion rather than

Prakash Singh Shekhawat


Gene ther­apy for sickle cell dis­ease  |  179
a coin­ci­dence: report of 4 cases and review of the lit­er­a­ture. Mod Pathol. 25. Grimley M, Asnani M, Shrestha A et  al. Early results from a phase 1/2
2019;32(12):1712-1726. study of Aru-1801 gene ther­apy for sickle cell dis­ease (SCD): manufactur­
21. Leonard A, Tisdale JF. A pause in gene ther­apy: reflecting on the unique ing pro­cess enhance­ments improve effi­cacy of a mod­i­fied gamma glo­bin
chal­lenges of sickle cell dis­ease. Mol Ther. 2021;29(4):1355-1356. len­ti­vi­rus vec­tor and reduced inten­sity con­di­tion­ing trans­plant. Blood.
22. Métais JY, Doerfler PA, Mayuranathan T, et al. Genome editing of HBG1 and 2020;136(suppl 1):20-21.
HBG2 to induce fetal hemo­glo­bin. Blood Adv. 2019;3(21):3379-3392.
23. Sullivan KM, Horwitz M, Osunkwo I, Shah N, Strouse JJ. Shared deci­sion-
mak­ing in hema­to­poi­etic stem cell trans­plan­ta­tion for sickle cell dis­ease.
Biol Blood Marrow Transplant. 2018;24(5):883-884.
24. Naik RP, Smith-Whitley K, Hassell KL, et al. Clinical out­comes asso­ci­ated
with sickle cell trait: a sys­tem­atic review. Ann Intern Med. 2018;169(9): © 2021 by The Amer­i­can Society of Hematology
619-627. DOI 10.1182/hema­tol­ogy.2021000250

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/174/1851456/174kanter.pdf by guest on 13 December 2021

180  |  Hematology 2021  |  ASH Education Program


EMERGING THERAPIES AND CONSIDERATIONS FOR SICKLE CELL DISEASE

Hematopoietic cell transplantation for sickle cell


disease: updates and future directions

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/181/1851576/181krishnamurti.pdf by guest on 13 December 2021


Lakshmanan Krishnamurti
Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Emory University, Atlanta, GA

Excellent outcomes in hematopoietic cell transplantation (HCT) from HLA-identical siblings, improvements in condition-
ing regimens, novel graft-versus-host disease prophylaxis, and the availability of alternative donors have all contributed
to the increased applicability and acceptability of HCT for sickle cell disease (SCD). In young children with symptomatic
SCD with an available HLA-identical related donor, HCT should be carefully considered. HCT from alternative donors is
typically undertaken only in patients with severe symptoms, causing or likely to cause organ damage, and in the context
of clinical trials. Patients undergoing HCT for SCD require careful counseling and preparation. They require careful moni-
toring of unique organ toxicities and complications during HCT. Patients must be prospectively followed for a prolonged
time to determine the long-term outcomes and late effects of HCT for SCD. Thus, there is a need for a universal, longi-
tudinal clinical registry to follow patients after HCT for SCD in conjunction with individuals who do not receive HCT to
compare outcomes. Antibody-based conditioning and ex-vivo umbilical cord blood expansion are likely to improve the
availability and acceptability of HCT. In addition, new disease-modifying drugs and the emerging option of the autol-
ogous transplantation of gene-modified hematopoietic progenitor cells are likely to expand the available therapeutic
options and make decision-making by patients, physicians, and caregivers even more complicated. Future efforts must
also focus on determining the impact of socioeconomic status on access to and outcomes of HCT and the long-term
impact of HCT on patients, families, and society.

LEARNING OBJECTIVES
• Discuss indications, conditioning, donor options, timing, outcomes, and decision­making in HCT for SCD
• Understand the impact of recipient ages and the availability of HLA­identical donors on outcomes of HCT for SCD
• Review emerging options in alternate­donor HCT for SCD.

L­glutamine has been shown to reduce the rate of VOE


CLINICAL CASE and related hospitalizations.2 Voxelotor has been shown
A 12­year­old girl with HbSS­type sickle cell disease (SCD) to increase the mean hemoglobin (Hb) level from baseline
has been having recurrent episodes of vaso­occlusive pain compared with placebo and may be particularly useful in
(VOE). She has been on hydroxyurea (HU) since the age of individuals who have continued anemia and hemolysis.3
9 months. Her clinical course remains severe despite an ade­ Crizanlizumab has been demonstrated to reduce the fre­
quate trial of L­glutamine 3 years ago and, more recently, quency of VOE.4 HCT, however, remains the only treatment
of voxelotor. She has an 8­year­old HLA­identical sibling. with curative intent. When performed in young patients
The patient’s pediatric hematologist inquires whether she from HLA­identical related donors, HCT results in excel­
should talk to this family about considering hematopoietic lent overall survival (OS) and event­free survival (EFS).5­11
cell transplantation (HCT). However, the lack of an available HLA­identical family
donor remains a significant limitation in the applicability of
matched sibling donor HCT. The optimization of support­
Introduction ive care, the development of novel conditioning regimens,
Comprehensive care and disease modification of SCD with and the availability of the options of HCT from alternative
HU can decrease morbidity and organ dysfunction and donors have enhanced the applicability of HCT for SCD.
improve health­related quality of life (QoL) and survival.1 However, it is a sobering fact that, despite its increase in
Prakash Singh Shekhawat
HCT for SCD | 181
Table 1. Risk score based on age and type of donor.

3-year prob­a­bil­ity/
inci­dence % (95% CI)
Death
Age, with­out
years Age score Type of donor Donor score Total score EFS GF Graft fail­ure ≤
≤12 0 HLA-matched sib­ling 0 0 92 (89-94)     2 (0-4) 6       (4-9)
0 HLA-matched rel­a­tive 2 2 62 (43-76)     8 (2-19) 30        (15-47)
0 Matched unre­lated donor 1 1 83 (69-91)     8 (2-18) 8 (2-18)
0 Mismatched unre­lated donor 2 2 68 (55-79)     5 (1-13) 27 (16-38)
≥13 1 HLA-matched sib­ling 0 1 87  (81-92)     7 (4-11) 5 (2-10)

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/181/1851576/181krishnamurti.pdf by guest on 13 December 2021


1 HLA mismatched rel­a­tive 2 3 52 (38-65)    10   (4-18) 38 (24-51)
1 Matched unre­lated donor 1 2 50    (34-64) 29 (17-43)  21   (10-33)
1 Mismatched unre­lated donor 2 3 49 (31-66) 23 (9-40) 28 (31-44)
GF, graft fail­ure. Reproduced with per­mis­sion from Brazauskas et al.12

the last decade,11 HCT has been applied to only a tiny frac­tion of patients and fre­quent VOE in 23% of patients.17 More recently,
patients with SCD, includ­ing those with severe dis­ease man­i­fes­ the most com­mon indi­ca­tion for HCT has been recur­rent VOE in
ta­tions. This arti­cle aims to review the cur­rent sta­tus of HCT for over 70% of cases.9,18-20
SCD and address future direc­tions in the field. The rea­sons for the shift in indi­ca­tions for HCT are unknown. It
is pos­si­ble that with the decline in the inci­dence of stroke,21 there
Considerations in deci­sion-mak­ing about HCT for SCD are fewer patients with stroke pres­ent and con­sid­er­ing HCT. It is
Recipient age and donor HLA match also pos­si­ble that a shift has occurred in the per­cep­tion of patients,
The diag­no­sis of SCD is made at birth, and chil­dren are often well care­giv­ers, and phy­si­cians regard­ing recur­rent VOE being an appro­
at a young age. However, the clin­i­cal course waxes and wanes and pri­ate indi­ca­tion for HCT.22-24 The num­ber of hos­pi­tal­i­za­tions or
progresses unpre­dict­ably with age. Families can usu­ally ascer­tain emer­gency room vis­its for SCD-asso­ci­ated pain has long been
very early if their child with SCD has a poten­tial HLA-iden­ti­cal sib­ con­sid­ered a sur­ro­gate mea­sure of the total bur­den of pain. How­
ling since sib­lings are often close in age. Thus, the cru­cial ques­tion ever, the fre­quency of health care uti­li­za­tion may be an inad­e­quate
is, at what age and when in the clin­i­cal course should HCT be mea­sure of the daily bur­den of pain since many patients man­age
con­sid­ered? Gluckman et al9 reported in a study com­bin­ing the most of their pain at home.25 A hos­pi­tal or emer­gency room visit
Euro­pean Society for Blood and Marrow Transplantation and Cen­ rep­re­sents a small frac­tion of the pain expe­ri­ence.26 Thus, some
ter for International Blood and Marrow Transplant Research reg­ patients with a severe bur­den of pain may not meet eli­gi­bil­ity cri­te­
is­tries that out­comes of HCT from HLA-iden­ti­cal related donors ria because they do not have fre­quent health care uti­li­za­tion. Acute
are excel­lent, but EFS decreases with increas­ing age at HCT (haz­ inter­mit­tent vaso-occlu­sive pain is the hall­mark of SCD, but more
ard ratio [HR], 1.09; P < .001). Cappelli et al8 reported 100% OS than half of the adults with SCD tran­si­tion from acute inter­mit­tent
and 93% EFS in chil­dren under 5 years of age. Brazauskas et al12 pain to chronic per­sis­tent pain. Chronic pain, defined by dura­tion
performed an anal­y­sis of 1425 patients with SCD who under­went as the pres­ence of pain on most days of the pre­vi­ous 6 months, is a
HCT between 2008 and 2017. They found that patients aged 12 sig­nif­i­cant cause of mor­bid­ity and impaired QoL in SCD.27 However,
or youn­ger with an HLA-matched sib­ling donor had the best out­ chronic pain may not affect all­indi­vid­u­als with the same degree of
come, with a 3-year EFS of 92%.12 Age at HCT and type of donor dis­abil­ity and inter­fer­ence with activ­i­ties.
were pre­dic­tive of EFS. Patients ≤12 years under­go­ing HCT from A def­i­ni­tion of chronic pain based on dura­tion alone does not
an HLA-iden­ti­cal sib­ling are the best risk group. Patients ≤12 years con­sider the mul­ti­ple dimen­sions of the con­di­tion or cap­ture the
receiv­ing HCT from an unre­lated donor and patients ≥13 years extent of asso­ci­ated dis­abil­ity. The US National Pain Strategy has
from an HLA-iden­ti­cal donor are at inter­me­di­ate risk. All other pro­posed high-impact chronic pain (HICP) as an extreme phe­
patients are in the high-risk group (Table 1). no­type of chronic pain asso­ci­ated with severe dis­abil­ity.28 HICP,
deter­mined by screen­ing patients for fre­quent daily pain and the
Indications for HCT pres­ence of dis­abil­ity,29 is begin­ning to be used as an eli­gi­bil­ity
HCT for SCD has been performed in patients with severe SCD- cri­te­rion for HCT for SCD even in the absence of fre­quent health
related com­pli­ca­tions. Common rea­sons to pro­ceed with HCT care uti­li­za­tion.28,29 However, more research is required on how
include a his­tory of stroke, the need for chronic blood trans­ to inte­grate screen­ing for HICP into clin­i­cal care and deter­mine
fu­sions with the atten­dant risks of trans­fu­sional iron over­load from elec­tronic health records if an indi­vid­ual has HICP. Post-HCT
and fea­tures of dis­ease sever­ity, and pre­dic­tors of pre­ma­ture patients with SCD show improve­ment in pain inter­fer­ence, opi­oid
mor­tal­ity, such as recur­rent VOE and recur­rent acute chest use, hos­pi­tal­i­za­tion, and QoL.18,30-33 A sub­group of patients with
syn­drome.13-16 In patients enrolled in early clin­i­cal tri­als of HCT the pre-HCT fea­tures of sig­nif­i­cantly higher pain bur­dens, anx­i­ety,
for SCD, the most com­mon indi­ca­tions were stroke in 57% of and the use of long-act­ing opi­oids before HCT have per­sis­tent

182  |  Hematology 2021  |  ASH Education Program


chronic pain beyond 1 year post HCT.33 Thus, the iden­ti­fi­ca­tion of early case series reported the use of a myeloablative com­bi­na­tion
HICP pre-HCT, care­ful prep­a­ra­tion of patients, includ­ing behav­ of busul­fan and cyclo­phos­pha­mide,17 more recent series describe
ioral health con­sul­ta­tion, and long-term mul­ti­modal reha­bil­i­ta­tion the use of RIC.9,36,42,43,47,48 The most com­ mon RIC approach is
post-HCT are cru­cial to opti­mize out­comes in these patients. substitut­ing cyclo­phos­pha­mide with fludarabine in com­bi­na­tion
with another agent,9 usu­ally an alkylator such as busul­fan or mel­
Donor con­sid­er­ations pha­lan.48 Several reduced-tox­ic­ity con­di­tion­ing reg­i­mens have
Type of donor been described, includ­ing the use of reduced doses of busul­fan
The major­ity of cases of HCT for SCD reported to inter­na­tional com­bined with fludarabine with or with­out cyclo­phos­pha­mide
reg­is­tries rep­re­sent HLA-iden­ti­cal sib­ling donor HCT.11 Outcomes or the sub­sti­tu­tion of busul­fan with treosulfan or with the addi­
of HCT from HLA-iden­ti­cal donors are supe­rior to those from alter­ tion of thio­tepa.18,49-52 Nonmyeloablative con­di­tion­ing with 200
nate donors,11,34 but the avail­abil­ity of HLA-iden­ti­cal donor HCT is cGy of total body irra­di­a­tion and fludarabine resulted in poor
severely lim­ited by the lack of suit­able fam­ily donors.35 The use of long-term engraft­ment.53,54 A com­bi­na­tion of total body irra­di­
alter­nate donors to expand the donor pool for patients with SCD a­tion (300 cGy) with alemtuzumab resulted in high OS and EFS

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/181/1851576/181krishnamurti.pdf by guest on 13 December 2021


HCT has been the sub­ject of inten­sive inves­ti­ga­tion. HCT from in adults and chil­dren.19,31,55,56 Pretransplant immu­no­sup­pres­sion
unre­lated donors results in sta­ble donor-derived hema­to­poi­e­sis with 2 courses of fludarabine and dexa­meth­a­sone to pre­vent
but is asso­ci­ated with sig­nif­i­cant HCT-related mor­bid­ity and mor­ graft fail­ure has been piloted in haploidentical- and URD HCT
tal­ity.36 The addi­tion of costimulatory block­ade using abatacept for for SCD.45,57
graft-ver­sus-host dis­ease (GVHD) pro­phy­laxis has shown prom­ise
in miti­gat­ing the risk of severe GVHD in HCT for SCD and is the Prophylaxis for GVHD
sub­ject of an ongo­ing mul­ti­cen­ter clin­i­cal trial (NCT 03924401).37 The deple­tion of T cells in vivo using either antithymocyte glob­
HCT from HLA-haploidentical famil­ial donors using in-vivo or ex- u­lin (ATG; 70.6%) or alemtuzumab (11.5%) has been used exten­
vivo T-cell deple­tion has been reported to be safe and effec­tive in sively in patients under­go­ing MRD HCT and may be impor­tant for
early-phase clin­i­cal tri­als and is the sub­ject of ongo­ing mul­ti­cen­ter the pre­ven­tion of graft fail­ure.9 Bernaudin et al reported that the
clin­i­cal tri­als (NCT 03263559, NCT04201210).20,38-41 use of ATG may decrease the rate of graft fail­ure from 22.6% to
3%.58 ATG has not been fre­quently used in MRD UCB trans­plan­ta­
Stem cell source tion (UCBT).59 The deple­tion of T cells in vivo with alemtuzumab
No study to date has com­pared the out­comes from HCT using has been used in MRD HCT and URD UCBT.19,42,43,48 The deple­tion
dif­fer­ent stem cell sources. A higher rate of nonengraftment led of T cells in vivo with posttransplant cyclo­phos­pha­mide with ATG
to the pre­ma­ture clo­sure of the unre­lated donor (URD) umbil­ or alemtuzumab has also been used for haploidentical HCT for

cal cord blood (UCB) arm of the BMT CTN 0601 study.42 A SCD.45,47,60-62 The deple­tion of T cells ex vivo with CD34+ selec­
recent case series sug­gests that the addi­tion of thio­tepa to the tion,63 CD3/CD19 deple­tion,64 or α/β T-cell recep­tor and CD19
reduced-inten­sity con­di­tion­ing (RIC) reg­i­men may enhance the deple­tion has also been used in haploidentical HCT for SCD.65 Ex
engraft­ment of UCB.43 Peripheral blood as a stem cell source has vivo α/β t-cell recep­tor and CD19 deple­tion may reduce the risk
been asso­ci­ated with a higher risk of chronic GVHD (CGVHD).11 of GVHD but may be asso­ci­ated with delayed immune recon­
sti­tu­tion and an increased risk of infec­tion and graft fail­ure.38,65
Donor char­ac­ter­is­tics The most com­monly used GVHD pro­phy­laxis con­sists of calci­
The impact of donor age in HCT for SCD has not been reported. neurin inhib­i­tors (CNIs), which are often com­bined with meth­o­
However, donor age has been reported as a risk fac­tor for CGVHD trex­ate (MTX) or mycophenolate mofetil.9 Locatelli et al reported
fol­low­ing HCT for leu­ke­mia.44 Thus, donor age is likely sig­nif­i­cant that the addi­tion of MTX fol­low­ing MRD UCBT reduced EFS.59
since GVHD does not con­vey any ben­e­fit in HCT for SCD. Increas­ Therefore, mycophenolate mofetil is substituted for MTX in MRD
ingly, patients with SCD are receiv­ing HCT from famil­ial haploiden­ UCBT.42,43 The addi­tion of selec­tive inhi­bi­tion of T-cell costimula­
tical donors, such as par­ents or older sib­lings; hence, it is cru­cial tion with abatacept can decrease GVHD and thus improve the
to deter­mine the con­tri­bu­tion of donor age to out­comes. In addi­ safety pro­file and appli­ca­bil­ity of HCT to SCD.37
tion, ABO major or minor incom­pat­i­bil­ity may add to the risk of
graft stem cell loss,42 delayed red cell engraft­ment decreased OS Cell dose con­sid­er­ations
[44], and pure red cell aplasia.45 Therefore, donor size is an impor­ For MRD UCB, a total nucle­ated cell (TNC) dose for UCB >3 × 107
tant con­sid­er­ation to ensure an ade­quate cell dose with­out com­ TNC/kg recip­i­ent weight is pref­er­a­ble.66 If the UCB cell dose is
pro­mis­ing donor safety. For pedi­at­ric donors for HCT for SCD, the low, a com­bi­na­tion of UCB and bone mar­row may be used.67 In
usual prac­tice is to accept a min­i­mum donor size ≥ 10 kg and age URD UCB, TNC >5 × 107/kg increased engraft­ment and dis­ease-
≥ 1 year. Donor-recip­i­ent cyto­meg­a­lo­vi­rus serostatus is matched free sur­vival.68 A tar­get cell dose of 4 × 108 to 5 × 108 TNC/kg for
(D−/R− or D+/R+) when­ever pos­si­ble to min­i­mize cyto­meg­a­lo­vi­ bone mar­row and 4 × 107 to 5 × 107 TNC/kg for UCB (prethaw) is
rus dis­ease risks. High-titer donor-directed HLA antibodies may recommended.
pre­dict an increased risk for graft rejec­tion. Therefore, recip­i­ents
should be screened for the pres­ence of donor-directed HLA anti­ Engraftment fol­low­ing HCT
bodies. If high-titer antibodies are found to be pres­ent, desen­si­ti­ Risk fac­tors for graft fail­ure include HLA mis­match, high titers
za­tion must be con­sid­ered before pro­ceed­ing to HCT.45,46 of donor-directed HLA antibodies, the inten­sity of con­di­tion­ing,
or the pres­ence of active infec­tion at the time of engraft­ment.69
Conditioning reg­i­men for HCT A com­bi­na­tion of donor chi­me­rism in the lym­phoid lin­e­age
Most often, matched related donor (MRD) HCT has been per­ (CD3) and mye­loid lin­e­age (CD15 or CD33), total Hb level, and
formed fol­low­ing a myeloablative con­di­tion­ing reg­i­men.9 While sickle cell hemo­glo­bin (HbS) per­cent­age are used to eval­u­ate the
Prakash Singh Shekhawat
HCT for SCD  |  183
robust­ness of engraft­ment and donor-derived hema­to­poi­e­sis. Cardiopulmonary
Whole-blood donor chi­me­rism of 11% to 74% may be asso­ci­ated In adult SCD patients, the com­bi­na­tion of echo­car­dio­graphic
with sta­ble donor-derived eryth­ro­poi­e­sis.70-72 Myeloid and lym­ tri­cus­pid regurgitant jet veloc­ity >3.0 m/s and brain natri­uretic
phoid lin­e­age-spe­cific chi­me­rism may pro­vide addi­tional infor­ma­ pep­tide >160 pg/mL is a strong pre­dic­tor of pre­ma­ture mor­tal­
tion, but ery­throid cell chi­me­rism assays are still being eval­u­ated ity.83,84 Following suc­cess­ful HCT there may be an improve­ment
in research stud­ies.73,74 Stable mye­loid chi­me­rism of at least 20% of tri­cus­pid regurgitant jet veloc­ity.18 Post HCT, obtaining an
to 25% is asso­ci­ated with sta­ble donor-derived eryth­ro­poi­e­sis.18,71,72 echo­car­dio­gram annu­ally for 5 years is recommended. In addi­
HbS >50% sug­ gests the imma­ nence of autol­ o­
gous recov­ ery. If tion, an eval­u­a­tion of car­diac iron by T2 MRI at 1 year after HCT
there is mixed or declin­ing donor chi­me­rism or an increas­ing HbS should be con­sid­ered in patients with mod­er­ate or severe iron
per­cent­age, closer mon­i­tor­ing with more fre­quent assess­ments of over­load at HCT.82
donor chi­me­rism may be nec­es­sary. Donor lym­pho­cyte infu­sions
are asso­ci­ated with a sig­nif­i­cant risk of GVHD, and their role in Unique infec­tion risks
improv­ing donor chi­me­rism is unknown. SCD patients undergo autoinfarction of the spleen variably with

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/181/1851576/181krishnamurti.pdf by guest on 13 December 2021


Most patients rejecting the allo­ graft recon­sti­
tute autol­ o­ age and have impair­ment of splenic func­tion and an increased
gous hema­to­poi­e­sis,58,71 even in the case of alter­na­tive-donor risk of pneu­mo­coc­cal sep­sis,85-87 which most often includes non­
HCT.18,36,43,60,71 Marrow aplasia or prolonged cytopenia are indi­ca­ vaccine sero­types.88 Overall, the risk of infec­tion in SCD patients
tions for an urgent sal­vage HCT.18 Consideration of a sec­ond HCT is increased due to splenic infarc­tion, defec­tive opsonization of
should be deferred for at least 6 months fol­low­ing a first HCT encap­su­lated organ­isms, impair­ment of T- and B-cell immune
that has resulted in graft fail­ure with autol­o­gous recon­sti­tu­tion. func­tion, and the pres­ence of infarcted devitalized bone.89 SCD
patients may recover splenic func­tion on HU fol­low­ing the insti­
Organ func­tion con­sid­er­ations spe­cific to HCT for SCD tu­tion of chronic trans­fu­sion or fol­low­ing suc­cess­ful HCT.90 How­
Neurological ever, patients who are older at the time of HCT and those with
In the ini­tial case series of patients who under­went MRD HCT, sei­ exten­sive CGVHD are at higher risk of poor post-HCT splenic
zures and hem­or­rhagic stroke were observed in 30% of patients.75 recov­ery.91 While pneu­mo­coc­cal infec­tions are rare fol­low­ing
The risk fac­tors for neu­ro­log­i­cal com­pli­ca­tions included (1) a his­ HCT for SCD,91 deaths due to over­whelm­ing pneu­mo­coc­cal sep­
tory of prior stroke, (2) hyper­ten­sion due to neu­ro­log­i­cal and renal sis have been reported. Therefore, pneu­mo­coc­cal pro­phy­laxis,
dys­func­tion exac­er­bated by CNIs and cor­ti­co­ste­roid use, (3) hem­ care­ful mon­i­tor­ing of splenic func­tion recov­ery post HCT, and
or­rhagic stroke in patients with preexisting cere­bral vasculopathy timely reimmunization starting with con­ju­gated pneu­mo­coc­cal
with post-HCT throm­bo­cy­to­pe­nia,58 and (4) pos­te­rior revers­ible vac­cines 6 months post trans­plant are impor­tant for the pre­ven­
enceph­a­lop­a­thy syn­drome (PRES) (22%-34%).36,76 There is an tion of seri­ous pneu­mo­coc­cal infec­tions.92
increased risk of PRES in SCD patients that is exac­er­bated dur­ing
HCT and may result in decreased OS and EFS.76,77 As com­pared to Management of trans­fu­sional iron over­load
indi­vid­u­als matched for age, sex, and race, indi­vid­u­als with SCD Patients may have trans­fu­sional iron over­load prior to HCT and
have lower blood pres­sure (BP).78 In patients with SCD, BP above may have also received sev­eral trans­fu­sions of packed red blood
the 50th per­cen­tile for age may be asso­ci­ated with an increased cells with the HCT. Patients who have received a life­time trans­
risk of stroke.78,79 Careful mon­i­tor­ing of BP and aggres­sive man­age­ fu­sion bur­den ≥10 trans­fu­sions and serum fer­ri­tin ≥1500 ng/mL
ment of hyper­ten­sion are par­tic­u­larly impor­tant when patients are are eval­u­ated pre-HCT for liver iron over­load and liver fibro­sis.
receiv­ing both cor­ti­co­ste­roids and CNIs since both drugs are likely MR of the abdo­men is performed for quan­ti­fic ­ a­tion of iron. In
to con­trib­ute to the devel­op­ment of hyper­ten­sion. In BMT CTN addi­tion, ultra­sound or MR elastography may be performed to
0601, a mul­ti­cen­ter trial of URD blood and mar­row trans­plan­ta­tion assess the degree of fibro­sis. In patients with evi­dence of severe
(BMT) for SCD, there was a high inci­dence of PRES.36 The GVHD iron over­load or liver fibro­sis, a hepatology con­sul­ta­tion and liver
pro­phy­laxis in this study included the use of pred­ni­sone through biopsy may be con­sid­ered for deter­min­ing the safety of pro­
day +28. Essential pre­cau­tions to pre­vent neu­ro­log­i­cal com­pli­ ceed­ing with HCT. Patients with cir­rho­sis, bridg­ing fibro­sis, or
ca­tions such as sei­zures or PRES include (1) starting anti­ep­i­lep­tic active hep­at­i­tis are too high risk and may not tol­er­ate an HCT
sei­zure pro­phy­laxis before con­di­tion­ing, espe­cially if busul­fan is con­di­tion­ing reg­i­men.93 In patients with base­line iron over­load,
used, and con­tinu­ing sei­zure pro­phy­laxis for the dura­tion of CNI the removal of excess body iron stores must be insti­tuted with
admin­is­tra­tion, (2) care­fully mon­i­tor­ing and strictly con­trol­ling oral iron che­la­tion, by monthly phle­bot­omy, or by a com­bi­na­tion
BP,17,75 with a tar­get BP within 10% of the median for age and sex thereof after mea­sure­ment of resid­ual iron over­load by serum fer­
for SCD patients as described by Pegelow et al,78 (3) maintaining ri­tin and MRI.94-97 Iron che­la­tion may be ini­ti­ated post HCT when
nor­mal mag­ne­sium lev­els by mag­ne­sium sup­ple­men­ta­tion,80,81 patients are off immu­no­sup­pres­sion, have no evi­dence of GVHD
and (4) admin­is­ter­ing pro­phy­lac­tic plate­let trans­fu­sions to main­ or drug-induced liver dam­age, and are trans­fu­sion inde­pen­dent.
tain a plate­let count >50 000/µL and red blood cell trans­fu­sion as
needed to main­tain an Hb level of 9 to 11 g/dL. Post HCT, the rec­ Renal
om­men­da­tion is to con­sider obtaining brain mag­netic res­o­nance Serum blood urea nitro­gen/cre­at­i­nine and glo­mer­u­lar fil­tra­
angi­og­ra­phy/imag­ing (MRA/MRI) at 1 and 2 years after HCT and tion rate or 24-hour cre­at­i­nine clear­ance and SCD-asso­ci­ated
then every 2 years as clin­i­cally indi­cated in patients with a his­tory pro­tein­uria are crit­i­cal con­sid­er­ations pre-HCT and fol­low-ups
of stroke, moyamoya pretransplant, PRES, or another neu­ro­tox­ic­ for recov­ ery post HCT.82 A prolonged course of CNIs places
ity dur­ing HCT.82 An age-appro­pri­ate neurocognitive eval­u­a­tion patients at a high risk of renal dys­func­tion. Patients with SCD
should be obtained at 1 year. It should be repeated every 2 years if with prior com­ ple­
ment-medi­ ated vas­ cu­lar injury due to their
there is a his­tory of a neu­ro­toxic com­pli­ca­tion dur­ing HCT.82 under­ly­ing pri­mary hemo­lytic dis­ease and addi­tional stress­ors

184  |  Hematology 2021  |  ASH Education Program


dur­ing the HCT pro­cess may develop pro­gres­sive endo­the­lial lev­els that result in deci­sional delays or uncertainties.114 The per­
injury and end-organ dys­func­tion.98 Case reports and small case cep­tion that the cur­rent dis­ease bur­den has become unac­cept­
series of trans­plant-asso­ci­ated throm­botic microangiopathy in able is a com­mon con­sid­er­ation for HCT for SCD among patients
patients with SCD and fol­low­ing HCT sug­gest the need to mon­ and their phy­si­cians.22-24,113 The avail­abil­ity of an HLA-iden­ti­cal sib­
i­tor trans­plant-asso­ci­ated throm­botic microangiopathy, con­ ling donor may be viewed as prov­i­den­tial by par­ents and is a
trol hyper­ten­sion, con­sider alter­na­tives to CNIs, and treat with sig­nif­i­cant fac­tor when con­sid­er­ing HCT. Some patients or care­
eculizumab when appro­pri­ate.99-103 giv­ers will not con­sider HCT at any level of risk.115,116 Almost three-
quar­ters of patients report being will­ing to take a mod­est risk of
Gonadal dam­age and impaired fer­til­ity ≥ 5% mor­tal­ity, whereas 57% are will­ing to take the ≥ 10% risk of
HCT recip­i­ents for SCD are at high risk of gonadal dam­age and GVHD. Following a suc­cess­ful HCT, SCD patients and care­giv­ers
infer­til­ity.104 However, data are lacking on the patient and care­ do not report deci­sional regret, even with active CGVHD.113
giver per­spec­tive on the impor­tance of fer­til­ity in mak­ing deci­
sions about HCT, the costs of and access to fer­til­ity pres­er­va­tion, The phy­si­cian per­spec­tive on deci­sion-mak­ing

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/181/1851576/181krishnamurti.pdf by guest on 13 December 2021


and the emo­tional and psy­cho­log­i­cal impact of gonadal dam­age Their past expe­ri­ences and the out­comes of pre­vi­ous patients
on the long-term sur­vi­vors of HCT.105 Sperm bank­ing in males and may influ­ence how phy­si­cians respond in mak­ing rec­om­men­da­
oocyte cryo­pres­er­va­tion in females are stan­dard pro­ce­dures tions to patients about seek­ing HCT con­sul­ta­tion. For exam­ple,
that can mit­i­gate the risk of infer­til­ity.106 Ovarian tis­sue bank­ing Bakshi et al, in a qual­i­ta­tive study of phy­si­cian deci­sion-mak­ing
is an accept­able fer­til­ity-pres­er­va­tion tech­nique that is no lon­ in dis­ease-mod­i­fy­ing ther­apy for SCD, found that the phy­si­cian’s
ger con­sid­ered exper­i­men­tal and is the only method to pre­serve per­cep­tion of the sever­ity of dis­ease and the abil­ity of a given
fer­til­ity for pre­pu­ber­tal girls.106 Testicular tis­sue cryo­pres­er­va­tion patient to adhere to the med­i­ca­tion and treat­ment reg­i­men is also
in pre­pu­ber­tal boys is under inves­ti­ga­tion.107,108 Fertility pres­ a sig­nif­i­cant con­sid­er­ation in their recommending HCT for SCD.23
er­va­tion pro­ce­dures pre-HCT must also con­sider that HU may
cause gonadal dam­age and have an adverse impact on male and Socioeconomic sta­tus and HCT for SCD
female fer­til­ity in SCD patients.109,110 Racial and socio­ eco­ nomic disparities may adversely affect
Med­ic­aid cov­er­age of fer­til­ity pres­er­va­tion is extremely lim­ access to and out­comes of HCT in minor­ity pop­u­la­tions.117,118
ited both in the scope of ben­e­fits and the num­ber of states that There is also a sub­ stan­
tial regional dis­ par­
ity in donor search
require such a ben­e­fit.111 For exam­ple, only 15 states require fer­til­ cov­er­age, trans­plant pro­ce­dures, hos­pi­tal­i­za­tions, med­i­ca­tions,
ity pres­er­va­tion cov­er­age in pri­vate insur­ance plans, and 5 states transportation, and lodg­ing pro­vided by dif­fer­ent states’ Med­
extend this ben­e­fit only to females. In addi­tion, state stat­utes ic­aid pro­grams for HCT.119 Mupfudze et al have described the
pro­vide var­i­able cov­er­age based on mar­i­tal sta­tus, diag­no­sis, bur­den of nav­i­gat­ing eli­gi­bil­ity for Med­ic­aid, the peer review
length of fer­til­ity prob­lems, and the mon­e­tary limit of the ben­e­fit. pro­cess for var­i­ous com­po­nents and pro­ce­dures for HCT, the dif­
fer­ence between fee-for-ser­vice and com­pre­hen­sively man­aged
Long-term and late effects of HCT plans, and the bur­den that HCT poses to fam­i­lies seek­ing HCT for
HCT can sta­bi­lize organ func­tion and ame­lio­rate man­i­fes­ta­tions SCD.120 Patients enrolled in Med­ic­aid had a lower EFS (HR, 2.36;
of SCD; patients need sys­tem­atic fol­low-up of mon­i­tor­ing SCD 95% CI, 1.44-3.85; P = .0006) and a higher cumu­la­tive inci­dence of
com­pli­ca­tions and the long-term and late effects of HCT accord­ graft fail­ure fol­low­ing HCT (HR, 2.57; 95% CI, 1.43-4.60; P = .0015)
ing to established con­sen­sus guide­lines.82,112 Further, most of the com­pared to pri­vately insured patients with SCD.121 Registry data
published lit­er­a­ture on the late effects of HCT for SCD is based pro­vide no clue to the fac­tors con­trib­ut­ing to worse out­comes
on HCT for malig­nan­cies. There is, there­fore, a need for a uni­ver­ among Med­ic­aid-insured patients. Afri­can Amer­i­cans, His­pan­ics,
sal, lon­gi­tu­di­nal clin­i­cal reg­is­try to fol­low out­comes after HCT and indi­vid­u­als with Med­ic­aid cov­er­age have been dem­on­strated
for SCD. to have worse out­comes fol­low­ing treat­ment for hema­to­log­i­cal
malig­nan­cies and fol­low­ing HCT.117,118,122-124 These find­ings pro­vide
Values and pref­er­ences of patients, fam­i­lies, the ratio­nale for fur­ther study of the com­plex inter­ac­tion of psy­
and care­giv­ers cho­so­cial func­tion­ing, health behav­iors, racial and eth­nic dispar­
The deci­sion to undergo HCT for SCD involves mak­ing com­plex ities, poor socio­eco­nomic sta­tus, and health care disparities on
trade-offs between the prom­ise of relief of dis­ease man­i­fes­ta­ out­comes of HCT in patients with SCD.117,118,122-124
tions, sta­bi­li­za­tion of organ func­tion, halt­ing of dis­ease pro­gres­
sion, and improve­ment of lon­gev­ity and QoL, on the one hand, Future direc­tions
with sub­stan­tial treat­ment bur­den and mor­bid­ity in the short Despite prog­ress in the con­di­tion­ing reg­i­mens, donor options,
term, and the risk of new long-term sequelae such as CGVHD, GVHD pro­phy­laxis, and out­comes of allo­ge­neic HCT over the last
infer­til­ity, and sub­se­quent malig­nancy, on the other hand.24 Many 25 years, HCT has been applied to a tiny pro­por­tion of patients
fac­tors are involved in patient and care­giver deci­sion-mak­ing for with SCD. Advances in sev­eral fields, includ­ing ame­lio­ra­tion of the
SCD.24,113,114 These include the unpre­dict­able onset and pro­gres­ risk of mor­bid­ity, mor­tal­ity, and long-term sequelae, are nec­es­
sion of dis­ease com­pli­ca­tions, the repeated need to seek health sary to increase the appli­ca­bil­ity and accept­abil­ity of HCT for SCD.
care, a poor QoL, a wors­en­ing of dis­ease-related com­pli­ca­tions, Antibody-based myeloablation approaches to trans­plant con­di­
the need to make major ther­a­peu­tic deci­sions, and con­cerns tion­ing through the use of anti-c-kit antibodies, anti-c-kit anti­body
about the long-term con­se­quences of SCD. Families that are con­ju­gated to the drug saporin, a ribo­some-inactivating pro­tein
aware of and have access to HCT, have strong fam­ily sup­port, with potent cell-cycle-inde­pen­dent cyto­toxic activ­ity, and c-kit
and have an avail­­able HLA-iden­ti­cal sib­ling donor are more likely targeted chi­me­ric anti­gen recep­tor T cells can reduce or elim­i­nate
to con­sider HCT.113 Overall, fam­i­lies face deci­sional con­flict at the need for che­mo­ther­apy-based con­di­tion­ing reg­i­mens.125-131
Prakash Singh Shekhawat
HCT for SCD  |  185
A nonchemotherapy con­di­tion­ing reg­i­men can fur­ther reduce Off-label drug use
acute mor­bid­ity related to mucositis and mar­row aplasia, gonadal Lakshmanan Krishnamurti: nothing to disclose.
tox­ic­ity, and sub­se­quent malig­nancy related to alkylators. The
refine­ment of GVHD pro­phy­laxis using posttransplant cyclo­phos­ Correspondence
pha­mide or costimulatory block­ade with abatacept can reduce Lakshmanan Krishnamurti, Children’s Healthcare of Atlanta-Egleston,
acute GVHD and CGVHD and their sequelae. UCB is lim­ited by 1405 Clifton Road NE, Atlanta, GA 30322; e-mail: lakshmanan.
both the avail­abil­ity of HLA-matched donors and the lim­ited cell krishnamurti@emory​­.edu
dose of avail­­able UCB units. Ex-vivo expan­sion of the cell dose of References
UCB units is likely to make more and pos­si­bly bet­ter HLA-matched 1. McGann PT, Ware RE. Hydroxyurea ther­apy for sickle cell ane­mia. Expert
units avail­­able. Advances in stem cell expan­sion and stem cell Opin Drug Saf. 2015;14(11):1749-1758.
engi­neer­ing, includ­ing iden­ti­fy­ing crit­i­cal cyto­kines, pro­teins, and 2. Cieri-Hutcherson NE, Hutcherson TC, Conway-Habes EE, Burns BN, White
NA. Systematic review of l-glu­ta­mine for pre­ven­tion of vaso-occlu­sive pain
small drug ago­nists, have led to clin­i­cal pro­to­cols for expanding
cri­sis in patients with sickle cell dis­ease. Pharmacotherapy. 2019;39(11):
hema­to­poi­etic stem cells and improved engraft­ment. URD UCBT

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/181/1851576/181krishnamurti.pdf by guest on 13 December 2021


1095-1104.
with omidubicel, a nic­o­tin­amide-based, ex vivo-expanded UCB 3. Blair HA. Voxelotor: first approval. Drugs. 2020;80(2):209-215.
prod­uct, is asso­ci­ated with rapid engraft­ment but a high rate of 4. Blair HA. Crizanlizumab: first approval. Drugs. 2020;80(1):79-84.
5. Sahdev I, Brochstein J, Werther N, Stiles J. Timing of alemtuzumab with
GVHD in patients with SCD.132 Ongoing clin­i­cal tri­als can refine
respect to day of bone mar­row infu­sion and its effects upon engraft­ment
the ex-vivo expan­sion of hema­to­poi­etic stem cells, with future and graft-ver­sus-host dis­ease in patients with sickle cell dis­ease: a sin­gle-
appli­ca­tions to SCD.133 Pharmacokinetics-directed pre­ci­sion dos­ insti­tu­tionalstudy. J Pediatr Hematol Oncol. 2020;42(8):e718-e722.
ing can improve the safety and effi­cacy of in-vivo T-cell deple­tion 6. Bernaudin F, Dalle JH, Bories D, et al; Société Française de Greffe de Moelle
with alemtuzumab and myeloablation with mel­pha­lan.134,135 et de Thérapie Cellulaire. Long-term event-free sur­ vival, chi­
me­ rism and
fer­til­ity out­comes in 234 patients with sickle-cell ane­mia youn­ger than 30
Impaired fer­til­ity is an unin­tended con­se­quence of the con­di­
years after myeloablative con­di­tion­ing and matched-sib­ling trans­plan­ta­tion
tion­ing reg­i­men. Improving the accept­abil­ity of HCT for SCD will in France. Haematologica. 2020;105(1):91-101.
require advo­cacy with state gov­ern­ments to expand access to a 7. Saraf SL, Rondelli D. Allogeneic hema­to­poi­etic stem cell trans­plan­ta­tion for
fer­til­ity pres­er­va­tion ben­e­fit, remove the spou­sal require­ment, adults with sickle cell dis­ease. J Clin Med. 2019;8(10):1565.
8. Cappelli B, Volt F, Tozatto-Maio K, et al; for Eurocord, the Cellular Therapy
expand the diag­nos­tic cri­te­ria, and include the ben­e­fit in Med­ic­aid
and Immunobiology Working Party, and the Paediatric Diseases Working Party
plans.111 Further study of improv­ing the aware­ness of, access to, and of the Euro­pean Society for Blood and Marrow Transplantation. Risk fac­tors
out­comes of HCT in under­served pop­u­la­tions can help us bet­ter and out­comes according to age at trans­plan­ta­tion with an HLA-iden­ti­cal
under­stand how to make HCT promptly avail­­able to a greater pro­ sib­ling for sickle cell dis­ease. Haematologica. 2019;104(12):e543-e546.
por­tion of SCD patients who could ben­e­fit from the pro­ce­dure. 9. Gluckman E, Cappelli B, Bernaudin F, et al; for Eurocord, the Pediatric Work­
ing Party of the Euro­pean Society for Blood and Marrow Transplantation,
The advances in HCT are occur­ring con­tem­po­ra­ne­ously with
and the Center for International Blood and Marrow Transplant Research.
explo­sive growth in clin­i­cal tri­als of novel dis­ease-mod­i­fy­ing Sickle cell dis­ease: an inter­na­tional sur­vey of results of HLA-iden­ti­cal sib­ling
ther­a­pies, on the one hand, and gene ther­apy strat­e­gies, on the hema­to­poi­etic stem cell trans­plan­ta­tion. Blood. 2017;129(11):1548-1556.
other hand. How the inter­ac­tion of these devel­op­ments is likely 10. Walters MC, De Castro LM, Sullivan KM, et al. Indications and results of HLA-
iden­ti­cal sib­ling hema­to­poi­etic cell trans­plan­ta­tion for sickle cell dis­ease.
to affect the future sta­tus of HCT in the man­age­ment of SCD is
Biol Blood Marrow Transplant. 2016;22(2):207-211.
unknown. Still, it is safe to say that we have embarked on a jour­ 11. Eapen M, Brazauskas R, Walters MC, et al. Effect of donor type and con­
ney of increased and improved ther­a­peu­tic options for patients di­tion­ing reg­i­men inten­sity on allo­ge­neic trans­plan­ta­tion out­comes in
with SCD. patients with sickle cell dis­ease: a ret­ro­spec­tive multicentre, cohort study.
Lancet Haematol. 2019;6(11):e585-e596.
12. Brazauskas R, Scigliuolo GM, Wang HL, et al. Risk score to pre­dict event-
free sur­ vival after hema­ to­
poi­etic cell trans­ plant for sickle cell dis­ ease.
Blood. 2020;136(5):623-626.
CLINICAL CASE (Con­t in­u ed) 13. Brandow AM, Panepinto JA. Hydroxyurea use in sickle cell dis­ease: the bat­
tle with low pre­scrip­tion rates, poor patient com­pli­ance and fears of toxic­
This 12-year-old girl with HbSS with fea­tures of severe dis­ease
ities. Expert Rev Hematol. 2010;3(3):255-260.
phe­no­type is at risk for the long-term com­pli­ca­tions of SCD. 14. Darbari DS, Wang Z, Kwak M, et al. Severe pain­ful vaso-occlu­sive cri­ses and
Her clin­i­cal course has remained severe despite a trial of HU, mor­tal­ity in a con­tem­po­rary adult sickle cell ane­mia cohort study. PLoS
L-glu­ta­mine, and voxelotor. She has excel­lent risk fac­tors for One. 2013;8(11):e79923.
15. Quinn CT, Lee NJ, Shull EP, Ahmad N, Rogers ZR, Buchanan GR. Prediction
HCT from her HLA-iden­ti­cal sib­ling. I would rec­om­mend that
of adverse out­comes in chil­dren with sickle cell ane­mia: a study of the
the hema­tol­o­gist have a series of con­ver­sa­tions with the fam­ily Dallas Newborn Cohort. Blood. 2008;111(2):544-548.
to edu­cate them about HCT and then refer them to HCT con­ 16. Prasad R, Hasan S, Castro O, Perlin E, Kim K. Long-term out­ comes in
sul­ta­tion and explore research pro­ce­dures for ovar­ian tis­sue patients with sickle cell dis­ease and fre­quent vaso-occlu­sive cri­ses. Am J
pres­er­va­tion. If the fam­ily pro­vi­des informed con­sent, we will Med Sci. 2003;325(3):107-109.
17. Walters MC, Patience M, Leisenring W, et al. Bone mar­row trans­plan­ta­tion
pro­ceed to HCT with a pretransplant trans­fu­sion to lower HbS.
for sickle cell dis­ease. N Engl J Med. 1996;335(6):369-376.
During HCT we will main­tain tar­get Hb and plate­let counts by 18. Krishnamurti L, Neuberg DS, Sullivan KM, et al. Bone mar­row trans­plan­ta­
trans­fu­sions, care­fully man­age BP, and main­tain nor­mal serum tion for ado­les­cents and young adults with sickle cell dis­ease: results of a
mag­ne­sium lev­els to pre­vent PRES. pro­spec­tive mul­ti­cen­ter pilot study. Am J Hematol. 2019;94(4):446-454.
19. Hsieh MM, Fitzhugh CD, Weitzel RP, et al. Nonmyeloablative HLA-matched
sib­ling allo­ge­neic hema­to­poi­etic stem cell trans­plan­ta­tion for severe sickle
cell phe­no­type. JAMA. 2014;312(1):48-56.
20. Bolaños-Meade J, Cooke KR, Gamper CJ, et al. Effect of increased dose of
Conflict-of-inter­est dis­clo­sure total body irra­di­a­tion on graft fail­ure asso­ci­ated with HLA-haploidentical
Lakshmanan Krishnamurti: no com­pet­ing finan­cial inter­ests to trans­plan­ta­tion in patients with severe haemoglobinopathies: a ­pro­spec­tive
declare. clin­i­cal trial. Lancet Haematol. 2019;6(4):e183-e193.

186  |  Hematology 2021  |  ASH Education Program


21. Fullerton HJ, Adams RJ, Zhao S, Johnston SC. Declining stroke rates in Cali­ 44. Watkins BK, Horan J, Storer B, Martin PJ, Carpenter PA, Flowers ME. Recip­
fornian chil­dren with sickle cell dis­ease. Blood. 2004;104(2):336-339. ient and donor age impact the risk of devel­op­ing chronic GvHD in chil­
22. Bakshi N, Katoch D, Sinha CB, et al. Assessment of patient and care­giver dren after allo­ge­neic hema­to­poi­etic trans­plant. Bone Marrow Transplant.
atti­tudes and approaches to deci­ sion-mak­ ing regard­ ing bone mar­ row 2017;52(4):625-626.
trans­plant for sickle cell dis­ease: a qual­i­ta­tive study. JAMA Netw Open. 45. Pawlowska AB, Cheng JC, Karras NA, et  al. HLA haploidentical stem cell
2020;3(5):e206742. trans­plant with pretransplant immu­no­sup­pres­sion for patients with sickle
23. Bakshi N, Sinha CB, Ross D, Khemani K, Loewenstein G, Krishnamurti L. Pro­ cell dis­ease. Biol Blood Marrow Transplant. 2018;24(1):185-189.
ponent or col­lab­o­ra­tive: phy­si­cian per­spec­tives and approaches to dis­ease 46. Ciurea SO, Cao K, Fernandez-Vina M, et al. The Euro­pean Society for Blood
mod­i­fy­ing ther­a­pies in sickle cell dis­ease. PLoS One. 2017;12(7):e0178413. and Marrow Transplantation (EBMT) Consensus Guidelines for the detec­
24. Sinha CB, Bakshi N, Ross D, Loewenstein G, Krishnamurti L. Primary care­ tion and treat­ment of donor-spe­cific anti-HLA antibodies (DSA) in hap­
giver deci­sion-mak­ing in hema­to­poi­etic cell trans­plan­ta­tion and gene ther­ loidentical hema­to­poi­etic cell trans­plan­ta­tion. Bone Marrow Transplant.
apy for sickle cell dis­ease. Pediatr Blood Cancer. 2021;68(1):e28749. 2018;53(5):521-534.
25. Smith WR, Penberthy LT, Bovbjerg VE, et  al. Daily assess­ment of pain in 47. Wiebking V, Hütker S, Schmid I, Immler S, Feuchtinger T, Albert MH. Reduced
adults with sickle cell dis­ease. Ann Intern Med. 2008;148(2):94-101. tox­ic­ity, myeloablative HLA-haploidentical hema­to­poi­etic stem cell trans­
26. Smith WR, Bovbjerg VE, Penberthy LT, et  al. Understanding pain and plan­ta­tion with post-trans­plan­ta­tion cyclo­phos­pha­mide for sickle cell dis­

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/181/1851576/181krishnamurti.pdf by guest on 13 December 2021


improv­ing man­age­ment of sickle cell dis­ease: the PiSCES study. J Natl Med ease. Ann Hematol. 2017;96(8):1373-1377.
Assoc. 2005;97(2):183-193. 48. King AA, Kamani N, Bunin N, et al. Successful matched sib­ling donor mar­
27. Field JJ, Ballas SK, Campbell CM, et al. AAAPT diag­nos­tic cri­te­ria for acute row trans­plan­ta­tion fol­low­ing reduced inten­sity con­di­tion­ing in chil­dren
sickle cell dis­ease pain. J Pain. 2019;20(7):746-759. with hemo­glo­bin­op­a­thies. Am J Hematol. 2015;90(12):1093-1098.
28. Von Korff M, Scher AI, Helmick C, et al. United States national pain strat­ 49. Gyurkocza B, Sandmaier BM. Conditioning reg­i­mens for hema­to­poi­etic cell
egy for pop­u­la­tion research: con­cepts, def­i­ni­tions, and pilot data. J Pain. trans­plan­ta­tion: one size does not fit all. Blood. 2014;124(3):344-353.
2016;17(10):1068-1080. 50. Bacigalupo A, Ballen K, Rizzo D, et al. Defining the inten­sity of con­di­tion­
29. Pitcher MH, Von Korff M, Bushnell MC, Porter L. Prevalence and pro­file of ing reg­i­mens: work­ing def­i­ni­tions. Biol Blood Marrow Transplant. 2009;15
high-impact chronic pain in the United States. J Pain. 2019;20(2):146-160. (12):1628-1633.
30. Han J, Holden CC, Ahluwalia AY, et al. Chronic opi­oid use can be reduced 51. Horan JT, Haight A, Dioguardi JL, et al. Using fludarabine to reduce expo­sure
or discontinued after haematopoietic stem cell trans­plan­ta­tion for sickle to alkylating agents in chil­dren with sickle cell dis­ease receiv­ing busul­fan,
cell dis­ease. Br J Haematol. 2020;191(3):e70-e72. cyclo­phos­pha­mide, and antithymocyte glob­u­lin trans­plant con­di­tion­ing:
31. Saraf SL, Oh AL, Patel PR, et al. Nonmyeloablative stem cell trans­plan­ta­ results of a dose de-esca­la­tion trial. Biol Blood Marrow Transplant.2015;21(5):
tion with alemtuzumab/low-dose irra­di­a­tion to cure and improve the qual­ 900-905.
ity of life of adults with sickle cell dis­ease. Biol Blood Marrow Transplant. 52. Strocchio L, Zecca M, Comoli P, et al. Treosulfan-based con­di­tion­ing reg­i­
2016;22(3):441-448. men for allo­ge­neic haematopoietic stem cell trans­plan­ta­tion in chil­dren with
32. Badawy SM, Beg U, Liem RI, Chaudhury S, Thompson AA. A sys­tem­atic sickle cell dis­ease. Br J Haematol. 2015;169(5):726-736.
review of qual­ity of life in sickle cell dis­ease and thal­as­se­mia after stem cell 53. Iannone R, Casella JF, Fuchs EJ, et  al. Results of min­i­mally toxic nonmy­
trans­plant or gene ther­apy. Blood Adv. 2021;5(2):570-583. eloablative trans­plan­ta­tion in patients with sickle cell ane­mia and beta-
33. Darbari DS, Liljencrantz J, Ikechi A, et al. Pain and opi­oid use after rever­sal thal­as­se­mia. Biol Blood Marrow Transplant. 2003;9(8):519-528.
of sickle cell dis­ease fol­low­ing HLA-matched sib­ling haematopoietic stem 54. Horan JT, Liesveld JL, Fenton P, Blumberg N, Walters MC. Hematopoietic
cell trans­plant. Br J Haematol. 2019;184(4):690-693. stem cell trans­plan­ta­tion for mul­ti­ply trans­fused patients with sickle cell
34. Gluckman E, Cappelli B, Scigliuolo GM, De la Fuente J, Corbacioglu S. Alter­ dis­ease and thal­as­se­mia after low-dose total body irra­di­a­tion, fludarabine,
native donor hema­to­poi­etic stem cell trans­plan­ta­tion for sickle cell dis­ease and rab­bit anti-thy­mo­cyte glob­u­lin. Bone Marrow Transplant. 2005;35(2):
in Europe. Hematol Oncol Stem Cell Ther. 2020;13(4):181-188. 171-177.
35. Mentzer WC, Heller S, Pearle PR, Hackney E, Vichinsky E. Availability of 55. Guilcher GM, Monagel D, Leaker M, et al. Alemtuzumab/low dose TBI con­
related donors for bone mar­row trans­plan­ta­tion in sickle cell ane­mia. Am J di­tion­ing facil­i­tates sta­ble long-term donor hema­to­poi­etic cell engraft­
Pediatr Hematol Oncol. 1994;16(1):27-29. ment from sib­ling donors in chil­dren with sickle cell dis­ease. Blood. 2017;
36. Shenoy S, Eapen M, Panepinto JA, et  al. A trial of unre­lated donor mar­ 130(suppl 1):4592.
row trans­plan­ta­tion for chil­dren with severe sickle cell dis­ease. Blood. 56. Guilcher GMT, Monagel DA, Nettel-Aguirre A, et  al. Nonmyeloablative
2016;128(21):2561-2567. matched sib­ling donor hema­to­poi­etic cell trans­plan­ta­tion in chil­dren and
37. Ngwube A, Shah N, Godder K, Jacobsohn D, Hulbert ML, Shenoy S. Abata­ ado­les­cents with sickle cell dis­ease. Biol Blood Marrow Transplant. 2019;
cept is effec­tive as GVHD pro­phy­laxis in unre­lated donor stem cell trans­ 25(6):1179-1186.
plan­ta­tion for chil­dren with severe sickle cell dis­ease. Blood Adv. 2020;4(16): 57. Kharya G, Bakane AN, Rauthan AM. Pretransplant mye­loid and immune
3894-3899. sup­pres­sion, reduced tox­ic­ity con­di­tion­ing with posttransplant cyclo­phos­
38. Foell J, Kleinschmidt K, Jakob M, Troeger A, Corbacioglu S. Alternative donor: pha­mide: ini­tial out­comes of novel approach for matched unre­lated donor
αß/CD19 T-cell-depleted haploidentical hema­to­poi­etic stem cell trans­plan­ hema­to­poi­etic stem cell trans­plant for hemo­glo­bin­op­a­thies. Pediatr Blood
ta­tion for sickle cell dis­ease. Hematol Oncol Stem Cell Ther. 2020;13(2): Cancer. 2021;68(4):e28909.
98-105. 58. Bernaudin F, Socie G, Kuentz M, et al; for the Société Française de Greffe
39. Foell J, Schulte JH, Pfirstinger B, et  al. Haploidentical CD3 or α/β T-cell de Moelle et de Thérapie Cellulair. Long-term results of related myeloab­
depleted HSCT in advanced stage sickle cell dis­ease. Bone Marrow Trans- lative stem-cell trans­plan­ta­tion to cure sickle cell dis­ease. Blood. 2007;
plant. 2019;54(11):1859-1867. 110(7):2749-2756.
40. Patel DA, Akinsete AM, Connelly JA, Kassim AA. T-cell deplete ver­sus T-cell 59. Locatelli F, Kabbara N, Ruggeri A, et al; Eurocord; Euro­pean Society for Blood
replete haploidentical hema­to­poi­etic stem cell trans­plan­ta­tion for sickle and Marrow Transplantation. Outcome of patients with hemo­glo­bin­op­a­thies
cell dis­ease: where are we? Expert Rev Hematol. 2019;12(9):733-752. given either cord blood or bone mar­row trans­plan­ta­tion from an HLA-iden­ti­cal
41. Patel DA, Dhedin N, Chen H, et  al. Early viral reactivation despite excel­ sib­ling. Blood. 2013;122(6):1072-1078.
lent immune recon­sti­tu­tion fol­low­ing haploidentical bone mar­row trans­ 60. Bolaños-Meade J, Fuchs EJ, Luznik L, et al. HLA-haploidentical bone mar­
plant with post-trans­plant cytoxan for sickle cell dis­ease. Transpl Infect row trans­plan­ta­tion with posttransplant cyclo­phos­pha­mide expands the
Dis. 2020;22(1):e13222. donor pool for patients with sickle cell dis­ease. Blood. 2012;120(22):4285-
42. Kamani NR, Walters MC, Carter S, et al. Unrelated donor cord blood trans­ 4291.
plan­ta­tion for chil­dren with severe sickle cell dis­ease: results of one cohort 61. Fitzhugh CD, Hsieh MM, Taylor T, et  al. Cyclophosphamide improves
from the phase II study from the Blood and Marrow Transplant Clinical engraft­ment in patients with SCD and severe organ dam­age who undergo
Trials Network (BMT CTN). Biol Blood Marrow Transplant. 2012;18(8):1265- haploidentical PBSCT. Blood Adv. 2017;1(11):652-661.
1272. 62. de la Fuente J, Dhedin N, Koyama T, et  al. Haploidentical bone mar­row
43. Abra­ham A, Cluster A, Jacobsohn D, et al. Unrelated umbil­i­cal cord blood trans­plan­ta­tion with post-trans­plan­ta­tion cyclo­phos­pha­mide plus thio­tepa
trans­plan­ta­tion for sickle cell dis­ease fol­low­ing reduced-inten­sity con­di­ improves donor engraft­ment in patients with sickle cell ane­mia: results
tion­ing: results of a phase I trial. Biol Blood Marrow Transplant. 2017;23(9): of an inter­na­tional learn­ing col­lab­o­ra­tive. Biol Blood Marrow Transplant.
1587-1592. 2019;25(6):1197-1209.
Prakash Singh Shekhawat
HCT for SCD  |  187
63. Gilman AL, Eckrich MJ, Epstein S, et  al. Alternative donor hema­to­poi­etic 84. Gladwin MT, Barst RJ, Gibbs JS, et al; walk-PHaSST Investigators and
stem cell trans­plan­ta­tion for sickle cell dis­ease. Blood Adv. 2017;1(16):1215- Patients. Risk fac­tors for death in 632 patients with sickle cell dis­ease in
1223. the United States and United Kingdom. PLoS One. 2014;9(7):e99489.
64. Foell J, Pfirstinger B, Rehe K, Wolff D, Holler E, Corbacioglu S. Haploidenti­ 85. Rogers ZR, Wang WC, Luo Z, et al; BABY HUG Investigators. Biomarkers of
cal stem cell trans­plan­ta­tion with CD3+−/CD19+− depleted periph­eral stem splenic func­tion in infants with sickle cell ane­mia: base­line data from the
cells for patients with advanced stage sickle cell dis­ease and no alter­na­tive BABY HUG trial. Blood. 2011;117(9):2614-2617.
donor: results of a pilot study. Bone Marrow Transplant. 2017;52(6):938- 86. Brown AK, Sleeper LA, Miller ST, Pegelow CH, Gill FM, Waclawiw MA; for the
940. Cooperative Study of Sickle Cell Disease. Reference val­ues and hema­to­logic
65. Gaziev J, Isgrò A, Sodani P, et al. Haploidentical HSCT for hemo­glo­bin­op­ changes from birth to 5 years in patients with sickle cell dis­ease. Arch
a­thies: improved out­comes with TCRαβ+/CD19+-depleted grafts. Blood Pediatr Adolesc Med. 1994;148(8):796-804.
Adv. 2018;2(3):263-270. 87. Gale HI, Bobbitt CA, Setty BN, et al. Expected sonographic appear­ance of
66. Locatelli F, Pagliara D. Allogeneic hema­to­poi­etic stem cell trans­plan­ta­tion the spleen in chil­dren and young adults with sickle cell dis­ease: an update.
in chil­dren with sickle cell dis­ease. Pediatr Blood Cancer. 2012;59(2):372- J Ultrasound Med. 2016;35(8):1735-1745.
376. 88. Martin OO, Moquist KL, Hennessy JM, Nelson SC. Invasive pneu­mo­coc­
67. Soni S, Boulad F, Cowan MJ, et  al. Combined umbil­i­cal cord blood and cal dis­ease in chil­dren with sickle cell dis­ease in the pneu­mo­coc­cal con

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/181/1851576/181krishnamurti.pdf by guest on 13 December 2021


bone mar­row from HLA-iden­ti­cal sib­ling donors for hema­to­poi­etic stem ­ju­gate vac­cine era. Pediatr Blood Cancer. 2018;65(1):e26713.
cell trans­plan­ta­tion in chil­dren with hemo­glo­bin­op­a­thies. Pediatr Blood 89. Ochocinski D, Dalal M, Black LV, et al. Life-threat­en­ing infec­tious com­pli­
Cancer. 2014;61(9):1690-1694. ca­tions in sickle cell dis­ease: a con­cise nar­ra­tive review. Front Pediatr.
68. Ruggeri A, Eapen M, Scaravadou A, et al; Eurocord Registry; Center for 2020;8(20 Feb­ru­ary):38.
International Blood and Marrow Transplant Research; New York Blood Cen­ 90. Khatib R, Rabah R, Sarnaik SA. The spleen in the sick­ling dis­or­ders: an
ter. Umbilical cord blood trans­plan­ta­tion for chil­dren with thal­as­se­mia and update. Pediatr Radiol. 2009;39(1):17-22.
sickle cell dis­ease. Biol Blood Marrow Transplant. 2011;17(9):1375-1382. 91. Nickel RS, Seashore E, Lane PA, et al. Improved splenic func­tion after hema­
69. Olsson R, Remberger M, Schaffer M, et al. Graft fail­ure in the mod­ern era to­poi­etic stem cell trans­plant for sickle cell dis­ease. Pediatr Blood Cancer.
of allo­ge­neic hema­to­poi­etic SCT. Bone Marrow Transplant. 2013;48(4): 2016;63(5):908-913.
537-543. 92. Tomblyn M, Chiller T, Einsele H, et al; Center for International Blood and
70. Walters MC, Patience M, Leisenring W, et al; Multicenter Investigation of Marrow Research; National Marrow Donor pro­gram; Euro­pean Society for
Bone Marrow Transplantation for Sickle Cell Disease. Stable mixed hema­to­ Blood and Marrow Transplantion; Amer­i­can Society of Blood and Marrow
poi­etic chi­me­rism after bone mar­row trans­plan­ta­tion for sickle cell ane­mia. Transplantation; Cana­dian Blood and Marrow Transplant Group; Infectious
Biol Blood Marrow Transplant. 2001;7(12):665-673. Diseases Society of America; Society for Healthcare Epidemiology of Amer­
71. Abra­ham A, Hsieh M, Eapen M, et al; National Institutes of Health; Cen­ ica; Association of Medical Microbiology and Infectious Disease Canada;
ter for International Blood and Marrow Transplant Research. Relationship Centers for Disease Control and Prevention. Guidelines for pre­vent­ing infec­
between mixed donor-recip­i­ent chi­me­rism and dis­ease recur­rence after tious com­pli­ca­tions among hema­to­poi­etic cell trans­plan­ta­tion recip­i­ents:
hema­to­poi­etic cell trans­plan­ta­tion for sickle cell dis­ease. Biol Blood Mar- a global per­spec­tive. Biol Blood Marrow Transplant. 2009;15(10):1143-1238.
row Transplant. 2017;23(12):2178-2183. 93. Ishak KG. Pathologic fea­ tures of chronic hep­ a­
ti­
tis. Am J Clin Pathol.
72. Fitzhugh CD, Cordes S, Taylor T, et al. At least 20% donor mye­loid chi­me­ 2000;113(1):40-55.
rism is nec­es­sary to reverse the sickle phe­no­type after allo­ge­neic HSCT. 94. Angelucci E, Pilo F. Management of iron over­load before, dur­ing, and after
Blood. 2017;130(17):1946-1948. hema­to­poi­etic stem cell trans­plan­ta­tion for thal­as­se­mia major: trans­plan­
73. Wu CJ, Gladwin M, Tisdale J, et  al. Mixed haematopoietic chi­me­rism for ta­tion and iron over­load. Ann NY Acad Sci. 2016;1368(1):115-121.
sickle cell dis­ease pre­vents intra­vas­cu­lar haemolysis. Br J Haematol. 95. Angelucci E, Muretto P, Lucarelli G, et al; for the Ital­ian Cooperative
2007;139(3):504-507. Group for Phlebotomy Treatment of Transplanted Thalassemia by Bone
74. Krishnamurti L, Kharbanda S, Biernacki MA, et  al. Stable long-term donor Marrow Transplantation. Phlebotomy to reduce iron over­load in patients
engraft­ment fol­low­ing reduced-inten­sity hema­to­poi­etic cell trans­plan­ta­tion cured of thal­as­se­mia by bone mar­row trans­plan­ta­tion. Blood. 1997;90(3):
for sickle cell dis­ease. Biol Blood Marrow Transplant. 2008;14(11):1270-1278. 994-998.
75. Walters MC, Sullivan KM, Bernaudin F, et al. Neurologic com­pli­ca­tions after 96. Giardini C, Galimberti M, Lucarelli G, et al. Desferrioxamine ther­apy accel­
allo­ge­neic mar­row trans­plan­ta­tion for sickle cell ane­mia. Blood. 1995;85(4): er­ates clear­ance of iron depos­its after bone mar­row trans­plan­ta­tion for
879-884. thal­as­sae­mia. Br J Haematol. 1995;89(4):868-873.
76. Gaziev J, Marziali S, Paciaroni K, et  al. Posterior revers­ible enceph­a­lop­a­ 97. Schettini F, De Mattia D, Sabato V, Del Vecchio GC. Compliance with iron-
thy syn­drome after hema­to­poi­etic cell trans­plan­ta­tion in chil­dren with che­la­tion treat­ment after bone mar­row trans­plan­ta­tion. Lancet. 1994;343
hemo­glo­bin­op­a­thies. Biol Blood Marrow Transplant. 2017;23(9):1531-1540. (8897):604.
77. Solh Z, Taccone MS, Marin S, Athale U, Breakey VR. Neurological PRESen­ 98. Jodele S. Double trou­ble: com­ple­ment-medi­ated throm­botic microangi­
tations in sickle cell patients are not always stroke: a review of pos­te­rior opathy in patients with hemo­glo­bin­op­a­thies after stem cell trans­plan­ta­
revers­ible enceph­a­lop­a­thy syn­drome in sickle cell dis­ease. Pediatr Blood tion. Pediatr Blood Cancer. 2017;64(9).
Cancer. 2016;63(6):983-989. 99. Abusin GA, Abu-Arja R, Bajwa RPS, Horwitz EM, Auletta JJ, Rangarajan HG.
78. Pegelow CH, Colangelo L, Steinberg M, et al. Natural his­tory of blood pres­ Severe trans­ plant-asso­ci­
ated throm­ botic microangiopathy in patients
sure in sickle cell dis­ease: risks for stroke and death asso­ci­ated with rel­a­ with hemo­glo­bin­op­a­thies. Pediatr Blood Cancer. 2017;64(9):e26503.
tive hyper­ten­sion in sickle cell ane­mia. Am J Med. 1997;102(2):171-177. 100. Bayer G, von Tokarski F, Thoreau B, et al. Etiology and out­comes of throm­
79. Kassim AA, Galadanci NA, Pruthi S, DeBaun MR. How I treat and man­age botic microangiopathies. Clin J Am Soc Nephrol. 2019;14(4):557-566.
strokes in sickle cell dis­ease. Blood. 2015;125(22):3401-3410. 101.  Bhunia N, Abu-Arja R, Bajwa RPS, Auletta JJ, Rangarajan HG. Successful
80. Thompson CB, June CH, Sullivan KM, Thomas ED. Association between cyc­ treat­ment with eculizumab for pos­te­rior revers­ible enceph­a­lop­a­thy syn­
losporin neu­ro­tox­ic­ity and hypomagnesaemia. Lancet. 1984;2(8412):1116- drome due to under­ly­ing trans­plant-asso­ci­ated throm­botic microangiop­
1120. athy in patients transplanted for sickle cell dis­ease. Pediatr Blood Cancer.
81. June CH, Thompson CB, Kennedy MS, Loughran TP Jr, Deeg HJ. Correlation 2019;66(10):e27912.
of hypo­mag­ne­se­mia with the onset of cyclo­spor­ine-asso­ci­ated hyper­ten­ 102. Davies JOJ, Hart AC, De La Fuente J, Bain BJ. Macrophage acti­va­tion syn­
sion in mar­row trans­plant patients. Transplantation. 1986;41(1):47-51. drome and post-trans­plant microangiopathy fol­low­ing haploidentical
82. Shenoy S, Gaziev J, Angelucci E, et  al. Late effects screen­ing guide­lines bone mar­row trans­plan­ta­tion for sickle cell ane­mia. Am J Hematol. 2018;​
after hema­to­poi­etic cell trans­plan­ta­tion (HCT) for hemo­glo­bin­op­a­thy: con­ 93(4):588-589.
sen­sus state­ment from the Second Pediatric Blood and Marrow Transplant 103. Maigne G, Ferlicot S, Galacteros F, et  al. Glomerular lesions in patients
Consortium inter­na­tional con­fer­ence on late effects after pedi­at­ric HCT. with sickle cell dis­ease. Medicine (Baltimore). 2010;89(1):18-27.
Biol Blood Marrow Transplant. 2018;24(7):1313-1321. 104. Walters MC, Hardy K, Edwards S, et al; Multicenter Study of Bone Marrow
83. Gladwin MT, Sachdev V, Jison ML, et  al. Pulmonary hyper­ ten­sion as a Transplantation for Sickle Cell Disease. Pulmonary, gonadal, and cen­tral
risk fac­tor for death in patients with sickle cell dis­ease. N Engl J Med. ner­vous sys­tem sta­tus after bone mar­row trans­plan­ta­tion for sickle cell
2004;350(9):886-895. dis­ease. Biol Blood Marrow Transplant. 2010;16(2):263-272.

188  |  Hematology 2021  |  ASH Education Program


105. 
Mishkin AD, Mapara MY, Barhaghi M, Reshef R. Fertility con­ cerns and 121. Abu Al Hamayel N, Waldfogel JM, Hannum SM, et al. Pain expe­ri­ences of
access to care for stem cell trans­plan­ta­tion can­di­dates with sickle cell adults with sickle cell dis­ease and hema­to­poi­etic stem cell trans­plan­ta­
dis­ease. Biol Blood Marrow Transplant. 2020;26(8):e192-e197. tion: a qual­i­ta­tive study. Pain Med. 2021;22(8):1753-1759.
106. Practice Committee of the Amer­i­can Society for Reproductive Medicine. 122. Isaac EI, Meisman AR, Drucker K, et al. The rela­tion­ship between health
Fertility pres­ er­
va­tion in patients under­ go­ing gonadotoxic ther­ apy or disparities, psy­cho­so­cial func­tion­ing and health out­comes in pedi­at­ric
gonad­ec­tomy: a com­mit­tee opin­ion. Fertil Steril. 2019;112(6):1022-1033. hema­tol­ogy-oncol­ogy and stem cell trans­plant pop­u­la­tions: rec­om­men­
107.  Brodigan K, Kapadia M, Frazier AL, et al. Safety of sur­gi­cal fer­til­ity pres­er­ da­tions for clin­i­cal care. Int J Environ Res Public Health. 2020;17(7):2218.
va­tion pro­ce­dures in chil­dren prior to hema­to­poi­etic stem cell trans­plant. 123. Patel MI, Ma Y, Mitchell B, Rhoads KF. How do dif­fer­ences in treat­ment
Transplant Cell Ther. 2021;27(8):696.e1-696696.e4. impact racial and eth­nic disparities in acute mye­loid leu­ke­mia? Cancer
108. Lautz TB, Harris CJ, Laronda MM, Erickson LL, Rowell EE. A fer­til­ity pres­ Epidemiol Biomarkers Prev. 2015;24(2):344-349.
er­va­tion toolkit for pedi­at­ric sur­geons car­ing for chil­dren with can­cer. 124. Kirtane K, Lee SJ. Racial and eth­nic disparities in hema­to­logic malig­nan­
Semin Pediatr Surg. 2019;28(6):150861. cies. Blood. 2017;130(15):1699-1705.
109. Joseph L, Jean C, Manceau S, et al. Effect of hydroxy­urea expo­sure before 125. Li Z, Czechowicz A, Scheck A, Rossi DJ, Mur­phy PM. Hematopoietic chi­me­
puberty on sperm param­e­ters in males with sickle cell dis­ease. Blood. rism and donor-spe­cific skin allo­graft tol­er­ance after non-genotoxic CD117
2021;137(6):826-829. anti­body-drug-con­ju­gate con­di­tion­ing in MHC-mismatched allotrans­

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/181/1851576/181krishnamurti.pdf by guest on 13 December 2021


110.  Pecker LH, Hussain S, Christianson MS, Lanzkron S. Hydroxycarbamide plantation. Nat Commun. 2019;10(1):616.
expo­sure and ovar­ian reserve in women with sickle cell dis­ease in the 126. Czechowicz A, Palchaudhuri R, Scheck A, et al. Selective hema­to­poi­etic
Multicenter Study of Hydroxycarbamide. Br J Haematol. 2020;191(5):880- stem cell abla­ tion using CD117-anti­ body-drug-con­ ju­
gates enables safe
887. and effec­tive trans­plan­ta­tion with immu­nity pres­er­va­tion. Nat Commun.
111.  Benoit M, Chiles K, Hsieh M. The land­scape of cov­er­age for fer­til­ity pres­ 2019;10(1):617.
er­va­tion in male pedi­at­ric patients. Urol Pract. 2018;5(3):198-204. 127.   Witt RG, Wang B, Nguyen QH, Eikani C, Mattis AN, MacKenzie TC. Deple­
tion of murine fetal hema­to­poi­etic stem cells with c-kit recep­tor and CD47
112. Shenoy S, Angelucci E, Arnold SD, et al. Current results and future research
block­ade improves neo­na­tal engraft­ment. Blood Adv. 2018;2(24):3602-
pri­or­i­ties in late effects after hema­to­poi­etic stem cell trans­plan­ta­tion for
3607.
chil­dren with sickle cell dis­ease and thal­as­se­mia: a con­sen­sus state­ment
128. Arai Y, Choi U, Corsino CI, et al. Myeloid con­di­tion­ing with c-kit-targeted
from the Second Pediatric Blood and Marrow Transplant Consortium
CAR-T cells enables donor stem cell engraft­ment. Mol Ther. 2018;26(5):
inter­na­tional con­fer­ence on late effects after pedi­at­ric hema­to­poi­etic
1181-1197.
stem cell trans­plan­ta­tion. Biol Blood Marrow Transplant. 2017;23(4):552-
129.  Yokoi T, Yokoi K, Akiyama K, et  al. Non-myeloablative preconditioning
561.
with ACK2 (anti-c-kit anti­body) is effi­cient in bone mar­row trans­plan­ta­tion
113. Khemani K, Ross D, Sinha C, Haight A, Bakshi N, Krishnamurti L. Experi­
for murine mod­els of mucopolysaccharidosis type II. Mol Genet Metab.
ences and deci­sion mak­ing in hema­to­poi­etic stem cell trans­plant in sickle
2016;​119(3):232-238.
cell dis­ease: patients’ and care­giv­ers’ per­spec­tives. Biol Blood Marrow
130. Xue X, Pech NK, Shelley WC, Srour EF, Yoder MC, Dinauer MC. Antibody
Transplant. 2018;24(5):1041-1048.
targeting KIT as pretransplantation con­di­tion­ing in immu­no­com­pe­tent
114. Schulz GL, Kelly KP, Holtmann M, Armer JM. Navigating deci­sional con­flict
mice. Blood. 2010;116(24):5419-5422.
as a fam­ily when fac­ing the deci­sion of stem cell trans­plant for a child or 131. Czechowicz A, Kraft D, Weissman IL, Bhattacharya D. Efficient trans­plan­
ado­les­cent with sickle cell dis­ease. Patient Educ Couns. 2021;104(5):1086- ta­tion via anti­body-based clear­ance of hema­to­poi­etic stem cell niches.
1093. Science. 2007;318(5854):1296-1299.
115. van Besien K, Koshy M, Anderson-Shaw L, et al. Allogeneic stem cell trans­ 132. Parikh S, Brochstein JA, Galamidi E, Schwarzbach A, Kurtzberg J. ­Allogeneic
plan­ta­tion for sickle cell dis­ease. A study of patients’ deci­sions. Bone Mar- stem cell trans­plan­ta­tion with omidubicel in sickle cell dis­ease. Blood
row Transplant. 2001;28(6):545-549. Adv. 2021;5(3):843-852.
116. Kodish E, Lantos J, Stocking C, Singer PA, Siegler M, Johnson FL. Bone mar­ 133. Zimran E, Papa L, Hoffman R. Ex vivo expan­sion of hema­to­poi­etic stem
row trans­plan­ta­tion for sickle cell dis­ease: a study of par­ents’ deci­sions. cells: finally transitioning from the lab to the clinic. Blood Rev. 2021;​50(2):​
N Engl J Med. 1991;325(19):1349-1353. 100853.
117. Majhail NS, Nayyar S, Santibañez ME, Mur­phy EA, Denzen EM. Racial dis­ 134. Chandra S, Mizuno K, Zhao J, et  al. Test-dose phar­ma­co­ki­net­ics guided
parities in hema­to­poi­etic cell trans­plan­ta­tion in the United States. Bone mel­pha­lan dose adjust­ment in reduced inten­sity con­di­tion­ing allo­ge­neic
Marrow Transplant. 2012;47(11):1385-1390. trans­plant for non-malig­nant dis­or­ders. Published online 1 June 2021. Br J
118. Majhail NS, Omondi NA, Denzen E, Mur­phy EA, Rizzo JD. Access to hema­ Clin Pharmacol.
to­poi­etic cell trans­plan­ta­tion in the United States. Biol Blood Marrow 135. Dong M, Emoto C, Fukuda T, et al. Model-informed pre­ci­sion dos­ing for
Transplant. 2010;16(8):1070-1075. alemtuzumab in pae­di­at­ric and young adult patients under­go­ing allo­ge­
119. Preussler JM, Farnia SH, Denzen EM, Majhail NS. Variation in Med­ ic­ neic haematopoietic cell trans­plan­ta­tion. Published online 28 June 2021.
aid cov­er­age for hema­to­poi­etic cell trans­plan­ta­tion. J Oncol Pract. Br J Clin Pharmacol. 2021.
2014;10(4):e196-e200.
120.  Mupfudze TG, Preussler JM, Sees JA, SanCartier M, Arnold SD, Devine S.
A qual­i­ta­tive anal­y­sis of state Med­ic­aid cov­er­age ben­e­fits for allo­ge­neic
hema­to­poi­etic cell trans­plan­ta­tion (alloHCT) for patients with sickle cell © 2021 by The Amer­i­can Society of Hematology
dis­ease (SCD). Transplant Cell Ther. 2021;27(4):345-351. DOI 10.1182/hema­tol­ogy.2021000251

Prakash Singh Shekhawat


HCT for SCD  |  189
EMERGING THERAPIES AND CONSIDERATIONS FOR SICKLE CELL DISEASE

EVIDENCE-BASED MINIREVIEW

In young children with severe sickle cell disease,


do the benefits of HLA-identical sibling donor HCT

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/190/1851641/190shah.pdf by guest on 13 December 2021


outweigh the risks?
Niketa Shah1 and Lakshmanan Krishnamurti2
Section of Pediatric Hematology/Oncology/BMT, Yale School of Medicine, New Haven, CT; and 2Aflac Cancer and Blood Disorders Center,
1

Children’s Healthcare of Atlanta, Emory University, Atlanta, GA

In case 1, a 14-month-old male child with sickle cell disease (SCD) was referred for evaluation for an allogeneic hematopoi-
etic stem cell transplant (HCT). The patient had a history of dactylitis 3 times in his first year of life and febrile episodes twice
at the consult. His 4-year-old sister was found to be human leukocyte antigen (HLA) identical. The patient was started on
hydroxyurea (HU) at 2.5 years of age. His parents again sought consultation when he was 5 years old because of concerns
about his medical condition. At the time, the patient had experienced 2 vaso-occlusive pain episodes (VOEs) requiring
hospitalization during the previous 2 years. He had also experienced intermittent pain crises requiring rest at home for
2 to 3 days. The child has not attended school in person due to the COVID-19 pandemic. The family is considering HCT
but is ambivalent about it because of potential toxicity. In case 2, an 8-year-old female child is 3 years out from HCT for
SCD from her HLA-identical sibling. Before HCT, despite receiving HU, she had experienced >5 VOEs requiring hospital-
ization and 2 episodes of acute chest syndromes in the previous 3 years. She had also been missing almost 50 days of
school days each year. After HCT, she is now attending school regularly and participating in all normal age-appropriate
activities. The parents believe that HCT has been transformative in their child’s life.

LEARNING OBJECTIVES
• Understand therapeutic options for a young child with SCD
• Describe the current outcomes of MSD HCT
• Critically appraise the role of MSD HCT for SCD in young children

Introduction Outcomes with comprehensive care and


Sickle cell disease (SCD) is an autosomal recessive inher- disease-modifying therapy for SCD
ited hemoglobinopathy. Over 300 000 affected babies are Advances such as newborn screening, penicillin prophy-
born each year with SCD, which is expected to increase to laxis, pneumococcal immunization, and comprehensive care
over 400 000 a year by 2050.1,2 Although the overwhelm- have improved survival in children with SCD. Hydroxyurea
ing majority of patients live in Africa, India, or the Middle (HU) has been proven to effectively decrease the frequency
East, growing numbers of individuals with SCD are encoun- of vaso-occlusive pain episodes (VOEs), acute chest syn-
tered in areas that were historically not endemic for malaria drome, frequency of transfusions, and death in multiple
because of population migrations. In the United States, it randomized controlled trials.6 In addition, HU is an effective
is estimated that 100 000 individuals are affected with substitute for chronic transfusions to prevent primary stroke
SCD.2-4 SCD is associated with a significant risk of morbidity among high-risk pediatric patients with SCD who have
and premature mortality.5 Thus, SCD is a significant public abnormal transcranial Doppler (TCD) flow velocity.5 Obser-
health problem worldwide that disproportionately affects vational studies have demonstrated a relationship between
populations residing in resource-poor settings. Hemato- HU use and decreased rates of hospitalization and blood
poietic cell transplantation (HCT) can ameliorate long-term transfusions.6 Thus, this relatively inexpensive orally admin-
SCD-related morbidity and its consequences. istered medication can be used globally to ameliorate SCD

190 | Hematology 2021 | ASH Education Program


com­pli­ca­tions and improve sur­vival. Nouraie et al7 reported the has been fur­ther improve­ment in the out­comes of HCT for SCD,
results of the pulmonary hypertension and the hypoxic response with OS and EFS in the range of 94% to 100% and 91% to 100%,
in sickle cell disease study that esti­mated that the sur­vival of a respec­ tively, with a min­ i­
mum risk of mor­ tal­
ity with reduced-
cohort of chil­dren, ado­les­cents, and young adults with SCD was inten­sity/tox­ic­ity reg­i­mens.20-24 An inter­na­tional, ret­ro­spec­tive,
99% to 18 years, and this declined to 94% at 25 years. Stroke was the reg­is­try-based anal­y­sis reported an OS of 92.9% and EFS of 91.4%
most com­mon cause of death. Elevated sys­tolic pul­mo­nary artery in 1000 chil­dren who under­went HCT between 1986 and 2013.21
pres­sure as reflected in triscupid regurgitant jet velocity (TRV) On mul­ti­var­i­ate anal­y­sis, sur­vival was bet­ter with bone mar­row
≥2.7 m/s, high serum fer­ri­tin con­cen­tra­tion, low forced expiratory than periph­eral blood stem cells graft and those who under­went
velocity (FEV1)/forced vital capacity, and high neu­tro­phil count trans­plan­ta­tion after 2006.21 Cappelli et al,22 in a study of recip­
were bio­mark­ers of increased risk of death, pre­dom­i­nantly after i­ent age as a pre­dic­tor of the out­come of MSD HCT, reported
the tran­si­tion to adult­hood. Recently, newer dis­ease-mod­i­fy­ing an OS and EFS of 100% and 93%, respec­tively, in chil­dren 0 to
agents approved by the US Food and Drug Administration (FDA) 5 years of age. Bernaudin et al25 reported out­comes of MSD HCT
offer added poten­tial to improve out­comes for patients with SCD. for SCD from 1988 to 2012 and described the improve­ment in
The new dis­ease-mod­i­fy­ing drugs include ­voxelotor, which binds out­come since 2005. They noted that for MSD since 2005, OS is

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/190/1851641/190shah.pdf by guest on 13 December 2021


to the N-ter­mi­nus of sickle hemoglobin to sta­bi­lize oxy­gen­ated 98.6% and EFS is 97.4%. The use of reduced-inten­sity or nonmy-
sick­ling and pre­vent sick­ling8; L-glu­ta­mine, which reduces oxi­da­ eloablative con­di­tion­ing strat­e­gies such as com­bin­ing alemtu-
tive stress and could result in fewer epi­sodes of VOE9; and crizan- zumab with fludarabine and mel­pha­lan,26 as well as low-dose
lizumab, which acts on cytoadherence of sickle red blood cells, total body irra­di­a­tion (300 cGy),20,27 has been asso­ci­ated with
leu­ko­cytes, and plate­lets to the endo­the­lium.10 These drugs offer high OS and EFS in both adults and chil­dren.
the poten­tial for fur­ther ame­lio­rat­ing com­pli­ca­tions of SCD and
pro­vide an alter­na­tive for HCT for dis­ease mod­i­fi­ca­tion. However, The risk-ben­e­fit trade-off in HCT for SCD
in the absence of long-term data, we do not know how the addi­ The deci­sion to pro­ceed with cura­tive options for SCD rep­re­sents
tional FDA-approved SCD ther­a­pies will mod­ify the clin­i­cal his­tory a com­plex risk-ben­e­fit trade-off. On one hand, sur­vival to adult­
of patients with SCD. hood is almost uni­ver­sal in chil­dren receiv­ing the best com­pre­
The improve­ment in sur­vival of chil­dren with SCD between hen­sive care. The dis­ease, how­ever, progresses in the major­ity to
1979 and 2005 seemed to mir­ror a sim­i­lar decrease in adult sur­ cumu­la­tive organ dam­age, mor­bid­ity, and mor­tal­ity in adult­hood.
vival dur­ing the same period.11 Although indi­vid­ual cen­ters in On the other hand, fol­low­ing HLA MSD HCT in young chil­dren,
high-resource set­tings report a median sur­vival of 58 to 67 years there is a high prob­a­bil­ity of sur­vival, with a poten­tial for alle­vi­at­
over­all, there has been lit­tle improve­ment in sur­vival for adults ing symp­toms related to SCD, sta­bi­liz­ing organ func­tion, and the
with SCD.12 In adults, there is a pro­gres­sive loss of pul­mo­nary pos­si­bil­ity of the patient being ­able to live rel­a­tively every­day life.
FEV1 at 49 cc/y.13 More than 61% of patients with SCD aged But HCT itself is asso­ci­ated with sub­stan­tial com­pli­ca­tions in the
18 to 30 years develop either micro- or macroalbuminuria.14 There short term, a sig­nif­i­cant bur­den of care that can be dis­rup­tive to
is a wors­en­ing change in tri­cus­pid regurgitant jet veloc­ity over the patient and care­giv­ers, and a small risk of mor­tal­ity.
time.15 There is neurocognitive decline and worse per­for­mance Furthermore, the long-term sequelae of HCT, such as graft-
IQ , processing speed, cog­ni­tive func­tion, and exec­u­tive func­ ver­sus-host dis­ease, infer­til­ity, and sub­se­quent malig­nancy, could
tion with age.16 Acute-on-chronic mor­bid­ity leads to reduced be unin­tended con­se­quences of this ther­apy (Figure 1). This deci­
health-related qual­ity of life (HRQOL), depres­sion and anx­i­ety, sional dilemma is also fur­ther compounded by the patient’s age,
dimin­ished edu­ca­tional and occu­pa­tional ful­fill­ment, and pre­ the sever­ity of the cur­rent clin­i­cal course, the type of donor
ma­ture mor­tal­ity, con­trib­ut­ing to health care costs..2,17,18 Disease- avail­­able, and the avail­abil­ity of other treat­ment options. Thus,
mod­i­fy­ing ther­a­pies and sup­port­ive care can improve out­comes par­ents con­sid­er­ing HCT for their young child face mul­ti­ple com­
but place a sub­stan­tial bur­den of care, require­ment of med­i­ca­ plex trade-offs: the pri­mary trade-off for a fam­ily of a 5-year-old
tion adher­ence, and inter­rup­tion of daily life.17 is sur­vival of the child in the decade when they age from 5 to 15
Thus, a child receiv­ing com­pre­hen­sive care in high-resource years. Although no study has com­pared HCT with stan­dard of
set­tings has an esti­mated 99% sur­vival into adult­hood. However, care in chil­dren with SCD, published stud­ies sug­gest that sur­vival
there may be pro­gres­sive organ dam­age, impaired qual­ity of life,
con­sid­er­able mor­bid­ity in child­hood, and risk of pre­ma­ture mor­
tal­ity in adult­hood. However, it is cru­cial to rec­og­nize that it is
not yet known if the addi­tional FDA-approved SCD ther­a­pies will
mod­ify the clin­i­cal his­tory of patients with SCD in adult­hood.

Outcomes after matched sib­ling donor bone mar­row


trans­plant
HCT for SCD was first performed in 1984 from a human leu­ko­
cyte anti­gen (HLA) matching sib­ling donor (MSD) to treat acute
mye­loid leu­ke­mia in a patient who also had SCD and paved the
way for a cura­tive option for SCD. The early trans­plant series
using myeloablative con­di­tion­ing, such as a trans­plant for malig­
nant dis­eases, showed over­all sur­vival (OS) and event-free sur­
vival (EFS) in the range of 77% to 95% and 93% to 95% fol­low­ing
HCT from an HLA MSD, respec­tively.18,19 In the past decade, there Figure 1. Risk-benefit paradigm for curative therapy for SCD.
Prakash Singh Shekhawat
HLA iden­ti­cal donor HCT in young chil­dren  |  191
to the next decade for a 5-year-old child with SCD fol­low­ing HCT who are youn­ger than 5 years have an EFS of 93% and an
stan­dard of care or MSD HCT may be com­pa­ra­ble. However, a OS of 100% com­pared with 89% and 95% for those aged 5 to 10
par­ent may also con­sider other trade-offs such as the out­comes years and 81% and 88% for those older than 15 years, respec­
of HCT in child­hood vs the poten­tial for mor­bid­ity and mor­tal­ tively.22 Gluckman et al21 report that for every 1-year incre­ment
ity in adult­hood, HRQOL with or with­out HCT, and cura­tive HCT in age, there was a 9% increase in the haz­ard ratio for treat­ment
vs chronic GVHD, infer­til­ity, car­dio­vas­cu­lar risk, and sub­se­quent fail­ure—that is, graft fail­ure or death. Similarly, for every 1-year
malig­nancy.28 How fam­i­lies resolve these deci­sional trade-offs is incre­ment in age, there was a 10% increase in the haz­ard ratio
likely to be highly indi­vid­u­al­ized and depen­dent on sev­eral fac­ for death.22 Risk of graft fail­ure, grade II to IV acute graft-ver­sus-
tors unique to the cir­cum­stances of the patient and fam­ily. To host dis­ease (GVHD), and chronic GVHD are low­est in chil­dren
assist the hema­tol­o­gist in the con­ver­sa­tion with the fam­ily of a youn­ger than 5 years at HCT. Gluckman et al21 reported from the
young child with SCD who has an HLA-iden­ti­cal sib­ling, we sum­ Euro­pean Society for Blood and Marrow Transplantation reg­is­try
ma­rize the avail­­able evi­dence as answers to crit­i­cal ques­tions that many cen­ters have diverse expe­ri­ence in performing HCT
that may arise in the minds of patients and their care­giv­ers as for SCD. This is mir­rored by the report by Bernaudin et al23 in a
they con­tem­plate HCT. sin­gle-cen­ter study with a 5-year EFS of 97.9% (95% con­fi­dence

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/190/1851641/190shah.pdf by guest on 13 December 2021


inter­val, 95.5%-100%). Brazauskas et al24 devel­oped a risk scor­ing
Who has been offered HCT for SCD? sys­tem that included age at HCT and donor type to pre­dict EFS
HCT for SCD has been offered to patients with severe SCD- after HCT for SCD. They con­cluded that patients 12 years and
related com­pli­ca­tions. Indications for HCT include stroke, the youn­ger with an HLA-iden­ti­cal fam­ily donor have the low­est risk.
need for chronic blood trans­ fu­sions to pre­
vent SCD-related Another pri­mary con­sid­er­ation in the tim­ing of HCT for SCD
com­pli­ca­tions, and severe dis­ease com­pli­ca­tions such as recur­ is the pres­ence or immi­nence of organ dam­age. Therefore, HCT
rent VOE, recur­rent acute chest syn­drome, sickle nephrop­a­thy, should pref­er­a­bly be offered to patients who have been pre-
or ret­i­nop­a­thy that at least affects vision in 1 eye or osteonecro- dicted to have a poor out­come but have not expe­ri­enced severe
sis.18,29 In early clin­i­cal tri­als of HCT for SCD, the most com­mon organ dam­age to avoid seri­ous side effects asso­ci­ated with HCT
indi­ca­tions were stroke in 57% of patients and fre­quent VOEs in pro­ce­dures. However, SCD organ dam­age is insid­i­ous, with pro­
23% of patients. The most com­mon indi­ca­tion for HCT recently gres­sive loss of FEV1 at 49 cc/y,13 61% of patients with SCD aged
has been recur­rent VOE in more than 70% of the cases.21 The 18 to 30 years devel­op­ing either micro- or macroalbuminuria,14
change in the most fre­quent indi­ca­tion for HCT may reflect the wors­ en­
ing changes in tri­ cus­
pid regurgitant jet veloc­ ity over
decrease in the inci­dence of stroke because of bet­ter com­pre­ time,15 and neurocognitive decline with poor per­for­mance IQ ,
hen­sive care, screen­ing for stroke risk, and pri­mary pre­ven­tion processing speed, cog­ni­tive func­tion, and exec­u­tive func­tion.16
of stroke by the insti­tu­tion of chronic blood trans­fu­sion. It is Because over­all out­comes of MSD HCT for SCD are best when
also pos­si­ble that phy­si­cians and patients are more accepting HCT is performed at age 5 years or youn­ger, the con­sen­sus opin­
of HCT for indi­vid­ua ­ ls with recur­rent VOE. Utilization of health ion of an Euro­pean Society for Blood and Marrow Transplantation
care for recur­rent VOE is being used as a marker of dis­ease expert panel is to con­sider MSD at a young age.33
sever­ity and a pre­dic­tor of long-term sur­vival in adults. Yet,
health care uti­li­za­tion for VOE is not an accu­rate mea­sure of What about long-term side effects and qual­ity of life after
pain bur­ den because many VOE epi­ sodes are man­ aged at HCT com­ pared with patients with SCD who have not
home. Thus, some patients may have chronic dis­abling pain received cura­tive ther­a­pies?
but may not access health care for var­i­ous rea­sons.30 There is, Improvement in HRQOL out­comes in patients with SCD fol­low­ing
there­fore, a need to bet­ter esti­mate pain bur­den to inform risk- HCT has been reported in mul­ti­ple stud­ies.34-39 This includes sig­
ben­e­fit strat­i­fi­ca­tion in con­sid­er­ation of HCT for SCD. An ongo­ nif­i­cant improve­ment in phys­i­cal, emo­tional, psy­cho­so­cial, gen­
ing clin­i­cal trial is study­ing HCT from HLA matched sib­ling HCT eral health, bodily pain, pain inter­fer­ence, and vital­ity domains
for chil­dren with the less severe clin­i­cal phe­no­type (ClinicalTri of HRQOL. It has also been reported that car­diac, neu­ro­logic,
als​­.gov NCT04018937). and neuropsychologic func­tions sta­bi­lize after HCT.19 In the most
recent report, for chil­dren with SCD requir­ing chronic trans­fu­sion
Is it pos­si­ble to offer MSD HCT to every child with SCD? because of per­sis­tently ele­vated TCD veloc­i­ties, MSD HSCT was
The appli­ca­bil­ity of HCT is lim­ited because only 14% to 18% of sig­nif­i­cantly asso­ci­ated with lower TCD veloc­i­ties at 1 year com­
patients with SCD have an avail­­able HLA-iden­ti­cal fam­ily donor.31 pared with stan­dard care.25 One of the sig­nif­i­cant risks fol­low­ing
Most sib­ling donors can have sickle cell trait. However, HCT from HCT is gonadal dam­age and infer­til­ity.19 Fertility pres­er­va­tion by
a donor with sickle cell trait is safe and effec­tive.31,32 Clinical tri­ sperm bank­ing or cryo­pres­er­va­tion of oocytes can poten­tially
als are eval­u­at­ing alter­nate donor HCT. An esti­mated 19% of the mit­i­gate the risk of infer­til­ity. Ovarian tis­sue cryo­pres­er­va­tion in
patients may find a suit­able HLA matched unre­lated donor,32 and pre­pu­ber­tal females is now con­sid­ered the stan­dard of clin­i­cal
most are likely to find a haploidentical fam­ily donor. If the safety care. Experimental pro­ ce­
dures such as tes­ tic­
u­lar tis­
sue cryo­
and effi­cacy of alter­nate donor HCT are proven, a more sig­nif­ pres­er­va­tion are still under inves­ti­ga­tion.40,41 On the other hand,
i­cant pro­por­tion of patients may be pro­vided HCT. Thus, only HU also adversely affects male and female fer­til­ity in patients
a small minor­ity of patients can be offered HCT from an HLA- with SCD.42,43 The risk of sub­se­quent malig­nancy fol­low­ing HCT
iden­ti­cal fam­ily donor. remains small.41,44,45 Late effects of HCT include car­dio­vas­cu­lar,
pul­mo­nary, and endo­crine com­pli­ca­tions; dys­func­tion of the thy­
What is the best time to per­form HCT? roid gland, gonads, liver, and kid­neys; infer­til­ity; iron over­load;
Age at HCT is the most crit­ ic ­al deter­
mi­
nant of out­ come in bone dis­eases; infec­tion; malig­nancy; and neuropsychological
patients with SCD with an avail­­able HLA MSD. Patients receiv­ing effects.46 Most of these late effects have been stud­ied in patients

192  |  Hematology 2021  |  ASH Education Program


under­go­ing HCT for can­cers or bone mar­row fail­ure syn­dromes. Some patients or care­giv­ers would not con­sider HCT at any
There is a need to gen­er­ate data on the long-term out­come of level of risk.61,62 Almost three-fourths of patients report being
HCT for SCD to address this ele­ment of uncer­tainty in the deci­ will­ing to take a mod­est risk of ≥5% mor­tal­ity risk, whereas 57%
sion-mak­ing regard­ing HCT for SCD.47,48 are ready to take ≥10% risk of GVHD. Some par­ents and care­
giv­ers may also have dif­fi­culty mak­ing deci­sions about HCT for
Is HCT cost-effec­tive? and on behalf of a child who is cur­rently doing well and may
Although the cost-effec­tive­ness of HCT for SCD is still being eval­u­ have to deal with long-term sequelae of HCT.28 However, fol­
ated, early data sug­gest HCT is asso­ci­ated with decreased hos­pi­ low­ing suc­cess­ful HCT, par­ents do not report deci­sional regret
tal­i­za­tion, health care uti­li­za­tion, and opi­oid uti­li­za­tion.49 In adults even if their child has had severe HCT-related com­pli­ca­tions.59
with SCD, Saraf et al50 reported lower health care costs by the sec­ Thus, the deci­ sion to undergo HCT is highly indi­ vid­
u­al­
ized
ond year post-HCT (median of $16 281 vs $64 634 pre-HSCT [P = .01] and is driven by the per­cep­tion of dis­ease sever­ity; the val­ues
vs $54 082 in the stan­dard-of-care group [P = .05]). They reported and pref­er­ences of patients, care­giv­ers, and their phy­si­cians
a median reduc­tion of –$20 833/patient/y (interquartile range, regard­ing dis­ease-mod­i­fy­ing options; and acute and long-term
–$67 078 to +$4442/patient/y) in health care costs com­pared sequelae of HCT.28,63

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/190/1851641/190shah.pdf by guest on 13 December 2021


to pre-HCT in the sec­ond year post-HSCT (P = .05). In chil­dren,
Arnold et al,51 while reviewing 2 years pre- and post-HCT data, Summary and rec­om­men­da­tions
reported sig­nif­i­cant reduc­tions in admis­sions (P < .001), length of Young chil­dren with SCD have an excel­lent out­look for sur­vival to
stay (P < .001), and cost (P = .008) for HCT patients who received adult­hood, and new drugs may fur­ther improve out­comes. How-
their HCT before the age of 10 years. ever, many chil­ dren may have severe dis­ ease man­ i­
fes­
ta­
tions,
organ dam­age, and impaired qual­ity of life. Disease pro­gres­sion
Should we wait for gene ther­apy clin­i­cal tri­als or for gene may be unpre­dict­able in adult­hood, with severe con­se­quences
ther­apy to be approved instead of con­sid­er­ing allo­ge­neic for the patient. HCT from HLA MSDs in young chil­dren is asso­ci­
HCT? ated with excel­lent OS and EFS, sta­bi­li­za­tion of organ func­tion,
Early phase clin­i­cal tri­als of autol­o­gous trans­plan­ta­tion of hema­ decreased health care uti­li­za­tion, and improved qual­ity of life.
to­poi­etic pro­gen­i­tors mod­i­fied by gene addi­tion or gene edit- These ben­e­fits come at the cost of acute mor­bid­ity, the sub­stan­
ing hold the prom­ise of long-term dis­ease ame­lio­ra­tion with­out tial short-term bur­den of care, a small risk of HCT-related mor­tal­
the need for an allo­ge­neic donor and asso­ci­ated risk of GVHD. ity, late sequelae such as infer­til­ity, and a slight increase in the
Gene ther­apy stud­ies have so far enrolled patients 12 years or risk of sub­se­quent malig­nancy.
older. Future stud­ies may offer enroll­ment to youn­ger chil­dren. Furthermore, with increas­ing age, there is a decline in the
The stud­ies typ­i­cally exclude chil­dren who have an avail­­able HLA out­comes of HCT. Parents and care­giv­ers approach these com­
MSD. The cur­rent clin­i­cal hold of the Hgb-206 gene ther­apy trial plex trade-offs in ways unique to their life sit­u­a­tions and expe­
(ClinicalTrials.gov: NCT02140554) after 2 par­tic­i­pants devel­oped ri­ences and their per­cep­tion of dis­ease bur­den. We, there­fore,
acute mye­loid leu­ke­mia under­scores the fact that there remain rec­om­mend rou­tine HLA typ­ing of full sib­lings and con­sul­ta­tion
many unknowns in this exper­i­men­tal ther­apy.52-58 Thus, until a via­ with a phy­si­cian knowl­edge­able about HCT as 1 com­po­nent of
ble, safe strat­egy has been implemented for the gene ther­apy com­pre­hen­sive and sup­port­ive care, which includes the use of
approach, HCT is still con­sid­ered the best cura­tive option for cur­rent dis­ease-mod­i­fy­ing ther­a­pies. The com­plex­ity of deci­
young chil­dren with SCD if they have an avail­­able HLA MSD, par­ sion-mak­ing regard­ing HCT neces­si­tates a bal­anced dis­cus­sion
tic­u­larly with reduced-inten­sity/tox­ic­ity con­di­tion­ing reg­i­mens. of the deci­sion with the fam­ily in shared deci­sion-mak­ing about
There have been no com­par­a­tive tri­als of allo­ge­neic HCT and HCT. Communication about the patient between the hema­tol­
autol­o­gous gene ther­apy for any other dis­eases. Thus, fur­ther ogy and HCT teams is also likely to be ben­e­fi­cial. Families may
stud­ies are required to deter­mine the rel­a­tive safety and effi­cacy also ben­e­fit from a con­sul­ta­tion with sup­port groups and peers
of the 2 treat­ment modal­i­ties. who may have nav­i­gated a sim­i­lar deci­sional dilemma.
The 8-year-old patient described in the sec­ ond vignette
Are there indi­vid­ual dif­fer­ences in how patients and fam­i­lies illus­trates such a pro­cess. The patient had severe dis­ease man­
con­sider the infor­ma­tion and make deci­sions about HCT? i­fes­ta­tions and impaired qual­ity of life. Routine HLA typ­ing had
The deci­ sion for HCT for SCD involves com­ plex deci­ sional revealed the avail­abil­ity of an HLA MSD. The fam­ily engaged in
trade-offs. Parents decid­ing to pro­ceed with HCT face many the con­sul­ta­tive and deci­sional pro­cess and decided to pro­ceed
dilem­mas such as the unpre­dict­able onset and pro­gres­sion of with HCT. Following HCT from the HLA MSD, the child is cur­rently
SCD com­pli­ca­tions, repeated need to seek health care with lead­ing a nor­mal life.
SCD, pos­si­ble poor qual­ity of life with SCD, the immi­nence of The tod­dler in the first vignette is begin­ning to man­i­fest
mak­ing a major ther­a­peu­tic deci­sion, and con­cerns about the com­pli­ca­tions of SCD. The par­ents, how­ever, are ambiv­a­lent
long-term con­se­quences of the dis­ease. The feel­ing that the about HCT. We would rec­om­mend con­tinu­ing com­pre­hen­sive
dis­ease bur­den has become unac­cept­able is fre­quently cited med­i­cal care, includ­ing the addi­tion of dis­ease-mod­i­fy­ing ther­
as a trig­ger for the paren­tal deci­sion.28,59,60 The avail­abil­ity of an apy and sup­port for treat­ment adher­ence and psy­cho­so­cial
HLA MSD may be viewed as prov­i­den­tial by some par­ents. Par- func­tion­ing. If the fam­ily were to elect to pro­ceed with HCT in
ents vary greatly in their will­ing­ness to accept risks asso­ci­ated the future, we rec­om­mend that HCT be offered in the con­text
with HCT to achieve poten­tial ben­e­fits. HCT-asso­ci­ated mor­ of a mul­ti­cen­ter clin­i­cal trial. Active con­sid­er­ation must also be
bid­ity, includ­ing mucositis, GVHD, risk of death, and infer­til­ity, is given to pro­vid­ing fer­til­ity pres­er­va­tion pro­ce­dures pre-HCT
a sig­nif­i­cant rea­son that gives fam­i­lies pause in accepting HCT and long-term fol­low-up post-HCT to mon­i­tor for late effects
options. Overall, fam­i­lies face sub­stan­tial deci­sional con­flict.60 of treat­ment.
Prakash Singh Shekhawat
HLA iden­ti­cal donor HCT in young chil­dren  |  193
Conflict-of-inter­est dis­clo­sure ner­vous sys­tem sta­tus after bone mar­row trans­plan­ta­tion for sickle cell dis­
ease. Biol Blood Marrow Transplant. 2010;16(2):263-272.
Niketa Shah: no com­pet­ing finan­cial inter­ests to declare.
20. Hsieh MM, Kang EM, Fitzhugh CD, et al. Allogeneic hema­to­poi­etic stem-cell
Lakshmanan Krishnamurti: no com­pet­ing finan­cial inter­ests to trans­plan­ta­tion for sickle cell dis­ease. N Engl J Med. 2009;361(24):2309-
declare. 2317.
21. Gluckman E, Cappelli B, Bernaudin F, et al; Eurocord, the Pediatric Work-
ing Party of the Euro­pean Society for Blood and Marrow Transplantation,
Off-label drug use and the Center for International Blood and Marrow Transplant Research.
Niketa Shah: no off-label drug use has been discussed. Sickle cell dis­ease: an inter­na­tional sur­vey of results of HLA-iden­ti­cal sib­
Lakshmanan Krishnamurti: no off-label drug use has been ling hema­to­poi­etic stem cell trans­plan­ta­tion. Blood. 2017;129(11):1548-
discussed. 1556.
22. Cappelli B, Volt F, Tozatto-Maio K, et al; Eurocord, the Cellular Therapy and
Immunobiology Working Party (CTIWP) and the Paediatric Diseases Work-
Correspondence ing Party (PDWP) of the EBMT. Risk fac­tors and out­comes according to
age at trans­plan­ta­tion with an HLA-iden­ti­cal sib­ling for sickle cell dis­ease.
Lakshmanan Krishnamurti, Children’s Healthcare of Atlanta–
Haematologica. 2019;104(12):e543-e546.
Egleston, 1405 Clifton Road NE, Atlanta, GA 30322; e-mail: 23. Bernaudin F, Dalle J-H, Bories D, et al; Société Française de Greffe de Moelle

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/190/1851641/190shah.pdf by guest on 13 December 2021


lakshmanan​­.krishnamurti@choa​­.org; lakshmanan.krishnamurti@ et de Thérapie Cellulaire. Long-term event-free sur­vival, chi­me­rism and
emory.edu. fer­til­ity out­comes in 234 patients with sickle-cell ane­mia youn­ger than
30 years after myeloablative con­di­tion­ing and matched-sib­ling trans­plan­
ta­tion in France. Haematologica. 2020;105(1):91-101.
References 24. Brazauskas R, Scigliuolo GM, Wang H-L, et al. Risk score to pre­dict event-
1. Stenger EO, Shenoy S, Krishnamurti L. How I treat sickle cell dis­ease with free sur­ vival after hema­to­poi­
etic cell trans­ plant for sickle cell dis­ ease.
hema­to­poi­etic cell trans­plan­ta­tion. Blood. 2019;134(25):2249-2260. Blood. 2020;136(5):623-626.
2. Hassell KL. Population esti­ma­tes of sickle cell dis­ease in the U.S. Am J Prev 25. Bernaudin F, Verlhac S, Peffault de Latour R, et al; DREPAGREFFE Trial Inves-
Med. 2010;38(4, suppl):S512-S521. tigators. Association of matched sib­ling donor hema­to­poi­etic stem cell
3. Hassell KL. Sickle cell dis­ease: a con­tin­ued call to action. Am J Prev Med. trans­plan­ta­tion with transcranial doppler veloc­i­ties in chil­dren with sickle
2016;51(1, suppl 1):S1-S2. cell ane­mia. JAMA. 2019;321(3):266-276.
4. Salinas Cisneros G, Thein SL. Recent advances in the treat­ment of sickle 26. King AA, Kamani N, Bunin N, et al. Successful matched sib­ling donor mar­
cell dis­ease. Front Physiol. 2020;11:435. row trans­plan­ta­tion fol­low­ing reduced inten­sity con­di­tion­ing in chil­dren
5. Ware RE, Davis BR, Schultz WH, et al. Hydroxycarbamide ver­sus chronic with hemo­glo­bin­op­a­thies. Am J Hematol. 2015;90(12):1093-1098.
trans­fu­sion for main­te­nance of transcranial Dopp­ler flow veloc­i­ties in chil­ 27. Hsieh MM, Fitzhugh CD, Weitzel RP, et al. Nonmyeloablative HLA-matched
dren with sickle cell anae­mia—TCD With Transfusions Changing to Hydroxy- sib­ling allo­ge­neic hema­to­poi­etic stem cell trans­plan­ta­tion for severe sickle
urea (TWiTCH): a multicentre, open-label, phase 3, non-infe­ri­or­ity trial. cell phe­no­type. JAMA. 2014;312(1):48-56.
Lancet.2016;387(10019):661-670. 28. Sinha CB, Bakshi N, Ross D, Loewenstein G, Krishnamurti L. Primary care­
6. Ware RE, McGann PT, Quinn CT. Hydroxyurea for chil­dren with sickle cell giver deci­sion-mak­ing in hema­to­poi­etic cell trans­plan­ta­tion and gene ther­
ane­mia: pre­scribe it early and often. Pediatr Blood Cancer. 2019;66(8): apy for sickle cell dis­ease. Pediatr Blood Cancer. 2021;68(1):e28749.
e27778. 29. Walters MC, Patience M, Leisenring W, et  al. Collaborative mul­ti­cen­ter
7. Nouraie M, Darbari DS, Rana S, et al. Tricuspid regur­gi­ta­tion veloc­ity and inves­ti­ga­tion of mar­row trans­plan­ta­tion for sickle cell dis­ease: cur­rent
other bio­mark­ers of mor­tal­ity in chil­dren, ado­les­cents and young adults results and future direc­tions. Biol Blood Marrow Transplant. 1997;3(6):310-
with sickle cell dis­ease in the United States: the PUSH study. Am J Hematol. 315.
2020;95(7):766-774. 30. Smith WR, Bovbjerg VE, Penberthy LT, et  al. Understanding pain and
8. Blair HA. Voxelotor: first approval. Drugs. 2020;80(2):209-215. improv­ing man­age­ment of sickle cell dis­ease: the PiSCES study. J Natl Med
9. Wilmore DW. Food and Drug Administration approval of glu­ta­mine for Assoc. 2005;97(2):183-193.
sickle cell dis­ease: suc­cess and pre­cau­tions in glu­ta­mine research. JPEN J 31. Mentzer WC, Heller S, Pearle PR, Hackney E, Vichinsky E. Availability of
Parenter Enteral Nutr. 2017;41(6):912-917. related donors for bone mar­row trans­plan­ta­tion in sickle cell ane­mia. Am J
10. Carden MA, Little J. Emerging dis­ease-mod­i­fy­ing ther­a­pies for sickle cell Pediatr Hematol Oncol. 1994;16(1):27-29.
dis­ease. Haematologica. 2019;104(9):1710-1719. 32. Gragert L, Eapen M, Williams E, et al. HLA match like­li­hoods for hema­to­poi­
11. Lanzkron S, Carroll CP, Haywood C Jr. Mortality rates and age at death from etic stem-cell grafts in the U.S. reg­is­try. N Engl J Med. 2014;371(4):339-348.
sickle cell dis­ease: U.S., 1979-2005. Public Health Rep. 2013;128(2):110-116. 33. Angelucci E, Matthes-Martin S, Baronciani D, et al; EBMT Inborn Error and
12. DeBaun MR, Ghafuri DL, Rodeghier M, et  al. Decreased median sur­vival EBMT Paediatric Working Parties. Hematopoietic stem cell trans­plan­ta­tion
of adults with sickle cell dis­ease after adjusting for left trun­ca­tion bias: a in thal­as­se­mia major and sickle cell dis­ease: indi­ca­tions and man­age­ment
pooled anal­y­sis. Blood. 2019;133(6):615-617. rec­om­men­da­tions from an inter­na­tional expert panel. Haematologica.
13. Field JJ, DeBaun MR, Yan Y, Strunk RC. Growth of lung func­tion in chil­dren 2014;99(5):811-820.
with sickle cell ane­mia. Pediatr Pulmonol. 2008;43(11):1061-1066. 34. Badawy SM, Beg U, Liem RI, Chaudhury S, Thompson AA. A sys­tem­atic
14. Guasch A, Navarrete J, Nass K, Zayas CF. Glomerular involve­ment in adults review of qual­ity of life in sickle cell dis­ease and thal­as­se­mia after stem cell
with sickle cell hemo­glo­bin­op­a­thies: prev­a­lence and clin­i­cal cor­re­lates of trans­plant or gene ther­apy. Blood Adv. 2021;5(2):570-583.
pro­gres­sive renal fail­ure. J Am Soc Nephrol. 2006;17(8):2228-2235. 35. Gallo AM, Patil C, Adeniyi T, Hsu LL, Rondelli D, Saraf S. Health-related
15. Gladwin MT, Sachdev V, Jison ML, et  al. Pulmonary hyper­ ten­
sion as a qual­ity of life and per­sonal life goals of adults with sickle cell dis­ease after
risk fac­tor for death in patients with sickle cell dis­ease. N Engl J Med. hema­to­poi­etic stem cell trans­plan­ta­tion. West J Nurs Res. 2019;41(4):555-
2004;350(9):886-895. 575.
16. Vichinsky EP, Neumayr LD, Gold JI, et al; Neuropsychological Dysfunction 36. Saraf SL, Oh AL, Patel PR, et al. Nonmyeloablative stem cell trans­plan­ta­
and Neuroimaging Adult Sickle Cell Anemia Study Group. Neuropsycho- tion with alemtuzumab/low-dose irra­di­a­tion to cure and improve the qual­
logical dys­func­tion and neuroimaging abnor­mal­i­ties in neu­ro­log­i­cally ity of life of adults with sickle cell dis­ease. Biol Blood Marrow Transplant.
intact adults with sickle cell ane­mia. JAMA. 2010;303(18):1823-1831. 2016;22(3):441-448.
17. Shah N, Bhor M, Xie L, et  al. Treatment pat­terns and eco­nomic bur­den 37. Krishnamurti L, Neuberg DS, Sullivan KM, et al. Bone mar­row trans­plan­
of sickle-cell dis­ ease patients pre­ scribed hydroxy­urea: a ret­ ro­spec­ tive ta­tion for ado­les­cents and young adults with sickle cell dis­ease: results
claims-based study. Health Qual Life Outcomes. 2019;17(1):155. of a pro­spec­tive mul­ti­cen­ter pilot study. Am J Hematol. 2019;94(4):446-
18. Walters MC, Patience M, Leisenring W, et al. Bone mar­row trans­plan­ta­tion 454.
for sickle cell dis­ease. N Engl J Med. 1996;335(6):369-376. 38. Arnold SD, Bhatia M, Horan J, Krishnamurti L. Haematopoietic stem cell trans­
19. Walters MC, Hardy K, Edwards S, et al; Multicenter Study of Bone Marrow plan­ta­tion for sickle cell dis­ease—cur­rent prac­tice and new approaches.
Transplantation for Sickle Cell Disease. Pulmonary, gonadal, and cen­tral Br J Haematol. 2016;174(4):515-525.

194  |  Hematology 2021  |  ASH Education Program


39. Bhatia M, Kolva E, Cimini L, et al. Health-related qual­ity of life after allo­ge­ 52. Brendel C, Williams DA. Current and future gene ther­a­pies for hemo­glo­
neic hema­to­poi­etic stem cell trans­plan­ta­tion for sickle cell dis­ease. Biol bin­op­a­thies. Curr Opin Hematol. 2020;27(3):149-154.
Blood Marrow Transplant. 2015;21(4):666-672. 53. Curtis SA, Shah NC. Gene ther­apy in sickle cell dis­ease: pos­si­ble util­ity and
40. Brodigan K, Kapadia M, Frazier AL, et al. Safety of sur­gi­cal fer­til­ity pres­er­ impact. Cleve Clin J Med. 2020;87(1):28-29.
va­tion pro­ce­dures in chil­dren prior to hema­to­poi­etic stem cell trans­plant. 54. Demirci S, Uchida N, Tisdale JF. Gene ther­apy for sickle cell dis­ease: an
Transplant Cell Ther. 2021;27(8):696.e1-696696.e4. update. Cytotherapy. 2018;20(7):899-910.
41. Lautz TB, Harris CJ, Laronda MM, Erickson LL, Rowell EE. A fer­til­ity pres­er­ 55. Frangoul H, Altshuler D, Cappellini MD, et al. CRISPR-Cas9 gene editing for
va­tion toolkit for pedi­at­ric sur­geons car­ing for chil­dren with can­cer. Semin sickle cell dis­ease and β-thal­as­se­mia. N Engl J Med. 2021;384(3):252-260.
Pediatr Surg. 2019;28(6):150861. 56. Ikawa Y, Miccio A, Magrin E, Kwiatkowski JL, Rivella S, Cavazzana M. Gene
42. Joseph L, Jean C, Manceau S, et al. Effect of hydroxy­urea expo­sure before ther­apy of hemo­glo­bin­op­a­thies: prog­ress and future chal­lenges. Hum Mol
puberty on sperm param­e­ters in males with sickle cell dis­ease. Blood. Genet. 2019;28(R1):R24-R30.
2021;137(6):826-829. 57. Jones RJ, DeBaun MR. Leukemia after gene ther­apy for sickle cell dis­ease:
43. Pecker LH, Hussain S, Christianson MS, Lanzkron S. Hydroxycarbamide inser­tional muta­gen­e­sis, busul­fan, both, or nei­ther. Blood. 2021;138(11):942-
expo­sure and ovar­ian reserve in women with sickle cell dis­ease in the Mul- 947.
ticenter Study of Hydroxycarbamide. Br J Haematol. 2020;191(5):880-887. 58. Leonard A, Tisdale J, Abra­ham A. Curative options for sickle cell dis­ease:
44. Kahn JM, Brazauskas R, Tecca HR, et  al. Subsequent neo­plasms and haploidentical stem cell trans­plan­ta­tion or gene ther­apy? Br J Haematol.
late mor­tal­ity in chil­dren under­go­ing allo­ge­neic trans­plan­ta­tion for 2020;189(3):408-423.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/190/1851641/190shah.pdf by guest on 13 December 2021


non­ma­lig­nant dis­eases. Blood Adv. 2020;4(9):2084-2094. 59. Khemani K, Ross D, Sinha C, Haight A, Bakshi N, Krishnamurti L. Experi-
45. Ghannam JY, Xu X, Maric I, et al. Baseline TP53 muta­tions in adults with SCD ences and deci­sion mak­ing in hema­to­poi­etic stem cell trans­plant in sickle
devel­op­ing mye­loid malig­nancy fol­low­ing hema­to­poi­etic cell trans­plan­ta­ cell dis­ease: patients’ and care­giv­ers’ per­spec­tives. Biol Blood Marrow
tion. Blood. 2020;135(14):1185-1188. Transplant. 2018;24(5):1041-1048.
46. Inamoto Y, Lee SJ. Late effects of blood and mar­row trans­plan­ta­tion. Hae- 60. Schulz GL, Kelly KP, Holtmann M, Armer JM. Navigating deci­sional con­flict
matologica. 2017;102(4):614-625. as a fam­ily when fac­ing the deci­sion of stem cell trans­plant for a child or
47. Shenoy S, Angelucci E, Arnold SD, et al. Current results and future research ado­les­cent with sickle cell dis­ease. Patient Educ Couns. 2021;104(5):1086-
pri­or­i­ties in late effects after hema­to­poi­etic stem cell trans­plan­ta­tion for 1093.
chil­dren with sickle cell dis­ease and thal­as­se­mia: a con­sen­sus state­ment 61. van Besien K, Koshy M, Anderson-Shaw L, et al. Allogeneic stem cell trans­
from the sec­ond pedi­at­ric blood and mar­row trans­plant con­sor­tium inter­ plan­ta­tion for sickle cell dis­ease: a study of patients’ deci­sions. Bone Mar-
na­tional con­fer­ence on late effects after pedi­at­ric hema­to­poi­etic stem cell row Transplant. 2001;28(6):545-549.
trans­plan­ta­tion. Biol Blood Marrow Transplant. 2017;23(4):552-561. 62. Kodish E, Lantos J, Stocking C, Singer PA, Siegler M, Johnson FL. Bone mar­
48. Shenoy S, Gaziev J, Angelucci E, et  al. Late effects screen­ing guide­lines row trans­plan­ta­tion for sickle cell dis­ease: a study of par­ents’ deci­sions.
after hema­to­poi­etic cell trans­plan­ta­tion (HCT) for hemo­glo­bin­op­a­thy: con­ N Engl J Med. 1991;325(19):1349-1353.
sen­sus state­ment from the sec­ond pedi­at­ric blood and mar­row trans­plant 63. Bakshi N, Katoch D, Sinha CB, et al. Assessment of patient and care­giver atti­
con­sor­tium inter­na­tional con­fer­ence on late effects after pedi­at­ric HCT. tudes and approaches to deci­sion-mak­ing regard­ing bone mar­row trans­
Biol Blood Marrow Transplant. 2018;24(7):1313-1321. plant for sickle cell dis­ease: a qual­i­ta­tive study. JAMA Netw Open. 2020;3(5):
49. Hsieh MM, Fitzhugh CD, Tisdale JF. Allogeneic hema­to­poi­etic stem cell trans­ e206742.
plan­ta­tion for sickle cell dis­ease: the time is now. Blood. 2011;118(5):1197-
1207.
50. Saraf SL, Ghimire K, Patel P, et al. Improved health care uti­li­za­tion and costs
in transplanted ver­sus non-transplanted adults with sickle cell dis­ease.
PLoS One. 2020;15(2):e0229710.
51. Arnold SD, Brazauskas R, He N, et al. Clinical risks and healthcare uti­li­za­
tion of hema­to­poi­etic cell trans­plan­ta­tion for sickle cell dis­ease in the USA © 2021 by The Amer­i­can Society of Hematology
using merged data­bases. Haematologica. 2017;102(11):1823-1832. DOI 10.1182/hema­tol­ogy.2021000322

Prakash Singh Shekhawat


HLA iden­ti­cal donor HCT in young chil­dren  |  195
EMERGING THERAPIES AND CONSIDERATIONS FOR SICKLE CELL DISEASE

Clinical trial considerations in sickle cell disease:


patient-reported outcomes, data elements,
and the stakeholder engagement framework

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/196/1851725/196badawy.pdf by guest on 13 December 2021


Sherif M. Badawy
Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL; and Division of Hematology, Oncology, and Stem Cell
Transplant, Ann and Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL

Patients with sickle cell disease (SCD) have significant impairment in their quality of life across the life span as a con-
sequence of serious disease burden with several SCD-related complications. A number of disease-modifying therapies
are currently available, yet long-term clinical benefits in real-world settings remain unclear. Over the past few years,
a number of important initiatives have been launched to optimize clinical trials in SCD in different ways, including:
(1) established panels through a partnership between the American Society of Hematology (ASH) and the US Food and
Drug Administration; (2) the ASH Research Collaborative SCD Clinical Trials Network; (3) the PhenX Toolkit (consensus
measures for Phenotypes and eXposures) in SCD; and (4) the Cure Sickle Cell Initiative, led by the National Heart, Lung,
and Blood Institute. Electronic patient-reported outcomes assessment is highly recommended, and patient-reported
outcomes (PROs) should be evaluated in all SCD trials and reported using Standard Protocol Items Recommendations
for Interventional Trials guidelines. Patient-centered outcomes research (PCOR) approaches and meaningful stakeholder
engagement throughout the process have the potential to optimize the execution and success of clinical trials in SCD
with considerable financial value. This article reviews several clinical trial considerations in SCD related to study design
and outcomes assessment as informed by recent initiatives as well as patient-centered research approaches and stake-
holder engagement. A proposed hematology stakeholder-engagement framework for clinical trials is also discussed.

LEARNING OBJECTIVES
• Review key considerations for SCD clinical trials related to PROs, medication adherence, developmental issues in
children, and the COVID­19 pandemic
• Review efforts to optimize clinical trials and outcomes assessment in SCD, such as ASH­FDA panels, the ASH Re­
search Collaborative Clinical Trials Network, the PhenX Toolkit, and the CureSCi
• Review the evidence for patient­centered research (PCR) and stakeholder engagement and their potential role in
successful trials and cost savings

CLINICAL CASE sickle cell research to help other sickle cell patients bene­
A 19­year­old man with hemoglobin SS disease presents ft from these therapies.
for his regular clinic visit. He has had no hospitalizations
over the past 5 years since he started taking daily hydroxy­
urea with good adherence. He believes that hydroxyurea Introduction
helped him a great deal with his quality of life, but he also Sickle cell disease (SCD) is an inherited hemoglobin dis­
understands that not every sickle cell patient feels the order affecting about 100 000 individuals in the United
same. He has learned about other approved and emerg­ States and more than 20 million people worldwide, mainly
ing therapies, and he is fascinated by the science behind of African descent.1,2 SCD is a chronic, debilitating medical
them. Given his interest in pursuing a career in medicine, condition that affects patients across their life span and is
he inquires about the possibility of getting involved in associated with signifcant morbidity and early mortality.2,3

196 | Hematology 2021 | ASH Education Program


SCD patients suf­fer from a num­ber of acute and chronic com­ and the Tobacco Regulatory Science Program of the National
pli­ca­tions, includ­ing pain epi­sodes, acute chest syn­drome, car­ Institutes of Health (NIH). In SCD, PhenX efforts have focused
dio­pul­mo­nary dis­ease, kid­ney dam­age, liver impair­ment, splenic on selecting high-qual­ity SCD-related out­come mea­sures to be
seques­ tra­
tion, avas­
cu­lar necro­ sis, stroke, priapism, and other included in the Toolkit (con­sen­sus mea­sures; www​­.phenxtoolkit​
end organ dam­age.2,3 These com­pli­ca­tions lead to sig­nif­i­cant ­.org), guided by the Sickle Cell Disease Research and Scientific
impair­ment in patient-reported out­comes (PROs) among chil­ Panel.19 Finally, the Cure Sickle Cell Initiative (CureSCi), led by
dren and adults with SCD, espe­cially in the phys­i­cal and psy­cho­ the NHLBI, has cen­tered on inno­vat­ing genetic ther­a­pies, nur­
so­cial domains.4-6 Patients with SCD have increased health care tur­ing a col­lab­o­ra­tive, PCR envi­ron­ment, and establishing data
uti­li­za­tion with fre­quent hos­pi­tal­i­za­tions and emer­gency depart­ stan­dards for SCD clin­i­cal tri­als.20 In par­tic­u­lar, inter­est in cura­tive
ment vis­its.7-9 Treatment approaches include pre­ven­tive strat­e­ ther­a­pies in the SCD com­mu­nity has been grow­ing as evi­dence
gies (eg, pen­i­cil­lin pro­phy­laxis, transcranial Dopp­ler screen­ing), has emerged and con­tin­ues to mate­ri­al­ize from ongo­ing clin­i­cal
acute man­age­ment (eg, opi­oids), dis­ease-mod­i­fy­ing ther­a­pies tri­als.
(ie, hydroxy­urea, L-glu­ta­mine, voxelotor, and crizanlizumab), and This arti­cle aims to review sev­eral clin­i­cal trial con­sid­er­ations

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/196/1851725/196badawy.pdf by guest on 13 December 2021


cura­tive options (eg, hema­to­poi­etic stem cell trans­plan­ta­tion, in SCD related to study design and out­ come assess­ ment as
gene ther­apy, or gene editing).3 informed by recent ini­tia­tives, in par­tic­u­lar PROs as well as PCR
Some ear­lier clin­i­cal tri­als in SCD faced a num­ber of logis­ti­ approaches and stake­holder engage­ment.
cal chal­lenges related to recruit­ment and reten­tion.10-12 Barriers
to suc­cess­ful trial com­ple­tion included con­cerns from par­ents Clinical trial con­sid­er­ations
as to the neces­sity of the research, patient belief that research PROs and e-PROs
was only needed for those with more severe dis­ease, or anx­i­ety PROs have been defined as “out­ comes reported directly by
related to pre­vi­ous research expe­ri­ence.11-13 A num­ber of facil­i­ta­ patients them­ selves and not interpreted by an observer.”21
tors were also iden­ti­fied from these ear­lier stud­ies, includ­ing edu­ Health-related qual­ity of life (HRQOL) is a PRO that is defined as
cat­ing peers, explaining trial ratio­nales more clearly, ­improv­ing “a mul­ti­di­men­sional con­cept that usu­ally includes self-report of
the read­abil­ity of con­sent/assent forms, explaining study pro­to­ the way in which phys­i­cal, emo­tional, social, or other domains
cols using vid­eos and other inno­va­tive illus­tra­tions, and leverag­ of well-being are affected by a dis­ease or its treat­ment.”21 A
ing patient-cen­ tered research (PCR) approaches that involve proxy- or par­ent/care­giver-reported out­come is also com­monly
patients, par­ents/care­giv­ers and other stake­hold­ers (here­to­fore used to eval­u­ate PROs and/or HRQOL among pedi­at­ric pop­u­
referred to as “stake­hold­ers”) across all­stages of the research la­tions. Major reg­u­la­tory author­i­ties have rec­og­nized the value
pro­cess, from plan­ning a study to the dis­sem­i­na­tion of find­ings.11-13 of includ­ing PROs eval­u­a­tion in clin­i­cal tri­als to inform clin­i­cal
Despite some ini­tial dif­fi­cul­ties in research, sev­eral tri­als in SCD deci­sion-mak­ing, phar­ma­ceu­ti­cal-label­ing claims, and prod­uct
to date have been suc­cess­fully com­pleted, lead­ing to sig­nif­i­cant reim­burse­ment.22 The inclu­sion of PROs in clin­i­cal trial pro­to­cols
prog­ress in the field of SCD with new ther­a­pies now approved should be well planned in advance and reported according to
by the US Food and Drug Administration (FDA), pro­vid­ing clin­i­ the Standard Protocol Items Recommendations for Interven­
cal ben­e­fits to many SCD patients. Currently, there are sev­eral tional Trials guide­lines.23 Evaluating PROs in clin­i­cal tri­als that
active, ongo­ing clin­i­cal tri­als with novel dis­ease-mod­i­fy­ing ther­ involve SCD patients pro­vi­des an oppor­tu­nity to mea­sure the
a­pies and cura­tive approaches, such as gene ther­apy, gene edit­ impact of a given treat­ment on their indi­vid­ual func­tion­ing and
ing, and hema­to­poi­etic stem cell trans­plan­ta­tion with var­i­ous well-being.5
reg­i­mens and donors.14,15 Nevertheless, efforts to improve clin­i­cal A num­ber of fac­tors should be con­sid­ered when assessing
tri­als, includ­ing involv­ing stake­hold­ers in tri­als, are still ongo­ing. PROs in SCD clin­i­cal tri­als, such as eli­gi­bil­ity cri­te­ria (eg, age), rel­e­
Over the past few years, a num­ber of impor­tant ini­tia­tives vant domains of inter­est, psy­cho­met­ric prop­er­ties (eg, respon­sive­
have been launched to opti­mize clin­i­cal tri­als in SCD in dif­fer­ ness, validity, reli­abil­ity, and floor/ceil­ing effects), min­i­mal clin­i­cally
ent ways. First, con­sen­sus rec­om­men­da­tions for evi­dence-based impor­tant dif­fer­ences, generic vs dis­ease-spe­cific approaches,
SCD end points have been devel­oped as a result of a col­lab­o­ par­tic­i­pants’ bur­den (ie, sur­vey length), and mode of admin­is­tra­
ra­tive effort from 7 pan­els of patients, cli­ni­cians, and research­ tion.5 Generic mea­sures pro­vide insight into the bur­den of SCD
ers established through a part­ner­ship between the Amer­i­can com­pared to healthy indi­vid­u­als and those with other chronic
Society of Hematology (ASH) and the FDA.16,17 Second, the ASH med­i­cal con­di­tions.5 On the other hand, dis­ease-spe­cific mea­
Research Collaborative SCD Clinical Trials Network has focused sures bet­ter exam­ine the dif­fer­ences and effects of treat­ments or
on build­ing part­ner­ships with the SCD com­mu­nity and stake­ inter­ven­tions across dif­fer­ent patient groups and within indi­vid­
hold­ers, establishing col­lab­o­ra­tion across SCD cen­ters, stream­ ual SCD patients.5 Thus, a com­bined approach using generic and
lining clin­i­cal trial oper­a­tions with a sin­gle insti­tu­tional review dis­ease-spe­cific instru­ments to eval­u­ate SCD patients’ PROs is
board approval, and facil­i­tat­ing data shar­ing with a cen­tral­ized highly recommended, with a pref­er­ence for patient self-report­
data repos­i­tory through the ASH Research Collaborative Data ing over proxy reporting when pos­si­ble.4,5 Further, the ASH-FDA
Hub.18 Third, the PhenX (Phenotypes and eXposures) pro­ject has panel for PROs has pro­vided detailed guid­ance and rec­om­men­
been funded by dif­fer­ent sources, includ­ing the National Human da­tions on PROs selec­tion in SCD clin­i­cal tri­als (Table 1).16
Genome Research Institute, the National Institute on Drug Finally, the mode of PROs eval­u­a­tion is another key con­sid­
Abuse, the Office of Behavioral and Social Sciences Research, er­ation. The use of elec­tronic approaches or e-PROs has been
the National Institute of Mental Health, the National Heart, Lung, recommended by the e-PRO con­sor­tiums of the International
and Blood Institute (NHLBI), the National Institute on Minority Society for Quality of Life Research, the Professional Society for
Health and Health Disparities, the National Cancer Institute, Health Economics and Outcomes Research, and the reg­u­la­tors

Prakash Singh Shekhawat


Clinical trial con­sid­er­ations in SCD  |  197
Table 1. PROs rec­om­men­da­tions in CureSCi CDEs (ver­sion 1.0)

Subdomain Population Measure Classification


Pain inten­sity Adults and chil­dren ≥8 years old NRS Core
VAS Supplemental
Pain impact/inter­fer­ence Adults, SCD-spe­cific ASCQ-Me Pain Impact Core
Children 5-18 years, SCD-spe­cific PedsQL Pain Impact SCD mod­ule Core
Children/adults, not SCD-spe­cific PROMIS Pain Interferencea,b Core
Pain: mixed Children 5-18 years, SCD-spe­cific PedsQL Pain and Hurt, SCD mod­ules Core
Painful cri­ses Adults, SCD-spe­cific ASCQ-Me Pain Episodes Core
Emotional impact of SCD Adults, SCD-spe­cific ASCQ-Me Emotional Impact Supplemental, highly recommended

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/196/1851725/196badawy.pdf by guest on 13 December 2021


Children, SCD-spe­cific PedsQL, SCD Module Emotions Supplemental, highly recommended
PedsQL, SCD Module Worrying Supplemental
Negative affect: mixed Children, not SCD-spe­cific PROMIS Physical Stress Experience1 Supplemental, highly recommended
Low mood Children/adults, not SCD-spe­cific PROMIS Emotional Distress: Depression 1,2
Supplemental, highly recommended
Anxiety Children/adults, not SCD-spe­cific PROMIS Emotional Distress: Anxiety1,2 Supplemental, highly recommended
Fatigue Children/adults, not SCD-spe­cific Pediatric/Adult PROMIS Fatigue1,2 Core
Children, SCD-spe­cific PedsQL Multidimensional Fatigue Scale Core
Sleep dis­tur­bance Children/adults, not SCD-spe­cific PROMIS Sleep Disturbance1,2 Supplemental, highly recommended
Adults, SCD-spe­cific ASCQ-Me Sleep Impact Supplemental, highly recommended
Adults, not SCD-spe­cific Pittsburgh Sleep Quality Index Supplemental
Epworth Sleepiness Scale Supplemental
Children, not SCD-spe­cific Epworth Sleepiness Scale (CHAD) Supplemental
General func­tion Adults, not SCD-spe­cific Cana­dian Occupational Performance Supplemental
Social func­tion Adults, SCD-spe­cific ASCQ-Me Social Functioning Impact Supplemental
Physical func­tion Adults, SCD-spe­cific ASCQ-Me Stiffness Impact Supplemental, highly recommended
Adults, not SCD-spe­cific PROMIS - Physical Function (PF) 12a2 Supplemental
Children, not SCD-spe­cific Pediatric PROMIS - PF Mobility1 Supplemental
Pediatric PROMIS - PF Upper Extremity1 Supplemental
Global health/QOL Adults, not SCD-spe­cific PROMIS 10 Global Health 2
Core
Children, not SCD-spe­cific PROMIS 7 + 2 Global Health* Core
Global cog­ni­tion Children, 0-3.5 years old Bayley-III Supplemental
Children, 2.5-7 years old WPPSI-IV (4th edi­tion) Supplemental
Children, 6-16 years old WISC-V (5th edi­tion) Supplemental
Adults Wechsler Adult Intelligence Scale Supplemental
Children and adults† NIH Toolbox Supplemental, highly recommended
Executive func­tion­ing Children 3-7, 8-11, and ≥12 years Flanker Inhibitory Control/Attention (NT) Supplemental, highly recommended
Dimensional Change Card Sort Test (NT) Supplemental, highly recommended
Children ≥9 years old Trail Making Test, parts A and B Supplemental
Children and adults, 8-89 years Delis-Kaplan Executive Function System Supplemental
Children and adults, 7-89 years Wisconsin Card Sort Test Supplemental
Processing speed Children ≥7 years old Pattern Comparison Processing Speed Test Supplemental, highly recommended
Adults Processing Speed Index Supplemental
Working mem­ory Children ≥7 years old List Sorting Working Memory Test (NT) Supplemental, highly recommended
*Pediatric PROMIS mea­sures are avail­­able for chil­dren self-report ≥8 years old and proxy report.
†Adult PROMIS mea­sures are avail­­able.
ASCQ-Me, Adult Sickle Cell Quality of Life Measurement Information System; CHAD, children and adolescents; NRS, Numeric Rating Scale; NT, NIH
Toolbox; PROMIS, Patient-Reported Outcomes Measurement Information System; VAS, Visual Analog Scale; WISC, Wechsler Intelligence Scale for
Children; WPPSI-IV, Wechsler Preschool and Primary Scale of Intelligence.
Publicly available at https:​­/​­/curesickle​­.org​­/sites​­/scdc​­/files​­/Doc​­/SC​­/Patient_Reported_Outcomes_Recommendations_Summary​­.pdf.

198  |  Hematology 2021  |  ASH Education Program


(eg, the FDA).23-25 e-PROs have the fol­low­ing advan­tages: (1) more shar­ing, and edu­cate young inves­ti­ga­tors on var­i­ous aspects of
pre­cise, com­plete, timely, and high-qual­ity data, (2) bet­ter adher­ clin­i­cal research meth­od­ol­ogy.20 Table 2 includes an over­view of
ence to study pro­to­col, (3) pos­si­ble PRO remind­ers and real- the pro­posed CDEs in CureSCi. In addi­tion, all­core data ele­ments
time mon­i­tor­ing, (4) less recall bias, (5) fewer data entry errors, that are essen­tial for the ini­ti­a­tion of any clin­i­cal research study
(6) lev­er­aged com­put­er­ized adap­tive test­ing when needed, in SCD are included in a Start-Up Resource Listing doc­u­ment.20
(7) inte­ grated skip pat­ terns for rel­e­vant ques­tions, (8) ligh­
ter The PhenX Toolkit for SCD is another key NHLBI-funded ini­tia­tive.
work­loads for staff, (9) pos­si­ble cost sav­ings and envi­ron­men­tal The goal of the PhenX Measures for SCD Research pro­ject is to
friend­li­ness with less paper print­ing, and (10) high accept­abil­ity help research­ers bet­ter under­stand the path­o­phys­i­­ol­ogy, nat­u­ral
rat­ings from patients.5,24,26 Given the ubiq­ui­tous access to smart­ his­tory, and treat­ment approaches for SCD. The PhenX Toolkit in
phones and tab­lets as well as the grow­ing evi­dence and accept­ SCD is a frame­work for out­comes assess­ment and data shar­ing
abil­ity of mobile health inter­ven­tions among SCD patients,27,28 across var­i­ous SCD research pro­jects that allows for poten­tial
e-PROs should be strongly con­ sid­
ered in SCD clin­ i­
cal tri­
als, com­par­i­sons across stud­ies.19
whether pro­vid­ing patients with a ded­i­cated device for e-PROs

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/196/1851725/196badawy.pdf by guest on 13 December 2021


or allowing patients to down­ load an app and use their own Developmental issues in pedi­at­ric tri­als
phone—an approach we call “Bring your own device,” or BYOD. Some issues should be con­ sid­
ered when plan­ ning out­ comes
assess­ment in an SCD clin­i­cal trial that involves chil­dren and
Medication adher­ence ado­les­cents, such as age-appro­pri­ate psy­cho­met­ric prop­er­ties
Adherence to any new med­i­ca­tion is a crit­i­cal com­po­nent of the for PROs, patient or proxy reports or both, and devel­op­men­tal
suc­cess of any clin­i­cal trial in SCD, yet often lit­tle atten­tion is level.12 Children and ado­les­cents expe­ri­ence many cog­ni­tive,
given to ways to mon­i­tor and opti­mize adher­ence dur­ing the psy­cho­log­i­cal, and phys­i­cal changes over time and show wide
course of a study. A num­ber of objec­tive and sub­jec­tive adher­ var­i­abil­ity in their emo­tional, social, atten­tional, and intel­lec­tual
ence mea­sures can be con­sid­ered in the set­ting of a clin­i­cal lev­els of devel­op­ment.12 These dif­fer­ences have impor­tant impli­
trial, which might vary based on the study design (eg, ran­dom­ ca­tions for pedi­at­ric clin­i­cal trial design and implementation,
ized con­trolled trial vs real-world com­par­a­tive effec­tive­ness espe­cially in behav­ioral and interventional areas, where a one-
trial). Objective mea­ sures of med­ i­
ca­
tion adher­ ence include size-fits-all­approach is far from ideal.
bio­chem­i­cal mea­sures (eg, drug lev­els, bio­mark­ers), elec­tronic
mon­i­tor­ing (eg, elec­tronic pill bot­tles or smartphone app logs), Patient-cen­tered research and stake­holder-engage­ment
directly observed ther­apy (ie, in-per­son or mobile), dig­i­tal pills frame­work in hema­tol­ogy
(eg, Pro­teus), pill counts, and phar­macy records (eg, pre­scrip­ Stakeholder involve­ment in dif­fer­ent stages of sickle cell research
tion refills).29 These mea­sures pro­vide a more accu­rate view of has been lim­ited, includ­ing in devel­op­ment, design, implemen­
a patient’s adher­ ence behav­ ior but require some addi­ tional tation, and dis­sem­i­na­tion. Most clin­i­cal tri­als in SCD have his­tor­
resources. In con­trast, sub­jec­tive mea­sures of med­i­ca­tion adher­ i­cally focused on sur­ro­gate end points, such as hos­pi­tal­i­za­tions
ence include a patient’s self-reported adher­ence, using sur­veys and emer­gency room vis­its, as mark­ers of dis­ease activ­ity, with
(eg, paper and pen­cil or elec­tronic) or inter­views, and phy­si­cian less empha­sis on PROs or patient-cen­tered out­comes research
assess­ments.29 These mea­sures are sim­ple, short, and inex­pen­ (PCOR). The prob­lem with this approach is the pos­si­bil­ity of
sive and can pro­vide insight into poten­tial adher­ence bar­ri­ers; miss­ing what patients and other stake­hold­ers care about the
nev­ er­the­ less, social desir­ abil­
ity and recall bias are impor­ tant most, mak­ing clin­i­cal trial find­ings less rel­e­vant to many of them,
con­sid­er­ations. It is worth not­ing that recent col­lab­o­ra­tive, mul­ at least in their view. The Patient-Centered Outcomes Research
ti­dis­ci­plin­ary efforts led to the devel­op­ment of the PROMIS Med­ Institute (PCORI) was established by the US Congress in 2010
ication Adherence Scale (PMAS), and its psy­cho­met­ric eval­u­a­tion to address this issue. Since then, PCORI has funded sev­eral SCD
is under­way in sev­eral ongo­ing tri­als.30 PMAS is listed in C
­ ureSCi’s pro­jects at dif­fer­ent stages and with a wide range of bud­gets
com­mon data ele­ments (CDEs). Given that both objec­tive and and scopes (Table 3). Other gov­ern­ment agencies also sup­port
sub­jec­tive mea­sures of adher­ence have the poten­tial to cap­ture PCOR pro­jects with var­i­ous lev­els of expected patient and stake­
var­i­ous aspects of med­i­ca­tion-tak­ing behav­ior, a mul­ti­modal holder involve­ ment (Table 4). The FDA Patient-Focused Drug
strat­ egy is highly recommended.29,31 Further, in ran­ dom­ ized Development ini­tia­tive is another key effort to include patients’
con­trolled tri­als eval­u­at­ing the effi­cacy of a new med­i­ca­tion in per­spec­tives on their med­i­cal con­di­tions, the symp­toms that
SCD, it is likely advan­ta­geous to use tools that can mon­i­tor and have the most impact on their daily lives, and the avail­­able ther­
enhance adher­ence behav­ior to opti­mize the clin­i­cal ben­e­fits of a­pies and to bet­ter under­stand the fac­tors that drive treat­ment
a given ther­apy for study par­tic­i­pants. In addi­tion, this can pro­ deci­sions and a will­ing­ness to par­tic­i­pate in clin­i­cal tri­als.25 More­
vide a more pre­cise assess­ment of the dif­fer­ences in study out­ over, PCOR high­ lighted impor­ tant out­comes that were often
comes based on expo­sure or adher­ence to either exper­i­men­tal overlooked by inves­ti­ga­tors, such as patient- or proxy-reported
drug vs pla­cebo or active com­par­a­tor. PROs, treat­ment sat­is­fac­tion, care­giver/par­ent bur­den, work
time off, transportation costs, and out-of-pocket costs.
CDEs (CureSCi and PhenX Toolkit for SCD) PCOR pro­ jects in SCD most often focus on inves­ ti­
ga­
tor-
One of the goals of the NHLBI-led CureSCi is to stan­dard­ize data ini­ti­ated com­par­a­tive effec­tive­ness tri­als eval­u­at­ing dif­fer­ent
col­lec­tion forms for all­clin­i­cal research stud­ies in SCD, includ­ing established treat­ment approaches. Involving patients and stake­
those with prom­is­ing genetic approaches.20 In 2021 the first set hold­ers in clin­i­cal trial deci­sions, using mea­sures such as PROs
of CDEs were assem­bled and final­ized. These CDEs serve as a and other out­comes, is crit­i­cal to ensure the rel­e­vance of these
crit­i­cal resource for SCD clin­i­cal tri­als in the effort to improve the assess­ments to the larger SCD com­mu­nity. Moreover, in 2017
effi­ciency of clin­i­cal stud­ies, enhance data qual­ity, enable data the Clinical Trials Transformation Initiative (CTTI) outlined, by
Prakash Singh Shekhawat
Clinical trial con­sid­er­ations in SCD  |  199
Table 2. NIH-NHLBI CureSCi CDEs (ver­sion 1.0)

Domain Subdomain Class Recommendations


Participant char­ac­ter­is­tics Demographics C Demographics
Baseline abnor­mal hema­to­poi­e­sis Behavioral his­tory short form
C
Transfusion his­tory Medical his­tory Surgical his­tory
General health his­tory
Behavioral his­tory Medical history supplemental elements
S
Hospitalization form Sleep assess­ment (ped form)
C Social sta­tus
Education school ques­tion­naire Social deter­mi­nants screen
Social his­tory
S ACEs screen chil­dren (1-17 years) ACEs screen adults (  ≥18 years)

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/196/1851725/196badawy.pdf by guest on 13 December 2021


Acute ane­mia Chronic ane­mia
Disease and treat­ment-
Asthma out­comes instru­ment rec­om­men­da­tions (highly recommended)
related events Asthma S
Asthma out­comes Over-read spi­rom­e­try report form
Fertility/bone C Endocrine, infer­til­ity, and bone health
6-min­ute walk test Pulmonary func­tion test
C
Lung dis­ease assess­ments guide­lines
Lung
PROMIS dyspnea func­tional lim­i­ta­tions
S
Pulmonary hyper­ten­sion PROMIS dyspnea sever­ity
Pain C Acute chest syn­drome SCD-related acute pain­ful epi­sodes
C Priapism core
Priapism
S Priapism Impact Profile (PIP) Priapism ques­tion­naire
Renal C Renal func­tion assess­ments
C Acute spleen Chronic spleen
Spleen
S Spleen assess­ment from the pedi­at­ric HU phase 3 clin­i­cal trial (BABY HUG)
Leg ulcers Retinopathy Avascular necro­sis
Other S
Chronic mal­nu­tri­tion Guidelines mal­nu­tri­tion iden­ti­fi­ca­tion
Assessments and
C Cardiac MRI Echocardiogram Brain MRI
exam­i­na­tions Imaging diag­nos­tics
S Functional MRI Brain MRA Imaging TCD
C Genetic diag­nos­tic test­ing Hemoglobin var­i­ant anal­y­sis
Laboratory tests
S Immune func­tion form Lab assess­ments-genet­ics/assays
Nonimaging S Electrocardiogram
Physical exam S Physical exam NIH Stroke Scale
Vital signs C Vital signs and blood gases
Treatment and
C Prior and con­com­i­tant med­i­ca­tions
inter­ven­tions Drugs
S PROMIS Medical Adherence Scale (PMAS) Asthma med­i­ca­tions list
Drug prod­uct Hematopoietic cel­lu­lar trans­plant infu­sion
C
Genetics and assays sum­mary of rec­om­men­da­tions
Therapies S Adhesion and vis­cos­ity Apheresis Conditioning reg­i­men

E Adhesion mol­e­cules assay

Cytopenia Genotoxicity Iron over­load


C
Adverse events and Infusion-related tox­ic­ity Infection form
toxicities
Adverse events New malig­nancy Toxicity form
S
Cellular ther­apy essen­tial data fol­low-up form

200  |  Hematology 2021  |  ASH Education Program


Table 2. (continued)

Domain Subdomain Class Recommendations


PROs C See details in Table 1
Outcomes and end points
Mortality C Death form
ACEs: adverse child­hood expe­ri­ences; Class: clas­si­fi­ca­tion; C: core; E: explor­atory; HU: hydroxy­urea; MRA: mag­netic res­o­nance angi­og­ra­phy; MRI:
mag­netic res­o­nance imag­ing; Ped: pedi­at­ric; S: sup­ple­men­tal; TCD: transcranial Dopp­ler.
Publicly available at https:​­/​­/curesickle​­.org​­/system​­/files​­/Sickle_Cell_Disease_CDE_Highlight_Summary​­.pdf.

phases of research, dif­fer­ent approaches to incor­po­rate patient Involving stake­hold­ers with diverse back­grounds pro­vi­des

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/196/1851725/196badawy.pdf by guest on 13 December 2021


and stake­holder input across the con­tin­uum of a clin­i­cal trial. the needed insight into per­sonal expe­ri­ences man­ag­ing SCD,
The CTTI also reported some exam­ples of poten­tial ben­e­fits for cul­tural con­sid­er­ations, adher­ence bar­ri­ers, and poten­tial strat­e­
PCOR, such as enhanc­ing the rel­e­vance of research ques­tions to gies to opti­mize the uptake of approved ther­a­pies as well as the
patients and stake­hold­ers, choos­ing the most appro­pri­ate pri­ research approach and accept­abil­ity of study assess­ments.33-35
mary and sec­ond­ary study out­comes, improv­ing strat­e­gies for An impor­tant part of stake­hold­ers’ engage­ment is clar­i­fy­ing the
engage­ment, recruit­ment, and reten­tion, addressing bar­ri­ers to sci­en­tific ratio­nale for the choice of study design and exam­in­ing
par­tic­i­pa­tion, keep­ing study bur­den to a min­i­mum, and opti­miz­ the fea­si­bil­ity of includ­ing spe­cific out­comes for a given trial with
ing over­all clin­i­cal trial expe­ri­ence.32 clear expec­ta­tions of time lines and lev­els of involve­ment.33-35

Table 3. Examples of SCD pro­jects funded by the PCORI

Project title Project type Budget Time line


We’ll Take the Village: Engaging the Community to Develop Better Health - Tier I Pipeline to pro­posal $15 000 2015
We’ll Take the Village: Engaging the Community to Better Health - Tier II Pipeline to pro­posal $25 000 2016-2017
OMPASS: COMmunity Participation to Advance the Sickle Cell Story Pipeline to pro­posal $50 000 2017-2018
National Sickle Cell Advocate Network (NSCAN) Engagement award $249 855 2016-2018
Tennessee Sickle Cell Disease Network Project Engagement award $249 963 2014-2017
Disseminating Results: Missed SCD Clinic Appointments and the Health Belief Model Engagement award $417 106 2019-2021
Automating Quality and Safety Benchmarking for Children: Meeting the Needs of Health PCORnet dem­on­stra­tion $1 264 641 2016-2021
Systems and Patients
Engaging Parents of Children With SCA and Providers in Shared-Decision Making for HU Research pro­ject $1 962 454 2017-2023
Comparative Effectiveness of a Decision Aid for Therapeutic Options in Sickle Cell Research pro­ject $2 143 228 2013-2018
Disease
PATient Navigator to rEduce Readmissions—The PArTNER Study Research pro­ject $2 054 803 2013-2018
Patient-Centered Comprehensive Medication Adherence Management System to Research pro­ject $2 148 331 2013-2018
Improve Effectiveness of Disease Modifying Therapy With HU in Patients With SCD
Comparing Two Ways to Help Patients With SCD Manage Pain (CaRISMA) Research pro­ject $4 343 821 2019-2024
Comparing Patient Centered Outcomes in the Management of Pain Between Research pro­ject $4 358 545 2014-2020
Emergency Departments and Dedicated Acute Care Facilities for Adults With SCD
National Pediatric Learning Health System (PEDSnet) - phase 1 PCORnet: CDRN (phase I) $6 459 893 2013-2015
Mid-South Clinical Data Research Network - phase 1 PCORnet: CDRN (phase 1) $6 672 017 2013-2015
Community Health Workers and Mobile Health for Emerging Adults Transitioning Research pro­ject $8 456 632 2017-2024
SCD Care (COMETS Trial)
Research Action for Health Network (REACHnet) PCORnet: CDRN (phase 2) $8 641 395 2015-2019
Comparative Effectiveness of Peer Mentoring Versus Structured Education-Based Research pro­ject $9 753 462 2017-2024
Transition Programming for the Management of Care Transitions in Emerging
Adults With SCD
Mid-South Clinical Data Research Network PCORnet: CDRN (phase 2) $10 064 128 2015-2019
Projects are orga­nized by level of funding sup­port from low to high.
CDRN, clin­i­cal data research net­work; HU, hydroxy­urea; PCORnet, National Patient-Centered Clinical Research Network.
Prakash Singh Shekhawat
Clinical trial con­sid­er­ations in SCD  |  201
Table 4. Various funding agencies, lev­els of patients, care­giver and stake­holder engage­ment, and poten­tial ben­efi
­ ts

Funding agency Level of engage­ment Benefits


Agency for Healthcare Research and Quality (AHRQ ) Desirable Possibly ben­e­fi­cial
Center for Medi­care and Med­ic­aid Services (CMS) Innovation Center Required Beneficial
National Institutes of Health (NIH) Potentially advan­ta­geous Possibly ben­e­fi­cial
Patient-Centered Outcomes Research Institute (PCORI) Expected Beneficial
Pharmaceutical com­pa­nies (indus­try) Potentially advan­ta­geous Beneficial
Professional soci­e­ties and orga­ni­za­tions Desirable Possibly ben­e­fi­cial
Note: Level of engage­ment is defined as the depth and the extent to which patients, care­giv­ers, and stake­hold­ers are engaged in dif­fer­ent stages of
a given research pro­ject that is pro­posed for funding by any of the listed agencies.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/196/1851725/196badawy.pdf by guest on 13 December 2021


Stakeholders may par­tic­i­pate in reg­u­lar study calls (eg, steering Figure 1  rep­re­sents  a pro­posed stake­holder engage­ment  frame-
com­mit­tee) and engage in detailed research dis­cus­sions in which work for clin­i­cal tri­als and research stud­ies in hema­tol­ogy, includ­ing
they can offer poten­tial solu­tions to unex­pected chal­lenges and SCD. The first layer (base of the pyr­am ­ id) includes the broader SCD
hur­dles hin­der­ing par­tic­i­pants’ recruit­ment, reten­tion, and fol­low- com­mu­nity, with online engage­ment ­strat­e­gies such as polls, sur­
up.33-35 Stakeholders may be given the oppor­tu­nity to con­trib­ute veys, blogs, social media, and dis­cus­sion boards. The sec­ond layer
to schol­arly prod­ucts from the research pro­ject and par­tic­i­ (mid­dle) involves more planned stake­holder engage­ment for in-
pate in edu­ca­tional ini­tia­tives for the dis­sem­i­na­tion of research depth insight into dif­fer­ent aspects of the research, with activ­i­ties
find­ings. such as com­mu­nity stu­dios, focus groups, and/or inter­views. This

Research
Partners

Planned Engagement
(community studios,
focus groups, interviews)

Broad Online Engagement (polls,


surveys, blogs, social media,
and discussion boards)

Paents Caregivers Researchers Clinicians Community

Professional Policy Payers


Industry
Sociees Maker

Figure 1. Hematology stakeholder-engagement framework in clinical trials and research studies.

202  |  Hematology 2021  |  ASH Education Program


engage­ment approach is essen­tial to pro­vide a safe envi­ron­ment plan might be help­ful to out­line the involve­ment of stake­hold­ers
for stake­hold­ers to give unbi­ased and crit­i­cal feed­back based on across dif­fer­ent stages of tri­als or research pro­jects.
their val­ues, expe­ri­ences, and back­grounds—espe­cially those who Historically, indus­ try-spon­
sored clin­ i­
cal tri­
als did not often
have no internet access, are not active on social media, or have lim­ include sig­nif­i­cant spon­sors of stake­holder engage­ment beyond
ited health lit­er­acy. Finally, the third layer (top) rep­re­sents research small pre­lim­i­nary stud­ies, and this hes­i­tancy may have been due
part­ners who are driv­ing the clin­i­cal trial or the research study, to a lack of famil­iar­ity with PCOR meth­od­ol­ogy and/or the unclear
includ­ing inves­ti­ga­tors, indus­try part­ners, and selected, actively return on invest­ment of study ben­e­fits. This has changed over
engaged patients and stake­hold­ers. This hema­tol­ogy stake­holder- the last decade, and cur­rently, a num­ber of ongo­ing clin­i­cal tri­
engage­ment frame­work (Figure 1) cap­tures var­i­ous poten­tial stake­ als in SCD have established advi­sory boards with dif­fer­ent stake­
hold­ers who either should or could be involved in clin­i­cal tri­als or hold­ ers, includ­ ing patients, care­ giv­ers, and advo­ cacy groups.
research stud­ies in hema­tol­ogy, with increas­ing lev­els of involve­ Furthermore, CTTI recently pro­posed an approach or a con­cep­
ment as we move toward the top of the pyr­a­mid. Engagement of tual finan­cial model to eval­u­ate the value of stake­holder engage­
all­these part­ners across dif­fer­ent lev­els of the pyr­a­mid, includ­ing ment in clin­i­cal tri­als.38 CTTI’s model is based on an esti­mated

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/196/1851725/196badawy.pdf by guest on 13 December 2021


stake­hold­ers and com­mu­nity orga­ni­za­tions, is essen­tial to ensure expected net pres­ent value (ENPV) incor­po­rat­ing cost, time, rev­
that clin­i­cal tri­als focus on mean­ing­ful out­comes for patients. This e­nue, and risk as cru­cial busi­ness driv­ers.38 In an exam­ple using an
frame­work also facil­i­tates col­lab­o­ra­tion and part­ner­ship between oncol­ogy devel­op­ment pro­gram, the authors reported a poten­
research­ers and stake­hold­ers while pri­or­i­tiz­ing out­comes of high tial mean­ing­ful impact of stake­holder engage­ment by avoiding
value to patients. pro­to­col amend­ments and enhanc­ing enroll­ment, reten­tion, and
Standardized train­ing may be needed to ensure that dif­fer­ent com­ple­tion of study assess­ments. This pos­i­tive impact in pre-
stake­hold­ers are equipped with the skill set and ade­quate prep­ phase 2 and pre-phase 3 tri­als was asso­ci­ated with an increase
a­ra­tion needed to be actively involved in the trial or the pro­ in NPV ($62 mil­lion and $65 mil­lion, respec­tively) and ENPV ($35
ject as research part­ners.33-35 A num­ber of PCOR com­pe­ten­cies mil­lion and $75 mil­ lion), adding sub­ stan­tial finan­
cial value to
and engage­ment prin­ci­ples have been reported in the lit­er­a­ture these tri­als. With a hypo­thet­i­cal ini­tial invest­ment of $100 000
(Table 5).36,37 Furthermore, establishing a detailed engage­ment ded­i­cated to opti­miz­ing stake­holder engage­ment strat­e­gies in

Table 5. PCR com­pe­ten­cies and prin­ci­ples

A. Competencies
I. Knowledge II. Skills III. Attitudes
Cultural con­text Communication Community val­ues
Knowledge about dis­ease Conflict man­age­ment Emotional intel­li­gence
Logistical con­sid­er­ations Critical think­ing General atti­tudes toward PCR
Participatory approaches Group par­tic­i­pa­tion Openness and trust
Agenda set­ting Leadership Personal attri­butes
Research meth­od­ol­ogy Project man­age­ment Personal growth
Understanding of data Teamwork Professional growth
Understanding PCR Prioritization Self-reflec­tion
B. Principles
I. Shared learn­ing expe­ri­ence Involvement of patients, care­giv­ers, and other stake­hold­ers in all­aspects of the research
Researchers and team mem­bers learn­ing about PCR meth­od­ol­ogy
Training for patients, care­giv­ers, and other stake­hold­ers on research prin­ci­ples
II. Collaborations Cultural sen­si­tiv­ity and mutual respect
Fair com­pen­sa­tion for effort and time
Inclusion and diver­sity for all­pro­ject-related activ­i­ties and part­ner­ships
Planning ahead for meet­ings, tasks, and mile­stones with real­is­tic time lines
III. Bidirectional rela­tion­ships Patient, care­giv­ers, and other stake­hold­ers are involved as research part­ners
Well-defined roles and strat­e­gies informed by col­lab­o­ra­tive dis­cus­sions
IV. Trustworthiness Clear and trans­par­ent com­mu­ni­ca­tions
Shared deci­sion-mak­ing pro­cess
Sharing infor­ma­tion and data openly

Prakash Singh Shekhawat


Clinical trial con­sid­er­ations in SCD  |  203
a clin­i­cal trial, there may be a return on invest­ment, in both NPV recommended to assess adher­ence out­comes. Developmental
and ENPV, that exceeds the invest­ment 500-fold.38 dif­fer­ences among chil­dren and ado­les­cents with SCD should
inform the study approach. Engaging patients and stake­hold­
COVID-19 pan­demic and clin­i­cal tri­als ers in SCD clin­i­cal tri­als in mean­ing­ful ways is crit­i­cal to ensure
More recently, the COVID-19 pan­demic has led to dis­rup­tions in that their voices are heard and that study designs and out­
our daily rou­tines, per­son­ally and pro­fes­sion­ally, in dif­fer­ent ways, comes are rel­e­vant to them, which is essen­tial for future suc­
includ­ing interrupting the exe­cu­tion of clin­i­cal tri­als.39,40 Most insti­ cess­ful dis­sem­i­na­tion and implementation. This engage­ment is a
tu­tions stopped new enroll­ments and allowed the con­tin­ua ­ ­tion dynamic, bidi­rec­tional com­mit­ment that is mutu­ally ben­e­fi­cial to
of interventional tri­als when there were poten­tial clin­i­cal ben­e­fits all­involved part­ners, and it has the poten­tial to improve health
for the par­tic­i­pants; how­ever, many reported chal­lenges related out­comes in the larger pop­u­la­tion of pedi­at­ric and adult SCD
to delayed and rescheduled study vis­its, pro­ce­dures, and assess­ patients. Stakeholders can play a major role in clos­ing the gap
ments and over­all dif­fi­culty reaching patients.40 Many of these between data-heavy research find­ings from clin­i­cal tri­als and
vul­ner­a­ble patients were at risk from expo­sure to COVID-19, and their impli­ca­tions in clin­i­cal prac­tice. It is crit­i­cal to keep stake­

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/196/1851725/196badawy.pdf by guest on 13 December 2021


some were inten­tion­ally avoiding health care facil­i­ties or obey­ing hold­ers engaged and inter­ested through­out the research pro­
stay-at-home-orders.39 This sit­u­a­tion high­lights the need for con­ cess, and the sus­tain­abil­ity of this part­ner­ship is key. Evaluating
sid­er­able adapt­abil­ity using a hybrid strat­egy in design­ing future the ­finan­cial value of stake­holder engage­ment is impor­tant to
clin­i­cal tri­als to com­plete all­ planned study pro­ce­dures.40 Some esti­mate the poten­tial cost sav­ings for SCD clin­i­cal tri­als, which
pro­posed strat­e­gies include (1) pri­or­i­tiz­ing pri­mary out­comes might be of ­con­sid­er­able finan­cial value. Timely adap­ta­tions to
over explor­ atory ones; (2) alter­ nat­
ing strat­e­
gies for out­ comes address unusual cir­cum­stances, such as the COVID-19 pan­demic,
assess­ment; (3) collecting remote data using phone inter­views or are often cru­cial.
online tools; (4) obtaining phone num­bers and e-mail addresses
for patients and 3 fam­ily mem­bers or friends to ensure maintained Acknowledgments
con­tact; (5) using dif­fer­ent meth­ods to con­tact par­tic­i­pants, This pro­ject was supported by a grant (K23HL150232, PI: Bad­
includ­ing text mes­sag­ing, phone calls, e-mail, or social media; awy) from the National Heart, Lung, and Blood Institute of the
(6) employing telemedicine; (7) arranging home vis­its by health National Institutes of Health. The con­tent is solely the respon­si­
care work­ers wear­ing per­sonal pro­tec­tive equip­ment; (8) allow­ bil­ity of the author and does not nec­es­sar­ily rep­re­sent the views
ing study med­ic ­ a­tions to be taken at home; (9) mak­ing use of of the National Institutes of Health.
con­cierge ser­vices; (10) using local instead of cen­tral lab facil­ I would like to thank Drs. Robert Liem, Jane Holl, David Cella,
i­ties; and (11) esca­lat­ing incen­tives.12,40,41 Other approaches Tonya Palermo, and Alexis Thompson for their advice while writ­
should be con­sid­ered to achieve the highest level of reten­tion ing this arti­cle.
and adher­ence to study inter­ven­tions, and the sta­tis­ti­cal anal­y­
sis plan for pri­mary and sec­ond­ary out­comes should be revised Conflict-of-inter­est dis­clo­sure
to reflect any expected mean­ing­ful effects or influ­ence relate Sherif M. Badawy: no com­pet­ing finan­cial inter­ests to declare.
to the pan­demic.41 For behav­ioral clin­ic ­ al tri­als, efforts should
be directed to lever­ age widely avail­­able and user-friendly
Off-label drug use
online sur­veys, data­bases, and web-based appli­ca­tions to opti­
Sherif M. Badawy: nothing to disclose.
mize e-consenting and remote enroll­ment, with com­ple­tion
of all­study assess­ments and deliv­ery of study inter­ven­tions
­conducted vir­tu­ally. Correspondence
Sherif M. Badawy, Ann and Robert H. Lurie Children’s Hospital of
Chicago, 225 E Chicago Ave, Box #30, Chicago, IL 60611; e-mail:
sbadawy@luriechildrens​­.org.
CLINICAL CASE (Con­t in­u ed)
References
The patient has been actively involved as an advi­sory board 1. Hassell KL. Population esti­ma­tes of sickle cell dis­ease in the U.S. Am J Prev
mem­ber in a few inves­ti­ga­tor-ini­ti­ated tri­als at our insti­tu­tion. He Med. 2010;38(4):S512-S521.
had sig­nif­i­cant input in these tri­als, such as feed­back on con­sent 2. Piel FB, Steinberg MH, Rees DC. Sickle cell dis­ ease. N Engl J Med.
2017;376(16):1561-1573.
and assent forms and the selec­tion of HRQOL domains that are
3. Kato GJ, Piel FB, Reid CD, et al. Sickle cell dis­ease. Nat Rev Dis Primers.
more rel­e­vant to SCD patients. In addi­tion, he was ­able to pro­ 2018;4(March):18010.
vide crit­i­cal insight to inform our efforts to develop and refine 4. Panepinto JA. Health-related qual­ity of life in patients with hemo­glo­bin­
a mobile app for ado­les­cents and young adults as a behav­ioral op­a­thies. Hematology Am Soc Hematol Educ Program. 2012;2012(Jan­u­
inter­ven­tion to mon­i­tor and improve med­i­ca­tion adher­ence and ary):284-289.
5. King AA, Badawy SM, Panepinto J, Anie K, Jonassaint C, Treadwell M. Psy­
HRQOL. His pas­sion for med­i­cine was reinforced, and he is deter­
chosocial bur­den in sickle cell dis­ease. In: Gladwin M, Novelli E, Kato G,
mined to become a hema­tol­o­gist car­ing for adult SCD patients. eds. Sickle Cell Disease. McGraw Hill; 2021 558-578.
6. Badawy SM, Thompson AA, Lai JS, Penedo FJ, Rychlik K, Liem RI. Health-
related qual­ity of life and adher­ence to hydroxy­urea in ado­les­cents and
Conclusions young adults with sickle cell dis­ease. Pediatr Blood Cancer. 2017;64(6):
e26369.
Several clin­i­cal trial con­sid­er­ations in SCD are key to suc­cess. 7. Raphael JL, Mei M, Mueller BU, Giordano T. High resource hos­pi­tal­i­za­tions
PROs are sig­nif­i­cantly impaired among SCD patients and should among chil­dren with vaso-occlu­sive cri­ses in sickle cell dis­ease. Pediatr
be included in all­clin­i­cal tri­als. A mul­ti­modal strat­egy is highly Blood Cancer. 2012;58(4):584-590.

204  |  Hematology 2021  |  ASH Education Program


8. Brousseau DC, Owens PL, Mosso AL, Panepinto JA, Steiner CA. Acute download; https://www.fda.gov/industry/prescription-drug-user-fee-​
care uti­li­za­tion and rehospitalizations for sickle cell dis­ease. JAMA. 2010;​ ame​nd​ments/​fda-led-patient-focused-drug-development-pfdd-public-
303(13):1288-1294. meetings#sicklecell.
9. Badawy SM, Thompson AA, Holl JL, Penedo FJ, Liem RI. Healthcare uti­li­ 26. Coons SJ, Eremenco S, Lundy JJ, O’Donohoe P, O’Gorman H, Malizia W.
za­tion and hydroxy­urea adher­ence in youth with sickle cell dis­ease. Pediatr Capturing patient-reported out­come (PRO) data elec­tron­i­cally: the past,
Hematol Oncol. 2018;35(5-6):297-308. pres­ ent, and prom­ ise of ePRO mea­ sure­ment in clin­i­
cal tri­
als. Patient.
10. Lebensburger JD, Hilliard LM, Pair LE, Oster R, Howard TH, Cutter GR. Sys­ 2015;8(4):301-309.
tematic review of interventional sickle cell tri­als reg­is­tered in ClinicalTrials​ 27. Badawy SM, Thompson AA, Liem RI. Technology access and smartphone
­.gov. Clin Trials. 2015;12(6):575-583. app pref­er­ences for med­i­ca­tion adher­ence in ado­les­cents and young
11. Lebensburger JD, Sidonio RF, Debaun MR, Safford MM, Howard TH, Scarinci adults with sickle cell dis­ease. Pediatr Blood Cancer. 2016;63(5):848-852.
IC. Exploring bar­ ri­
ers and facil­i­
ta­
tors to clin­

cal trial enroll­
ment in the 28. Badawy SM, Cronin RM, Hankins J, et al. Patient-cen­tered eHealth inter­ven­
con­ text of sickle cell ane­ mia and hydroxy­ urea. Pediatr Blood Cancer. tions for chil­dren, ado­les­cents, and adults with sickle cell dis­ease: sys­tem­
2013;60(8):1333-1337. atic review. J Med Inter­net Res. 2018;20(7):e10940.
12. Palermo TM, Kashikar-Zuck S, Friedrichsdorf SJ, Powers SW. Special con­sid­ 29. Lehmann A, Aslani P, Ahmed R, et  al. Assessing med­i­ca­tion adher­ence:
er­ations in conducting clin­i­cal tri­als of chronic pain man­age­ment inter­ven­ options to con­sider. Int J Clin Pharm. 2014;36(1):55-69.
tions in chil­dren and ado­les­cents and their fam­i­lies. Pain Rep. 2019;4(3):e649. 30. Peipert JD, Badawy SM, Baik SH, et  al. Development of the NIH patient-

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/196/1851725/196badawy.pdf by guest on 13 December 2021


13. Liem RI, Cole AH, Pelligra SA, Mason M, Thompson AA. Parental atti­tudes reported out­comes mea­sure­ment infor­ma­tion sys­tem (PROMIS) ­med­i­ca­tion
toward research par­tic­i­pa­tion in pedi­at­ric sickle cell dis­ease. Pediatr Blood adher­ence scale (PMAS). Patient Prefer Adherence. 2020;14(9 June):
Cancer. 2010;55(1):129-133. 971-983.
14. Carden MA, Little J. Emerging dis­ease-mod­i­fy­ing ther­a­pies for sickle cell 31. Hansen RA, Kim MM, Song L, Tu W, Wu J, Murray MD. Comparison of meth­
dis­ease. Haematologica. 2019;104(9):1710-1719. ods to assess med­i­ca­tion adher­ence and clas­sify nonadherence. Ann Phar-
15. Leonard A, Tisdale J, Abra­ham A. Curative options for sickle cell dis­ease: macother. 2009;43(3):413-422.
haploidentical stem cell trans­plan­ta­tion or gene ther­apy? Br J Haematol. 32. Bloom D, Beetsch J, Harker M, et al. The rules of engage­ment: CTTI rec­om­
2020;189(3):408-423. men­da­tions for suc­cess­ful col­lab­o­ra­tions between spon­sors and patient
16. Farrell AT, Panepinto J, Carroll CP, et al. End points for sickle cell dis­ease groups around clin­i­cal tri­als. Ther Innov Regul Sci. 2018;52(2):206-213.
clin­i­cal tri­als: patient-reported out­comes, pain, and the brain. Blood Adv. 33. Concannon TW, Grant S, Welch V, et al; Multi Stakeholder ­Engagement
2019;3(23):3982-4001. Consortium. Practical guid­
­ ance for involv­ing stake­ hold­
ers in health
17. Farrell AT, Panepinto J, Desai AA, et  al. End points for sickle cell dis­ease research. J Gen Intern Med. 2019;34(3):458-463.
clin­i­cal tri­als: renal and car­dio­pul­mo­nary, cure, and low-resource set­tings. 34. Forsythe L, Heckert A, Margolis MK, Schrandt S, Frank L. Methods and
Blood Adv. 2019;3(23):4002-4020. impact of engage­ ment in research, from the­ ory to prac­ tice and back
18. Amer­i­can Society of Hematology Research Collaborative. Clinical tri­als net­ again: early find­ings from the Patient-Centered Outcomes Research Insti­
work. Accessed 3 Octo­ber 2020. https:​­/​­/www​­.ashre​searchcollaborative​ tute. Qual Life Res. 2018;27(1):17-31.
­.org​­/s​­/​clinical​­-trials​­-network. 35. Forsythe LP, Ellis LE, Edmundson L, et al. Patient and stake­holder engage­
19. Eckman JR, Hassell KL, Huggins W, et al. Standard mea­sures for sickle cell ment in the PCORI pilot pro­jects: descrip­tion and les­sons learned. J Gen
dis­ease research: the PhenX Toolkit sickle cell dis­ease col­lec­tions. Blood Intern Med. 2016;31(1):13-21.
Adv. 2017;1(27):2703-2711. 36. Frisch N, Atherton P, Doyle-Waters MM, et  al. Patient-ori­ented research
20. National Heart, Lung, and Blood Institute. Cure sickle cell ini­ tia­
tive. com­pe­ten­cies in health (PORCH) for research­ers, patients, healthcare pro­
Accessed 3 Octo­ber 2021. https:​­/​­/www​­.nhlbi​­.nih​­.gov​­/science​­/cure​­-sickle​ vid­ers, and deci­sion-mak­ers: results of a scop­ing review. Res Involv Enga-
­-cell​­-initiative. gem. 2020;6(10 Feb­ru­ary):4.
21. Calvert M, Blazeby J, Altman DG, et al; Consolidated Standards of Report­ 37. Sheridan S, Schrandt S, Forsythe L, Hilliard TS, Paez KA; Advisory Panel
ing Trials Patient-Reported Outcomes Group. Reporting of patient- on Patient Engagement (2013 Inaugural Panel). The PCORI engage­ment
reported out­comes in ran­dom­ized tri­als: the CONSORT PRO exten­sion. rubric: prom­ is­
ing prac­ tices for partnering in research. Ann Fam Med.
JAMA. 2013;309(8):814-822. 2017;15(2):165-170.
22. Tunis SR, Stryer DB, Clancy CM. Practical clin­i­cal tri­als: increas­ing the value 38. Levitan B, Getz K, Eisenstein EL, et al. Assessing the finan­cial value of patient
of clin­i­cal research for deci­sion mak­ing in clin­i­cal and health pol­icy. JAMA. engage­ ment: a quan­ ti­
ta­
tive approach from CTTI’s patient groups and
2003;290(12):1624-1632. clin­i­cal tri­als pro­ject. Ther Innov Regul Sci. 2018;52(2):220-229.
23. Calvert M, Kyte D, Mercieca-Bebber R, et al; Standard Protocol Items: Rec­ 39. van Dorn A. COVID-19 and readjusting clin­ i­
cal tri­
als. Lancet. 2020;
ommendations for Clinical Trials Patient-Reported Outcome Group. Guide­ 396(10250):523-524.
lines for inclu­sion of patient-reported out­comes in clin­i­cal trial pro­to­cols: 40. McDermott MM, Newman AB. Preserving clin­i­cal trial integ­rity dur­ing the
the SPIRIT-PRO exten­sion. JAMA. 2018;319(5):483-494. coronavirus pan­demic. JAMA. 2020;323(21):2135-2136.
24. Coons SJ, Gwaltney CJ, Hays RD, et al; International Society for Pharma­ 41. Flem­ing TR, Labriola D, Wittes J. Conducting clin­i­cal research dur­ing the
coeconomics and Outcomes Research ePRO Task Force. Recommenda­ COVID-19 pan­demic: protecting sci­en­tific integ­rity. JAMA. 2020;324(1):
tions on evi­dence needed to sup­port mea­sure­ment equiv­a­lence between 33-34.
elec­tronic and paper-based patient-reported out­come (PRO) mea­sures:
ISPOR ePRO Good Research Practices Task Force report. Value Health.
2009;12(4):419-429.
25. US Food and Drug Administration. The voice of the patient: a series of
reports from the U.S. Food and Drug Administration’s (FDA’s) patient- © 2021 by The Amer­i­can Society of Hematology
focused drug devel­op­ment ini­tia­tive. https://www.fda.gov/media/89898/ DOI 10.1182/hema­tol­ogy.2021000252

Prakash Singh Shekhawat


Clinical trial con­sid­er­ations in SCD  |  205
HEMOPHILIA UPDATE: OUR CUP RUNNETH OVER

How do we optimally utilize factor concentrates


in persons with hemophilia?

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/206/1851572/206lim.pdf by guest on 13 December 2021


Ming Y. Lim
Division of Hematology and Hematologic Malignancies, Department of Internal Medicine, University of Utah, Salt Lake City, UT

The current mainstay of therapy for hemophilia is to replace the deficient clotting factor with the intravenous administra-
tion of exogenous clotting factor concentrates. Prophylaxis factor replacement therapy is now considered the standard
of care in both pediatric and adult patients with hemophilia with a severe phenotype to protect musculoskeletal health
and improve quality of life. Heterogeneity in bleeding presentation among patients with hemophilia due to genetic,
environmental, and treatment-related factors has been well described. Accordingly, the World Federation of Hemophilia
recommends an individualized prophylaxis regimen that considers the factors mentioned above to meet the clinical
needs of the patient, which can vary over time. This review focuses on the practical points of choosing the type of factor
concentrate, dose, and interval while evaluating appropriate target trough factor levels and bleeding triggers such as
level of physical activity and joint status. We also discuss the use of a pharmacokinetics assessment and its incorporation
in the clinic for a tailored approach toward individualized management. Overall, adopting an individualized prophylaxis
regimen leads to an optimal utilization of factor concentrates with maximum efficacy and minimum waste.

LEARNING OBJECTIVES
• Describe the different environmental and treatment-related variables involved in determining an appropriate pro-
phylaxis regimen
• Understand the importance of an individualized prophylaxis regimen that meets the clinical needs of hemophilia
patients

CLINICAL CASE Introduction


A 15-year-old adolescent boy with moderate hemophilia A HA and hemophilia B (HB) are X-linked bleeding disorders
(HA; baseline factor VIII 2%) presented to a hematology clinic that result from decreased or deficient plasma clotting fac-
to discuss whether he should start prophylaxis with clotting tors VIII (FVIII) and IX (FIX), respectively. The severity of
factor concentrates (CFCs). He was diagnosed at the age of hemophilia has traditionally been defined based on the de-
3 by his astute pediatrician when he developed severe right gree of the clotting factor deficiency, with levels of >5% to
knee hemarthrosis after tripping while running. Throughout 40% classified as mild hemophilia, 1% to 5% as moderate,
his childhood and adolescent years, he required infrequent and <1% as severe.1 The severity and frequency of bleeding
on-demand administration of CFCs predominantly due to in hemophilia typically correlate with the degree of the
trauma-induced acute knee hemarthrosis from playing soft- clotting factor deficiency (Table 1).
ball and basketball. He recalled 3 episodes of spontaneous The mainstay of therapy for hemophilia is to replace
knee hemarthrosis occurring about once per year in the last the deficient clotting factor, usually by intravenous admin-
3 years. He was now in high school and wanted to play bas- istration of exogenous CFC. This can be episodic to treat
ketball competitively, with 3 practice games per week and bleeding events or on a regular basis (prophylaxis) to pre-
more during tournament season. He was concerned about vent bleeding episodes.1 Heterogeneity in bleeding pre-
his increased risk of bleeding given his history of spontane- sentation between patients with similar severity of disease
ous joint bleeds and the high likelihood of musculoskeletal or between patients with HA and HB is well recognized.2-4
injuries, such as sprained ankles and knee ligament tears, Similarly, patients with moderate hemophilia can present
that can occur while playing basketball. phenotypically as those with severe disease.5 The basis
for this phenotypic variation is multifactorial and includes

206 | Hematology 2021 | ASH Education Program


Table 1.  Association of bleed­ing man­i­fes­ta­tions with sever­ity Types of fac­tor con­cen­trates
of dis­ease based on plasma clot­ting fac­tor lev­els For a rare dis­ or­der, the arma­ men­ tar­
ium of CFCs avail­­able is
note­wor­thy, with a wide range of prod­ucts in use around the
Severity Plasma clot­ting fac­tor lev­els Bleeding man­i­fes­ta­tions world. The 2 main types of SHL CFC are the virally inactivated
Mild 5%-<40% of nor­mal Bleeding with major plasma-derived lyoph­i­lized fac­tor con­cen­trates and the recom­
trauma or sur­gery; rare bi­nant fac­tor con­cen­trates manufactured from genet­i­cally engi-
spon­ta­ne­ous bleed­ing neered cells; both have high clin­i­cal effi­cacy and safety.18 Table 2
Moderate 1%-5% of nor­mal Bleeding with minor trauma lists the CFCs cur­rently licensed for use in the United States. An
or sur­gery; occa­sional updated list of all­cur­rently avail­­able prod­ucts glob­ally and their
spon­ta­ne­ous bleed­ing manufactur­ing details are avail­­able in the WFH Online Registry of
Severe <1% of nor­mal Spontaneous bleed­ing ­ Clotting Factor Concentrates.19
pre­dom­i­nantly in the joints For HA patients, there is a clin­i­cal con­cern that recom­bi­nant
or mus­cles but can occur FVIII con­cen­trates may con­trib­ute to a higher rate of inhib­i­tor
any­where, includ­ing
devel­op­ment in pre­vi­ously untreated patients (PUPs) com­pared

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/206/1851572/206lim.pdf by guest on 13 December 2021


life-threat­en­ing intra­cra­nial
bleed to plasma-derived FVIII con­ cen­trates, although this remains
highly con­tro­ver­sial.20,21 Conversely, the risk of inhib­i­tor devel­
Adapted with permission from Srivastava, A, Santagostino, E, Dougall, A,
et al. WFH Guidelines for the Management of Hemophilia, 3rd edition.9 op­ment in HB patients is not thought to be related to the type
© 2020 World Federation of Hemophilia. Haemophilia © 2020 John of CFC.9 Inhibitors are immu­no­glob­u­lin G (IgG) alloantibodies to
Wiley & Sons Ltd. exog­e­nous FVIII or FIX that neu­tral­ize the func­tion of infused CFC
and mostly occur within the first 50 expo­sures.1,22 An expo­sure is
(1) genetic fac­tors such as the F8/F9 geno­type (null vs nonnull defined as any infu­sion of a FVIII/FIX-containing prod­uct in a 24-
var­i­ants),6 con­com­i­tant thrombophilic gene var­i­ants or other hour period.1 Meta-ana­ly­ses of obser­va­tional stud­ies, includ­ing
bleed­ing dis­or­ders,7 and intrin­sic pro- and anti­co­ag­u­lant activ­ patient-level data, found no dif­fer­ences in inhib­i­tor rates when
ity,6 and (2) envi­ron­men­tal fac­tors that can vary for an indi­vid­ com­par­ing all­ plasma-derived FVIII con­cen­trates to all­ recom­bi­
ual over time, such as func­tional sta­tus, lev­els and pat­terns of nant FVIII con­cen­trates.20,23,24 In con­trast, the much-awaited Sur-
phys­i­cal activ­ity, and joint sta­tus.8 Rather than a one-size-fits-all­ vey of Inhibitors in Plasma-Product Exposed Toddlers (SIPPET)
approach, the opti­mal uti­li­za­tion of CFC takes into account both study, the only pro­spec­tive, ran­dom­ized con­trolled trial, found
genetic and envi­ron­men­tal fac­tors as well as treat­ment-related that PUPs treated with recom­bi­nant FVIII prod­ucts had a higher
fac­tors such as fac­tor type and dose, indi­vid­ual and prod­uct cumu­ la­
tive inci­
dence of inhib­ i­
tors (44.5%; 95% CI, 34.7-54.3)
phar­ma­co­ki­net­ics (PK) of fac­tor, and indi­vid­ual pref­er­ence and com­pared with those treated with plasma-derived FVIII prod­ucts
adher­ence (Figure 1). (26.8%; 95% CI, 18.4-35.2).21 The gen­er­al­iz­abil­ity and appli­ca­bil­
In this review, we focus on pro­phy­laxis fac­tor replace­ment ity of SIPPET have been widely debated in numer­ous con­fer­ences
ther­apy and prac­ti­cal points around choos­ing the type of con­ and review arti­cles and is beyond the scope of this review. The
cen­trate, dose, and inter­val while con­sid­er­ing trough fac­tor lev­ inter­ested reader is directed to the National Hemophilia Foun-
els and bleed­ing trig­gers. Finally, we briefly dis­cuss the use of dation’s Medical and Scientific Advisory Council doc­u­ment 243
PK assess­ment for indi­vid­u­al­ized man­age­ment and its incor­po­ for a detailed expla­na­tion of the dif­fer­ences between SIPPET
ra­tion in the clinic. The use of CFC in the man­age­ment of hemo­ and prior obser­va­tional stud­ies and their rec­om­men­da­tions.25
philia patients with inhib­i­tors and the opti­mal uti­li­za­tion of fac­tor Additionally, the Euro­ pean Medicines Agency Pharmacovigi-
replace­ment ther­apy dur­ing sur­gery are beyond the scope of lance Risk Assessment Committee con­cluded in 2017 that no
this paper. Readers are directed to the World Federation of clear evi­dence exists of inhib­i­tor risk dif­fer­ences between plas-
Hemophilia (WFH) guide­lines on the dos­age and dura­tion of CFC ma-derived and recom­bi­nant fac­tor VIII con­cen­trates.26 Ongo-
cov­er­age for major and minor sur­ger­ies (Srivastava et al; Table ing research through the Amer­i­can Thrombosis and Hemostasis
7-2).9 During the perioperative period, both stan­dard half-life Network 8 PUPs Matter study, a lon­gi­tu­di­nal obser­va­tional study
(SHL) and extended half-life (EHL) CFC can be given via con­tin­u­ collecting treat­ment and inhib­i­tor data on chil­dren with mod­
ous infu­sion or inter­mit­tent bolus.10-12 er­ate and severe HA or HB born on or after 1 Jan­u­ary 2010, may
help pro­vide addi­tional clar­ity on this issue.27 At this time, the
Prophylaxis as the stan­dard of care WFH does not express a pref­er­ence for prod­uct type and rec­
Since com­ ple­
tion of the Joint Outcome Study,13 and ESPRIT14 om­mends prod­uct selec­tion based on local avail­abil­ity, cost, and
study for children with severe HA, and the SPINART study15 for pro­vider/patient pref­er­ence.9
adolescents and adults with severe HA, the use of pro­phy­laxis For pre­vi­ously treated patients, clas­si­cally defined as hav­
fac­tor replace­ment ther­apy is now con­sid­ered the stan­dard of ing received >150 expo­sure (although oth­ers have con­sid­ered
care. The recently updated WFH guide­lines rec­om­mend that it to be >50 expo­sure),28 the risk of inhib­i­tor devel­op­ment is
pedi­at­ric and adult patients with hemo­philia with a severe phe­ low.22 Additionally, stud­ies indi­cate no increased risk of inhib­
no­type (to include those with mod­er­ate hemo­philia with a se- i­tor devel­op­ment when switching to another type or brand
vere phe­no­type) be on pro­phy­laxis to pre­vent spon­ta­ne­ous and of fac­tor.29,30 Whether to use plasma-derived vs recom­bi­nant
break­through bleed­ing at all­times.9 Specifically for chil­dren, the fac­tor con­cen­trates is a mat­ter of dealer’s choice. Practically,
WFH guide­lines rec­om­mend the early ini­ti­a­tion of pro­phy­laxis regional avail­abil­ity and pay­ers (gov­ern­ment or insur­ance
ide­ally before age 3, as this has been shown to have bet­ter long- com­pa­nies) pref­er­ence due to nego­ti­ated pro­cure­ment dis­
term joint out­comes com­pared to starting after age 6.9,16,17 counts (as well as out-of-pocket cost for US patients) play a

Prakash Singh Shekhawat


Optimal pro­phy­laxis fac­tor replace­ment ther­apy | 207
Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/206/1851572/206lim.pdf by guest on 13 December 2021
Figure 1.  Factors that lead to optimal utilization of CFC. Created with BioRender​.com.

larger role in deter­min­ing the spe­cific prod­uct of CFC to pre­ a starting reg­i­men of one of the afore­men­tioned approaches
scribe within each type. is selected that is mutu­ ally accept­
able to both cli­
ni­
cian and
patient/care­giver and then tai­lored (esca­lated or de-esca­lated)
Dose and inter­val to the patient’s clin­i­cal needs.
The dose and inter­val of a pro­phy­laxis reg­i­men depend on the
half-life of the CFC—the time for fac­tor con­cen­tra­tion to reach Target trough lev­els
half of its orig­i­nal peak con­cen­tra­tion. In adults, the half-life can Historically, the goal of pro­ phy­laxis replace­
ment ther­ apy has
range between 8 and 12 hours for FVIII con­cen­trates and 18 and been to tar­get a trough fac­tor level of at least 1% (ie, convert-
30 hours for FIX con­cen­trates, depending on the brand of fac­ ing a patient with severe hemo­philia to the typ­i­cal bleed­ing
tor; in chil­dren the half-life is shorter.31,32 Although most man­u­ ­phe­no­type of mod­er­ate hemo­philia) to pre­vent spon­ta­ne­ous
fac­tur­ers pro­vide recommended starting doses for pro­phy­laxis joint bleed and to reduce dam­age to mus­cu­lo­skel­et­ al health.9,31,34
(Table 2), mul­ti­ple reg­im­ ens exist and vary widely. These reg­i­ However, given the afore­men­tioned phe­no­typic var­i­a­tions ob-
mens range from a high-dose approach that involves the admin­ served, the tra­di­tional tar­get trough level of 1% does not pre­vent
is­tra­tion of 25 to 40 IU/kg per dose every other day (for SHL FVIII bleed­ing in all­hemo­philia patients.35 Also, for patients with mod­
con­cen­trates) or twice per week (for SHL FIX con­cen­trates) to a er­ate hemo­philia with fre­quent spon­ta­ne­ous bleed­ing (a severe
low-dose approach that involves once-weekly or twice-weekly phe­no­type), a tar­get trough level of 1% is clin­i­cally unhelp­ful.36
infu­sion and/or using lower doses to a reg­i­men in between these So what should the tar­get trough level be to achieve zero
2 approaches. The low-dose approach is pre­dom­i­nantly used in bleeds for all­and is this attain­­able? Studies in mild and mod­er­
resource-constrained countries where access to CFCs is lim­ited. ate hemo­ philia patients observed that the annual num­ ber of
The ben­e­fits of such an approach over epi­sodic treat­ment have joint bleeds decreases as base­line fac­tor activ­ity increases and
been dem­on­strated with­out incur­ring a huge finan­cial bur­den.33 approaches zero with a base­line fac­tor activ­ity of >15% to 30%.4,37
Thus, depending on local resources and eco­nomic con­straints, Predictive mod­el­ing stud­ies in severe HA patients on pro­phy­laxis

208  |  Hematology 2021  |  ASH Education Program


Table 2.  CFCs licensed in the US to treat HA and HB

Brand (non­pro­pri­etary name) Technology Manufacturer-recommended starting dose for pro­phy­laxis use*
Factor VIII prod­ucts
  Kogenate FS® (octocog alfa) Second gen­er­a­tion: full-length recom­bi­nant Adults: 25 IU/kg 3 times weekly; chil­dren: 25 IU/kg every other day
 Advate® (octocog alfa) Third gen­er­a­tion: full-length recom­bi­nant 20-40 IU/kg every other day
 Kovaltry (octocog alfa)
®
Third gen­er­a­tion: full-length recom­bi­nant Adults/ado­les­cents: 20-40 IU/kg 2-3 times weekly; chil­dren ≤12
years: 25-50 IU/kg 2 times weekly, 3 times weekly, or every
other day
 NovoEight® (turoctocog alfa) Third gen­er­a­tion: B-domain trun­cated Adults/ado­les­cents: 20-50 IU/kg 3 times weekly or 20-40 IU/kg
every other day; chil­dren <12 years: 25-60 IU/kg 3 times weekly
or 25-50 IU/kg every other day
  Xyntha®/ReFacto AF® Third gen­er­a­tion: B-domain deleted Adults/ado­les­cents: 30 IU/kg 3 times weekly; chil­dren <12 years:

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/206/1851572/206lim.pdf by guest on 13 December 2021


(moroctocog alfa) 25 IU/kg every other day
 Nuwiq® (simoctocog alfa) Fourth gen­er­a­tion: B-domain deleted Adults/ado­les­cents: 30-40 IU/kg every other day; chil­dren <12
years: 30-50 IU/kg every other day or 3 times weekly
  Hemofil M® Plasma-derived immunoaffinity-puri­fied FVIII Not pro­vided
 Koate ®
Plasma-derived containing FVIII and von Not pro­vided
Willebrand fac­tor
 Humate-P® Plasma-derived containing FVIII and von Not pro­vided
Willebrand fac­tor
 Alphanate® Plasma-derived containing FVIII and von Not pro­vided
Willebrand fac­tor
Factor IX prod­ucts
 BeneFIX® (nonacog alfa)† Recombinant FIX Age ≥16 years: 100 IU/kg weekly
 Ixinity® (trenonacog alfa)‡ Recombinant FIX Adults: 40-70 IU/kg twice weekly
 Rixubis® (nonacog gamma) Recombinant FIX Age ≥12 years: 40-60 IU/kg twice weekly; chil­dren <12 years:
60-80 IU/kg twice weekly
 Alphanine Plasma derived Not pro­vided
*Recommended doses based on US pack­age insert.
†In the US, BeneFIX® is approved for pro­phy­laxis use in those aged ≥16 years only, whereas in the EU it is approved for pro­phy­laxis use at all­ages.
‡In the US, Ixinity® is approved for pro­phy­laxis use in adults only. It is not approved in the EU.

fac­tor replace­ment ther­apy projected that every 1% rise in trough activ­i­ties are lacking. A Delphi con­sen­sus state­ment suggested a
FVIII lev­els resulted in a 2% increase in the num­ber of patients who fac­tor level of 3% to 5% for mild phys­i­cal activ­ity and 5% to 15%
might achieve zero bleeds in a year.38 Accordingly, most cli­ni­cians for higher-risk phys­i­cal activ­ity,42 whereas a more recent expert
favor targeting a higher trough level (>3%-5% or higher) but are solic­i­ta­tion exer­cise suggested much higher lev­els of up to 40%
prohibited from doing so due to the con­straints of SHL CFCs that to 50% (Table 3).43 Regardless of fac­tor lev­els, addi­tional strat­e­
require almost daily infu­sions to achieve this level. In addi­tion to gies includ­ing proper coaching and super­vi­sion, appro­pri­ate use
the bur­den of daily dos­ing, this strat­egy is also cost-pro­hib­i­tive. of safety equip­ment, and suit­able foot­wear are equally impor­tant
to max­i­mize safety for these patients.44 To ensure safe par­tic­i­pa­
Bleeding trig­gers tion, a well-informed dis­cus­sion among the health care pro­vider,
The level and pat­tern of phys­i­cal activ­ity as well as under­ly­ing patient, par­ents, and coaches should take place. The National
mus­cu­lo­skel­e­tal health can act as bleed­ing trig­gers, con­trib­ut­ Hemophilia Foundation’s Playing It Safe guide may be help­ful in
ing to the bleed­ing phe­no­type. School-aged chil­dren and col­ these dis­cus­sions, as it cat­e­go­rizes the level of risk and pro­vi­des
lege stu­dents often engage in sports and should be encour­aged infor­ma­tion on safety mea­sures for >80 sport­ing activ­i­ties.39
to do so, even if some restric­tions or mod­i­fi­ca­tions are nec­es­ Hemophilia patients who have under­ ly­ing joint dam­ age
sary.39 Several ret­ro­spec­tive stud­ies con­cluded that par­tic­i­pa­tion (tar­get joints) from prior bleeds at a young age usu­ally require
in orga­nized sports (includ­ing high-risk sports such as bas­ket­ball higher fac­ tor trough lev­ els and hence higher fac­ tor use to
and foot­ball) by chil­dren and ado­les­cents with severe hemo­philia pre­vent future bleeds. The age at first joint bleed is an early
on pro­phy­laxis fac­tor replace­ment ther­apy was not asso­ci­ated indi­ca­tor of the patient’s bleed­ing phe­no­type.2 Patients who
with increased bleed­ing com­pli­ca­tions.40,41 However, depending expe­ri­ence their first joint bleed ear­lier in life tend to have
on the activ­ity, a change in the pro­phy­laxis dose and/or sched­ule higher annual CFC uti­li­za­tion and to develop more arthrop­a­thy
or an addi­tional preactivity fac­tor infu­sion may be indi­cated. Even in later years than patients who have their first joint bleed at a
though it is widely agreed that higher fac­tor lev­els are required later age.2 This high­lights the impor­tance of early pro­phy­laxis,
with higher-risk phys­i­cal activ­i­ties, clin­i­cal data on what con­sti­ as even a few bleeds prior to starting pro­phy­laxis can con­trib­
tutes a pro­tec­tive fac­tor level to safely par­tic­i­pate in high-risk ute to long-term joint dam­age.13
Prakash Singh Shekhawat
Optimal pro­phy­laxis fac­tor replace­ment ther­apy | 209
Table 3.  Estimates of min­i­mum fac­tor lev­els cor­re­spond­ing to National Hemophilia Foundation categories of risks
for phys­i­cal activ­i­ties

Minimum fac­tor lev­els (Mean ± SD)†


Risk level Examples of phys­i­cal activ­i­ties* Without joint mor­bid­ity With joint mor­bid­ity‡
1 – low Aquatics, archery, ellip­ti­cal machine, frisbee, golf, hik­ing, tai chi, 4  ±  2 7  ±  2
snor­kel­ing, sta­tion­ary bike, step­per, swim­ming, walk­ing
1.5 – low-mod­er­ate Baseball, bas­ket­ball, bicy­cling, body-sculpting class, canoe­ing, 7  ±  2 11  ±  2
cheerleading, cir­cuit train­ing, dance, fish­ing, frisbee, hik­ing,
horse­back rid­ing, indoor-cycling class, kay­ak­ing, Pilates, indoor
rock climbing, row­ing, row­ing machine, non­mo­tor­ized scooter,
skate­board­ing, ice skat­ing, inline skat­ing, ski machine, soft­ball,
step­per, strength train­ing, T-ball, tread­mill, yoga, Zumba class
2 – mod­er­ate Baseball, bas­ket­ball, bicy­cling, boot camp work­out class, 11  ±  2 12  ±  2

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/206/1851572/206lim.pdf by guest on 13 December 2021


bowl­ing, canoe­ing, cardio kick­box­ing class, cheerleading,
dance, div­ing, fish­ing, touch foot­ball, gym­nas­tics, CrossFit,
horse­back rid­ing, indoor-cycling class, Jet Ski, jumping rope,
kay­ak­ing, mar­tial arts, Pilates, river rafting, rock climbing,
run­ning/­­jog­ging, motor­ized and non­mo­tor­ized scoot­ers,
scuba div­ing, skate­board­ing, ice skat­ing, cross-coun­try ski­ing,
water ski­ing, soc­cer, soft­ball, surf­ing, ten­nis, track and field,
vol­ley­ball, yoga, Zumba class
2.5 – mod­er­ate-high Baseball, bas­ket­ball, bicy­cling, bounce houses, canoe­ing, 19  ±  2 21  ±  2
cheerleading, dance, div­ing, gym­nas­tics, CrossFit, hockey,
horse­back rid­ing, Jet Ski, kay­ak­ing, mar­tial arts, moun­tain bik­
ing, rac­quet­ball, out­door rock climbing, motor­ized and
non­mo­tor­ized scooter, scuba div­ing, skate­board­ing, ice
skat­ing, down­hill ski­ing, water ski­ing, snow­board­ing, soc­cer,
soft­ball, surf­i ng, track and field, tram­po­line, vol­ley­ball,
water polo
3 – high Bicycling, BMX rac­ing, bounce houses, box­ing, dance, div­ing, 38  ±  2 47  ±  2
tackle foot­ball, gym­nas­tics, CrossFit, hockey, Jet Ski, lacrosse,
mar­tial arts, dirt bikes, powerlifting, out­door rock climbing,
rodeo, rugby, snow­mo­bil­ing, soc­cer, tram­po­line, wres­tling
* Examples of phys­i­cal activ­i­ties based on National Hemophilia Foundation categories of risk. Depending on the inten­sity of the phys­i­cal activ­ity,
some activ­i­ties are in mul­ti­ple risk-level categories.39
†Minimum fac­tor lev­els for par­tic­i­pa­tion in phys­i­cal activ­i­ties based on an expert elic­i­ta­tion exer­cise.43
‡Joint mor­bid­ity was defined as hav­ing one or more “dam­aged joints”—ie, any joint per­ma­nently dam­aged as a result of the patient’s bleed­ing dis­or­
der or asso­ci­ated with chronic joint pain and/or lim­ited range of move­ment due to com­pro­mised joint integ­rity.43

CLINICAL CASE (Con­t in­u ed) occur­ring about twice a year, which he treated using his pro­
The deci­sion was made to start the patient on pro­phy­laxis fac­ phy­laxis 25 IU/kg dose each time. He then started col­lege and
tor replace­ ment ther­apy, given his his­ tory of 3 spon­ ta­
ne­ous returned to playing bas­ket­ball casu­ally. Because of this and a
joint bleeds. He had received recom­bi­nant SHL FVIII CFC in the part-time job to sup­port his tuition, his adher­ence to the pro­
past for treat­ment of his acute joint bleeds and opted to con­ phy­laxis regimen dropped. He missed a dose about once every
tinue with the same prod­uct. The prod­uct was on his insur­ance 2 weeks due to com­pet­ing demands on his time. He began to
com­pany’s pre­ferred drug list and was on for­mu­lary at the l­ocal expe­ri­ence more break­through joint bleeds, par­tic­u­larly in his
hos­pi­tal. He started at a dose of 25 IU/kg 3 times per week, right knee, about once every month. He returned to the clinic
to be infused prior to his bas­ket­ball prac­tices on Tues­day and to dis­cuss his options.
Thurs­day even­ings and Sat­ur­day morn­ing, to reduce the risk of
bleed­ing from bas­ket­ball injury and to reduce spon­ta­ne­ous joint
bleeds at all­times. Occasionally, when he had addi­tional bas­ket­ EHL CFCs
ball matches on Sat­ur­day even­ing, he would infuse 40 IU/kg on The high treat­ment bur­den asso­ci­ated with pro­phy­laxis fac­tor
Sat­ur­day morn­ing instead. In addi­tion, strat­e­gies to min­i­mize his replace­ment ther­apy, given the fre­quency of infu­sions using
injury risk by using eye pro­tec­tion, elbow pads and kneepads, SHL CFC, often leads to a less than opti­mal degree of adher­
mouth guards, ath­letic sup­port­ers, and proper foot­wear were ence. In the last decade, EHL prod­ucts were engineered to re-
discussed with the patient. He learned how to self-infuse, and his duce infu­sion fre­quency and to main­tain a higher fac­tor trough
bas­ket­ball coach also received infu­sion train­ing. ­level for bet­ter pro­tec­tion against spon­ta­ne­ous bleed­ing. This
Over the next 3 years, he con­tin­ued on the same reg­i­men was achieved using tech­ niques includ­ ing fusion with either
and reported no injury-related bleeds from playing bas­ket­ball. albu­min or the mono­meric Fc frag­ment of immu­no­glob­u­lin G1
He noted break­through joint bleeds involv­ing the right knee (IgG1) or con­ju­ga­tion with poly­eth­yl­ene gly­col (PEG). In the first

210  |  Hematology 2021  |  ASH Education Program


Table 4.  Characteristics of EHL CFCs

Manufacturer-recommended
Licensed for starting dose for Recommended assay for
Brand (non­pro­pri­etary name) Technology pro­phy­laxis use pro­phy­laxis use* mon­i­tor­ing
Factor VIII prod­ucts
  Eloctate®/Elocta® B-domain deleted FVIII fused US & EU: all­ages Adults: 50 IU/kg every 4 days; One stage or chro­mo­genic
(efmoroctocog alfa) to Fc por­tion of IgG1 chil­dren <6 years: 50 IU/kg
twice weekly
  Adynovate®/Adynovi® Full-length recom­bi­nant FVIII US: all­; EU: ≥12 years Adolescents/adults: 40-50 One stage or chro­mo­genic
(rurioctocog alfa pegol) with 20 kDa PEG IU/kg 2 times weekly;
chil­dren <12 years: 55 IU/kg
2 times weekly
  Jivi® (damoctocog alfa pegol) B-domain deleted FVIII with US & EU: ≥12 years 30–40 IU/kg twice weekly Chromogenic or one stage

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/206/1851572/206lim.pdf by guest on 13 December 2021


60 kDa PEG with val­i­dated reagents†
  Esperoct® (turoctocog alfa B-domain trun­cated FVIII US: all­; EU: ≥12 years Adolescents/adults: 50 Chromogenic or one stage
pegol) with 40 kDa PEG IU/kg every 4 days; with val­i­dated reagents†
chil­dren <12 years:
65 IU/kg twice weekly
  Afstyla® (lonoctocog alfa) Single-chain recom­bi­nant US & EU: all­ages Adults: 20-50 IU/kg 2-3 Chromogenic assay
FVIII times weekly; chil­dren <12
years: 30-50 IU/kg
2-3 times weekly
Factor IX prod­ucts
  Alprolix® (efrenonacog alfa) Recombinant FIX fused to Fc US & EU: all­ages Adolescents/adults: 50 IU/kg Chromogenic or one stage
por­tion of IgG1 weekly or 100 IU/kg every with val­i­dated reagents†
10 days; chil­dren <12 years:
60 IU/kg weekly
  Idelvion® (albutrepenonacog Recombinant FIX fused to US & EU: all­ages Adolescents/adults: 25-40 One stage with val­i­dated
alfa) albu­min IU/kg weekly; chil­dren <12 reagents†
years: 40-55 IU/kg weekly
  Rebinyn®/Refixia® (nonacog Recombinant FIX with 40 kDa EU: ≥12 years 40  IU/kg weekly Chromogenic or one stage
beta pegol) PEG with val­i­dated reagents†
*Recommended doses based on US pack­age insert except for Refixia®, which is licensed for pro­phy­laxis use in the EU only.
†Assay for mon­i­tor­ing based on World Federation of Hemophilia rec­om­men­da­tions.9 Further details on val­i­dated reagents are avail­­able at Gray et al
and Kitchen et al.51,52

tech­nique, the neo­na­tal Fc recep­tor inhib­its lyso­somal deg­ra­ use (Table 4).51,52 Hence, the lack of appro­pri­ate FVIII/FIX assays
da­tion of fusion pro­teins and recy­cles them back into the cir­cu­ for mon­i­tor­ing may also influ­ence prod­uct selec­tion.
la­tion,45 whereas PEGylation delays the deg­ra­da­tion and renal The avail­abil­ity of EHL CFCs has decreased the treat­ment bur­
elim­i­na­tion of its attached clot­ting fac­tor.46 A novel strat­egy to den, espe­cially for patients with HB, lead­ing to high adher­ence
increase the sta­bil­ity and affin­ity of FVIII to von Willebrand fac­ rates for pro­phy­laxis.53 The prolonged half-life of EHL CFCs trans­
tor (VWF) to reduce the risk of inhib­i­tor devel­op­ment led to lates to dos­ing twice per week or every 4 days for FVIII CFC and
the devel­op­ment of sin­gle-chain forms of FVIII.47 This strat­egy once every 7 to 14 days for FIX CFC. The dis­crep­ancy in dos­ing
resulted in the added ben­e­fit of a favor­able half-life, which al- inter­vals is due to the depen­dence of FVIII on the half-life of its
lows for dos­ing twice per week, sim­i­lar to other EHL FVIII prod­ chap­er­one VWF; thus, the prolonged half-life is mod­est at 1.5 to
ucts.48 Although this strat­egy was not intended to extend the 1.7 times the half-life of SHL FVIII CFC. This lim­i­ta­tion has been
half-life, it is included as an EHL prod­uct for the sake of com­ over­come by a new class of FVIII that builds on the Fc fusion
plete­ness. All cur­rently avail­­able EHL CFCs have been shown to tech­nol­ogy by adding a region of VWF and XTEN® poly­pep­tides,
be effi­ca­cious in the pre­ven­tion and treat­ment of bleeds with phys­i­cally decoupling it from endog­e­nous VWF.54 Early-phase
no evi­dence of any clin­i­cal safety issues,9,49 albeit the­o­ret­i­cal clin­i­cal stud­ies dem­on­strated a 3- to 4-fold increase in half-life
con­ cerns remain regard­ ing life­
long use of PEGylated CFC.50 observed, pos­si­bly reduc­ing the dos­ing fre­quency of FVIII for
This has led to vary­ing reg­u­la­tory approval for some PEGylated pro­phy­laxis to once a week.55 Phase 3 clin­i­cal tri­als are ongo­ing,
prod­ucts for pro­phy­laxis use in the pedi­at­ric pop­u­la­tion (Ta- with results expected in 2022 (NCT04161495).
ble 4). When selecting an EHL CFC, the same prac­ti­cal mea­ In selected HA patients who require higher trough lev­ els,
sures apply—local avail­abil­ity, payer pref­er­ence, and cost. For a switch to EHL CFCs admin­is­tered at the reg­u­lar dose inter­
EHL prod­ucts, an addi­tional deci­sion point for cli­ni­cians is the val (ie, every other day) used for SHL CFCs may be appro­pri­ate.
abil­ity or avail­abil­ity of local assays to mon­i­tor FVIII/FIX lev­els The pre­sumed ben­e­fit predicted from targeting higher trough
accu­rately. Certain EHL CFCs require chro­mo­genic fac­tor as- lev­els was con­firmed in the recently published PROPEL study
says or one-stage FVIII/FIX assays that have been val­i­dated for that ran­dom­ized 115 severe HA patients on pro­phy­laxis fac­tor
Prakash Singh Shekhawat
Optimal pro­phy­laxis fac­tor replace­ment ther­apy | 211
Table 5.  A prac­ti­cal guide for adopting PK assess­ment in the clinic

Question Answer
How should I sched­ule the clinic visit? Time/day of infu­sion and clinic visit should be coor­di­nated to ensure fac­tor lev­els can be drawn at
required time points to min­i­mize mul­ti­ple patient vis­its and incon­ve­nience
What are the ideal mea­sure­ment time points?* For SHL FVIII – predose, 2-4 hours, 24 ± 4 hours, 48 ± 6 hours; for SHL FIX – predose, any time on day
2 and 3; for EHL – as above and add a time point at 60-84 hours for FVIII and at 2-14 days for FIX
What pop­u­la­tion PK soft­ware is avail­­able? A glob­ally acces­si­ble online tool, such as WAPPS-Hemo, or prod­uct-spe­cific tools by the prod­uct
man­u­fac­tur­ers
What infor­ma­tion is required?* Age, base­line fac­tor level, prod­uct name, total dose admin­is­tered, body weight, height, infu­sion
day, infu­sion time, postinfusion fac­tor lev­els, and tim­ing of sam­ples
What do I do with the results? Share and explain results with patients/care­giv­ers; tai­lor treat­ment reg­i­men based on results,
if appro­pri­ate; inte­grate results into patient’s med­i­cal records

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/206/1851572/206lim.pdf by guest on 13 December 2021


*Ideal time points and infor­ma­tion required are based on WAPPS-Hemo (www​­.wapps​­-hemo​­.org). Please see prod­uct man­uf­ac­tur­ers’ man­ual if using
prod­uct-spe­cific tools. Adapted with permission from Hermans C, Dolan G. Therapeutic Advances in Hematology (Volume 11).66 © The Author(s),
2020; Reprinted by Permission of SAGE Publications.

replace­ment ther­apy using rurioctocog alfa pegol (an EHL prod­ tant esti­ma­tes in the PK pro­file are the time to a targeted (or
uct with a half-life of 14 to 16 hours) to 2 FVIII trough lev­els, 1% trough) fac­tor level or the fac­tor level at a spe­cific time after the
to 3% vs 8% to 12%.56 As expected, dur­ing the 6-month study infu­sion.64,65,67 One can then sim­u­late the effect of using dif­fer­ent
period 85% of patients in the higher-trough arm achieved zero doses and/or infu­sion fre­quen­cies to achieve the desired fac­tor
spon­ta­ne­ous joint bleeds vs 65% in the lower-trough arm. How- level or to deter­mine the appro­pri­ate­ness of performing a phys­i­
ever, achiev­ing the higher tar­get trough lev­els required a higher cal activ­ity given the fac­tor level at a spe­cific time. In my expe­ri­
bur­den of ther­apy, with 72.4% need­ing infu­sion every 24 to 48 ence, the abil­ity to deter­mine the lat­ter has pro­vided sig­nif­i­cant
hours vs 19.3% in the lower-trough arm. Notably, 60% of patients peace of mind to hemo­philia patients with active life­styles.
in the ­lower-trough arm expe­ri­enced zero spon­ta­ne­ous bleeds,
whereas 24% of those in the higher-trough arm con­tin­ued to
have spon­ta­ne­ous bleeds. This sup­ports 2 key points: (1) bleed­
ing events decrease as tar­get trough fac­tor level increases, and CLINICAL CASE (Con­tin­ued)
(2) there is a need for per­son­al­ized treat­ment. The lat­ter indi­ We discussed switching to the EHL ver­sion of the patient’s
rectly answers the ques­tion that the appro­pri­ate tar­get trough cur­rent SHL CFC. He declined to undergo a PK assess­ment on
level is that at which the patient expe­ri­ences zero bleeds while his cur­rent SHL reg­i­men but was agree­­able to doing so after
pre­serv­ing an active (or sed­en­tary) life­style. switching. He was started on an EHL CFC at 50 IU/kg once
every 4 days. After 2 months on the new reg­im ­ en, he came to
Individual PK assess­ment the clinic for a PK eval­u­a­tion that showed a half-life of 16.25
For patients with HA, there is a wide var­i­a­tion of interpatient PK hours and an esti­mated trough level of 3.3% prior to the next
han­dling of infused fac­tor depen­dent on age, body mass, blood dose. One year later, he reports zero spon­ta­ne­ous joint bleeds.
group, and VWF lev­els.57-60 PK stud­ies on interpatient han­dling
of FIX CFC have been fewer, but it is thought to have sim­i­lar
interpatient var­i­abil­ity.61,62 Hence, to opti­mize pro­phy­laxis and Monitoring real-world hemo­static effec­tive­ness
fac­tor uti­li­za­tion, the WFH cur­rently rec­om­mends indi­vid­u­al­ized Even though all­avail­­able CFCs have been shown to be effi­ca­
PK assess­ment.9 cious in clin­i­cal tri­als, con­tin­u­ous mon­i­tor­ing of hemo­static
Traditionally, obtaining PK eval­u­a­tion was a huge incon­ve­ out­comes in clin­i­cal prac­tice is recommended. Recently, it has
nience due to the need for a wash­out period (abstaining for 72 been reported that a small sub­set of HB patients on EHL FIX
hours from FVIII CFC use and 96-120 hours for FIX CFC use, thus CFCs for pro­phy­laxis expe­ri­enced unex­pected spon­ta­ne­ous
intro­duc­ing eth­i­cal con­cerns for increased bleed­ing risk) and break­through bleed­ing despite ade­quate FIX lev­els.68,69 These
fre­quent fac­tor mea­sure­ments for 48 to 72 hours after infu­sion patients had to switch back to SHL CFCs, switch to a dif­fer­ent
of a pro­phy­laxis dose. Nowadays, the avail­abil­ity of ­pop­u­la­tion-​ EHL CFC, or reduce the dos­ing inter­val from 14 days to 7 to 10
based PK mod­els, such as WAPPS-Hemo (www​­.wapps​­-hemo​ days to main­tain ade­quate hemo­static con­trol. The cause of this
­.org), enables a Bayes­ian esti­ma­tion of indi­vid­ual PK from only 2 ­unex­pected hemo­static out­come remains unclear, and fur­ther
or 3 fac­tor mea­sure­ments with­out a wash­out period and allows research is warranted.68,69 Yet this high­lights the need for con­tin­
for the increased use of PK mon­i­tor­ing in rou­tine clin­i­cal prac­ u­ous assess­ment of real-world hemo­static out­comes out­side of
tice.63,64 A prac­ti­cal guide for adopting PK assess­ ment in the clin­i­cal tri­als when newly approved treat­ment options for hemo­
clinic is shown in Table 5. philia are intro­duced in rou­tine clin­i­cal prac­tice.
A typ­i­cal PK pro­file con­tains many PK param­e­ters, includ­ing
area under the curve, in vivo recov­ery, half-life, and clear­ance. Conclusion
Detailed descrip­tions of these PK param­e­ters are described in In the era of pre­ci­sion med­i­cine, it is clear that pro­phy­laxis fac­tor
recently published review arti­cles by Delavenne and Dargaud, replace­ment ther­apy should be indi­vid­u­al­ized for opti­mal fac­
Hermans and Dolan, and Iorio.65-67 Practically, the most impor­ tor uti­li­za­tion and that “one reg­i­men does not fit all­.” Different

212  |  Hematology 2021  |  ASH Education Program


aspects of the treat­ ment, includ­ing prod­ uct type, dose, and 10. Pabinger I, Mamonov V, Windyga J, et al. Results of a ran­dom­ized phase
inter­val, as well as targeted trough lev­els and bleed­ing trig­ III/IV trial com­par­ing inter­mit­tent bolus ver­sus con­tin­u­ous infu­sion of anti-
haemophilic fac­ tor (recom­ bi­
nant) in adults with severe or mod­ er­
ately
gers, are involved in tai­lor­ing the reg­i­men to the clin­i­cal needs severe haemophilia A under­go­ing major ortho­pae­dic sur­gery. Haemo-
of the patient. One aspect not explic­itly discussed is that effec­ philia. 2021;27(3):e331-e339.
tive pro­phy­laxis is an ongo­ing col­lab­o­ra­tive effort that relies on 11. Pan-Petesch B, Nagao A, Karim FA, et al. Efficacy and safety of rIX-FP in
shared deci­sion-mak­ing between the patient and the cli­ni­cian. sur­gery: an update from a phase 3b exten­sion study. Thromb Res. 2020;​
193(Sep­tem­ber):139-141.
All of the afore­men­tioned con­sid­er­ations are irrel­e­vant if the pa-
12. Santagostino E, Lalezari S, Reding MT, et al. Safety and effi­cacy of BAY 94-
tient’s pref­er­ences and val­ues are not taken into account. As the 9027, an extended-half-life fac­tor VIII, dur­ing minor sur­gi­cal pro­ce­dures in
com­plex­ity of treat­ment options increases with the avail­abil­ity patients with severe haemophilia A. Haemophilia. 2021;27(4):e559-e562.
of non­fac­tor ther­a­pies and gene ther­apy, it is crit­i­cal that both 13. Manco-Johnson MJ, Abshire TC, Shapiro AD, et al. Prophylaxis ver­sus epi­
patients and cli­ni­cians are actively involved in this col­lab­o­ra­tive sodic treat­ment to pre­vent joint dis­ease in boys with severe hemo­philia. N
Engl J Med. 2007;357(6):535-544.
pro­cess to opti­mize treat­ment and over­all patient out­comes. 14. Gringeri A, Lundin B, von Mackensen S, Mantovani L, Mannucci PM; ESPRIT
Study Group. A ran­dom­ized clin­i­cal trial of pro­phy­laxis in chil­dren with
Acknowledgment hemo­philia A (the ESPRIT Study). J Thromb Haemost. 2011;9(4):700-710.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/206/1851572/206lim.pdf by guest on 13 December 2021


Ming Y. Lim received a 2018 Mentored Research Award from the 15. Manco-Johnson MJ, Lundin B, Funk S, et  al. Effect of late pro­ phy­laxis
Hemostasis and Thrombosis Research Society, which was sup- in hemo­ philia on joint sta­ tus: a ran­ dom­ ized trial. J Thromb Haemost.
ported by an edu­ca­tional grant from Bioverativ, a Sanofi com­pany. 2017;15(11):2115-2124.
16. Warren BB, Thornhill D, Stein J, et al. Young adult out­comes of child­hood
pro­phy­laxis for severe hemo­philia A: results of the Joint Outcome Continu-
Conflict-of-inter­est dis­clo­sure
ation Study. Blood Adv. 2020;4(11):2451-2459.
Ming Y. Lim: hon­o­rar­ium for par­tic­i­pa­tion in advi­sory boards: Sa- 17. Manco-Johnson MJ, Soucie JM, Gill JC; Joint Outcomes Committee of the
nofi Genzyme, Argenx, Dova Pharmaceuticals, Hema Biologics; Universal Data Collection, US Hemophilia Treatment Center Network. Pro-
hon­o­rar­ium and travel expenses for edu­ca­tional par­tic­i­pa­tion: phylaxis usage, bleed­ing rates, and joint out­comes of hemo­philia, 1999 to
Hemostasis and Thrombosis Research Society Trainee Workshop 2010: a sur­veil­lance pro­ject. Blood. 2017;129(17):2368-2374.
18. Farrugia A. Guide for the Assessment of Clotting Factor Concentrates. 3rd
supported by Novo Nordisk. ed. World Federation of Hemophilia; 2017. Accessed 10 May 2021. https:​­/​­/
www1​­.wfh​­.org​­/publication​­/files​­/pdf​­-1271​­.pdf.
Off-label drug use 19. World Federation of Hemophilia. World Federation of Hemophilia online
Ming Y. Lim: nothing to disclose. reg­is­try of clot­ting fac­tor con­cen­trates. Accessed 10 May 2021. https:​­/​­/
www1​­.wfh​­.org​­/custom​­/CFC​­/index​­.html.
Correspondence 20. Gouw SC, van der Bom JG, Ljung R, et al; PedNet and RODIN Study Group.
Ming Y. Lim, Division of Hematology and Hematologic Malignan- Factor VIII prod­ucts and inhib­i­tor devel­op­ment in severe hemo­philia A. N
cies, Department of Internal Medicine, University of Utah, 2000 Engl J Med. 2013;368(3):231-239.
21. Peyvandi F, Mannucci PM, Garagiola I, et al. A ran­dom­ized trial of fac­tor VIII
Circle of Hope, Rm 4126, Salt Lake City, UT 84112; e-mail: ming​ and neu­tral­iz­ing antibodies in hemo­philia A. N Engl J Med. 2016;374(21):​
­.lim@hsc​­.utah​­.edu. 2054-2064.
22. Wight J, Paisley S. The epi­de­mi­­ol­ogy of inhib­i­tors in haemophilia A: a sys­
References tem­atic review. Haemophilia. 2003;9(4):418-435.
1. Blanchette VS, Key NS, Ljung LR, et al; Subcommittee on Factor VIII, Factor 23. Franchini M, Coppola A, Rocino A, et al; Ital­ian Association of Hemophilia
IX and Rare Coagulation Disorders of the Scientific and Standardization Centers Working Group. Systematic review of the role of FVIII con­cen­trates
Committee of the International Society on Thrombosis and Hemostasis. in inhib­ i­
tor devel­ op­ment in pre­ vi­
ously untreated patients with severe
Definitions in hemo­ philia: com­mu­ni­
ca­
tion from the SSC of the ISTH. J hemo­philia a: a 2013 update. Semin Thromb Hemost. 2013;39(7):752-766.
Thromb Haemost. 2014;12(11):1935-1939. 24. Marcucci M, Mancuso ME, Santagostino E, et al. Type and inten­sity of FVIII
2. van Dijk K, Fischer K, van der Bom JG, Grobbee DE, van den Berg HM. Vari- expo­sure on inhib­i­tor devel­op­ment in PUPs with haemophilia A: a patient-
ability in clin­i­cal phe­no­type of severe haemophilia: the role of the first joint level meta-anal­y­sis. Thromb Haemost. 2015;113(5):958-967.
bleed. Haemophilia. 2005;11(5):438-443. 25. National Hemophilia Foundation. Recommendation on SIPPET (sur­vey of
3. Castaman G, Matino D. Hemophilia A and B: molec­u­lar and clin­i­cal sim­i­lar­i­ inhib­i­tors in plasma-prod­uct-exposed tod­dlers): results and rec­om­men­da­
ties and dif­fer­ences. Haematologica. 2019;104(9):1702-1709. tions for treat­ment prod­ucts for pre­vi­ously untreated patients with hemo­
4. Soucie JM, Monahan PE, Kulkarni R, Konkle BA, Mazepa MA; US Hemophilia philia A. Accessed 10 May 2021. https:​­/​­/www​­.hemophilia​­.org​­/healthcare​
Treatment Center Network. The fre­quency of joint hem­or­rhages and pro­ ­-professionals​­/guidelines​­-on​­-care​­/masac​­-documents​­/masac​­-document-​
ce­dures in nonsevere hemo­philia A vs B. Blood Adv. 2018;2(16):2136-2144. ­243​­-recommendation​­-on​­-sippet​­-survey​­-of​­-inhibitors​­-in​­-plasma​­-product​
5. Scott MJ, Xiang H, Hart DP, et  al. Treatment reg­i­mens and out­comes in ­-exposed​­-toddlers​­-results​­-and​­-recommendations​­-for​­-treatment​­-products​
severe and mod­er­ate haemophilia A in the UK: the THUNDER study. Hae- ­-for​­-previously​­-untreated​­-patients​­-with.
mophilia. 2019;25(2):205-212. 26. Euro­pean Medicines Agency. PRAC con­firms its pre­vi­ous con­clu­sion
6. Santagostino E, Mancuso ME, Tripodi A, et al. Severe hemo­philia with mild on risk of inhib­i­tor devel­op­ment with fac­tor VIII med­i­cines. Accessed 31
bleed­ing phe­no­type: molec­u­lar char­ac­ter­iza­tion and global coag­u­la­tion August 2021. https:​­/​­/www​­.ema​­.europa​­.eu​­/en​­/documents​­/referral​­/factor​
pro­file. J Thromb Haemost. 2010;8(4):737-743. ­-viii​­-article​­-31​­-referral​­-prac​­-confirms​­-its​­-previous​­-conclusion​­-risk​­-inhibitor​
7. Bos MH, Meijerman DW, van der Zwaan C, Mertens K. Does acti­vated pro­ ­-development_en​­.pdf.
tein C-resis­tant fac­tor V con­trib­ute to throm­bin gen­er­a­tion in hemo­philic 27. Thornburg C, Friedman K, Guerrera M, et al. U.S. cohort study of pre­vi­ously
plasma? J Thromb Haemost. 2005;3(3):522-530. untreated patients with con­gen­i­tal hemo­philia (ATHN 8: PUPs study): asso­
8. Zhou JY, Barnes RFW, Foster G, Iorio A, Cramer TJ, von Drygalski A. Joint ci­a­tion between fam­ily his­tory and age of diag­no­sis. Blood. 2020;136(suppl
bleed­ing ten­den­cies in adult patients with hemo­philia: it’s not all­phar­ma­ 1):39-40.
co­ki­net­ics. Clin Appl Thromb Hemost. 2019;25­(Jan­u­ary-Decem­ber):107602 28. White GC, DiMichele D, Mertens K, et  al. Utilization of pre­vi­ously treated
9619862052. patients (PTPs), non­ in­ fected patients (NIPs), and pre­ vi­
ously untreated
9. Srivastava A, Santagostino E, Dougall A, et al; WFH Guidelines for the Man- patients (PUPs) in the eval­u­a­tion of new fac­tor VIII and fac­tor IX con­cen­trates:
agement of Hemophilia pan­el­ists and coau­thors. WFH Guidelines for the rec­om­men­da­tion of the Scientific Subcommittee on Factor VIII and Factor IX
Management of Hemophilia, 3rd edi­tion. Haemophilia. 2020;26(suppl 6):1– of the Scientific and Standardization Committee of the International Society
158. on Thrombosis and Haemostasis. Thromb Haemost. 1999;81(3):462.

Prakash Singh Shekhawat


Optimal pro­phy­laxis fac­tor replace­ment ther­apy | 213
29. Dubé E, Bonnefoy A, Merlen C, et al. A pro­spec­tive sur­veil­lance study of 51. Gray E, Kitchen S, Bowyer A, et  al. Laboratory mea­ sure­ment of fac­
inhib­i­tor devel­op­ment in haemophilia A patients fol­low­ing a pop­u­la­tion tor replace­ ment ther­ a­
pies in the treat­ ment of con­ gen­ i­
tal haemophilia:
switch to a third-gen­er­a­tion B-domain-deleted recom­bi­nant fac­tor VIII. a United Kingdom Haemophilia Centre Doctors’ Organisation guide­line.
Haemophilia. 2018;24(2):236-244. Haemophilia. 2020;26(1):6-16.
30. Sidonio RF, Cheng D, Guelcher C, et al. Lack of inhib­i­tor devel­op­ment in 52. Kitchen S, Tiefenbacher S, Gosselin R. Factor activ­ity assays for mon­it­ or­ing
the Amer­i­can throm­bo­sis and hemo­sta­sis net­work (ATHN)-2 fac­tor switch- extended half-life FVIII and fac­tor IX replace­ment ther­a­pies. Semin Thromb
ing study: pre­lim­i­nary report of pri­mary out­come. Blood. 2019;134(suppl Hemost. 2017;43(3):331-337.
1):1114. 53. Mancuso ME, Oldenburg J, Boggio L, et al. High adher­ence to pro­phy­laxis
31. Collins PW, Björkman S, Fischer K, et  al. Factor VIII require­ ment to reg­i­mens in haemophilia B patients receiv­ing rIX-FP: evi­dence from clin­ic ­ al
main­tain a tar­get plasma level in the pro­phy­lac­tic treat­ment of severe tri­als and real-world prac­tice. Haemophilia. 2020;26(4):637-642.
hemo­philia A: influ­ences of var­i­ance in phar­ma­co­ki­net­ics and treat­ment 54. Seth Chhabra E, Liu T, Kulman J, et al. BIVV001, a new class of fac­tor VIII
reg­i­mens. J Thromb Haemost. 2010;8(2):269-275. replace­ment for hemo­philia A that is inde­pen­dent of von Willebrand fac­tor
32. Collins PW, Fischer K, Morfini M, Blanchette VS, Björkman S; International in pri­ma­tes and mice. Blood. 2020;135(17):1484-1496.
Prophylaxis Study Group Pharmacokinetics Expert Working Group. Impli- 55. Konkle BA, Shapiro AD, Quon DV, et al. BIVV001 fusion pro­tein as fac­tor VIII
cations of coag­u­la­tion fac­tor VIII and IX phar­ma­co­ki­net­ics in the pro­phy­lac­ replace­ment ther­apy for hemo­philia A. N Engl J Med. 2020;383(11):1018–
tic treat­ment of haemophilia. Haemophilia. 2011;17(1):2-10. 1027.
33. Gouider E, Jouini L, Achour M, et al. Low dose pro­phy­laxis in Tunisian chil­­ 56. Klamroth R, Windyga J, Radulescu V, et  al. Rurioctocog alfa pegol

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/206/1851572/206lim.pdf by guest on 13 December 2021


dren with haemophilia. Haemophilia. 2017;23(1):77-81. PK-guided pro­ phy­laxis in hemo­ philia A: results from the phase 3
34. Collins PW, Blanchette VS, Fischer K, et al; rAHF-PFM Study Group. Break- PROPEL study. Blood. 2021;137(13):1818-1827.
through bleed­ ing in rela­ tion to predicted fac­ tor VIII lev­
els in patients 57. Björkman S, Folkesson A, Jönsson S. Pharmacokinetics and dose require­
receiv­ing pro­phy­lac­tic treat­ment for severe hemo­philia A. J Thromb Hae- ments of fac­tor VIII over the age range 3-74 years: a pop­ul­a­tion anal­y­sis
most. 2009;7(3):413-420. based on 50 patients with long-term pro­phy­lac­tic treat­ment for haemo-
35. Ahnström J, Berntorp E, Lindvall K, Björkman S. A 6-year fol­low-up of dos­ philia A. Eur J Clin Pharmacol. 2009;65(10):989-998.
ing, coag­u­la­tion fac­tor lev­els and bleed­ings in rela­tion to joint sta­tus in the 58. Fischer K, Pendu R, van Schooten CJ, et al. Models for pre­dic­tion of fac­tor
pro­phy­lac­tic treat­ment of haemophilia. Haemophilia. 2004;10(6):689-697. VIII half-life in severe haemophiliacs: dis­tinct approaches for blood group
36. den Uijl I, Biesma D, Grobbee D, Fischer K. Outcome in mod­er­ate haemo- O and non-O patients. PLoS One. 2009;4(8):e6745.
philia. Blood Transfus. 2014;12(suppl 1):s330-s336. 59. Henrard S, Speybroeck N, Hermans C. Impact of being under­weight or
37. den Uijl IE, Fischer K, Van Der Bom JG, Grobbee DE, Rosendaal FR, Plug I. over­weight on fac­tor VIII dos­ing in hemo­philia A patients. Haematologica.
Analysis of low fre­quency bleed­ing data: the asso­ci­a­tion of joint bleeds 2013;98(9):1481-1486.
according to base­line FVIII activ­ity lev­els. Haemophilia. 2011;17(1):41- 60. Tang L, Leong L, Sim D, et al. von Willebrand fac­tor con­trib­utes to lon­ger
44. half-life of PEGylated fac­tor VIII in vivo. Haemophilia. 2013;19(4):539-545.
38. Chowdary P, Fischer K, Collins PW, et  al. Modeling to pre­dict fac­tor VIII 61. Björkman S, Ahlén V. Population phar­ma­co­ki­net­ics of plasma-derived fac­
lev­els asso­ci­ated with zero bleeds in patients with severe hemo­philia A tor IX in adult patients with haemophilia B: impli­ca­tions for dos­ing in pro­
ini­ti­ated on ter­tiary pro­phy­laxis. Thromb Haemost. 2020;120(5):728-736. phy­laxis. Eur J Clin Pharmacol. 2012;68(6):969-977.
39. Anderson A, Forsyth A; National Hemophilia Foundation. Playing It Safe: 62. Iorio A, Fischer K, Blanchette V, et al; Pharmacokinetic Expert Work-
Bleeding Disorders, Sports and Exercise. National Hemophilia Founda- ing Group of the International Prophylaxis Study Group. Tailoring treat­
tion; 2017. Accessed 10 May 2021. https:​­/​­/www​­.changinghemophilia​­.com​ ment of haemophilia B: account­ ing for the dis­ tri­
bu­tion and clear­ ance
­/content​­/dam​­/biopharm​­/changinghemophilia​­/managing​­-hemophilia​­/ of stan­dard and extended half-life FIX con­cen­trates. Thromb Haemost.
lifestyle​­-tips​­/sports​­-and​­-exercise​­/Playing​­-It​­-Safe_0​­.pdf. 2017;117(6):1023-1030.
40. Ross C, Goldenberg NA, Hund D, Manco-Johnson MJ. Athletic par­tic­i­pa­ 63. WAPPS-Hemo Research Network. Web-acces­si­ble pop­u­la­tion phar­ma­co­
tion in severe hemo­philia: bleed­ing and joint out­comes in chil­dren on pro­ ki­netic ser­vice—hemo­philia. Accessed 14 May 2021. https:​­/​­/www​­.wapps​
phy­­laxis. Pediatrics. 2009;124(5):1267-1272. ­-hemo​­.org.
41. Versloot O, Timmer MA, de Kleijn P, et al. Sports par­tic­i­pa­tion and sports 64. Iorio A, Blanchette V, Blatny J, Collins P, Fischer K, Neufeld E. Estimating
injuries in Dutch boys with haemophilia. Scand J Med Sci Sports. 2020;30(7):​ and interpreting the phar­ma­co­ki­netic pro­files of indi­vid­ual patients with
1256-1264. hemo­philia A or B using a pop­u­la­tion phar­ma­co­ki­netic approach: com­mu­
42. Iorio A, Iserman E, Blanchette V, et al. Target plasma fac­tor lev­els for per­ ni­ca­tion from the SSC of the ISTH. J Thromb Haemost. 2017;15(12):2461–
son­al­ized treat­ment in haemophilia: a Delphi con­sen­sus state­ment. Hae- 2465.
mophilia. 2017;23(3):e170-e179. 65. Delavenne X, Dargaud Y. Pharmacokinetics for haemophilia treat­ers: mean­
43. Martin AP, Burke T, Asghar S, Noone D, Pedra G, O’Hara J. Understanding ing of PK param­e­ters, inter­pre­ta­tion pit­falls, and use in the clinic. Thromb
min­i­mum and ideal fac­tor lev­els for par­tic­i­pa­tion in phys­i­cal activ­i­ties by Res. 2020;192(May):52-60.
peo­ple with haemophilia: an expert elic­i­ta­tion exer­cise. Haemophilia. 66. Hermans C, Dolan G. Pharmacokinetics in rou­ tine haemophilia clin­ i­
cal
2020;26(4):711-717. prac­tice: ratio­nale and modal­i­ties—a prac­ti­cal review. Ther Adv Hematol.
44. McGee S, Raffini L, Witmer C. Organized sports par­tic­i­pa­tion and the asso­ 2020;11(Octo­ber):2040620720966888.
ci­a­tion with injury in pae­di­at­ric patients with haemophilia. Haemophilia. 67. Iorio A. Using phar­ma­co­ki­net­ics to indi­vid­u­al­ize hemo­philia ther­apy.
2015;21(4):538-542. Hematology Am Soc Hematol Educ Program. 2017;2017(1):595-604.
45. Roopenian DC, Akilesh S. FcRn: the neo­na­tal Fc recep­tor comes of age. Nat 68. Kleiboer B, Nielsen B, Ma AD, Abajas Y, Monroe DM, Key NS. Excessive
Rev Immunol. 2007;7(9):715-725. break­ through bleed­ ing in haemophilia B patients on fac­ tor IX-albu­
min
46. Ivens IA, Baumann A, McDonald TA, Humphries TJ, Michaels LA, Mathew fusion pro­tein pro­phy­lac­tic ther­apy: a sin­gle cen­tre case series. Haemo-
P. PEGylated ther­a­peu­tic pro­teins for haemophilia treat­ment: a review for philia. 2020;26(1):e23-e25.
haemophilia care­giv­ers. Haemophilia. 2013;19(1):11-20. 69. Malec LM, Croteau SE, Callaghan MU, Sidonio RF Jr. Spontaneous bleed­
47. Mahlangu J, Kuliczkowski K, Karim FA, et al; AFFINITY Investigators. Efficacy ing and poor bleed­ing response with extended half-life fac­tor IX prod­
and safety of rVIII-sin­gle chain: results of a phase 1/3 mul­ti­cen­ter clin­i­cal ucts: a sur­vey of select US haemophilia treat­ment cen­tres. Haemophilia.
trial in severe hemo­philia A. Blood. 2016;128(5):630-637. 2020;26(3):e128-e129.
48. Zhang Y, Roberts J, Tortorici M, et  al. Population phar­ma­co­ki­net­ics of
recom­bi­nant coag­u­la­tion fac­tor VIII-sin­gle chain in patients with severe
hemo­philia A. J Thromb Haemost. 2017;15(6):1106-1114.
49. Mahlangu J, Young G, Hermans C, Blanchette V, Berntorp E, Santagostino
E. Defining extended half-life rFVIII—a crit­i­cal review of the evi­dence. Hae-
mophilia. 2018;24(3):348-358.
50. Lubich C, Allacher P, de la Rosa M, et al. The mys­tery of antibodies against
poly­eth­yl­ene gly­col (PEG)—what do we know? Pharm Res. 2016;33(9):​ © 2021 by The Amer­i­can Society of Hematology
2239-2249. DOI 10.1182/hema­tol­ogy.2021000310

214  |  Hematology 2021  |  ASH Education Program


HEMOPHILIA UPDATE: OUR CUP RUNNETH OVER

EVIDENCE-BASED MINIREVIEW

For overweight or obese persons with


hemophilia A, should factor VIII dosing be based

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/215/1851813/215machin.pdf by guest on 13 December 2021


on ideal or actual body weight?
Nicoletta Machin1,2 and Ming Y. Lim3
Department of Medicine, Division of Hematology/Oncology, University of Pittsburgh, Pittsburgh, PA; 2Hemophilia Center of Western Pennsylvania,
1

Pittsburgh, PA; and 3Department of Internal Medicine, Division of Hematology and Hematologic Malignancies, University of Utah, Salt Lake City, UT

LEARNING OBJECTIVES
• Review evidence of the impact of body weight on FVIII in vivo recovery
• Review evidence of using ideal body weight for FVIII dosing in overweight and obese persons with hemophilia A

CLINICAL CASE the person’s physical build does not markedly differ from
A 35­year­old man with severe hemophilia A and a body mass the average. Despite this caveat, the IVR of 2 has been
index (BMI) of 38 (height, 180 cm; weight, 123 kg) presents adopted widely for FVIII dose calculations irrespective of
for his annual comprehensive care visit. He is on prophylaxis body composition. With over 31% of the US hemophilia
using factor replacement therapy with a recombinant stan­ population classified as obese, the appropriateness of an
dard half­life coagulation factor VIII (FVIII) product that is IVR of 2 as a “one­size­fits­all” approach in this population
dosed according to actual body weight (ABW). He reports is questionable.2
zero spontaneous joint bleeds over the past 12 months. He
has an upcoming elective laparoscopic cholecystectomy Impact of body composition on FVIII IVR
for symptomatic cholelithiasis. His hematologist wonders if In persons who are overweight or obese, the rise in BW is
his perioperative dosing as well as prophylaxis dosing of primarily the result of increased adipose tissue, which con­
FVIII should be adjusted considering his increased BW. tains less vascular space than lean mass. The plasma volume
per kilogram of BW decreases as BW increases, resulting in
a lower plasma volume per kilogram of BW.3 Since FVIII is
Introduction primarily restricted to the vascular compartment, the same
For persons with hemophilia A, factor replacement therapy amount of FVIII concentrate (in units per kilogram) infused
with FVIII concentrates is the cornerstone of treatment for in an obese person likely results in a higher circulating plas­
managing and preventing bleeding episodes. FVIII concen­ ma FVIII level compared to a nonobese person.4
trate is typically dosed on a per­kilogram basis. The number Several clinical studies have demonstrated that FVIII IVR
of FVIII units needed to achieve a desired circulating FVIII increases with increasing BMI and BW (Table 1).5-8 The IVR
level is empirically calculated using the following formula: is calculated by

FVIII dose =
(
BW in kg  ×  desired FVIII increase  IU
dL ) IVR =
(
BW in kg  ×   observed FVIII recovery in IU
dL
−preinfusion FVIII in IU
dL  )
2 FVIII dose in IU

This formula is based on a FVIII in vivo recovery (IVR) of In addition, a population pharmacokinetic (PK) model­
2, which assumes that each unit of FVIII concentrate infused ing study found that changes in ideal body weight (IBW)
per kilogram of BW increases the circulating FVIII level by account for the most significant amount of interindivid­
2 IU/dL.1 The IVR of 2 was first described in 1981 by Ingram ual FVIII PK variability.9 Given these findings, this minire­
based on data from 19 persons with hemophilia A with an view seeks to compare the evidence for IBW vs ABW for
ABW between 27 and 91 kg.1 He concluded that the dose the dosing of FVIII in overweight and obese persons with
calculation using a plasma volume Prakash
of 0.5  dL/kgSingh hemophilia A.
appliesShekhawat
if

IBW vs ABW dosing for FVIII | 215


Table 1. Summary of clin­i­cal stud­ies eval­u­at­ing FVIII IVR strat­i­fied by BMI

Study design Intervention (median IU, Strongest pre­dic­tor


Reference (age, years) n range) BMI groups (kg/m2) (n) IVR (IU dL−1/IU kg−1) of IVR
Henrard et al5 Prospective obser­va­tional 46 A dose of rFVIII (2000, 18.5-24.9 (26) 1.88 BW
(mean, 40.4 ± 12.3) 980-4200) 25.0-29.9 (14) 2.30
≥30.0 (6) 2.70
Henrard et al6 Retrospective pooled 201 A dose of rFVIII (3745, <18.5 (9) 1.72 BMI
anal­y­sis of 8 PK tri­als 1953-8794) 18.5-24.9 (105) 2.03
(median, 26; IQR, 25.0-29.9 (52) 2.18
21-38) >30.0 (35) 2.68
Henrard et al7 Retrospective pool 66 A dose of rFVIII (2778, Normal (43) 1.93 BMI-for-age
anal­y­sis of 6 PK tri­als 1675-5420) Overweight (7) 2.12
(median, 14.5; IQR, Obese (16)b 2.65

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/215/1851813/215machin.pdf by guest on 13 December 2021


12.8-15.6)a
Tiede et al8 Prospective obser­va­tional 35 rFVIII 50 IU/kg by ABW <18.5 (5) 2.2c BMI
(mean, 37.4; range, 18.5-24.9 (7) 2.9c
23.0–57.0) 25.0-29.9 (9) 2.9c
30.0-34.9 (7) 3.2c
≥35 (7) 3.5c
a
Only trial in chil­dren.
b
Based on the BMI-for-age per­cen­tiles (nor­mal, 5th-84th; over­weight, 85th-94th; obese, >95th).
c
End point reported as IVR at 30 min­utes using a geo­met­ric mean.
IQR, interquartile range.

Methods 41.8) and com­pared actual FVIII recov­ery lev­els with expected-
We conducted a sys­tem­atic search of MEDLINE, Embase, and the recov­ery lev­els after a FVIII 50 IU/kg dose.11 Participants used
Cochrane Database of Systematic Reviews from 1 Jan­u­ary 1981 their per­sonal brand of fac­tor, and both stan­dard and extended
to 10 May 2021. Our search included MeSH and the key words half-life prod­ ucts were included. With ABW dos­ ing, 15 of 16
“hemo­philia A,” “phar­ma­co­ki­netic,” and “dose” and yielded 830 patients achieved above-expected recov­ery rang­ing from 0.1 to
abstracts (Figure 1). References of the resulting stud­ies and nar­ 0.96 above expected (mean, 0.44). In the IBW dos­ing arm, 8 of 16
ra­tive reviews were screened. There were 594 unique arti­cles patients achieved above-expected recov­ery (range, 0.01-0.37).
after 236 dupli­cates were removed. Of the unique arti­cles, 181 Of the 8 patients who did not achieve above-expected recov­ery,
were review papers, 117 were eval­ua ­ t­ing new fac­tor con­cen­ 6 were within 10% from expected-recov­ery.
trates, 87 were case reports and alter­na­tive trial designs, 68 in­ The sec­ond cross­over trial, a 3 × 3 × 3 design of ABW vs IBW vs
volved non­fac­tor ther­apy, 49 involved inhib­i­tor patients, 38 were lean body mass (LBM), was conducted in 19 adults (mean BMI,
not on hemo­philia A, and 27 had no weight infor­ma­tion. Full-text 29.5) to deter­mine which descrip­tors of BW achieved the tar­
reviews by 2 inde­pen­dent review­ers were conducted on the 27 geted IVR of 2 with bet­ter pre­ci­sion.12 The mean IVR postrecom­
remaining arti­cles, and 3 stud­ies were ulti­mately included: 1 ret­ binant fac­tor VIII (rFVIII) infu­sion for ABW-, LBM-, and IBW-based
ro­spec­tive obser­va­tional study and 2 cross­over tri­als.10-12 dos­ing was 2.46, 2.22, and 2.29, respec­tively. The pro­por­tion of
par­tic­i­pants with an IVR of 2.00% ± 10% was 31.1%, 44.2%, and
Results 49.0% for ABW-, LBM-, and IBW-based dos­ing, respec­tively.
All three stud­ies used dif­fer­ent out­come mea­sures: peak FVIII
lev­els after a 50 IU/kg dose and clin­i­cal effec­tive­ness10; per­cent­ Discussion
age of actual FVIII recov­ery lev­els over expected lev­els after a Our sys­tem­atic minireview iden­ti­fied 2 pro­spec­tive cross­over
50 IU/kg dose11; and FVIII IVR (Table 2).12 Graham et al performed stud­ies that directly com­pared IBW to ABW dos­ing of FVIII con­
a ret­ro­spec­tive study reporting peak FVIII lev­els for 6 adults cen­trates and 1 ret­ro­spec­tive study that com­pared IBW dos­ing
with BMIs ≥30 treated with recom­bi­nant FVIII 50 IU/kg dosed to his­tor­i­cal ABW dos­ing in over­weight and obese per­sons with
by IBW.10 These patients had been previously receiving recom­ hemo­philia A.10-12 A con­cern with using IBW is the risk of under­
binant FVIII at 50 IU/kg dosed by ABW for prophylaxis or were dosing, resulting in sub­ther­a­peu­tic FVIII lev­els and increased risk
undergoing surgery. With IBW dos­ ing, 5 of the 6 patients of bleed­ ing. Overall, these 3 stud­ ies dem­ on­strated favor­
able
achieved expected mean peak FVIII lev­els of 100%. The patient out­comes using dif­fer­ent PK mea­sures when dosed by IBW and
with the low­est peak level (78%) had the highest weight (151   kg, sup­port the use of IBW to per­form indi­vid­u­al­ized PK anal­y­sis in
BMI 45.8) and was retreated with IBW plus 25%, resulting in a over­weight and obese per­sons with hemo­philia A.
peak FVIII level of 107%. During the 3-month pro­phy­laxis and/or Despite favor­able PK mea­sures, a clin­i­cal con­cern is hemo­static
sur­gi­cal period for each patient, there was no increase in spon­ta­ effec­tive­ness when using IBW for dos­ing FVIII con­cen­trates. Of
ne­ous or trau­matic bleeds and no unex­pected bleed­ing or trans­ the stud­ies iden­ti­fied, only the ret­ro­spec­tive study by Graham et
fu­sion require­ment with sur­gery. al pro­vided 3 months of data on hemostastic effec­tive­ness for the
Blair et al conducted a cross­over trial of both ABW and IBW IBW dos­ing strat­egy.10 Even though the hemo­static effec­tive­ness
dos­ing in 16 adults and chil­dren (median BMI 33.1; range, 25.6- of IBW dos­ing was favor­able in both the perioperative and pro­

216  |  Hematology 2021  |  ASH Education Program


830 records idenfied through MEDLINE (156),
EMBASE (459) and Cochrane database (215)

594 records screened by abstract a‚er 236


duplicates removed 567 arcles excluded

• Review papers (181)

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/215/1851813/215machin.pdf by guest on 13 December 2021


• New factor product study (117)
• Other trial designs/ case reports (87)
• Nonfactor therapy (68)
• Inhibitor paents (49)
• Wrong disease (38)
27 full-text reviewed for eligibility • No weight informaon (27)

24 arcles excluded

• No weight comparison (18)


• Review papers (2)
• Other trial designs (3)
• Alternave dosing schedule (1)

3 arcles included

Figure 1. Study flow diagram.

Table 2. Findings of 3 stud­ies eval­u­at­ing out­comes based on IBW

Mean (range)
Peak FVIII level or Achieved peak FVIII
Study design expected recov­ery level or expected
Reference (age, years) n Intervention ABW (kg) BMI IBW or IVR recov­ery or IVR
Grahamet al 10
Retrospective 6 rFVIII 50 IU/kg 116 (91-151) 36.0 (30.0-45.8) 74.4 (67.0– Peak FVIII level 100% 83% (5 of 6)a
(median, by IBW 84.5) (78-123)
33; range,
25-47)
Blair et al11 Prospective 16 FVIII 50 IU/kg NR Median, 33.1 (25.6-41.8) NR ABW, 140% expected ABW, 94% (15 of 16)b;
cross­over (±20%) by recov­ery (83-196); IBW, 87.5% (14 of 16)b
(median, both ABW IBW, 100% expected
21.5; range, and IBW recov­ery (56-137)
12-53)
Seaman et al12 Prospective 19 A dose of rFVIII 93.0 ± 13.6 29.2 ± 3.5 (Max, 38.3) 73.6 ± 6.3 ABW: IVR, 2.46; ABW, 31.1%c;
cross­over based on IBW: IVR, 2.29; IBW, 49.0%c;
(mean, ABW, IBW, LBM: IVR, 2.22 LBM, 44.2%c
34.6 ± 11.3) and LBM and
an IVR of 2.0
a
Above expected peak FVIII level of 100% or within 10% below expected peak FVIII level.
b
Above expected recov­ery or within 10% below expected recov­ery.
c
Within 10% of targeted IVR of 2.00.
LBW, lean body weight.
Prakash Singh Shekhawat
IBW vs ABW dos­ing for FVIII  |  217
phy­laxis set­ting, this study was lim­ited by the small sam­ple size of sis Research Society Trainee Workshop supported by Novo Nordisk.
6 adults. Particularly in the perioperative set­ting, the appli­ca­tion Nicoletta Machin: research funding: Takeda Pharmaceuticals.
of IBW dos­ing seems appeal­ing to avoid over­treat­ment with the
unnec­es­sary high FVIII peaks asso­ci­ated with ABW dos­ing, which Off-label drug use
could lead to com­pli­ca­tions such as throm­bo­sis and inhib­i­tor Ming Y. Lim: nothing to disclose.
devel­op­ment, espe­cially in per­sons with nonsevere hemo­philia Nicoletta Machin: nothing to disclose.
A who have life­long inhib­i­tor risk.13 A case-con­trol study found
that inten­sive FVIII treat­ment was a risk fac­tor for inhib­it­ or devel­ Correspondence
op­ment in per­sons with nonsevere hemo­philia A even after more Ming Y. Lim, Division of Hematology and Hematologic Malignan­
than 50 expo­sure days.14 Larger cohort stud­ies with long-term cies, Department of Internal Medicine, University of Utah, 2000
fol­low-up are crit­i­cal to fully eval­u­ate the effec­tive­ness of dos­ing Circle of Hope, Rm 4126, Salt Lake City, UT 84112; e-mail: ming​
FVIII by IBW in both the perioperative and pro­phy­laxis set­ting. ­.lim@hsc​­.utah​­.edu.
Additionally, given the cost of FVIII con­cen­trates, dos­ing based

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/215/1851813/215machin.pdf by guest on 13 December 2021


on IBW is a cost-sav­ings approach if hemo­static effec­tive­ness is References
com­pa­ra­ble to ABW dos­ing. Graham et al cal­cu­lated that IBW 1. Ingram GI. Calculating the dose of fac­tor VIII in the man­age­ment of haemo­
philia. Br J Haematol. 1981;48(2):351-354.
dos­ing resulted in a 48.9% reduc­tion in FVIII usage dur­ing the
2. Wilding J, Zourikian N, Di Minno M, et al. Obesity in the global haemophilia
3-month period, with an annu­al­ized mean sav­ings of $133,000 per pop­u­la­tion: prev­a­lence, impli­ca­tions and expert opin­ions for weight man­
patient.10 However, with­out robust effec­tive­ness data, it is dif­fi­ age­ment. Obes Rev. 2018;19(11):1569-1584.
cult to judge the cost-effec­tive­ness of IBW dos­ing at this time. 3. Feldschuh J, Enson Y. Prediction of the nor­mal blood vol­ume: rela­tion of
While body com­po­si­tion has a dem­on­strated impact on FVIII blood vol­ume to body hab­i­tus. Circulation. 1977;56(suppl 4, pt 1):605-612.
4. Björkman S, Berntorp E. Pharmacokinetics of coag­u­la­tion fac­tors: clin­i­
PKs, it should be remem­bered that other fac­tors may play a role cal rel­e­vance for patients with haemophilia. Clin Pharmacokinet. 2001;​
and should also be con­sid­ered.15 Age has been iden­ti­fied as an 40(11):815-832.
inde­pen­dent mod­i­fier of FVIII clear­ance, with increased clear­ 5. Henrard S, Speybroeck N, Hermans C. Body weight and fat mass index as
ance seen in early child­hood.16 von Willebrand fac­tor lev­els and strong pre­dic­tors of fac­tor VIII in vivo recov­ery in adults with hemo­philia A.
J Thromb Haemost. 2011;9(9):1784-1790.
the ABO blood group are noted to alter FVIII clear­ance but were
6. Henrard S, Speybroeck N, Hermans C. Impact of being under­weight or
not found to be sig­nif­i­cant deter­mi­na­tes of IVR in the stud­ies over­weight on fac­tor VIII dos­ing in hemo­philia A patients. Haematologica.
reviewed.5,6,8 Individual PKs also vary between fac­tor concen­ 2013;98(9):1481-1486.
trate products (recom­ bi­
nant, plasma-derived, and extended 7. Henrard S, Hermans C. Impact of being over­weight on fac­tor VIII dos­ing in
half-life), and the stud­ies included in this review only eval­u­ated chil­dren with haemophilia A. Haemophilia. 2016;22(3):361-367.
8. Tiede A, Cid AR, Goldmann G, et al. Body mass index best pre­dicts recov­
recom­ bi­
nant prod­ ucts. Overall, even though numer­ ous indi­ ery of recom­bi­nant fac­tor VIII in under­weight to obese patients with severe
vid­ual fac­tors can account for inter­in­di­vid­ual PK pro­files, body haemophilia A. Thromb Haemost. 2020;120(2):277-288.
com­po­si­tions appear to be the stron­gest deter­mi­nate.5,6,8 9. van Moort I, Preijers T, Hazendonk HCAM, et al; OPTI-CLOT Study Group.
In sum­mary, although con­clu­sive rec­om­men­da­tions can­not Dosing of fac­tor VIII con­cen­trate by ideal body weight is more accu­rate
in over­weight and obese haemophilia A patients. Br J Clin Pharmacol.
be made based on the small num­ber of stud­ies and the lack of
2021;87(6):2602-2613.
stan­dard­i­za­tion of out­comes, we sug­gest the fol­low­ing: 10. Graham A, Jaworski K. Pharmacokinetic anal­y­sis of anti-hemo­philic fac­tor in
the obese patient. Haemophilia. 2014;20(2):226-229.
1. For overweight and obese persons with hemophilia A, we
11. Blair A, Felgenhauer J, Recht M, Kruse-Jarres R. Comparison of ideal ver­sus
suggest that an individualized PK analysis be performed using actual body weight fac­tor dos­ing in hemo­philia A [ab­stract]. Haemophilia.
IBW to determine adequate FVIII in vivo recovery (Grade 2B). 25(S2): 3-77.
2. If there is adequate FVIII in vivo recovery, we suggest using an 12. Seaman CD, Yabes JG, Lalama CM, Ragni MV. Factor VIII con­cen­trate dos­ing
IBW dosing strategy in the perioperative setting (Grade 2C). with lean body mass, ideal body weight and total body weight in over­
weight and obe­sity: a ran­dom­ized, con­trolled, open-label, 3 × 3 cross­over
3. If there is adequate FVIII in vivo recovery, we suggest using trial. Haemophilia. 2021;27(3):351-357.
an IBW dosing strategy in the prophylaxis setting (Grade 2C). 13. Eckhardt CL, van Velzen AS, Peters M, et al; INSIGHT Study Group. Fac­
We empha­size the need for larger and long-term clin­i­cal tor VIII gene (F8) muta­tion and risk of inhib­i­tor devel­op­ment in nonsevere
stud­ies to deter­mine the hemo­static effec­tive­ness of using hemo­philia A. Blood. 2013;122(11):1954-1962.
14. Abdi A, Eckhardt CL, van Velzen AS, et al; INSIGHT Study Group. Treat­
an IBW dos­ing strat­egy in the perioperative and pro­phy­laxis
ment-related risk fac­tors for inhib­i­tor devel­op­ment in non-severe hemo­
set­ting. philia A after 50 cumu­la­tive expo­sure days: a case-con­trol study. J Thromb
Haemost. 2021;19(9):2171-2181.
15. Iorio A. Using phar­ma­co­ki­net­ics to indi­vid­u­al­ize hemo­philia ther­apy. Hema-
Acknowledgment  tology Am Soc Hematol Educ Program. 2017;2017(1):595-604.
Ming Y. Lim received a 2018 Mentored Research Award from the 16. Björkman S. Comparative phar­ma­co­ki­net­ics of fac­tor VIII and recom­bi­nant
Hemostasis and Thrombosis Research Society, which was sup­ fac­tor IX: for which coag­u­la­tion fac­tors should half-life change with age?
Haemophilia. 2013;19(6):882-886.
ported by an edu­ca­tional grant from Bioverativ, a Sanofi com­pany.

Conflict-of-inter­est dis­clo­sure
Ming Y. Lim: advi­sory board: Sanofi Genzyme, Argenx, Dova Phar­ © 2021 by The Amer­i­can Society of Hematology
maceuticals, Hema Biologics; hon­o­raria: Hemostasis and Thrombo­ DOI 10.1182/hema­tol­ogy.2021000317

218  |  Hematology 2021  |  ASH Education Program


HEMOPHILIA UPDATE: OUR CUP RUNNETH OVER

Factor-mimetic and rebalancing therapies


in hemophilia A and B: the end of factor
concentrates?

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/219/1851769/219ellsworth.pdf by guest on 13 December 2021


Patrick Ellsworth and Alice Ma
Department of Medicine, Division of Hematology, University of North Carolina, Chapel Hill, NC

Hemophilia A (HA) and B are inherited bleeding disorders caused by a deficiency of factor VIII or factor IX, respectively.
The current standard of care is the administration of recombinant or purified factor. However, this treatment strategy
still results in a high economic and personal burden to patients, which is further exacerbated by the development of
inhibitors—alloantibodies to factor. The treatment landscape is changing, with nonfactor therapeutics playing an increas-
ing role in what we consider to be the standard of care. Emicizumab, a bispecific antibody that mimics the function of
factor VIIIa, is the first such nonfactor therapy to gain US Food and Drug Administration approval and is rapidly changing
the paradigm for HA treatment. Other therapies on the horizon seek to target anticoagulant proteins in the coagulation
cascade, thus “rebalancing” a hemorrhagic tendency by introducing a thrombotic tendency. This intricate hemostatic
balancing act promises great things for patients in need of more treatment options, but are these other therapies going
to replace factor therapy? In light of the many challenges facing these therapies, should they be viewed as a replacement
of our current standard of care? This review discusses the background, rationale, and potential of nonfactor therapies as
well as the anticipated pitfalls and limitations. This is done in the context of a review of our current understanding of the
many aspects of the coagulation system.

LEARNING OBJECTIVES
• Describe the basis of emicizumab action
• Describe potential targets used in rebalancing therapies for hemophilia treatment

Introduction in hemophilia in subsequent years.3 However, even with


Congenital hemophilia A (HA) and B (HB) are inherited infectious concerns all but eradicated in new patients,
bleeding disorders caused by a deficiency of factor VIII treatment with factor products poses a risk of developing
(FVIII) and factor IX (FIX), respectively. Many barriers have inhibitors, which are neutralizing alloantibodies to exog­
been overcome leading up to our current standard of care enous FVIII or FIX proteins recognized as foreign by the
in the form of modern factor therapies.1 body’s immune system.
The administration of factor concentrates (derived Inhibitor development remains a significant complica­
from human plasma) to replace the missing factor is the tion in the treatment of patients with HA and HB and leads
most straightforward treatment approach and started in to bleeding despite factor therapy. The phenomenon is
the 1970s. However, many hemophilia patients fell victim more common in severe HA than in nonsevere HA, with an
to the HIV pandemic, when contaminated factor products incidence of 25% to 30%.4 In HB, inhibitor incidence is 3% to
infected many patients. With the sequencing and cloning 5% in general but is higher in populations enriched for null
of the FVIII and FIX genes in the 1980s and technological mutations.5 Even with attempted immune tolerance induc­
advances to inactivate and purify concentrates, modern tion and immunosuppression, inhibitors recur in up to 30%
recombinant and plasma­derived factor products ushered of HA and 20% of HB patients.5-7 First­line bleed treatment in
in a new era of treatment.1,2 Recombinant and plasma­ inhibitor patients is generally with bypassing agents (BPA)
derived factor products have ascended as the major hemo­ such as recombinant activated factor VII (rFVIIa) or activated
philia treatment and have remained the standard of care prothrombin complex concentrates (aPCC), which have
Prakash Singh Shekhawat
Treating hemophilia without factor? | 219
sim­i­lar effi­cacy and side effect pro­files in ret­ro­spec­tive ana­ly­ses. func­tion of FVIII by bring­ing FIXa and FX into close enough prox­
Although gen­er­ally effec­tive, BPA can­not be mon­i­tored by stan­ im­ity to facil­i­tate FX acti­va­tion.
dard lab assays and have a reported fail­ure rate rang­ing from 7% First approved for bleed pro­phy­laxis in HA with inhib­it­ors in
to 11.6% and throm­bo­sis rates between 4% and 6.5%.8 As such, the US by the US Food and Drug Administration in 2018, emici­
non­fac­tor alter­na­tives to cir­cum­vent these com­pli­ca­tions have zumab has been appro­pri­ately rec­og­nized as sig­nal­ing a new
been of inter­est. These ther­a­pies are part of 2 broad categories: era of HA treat­ment. In all­phase 3 tri­als, emicizumab pro­phy­laxis
fac­tor mimet­ics and rebalancing ther­a­pies. This review discusses has led to dras­tic reduc­tions in annu­al­ized bleed­ing rates (ABRs),
the indi­vid­ual drugs either cur­rently in use, in trial, or in pre­clin­ with median ABRs of 2.6 for all­dos­ ing reg­i­
mens in patients
i­cal inves­ti­ga­tion. A dis­cus­sion of gene ther­apy, desmopressin, through the HAVEN 1 through 4 tri­als and with over 80% of par­
and antifibrinolytics is omit­ted in favor of a focus on the basic tic­i­pants expe­ri­enc­ing no bleeds after week 24 of ther­apy.9,10
sci­ence, ratio­nale, and con­sid­er­ations of these emerg­ing non­ Considering data show­ing diminishing FVIII expres­sion months
fac­tor ther­a­pies. to years after adeno-asso­ci­ated virus gene ther­apy,11 emicizumab
or a sim­i­lar mimetic ther­apy may well be the dom­i­nant par­a­digm

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/219/1851769/219ellsworth.pdf by guest on 13 December 2021


in HA treat­ment for some time. In clin­i­cal prac­tice it is being suc­
cess­fully used in infants and other pre­vi­ously untreated patients,
CLINICAL CASES
who now grow up with­out bleed­ing or fac­tor expo­sure. Given
Case 1: A 10-year-old boy with severe HB devel­oped an inhib­i­tor the low inci­dence of anti­drug antibodies, the ease of admin­is­
at age 4, suc­cess­fully com­plet­ing immune tol­er­ance induc­tion tra­tion with min­i­mal instruc­tion, and the extremely long half-life,
at that time. He expe­ri­enced bleed­ing while on pro­phy­lac­tic patients may expe­ri­ence a de facto cure with reg­u­lar admin­is­
recom­bi­nant FIX infu­sions years later and was found to have a tra­tion.
recur­rence of his inhib­i­tor. He had a cen­tral line inserted for BPA However, much remains unanswered in emicizumab use, and
use while restarting immu­no­sup­pres­sion. He still has evi­dence of opti­mal man­age­ment requires an expe­ri­enced hema­tol­o­gist.
an inhib­i­tor, and his par­ents are ask­ing about alter­na­tive ther­a­ Standard coag­u­la­tion tests and thus one-stage, clot-based FVIII
pies, espe­cially those that could allow him to live with­out cen­tral activ­ity assays (FVIII OSCA) are ren­dered unre­li­able due to the
venous access and such fre­quent infu­sion cen­ter vis­its. inde­pen­dence of emicizumab from throm­bin gen­er­a­tion in ini­ti­
Case 2: A 26-year-old man has severe HA and has not been on fac­ at­ing its activ­ity.
tor pro­phy­laxis since age 12. He is afraid of bleed­ing and avoids Thromboses and throm­botic microangiopathy were observed
var­i­ous activ­i­ties out of cau­tion but still expe­ri­ences spon­ta­ne­ous in early tri­als, and all­were asso­ci­ated with the con­com­i­tant use
bleed­ing, pre­dom­i­nantly in his ankles. He never learned to infuse of aPCC.10 The mech­a­nism of this poten­tially dev­as­tat­ing adverse
fac­tor and states that he is afraid of needles. Additionally, he has effect is still unknown, though elu­ci­dat­ing this may yield insights
no insur­ance cov­er­age. He is ask­ing if there are now any options help­ful for cli­ni­cians treating with emicizumab in gen­eral.
other than infus­ing intra­ve­nous fac­tor to treat his hemo­philia. Other anti­body fac­tor VIIIa mimet­ics with the same mech­
a­nism of action, bind­ing fac­tors IXa and X in close prox­im­ity,
lead­ing to throm­bin-inde­pen­dent FXa acti­va­tion, are in devel­
op­ment. Mim8 (Novo Nordisk, Bagsvaerd, Denmark) is a next-
Hemostatic tar­gets of non­fac­tor ther­a­pies
gen­ er­
at­ion bispecific anti­ body to FIXa and X that has been
A review of the clot­ting cas­cade and its role in the hemo­philia
shown in vivo and in vitro to potently pro­mote throm­bin gen­er­a­
phe­no­type is nec­es­sary to a dis­cus­sion of mimetic and rebalanc­
tion in the absence of FVIII.12 It is cur­rently being inves­ti­gated as
ing ther­a­pies.
a next-gen­er­a­tion FVIII mimetic in a phase 1/2 trial (clinicaltrials​
Hemostasis is ini­ti­ated by the extrin­sic path­way of the coag­
­.gov). Another next-gen­er­at­ ion bispecific FVIII mimetic in devel­
u­la­tion cas­cade and then ampli­fied by the con­tact, or intrin­sic,
op­ment is BS-027125 (Bioverativ, Waltham, MA), cur­rently in pre­
path­way. These inter­ac­tions gen­er­ate throm­bin, which then
clin­i­cal eval­u­a­tion.13,14
cleaves to fibrin­o­gen to form a sta­ble fibrin clot (Figure 1).
FVIII is a cofac­tor rather than a ser­ine pro­te­ase, mak­ing it
Given the pri­macy of the extrin­sic, or ini­ti­a­tion, path­way in
ame­na­ble to ther­apy replacing it with a non­fac­tor com­pound,
phys­i­o­logic hemo­sta­sis, why should a defi­ciency of FVIII or FIX
leav­ing HB treat­ment want­ing a sim­i­lar par­a­digm change. Fortu­
lead to such severe bleed­ing? Because once fibrin depo­si­tion
nately, inge­nious manip­u­la­tion of the clot­ting cas­cade prom­ises
begins and throm­bin is formed, throm­bin also acts “upstream”
a sim­i­lar new era for HB. Approaches with roots in obser­va­tional
to acti­vate addi­tional FVIII, FXI, and FV, lead­ing to more throm­bin
sci­ence are driv­ ing emerg­ ing hemo­ philia treat­ments known
gen­er­a­tion via the intrin­sic path­way (also called the ampli­fi­ca­
as rebalancing ther­a­pies. These ther­a­pies seek to “rebalance”
tion path­way) in what is referred to as the throm­bin burst. With­
coag­ u­la­
tion to a more nor­ mal state by alter­ ing the inher­
ent
out FVIII or FIX, this phe­nom­e­non does not occur, and throm­bin
phys­i­o­logic mod­u­la­tion of coag­u­la­tion (Table 1).
gen­er­at­ ion remains too mea­ger to form a sta­ble fibrin clot. The
objec­tive of either a fac­tor mimetic or a rebalancing ther­apy is
to restore throm­bin gen­er­a­tion in patients with hemo­philia, thus Rebalancing ther­a­pies as a treat­ment for hemo­philia
achiev­ing clin­i­cal hemo­sta­sis with­out throm­botic com­pli­ca­tions. siRNA ther­a­peu­tics
With the advent of genome sequenc­ing, sci­en­tists iden­ti­fied
Factor mimetic ther­apy patients with severe hemo­philia who coinherited var­i­ous pro­
Emicizumab (Genentech, San Francisco, CA) is a human­ ized, thrombotic gene muta­tions and displayed a milder phe­no­type.15
bispecific, mono­clo­nal immu­no­glob­u­lin G4 anti­body (mAb) that A pre­pon­der­ance of evi­dence exists for coinheritance of FVIII
binds to acti­vated FIX (FIXa) and FX, thereby performing the and pro­throm­bin muta­tions, so it is per­haps fit­ting that target­

220  |  Hematology 2021  |  ASH Education Program


Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/219/1851769/219ellsworth.pdf by guest on 13 December 2021
Figure 1.  Coagulation cascade. A simplified representation of the “coagulation cascade”. Note the role that the tenase complexes
play in thrombin generation. The intrinsic tenase complex consists of factors VIIIa, IXa, and X on a phospholipid surface with phos­
phatidylserine exposure, usually a platelet, and facilitates the activation of factor X. The extrinsic tenase complex consists of factor
VIIa, tissue factor, and factor X, likewise leading to the activation of factor X. Note the many feedback mechanisms of activation that
thrombin performs. Although the generation and exposure of TF at the site of vascular endothelial is the primary initiator of coagu­
lation via the extrinsic pathway, the intrinsic tenase pathway is important because active TF has only limited availability in vivo and
TFPI’s constitutive activity inhibits the extrinsic tenase complex from generating adequate thrombin for a stable clot (see reference 19
for a more detailed treatment of this topic). In a PTT test, a test on which clot-based factor assays are built, phospholipid and calcium
are added to a sample anticoagulated with sodium citrate (a calcium chelator that inhibits the Ca++ dependent steps as noted in the
figure). Thrombin is added to the assay and further generated by the thrombin burst (see text). The activation of factor VIII or IX is
the rate-limiting step in the assay. Factors are labeled by their traditional roman numeral. TF, tissue factor; TFPI, tissue factor pathway
inhibitor; EPCR, endothelial protein C receptor; APC, activated protein C.

ing the func­tion of throm­bin would be an early con­tender for a lead­ing to increased total throm­bin gen­er­ated with a hemo­static
rebalancing ther­apy. chal­lenge.16,17
Antithrombin (AT) is an endog­ e­
nous pro­
tein that nat­ u­rally In early-phase clin­i­cal tri­als to date, doses of fitusiran were
reg­u­
lates the func­ tion of active throm­
bin. Fitusiran (Alnylam, targeted to lower AT lev­els to 20%, which nor­mal­izes throm­bin
Cambridge, MA) is a small-mol­e­cule RNA inter­fer­ence ther­a­peu­ gen­er­a­tion and reduces bleed­ing. However, tri­als were briefly
tic that acts by bind­ing and degrading the mRNA encoding AT, put on hold in 2017 after a patient died after devel­op­ing a dural
Prakash Singh Shekhawat
Treating hemo­philia with­out fac­tor? | 221
Table 1. Summary of non-factor therapies

Name of Drug Mechanism ofAc on CurrentStatus Route ofAdministra on DosingSchedule

Factor VIIIMimecs

Emicizumab1 Humanized monoclonal IgG4 anbody with specificity to FIXa and FX FDA approved in US, EMA approved in Europe Subcutaneous injecon Generally weekly or every other week, though approved for intervals
up to every 4 weeks

Mim82 Monoclonal anbody specific to FIXa and FX Phase 1/2 trial Subcutaneous injecon Weekly and monthly injecons are being evaluated in the phase 2
poron of the study.3

BS-0271254 Monoclonal anbody specific to FIXa and FX Preclinical evaluaon TBD TBD

siRNA Therapies

Fitusiran3,5 siRNA against AT3 mRNA, leading to decreased AT protein translaon Phase 2 and 3 trials ongoing Subcutaneous injecon Monthly

siRNA against Protein S mRNA, leading to decreased Protein S translaon. Preclinical/animal models TBD TBD

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/219/1851769/219ellsworth.pdf by guest on 13 December 2021


Protein S siRNA (unnamed)7

TFPI Inhibitors

Concizumab8 Humanized monoclonal IgG4 against TFPI, Kunitz domain 2 Phase 2 and 3 trials ongoing. Phase 3 trials in paents without Subcutaneous injecon Daily
inhibitors are sll recruing.

Befovacimab(BAY1093884)8 Humanized monoclonal IgG4 against TFPI, Kunitz domains 1 and 2 Phase 2 trial terminated due to 2 paents with arterial thrombi Subcutaneous injecon Weekly

Marstacimab(PF-06741086)9 Monoclonal Ab against TFPI Phase 1 – 3 trials are accruing Subcutaneous injecon Weekly

MG11133,8 Monoclonal Ab against TFPI Phase 1 trials in healthy volunteers. Preclinical, non-human Subcutaneous injecon Weekly
primate models showed decreased bleeding.

BAX49910 Aptamer disrupng TFPI binding to extrinsic tenase complex. Phase 1/2 studies terminated due to bleeding in subjects. Intravenous N/A

Other Serine Protease Targets

Serpin PC3,11 Alpha-1-antrypsin-like serine protease which inhibits acvated protein C Phase 1/2 trial evaluang in hemophilia A and B paents Subcutaneous or intravenous TBD

HAPC157312 Monoclonal anbody to acvated protein C Preclinical, non-human primate models have shown decreased TBD TBD
bleeding.

Protein Z related protease Inacve, mutant protein Z, prevenng protein Z (a cofactor) binding to Preclinical, in vitro thrombin generaon is enhanced. TBD TBD
inhibitor (unnamed)13 protein Z-dependent protease inhibitor, prevenng FXa inhibion by the
endogenous complex.
Protease Nexin-1 inhibitor Endogenous glycoprotein that is secreted by acvated platelets and inhibits Preclinical murine knockout model, no specific drug in TBD TBD
(unnamed)14 FXIa and thrombin, among other coagulaon factors. development.

This table includes currently approved therapeucs (emicizumab), those in advanced phase trials (fitusiran, concizumab, marstacimab), and others which are either in preclinical invesgaon or suspended invesgaon as far as the authors are currently aware. References for informaon
provided in text. Paent convenience and acceptability are important consideraons and noted in this table. An-TFPI mAb drugs are administered by daily subcutaneous injecons, making them less convenient than siRNA promises to be. Ulmately though, physician opinion, paent
symptoms and personal preferences and values will decide appropriate therapy.
FVIII - Factor VIII, FIXa - acvated factor IX, FX - Factor X, siRNA - small interfering RNA, mRNA - messenger RNA, TFPI - ssue factor pathway inhibitor

1. Callaghan MU, Negrier C, Paz-Priel I, et al. Long-term outcomes with emicizumab prophylaxis for hemophilia A with or without FVIII inhibitors from the HAVEN 1-4 studies. Blood. 2021;137(16):2231-2242. doi:10.1182/blood.2020009217
2. Kjellev SL, Østergaard H, Greisen PJ, et al. Mim8 - a Next-Generaon FVIII Mimec Bi-Specific Anbody - Potently Restores the Hemostac Capacity in Hemophilia a SeŸngs in Vitro and In Vivo. Blood. 2019;134(Supplement_1):96-96. doi:10.1182/blood-2019-122817
3. clinicaltrials.gov
4. Aleman MM, Jindal S, Leksa N, Peters R, Salas J. Phospholipid-Independent Acvity of Fviiia Mimec Bispecific Anbodies in Plasma. Blood. 2018;132(Supplement 1):2461-2461. doi:10.1182/blood-2018-99-119226
5. Machin N, Ragni MV. An invesgaonal RNAi therapeuc targeng anthrombin for the treatment of hemophilia A and B. J Blood Med. 2018;9:135-140. doi:10.2147/JBM.S159297
6. Pasi KJ, Lissitchkov T, Mamonov V, et al. Targeng of anthrombin in hemophilia A or B with invesgaonal siRNA therapeuc fitusiran - results of the phase 1 inhibitor cohort. J Thromb Haemost. February 2021. doi:10.1111/jth.15270
7. Prince R, Bologna L, ManeŸ M, et al. Targeng ancoagulant protein S to improve hemostasis in hemophilia. Blood. 2018;131(12):1360-1371. doi:10.1182/blood-2017-09-800326
8. Mahlangu JN. Progress in the Development of An-ssue Factor Pathway Inhibitors for Haemophilia Management. Front Med (Lausanne). 2021;8:670526. doi:10.3389/fmed.2021.670526
9. Patel-He¥ S, Marn EJ, Mohammed BM, et al. Marstacimab, a ssue factor pathway inhibitor neutralizing anbody, improves coagulaon parameters of ex vivo dosed haemophilic blood and plasmas. Haemophilia. 2019;25(5):797-806. doi:10.1111/hae.13820
10. Spadarella G, Di Minno A, Milan G, et al. Paradigm shi¦ for the treatment of hereditary haemophilia: Towards precision medicine. Blood Rev. 2020;39:100618. doi:10.1016/j.blre.2019.100618
11. Weyand AC, Pipe SW. New therapies for hemophilia. Blood. 2019;133(5):389-398. doi:10.1182/blood-2018-08-872291
12. Zhao X-Y, Wilmen A, Wang D, et al. Targeted inhibion of acvated protein C by a non-acve-site inhibitory anbody to treat hemophilia. Nat Commun. 2020;11(1):2992. doi:10.1038/s41467-020-16720-9
13. Aymonnier K, Kawecki C, Arocas V, Boula¦ali Y, Bouton MC. Serpins, new therapeuc targets for hemophilia. Thromb Haemost. 2021;121(3):261-269. doi:10.1055/s-0040-1716751

sinus throm­bo­sis fol­low­ing high-dose rFVIII admin­is­tra­tion to Another pre­clin­i­cal siRNA ther­a­peu­tic shares fitusiran’s mech­
treat a bleed dur­ing the open-label exten­sion of the trial.17 The a­nism but silences pro­tein S expres­sion rather than AT. It is in
hold was lifted after pro­to­col amend­ments were made. The drug pre­clin­i­cal inves­ti­ga­tion for even­tual use in hemo­philia.19
had no throm­botic com­pli­ca­tions in a phase 1 cohort of patients
with inhib­i­tors,18 and results of the phase 2 trial have not been Tissue fac­tor path­way inhib­i­tors
published. Subsequently, fol­low­ing fur­ther non­fa­tal throm­botic Another way to exploit inher­ent mech­a­nisms to pro­mote hemo­
events, clin­i­cal tri­als were again held, and doses were reduced in sta­sis is by blocking tis­sue fac­tor path­way inhib­i­tor (TFPI), an
Octo­ber 2020. Revised dos­ing and AT tar­gets have been adopted endog­e­nous ser­ine pro­te­ase inhib­i­tor that pre­vents the acti­va­
for ongo­ing phase 3 par­tic­i­pants (clinicaltrials​­.gov). tion of FX by the TF-FVIIa com­plex, thus lim­it­ing the degree of
In tri­als, fitusiran is now admin­is­tered as a sub­cu­ta­ne­ous injec­ throm­bin gen­er­a­tion via the con­tact path­way. By disrupting TFPI
tion every other month. Since the decrease in AT affects the com­ bind­ing to this extrin­sic tenase com­plex, Xa and throm­bin gen­er­
mon path­way, this can treat either HA or HB, poten­tially with or a­tion are increased. This approach has been suc­cess­ful via mono­
with­out inhib­i­tors. clo­nal antibodies to var­i­ous domains of the TFPI mol­ec ­ ule.20

222  |  Hematology 2021  |  ASH Education Program


Concizumab (Novo Nordisk) is a human­ized immu­no­glob­u­lin some mAb according to many met­rics for rare dis­eases, equity in
G4 anti-TFPI anti­body to the sec­ond Kunitz (K2) domain of TFPI. access to treat­ment with these expen­sive med­i­ca­tions must be
Building on obser­va­tional data in humans, ani­mal mod­els dem­ ensured for these ben­e­fits to be real­ized.30
on­strated restored throm­bin gen­er­a­tion and decreased bleed­ Some authors have found that mAb costs for malig­nant and
ing despite deficiencies in FVIII or FIX.21 Phase 1 and 2 tri­als in non­ma­lig­nant hema­to­logic dis­or­ders skew higher than those
hemo­philia patients with­out inhib­i­tors dem­on­strated reduced marketed for use in other dis­or­ders.31 However, emicizumab has
ABRs and no throm­bo­em­bolic events. Phase 3 tri­als in HA and HB actu­ally been shown to be highly cost-effec­tive in sev­eral real-
were tem­po­rar­ily suspended due to non­fa­tal throm­botic events world ana­ly­ses, owing to its superb effi­cacy and the already high
in early 2020 but have since resumed. In addi­tion, anti­drug anti­ cost of hemo­philia care with fac­tor infu­sions and inhib­i­tor devel­
bodies have been observed in tri­als for concizumab.22,23 op­ment.32,33 Current data describe an existing high bur­den in
Befovacimab (BAY1093884, Bayer) is spe­cific to both K2 and treating those patients who fail fac­tor ther­apy or can­not secure
K1 domains, blocking TFPI bind­ing to both FVIIa and FX in the access to effec­tive fac­tor ther­apy.34,35 In this con­text, costs of
extrin­sic tenase com­plex and enhanc­ing throm­bin gen­er­a­tion new ther­a­pies may be off­set by a reduced bur­den of the com­

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/219/1851769/219ellsworth.pdf by guest on 13 December 2021


despite deficiencies in the con­tact path­way. In an early study, it pli­ca­tions of stan­dard ther­apy. However, con­cerns remain about
decreased bleed­ing epi­sodes, but the trial was ter­mi­nated when the long-term effects of these ther­a­pies, which may carry hid­den
2 patients devel­oped cere­bral arte­rial thrombi, and one devel­ eco­nomic costs and dis­ad­van­tages.
oped a cere­bral venous throm­bus.22,23
PF-06741086 (Marstacimab, Pfizer) has been shown to nor­ Concerns and pit­falls
mal­ize coag­u­la­tion in hemo­philia patient plas­mas ex vivo and is For fac­tor mimetic and rebalancing ther­a­pies that have reached
being eval­u­ated in HA and HB patients (clinicaltrials​­.gov). There a suf­fi­ciently advanced stage in clin­i­cal tri­als, a reduc­tion in ABRs
have been no throm­botic com­pli­ca­tions to date, with reduc­ has indeed been noted. Note that this effi­cacy has been shown
tion of ABRs to zero in most par­ tic­

pants.22,24 MG1113 (Green thus far in pro­phy­laxis only, includ­ing in emicizumab. This is to
Cross Corporation) is another TFPI mAb like­wise being tested be expected con­sid­er­ing the mech­a­nism of action of these ther­
in healthy vol­un­teers (clinicaltrials​­.gov), with non­hu­man pri­mate a­peu­tics but is impor­tant in eval­u­at­ing the lim­i­ta­tions and poten­
mod­els hav­ing dem­on­strated in vivo reduc­tion of blood loss.22 tial appli­ca­tion of these new and emerg­ing drugs.
An aptamer derived from recom­bi­nant human TFPI (BAX499, As noted in the dis­cus­sion of these agents, every non­fac­tor
Takeda) was also devel­oped and found to effi­ciently inhibit TFPI ther­apy that has reached clin­i­cal trial has had throm­botic side
in vitro and in vivo, with dose-depen­dent increases in throm­ effects with the excep­tion of marstacizumab.23,29,36 With emici­
bin gen­er­a­tion and decreased bleed­ing in ani­mal mod­els and zumab, all­throm­ bo­ ses and throm­ botic microangiopathy epi­
in early-phase human tri­als.15,25 Development is on hold due to sodes were asso­ci­ated with aPCC use, and safety oth­er­wise is
bleed­ing in sub­jects.15 becom­ing well established.10 But safety in the set­ting of sur­ger­
ies and trauma remains an unre­solved prob­lem. Although the
Other ser­ine pro­te­ase tar­gets cur­rent aim of non­fac­tor ther­apy is pro­phy­laxis, the con­com­i­tant
SerpinPC is an alpha-1-antitrypsin-like ser­ine pro­te­ase mod­i­fied man­age­ment of perioperative and trauma-related bleed­ing is of
to inhibit activated protein C (APC).26 By pre­vent­ing the action grow­ing inter­est,35-37 with con­sen­sus that fac­tor con­cen­trate use
of APC, which inhib­its FV acti­va­tion, throm­bin gen­er­a­tion is like­ is still nec­es­sary in these cases. Though stan­dard­ized, opti­mized
wise restored despite fac­tor deficiencies in the con­tact sys­tem approaches do not yet exist, cur­rent con­sen­sus guide­lines pro­
via enhanced com­mon path­way activ­ity (Figure 1).27 This is cur­ vide detailed guid­ance based on real-world use thus far.35,38,39
rently in a phase 1/2 trial, with approx­i­ma­tely 50 par­tic­i­pants Additionally, expe­ri­ence in com­pet­i­tive ath­letes is lacking,
hav­ing both HA and HB (clinicaltrials​­.gov). Recently, a mono­clo­ though expert opin­ion gen­er­ally con­sid­ers monotherapy insuf­fi­
nal anti­body to acti­vated pro­tein C (HAPC1573) has been shown cient if break­through bleed­ing is observed.23,37
in non­hu­man pri­ma­tes to pre­vent bleed­ing.28 Because mimetic and rebalancing ther­a­pies alter hemo­sta­sis
Protein Z (PZ)-related pro­te­ase inhib­i­tor is an inac­tive mutant either inde­pen­dently or by directly mod­i­fy­ing reg­u­la­tory mech­a­
PZ, cofac­tor to PZ-depen­dent pro­te­ase inhib­i­tor. Endogenously, nisms of coag­u­la­tion, patient selec­tion will also need to take into
the PZ-depen­dent pro­te­ase inhib­i­tor com­plex inhib­its FXa. By account throm­botic risk fac­tors such as obe­sity, existing car­dio­
pre­vent­ing this com­plex for­ma­tion through var­i­ous meth­ods, vas­cu­lar dis­ease, dia­be­tes, and other con­di­tions.
pre­clin­i­cal data have shown increased throm­bin gen­er­a­tion in Aside from effi­cacy and safety in gen­eral, mean­ing­ful trial end
vitro.29 points are essen­tial for emerg­ing ther­a­pies. For exam­ple, some
Finally, pro­ te­
ase nexin-1 has been pro­ posed as a tar­ get. extended half-life fac­tor prod­ucts, despite show­ing ade­quate
Endogenously, this gly­co­pro­tein is expressed by acti­vated plate­ trough lev­els in clin­i­cal trial, have been found in real-world use
lets and inhib­its FXIa, throm­bin, and other fac­tors. Hemophilic to have infe­rior bleed­ing response.38 The approval and wide clin­
mice showed decreased bleed­ing in pro­te­ase nexin-1 knock­out i­cal adop­tion of emicizumab reflect its proven effi­cacy in reduc­
mod­els, mak­ing this another prom­is­ing tar­get.29 ing ABRs in patients with and with­out inhib­i­tors.39 As mon­i­tor­ing
will not be as straight­for­ward as with fac­tor replace­ment, ABR
The eco­nom­ics of non­fac­tor ther­apy reduc­tion must remain the end point of choice in non­fac­tor ther­
Although poten­tially par­a­digm chang­ing for patients, non­fac­tor apy tri­als.15,23,40
ther­a­pies may come at a par­a­digm-chang­ing price. Cost ana­ly­ses The mon­i­tor­ing of non­fac­tor ther­a­pies is com­pli­cated and
of new mono­clo­nal antibodies for var­i­ous indi­ca­tions are com­mon impos­si­ble with stan­dard screen­ing tests and assays, a promi­
in the lit­er­a­ture. Although ana­ly­ses sug­gest cost-effec­tive­ness for nent sub­ject of con­cern.40 This is because these drugs exert their

Prakash Singh Shekhawat


Treating hemo­philia with­out fac­tor? | 223
effect inde­pen­dently of acti­va­tion by throm­bin itself, the reac­ of hemo­philia—a chap­ter of pre­ci­sion treat­ment, in which many
tion noted above being the rate-lim­it­ing step in such clot-based roads lead to a life unen­cum­bered by the threat of bleed­ing.
assays,39 or by directly mod­i­fy­ing throm­bin gen­er­a­tion. Though
OSCA and chro­mo­genic fac­tor assays are widely avail­­able, val­i­ Conflict-of-inter­est dis­clo­sure
dated mon­i­tor­ing of non­fac­tor agents is forth­com­ing. Patrick Ellsworth: Is an NHF-Takeda Clinical Research Fellowship
Patients started on emicizumab while still hav­ ing active award recipient.
inhib­i­tors are another pop­u­la­tion who will ben­e­fit from fur­ther Alice Ma: research funding and hon­or­ aria: Takeda.
research to opti­mize inhib­i­tor erad­i­ca­tion, with approaches
likely to dif­fer between pedi­at­ric and adult patients.41,42 Alloan­ Off-label drug use
tibodies to the drugs them­selves and inhib­it­ or recur­rence while Patrick Ellsworth: no off-label drug use has been discussed.
on mimetic ther­apy are emerg­ing issues as well.29,43,44 Alice Ma: no off-label drug use has been discussed.
Other con­cerns with the advent of fac­tor-mimetic ther­ap ­ ies,
espe­cially in long-term use, are that they do not per­form some Correspondence

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/219/1851769/219ellsworth.pdf by guest on 13 December 2021


impor­tant func­tions of the fac­tor mol­e­cules in maintaining vas­ Alice Ma, University of North Carolina at Chapel Hill, Houpt Bldg,
cu­lar integ­rity and interacting with the cel­lu­lar com­po­nents of CB# 7305, 170 Manning Dr, 3rd Fl, Chapel Hill, NC 27599-7305;
blood.39 Additionally, TFPI knock­ out mice develop pre­ ma­ture e-mail: alice_ma@med​­.unc​­.edu.
ath­ero­scle­ro­sis, suggesting that mod­u­lat­ing the hemo­static sys­
tem could have unin­tended con­se­quences out­side of the acute References
hemo­static effect.45 Indeed, the act of rebalancing a sys­tem that 1. Franchini M, Mannucci PM. The his­ tory of hemo­ philia. Semin Thromb
exists in such a com­plex bal­ance is bound to be a dif­fi­cult prop­ Hemost. 2014;40(5):571-576.
o­si­tion, and long-term fol­low-up data will be crit­i­cal in all­emerg­ 2. Mannucci PM. Hemophilia ther­apy: the future has begun. Haematologica.
2020;105(3):545-553.
ing ther­a­pies. 3. Aledort L, Mannucci PM, Schramm W, Tarantino M. Factor VIII replace­ment
Factor mimetic and rebalancing ther­ a­
pies will con­ tinue to is still the stan­dard of care in haemophilia A. Blood Transfus. 2019;17(6):479-
coevolve with fac­tor ther­apy. These ther­ap ­ ies and the accom­ 486.
pa­ny­ing com­plex­ity involved in their use will con­tinue to require 4. Witmer C, Young G. Factor VIII inhib­i­tors in hemo­philia A: ratio­nale and lat­est
evi­dence. Ther Adv Hematol. 2013;4(1):59-72.
com­pre­hen­sive, expert care. The myr­iad offer­ings will match the
5. Dolan G, Benson G, Duffy A, et al. Haemophilia B: where are we now and
com­plex needs of our patients and even­tu­ally lead to supe­rior what does the future hold? Blood Rev. 2018;32(1):52-60.
care over­all. While expert opin­ion still regards fac­tor replace­ment 6. Santoro C, Quintavalle G, Castaman G, et  al. Inhibitors in hemo­philia B.
as the cur­rent stan­dard of care for hemo­philia,3,37 this “stan­dard” is Semin Thromb Hemost. 2018;44(6):578-589.
con­tinu­ing to change. Future ther­apy, pro­tean as it may be, has a 7. Antun A, Monahan PE, Manco-Johnson MJ, et al. Inhibitor recur­rence after
immune tol­er­ance induc­tion: a mul­ti­cen­ter ret­ro­spec­tive cohort study.
com­mon goal: a stan­dard of result that our patients do not bleed. J Thromb Haemost. 2015;13(11):1980-1988.
8. Ma AD, Kessler CM, Al-Mondhiry HA, Gut RZ, Cooper DL. US expe­ri­ence
Final thoughts with recom­ bi­
nant fac­tor VIIa for sur­ gery and other inva­ sive pro­
ce­
dures
There will be a need for fac­tor replace­ment ther­apy in the treat­ in acquired haemophilia: anal­ y­
sis from the Hemostasis and Thrombosis
Research Society Registry. Haemophilia. 2016;22(1):e18-e24.
ment of hemo­philia for the fore­see­able future, but non­fac­tor ther­
9. Yada K, Nogami K. Spotlight on emicizumab in the man­ age­ment of
a­pies ful­fill a great need and rep­re­sent a new phase of hemo­philia hemo­philia A: patient selec­tion and spe­cial con­sid­er­ations. J Blood Med.
care. Some of those needs not cur­rently ame­na­ble to non­fac­tor 2019;10(2 July):171-181.
ther­ap­ ies include the man­age­ment of sur­ger­ies and trauma. 10. Callaghan MU, Negrier C, Paz-Priel I, et al. Long-term out­comes with emi­
Are non­fac­tor ther­a­pies the end of fac­tor ther­apy? Perhaps cizumab pro­phy­laxis for hemo­philia A with or with­out FVIII inhib­i­tors from
the HAVEN 1-4 stud­ies. Blood. 2021;137(16):2231-2242.
the more impor­tant ques­tion is whether they should be the end 11. Pasi KJ, Rangarajan S, Mitchell N, et al. Multiyear fol­low-up of AAV5-hFVIII-
of fac­tor ther­apy. Preliminary data seem to sug­gest that alter­ing SQ gene ther­apy for hemo­philia A. N Engl J Med. 2020;382(1):29-40.
the hemo­static bal­ance is a fraught prop­o­si­tion, and ulti­mately, 12. Kjellev SL, Østergaard H, Greisen PJ, et al. Mim8—a next-gen­er­a­tion FVIII
the best option is that which yields the best result for a given mimetic bi-spe­cific anti­body—potently restores the hemo­static capac­ity
in hemo­philia a set­tings in vitro and in vivo. Blood. 2019;134(suppl 1):96.
patient. Some patients will not want new options to replace cur­
13. Leksa NC, Aleman MM, Goodman AG, Rabinovich D, Peters R, Salas J.
rent treat­ments. Maybe they will be reassured by the num­ber of Intrinsic dif­ fer­
ences between FVIIIa mimetic bispecific antibodies and
choices. Clinicians should find reas­sur­ance in this as well. FVIII pre­vent assign­ment of FVIII-equiv­a­lence. J Thromb Haemost. 2019;​
Returning to our intro­duc­tory cases, are they can­di­dates for 17(7):1044-1052.
non­fac­tor ther­a­pies? Certainly, our young man in Case 1 would 14. Aleman MM, Jindal S, Leksa N, Peters R, Salas J. Phospholipid-inde­pen­dent
activ­ity of Fviiia mimetic bispecific antibodies in plasma. Blood. 2018;132
clearly ben­e­fit from ther­apy to free him from fre­quent BPA use in (­suppl 1):2461.
a cen­tral line and the risks of ongo­ing immu­no­sup­pres­sion. Our 15. Spadarella G, Di Minno A, Milan G, et  al. Paradigm shift for the treat­
gen­tle­man from Case 2 may have more to think about. Someone ment of hered­i­tary haemophilia: towards pre­ci­sion med­i­cine. Blood Rev.
who has never had fac­tor ther­apy opti­mized lacks a com­pel­ling 2020;39(Jan­u­ary):100618.
16. Pasi KJ, Rangarajan S, Georgiev P, et al. Targeting of anti­throm­bin in hemo­
rea­son to con­sider newer ther­apy, though the deci­sion would
philia A or B with RNAi ther­apy. N Engl J Med. 2017;377(9):819-828.
ulti­mately rest on his val­ues and per­cep­tion of his qual­ity of life. As 17. Machin N, Ragni MV. An inves­ti­ga­tional RNAi ther­a­peu­tic targeting anti­
we have seen with emicizumab, even patients with­out inhib­i­tors throm­bin for the treat­ment of hemo­philia A and B. J Blood Med. 2018;9(22
and other dif­fi­cul­ties have benefited from non­fac­tor treat­ment. August):135-140.
Also, the sim­plic­ity of sub­cu­ta­ne­ous dos­ing in a patient who has 18. Pasi KJ, Lissitchkov T, Mamonov V, et al. Targeting of anti­throm­bin in hemo­
philia A or B with inves­ti­ga­tional siRNA ther­a­peu­tic fitusiran—results of the
not learned self-infu­sion is likely to improve bleed pro­phy­laxis. phase 1 inhib­i­tor cohort. J Thromb Haemost. 2021;19(6):1436-1446.
Even if these non­fac­tor tools are not the end of fac­tor ther­apy, 19. Prince R, Bologna L, Manetti M, et al. Targeting anti­co­ag­u­lant pro­tein S to
they do seem poised to inau­gu­rate a new chap­ter in the his­tory improve hemo­sta­sis in hemo­philia. Blood. 2018;131(12):1360-1371.

224  |  Hematology 2021  |  ASH Education Program


20. Chowdary P. Inhibition of tis­ sue fac­tor path­
way inhib­i­
tor (TFPI) as a 35. D’Angiolella LS, Cortesi PA, Rocino A, et  al. The socio­eco­nomic bur­den
treat­ment for haemophilia: ratio­nale with focus on concizumab. Drugs. of patients affected by hemo­philia with inhib­i­tors. Eur J Haematol. 2018;​
2018;78(9):881-890. 101(4):​435-456.
21. Morfini M, Marchesini E. The avail­abil­ity of new drugs for hemo­philia treat­ 36. Lillicrap D, Fijnvandraat K, Young G, Mancuso ME. Patients with hemo­philia
ment. Expert Rev Clin Pharmacol. 2020;13(7):721-738. A and inhib­i­tors: pre­ven­tion and evolv­ing treat­ment par­a­digms. Expert Rev
22. Mahlangu JN. Progress in the devel­op­ment of anti-tis­sue fac­tor path­way Hematol. 2020;13(4):313-321.
inhib­i­tors for haemophilia man­age­ment. Front Med (Lausanne). 2021;8(5 37. Srivastava A, Santagostino E, Dougall A, et al; World Federation of Hemo­
May):670526. philia Guidelines for the Management of Hemophilia pan­ el­ists and co-
23. Jiménez-Yuste V, Auerswald G, Benson G, et  al. Practical con­sid­er­ations authors. WFH Guidelines for the Management of Hemophilia, 3rd edi­tion.
for non­fac­tor-replace­ment ther­a­pies in the treat­ment of haemophilia with Haemophilia. 2020;26 (suppl 6):1-158.
inhib­i­tors. Haemophilia. 2021;27(3):340-350. 38. Malec LM, Croteau SE, Callaghan MU, Sidonio RF Jr. Spontaneous bleed­
24. Patel-Hett S, Martin EJ, Mohammed BM, et  al. Marstacimab, a tis­sue fac­ ing and poor bleed­ing response with extended half-life fac­tor IX prod­
tor path­way inhib­i­tor neu­tral­iz­ing anti­body, improves coag­u­la­tion param­ ucts: a sur­vey of select US haemophilia treat­ment cen­tres. Haemophilia.
e­ters of ex vivo dosed haemophilic blood and plas­ mas. Haemophilia. 2020;26(3):e128-e129.
2019;25(5):797-806. 39. Lenting PJ, Denis CV, Christophe OD. Emicizumab, a bispecific anti­body
25. Miesbach W, Eladly F. Current and future options of haemophilia A treat­ rec­og­niz­ing coag­u­la­tion fac­tors IX and X: how does it actu­ally com­pare to

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/219/1851769/219ellsworth.pdf by guest on 13 December 2021


ments. Expert Opin Biol Ther. 2021:1 April:1-8. fac­tor VIII? Blood. 2017;130(23):2463-2468.
26. Pelland-Marcotte MC, Carcao MD. Hemophilia in a chang­ ing treat­ ment 40. Lenting PJ. Laboratory mon­i­tor­ing of hemo­philia A treat­ments: new chal­
land­scape. Hematol Oncol Clin North Am. 2019;33(3):409-423. lenges. Blood Adv. 2020;4(9):2111-2118.
27. Weyand AC, Pipe SW. New ther­a­pies for hemo­philia. Blood. 2019;133(5):389- 41. Escuriola-Ettingshausen C, Auerswald G, Königs C, et  al. Optimizing the
398. man­age­ment of patients with haemophilia A and inhib­i­tors in the era of
28. Zhao XY, Wilmen A, Wang D, et al. Targeted inhi­bi­tion of acti­vated pro­tein emicizumab: rec­ om­ men­ da­tions from a Ger­ man expert panel. Haemo-
C by a non-active-site inhib­i­tory anti­body to treat hemo­philia. Nat Com- philia. 2021;27(3):e305-e313.
mun. 2020;11(1):2992. 42. Linari S, Castaman G. Concomitant use of rFVIIa and emicizumab in peo­ple
29. Aymonnier K, Kawecki C, Arocas V, Boulaftali Y, Bouton MC. Serpins, new with hemo­philia A with inhib­i­tors: cur­rent per­spec­tives and emerg­ing clin­
ther­a­peu­tic tar­gets for hemo­philia. Thromb Haemost. 2021;121(3):261-269. i­cal evi­dence. Ther Clin Risk Manag. 2020;16(May 22):461-469.
30. Park T, Griggs SK, Suh DC. Cost effec­tive­ness of mono­clo­nal anti­body ther­ 43. Harroche A, Sefiane T, Desvages M, et al. Non-inhib­i­tory antibodies induc­ing
apy for rare dis­eases: a sys­tem­atic review. BioDrugs. 2015;29(4):259-274. increased emicizumab clear­ance in a severe haemophilia A inhib­i­tor patient.
31. Hernandez I, Bott SW, Patel AS, et al. Pricing of mono­clo­nal anti­body ther­ Haematologica. 2021;106(8):2287-2290.
a­pies: higher if used for can­cer? Am J Manag Care. 2018;24(2):109-112. 44. Capdevila L, Borgel D, Lasne D, et al. Reappearance of inhib­i­tor in a toler­
32. Cortesi PA, Castaman G, Trifirò G, et  al. Cost-effec­tive­ness and bud­get ized patient with severe haemophilia A dur­ing FVIII-free emicizumab ther­
impact of emicizumab pro­phy­laxis in haemophilia A patients with inhib­i­ apy. Haemophilia. 2021;27(4):e581-e584.
tors. Thromb Haemost. 2020;120(2):216-228. 45. Xiao J, Jin K, Wang J, et al. Conditional knock­out of TFPI-1 in VSMCs of mice
33. Polack B, Trossaërt M, Cousin M, Baffert S, Pruvot A, Godard C. Cost- accel­er­ates ath­ero­scle­ro­sis by enhanc­ing AMOT/YAP path­way. Int J Car-
effec­tive­ness of emicizumab vs bypassing agents in the pre­ ven­
tion of diol. 2017;228(Feb­ru­ary):605-614.
bleed­ing epi­sodes in haemophilia A patients with anti-FVIII inhib­i­tors in
France. Haemophilia. 2021;27(1):e1-e11.
34. Buckner TW, Bocharova I, Hagan K, et al. Health care resource uti­li­za­tion
and cost bur­den of hemo­philia B in the United States. Blood Adv. 2021;5(7):​ © 2021 by The Amer­i­can Society of Hematology
1954-1962. DOI 10.1182/hema­tol­ogy.2021000253

Prakash Singh Shekhawat


Treating hemo­philia with­out fac­tor? | 225
HEMOPHILIA UPDATE: OUR CUP RUNNETH OVER

Hemophilia gene therapy: ushering in a new


treatment paradigm?

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/226/1851420/226george.pdf by guest on 13 December 2021


Lindsey A. George
Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA; and Division of Hematology and
Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children’s Hospital of Philadelphia, Philadelphia, PA

After 3 decades of clinical trials, repeated proof-of-concept success has now been demonstrated in hemophilia A and
B gene therapy. Current clinical hemophilia gene therapy efforts are largely focused on the use of systemically admin-
istered recombinant adeno-associated viral (rAAV) vectors for F8 or F9 gene addition. With multiple ongoing trials,
including licensing studies in hemophilia A and B, many are cautiously optimistic that the first AAV vectors will obtain reg-
ulatory approval within approximately 1 year. While supported optimism suggests that the goal of gene therapy to alter
the paradigm of hemophilia care may soon be realized, a number of outstanding questions have emerged from clinical
trial that are in need of answers to harness the full potential of gene therapy for hemophilia patients. This article reviews
the use of AAV vector gene addition approaches for hemophilia A and B, focusing specifically on information to review
in the process of obtaining informed consent for hemophilia patients prior to clinical trial enrollment or administering a
licensed AAV vector.

LEARNING OBJECTIVES
• Understand information important to review with hemophilia patients prior to consenting for a clinical trial
or approved AAV vector
• Differentiate information that is well understand from information that remains incompletely understood

occurring AAV, which is nonpathogenic in humans and rep-


CLINICAL CASE lication defective. rAAV vectors are produced in cell culture
A 62-year-old man with severe hemophilia A (HA) with a his- using either HEK293 mammalian cells or baclovirus/Sl9 insect
tory of an inhibitor, eradicated by immune tolerance induc- cell culture. The design and manufacturing of rAAV vectors
tion, on emicizumab prophylaxis is interested in enrolling was recently reviewed by Li and Samulski.1
in a HA gene therapy trial or receiving a licensed adeno- While hepatocytes are the natural site of factor IX (FIX)
associated viral (AAV) vector that will be available soon. synthesis, liver sinusoidal endothelial cells are the pre-
His comorbidities include 3 target joints, a prior 2-decade dominate site of factor VIII (FVIII) synthesis.2 Most HA rAAV
history of the hepatitis C virus (HCV) infection with stage 2 vectors contain a B-domain deleted FVIII, FVIII-SQ trans-
liver fibrosis, and HIV well controlled on antiretroviral med- gene that retains full procoagulant function and accom-
ications. He wants to know what is known and unknown modates AAV vector packaging constraints (4.7 kB).3 This
about hemophilia gene therapy. exact amino acid sequence has been used for recombinant
protein HA therapy for 2 decades (eg, Xyntha/ReFacto®,
Pfizer) without evidence of increased risk of inhibitor for-
Introduction: What are the current approaches mation. A single trial is using an alternative B-domain
for hemophilia gene therapy? deleted FVIII, FVIII-V3 that consists of the SQ linker with 6
Clinical approaches in hemophilia gene therapy nearly exclu- additional N-linked glycosylation sites to improve expres-
sively use systemically delivered recombinant AAV (rAAV) sion.4,5 All enrolling hemophilia B (HB) trials have adapted
vectors to target hepatocyte expression of an episom- the use of the FIX-Padua (FIX-R338L) mutation, which is a
ally maintained transgene expressed under a hepatocyte naturally occurring missense mutation with approximately
promoter (Table 1). The vector is engineered from naturally 8-fold greater specific activity relative to wild-type FIX.6-10

226 | Hematology 2021 | ASH Education Program


Table 1. Ongoing hemo­philia A and B gene ther­apy tri­als

Manufacturing Capsid
Sponsor Clinicaltrials​­.gov plat­form Transgene sero­type Dose (vg/kg) Phase Ref
HB
  UCL/St. Jude NCT00979238 Mammalian scFIX AAV8 2 × 1011 to 2 × 1012 1/2 16,45

 Pfizer/Spark NCT03681273 Mammalian ssFIX-R338L SPK100 5 × 1011 1/2, 3 9

NCT03307980
 uniQure NCT02396342 Sl9 (insect) ssFIX-R338L AAV5 2 × 1013 3 8

 Freeline NCT03369444 Mammalian scFIX-R338L AAVS3 7.5 × 10 11 10

9.5 × 1011
HA
 BioMarin NCT02576795 Sl9 (insect) ssFVIII-SQ AAV5 6 × 1013 1/2, 3 11,15

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/226/1851420/226george.pdf by guest on 13 December 2021


NCT03392974
NCT03370913
 Sangamo/Pfizer NCT03061201 Sl9 (insect) ssFVIII-SQ AAV6 3 × 1013 1/2, 3 18

 Spark NCT03003533 Mammalian ssFVIII-SQ LK03 5 × 10 to 2 × 10


11 12
1/2 14

NCT03432520
  UCL/St. Jude NCT03001830 Mammalian ssFVIII-V3 AAV8 6 × 1011 to 6 × 1012 1/2 4,5

 Bayer/Ultragenyx NCT03588299 Mammalian ssFVIII-SQ AAVhu37 5 × 10 to 2 × 10


12 13
1/2 58

 Takeda NCT03370172 Mammalian ssFVIII-SQ AAV8 1/2


sc, self-com­ple­men­tary; ss, sin­gle stranded; UCL, University College London.

What are the short- and long-term rAAV safety agents have been used con­cur­rently with glu­co­cor­ti­coids or in
con­sid­er­ations? iso­la­tion to main­tain transgene expres­sion.10,13,14 Additional study
In the 300-fold range of rAAV vec­ tor doses (2 × 1011 to 6 × 1013 is needed to under­stand which reg­i­men best main­tains transgene
vg/kg) thus far eval­u­ated in hemo­philia clin­i­cal tri­als, there have expres­sion. Further still, because some stud­ies have employed
been no major safety con­cerns. glu­co­cor­ti­coids while simul­ta­neously reporting that par­tic­i­pants
In the short term, a small num­ber of sub­jects across 3 tri­als had no cel­lu­lar immune response, addi­tional study is needed to
at rAAV vec­tor doses rang­ing from 2 × 1012 to 6 × 1013 vg/kg have dis­tin­guish a cel­lu­lar immune response vs other poten­tial eti­­ol­o­
expe­ri­enced acute vec­tor infu­sion reac­tions, includ­ing a sin­gle gies for ele­vated trans­am­i­nases.12,13,15
inci­dence of ana­phy­laxis and a cou­ple of inci­dences of either While safety in hemo­philia has been excel­lent, rAAV vec­tors
myal­gias, fever, and/or hypo­ten­sion of unclear eti­­ol­ogy that may devel­oped for other mono­genic dis­or­ders sug­gest there may
be con­sis­tent with an innate immune response to rAAV11-14; these be dose-lim­it­ing hepatic toxicities with sys­temic rAAV vec­tor
events have not clearly cor­re­lated with clin­i­cal out­comes post doses >1 × 1014 vg/kg. Specifically, Zolgensma (Novartis), an
vec­tor. rAAV9 vec­tor and the first licensed sys­temic rAAV vec­tor, deliv­
Because AAV effi­ciently tar­gets the liver, the bulk of safety con­ ered at a dose of 1.1 × 1014 vg/kg dem­on­strated not only remark­
sid­er­ations of sys­temic rAAV deliv­ery have focused on hep­a­to­tox­ able effi­cacy for spi­nal mus­cu­lar atro­phy but also evi­dence of
ic­ity. Within hemo­philia tri­als, a sin­gle study outlined multimonth hepatic tox­ic­ity in clin­i­cal trial such that it was approved with a
trans­am­i­nase ele­va­tions post vec­tor of unclear eti­­ol­ogy that were boxed safety warn­ing for acute liver injury. Subsequently, a case
not asso­ci­ated with liver dys­func­tion and resolved.11,15 Beyond report outlined 2 chil­dren presenting 6 to 8 weeks post Zol-
this, most trans­am­i­nase ele­va­tions observed in hemo­philia tri­als gensma infu­sion with acute liver fail­ure that resolved with glu­
occurred in the set­ting of an immune response to the rAAV cap­ co­cor­ti­coid inter­ven­tion.20 While the exact eti­­ol­ogy is unclear,
sid.9,14,16-18 This AAV cap­sid immune response is hypoth­e­sized to the tim­ing post vec­tor, liver biopsy spec­i­mens dem­on­strat­ing
occur when CD8+ T cells rec­og­nize cap­sid pep­tides on the sur­ CD8+ T-cell infil­tra­tion, and respon­sive­ness to ste­roids sug­gest
face of trans­duced hepa­to­cytes, trig­ger­ing a cel­lu­lar immune an immune-medi­ated pro­cess that is pos­si­bly con­sis­tent with
response that results in clear­ance of the trans­duced cells.19 While an rAAV cap­sid immune response. These obser­va­tions sup­port
the AAV cap­sid immune response has not posed safety con­cerns the pos­si­bil­ity that the rAAV cap­sid immune response has safety
in hemo­philia, it has dimin­ished or prevented effi­cacy in some impli­ca­tions and sup­port using the low­est pos­si­ble rAAV vec­tor
tri­als.9,16-18 Close mon­i­tor­ing for a cap­sid immune response within dose to min­i­mize safety con­cerns and max­i­mize effi­cacy. Addi-
the first 3 months or so post vec­tor clas­si­cally includes fac­tor tionally, a study using an rAAV8 vec­tor for X-linked myotubular
assays, liver func­ tion stud­ ies, and periph­ eral blood mono­ nu­ myop­a­thy reported that 3 of 17 boys in its 3.5 × 1014 vg/kg dose
clear cell inter­feron-γ anticapsid enzyme-linked immu­no­sor­bent cohort devel­oped liver fail­ure approx­i­ma­tely 6 weeks post vec­
spot assays that, when interpreted as con­sis­tent with a cap­sid tor and ulti­mately succumbed to com­pli­ca­tions.21 The gene ther­
immune response, trig­ger inter­ven­tion with immune-mod­u­lat­ing apy com­mu­nity eagerly awaits a com­pos­ite anal­y­sis of these
ther­a­pies to main­tain transgene expres­sion; glu­co­cor­ti­coids have obser­va­tions. Nonetheless, these tragic events sup­port pos­si­ble
been used pre­dom­i­nantly,9,16-18 but other immune-mod­u­lat­ing dose-lim­it­ing toxicities of sys­temic rAAV vec­tors at doses >1 × 1014
Prakash Singh Shekhawat
Update on clin­i­cal hemo­philia gene ther­apy  |  227
vg/kg, which are >1.6- to 500-fold higher than the rAAV doses
used in hemo­philia.
Finally, dor­sal root gan­glia (DRG) tox­ic­ity has recently emerged
as a poten­tial short- or long-term safety con­cern. Overwhelm-
ingly, these obser­va­tions are his­to­log­i­cal find­ings in large-ani­mal
mod­els. Specifically, a meta-anal­y­sis of NHP data (n = 33 vec­tors,
n = 256 NHPs) by a sin­gle lab­o­ra­tory fol­low­ing het­ero­ge­neous
routes of admin­is­tra­tion (intra­the­cal, intracisternal magna, and
sys­temic), vec­tor doses, AAV cap­sids, and transgenes reported
no or mild his­to­log­i­cal evi­dence of DRG tox­ic­ity in most ani­mals
that appeared to be dose depen­dent and more com­mon with
direct cen­tral ner­vous sys­tem admin­is­tra­tion.22 Importantly, clin­
i­cal symp­tom­atol­ogy in only 2 ani­mals was attrib­uted to DRG

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/226/1851420/226george.pdf by guest on 13 December 2021


tox­ic­ity. Subsequent NHP stud­ies by the same group suggested
DRG tox­ic­ity may be related to an unfolded pro­tein response
abro­gated when inhibiting transgene expres­sion in the DRG with,
as yet, unclear clin­i­cal trans­la­tion.23 Thus far, in the full cadre of
clin­ic­ al AAV tri­als span­ning 3 decades, var­io ­ us routes of admin­
is­tra­tion for a vari­ety of dis­or­ders, and an approx­i­mate 1000-fold
range of rAAV vec­tor doses,24 no DRG tox­ic­ity clin­i­cal symp­toms
have been observed out­side of a sin­gle recently reported case.
A trial par­tic­i­pant who received an rAAVrh10 vec­tor (a cap­sid not Figure 1. Biochemical characterization of FIX-Padua (FIX-R338L).
cur­rently used for hemo­philia) by intra­the­cal injec­tion to deliver Data support that FIX-R338L requires factor VIIIa (FVIIIa) cofactor
microRNA for amyotrophic lat­eral scle­ro­sis presented with symp­ function for enzymatic activity to generate factor Xa (FXa) from
toms and imag­ing con­sis­tent with DRG tox­ic­ity; symp­toms FX and is similarly activated for factor XIa (FIXa) and inactivated
dimin­ished with glu­co­cor­ti­coid and sup­port­ive care inter­ven­tion by antithrombin (AT). Modified with permission from Samelson-
but had not been com­pletely ame­lio­rated at the time of pub­ Jones et al.7
li­ca­tion.25 Whether DRG tox­ic­ity will be route of admin­is­tra­tion,
cap­sid, or rAAV vec­tor dose depen­dent and/or a poten­tial rAAV
vec­tor plat­form tox­ic­ity is unclear. infec­tion and other risk fac­tors for HCC devel­oped HCC approx­
The major iden­ti­fied long-term safety con­cerns of sys­temic i­ma­tely 1 year fol­low­ing vec­tor infu­sion. Recently presented
rAAV vec­tors are the risks of hep­a­to­tox­ic­ity and genotoxicity. tumor sequenc­ing data dem­on­strated the expected ran­dom,
Direct sequenc­ing data in ani­mals and humans dem­on­strate that low-fre­quency AAV inte­gra­tions with­out evi­dence of clonality
AAV inte­gra­tion can occur and has a pro­pen­sity for sites of active as well as well-described HCC-asso­ ci­
ated genetic changes,
tran­scrip­tion.26-30 Data in neo­na­tal mice dem­on­strated AAV inte­ suggesting that rAAV vec­tor admin­is­tra­tion did not con­trib­ute
gra­tion that disrupted the murine Rian locus (that does not have to HCC devel­op­ment.33 There is thus far no clin­i­cal evi­dence to
a human ortholog) and resulted in near 100% pen­e­trance of hepa­ sug­gest that sys­temic rAAV vec­tor deliv­ery is a risk fac­tor for
to­cel­lu­lar car­ci­noma (HCC).29 A sub­se­quent murine study dem­ the devel­op­ment of HCC. Ultimately, cohorts out­side the adult
on­strated that the risk of HCC cor­re­lated directly with the rAAV hemo­philia pop­ul­a­tion may more closely mimic the risk fac­tors
vec­tor dose and degree of cel­lu­lar divi­sion and that hepa­to­cyte- iden­ti­fied in mice for the devel­op­ment of HCC post sys­temic
spe­ cific enhancer and pro­ moter ele­
ments protected against rAAV vec­ tor infu­
sion. Specifically, infants and tod­dlers (who
onco­gen­e­sis, while non-hepa­to­cyte-spe­cific ele­ments increased have inher­ent greater degrees of cel­lu­lar divi­sion than adults)
HCC risk.27 This would thus sup­port using the low­est pos­si­ble who receive higher sys­temic rAAV vec­tor doses that con­tain
effec­tive vec­tor dose with hepa­to­cyte-spe­cific pro­moter and ubiq­ui­tous pro­mot­ers will be impor­tant pop­u­la­tions to fol­low
enhancer ele­ments to min­i­mize genotoxicity risk. Recently, stud­ to eval­ua ­ te long-term AAV genotoxicity.
ies in HA dogs followed for 10 years post rAAV vec­tor dem­on­
strated evi­dence of clonal pro­lif­er­a­tion at sites of AAV inte­gra­tion What are the transgene safety con­sid­er­ations?
in liver tis­sue with­out tumor­i­gen­e­sis but were the first large-ani­ The gen­ er­ ally iden­ti­
fied safety con­
cerns related to transgene
mal data to sup­port the the­o­ret­i­cal risk of genotoxicity.30 expres­sion include an immu­no­log­i­cal response to the expressed
Nearly all­severe hemo­philia patients >40 years of age con- pro­tein and direct transgene tox­ic­ity. Taking the lat­ter first, epi­
tracted iat­ ro­genic hep­ a­ti­
tis B virus and/or HCV, which are de­mi­o­log­i­cal stud­ies have outlined that supraphysiologic FIX:C
known risk fac­tors for HCC such that the hemo­philia pop­ul­a­tion has a mod­er­ate, inde­pen­dent risk of venous throm­bo­sis (odds
has an increased risk of HCC rel­a­tive to the gen­eral pop­ul­a­ ratio, 1.8-4.0) rel­a­tive to supraphysiologic FVIII:C (OR, 8.8-21.3).34
tion.31 Published 15-year fol­low-up data of the first par­tic­i­pants Biochemical data sup­port that FIX-R338L func­tion, like wild-type
to receive a sys­temic rAAV vec­tor (HB sub­jects) dem­on­strated FIX, requires FVIIIa cofac­tor func­tion for enzy­matic activ­ity and
no evi­dence of long-term hepatoxicity, but the report is lim­ited is sim­i­
larly acti­ vated by FXIa and inactivated by anti­ throm­ bin
in inter­pre­ta­tion due to small cohort size and the absence of (Figure 1), supporting that FIX-R338L is reg­u­lated the same as wild-
detect­able transgene expres­sion or direct sequenc­ing data to type FIX7 and is not inher­ently not prothrombotic. Consistent with
con­firm the per­sis­tence of trans­duced hepa­to­cytes.32 A sin­gle this, observed throm­botic events in mice expressing w ­ t-FIX or
clin­ic­ al trial par­tic­ip
­ ant in an HB trial with prior long-term HCV FIX-R338L cor­re­lated with the degree of supratherapeutic FIX:C,

228  |  Hematology 2021  |  ASH Education Program


Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/226/1851420/226george.pdf by guest on 13 December 2021
Figure 2. Aggregated reported annualized bleeding rate data for hemophilia A and B clinical trials before (blue) and after (maroon)
receiving the described recombinant AAV vectors. The dotted line denotes an annualized bleeding rate of 1.AAV8-wt-FIX, also known
as scAAV2/8-LP1-hFIXco. Data from Nathwani et al.45 AMT-060 data from Miesbach et al.12 AMT-061, now etranocogene dezaparvovec,
data from Pipe et al.8 SPK-9001, now fidancogene elaparvovec, data from George et al.9 BMN270, now valoctocogene roxaparvovec,
data from Pasi et al.11 SPK-8011 data from George et al.14

inde­ pen­ dent of the expressed transgene.35 Concurrently, HB fac­tor expo­sures). Nonetheless, inhib­i­tor risk post gene ther­apy
gene ther­apy tri­als using the FIX-R338L transgene achieved FIX:C is gen­er­ally thought to be unlikely because of small- and large-
in the range of mild or nor­mal HB with­out safety con­cerns. A sin­ ani­mal data that dem­on­strate the abil­ity of hepa­to­cyte-directed
gle FIX-R338L trial reported throm­bo­sis in a sub­ject who achieved gene ther­apy to induce tol­er­ance to FVIII, FIX, or FIX-R338L.35-37
supraphysiologic FIX:C with mul­ti­ple prothrombotic comorbid- These ani­mal stud­ies show that future gene trans­fer may ulti­
ities (obe­sity, kid­ney fail­ure with an arte­rio­ve­nous fis­tula).10 This mately be an effec­tive means for FVIII tol­er­ance induc­tion in
obser­va­tion when paired with epi­de­mi­o­log­i­cal data of supraphys- HA patients with inhib­i­tors; ini­ti­at­ing clin­i­cal tri­als for tol­er­ance
iologic fac­tor activ­ity and venous throm­bo­sis risk under­scores induc­tion was recently supported by con­sen­sus rec­om­men­da­
the impor­tance of maintaining expres­sion within a ther­a­peu­tic tions fol­low­ing a National Heart, Lung, and Blood Institute State
­win­dow. In addi­tion to throm­bo­sis risk, the expres­sion of FVIII or of the Science of FVIII inhib­i­tors.38
FIX-R388L could impart direct cel­lu­lar tox­ic­ity, which is one pos­si­
ble cause of the unex­pected declin­ing FVIII expres­sion observed What are the pre­lim­i­nary effi­cacy data?
in the first HA gene ther­apy trial detailed below. The approx­i­mate homo­ge­neous and remark­able phe­no­typic
While mul­ti­ple immu­no­log­i­cal responses to the expressed ame­lio­ra­tion that has been observed with het­ero­ge­neous trans-
transgene are pos­si­ble, the most concerning is the devel­op­ment gene-derived fac­ tor activ­
ity supporting fac­ tor activ­
ity itself
of inhib­i­tory antibodies, which have not been observed in clin­i­ should not be the sole distinguishing fea­ ture of clin­

cal pro­
cal trial. However, cur­rent trial enroll­ment includes patients who grams (Figure 2). Specifically, cur­rently avail­­able data are con­sis­
are the least likely to develop an inhib­i­tor (eg, par­tic­i­pants with tent with HA nat­u­ral his­tory stud­ies that sug­gest fac­tor activ­ity
prior or cur­rent inhib­i­tors are excluded and must have >50-150 >10% ame­lio­rates spon­ta­ne­ous bleed­ing; how­ever, higher fac­tor
Prakash Singh Shekhawat
Update on clin­i­cal hemo­philia gene ther­apy  |  229
Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/226/1851420/226george.pdf by guest on 13 December 2021
Figure 3. FVIII activity 1 and 2 years post vector infusion in trial participants who received a therapeutic vector dose (6 × 1013 vg/kg
of AAV5-hFVIII vs 5 × 1011 to 2 × 1012 vg/kg of SPK 8011) that maintained expression and were followed >2 years post vector. FVIII ac-
tivity is reported by 1-stage assay for SPK-8011 participants and CSA in participants who received AAV5-hFVIII. Data for AAV5-hFVIII
are from Pasi et al11 and data for SPK-8011 are from George et al.14

activ­ity is likely required among patients with marked preexist- 8 years post gene trans­fer.16,30,42,45,46 In con­trast, the first phase
ing joint dis­ease. Most tri­als report an unpre­dict­able range of 1/2 HA gene ther­apy trial of valoctocogene roxaparvovec
expres­sion of 5- to 10-fold var­ia ­ bil­ity (some tri­als up to 50- to at a dose of 6 × 1013 vg/kg dem­on­strated an aver­age loss of
100-fold) such that it is not cur­rently pos­si­ble to tar­get expres­ approx­i­ma­tely 40% of transgene expres­sion from year 1 to
sion based on indi­vid­ual patient needs.9,10,11,13,14,39 Correspondingly, year 2 post vec­tor (n = 7)11,15; sim­i­lar obser­va­tions were observed
an up to 7-fold var­i­abil­ity in trans­duc­tion was dem­on­strated in a among the 17 phase 3 par­tic­i­pants administered the same vec­
chi­me­ric murine model of human hepa­to­cytes.40 This degree of tor dose who were followed for >2 years.13 Available phase
var­i­able expres­sion in a clonal mouse pop­u­la­tion is con­sis­tent 1/2 data of par­tic­i­pants 5 years post valoctocogene roxapar-
with the mul­ ti­
ple var­
i­ables that exist from vec­ tor infu­
sion to vovec dem­on­strated a con­tin­ued, but lesser, decline in FVIII
transgene expres­sion1 that may neces­si­tate tol­er­ance for a range expres­sion in years 2 to 5.47 In con­trast, a phase 1/2 study of
of transgene expres­sion, albeit hope­fully to a lesser degree than SPK-8011 for HA at vec­tor doses 5 × 1011 to 2 × 1012 vg/kg dem­
has thus far been reported. This under­scores the need for an on­strated dif­fer­ent phar­ma­co­ki­net­ics of expres­sion such that
accu­rate mea­sure­ment of in vivo activ­ity of transgene hemo­ the 12 par­tic­i­pants with sustained expres­sion out­side the cel­
static func­tion to define the range of tol­er­a­ble safe and effi­ca­ lu­lar immune response and followed for >2 years dem­ on­
cious expres­sion. strated no appar­ent decline in expres­sion from year 1 to year 2
In HA and HB efforts, there is a dis­crep­ancy in transgene- (Figure 3).14 Some of these sub­jects have been followed for up to
derived fac­tor activ­ity by one-state assay (OSA) and chro­mo­genic 4 years and dem­on­strate gen­er­ally sta­ble FVIII expres­sion after
assay (CSA). Transgene-derived FVIII:C by OSA mea­sures approx­ 1 year post vec­tor; these pre­lim­i­nary SPK-8011 data gen­er­ally
i­ma­tely 1.6-fold higher than CSA in humans15,39-42 and mice,43 which sup­port the cur­rent strat­egy of targeting hepa­to­cytes for mul­
dif­fers from the expe­ri­ence with recom­bi­nant pro­tein prod­ucts ti­year sta­ble FVIII expres­sion. Given that the transgene is main-
of the same amino acid sequence. Which assay cor­re­lates with tained epi­som­ally, some degree of declin­ing expres­sion over
in vivo hemo­static ben­e­fit is unclear. Current tri­als tar­get exog­e­ mul­ti­year fol­low-up may be antic­i­pated, although it is unclear
nous FVIII expres­sion, and whether this mildly alters bio­chem­i­cal how rap­idly, but the mech­a­nism of loss or the more marked
prop­er­ties of the pro­tein is hypoth­e­sized (eg, changes in von Wil- decline in FVIII expres­sion from year 1 to year 2 observed with
lebrand fac­tor affin­ity or FVIII cleav­age by throm­bin or fac­tor Xa) valoctocogene roxaparvovec is unknown. Possible hypoth­ e­
but not deter­mined. Similarly, FIX-R338L deter­mined that FIX:C ses include (1) an unfolded pro­tein response, which has been
mea­sures higher by OSA vs CSA FIX:C; how­ever, unlike HA, this described in mam­ma­lian expres­sion sys­tems of recom­bi­nant
obser­va­tion is also maintained with recom­bi­nant FIX-R338L and FVIII pro­duc­tion and in mouse mod­els of supratherapeutic but
con­sis­tent with the avail­­able bio­chem­i­cal under­stand­ing of FIX- not low lev­els of FVIII expres­sion post rAAV48-50; (2) pro­moter
R338L.44 Additionally, unlike HA tri­als, OSA deter­mined that FIX:C silenc­ing; (3) an ongo­ing unde­tected/unmit­i­gated immune
from FIX-R338L expres­sion dif­fers by ini­ti­at­ing reagents.44 response to AAV or the transgene; or (4) fail­ure to form sta­ble
concatermized epi­somal DNA due to prop­er­ties of the vec­tor.
How long will the ther­apy last?
Thus far, clin­i­cal trial efforts in HB gene trans­fer mir­ror hemo­ What do AAV neu­tral­iz­ing antibodies mean?
philia canine mod­ els of rAAV-medi­ ated gene trans­ fer that AAV neu­tral­iz­ing antibodies (NAb) are incor­po­rated into most
dem­on­strated no decline in FVIII or FIX expres­sion for up to eli­gi­bil­ity cri­te­ria in hemo­philia and other dis­ease mod­els

230  |  Hematology 2021  |  ASH Education Program


Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/226/1851420/226george.pdf by guest on 13 December 2021
Figure 4. Summary of known and unknown variables for recombinant AAV gene edition clinical efforts for in hemophilia A and B.

using sys­temic AAV vec­tors. While results of the assay have What other approaches are in devel­op­ment?
an impact on eli­gi­bil­ity, the assays are not stan­dard­ized, can Safely achiev­ing sustained, sta­ble, and pre­dict­able FVIII or FIX
yield highly var­i­able results depending on the assay meth­ expres­ sion, even in the pres­ ence of an immune response, is
ods (eg, trans­duc­tion vs enzyme-linked immu­no­sor­bent assay an unre­al­ized goal of hemo­philia gene ther­apy. Beyond gene
based, employed multiplicity of infection, etc), and do not addi­tion approaches, gene editing using lipid nanoparticles to
always have a clear cor­re­la­tion with clin­i­cal out­come. None- deliver mRNA encoding Cas9 and gRNA and a donor FIX cDNA
theless, most avail­­able data sup­port the fact that high-titer tem­plate via an rAAV vec­tor to knockin F9 into the albu­men
AAV NAb pre­clude tar­get tis­sue trans­duc­tion of a sys­tem­i­ locus have dem­on­strated nor­mal lev­els of FIX expres­sion in an
cally admin­is­tered rAAV vec­tor and thus limit effi­cacy. A sin­gle NHP model.52 In addi­tion, lentiviral vec­tors for either sys­temic
AAV5 trial using a dose of 2 × 10 vg/kg is enroll­ing par­tic­i­pants infu­sion,53 ex vivo trans­duc­tion of hema­to­poi­etic stem cells,54-56
inde­pen­dent of AAV NAb sta­tus and has dem­on­strated trans- or induced plu­rip­o­tent stem cells are being pur­sued in pre­clin­
gene expres­sion in all­ par­tic­i­pants except the par­tic­i­pant with i­cal inves­ti­ga­tion and early-phase clin­i­cal trial (clinicaltrials.gov;
the highest mea­sured AAV NAb (>1:3000).8,13 These data chal­ NCT03818763).57
lenge existing notions that would oth­er­wise pre­dict preex-
isting AAV NAb to pre­clude effi­cacy. Details of the AAV NAb, Will hemo­philia gene ther­apy ever be main­stream?
thus far unre­ported, are nec­es­sary to inter­pret the data and Building on the remark­able successes the field has seen over the
whether these data are spe­cific to rAAV5 and/or the vec­tor past approx­i­ma­tely 5 years will be pred­i­cated on the thought­ful
dose used or may be par­tially explained by meth­ods used to inves­ti­ga­tion of ques­tions that have emerged from clin­i­cal tri­als.
per­form the AAV NAb. Optimism in the field will con­tinue to prog­ress, buoyed by 3 gen­
Following rAAV vec­tor admin­is­tra­tion, patients uni­ver­sally eral strengths: (1) a strong under­stand­ing of the molec­u­lar and
develop per­ sis­
tent, multiserotype cross-reac­ tive AAV NAb bio­chem­i­cal basis of HA and HB, (2) an orga­nized patient advo­
that avail­­able data sug­gest would pre­clude repeat rAAV vec­ cacy and phy­si­cian pro­vider net­work that fos­ters basic research
tor effi­cacy.32,51 This under­scores that patients inter­ ested in and clin­i­cal tri­als, and (3) a rap­idly expanding knowl­edge base in
gene ther­apy must rec­og­nize that the cur­rent state of clin­i­cal gene ther­apy with sig­nif­i­cant finan­cial invest­ment. Collectively,
devel­op­ment likely only per­mits 1 sys­temic rAAV vec­tor infu­ these strengths ensure that gene ther­apy will ful­fill its prom­ise to
sion. Thus, outlining the cur­rent known and unknown infor­ma­ alter the par­a­digm of hemo­philia care.
tion regard­ing rAAV gene ther­apy for hemo­philia is essen­tial
to pro­vide true informed con­sent for both clin­i­cal tri­als and Acknowledgments 
antic­i­pated soon-to-be licensed vec­tors (Figure 4); this will be This work was supported by NIH/NHLBI K08 HL 146991 (L. A.
nec­es­sary to avoid “buyer’s remorse” should one receive an George).
rAAV vec­tor that is ulti­mately found to be infe­rior to another Dr. George thanks Ben­ja­min Samelson-Jones, MD/PhD, for his
rAAV vec­tor. thought­ful review of the arti­cle.
Prakash Singh Shekhawat
Update on clin­i­cal hemo­philia gene ther­apy  |  231
Conflict-of-inter­est dis­clo­sure 19. Mingozzi F, Maus MV, Hui DJ, et  al. CD8(+) T-cell responses to adeno-
asso­ci­ated virus cap­sid in humans. Nat Med. 2007;13(4):419-422.
Lindsey A. George: sci­en­tific advi­sory board mem­ber: STRM.Bio;
20. Feldman AG, Parsons JA, Dutmer CM, et al. Subacute liver fail­ure fol­low­ing
data safety mon­i­tor­ing com­mit­tee mem­ber: Avrobio; con­sul­tancy: gene replace­ment ther­apy for spi­nal mus­cu­lar atro­phy type 1. J Pediatr.
Intellia, Biomarin, Pfizer, Bayer. 2020;225(Octo­ber):252-258.e1258e1.
21. Shieh PB, Bönnemann CG, Müller-Felber W, et  al. Re: “Moving Forward
Off-label drug use After Two Deaths in a Gene Therapy Trial of Myotubular Myopathy” by Wil-
son and Flotte. Hum Gene Ther. 2020;31(15-16):787.
Lindsey A. George: glucocorticoids were discussed.
22. Hordeaux J, Buza EL, Dyer C, et al. Adeno-asso­ci­ated virus-induced dor­sal
root gan­glion pathol­ogy. Hum Gene Ther. 2020;31(15-16):808-818.
Correspondence 23. Hordeaux J, Buza EL, Jeffrey B, et al. MicroRNA-medi­ated inhi­bi­tion of trans-
gene expres­sion reduces dor­sal root gan­glion tox­ic­ity by AAV vec­tors in
Lindsey A. George, University of Pennsylvania School of Med-
pri­ma­tes. Sci Transl Med. 2020;12(569):eaba9188.
icine, Colket Translational Research Bldg, Rm 5016, 3501 Civic 24. Wang D, Tai PWL, Gao G. Adeno-asso­ci­ated virus vec­tor as a plat­form for
Center Blvd, Philadelphia, PA 19104; e-mail: georgel@email​­.chop​ gene ther­apy deliv­ery. Nat Rev Drug Discov. 2019;18(5):358-378.
­.edu. 25. Mueller C, Berry JD, McKenna-Yasek DM, et  al. SOD1 sup­ pres­sion with

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/226/1851420/226george.pdf by guest on 13 December 2021


adeno-asso­ ci­ated virus and microRNA in famil­ ial ALS. N Engl J Med.
2020;383(2):151-158.
References 26. Nault JC, Datta S, Imbeaud S, et al. Recurrent AAV2-related inser­tional muta­
1. Li C, Samulski RJ. Engineering adeno-asso­ci­ated virus vec­tors for gene gen­e­sis in human hepa­to­cel­lu­lar car­ci­no­mas. Nat Genet. 2015;47(10):1187-
ther­apy. Nat Rev Genet. 2020;21(4):255-272. 1193.
2. Shahani T, Covens K, Lavend’homme R, et al. Human liver sinu­soi­dal endo­ 27. Chandler RJ, LaFave MC, Varshney GK, et  al. Vector design influ­ences
the­lial cells but not hepa­to­cytes con­tain fac­tor VIII. J Thromb Haemost. hepatic genotoxicity after adeno-asso­ ci­ated virus gene ther­ apy. J Clin
2014;12(1):36-42. Invest. 2015;125(2):870-880.
3. Pittman DD, Alderman EM, Tomkinson KN, Wang JH, Giles AR, Kaufman 28. Nakai H, Montini E, Fuess S, Storm TA, Grompe M, Kay MA. AAV sero­type
RJ. Biochemical, immu­no­log­i­cal, and in vivo func­tional char­ac­ter­iza­tion of 2 vec­tors pref­er­en­tially inte­grate into active genes in mice. Nat Genet.
B-domain-deleted fac­tor VIII. Blood. 1993;81(11):2925-2935. 2003;34(3):297-302.
4. McIn­tosh J, Lenting PJ, Rosales C, et al. Therapeutic lev­els of FVIII fol­low­ing 29. Donsante A, Miller DG, Li Y, et  al. AAV vec­tor inte­gra­tion sites in mouse
a sin­gle periph­eral vein admin­is­tra­tion of rAAV vec­tor encoding a novel hepa­to­cel­lu­lar car­ci­noma. Science. 2007;317(5837):477.
human fac­tor VIII var­i­ant. Blood. 2013;121(17):3335-3344. 30. Nguyen GN, Everett JK, Kafle S, et al. A long-term study of AAV gene ther­
5. Nathwani AC, Tuddenham E, Chowdary P, McIn­tosh J, et al. GO-8: pre­lim­ apy in dogs with hemo­philia A identifies clonal expan­sions of trans­duced
i­nary results of a phase I/II dose esca­la­tion trial of gene ther­apy for hae- liver cells. Nat Biotechnol. 2021;39(1):47-55.
mophilia A using a novel human fac­tor VIII var­i­ant. Blood. 2018;132(suppl 1): 31. Mazepa MA, Monahan PE, Baker JR, Riske BK, Soucie JM; US Hemophilia Treat-
489. ment Center Network. Men with severe hemo­philia in the United States:
6. Simioni P, Tormene D, Tognin G, et al. X-linked thrombophilia with a mutant birth cohort anal­ y­sis of a large national data­ base. Blood. 2016;127(24):
fac­tor IX (fac­tor IX Padua). N Engl J Med. 2009;361(17):1671-1675. 3073-3081.
7. Samelson-Jones BJ, Finn JD, George LA, Camire RM, Arruda VR. Hyperactiv- 32. George LA, Ragni MV, Rasko JEJ, et  al. Long-term fol­low-up of the first
ity of fac­tor IX Padua (R338L) depends on fac­tor VIIIa cofac­tor activ­ity. JCI in human intra­vas­cu­lar deliv­ery of AAV for gene trans­fer: AAV2-hFIX16 for
Insight. 2019;4(14):e128683. severe hemo­philia B. Mol Ther. 2020;28(9):2073-2082.
8. Pipe SW, Recht M, Key NS, et al. HOPE-B inves­ti­ga­tors 52 week effi­cacy and 33. Schmidt M, Foster GM, Coppens M, Thomsen H, et  al. Liver safety case
safety of etranacogene dezaparvovec in adults with severe or mod­er­ate- report from the phase 3 HOPE-B gene ther­apy trial in adults with hemo­philia
severe hemo­philia B: data from the phase 3 HOPE-B gene ther­apy trial B [ab­stract]. Res Pract Thromb Haemost. 2021;5(suppl 1). Abstract OC 67.4.
[ab­stract]. Res Pract Thromb Haemost. 2021;5(suppl 2). 34. Rietveld IM, Lijfering WM, le Cessie S, et al. High lev­els of coag­u­la­tion fac­
9. George LA, Sullivan SK, Giermasz A, et al. Hemophilia B gene ther­apy with a tors and venous throm­bo­sis risk: stron­gest asso­ci­a­tion for fac­tor VIII and
high-spe­cific-activ­ity fac­tor IX var­i­ant. N Engl J Med. 2017;377(23):2215-2227. von Willebrand fac­tor. J Thromb Haemost. 2019;17(1):99-109.
10. Chowdary P, Makris M, Evans G, et  al. A novel adeno asso­ci­ated virus 35. Crudele JM, Finn JD, Siner JI, et al. AAV liver expres­sion of FIX-Padua pre­
(AAV) gene ther­apy (FLT180a) achieves nor­mal FIX activ­ity lev­els in severe vents and erad­i­cates FIX inhib­i­tor with­out increas­ing thrombogenicity in
hemo­philia B (HB) patients (B-AMAZE study). Res Pract Thromb Haemost. hemo­philia B dogs and mice. Blood. 2015;125(10):1553-1561.
2020;4(suppl 2):17. 36. Finn JD, Ozelo MC, Sabatino DE, et al. Eradication of neu­tral­iz­ing antibod-
11. Pasi KJ, Rangarajan S, Mitchell N, et al. Multiyear fol­low-up of AAV5-hFVIII- ies to fac­tor VIII in canine hemo­philia A after liver gene ther­apy. Blood.
SQ gene ther­apy for hemo­philia A. N Engl J Med. 2020;382(1):29-40. 2010;116(26):5842-5848.
12. Miesbach W, Meijer K, Coppens M, et  al. Gene ther­apy with adeno- 37. Mingozzi F, Hasbrouck NC, Basner-Tschakarjan E, et al. Modulation of tol­er­
asso­ci­ated virus vec­tor 5-human fac­tor IX in adults with hemo­philia B. ance to the transgene prod­uct in a non­hu­man pri­mate model of AAV-medi­
Blood. 2018;131(9):1022-1031. ated gene trans­fer to liver. Blood. 2007;110(7):2334-2341.
13. Ozelo M, Mahlangu J, Pasi K, Giermasz A, et al; GENEr8-1 Trial Group. Efficacy 38. Ragni MV, George LA; for Members of Working Group 1, the National Heart,
and safety of valoctocogene roxaparvovec adeno-asso­ci­ated virus gene Lung, and Blood Institute State of the Science Workshop on Factor VIII Inhib-
trans­fer for severe hemo­philia A: results from the phase 3 GENEr8-1 trial itors: Generating a National Blueprint for Future Research. The national blue­
[ab­stract]. Res Pract Thromb Haemost. 2021;5(suppl 1). Abstract OC 26.1. print for future fac­tor VIII inhib­i­tor clin­i­cal tri­als: NHLBI State of the Science
14. George LA, Monahan PE, Eyster ME, Sullivan SK, et al. Phase I/II trial of SPK- (SOS) Workshop on fac­tor VIII inhib­i­tors. Haemophilia. 2019;25(4):581-589.
8011: sta­ble and dura­ble FVIII expres­sion after AAV gene trans­fer for hemo­ 39. Konkle BA, Stine K, Visweshwar N, Harrington TJ, et al. Updated fol­low-up
philia A [ab­stract]. Res Pract Thromb Haemost. 2021;5(suppl 1). Abstract of the Alta study, a phase 1/2, open-label, adap­tive, dose-rang­ing study to
OC 67.2. assess the safety and tol­er­ab ­ il­ity of SB-525 gene ther­apy. Blood. 2019;
15. Rangarajan S, Walsh L, Lester W, et  al. AAV5-fac­tor VIII gene trans­fer in 134(suppl 1):2060.
severe hemo­philia A. N Engl J Med. 2017;377(26):2519-2530. 40. Zou C, Vercauteren KOA, Michailidis E, et  al. Experimental var­i­ables that
16. Nathwani AC, Tuddenham EG, Rangarajan S, et al. Adenovirus-asso­ci­ated affect human hepa­to­cyte AAV trans­duc­tion in liver chi­me­ric mice. Mol Ther
virus vec­tor-medi­ated gene trans­fer in hemo­philia B. N Engl J Med. 2011; Methods Clin Dev. 2020;18(Sep­tem­ber):189-198.
365(25):2357-2365. 41. Rosen S, Tiefenbacher S, Robinson M, et al. Activity of transgene-pro­duced
17. Manno CS, Pierce GF, Arruda VR, et al. Successful trans­duc­tion of liver in B-domain-deleted fac­tor VIII in human plasma fol­low­ing AAV5 gene ther­
hemo­philia by AAV-fac­tor IX and lim­i­ta­tions imposed by the host immune apy. Blood. 2020;136(22):2524-2534.
response. Nat Med. 2006;12(3):342-347. 42. Nathwani AC, Tuddenham E, Chowdary P, et al. Adeno-asso­ci­ated medi­
18. Konkle BA, Walsh CE, Escobar MA, et al. BAX 335 hemo­philia B gene ther­ ated gene trans­fer for hemo­philia B: 8 year fol­low up and impact of remov­
apy clin­ i­
cal trial results: poten­ tial impact of CpG sequences on gene ing” empty viral par­ti­cles” on safety and effi­cacy of gene trans­fer. Blood.
expres­sion. Blood. 2021;137(6):763-774. 2019;132(suppl 1):491.

232  |  Hematology 2021  |  ASH Education Program


43. Sternberg AR, Davidson RJ, Samelson-Jones BJ, George L. In vitro and in 51. Long BR, Veron P, Kuranda K, et al. Early phase clin­i­cal immu­no­ge­nic­ity of
vivo mod­els to under­stand one-stage and chro­mo­genic fac­tor VIII activ­ity valoctocogene roxaparvovec, an AAV5-medi­ated gene ther­apy for hemo­
assay dis­crep­ancy of hepa­to­cyte-derived fac­tor VIII [ab­stract]. Res Pract philia A. Mol Ther. 2021;29(2):597-610.
Thromb Haemost. 2021;5(suppl 1). Abstract OC 75.3. 52. Huang H MEC, Bialek P, Wang C, et  al. CRISPR/Cas9-medi­ated targeted
44. Robinson MM, George LA, Carr ME, et al. Factor IX assay discrepancies in inser­tion of human F9 achieves ther­a­peu­tic cir­cu­lat­ing pro­tein lev­els in mice
the set­ting of liver gene ther­apy using a hyper­func­tional var­i­ant fac­tor IX- and non-human pri­ma­tes [ab­stract]. Mol Ther. 2019;27(April):7. Abstract 11.
Padua. J Thromb Haemost. 2021;19(5):1212-1218. 53. Cantore A, Ranzani M, Bartholomae CC, et al. Liver-directed lentiviral gene
45. Nathwani AC, Reiss UM, Tuddenham EG, et al. Long-term safety and effi­cacy ther­apy in a dog model of hemo­philia B. Sci Transl Med. 2015;7(277):277ra28.
of fac­tor IX gene ther­apy in hemo­philia B. N Engl J Med. 2014;371(21):1994- 54. Du LM, Nurden P, Nurden AT, et  al. Platelet-targeted gene ther­apy with
2004. human fac­tor VIII establishes haemostasis in dogs with haemophilia A. Nat
46. Niemeyer GP, Herzog RW, Mount J, et al. Long-term cor­rec­tion of inhib­i­ Commun. 2013;4(Novem­ber):2773.
tor-prone hemo­philia B dogs treated with liver-directed AAV2-medi­ated 55. Shi Q , Wilcox DA, Fahs SA, et al. Factor VIII ectop­i­cally targeted to plate­
fac­tor IX gene ther­apy. Blood. 2009;113(4):797-806. lets is ther­a­peu­tic in hemo­philia A with high-titer inhib­i­tory antibodies. J
47. Pasi KR, Rangarajan S, TM Robinson, Lester W, et al. Hemostatic response Clin Invest. 2006;116(7):1974-1982.
is maintained for up to 5 years fol­low­ing treat­ment with valoctocogene 56. Greene TK, Wang C, Hirsch JD, et al. In vivo effi­cacy of plate­let-deliv­ered,
roxaparvovec, an AAV5-hFVIII-SQ gene ther­apy for severe hemo­philia A high spe­cific activ­ity fac­tor VIII var­i­ants. Blood. 2010;116(26):6114-6122.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/226/1851420/226george.pdf by guest on 13 December 2021


[ab­stract]. Res Pract Thromb Haemost. 2021;5(suppl 1). Abstract OC 67.1. 57. Follenzi A, Benten D, Novikoff P, Faulkner L, Raut S, Gupta S. Transplanted
48. Lange AM, Altynova ES, Nguyen GN, Sabatino DE. Overexpression of fac­ endo­the­lial cells repopulate the liver endo­the­lium and cor­rect the phe­no­
tor VIII after AAV deliv­ery is tran­siently asso­ci­ated with cel­lu­lar stress type of hemo­philia A mice. J Clin Invest. 2008;118(3):935-945.
in hemo­philia A mice. Mol Ther Methods Clin Dev. 2016;3(28 Sep­tem­ 58. Pipe S, Becka M, Detering E, Vanevski K, Lissitchkov T. First-in-human gene
ber):16064. ther­apy study of AAVhu37 cap­sid vec­tor tech­nol­ogy in severe hemo­philia
49. Zolotukhin I, Markusic DM, Palaschak B, Hoffman BE, Srikanthan MA, A. Blood. 2019;134(suppl 1):4630.
Herzog RW. Potential for cel­lu­lar stress response to hepatic fac­tor VIII
expres­sion from AAV vec­tor. Mol Ther Methods Clin Dev. 2016;3(28 Sep­
tem­ber):16063.
50. Poothong J, Pottekat A, Siirin M, et al. Factor VIII exhib­its chap­er­one-depen­
dent and glu­cose-reg­u­lated revers­ible amy­loid for­ma­tion in the endo­plas­ © 2021 by The Amer­i­can Society of Hematology
mic retic­u­lum. Blood. 2020;135(21):1899-1911. DOI 10.1182/hema­tol­ogy.2021000254

Prakash Singh Shekhawat


Update on clin­i­cal hemo­philia gene ther­apy  |  233
HODGKIN LYMPHOMA: CURE AND OPTIMAL SURVIVORSHIP

Controversies in the management of early-stage


Hodgkin lymphoma

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/234/1851889/234blum.pdf by guest on 13 December 2021


Kristie A. Blum
Department of Hematology and Medical Oncology, Emory Winship Cancer Institute, Atlanta, GA

Positron emission tomography (PET)–adapted chemotherapy and radiotherapy approaches are currently used for the ini-
tial treatment of early-stage Hodgkin lymphoma (HL) with progression-free survival and overall survival exceeding 85%
and 95%, respectively. However, despite general agreement on the prognostic value of interim PET in HL, frontline treat-
ment approaches vary among institutions with respect to how pretreatment clinical risk factors determine treatment
selection, the definition of PET negativity, which chemotherapy regimen to initiate and how many cycles to administer,
and when to incorporate radiation. Furthermore, as recent trials have confirmed improved efficacy and manageable
toxicity when brentuximab and checkpoint inhibitors are combined with frontline regimens such as doxorubicin, vinblas-
tine, and dacarbazine in advanced-stage HL, these agents are now under evaluation as frontline therapy in early-stage
HL. A number of issues will affect the use of these agents in early-stage HL, including the costs, early and late toxicities
with these agents, patient population (favorable or unfavorable risk groups), how to incorporate them (concurrently or
sequentially), and whether they can ultimately replace cytotoxic therapy with similar efficacy and fewer late effects.
Future treatment paradigms for early-stage HL may change significantly once randomized studies are completed incor-
porating these agents into frontline therapy. Ideally, frontline use of brentuximab and checkpoint inhibitors in early-stage
HL will result in improved outcomes compared with current PET-adapted approaches with decreased risks of late toxici-
ties that continue to afflict long-term survivors of HL.

LEARNING OBJECTIVES
• Describe current frontline treatment approaches in patients with early-stage HL
• Describe recent clinical trials and controversies surrounding the incorporation of brentuximab and checkpoint inhib-
itors into frontline therapy in early-stage HL

white blood cell count of 12,400/microliter, hemoglobin of


CLINICAL CASE 11.9g/dL, platelet count of 471 000, and an elevated eryth-
A 20-year-old woman has bilateral cervical lymph node rocyte sedimentation rate (ESR) of 77mm/h. Therefore, she
enlargement but no fevers, night sweats, or weight loss. has nonbulky stage IIA classic HL, with adverse features
She was initially treated with antibiotics and steroids by her including involvement of 5 nodal sites and an elevated ESR.
primary care physician without improvement. Core needle
biopsy specimen of a right cervical lymph node was suspi-
cious but not diagnostic for classic Hodgkin lymphoma (HL). Introduction
Excisional lymph node biopsy confirmed classic HL, and A number of controversies surround treatment decisions
positron emission tomography (PET) scan demonstrated in patients diagnosed with early-stage HL, and institutions
numerous cervical, supraclavicular, mediastinal, and axillary vary in their approaches to these patients. Clinical trials that
nodes, including a left posterior cervical node of 1.2×1cm guide treatment decisions in early-stage HL differ in the
with standardized uptake value (SUV) 7.3, a right supracla- designation of pretreatment risk factors, eligibility criteria,
vicular node measuring 2.7×3.5cm with SUV 10.6, a medias- definition of response based on interim PET, and how novel
tinal node measuring 2.3×1.3cm with SUV 5.5, and right and agents are incorporated. In addition, long-term follow-up
left axillary lymphadenopathy. No intra-abdominal or splenic is lacking from many of these trials, and recent studies con-
uptake was noted. Her laboratory testing demonstrated a tinue to show that patients with classic HL remain at risk

234 | Hematology 2021 | ASH Education Program


for late com­pli­ca­tions from ther­apy despite improve­ments and and 9 of 11 solid tumors occurred within the radi­a­tion field. Most
reduc­tions in ther­apy. Therefore, cur­rent treat­ment approaches solid tumors were pap­il­lary thy­roid car­ci­noma, but there was
need to bal­ance the com­pet­ing risks of achiev­ing high clin­i­cal 1 breast can­cer that did not develop until 13 years after study
effi­cacy while min­i­miz­ing late tox­ic­ity. This review sum­ma­rizes enroll­ment. Therefore, it is likely with lon­ger fol­low-up that addi­
recent data regard­ing late toxicities in patients with early-stage tional sec­ond­ary neo­plasms, includ­ing breast and lung can­cers,
HL, PET-adapted ther­a­peu­tic approaches for these patients, the will be observed despite a PET-adapted approach that lim­its
treat­ment of bulky HL, and the incor­po­ra­tion of brentuximab both che­mo­ther­apy and radi­a­tion doses.
and check­point inhib­i­tors into the treat­ment of early-stage HL. In 3905 Dutch patients aged 15 to 50 years who were treated
In addi­tion, the review high­lights ongo­ing ques­tions in the field, between 1965 and 2000, with 60.5% receiv­ing CMT, the cumu­
includ­ing which che­mo­ther­apy back­bone to ini­ti­ate, the num­ber la­tive inci­dence of sub­se­quent solid neo­plasms did not dif­fer
of treat­ment cycles to admin­is­ter, def­i­ni­tions of PET neg­a­tiv­ity, sig­nif­i­cantly (P = .71) among treat­ment eras: 1965 to 1976, 1977 to
when to incor­po­rate radio­ther­apy, how to treat high-risk pa- 1988, and 1989 to 2000.3 In addi­tion, the risk for any sec­ond can­
tients who are interim or end-of-ther­apy PET pos­i­tive, treat­ment cer remained high for up to 40 years after treat­ment for HL, with
of bulky HL, and how to include brentuximab and check­point a 48.5% cumu­la­tive inci­dence. Bright et al4 also showed that the

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/234/1851889/234blum.pdf by guest on 13 December 2021


inhib­i­tors in front­line ther­apy. risk of solid tumors con­tin­ues to increase annu­ally, even up to
35 years after treat­ment for HL. In their study of 16 971 sur­vi­vors
Late toxicities of treat­ment of clas­sic HL of HL aged 15 to 39 years who were treated from 1971 to 2006,
in the mod­ern era the cumu­la­tive inci­dence of sub­se­quent pri­mary neo­plasms was
Despite advances in front­line che­mo­ther­apy and radio­ther­apy 0.9% and 26.6% in females and 0.6% and 16.5% in males at 10 and
for patients with early-stage HL, these patients still expe­ri­ence 35 years from diag­no­sis. Taken together, these stud­ies all­dem­
a life­long risk of sec­ond­ary malig­nan­cies and car­dio­vas­cu­lar on­strate that the risk of sec­ond neo­plasms con­tin­ues to rise with
dis­ease. In an anal­y­sis of Surveillance, Epidemiology, and End long-term fol­low-up and that most of these sec­ond solid tumors
Results data on 20,007 sur­vi­vors of HL aged 20 to 74 years diag­ are not observed until after at least 10 years from diag­no­sis. In
nosed from 2000 to 2015, Dores et al1 ascertained that the risk of addi­tion, the risk of sec­ond malig­nant neo­plasms con­tin­ues to
death due to noncancer causes and sec­ond neo­plasms remains per­sist in the cur­rent treat­ment era despite improved che­mo­
sig­nif­i­cantly ele­vated over the gen­eral US pop­u­la­tion, even in ther­apy and reduced radi­a­tion expo­sure.
the mod­ern treat­ment era. In this study, 60% of patients had
stage I/II dis­ease, and median fol­low-up was 8 years. Noncancer PET-adapted front­line treat­ment
causes of mor­tal­ity in early-stage patients included inter­sti­tial As a result of the persisting evi­dence of late com­pli­ca­tions even
lung dis­ease, infec­tion, benign hema­to­logic dis­ease (cytopenias with mod­ern che­mo­ther­apy and radi­a­tion tech­niques and with
and clot­ ting), heart dis­ease, and dia­be­tes.1 Other neo­ plasms the proven prog­nos­tic sig­nif­i­cance of interim PET in early-stage
accounted for 25% of all­nonlymphoma deaths regard­ less of HL,5 many ado­les­cent and young adult (AYA) and adult patients
stage, with 145 cases of sec­ond­ary neo­plasms in early-stage and with early-stage HL cur­rently receive PET-adapted ther­apy in
144 cases in advanced-stage patients. Stage-spe­cific mor­tal­ity an effort to fur­ther reduce che­mo­ther­apy and radi­a­tion expo­
trends are declin­ing in the mod­ern treat­ment era with all­-cause, sure. The stud­ies supporting PET-adapted ther­apy dif­fer slightly
noncancer, and other neo­plasm stan­dard­ized mor­tal­ity ratios of in eli­gi­bil­ity cri­te­ria, in the treat­ment reg­i­men, and in defin­ing
7.0, 2.9, and 2.9 for early-stage patients treated from 1983 to 1991 PET neg­a­tiv­ity, mak­ing com­par­i­sons across stud­ies chal­leng­ing.
vs 2.9, 1.5, and 1.6 treated from 2001 to 2009, respec­tively. How- Table 1 sum­ma­rizes these stud­ies that cur­rently guide treat­ment
ever, fol­low-up is short for those patients treated from 2001 to in AYA and adult HL.
2009, and these mor­tal­ity rates still exceed rates in the gen­eral In the UK RAPID study,7 602 nonbulky stage I to IIA patients
pop­u­la­tion. (32.3% unfa­vor­able by German Hodgkin Study Group cri­te­ria)
Focusing on sec­ond­ary neo­plasms, the Children’s Oncology received 3 cycles of doxorubicin, bleomycin, vinblastine, and
Group published a 10-year fol­low-up on AHOD0031, a trial of 1711 dacarbazine followed by a PET scan. Patients with a Deauville
patients aged up to 21 years treated with response-adapted score of 1 to 2 were con­ sid­ered PET neg­ a­
tive and ran­
dom­
ther­apy.2 All patients ini­tially received doxo­ru­bi­cin, bleomycin, ized to either 30 Gy IFRT or no fur­ther treat­ment, and patients
vin­cris­tine, etoposide, pred­ni­sone, and cyclo­phos­pha­mide, and with a Deauville score of 3 to 5 under­went a fourth cycle of
rapid early respond­ers (deter­mined by PET) were ran­domly allo­ ABVD and IFRT. Seventy-four per­cent of patients were PET
cated to 21 Gy involved field radio­ther­apy (IFRT) or obser­va­tion. ­neg­a­tive, and 3-year pro­gres­sion-free sur­vival (PFS) was 94.6%
Slow respond­ers were ran­dom­ized to IFRT or dexa­meth­a­sone, with IFRT and 90.8% with obser­va­tion (P = .02). No OS ben­e­fit
etoposide, cis­platin, and cytarabine plus IFRT. Ten-year event- was observed, with a 3-year OS of 97.1% com­pared with 99%
free sur­vival and over­all sur­vival (OS) were not sig­nif­i­cantly dif­ with and with­out radi­a­tion (P = .27). For the PET-pos­i­tive patients,
fer­ent in the rapid early respond­ers treated with obser­va­tion 3-year PFS was 83%. In an anal­y­sis of the RAPID study by Deau-
vs IFRT and also not dif­fer­ent in slow respond­ers treated with ville score,7 infe­rior out­comes were seen only in patients with
dexa­meth­a­sone, etoposide, cis­platin, and cytarabine plus IFRT scores of 5, with 5-year PFS of 91.5%, 91.1%, 95.3%, 87.5%, and
vs IFRT alone. The cumu­la­tive inci­dence of sec­ond neo­plasms 61.9% in patients with scores of 1, 2, 3, 4, and 5, respec­tively.
was 1.3% with 17 sec­ond malig­nan­cies (3 cases of acute mye­ Cancer and Leukemia Group B 50604 exam­ ined a PET-
loid leu­ke­mia, 11 solid tumors, and 3 cases of NHL). Sixteen of adapted approach in patients aged 18 to 60 years with nonbulky
these malig­nan­cies occurred in patients treated with com­bined stage I and II HL with and with­out B-symp­toms.8 All patients
modal­ity ther­apy (CMT) with doxo­ru­bi­cin, bleomycin, vin­cris­tine, received 2 cycles of ABVD, interim PET, and, if PET neg­a­ tive
etoposide, pred­ni­sone, and cyclo­phos­pha­mide plus 21 Gy IFRT, (Deauville scores 1-3), 2 addi­tional ABVD cycles with­out IFRT.
Prakash Singh Shekhawat
Management of early-stage HL  | 235
Table 1. PET-adapted ther­a­peu­tic tri­als in early-stage HL

Study (median fol­low-up) Patient pop­u­la­tion (N) Risk fac­tors at enroll­ment PET neg­a­tive Treatment arms PFS or FFTF OS
6
UK RAPID (5.0 years) Stage IA or IIA (n = 602) Nonbulky Deauville 1-2 PET neg: ABVD × 3 3-year 90.8% 3-year 99.0%
PET neg: ABVD × 3 + 30 Gy IFRT 3-year 94.6% 3-year 97.1%
PET pos: ABVD × 4 + 30 Gy IFRT 3-year 83%
CALGB 506048 (3.8 years) Stage I or II (n = 164) Nonbulky Deauville 1-3 PET neg: ABVD × 4 3-year PFS 91%
PET pos: ABVD × 2, eBEACOPP × 2, 3-year PFS 66%
30 Gy IFRT
EORTC9 (4.5-5.1 years) Stage I or II (n = 1950) Favorable (no risk fac­tors) Deauville 1-2 (F) PET neg: ABVD × 3 + 30 Gy INRT 5-year PFS 99% 5-year OS 100%

236  |  Hematology 2021  |  ASH Education Program


Unfavorable (any of the fol­low­ing) (F) PET neg: ABVD × 4 5-year PFS 87.1% 5-year OS 99.6%
1. Age ≥50
(U) PET neg: ABVD × 4 + 30 Gy INRT 5-year PFS 92.1% 5-year OS 96.7%
2. Bulk
(U) PET neg: ABVD × 6 5-year 89.6% 5-year OS 98.3%
3. >3 nodal sites
4. ESR ≥50 (or 30 if B-symp­toms) (F/U) PET pos: ABVD × 4 + 30 Gy INRT 5-year PFS 77.4% 5 year OS 89.3%
(F/U) PET pos: ABVD × 2, eBEACOPP × 2, 5-year PFS 90.6% 5 year OS 96.0%
30 Gy INRT
GHSG HD1610 (45 months) Stage I or II (N = 1150) Favorable (none of the fol­low­ing Deauville 1-2 PET neg: ABVD × 2 + 20 Gy IFRT 5-year PFS 93.4% 5-year OS 98.1%
risk fac­tors): PET neg: ABVD × 2 5-year PFS 86.1% 5-year OS 98.4%
1. Bulk PET pos: ABVD × 2 + 20 Gy IFRT 5-year PFS 88.4% 5-year OS 97.9%
2. Extranodal sites
3. >2 nodal areas
4. ESR ≥50 (or 30 if B-symp­toms)
GHSG HD1711 (46.2 months) Stage I or II (N = 1100) Unfavorable (with one of the Deauville 1-2 PET neg­a­tive: eBEACOPP/ABVD × 4 +  5-year PFS 97.7% 5-year OS 98.7%
fol­low­ing risk fac­tors): 30 Gy IFRT 5-year PFS 95.9% 5-year OS 98.8%
1. Bulk PET neg: eBEACOPP/ABVD × 4 5-year PFS 94.2% 5-year OS 99.2%
2. Extranodal sites PET pos: eBEACOPP/ABVD × 4 + 30 Gy
3. >2 nodal areas IFRT
4. ESR ≥50 (or 30 if B-symp­toms)
eBEACOPP, esca­lated bleomycin, etoposide, doxo­ru­bi­cin, cyclo­phos­pha­mide, vin­cris­tine, pro­car­ba­zine and pred­ni­sone; EN, XXX; F, favor­able; FFTF, free­dom from treat­ment fail­ure; neg, neg­a­
tive; pos, pos­i­tive; U, unfa­vor­able.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/234/1851889/234blum.pdf by guest on 13 December 2021


PET-pos­i­tive patients switched to esca­lated bleomycin, etopo- PET-neg­a­tive patients. On the basis of the sec­ond­ary ana­ly­ses of
side, doxo­ru­bi­cin, cyclo­phos­pha­mide, vin­cris­tine, pro­car­ba­zine, the asso­ci­a­tion of PFS by Deauville score from the RAPID,11 HD16,9
and pred­ni­sone (BEACOPP) with IFRT. In total, 164 patients were and HD1710 stud­ies, which dem­on­strated sig­nif­i­cantly worse PFS
enrolled, 26% with B-symp­toms and 58% unfa­vor­able by GHSG only in patients with Deauville scores of 4 to 5, I define PET neg­a­
cri­te­ria. With the expan­sion of PET neg­a­tiv­ity to Deauville scores tive as Deauville scores of 1 to 3 and often omit radio­ther­apy even
of 1 to 3, 91% of patients were PET neg­a­tive com­pared with 76% in patients with a Deauville score of 3. With this approach, more
if Deauville scores of 1 to 2 were used. Three-year PFS was 91% than 90% of patients with early-stage HL can avoid radio­ther­apy.
for PET-neg­a­tive patients and 66% for PET-pos­i­tive patients. For However, as all­patients in the RAPID, HD16, and HD17 tri­als with
patients with Deauville scores of 1 to 2 (n = 113), 3 (n = 22), and 4 to a Deauville score of 3 received CMT and CALGB 50604 dem­on­
5 (n = 14), 3-year PFS was 94%, 77%, and 67%, respec­tively. strated an infe­rior PFS of 77% in 22 patients with a Deauville score
The larg­est PET-adapted study, European Organization for of 3 treated with che­mo­ther­apy alone, the use of radio­ther­apy
Research and Treatment of Cancer H10, enrolled 1950 stage I to II is cer­tainly jus­ti­fied in patients with a Deauville score of 3 and
patients, with 754 favor­able and 1196 unfa­vor­able HL.9 In the con­ should be discussed with the patient and a mul­ ti­
dis­
ci­
plin­
ary
trol arms, treat­ment consisted of 3 (favor­able) or 4 (unfa­vor­able) treat­ment team and weighed against poten­tial late effects.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/234/1851889/234blum.pdf by guest on 13 December 2021


ABVD cycles and involved nodal radio­ther­apy (INRT), regard­ In addi­ tion to the ongo­ ing con­ tro­
versy over PET-directed
less of PET results. In the exper­i­men­tal arms, patients received radio­ther­apy, these tri­als also high­light other debates in early-​
2 cycles of ABVD, interim PET, and, if PET neg­a­tive (Deauville stage HL, includ­ing num­ber of che­mo­ther­apy cycles, which reg­i­
scores 1-2), 2 (favor­able) or 4 (unfa­vor­able) addi­tional cycles of men to start (ABVD or esca­lated BEACOPP), and treat­ment of
ABVD. PET-pos­ i­
tive patients switched to esca­ lated BEACOPP PET-pos­i­tive patients. Although the low-risk patient could receive
and INRT. Eighty-seven per­cent of favor­able and 77.6% of unfa­ as few as 2 to 4 ABVD cycles, unfa­vor­able patients in the EORTC
vor­able patients were PET neg­a­tive. Five-year PFS rates in the and HD17 tri­als seem to have improved out­comes with 6 cycles of
favor­able PET-neg­a­tive patients were 99% with 3 cycles of ABVD ABVD or esca­ lated BEACOPP. In my own prac­ tice, I tend to
and INRT vs 87.1% with 4 cycles of ABVD alone. In unfa­vor­able admin­is­ter 3 to 4 cycles of ABVD alone in nonbulky, unfa­vor­able
PET-neg­a­tive patients, 5-year PFS was 92.1% with 4 cycles of patients who are interim PET neg­a­tive based on the UK Rapid and
ABVD and INRT vs 89.6% with 6 cycles of ABVD. In both favor­ CALGB stud­ies but rec­og­nize that 6 cycles can also be con­sid­
able and unfa­vor­able cohorts, noninferiority for che­mo­ther­apy ered for these patients, par­tic­u­larly if omit­ting radio­ther­apy.
alone could not be dem­on­strated. Five-year OS was not sig­nif­ For those patients with interim PET scores of 4 to 5, it remains
i­cantly dif­fer­ent at 99.6% and 98.3% with ABVD only and 100% unclear if these patients should switch ther­apy from ABVD to
and 96.7% with ABVD and INRT in favor­able and unfa­vor­able esca­lated BEACOPP. In EORTC H10, esca­lated BEACOPP and INRT
patients, respec­tively. For PET-pos­i­tive patients, 5-year PFS with in interim PET-pos­i­tive patients improved PFS to 90.6% vs 77.4%
esca­lated BEACOPP was 90.6% com­pared with 77.4% with 3 to 4 with ABVD and INFRT. However, in the RAPID and HD16 tri­als,
cycles of ABVD and INRT (P = .002), with no OS ben­e­fit (P = .062). PET-pos­i­tive patients received ABVD and IFRT with­out che­mo­
The GHSG conducted the HD16 trial in 1150 patients with favor­ ther­apy inten­si­fi­ca­tion, and PFS was 87.6% and 88.4%, respec­
able stage I to II HL exam­in­ing 2 cycles of ABVD and 20 Gy IFRT tively. In my own prac­tice, I avoid the use of esca­lated BEACOPP
com­pared with PET-guided treat­ment with 2 cycles of ABVD, PET, even in interim PET-pos­i­tive patients due to the poten­tial risks of
and no radio­ther­apy if PET neg­a­tive (Deauville scores 1-2) and 20 infer­til­ity and sec­ond­ary malig­nan­cies with the reg­i­men. I typ­i­
Gy IFRT if PET pos­i­tive.10 In PET-neg­a­tive patients, 5-year PFS was cally rec­om­mend IFRT for a Deauville score of 4 and biopsy fol-
93.4% with CMT and 86.1% with ABVD alone (P = .04). No OS ben­ lowed by sal­vage che­mo­ther­apy and autol­o­gous trans­plant for
e­fit was noted with 5-year OS of 98.1% with CMT and 98.4% with a Deauville score of 5.
ABVD (P = .12). Notably, PFS was sig­nif­i­cantly worse in patients
with Deauville scores of 4 to 5, with 5-year PFS of 80.9%, 93.1%, Treatment of bulky dis­ease
and 93.2% for scores of 4 to 5, 1 to 3, and 1 to 2, respec­tively. In bulky HL, 4 stud­ies sug­gest that radi­a­tion can be elim­i­nated
The GHSG HD17 trial exam­ined 2 cycles of esca­lated BEACOPP in PET-neg­a­tive patients with­out com­pro­mis­ing out­comes. The
plus 2 cycles of ABVD followed by 30 Gy IFRT in unfa­vor­able ear- Brit­ish Colombia Cancer Agency omit­ted radio­ther­apy in patients
ly-stage HL com­pared with an exper­i­men­tal arm omit­ting IFRT in with stage I to II bulky, IIB, and III to IV HL who were PET neg­
PET-neg­a­tive (Deauville scores 1-2) patients after 4 cycles of che­ a­tive (Deauville scores 1-3) after 6 cycles of ABVD.12 Eighty-four
mo­ther­apy. In the PET-neg­a­tive patients, 5-year PFS was 97.7%
11
per­cent were PET neg­a­tive and did not receive radi­a­tion. Five-
with CMT, not sta­tis­ti­cally dif­fer­ent com­pared with 95.9% with year free­dom from treat­ment fail­ure was 89% for PET-neg­a­tive
che­mo­ther­apy alone. As in the HD16 trial, a Deauville score of 4 com­pared with 56% for PET-pos­i­tive patients. In PET-neg­a­tive
to 5 was a sig­nif­i­cant risk fac­tor for poor PFS, whereas scores of patients with bulk (n = 112), 5-year free­dom from treat­ment fail­
1 to 3 were not asso­ci­ated with PFS in the mul­ti­var­i­able model. ure was 89% com­pared with 88.5% for PET-neg­a­tive nonbulky
Although the RAPID, EORTC H10, and GHSG HD16 tri­als all­ dis­ease (n = 152).
failed to show noninferiority in PFS with PET-adapted omis­sion In CALGB 50801, 101 patients with bulky stage I to II HL were
of radio­ther­apy com­pared with CMT, par­tic­u­larly in favor­able treated with 2 cycles of ABVD followed by interim PET.13 PET-neg­
patients, there was no OS ben­e­fit with inclu­sion of radi­a­tion. a­tive (Deauville scores 1-3) patients received 4 addi­tional cycles
Therefore, the treating phy­si­cian needs to weigh the risks of of ABVD and no radi­a­tion, and PET-pos­i­tive patients received 4
poten­ tial late toxicities with radio­ ther­apy against the risk of cycles of esca­lated BEACOPP and 30 Gy involved site radio­ther­
relapse in their patients when deter­min­ing if radio­ther­apy should apy (ISRT). Seventy-eight per­cent were PET neg­a­tive, and 3-year
be included. Based on data show­ing con­tin­ued risk of late sec­ PFS was 93.1% com­pared with 89.7% for PET-pos­i­tive patients,
ond malig­nan­cies with mod­ern radio­ther­apy tech­niques and the confirming that a PET-adapted approach elim­i­nat­ing radi­a­tion
lack of OS ben­e­fit with IFRT, my prac­tPrakash
ice is to omitSingh Shekhawat
radio­ther­
apy in leads to dura­ble remis­sions in bulky HL.

Management of early-stage HL  | 237


In the Gruppo Italiano Terapie Innovative nei Linfomi/Fonda- pleted a phase 2 study of 3 doses of pembrolizumab followed by
zione Italiana Linfomi HD 0607 study,14 patients with stage IIB to 4 to 6 cycles of AVD with­out radio­ther­apy in 12 early-stage and 18
IVB HL who were PET neg­a­tive after 6 cycles of ABVD and who advanced-stage patients, includ­ing 10 patients with bulky tumors
had a large nodal mass 5 cm or larger under­went ran­dom­i­za­tion more than 10 cm.24 After 3 cycles of pembrolizumab, the CR rate
to 30 Gy IFRT or no fur­ther treat­ment. In the 296 PET-neg­a­tive was 37% and improved to 100% after 2 cycles of AVD. Two-year
patients with a large nodal mass, there was no sig­nif­i­cant dif­ PFS was 100%. Grade 3 to 4 events included neutropenia in 3
fer­ence in 3-year PFS with a PFS of 93% with­out radio­ther­apy patients and transaminitis, lymphopenia, diar­rhea, and Bell palsy
com­pared with 97% with IFRT (P = .29). Even when lim­it­ing the in 1 patient each.
anal­y­sis to patients with bulk more than 10 cm, 3-year PFS was Therefore, BV and check­ point inhib­ i­
tors appear safe and
94% with IFRT and 86% for obser­va­tion (P = .34). Last, the UK effec­tive in early-stage and bulky HL with PFS exceed­ing 90%;
RATHL study included 500 patients with bulky or high-risk how­ever; large ran­dom­ized tri­als are nec­es­sary to estab­lish the
stage II HL and dem­on­strated a 90.9% PFS in these patients if safety, cost-effec­tive­ness, and long-term effi­cacy in com­par­i­
PET neg­a­tive after 6 cycles of ABVD.15 Therefore, based on these son to cur­rent PET-adapted ABVD ther­apy. Although sequen­tial
stud­ies, I omit IFRT in patients with bulky early-stage HL who admin­is­tra­tion may min­i­mize tox­ic­ity, these prolonged treat­

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/234/1851889/234blum.pdf by guest on 13 December 2021


achieve a neg­a­tive PET. ment approaches may lead to sig­nif­i­cant edu­ca­tion, childcare,
finan­cial, and employ­ment con­straints in an AYA pop­u­la­tion.
Incorporation of novel agents Alternatively, as dem­on­strated by Abramson et al19 and Allen et
Unlike advanced-stage HL in which large ran­dom­ized stud­ies al,24 finan­cial costs and treat­ment risks may be min­i­mized with
have exam­ined brentuximab vedotin (BV) and check­point inhib­ 1 to 3 lead-in cycles of BV or check­point inhib­i­tor prior to AVD,
i­tors com­bined with doxo­ru­bi­cin, vin­blas­tine, and dacarbazine and these lim­ited approaches should also be eval­u­ated. In con­
(AVD) or BEACOPP,16-18 only small stud­ies have been com­pleted clu­sion, although it is likely that the par­a­digm of treat­ment of
incor­po­rat­ing these agents into front­line ther­apy for early-stage early-stage HL will likely shift to incor­po­rate BV and check­point
HL. Abramson et al19 eval­u­ated com­bined AVD/BV with­out radio­ inhib­i­tors front­line, which agents to use, how to incor­po­rate
ther­apy in 34 patients with nonbulky early-stage HL. Patients re- them (lead-in, sequen­tial, or con­cur­rent admin­is­tra­tion), which
ceived 1 cycle of BV alone on days 1 and 15 followed by 4 cycles reg­i­men (AVD or BEACOPP) to com­bine with, and whether con­
of com­ bined AVD/BV. The com­ plete response (CR) rate was ven­tional cyto­toxic ther­apy can ulti­mately be elim­in ­ ated are
52% after the lead-in cycle of BV and 97% after 2 AVD/BV cycles. still under inves­ti­ga­tion.
Three-year PFS was 94%. Grade 3 to 4 events included sen­sory
neu­rop­at­hy (23%), neutropenia (62%), and febrile neutropenia
(35%), although neutropenic fever declined once growth fac­tor
sup­port was man­dated. Park and col­leagues exam­ined con­sol­i­ CLINICAL CASE (Con­tin­ued)
da­tion of patients with nonbulky (defined as ≤7.5 cm) early-stage
HL with 6 cycles of BV after 2 to 6 cycles of ABVD.20 Of the 41 The 20-year-old woman with nonbulky stage IIA clas­sic HL with
patients enrolled, 36 com­pleted the planned 6 BV doses. With 2 adverse risk fac­tors (5 nodal sites and ele­vated ESR) started
a con­sol­i­da­tion approach, 95% achieved a CR, and 3-year PFS ABVD che­mo­ther­apy, and after 2 cycles, PET was performed,
was 92%. dem­on­strat­ing a right supraclavicular node of 2.1 × 1.5 cm with
Kumar et al21 conducted a pilot study of CMT using AVD/BV SUV 5.9 com­pared with 2.9 × 3 cm with SUV 7.3 pre­vi­ously, new
and 30 Gy ISRT in 30 patients with early unfa­vor­able HL. For- right axil­lary lymph node of 1.5 × 1.1 cm with SUV 7.5, and wors­
ty-seven per­cent of patients had bulky dis­ease. Twenty-seven en­ing ante­rior medi­as­ti­nal nodal mass with SUV 10.2 com­pared
patients achieved CR, and 1-year PFS was 93.3%. This trial with 6.3 pre­ vi­
ously (Deauville score 5). Core needle biopsy
recently enrolled 3 addi­tional cohorts test­ing the fea­si­bil­ity of spec­ i­
men of the supraclavicular node con­ firmed per­ sis­
tent
reduc­ing the ISRT dose to 20 Gy, reduc­ing the radi­a­tion field clas­sic HL. She stopped ABVD ther­apy and began 4 cycles of
with con­sol­i­da­tion vol­ume radi­a­tion, and elim­i­nat­ing ISRT.22 To com­bined BV/nivolumab sal­vage ther­apy. After 4 cycles, she
date, 27% of the 117 patients enrolled across all 4 cohorts had achieved a CR with a Deauville score of 3. She proceeded to
bulky dis­ease, and CR rates were 93% to 97% in the CMT cohorts autol­o­gous trans­plant followed by 30 Gy pro­ton ther­apy to
and 97% with AVD/BV alone. With a median fol­low-up of 3.8 bilat­eral neck, medi­as­ti­num, and axil­lary nodal sites. She ini­ti­
years, 2-year PFS was 94% in all­patients, with a 2-year PFS of ated BV main­te­nance after radi­a­tion and remains on treat­ment
96.6% in the che­mo­ther­apy alone arm, although fol­low-up in this for a planned max­i­mum of 16 cycles.
no radio­ther­apy cohort is still short at 2.2 years.
Two stud­ies have explored check­point inhib­it­ ors in early-stage
HL. The GHSG conducted a trial in 109 patients with unfa­vor­able Conflict-of-inter­est dis­clo­sure
or bulky HL exam­in­ing con­cur­rent nivolumab/AVD for 4 cycles or Kristie A. Blum: no com­pet­ing finan­cial inter­ests to declare.
sequen­tial ther­apy with 4 nivolumab cycles, 2 nivolumab/AVD
cycles, and 2 AVD cycles.23 All patients received 30 Gy ISRT. Off-label drug use
Eighty-seven per­cent and 26% of patients achieved a CR after 2 Kristie A. Blum: Brentuximab, nivolumab, and pembrolizumab are
nivolumab/AVD cycles or 4 nivolumab cycles, respec­tively. At the not FDA approved as frontline therapy in early-stage HL.
com­ple­tion of treat­ment, the CR rates were 83% and 84%, and
2-year PFS was 100% and 98% in the com­bi­na­tion and sequen­tial Correspondence
arms, respec­tively. Grade 3 to 4 toxicities were sim­il­ar with both Kristie A. Blum, Department of Hematology and Medical Oncolo-
approaches, and hypo­ thy­
roid­
ism was the most fre­ quent late gy, Emory Winship Cancer Institute, 1365 Clifton Road NE, B4013,
effect, persisting in 17% of patients. Allen and col­leagues com­ Atlanta, GA 30322; e-mail: kablum@emory​­.edu.

238  |  Hematology 2021  |  ASH Education Program


References 13. LaCasce AS, Dockter T, Ruppert AS, et  al. CALGB 50801 (Alliance): PET
1. Dores GM, Curtis RE, Dalal NH, et al. Cause-spe­cific mor­tal­ity fol­low­ing ini­ adapted ther­apy in bulky stage I/II clas­sic Hodgkin lym­phoma (cHL). J Clin
tial che­mo­ther­apy in a pop­u­la­tion-based cohort of patients with clas­si­cal Oncol. 2021;39(15, suppl):7507.
Hodgkin lym­phoma, 2000-2016. J Clin Oncol. 2020;38(35):4149-4162. 14. Gallamini A, Tarella C, Viviani S, et al. Early che­mo­ther­apy inten­si­fi­ca­tion
2. Giulino-Roth L, Pei Q , Buxton A, et al. Subsequent malig­nant neo­plasms with esca­lated BEACOPP in patients with advanced-stage Hodgkin lym­
among chil­dren with Hodgkin lym­phoma: a report from the Children’s phoma with a pos­i­tive interim pos­i­tron emis­sion tomog­ra­phy/com­puted
Oncology Group. Blood. 2021;137(11):1449-1456. tomog­ra­phy scan after two ABVD cycles: long-term results of the GITIL/
3. Schaapveld M, Aleman BM, van Eggermond AM, et al. Second can­cer risk FIL HD 0607 trial. J Clin Oncol. 2018;36(5):454-462.
up to 40 years after treat­ment for Hodgkin’s lym­phoma. N Engl J Med. 15. Johnson P, Federico M, Kirkwood A, et al. Adapted treat­ment guided by
2015;373(26):2499-2511. interim PET-CT scan in advanced Hodgkin’s lym­ phoma. N Engl J Med.
4. Bright CJ, Reulen RC, Winter DL, et  al. Risk of sub­se­quent pri­mary neo­ 2016;374(25):2419-2429.
plasms in sur­vi­vors of ado­les­cent and young adult can­cer (Teenage and 16. Eichenauer DA, Plütschow A, Kreissl S, et al. Incorporation of brentuximab
Young Adult Cancer Survivor Study): a pop­u­la­tion-based, cohort study. vedotin into first-line treat­ ment of advanced clas­ si­
cal Hodgkin’s lym­
Lancet Oncol. 2019;20(4):531-545. phoma: final anal­y­sis of a phase 2 randomised trial by the Ger­man Hodgkin
5. Zinzani PL, Rigacci L, Stefoni V, et  al. Early interim 18F-FDG PET in Hod- Study Group. Lancet Oncol. 2017;18(12):1680-1687.
gkin’s lym­phoma: eval­u­a­tion on 304 patients. Eur J Nucl Med Mol Imaging. 17. Straus DJ, Długosz-Danecka M, Alekseev S, et al. Brentuximab vedotin with
che­mo­ther­apy for stage III/IV clas­si­cal Hodgkin lym­phoma: 3-year update

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/234/1851889/234blum.pdf by guest on 13 December 2021


2012;39(1):4-12.
6. Radford J, Illidge T, Counsell N, et al. Results of a trial of PET-directed ther­ of the ECHELON-1 study. Blood. 2020;135(10):735-742.
apy for early-stage Hodgkin’s lym­phoma. N Engl J Med. 2015;372(17):1598- 18. Ramchandren R, Domingo-Domènech E, Rueda A, et  al. Nivolumab for
1607. newly diag­nosed advanced-stage clas­sic Hodgkin lym­phoma: safety and
7. Barrington SF, Phil­lips EH, Counsell N, et al. Positron emis­sion tomog­ra­phy effi­cacy in the phase II checkMate 205 Study. J Clin Oncol. 2019;37(23):1997-
score has greater prog­nos­tic sig­nif­i­cance than pre­treat­ment risk strat­i­fi­ca­ 2007.
tion in early-stage Hodgkin lym­phoma in the UK RAPID Study. J Clin Oncol. 19. Abramson JS, Arnason JE, LaCasce AS, et  al. Brentuximab vedotin, doxo­
2019;37(20):1732-1741. ru­bi­cin, vin­blas­tine, and dacarbazine for nonbulky lim­ited-stage clas­si­cal
8. Straus DJ, Jung S-H, Pitcher B, et al. CALGB 50604: risk-adapted treat­ment Hodgkin lym­phoma. Blood. 2019;134(7):606-613.
of nonbulky early-stage Hodgkin lym­phoma based on interim PET. Blood. 20. Park SI, Shea TC, Olajide O, et  al. ABVD followed by BV con­sol­i­da­tion in
2019;132(10):1013-1021. risk-strat­i­fied patients with lim­ited-stage Hodgkin lym­phoma. Blood Adv.
9. André MPE, Girinsky T, Federico M, et al. Early pos­i­tron emis­sion tomog­ 2020;4(11):2548-2555.
ra­phy response-adapted treat­ment in stage I and II Hodgkin lym­phoma: 21. Kumar A, Casulo C, Yahalom J, et al. Brentuximab vedotin and AVD followed
final results of the ran­dom­ized EORTC/LYSA/FIL H10 trial. J Clin Oncol. by involved-site radio­ther­apy in early stage, unfa­vor­able risk Hodgkin lym­
2017;35(16):1786-1794. phoma. Blood. 2016;128(11):1458-1464.
10. Fuchs M, Goergen H, Kobe C, et al. Positron emis­sion tomog­ra­phy-guided 22. Kumar A, Casulo C, Advani RH, et al. Brentuximab vedotin com­bined with
treat­ment in early-stage favor­able Hodgkin lym­phoma: final results of the che­mo­ther­apy in patients with newly diag­nosed early-stage, unfa­vor­able-
inter­na­
tional, ran­
dom­ ized phase III HD16 trial by the Ger­ man Hodgkin risk Hodgkin lym­phoma. J Clin Oncol. 2021;39(20):2257-2265.
23. Bröckelmann PJ, Goergen H, Keller U, et al. Efficacy of nivolumab and AVD in
Study Group. J Clin Oncol. 2019;37(31):2835-2845.
early-stage unfa­vor­able clas­sic Hodgkin lym­phoma: the ran­dom­ized phase
11. Borchmann P, Plütschow A, Kobe C, et al. PET-guided omis­sion of radio­ther­
2 Ger­man Hodgkin Study Group NIVAHL trial. JAMA Oncol. 2020;6(6):872.
apy in early-stage unfavourable Hodgkin lym­phoma (GHSG HD17): a multi-
24. Allen PB, Savas H, Evens AM, et  al. Pembrolizumab followed by AVD in
centre, open-label, randomised, phase 3 trial. Lancet Oncol. 2021;22(2):
untreated early unfa­vor­able and advanced-stage clas­si­cal Hodgkin lym­
223-234.
phoma. Blood. 2021;137(10):1318-1326.
12. Savage KJ, Connors JM, Villa DR, et al. Advanced stage clas­si­cal Hodgkin
lym­phoma patients with a neg­a­tive PET-scan fol­low­ing treat­ment with
ABVD have excel­lent out­comes with­out the need for consolidative radio­ © 2021 by The Amer­i­can Society of Hematology
ther­apy regard­less of dis­ease bulk at pre­sen­ta­tion. Blood. 2015;126(123):579. DOI 10.1182/hema­tol­ogy.2021000255

Prakash Singh Shekhawat


Management of early-stage HL  | 239
HODGKIN LYMPHOMA: CURE AND OPTIMAL SURVIVORSHIP

How to choose first salvage therapy


in Hodgkin lymphoma: traditional
chemotherapy vs novel agents

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/240/1851809/240driessen.pdf by guest on 13 December 2021


Julia Driessen,1 Sanne H. Tonino,1 Alison J. Moskowitz,2 and Marie José Kersten1
Department of Hematology, Amsterdam UMC, University of Amsterdam, LYMMCARE, Cancer Center Amsterdam, Amsterdam, The Netherlands;
1

and 2Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY

Approximately 10% to 30% of patients with classical Hodgkin lymphoma (cHL) develop relapsed or refractory (R/R)
disease. Of those patients, 50% to 60% show long-term progression-free survival after standard salvage chemotherapy
followed by high-dose chemotherapy (HDCT) and autologous stem cell transplant (ASCT). In the past decade, novel
therapies have been developed, such as the CD30-directed antibody–drug conjugate brentuximab vedotin and immune
checkpoint inhibitors, which have greatly extended the treatment possibilities for patients with R/R cHL. Several phase
1/2 clinical trials have shown promising results of these new drugs as monotherapy or in combination with chemother-
apy, but unfortunately, very few randomized phase 3 trials have been performed in this setting, making it difficult to give
evidence-based recommendations for optimal treatment sequencing. Two important goals for the improvement in the
treatment of R/R cHL can be identified: (1) increasing long-term progression-free and overall survival by optimizing risk-
adapted treatment and (2) decreasing toxicity in patients with a low risk of relapse of disease by evaluating the need for
HDCT/ASCT in these patients. In this review, we discuss treatment options for patients with R/R cHL in different settings:
patients with a first relapse, primary refractory disease, and in patients who are ineligible or unfit for ASCT. Results of
clinical trials investigating novel therapies or strategies published over the past 5 years are summarized.

LEARNING OBJECTIVES
• Describe current and emerging therapies for patients with R/R Hodgkin lymphoma
• Understand the importance for patients with R/R Hodgkin lymphoma to achieve a CMR before HDCT/ASCT

phamide, vincristine, procarbazine, and prednisone. After 2


CLINICAL CASE cycles, a CMR was reached and the patient received consol-
A 30-year-old woman presented with a persistent painless idative involved node radiotherapy (30 Gy).
lump in the neck without B-symptoms. A biopsy of a right Unfortunately, 1 year later, the patient presented with
supraclavicular node was performed, which showed a clas- night sweats and severe itching. Imaging revealed exten-
sical Hodgkin lymphoma (cHL). An 18F-fluorodeoxyglucose sive lymphadenopathy above and below the diaphragm,
positron emission tomography (PET)–computed tomogra- and a biopsy confrmed the relapse. Salvage chemother-
phy (CT) scan revealed lymphadenopathy bilaterally in the apy with dexamethasone, high-dose cytarabine, and cis-
supraclavicular and infraclavicular region, retrosternally, platin (DHAP) was initiated, which resulted in a CMR after
and in the mediastinum; hence, cHL stage IIA unfavorable 2 cycles, and stem cells were mobilized and collected
was diagnosed. after a third cycle of DHAP with the intention to proceed
After oocyte preservation, treatment with adriamycin, to high-dose chemotherapy (HDCT) followed by autolo-
bleomycin, vinblastine, and dacarbazine was initiated with gous stem cell transplant (ASCT) rescue.
the intention to administer a total of 6 cycles in case of a com-
plete metabolic response (CMR) after 2 cycles. However, the
interim PET-scan showed only a partial metabolic response Introduction
(PR) (Deauville score 4), and the treatment was intensifed Approximately 10% to 30% of patients with cHL will relapse or
to escalated bleomycin, etoposide, adriamycin, cyclophos- are primary refractory (R/R) to frst-line treatment. Standard

240 | Hematology 2021 | ASH Education Program


Table 1. Overview of first-sal­vage che­mo­ther­apy reg­i­mens since 2010

Refractory,
Study N Intervention n (%) CR pre-ASCT ORR pre-ASCT PFS OS
Josting et al 279 DHAP 0 (0) CT: 24% CT: 71% 3 years: 69% (no 3 years: 85% (no
(2010)13 (RCT) sig­nif­i­cant dif­fer­ence sig­nif­i­cant dif­fer­ence
between arms) between arms)
Moskowitz et al 105 ICE 48 (46) PET/gal­lium: 61% CT: 59% 4 years: 56% 4 years: 72%
(2010)14 CT: 33%
Moskowitz et al 97 ICE + GVD 41 (42) PET: 60% after ICE — 51 months: 70% 51 months: 80%
(2012)11 (PET-adapted 78% after GVD
sequen­tial)
Labrador et al 82 ESHAP 41 (50) PET/gal­lium: 50% PET/ gal­lium: 67% Median PFS: 56 months 5 years: 73%

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/240/1851809/240driessen.pdf by guest on 13 December 2021


(2014)15 (ret­
ro­spec­tive)
Santoro et al 58 BeGEV 27 (46) PET: 73% PET: 83% 5 years: 59% 5 years: 78%
(2016)16
BeGEV, bendamustine, gemcitabine, and vinorelbine.

sal­vage che­mo­ther­apy and ­con­sol­i­da­tion with HDCT/ASCT leads ESHAP) (Table 1). However, ran­dom­ized con­trolled tri­als (RCTs)
to long-term pro­gres­sion-free sur­vival (PFS) in about 50% to 60% com­par­ing dif­fer­ent sal­vage che­mo­ther­apy reg­i­mens or a PET-
of patients. Until recently, patients who relapsed after ASCT or adapted approach are lacking.
were inel­i­gi­ble for ASCT had lim­ited treat­ment options, and 50% of
those patients even­tu­ally died of the dis­ease.1 In the past decades, BV and check­point inhib­i­tors
sev­eral novel ther­a­peu­tic options for patients with R/R cHL have cHL is char­ ac­
ter­
ized by the pres­ ence of a minor­ity of bi- or
become avail­­able, includ­ing brentuximab vedotin (BV) and immune mul­ ti­
nu­cle­ated Hodgkin and Reed-Sternberg (HRS) cells that
check­point inhib­i­tors (CPIs), lead­ing to high CMR rates pre-ASCT, uni­ver­sally express CD30 in an inflam­ma­tory tumor micro­en­vi­
espe­cially when com­bined with che­mo­ther­apy.2 Achieving a CMR ron­ment. BV is an anti-CD30 mono­clo­nal anti­body con­ju­gated
prior to ASCT appears to be the most impor­tant prog­nos­tic fac­tor to the micro­tu­bule-disrupting agent monomethyl auristatin-E.17
for PFS.3-10 Therefore, a risk- and PET-adapted treat­ment approach PD-L1 and PD-L2 are upregulated by HRS cells in about 90%
could prob­a­bly lead to higher cure rates.11 On the other hand, the of patients and induce T-cell exhaus­tion, which con­trib­utes to
bur­den of late toxicities related to HDCT, such as sec­ond­ary malig­ immune escape of HRS cells.18 CPIs are mono­clo­nal antibodies
nan­cies and infer­til­ity, is con­sid­er­able, espe­cially as the dis­ease that block the inter­ac­tion between inhib­i­tory ligands such as PD-
typ­i­cally affects patients early in life. For this rea­son, decreas­ing L1 and PD-L2 on the tumor cells and PD-1 recep­tors on immune
tox­ic­ity is one of the main goals in the treat­ment of R/R cHL.12 effec­tor cells.
In this edu­ca­tional ses­sion, we dis­cuss the results of stud­ies Several stud­ies have inves­ti­gated the use of BV in com­bi­na­
that incor­po­rated novel ther­a­pies and response-adapted treat­ tion with che­mo­ther­apy as first sal­vage reg­i­men and showed
ment and how this could be implemented in stan­dard prac­tice high CMR rates prior to ASCT of up to 83%, with 2-year PFS
to improve out­comes for patients with R/R cHL. rates rang­ ing from 63% to 81% (Table 2).3-10 In 5 stud­ ies, BV
was com­bined with che­mo­ther­apy in 2 to 6 cycles followed by
Treatment for patients with a first relapse or pri­mary ASCT in patients with PR or CMR, whereas in 2 stud­ies, patients
refrac­tory dis­ease after first-line treat­ment were treated ini­tially with 4 to 6 admin­is­tra­tions of BV mono-
Conventional sal­vage che­mo­ther­apy results in pre-ASCT com­ therapy; patients with a CMR could pro­ceed directly to ASCT,
plete response (CR) rates of about 20% to 25% and over­ all whereas patients with a PR received addi­tional sal­vage che­mo­
response rates (ORRs) of 60% to 70%, based on eval­u­a­tion by ther­apy with­out BV.4,5 This PET-adapted approach is inter­est­ing
CT scan.13 More recent stud­ies reporting response rates based because approx­i­ma­tely 30% to 50% of patients could pro­ceed
on func­tional imag­ing using PET or gal­lium showed CMR rates of to ASCT after BV monotherapy only, thereby avoiding tox­ic­ity
50% to 60% after ifosfamide, carboplatin, and etoposide (ICE) from sal­vage che­mo­ther­apy in these patients. Moreover, a trial
or etoposide, meth­yl­pred­nis­o­lone, high-dose cytarabine, and inves­ti­gat­ing the com­bi­na­tion of BV and the CPI nivolumab as
cis­platin (ESHAP). Even higher CMR rates were reported for the pre-ASCT sal­vage reg­i­men showed that 67 of 91 patients could
bendamustine, gemcitabine, and vinorelbine reg­i­men (73%) and pro­ceed directly to ASCT after BV-nivolumab, with­out sal­vage
a sequen­tial ICE–gemcitabine, vinorelbine, and lipo­so­mal doxo­ che­mo­ther­apy. The study revealed low tox­ic­ity of this reg­i­men
ru­bi­cin (GVD) approach (78%) in which patients with no CR on com­pared with sal­vage che­mo­ther­apy.10
ICE received addi­ tional che­mo­ther­
apy with GVD before pro­ Thus far, in stud­ies that incor­po­rated a PET-adapted strat­egy,
ceed­ing to ASCT.11,14-16 PFS ranges between 50% and 60% with an PFS seems to be sim­i­lar for patients who proceeded to ASCT
over­all sur­vival (OS) of 70% to 80% at 5 years.11,13-16 Overall, there directly after hav­ing a CMR on BV, BV-nivolumab, or ICE alone as
seem to be no sig­nif­i­cant dif­fer­ences with regard to out­come for those patients who needed addi­tional sal­vage che­mo­ther­
between the most com­monly used reg­i­mens (ie, ICE/DHAP/ apy to achieve a CMR.4,5,10 This con­firms that the most i­mpor­tant
Prakash Singh Shekhawat
Salvage treatment in relapsed/refractory Hodgkin lymphoma  |  241
Table 2. Overview of first-sal­vage reg­i­mens containing BV or CPI

Refractory, CMR pre-ASCT,


Study N Intervention Schedule n (%) n (%) 2-year PFS 2-year OS
Moskowitz et al 65 BV + sequen­tial ICE BV 1.2 mg/kg d1, 8, 15 of 34 (52) 54 (83) 82% 97%
(2017)4 28-d cycles, 2 cycles.
ICE sal­vage in case of
Deauville >3.
Herrera et al 57 BV + sequen­tial ICE/GVD BV 1.8 mg/kg every 21 d, 4 35 (61) 37 (65) 67% 93%
(2018)5 cycles. Last 2 cycles BV
esca­la­tion to 2.4 mg/kg
in n = 8 patients with
PR/SD. Salvage che­mo­
ther­apy at dis­cre­tion of

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/240/1851809/240driessen.pdf by guest on 13 December 2021


treating phy­si­cian.
Cole et al 45 BV + gemcitabine BV 1.8 mg/kg on d1 and d8 29 (64) 28 (67) — 1 year: 95%
(2018)6 every 21 days, 4 cycles.
In com­bi­na­tion with
gemcitabine.
LaCasce et al 55 BV + bendamustine BV 1.8 mg/kg every 21 d, 28 (51) 39 (74) 63% 94%
(2018)9 2-6 cycles. In com­bi­na­
tion with bendamustine.
Post-ASCT BV
monotherapy main­te­
nance up to 16 cycles.
Garcia-Sanz 66 BV + ESHAP BV 1.8 mg/kg every 21 40 (61) 46 (70) 71% 90%
et al (2019)8 days, 4 cycles. In com­
bi­na­tion with 3 cycles of
ESHAP.
Broccoli et al 40 BV + bendamustine BV 1.8 mg/kg every 21 20 (50) 30 (79) 68% 97%
(2019)7 days, 4-6 cycles. In
com­bi­na­tion with
bendamustine.
Abuelgasim 28 BV + IGEV BV 1.8 mg/kg every 12 (43) includ­ing 70% 73.5% (100% for 87.1% (100% for
et al (2019)19 21 days, 2-4 cycles. In n = 14 with >1 patients with patients with
com­bi­na­tion with IGEV. line of ther­apy. first relapse) first relapse)
64% received BV con­sol­
i­da­tion after ASCT.
Kersten et al 67 BV + DHAP BV 1.8 mg/kg every 21 30 (45) 53 (82) 78% 96%
(2021)3 days, 3 cycles. In com­bi­
na­tion with DHAP.
Advani et al 91 BV + nivolumab BV 1.8 mg/kg and 38 (42) 61 (67) 78% 93%
(2021)10 nivolumab 3.0 mg/kg
every 21 days, 4 cycles.
Moskowitz et al 39 Pembrolizumab + GVD Pembrolizumab 200 mg 16 (41) 36 (95) 1 year: 100% 1 year: 100%
(2021)20 every 21 days, 4 cycles.
In com­bi­na­tion with
GVD.
d, day; IGEV, ifosfamide, gemcitabine, vinorelbine, and pred­nis­o­lone; SD, sta­ble dis­ease.

goal is to achieve a CMR before ASCT and that patients who is asso­ci­ated with imme­di­ate tox­ic­ity such as cytopenias and
do not respond ini­tially can poten­tially be res­cued with addi­ mucositis and long-term tox­ic­ity such as infer­til­ity and sec­ond­ary
tional sal­vage che­mo­ther­apy and pro­ceed to ASCT if they sub­ malig­nan­cies.12 Superiority for HDCT/ASCT over mini-carmustine,
se­quently reach a CMR. etoposide, cytarabine, and melphalan (BEAM) (ie, reduced-dose
Figure 1 pro­poses a flow­chart of PET-adapted treat­ment in BEAM with­out ASCT) in R/R cHL was shown in 2 ran­dom­ized
patients with R/R cHL. Because BV and CPI are not yet avail­­able con­trolled tri­als (RCTs) in 1993 and 2002.21,22 However, in these
or reim­bursed as first sal­vage treat­ment in many countries, sal­ tri­als, patients did not receive any sal­vage che­mo­ther­apy before
vage che­mo­ther­apy is often still the stan­dard approach. BEAM or mini-BEAM. In addi­tion, with the advent of effec­tive
drugs such as BV and CPIs, a sub­set of patients may be cured
High-dose che­mo­ther­apy and ASCT with sal­vage treat­ment alone.17
With increas­ing CMR rates pre-ASCT, one might ques­tion the need A recently published study using pembrolizumab and GVD
for con­sol­i­da­tion with HDCT/ASCT in all­patients. HDCT/ASCT che­ mo­ ther­
apy followed by HDCT/ASCT showed a very high

242  |  Hematology 2021  |  ASH Education Program


Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/240/1851809/240driessen.pdf by guest on 13 December 2021

Figure 1. Flowchart of treatment for R/R cHL. IGEV, ifosfamide, gemcitabine, vinorelbine, and prednisolone; SCT, stem cell transplant.

­ re-ASCT CMR rate of 95%, and with a median fol­low-up of 1 year,


p There is a high unmet need for RCTs in the pre-ASCT R/R
no pro­gres­sions have occurred.20 This study has now started a set­ting. Future stud­ies should focus on the opti­mal sequence of
sec­ond part in which patients with a CMR after 2 cycles of pem- using CPIs and BV in sal­vage treat­ment and con­sol­i­da­tion strat­e­
brolizumab-GVD con­tinue with 2 addi­tional cycles of pembroli- gies to induce high CMR rates while at the same time min­i­miz­ing
zumab-GVD followed by pembrolizumab con­sol­i­da­tion instead early and late tox­ic­ity. Eventually, an RCT should be performed
of HDCT/ASCT. Results of this rev­ o­
lu­
tion­
ary approach could to estab­lish the role of risk- and PET-adapted treat­ment in R/R
change the treat­ment of patients with R/R cHL sig­nif­i­cantly. cHL, includ­ing the role of HDCT/ASCT.
Prakash Singh Shekhawat
Salvage treatment in relapsed/refractory Hodgkin lymphoma  |  243
Patients with pri­mary refrac­tory dis­ease risk patients with CPI and/or BV vs reserv­ing these treat­ments
Primary refrac­tory dis­ease and a short inter­val between first-line for a sub­se­quent relapse.
treat­ment and relapse are impor­tant poor prog­nos­tic fac­tors
for response to sal­vage ther­apy and PFS.3,11,14 CMR rates to sal­ BV and CPI treat­ment for patients who relapse after ASCT
vage ther­apy for refrac­tory patients are usu­ally lower com­pared or are inel­i­gi­ble for ASCT
with relapsed patients: 73% vs 86% after DHAP, 64% vs 84% after Patients who relapse after ASCT or are inel­i­gi­ble for ASCT due to
BV-bendamustine, and 53% vs 77% after BV-nivolumab, respec­ che­mo­ther­apy-resis­tant dis­ease gen­er­ally have a poor prog­no­
tively.3,9,10 However, in patients who do achieve a CMR to sal­vage sis.1 In Table 3, we sum­ma­rize the most impor­tant recent stud­ies
ther­apy and pro­ceed to ASCT, post-ASCT PFS for pri­mary refrac­ in patients with R/R cHL who have pro­gres­sion after at least 1
tory and relapsed patients is sim­i­lar.3,11 A ret­ro­spec­tive anal­y­sis in line of sal­vage treat­ment. The first break­through in the post-ASCT
78 patients who progressed on one or more sal­vage reg­i­mens set­ting was the appli­ca­tion of monotherapy with BV in heavily
and who were treated with reg­i­mens containing CPI showed pretreated R/R patients, which showed an ORR of 75% and a
favor­able results, with 59% of patients achiev­ing a CMR and a CMR rate of 34% with a median PFS of 20.5 months in those with

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/240/1851809/240driessen.pdf by guest on 13 December 2021


post-ASCT PFS of 81% at 18 months.23 This sug­gests that treat­ a CMR. The PFS rate at 5 years, how­ever, was only 22% with an
ment with CPI may improve chemosensitivity of pre­vi­ously che- OS of 41%, high­light­ing the need for addi­tional treat­ment options
morefractory dis­ease. Interestingly, pre-ASCT CMR sta­tus was (Table 3).17
not sig­nif­i­cantly prog­nos­tic for post-ASCT PFS in this cohort, A study that investigated pembrolizumab monotherapy showed
suggesting a PR might be suf­fi­cient for pro­ceed­ing to ASCT after an ORR of 69% and a CMR rate of 22%, with a 2-year PFS of 31%
CPI in this patient pop­u­la­tion. and OS of 91%.2 Several dif­fer­ent CPIs and also com­bi­na­tions of
CPIs have been inves­ti­gated in R/R cHL.30,31,33,34 In a phase 1 trial, 64
Patients with a late relapse patients were ran­dom­ized between ipilimumab-BV, nivolumab-BV,
Although most relapses after first-line treat­ment occur within and tri­ple ther­apy with ipilimumab-nivolumab-BV. The trial showed
the first 2 years, a minor­ity of patients has a late relapse more dif­fer­ences in tox­ic­ity pro­file and effi­cacy between the 3 reg­i­mens,
than 5 years after first-line treat­ment.24 A large ret­ro­spec­tive with the highest per­cent­age of grade 3/4 adverse events in the
anal­y­sis showed that late relapses occur more fre­quently in trip­let and ipilimumab-BV group, whereas the highest ORR and
patients with early-stage favor­ able dis­
ease com­ pared with CMR rates were found in the trip­let and nivolumab-BV group.34
patients with unfa­vor­able or advanced-stage dis­ease. Whether In a recently published head-to-head com­par­i­son of mono-
these relapses rep­re­sent true relapses or a new sec­ond cHL therapy with pembrolizumab vs monotherapy with BV, pembroli-
man­i­fes­ta­tion, pos­si­bly due to genetic or envi­ron­men­tal risk zumab showed a sig­nif­i­cantly higher median PFS of 13.2 months
fac­tors in these patients, remains largely unknown. About half vs 8.3 months for BV.35 The inci­dence of adverse events was com­
of these patients were treated with ASCT, which was asso­ci­ pa­ra­ble between the 2 groups, with immune-medi­ated adverse
ated with favor­able PFS and OS com­pared with other sal­vage events in the pembrolizumab arm and neu­rop­a­thy in the BV arm.
ther­ap­ ies; how­ever, non-ASCT approaches, such as those using Importantly, in patients who received ear­lier treat­ment with
com­bi­na­tion che­mo­ther­apy or occa­sion­ally radi­a­tion alone, BV or CPI, response rates seem to be sim­i­lar to patients who
could be con­sid­ered depending on the patient’s ini­tial treat­ have not received BV or CPI before. A ret­ro­spec­tive study eval­
ment and under­ly­ing comorbidities.24 u­ated 18 patients with R/R cHL and 10 patients with R/R ana­
plas­tic large cell lym­phoma who received treat­ment with BV
Maintenance treat­ment after ASCT in 2 lines and showed CMR and ORR that are com­pa­ra­ble with
For patients with a high risk of relapse after ASCT, main­te­nance patients who received BV for the first time.37 Retreatment with
treat­ment with BV can be con­sid­ered. In a study investigating CPI in 78 patients with R/R cHL who relapsed after nivolumab
BV maintenance, 329 patients with unfavorable risk R/R cHL also showed com­pa­ra­ble effi­cacy.38
(defined as primary refractory disease, relapse <1 year, or extran-
odal disease) received either up to 16 cycles of BV maintenance Role of radio­ther­apy in the man­age­ment of R/R cHL
or placebo after ASCT.25 The study showed improve­ment in PFS The role of radio­ther­apy in the R/R set­ting has not been revis-
in patients receiv­ing BV main­te­nance, with a 5-year PFS of 59% ited well in this era of novel treat­ment options. Radiotherapy
vs 41% for pla­cebo. However, there was no dif­fer­ence in OS, can be used pre-ASCT or post-ASCT on resid­ual lesions or in
prob­a­bly because 87% of patients who relapsed in the pla­cebo patients with extranodal or bulky dis­ease and as part of the
arm received BV at the sub­se­quent relapse. Therefore, the use con­di­tion­ing reg­i­men using total lym­phoid irra­di­a­tion, but
of BV main­te­nance after ASCT could poten­tially be restricted to com­par­a­tive data about effi­cacy of radio­ther­apy in these set­
patients with at least 2 risk fac­tors, or alter­na­tively, its use could tings are scarce and out­dated.39 Earlier stud­ies have shown
be delayed until progression. With the increas­ing use of BV in that patients who receive radio­ther­apy have a decreased risk
the first-line set­ting, it is also impor­tant to inves­ti­gate whether of local recur­rence, and thus for patients with lim­ited-stage
patients who relapse after BV in com­bi­na­tion with che­mo­ther­apy dis­ease at relapse, radio­ther­apy may be an effec­tive option.39
will still show advan­tage of BV maintenance after ASCT.26 Alter- Using radio­ther­apy in patients who have a PR pre-ASCT would
natively, CPI could be used as main­te­nance treat­ment; a small be an inter­est­ing strat­egy to increase the CMR rate, and stud­ies
phase 2 trial in 30 high-risk patients showed high post-ASCT inves­ti­gat­ing this approach are warranted. In addi­tion, the syn­
PFS.27 The com­bi­na­tion of CPI and BV main­te­nance in 59 high- er­gis­tic effects of radi­a­tion with immu­no­ther­apy, as described
risk patients has also shown prom­is­ing results, with 5 patients in a few case reports, should be inves­ti­gated more exten­sively
with PR converting to CR dur­ing main­te­nance.28 Further stud­ies and could be an option for patients who relapse after ASCT or
should inves­ti­gate the role of post-ASCT main­te­nance in high- are inel­i­gi­ble for ASCT.

244  |  Hematology 2021  |  ASH Education Program


Table 3. Overview of recently reported trial results incor­po­rat­ing BV or CPIs for patients after ≥1 line of sal­vage treat­ment

Study N Intervention CMR ORR PFS OS


Chen et al (2016)17 102 BV 34% 75% 5 years: 22% 5 years: 41%
O’Connor et al (2018) 29
65 BV + bendamustine 32% 71% 2 years: 50%* 2 years: 65%*
Armand et al (2018)18 243 Nivolumab 16% 69% 1 year: 50%* 1 year: 92%
Chen et al (2019)2 210 Pembrolizumab 27.6% 71.9% 2 years: 31.3% 2 years: 90.9%
Shi et al (2019)30 92 Sintilimab 34% 80.4% 6 month: 77.6% 6 month: 100%
Song et al (2019) 31
75 Camrelizumab 28% 76% 9 month: 76.6% 9 month: 96%*
Armand et al (2020)32 31 Pembrolizumab 19% 58% 2 years: 30% 2 years: 87%
Song et al (2020) 33
70 Tislelizumab 63.9% 87.1% 9 month: 74.5% 9 month: 98.6%

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/240/1851809/240driessen.pdf by guest on 13 December 2021


Diefenbach et al (2020)34 64 Ipilimumab + BV vs 60%/65%/84% 80%/94%/95% 1 year: 59%/77%/87% 1 year:
(includ­ing n = 26 first nivolumab + BV 90%*/80%*/95%*/
relapse) vs ipilimumab +
nivolumab + BV
Kuruvilla et al (2021)35 304 Pembrolizumab (n = 151) 25% vs 24% 66% vs 54% 2 years: 35% vs 25% —
vs BV (n = 152)
Liu et al (2021)36 61 Camrelizumab 79% vs 32% for 95% vs 89% 2 years: 67% vs 42% 2 years: 100%
monotherapy vs camrelizumab +
camrelizumab + decitabine vs
decitabine camrelizumab
*Data have been extracted from avail­­able Kaplan-Meier curves using WebPlot Digitizer.

General con­sid­er­ations Conflict-of-inter­est dis­clo­sure


In con­clu­sion, the pri­mary goal of treat­ment for patients with Julia Driessen: travel sup­port from Takeda.
R/R cHL is to achieve a CMR before HDCT/ASCT as this sig­nif­ Sanne H. Tonino: no rel­e­vant dis­clo­sures.
i­cantly cor­re­lates with a favor­able out­come after ASCT. Using Alison J. Moskowitz: hon­o­raria: Seattle Genetics; con­sul­ting or advi­
a sequen­tial approach, treat­ment inten­sity and tox­ic­ity can be sory role: Seattle Genetics, Takeda, Imbrium Therapeutics, Merck,
reduced in a sub­set of fast-responding patients. Patients who are Janpix, Kyowa Kirin International, miRagen, ADC Therapeutics, Bris-
inel­i­gi­ble for ASCT or relapse after ASCT can be treated with BV tol Myers Squibb; research funding: Incyte, Seattle Genetics, Merck,
or CPIs. There is a need to develop novel ther­a­pies to increase Bristol Myers Squibb, miRagen, ADC Therapeutics, BeiGene.
response rates with­out increas­ing tox­ic­ity. One of the next goals Marie José Kersten: hon­o­raria for con­sul­ting or advi­sory boards:
for clin­i­cal tri­als is to inves­ti­gate which patients can pos­si­bly Kite/Gilead, Novartis, BMS/Celgene, Miltenyi Biotech, Research
be cured with­out HDCT/ASCT. Risk-strat­i­fied and PET-adapted funding Takeda, Roche, Celgene, Kite/Gilead.
pro­spec­tive stud­ies could help achieve this goal. Optimized risk
strat­i­fi­ca­tion and response eval­u­a­tion will guide future treat­ment Off-label drug use
deci­sions and will help to find the right treat­ment for the right Julia Driessen: nothing to disclose.
patient. Sanne H. Tonino: nothing to disclose.
Alison J. Moskowitz: nothing to disclose.
Marie José Kersten: nothing to disclose.

CLINICAL CASE (Con­t in­u ed) Correspondence


Despite the ini­tial CMR after 2 cycles of DHAP, soon after the Marie José Kersten, Amsterdam UMC, University of Amsterdam,
third and last cycle of DHAP, B-symp­toms and itching returned Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; e-mail:
and a relapse was again con­firmed by PET-CT and a biopsy. m​­. j​­.kersten@amsterdamumc​­.nl.
Given the poor prog­no­sis in this patient with chemorefrac-
tory dis­ease, com­bi­na­tion treat­ment with BV and nivolumab was References
started based on the encour­ag­ing results of a phase 1/2 trial.10 1. von Tresckow B, Müller H, Eichenauer DA, et al. Outcome and risk fac­
Already after 1 cycle of BV-nivolumab, her B-symp­toms disap- tors of patients with Hodgkin lym­phoma who relapse or prog­ress after
autol­o­gous stem cell trans­plant. Leuk Lymphoma. 2014;55(8):1922-
peared, and after 4 cycles, she reached a CMR. We decided to 1924.
pro­ceed to haploidentical ASCT, because the patient ini­ tially 2. Chen R, Zinzani PL, Lee HJ, et  al. Pembrolizumab in relapsed or refrac­
progressed dur­ing sal­vage che­mo­ther­apy. As men­tioned above, tory Hodgkin lym­phoma: 2-year fol­low-up of KEYNOTE-087. Blood. 2019;
though, emerg­ing data on the role of ASCT after CPI for patients 134(14):1144-1153.
3. Kersten MJ, Driessen J, Zijlstra JM, et al. Combining brentuximab vedotin with
refrac­tory to mul­ti­ple prior lines of che­mo­ther­apy sug­gest that
dexa­meth­a­sone, high-dose cytarabine and cis­platin as sal­vage treat­ment
ASCT could be con­sid­ered in this set­ting as well.23 in relapsed or refrac­tory Hodgkin lym­phoma: the phase II ­HOVON/LLPC
trans­plant BRaVE study. Haematologica. 2021;106(4):1129-1137.
Prakash Singh Shekhawat
Salvage treatment in relapsed/refractory Hodgkin lymphoma  |  245
4. Moskowitz AJ, Schöder H, Gavane S, et al. Prognostic sig­nif­i­cance of base­ dis­ ease: results of a BNLI randomised trial. Lancet. 1993;341(8852):
line met­ ab
­ olic tumor vol­ume in relapsed and refrac­ tory Hodgkin lym­ 1051-1054.
phoma. Blood. 2017;130(20):2196-2203. 23. Merryman RW, Redd RA, Nishihori T, et al. Autologous stem cell trans­plan­
5. Herrera AF, Palmer J, Martin P, et al. Autologous stem-cell trans­plan­ta­tion ta­tion after anti-PD-1 ther­apy for mul­ti­ply relapsed or refrac­tory Hodgkin
after sec­ond-line brentuximab vedotin in relapsed or refrac­tory Hodgkin lym­phoma. Blood Adv. 2021;5(6):1648-1659.
lym­phoma. Ann Oncol. 2018;29(3):724-730. 24. Bröckelmann PJ, Goergen H, Kohnhorst C, et al. Late relapse of clas­si­cal
6. Cole PD, McCarten KM, Pei Q , et  al. Brentuximab vedotin with gemcit- Hodgkin lym­phoma: an anal­y­sis of the Ger­man Hodgkin Study Group HD7
abine for pae­di­at­ric and young adult patients with relapsed or refrac­tory to HD12 tri­als. J Clin Oncol. 2017;35(13):1444-1450.
Hodgkin’s lym­phoma (AHOD1221): a Children’s Oncology Group, multi- 25. Moskowitz CH, Nademanee A, Masszi T, et al; AETHERA Study Group.
centre sin­gle-arm, phase 1-2 trial. Lancet Oncol. 2018;19(9):1229-1238. Brentuximab vedotin as con­sol­i­da­tion ther­apy after autol­o­gous stem-cell
7. Broccoli A, Argnani L, Botto B, et al; Fondazione Italiana Linfomi ONLUS. trans­plan­ta­tion in patients with Hodgkin’s lym­phoma at risk of relapse or
First sal­vage treat­ment with bendamustine and brentuximab vedotin in pro­gres­sion (AETHERA): a randomised, dou­ble-blind, pla­cebo-con­trolled,
Hodgkin lym­phoma: a phase 2 study of the Fondazione Italiana Linfomi. phase 3 trial. Lancet. 2015;385(9980):1853-1862.
Blood Cancer J. 2019;9(12):100. 26. Connors JM, Jurczak W, Straus DJ, et al; ECHELON-1 Study Group. Brentux-
8. Garcia-Sanz R, Sureda A, de la Cruz F, et  al. Brentuximab vedotin and imab vedotin with che­mo­ther­apy for stage III or IV Hodgkin’s lym­phoma.
ESHAP is highly effec­tive as sec­ond-line ther­apy for Hodgkin lym­phoma N Engl J Med. 2018;378(4):331-344.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/240/1851809/240driessen.pdf by guest on 13 December 2021


patients (long-term results of a trial by the Span­ish GELTAMO Group). Ann 27. Armand P, Chen Y-B, Redd R-A, et al. PD-1 block­ade with pembrolizumab
Oncol. 2019;30(4):612-620. for clas­si­cal Hodgkin lym­phoma after autol­o­gous stem cell trans­plan­ta­tion.
9. LaCasce AS, Bociek RG, Sawas A, et al. Brentuximab vedotin plus benda- Blood. 2019;134(1):22-29.
mustine: a highly active first sal­vage reg­i­men for relapsed or refrac­tory 28. Herrera AF, Chen L, Nieto Y, et al. Consolidation with nivolumab and bren-
Hodgkin lym­phoma. Blood. 2018;132(1):40-48. tuximab vedotin after autol­o­gous hema­to­poi­etic cell trans­plan­ta­tion in
10. Advani R, Moskowitz AJ, Bartlett NL, et al. Brentuximab vedotin in com­bi­ patients with high-risk Hodgkin lym­phoma. Blood. 2020;136(suppl 1):19-20.
na­tion with nivolumab in relapsed or refrac­tory Hodgkin lym­phoma: 3-year 29. O’Connor OA, Lue JK, Sawas A, et  al. Brentuximab vedotin plus benda-
study results. Blood. 2021;138(6):427-438. mustine in relapsed or refrac­tory Hodgkin’s lym­phoma: an inter­na­tional,
11. Moskowitz CH, Matasar MJ, Zelenetz AD, et al. Normalization of pre-ASCT, multicentre, sin­gle-arm, phase 1-2 trial. Lancet Oncol. 2018;19(2):257-266.
FDG-PET imag­ing with sec­ond-line, non-cross-resis­tant, che­mo­ther­apy 30. Shi Y, Su H, Song Y, et al. Safety and activ­ity of sintilimab in patients with
pro­ grams improves event-free sur­ vival in patients with Hodgkin lym­ relapsed or refrac­tory clas­si­cal Hodgkin lym­phoma (ORIENT-1): a multi-
phoma. Blood. 2012;119(7):1665-1670. centre, sin­gle-arm, phase 2 trial. Lancet Haematol. 2019;6(1):e12-e19.
12. Myers RM, Hill BT, Shaw BE, et al. Long-term out­comes among 2-year sur­ 31. Song Y, Wu J, Chen X, et  al. A sin­gle-arm, mul­ti­cen­ter, phase II study
vi­vors of autol­o­gous hema­to­poi­etic cell trans­plan­ta­tion for Hodgkin and of camrelizumab in relapsed or refrac­tory clas­si­cal Hodgkin lym­phoma.
dif­fuse large b-cell lym­phoma. Cancer. 2018;124(4):816-825. Clin Cancer Res. 2019;25(24):7363-7369.
13. Josting A, Müller H, Borchmann P, et  al. Dose inten­sity of che­mo­ther­apy 32. Armand P, Kuruvilla J, Michot J-M, et al. KEYNOTE-013 4-year fol­low-up of
in patients with relapsed Hodgkin’s lym­phoma. J Clin Oncol. 2010;28(34): pembrolizumab in clas­si­cal Hodgkin lym­phoma after brentuximab vedotin
5074-5080. fail­ure. Blood Adv. 2020;4(12):2617-2622.
14. Moskowitz CH, Yahalom J, Zelenetz AD, et al. High-dose chemo-radio­ther­ 33. Song Y, Gao Q , Zhang H, et al. Treatment of relapsed or refrac­tory clas­si­cal
apy for relapsed or refrac­tory Hodgkin lym­phoma and the sig­nif­i­cance of Hodgkin lym­phoma with the anti-PD-1, tislelizumab: results of a phase 2,
pre-trans­plant func­tional imag­ing. Br J Haematol. 2010;148(6):890-897. sin­gle-arm, mul­ti­cen­ter study. Leukemia. 2020;34(2):533-542.
15. Labrador J, Cabrero-Calvo M, Pérez-López E, et  al. ESHAP as sal­ vage 34. Diefenbach CS, Hong F, Ambinder RF, et  al. Ipilimumab, nivolumab, and
ther­apy for relapsed or refrac­tory Hodgkin’s lym­phoma. Ann Hematol. brentuximab vedotin com­bi­na­tion ther­a­pies in patients with relapsed or
2014;93(10):1745-1753. refrac­tory Hodgkin lym­ phoma: phase 1 results of an open-label, multi-
16. Santoro A, Mazza R, Pulsoni A, et al. Five-year results of the BEGEV sal­vage centre, phase ½ trial. Lancet Haematol. 2020;7(9):e660-e670.
reg­i­men in relapsed/refrac­tory clas­si­cal Hodgkin lym­phoma. Blood Adv. 35. Kuruvilla J, Ramchandren R, Santoro A, et al; KEYNOTE-204 inves­ ti­
ga­
2020;4(1):136-140. tors. Pembrolizumab ver­sus brentuximab vedotin in relapsed or refrac­
17. Chen R, Gopal AK, Smith SE, et al. Five-year sur­vival and dura­bil­ity results tory clas­si­cal Hodgkin lym­phoma (KEYNOTE-204): an interim anal­y­sis of a
of brentuximab vedotin in patients with relapsed or refrac­tory Hodgkin multicentre, randomised, open-label, phase 3 study. Lancet Oncol. 2021;
lym­phoma. Blood. 2016;128(12):1562-1566. 22(4):512-524.
18. Armand P, Engert A, Younes A, et  al. Nivolumab for relapsed/refrac­tory 36. Liu Y, Wang C, Li X, et al. Improved clin­ic
­ al out­come in a ran­dom­ized phase II
clas­sic Hodgkin lym­phoma after fail­ure of autol­o­gous hema­to­poi­etic cell study of anti-PD-1 camrelizumab plus decitabine in relapsed/refrac­tory Hod-
trans­plan­ta­tion: extended fol­low-up of the multicohort sin­gle-arm phase II gkin lym­phoma. J Immunother Cancer. 2021;9(4):e002347.
checkmate 205 trial. J Clin Oncol. 2018;36(14):1428-1439. 37. Fukuhara N, Yamamoto G, Tsujimura H, et al. Retreatment with brentuximab
19. Abuelgasim KA, Alzahrani M, Alsharhan Y, et al. Chemoimmunotherapy with vedotin in patients with relapsed/refrac­tory clas­si­cal Hodgkin lym­phoma
brentuximab vedotin com­bined with ifosfamide, gemcitabine, and vinorel- or sys­temic ana­plas­tic large-cell lym­phoma: a mul­ti­cen­ter ret­ro­spec­tive
bine is highly active in relapsed or refrac­tory clas­si­cal Hodgkin lym­phoma. study. Leuk Lymphoma. 2020;61(1):176-180.
Bone Marrow Transplant. 2019;54(7):1168-1172. 38. Manson G, Brice P, Herbaux C, et  al. Efficacy of anti-PD1 re-treat­ment in
20. Moskowitz AJ, Shah G, Schöder H, et  al. Phase II trial of pembrolizumab patients with Hodgkin lym­phoma who relapsed after anti-PD1 dis­con­tin­
plus gemcitabine, vinorelbine, and lipo­so­mal doxo­ru­bi­cin as sec­ond-line u­a­tion. Haematologica. 2020;105(11):2664-2666.
ther­apy for relapsed or refrac­tory clas­si­cal Hodgkin lym­phoma [published 39. Constine LS, Yahalom J, Ng AK, et  al. The role of radi­ a­tion ther­
apy in
online 25 June 2021]. J Clin Oncol. patients with relapsed or refrac­tory Hodgkin lym­phoma: guide­lines from
21. Schmitz N, Pfistner B, Sextro M, et al; Ger­man Hodgkin’s Lymphoma Study the inter­na­tional lym­phoma radi­a­tion oncol­ogy group. Int J Radiat Oncol
Group; Lymphoma Working Party of the Euro­pean Group for Blood and Biol Phys. 2018;100(5):1100-1118.
Marrow Transplantation. Aggressive con­ven­tional che­mo­ther­apy com­
pared with high-dose che­mo­ther­apy with autol­o­gous haemopoietic stem-
cell trans­plan­ta­tion for relapsed chemosensitive Hodgkin’s dis­ease: a
randomised trial. Lancet. 2002;359(9323):2065-2071.
22. Linch DC, Winfield D, Goldstone AH, et al. Dose inten­si­fi­ca­tion with autol­ © 2021 by The Amer­i­can Society of Hematology
o­gous bone-mar­row trans­plan­ta­tion in relapsed and resis­tant Hodgkin’s DOI 10.1182/hema­tol­ogy.2021000311

246  |  Hematology 2021  |  ASH Education Program


HODGKIN LYMPHOMA: CURE AND OPTIMAL SURVIVORSHIP

Double-refractory Hodgkin lymphoma:


tackling relapse after brentuximab vedotin
and checkpoint inhibitors

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/247/1851712/247epperla.pdf by guest on 13 December 2021


Narendranath Epperla1 and Mehdi Hamadani2
Division of Hematology, Department of Medicine, The James Cancer Hospital and Solove Research Institute, Ohio State University, Columbus, OH;
1

and 2Blood and Marrow Transplant Program and Cellular Therapy Program, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI

The approval of brentuximab vedotin (BV) and checkpoint inhibitors (CPI) has revolutionized the management of relapsed/
refractory classical Hodgkin lymphoma (cHL) patients. In recent years these agents have rapidly moved to earlier lines
of therapy, including post-autologous hematopoietic cell transplant (auto-HCT) consolidation, pre-HCT salvage, and the
frontline treatment setting. This shift in practice means that double-refractory (refractory to both BV and CPI) cHL is
becoming an increasingly common clinical problem. In patients who are not eligible for clinical trials, conventional cyto-
toxic and targeted therapies (off label) may be a potential option. In patients who are transplant eligible, early referral to
allogeneic HCT should be considered given the significant improvement in transplant outcomes in the contemporary era.
Cellular therapy options including CD30.chimeric antigen receptor T cells, Epstein-Barr virus-directed cytotoxic T cells,
and CD16A/30 bispecific natural killer cell engagers appear promising and are currently in clinical trials.

LEARNING OBJECTIVES
• Identify promising therapeutic agents for cHL refractory to both BV and checkpoint inhibitors
• Highlight contemporary outcomes of allogeneic transplantation in patients with heavily pretreated cHL
• Recognize emerging cellular therapies and summarize the results of clinical trials evaluating these therapies

CLINICAL CASE Introduction


A 32-year-old Caucasian man was diagnosed with stage IV-B Although most patients with cHL are cured with initial che-
classical Hodgkin lymphoma (cHL). He achieved a complete motherapy, up to 20% to 25% of patients will relapse or
metabolic remission following six cycles of frst-line treat- have primary refractory (R/R) disease.1 While high-dose
ment with adriamycin, vinblastine, bleomycin, and dacar- therapy and an auto-HCT consolidation can cure cHL
bazine. The patient experienced a biopsy-proven relapse patients responding to subsequent treatments, only about
9 months after fnishing this chemotherapy. At the time of half of these patients will attain durable disease control.2,3
relapse, the patient had extranodal involvement in the axial The advent of novel agents, especially BV and checkpoint
skeleton and liver. His disease remained refractory to second- inhibitors (CPI), in the past decade has not only revolution-
line treatment with ifosfamide, carboplatin, and etoposide. ized the management of R/R cHL patients; in recent years
The patient achieved a second complete metabolic remis- these agents have rapidly moved to earlier lines of therapy,
sion with a brentuximab vedotin (BV) and nivolumab com- including post-auto-HCT consolidation (Figure 1 depicts
bination and subsequently went on to receive autologous the increased utilization of BV consolidation post-auto-HCT
hematopoietic cell transplantation (auto-HCT) followedby in the United States based on reporting to the Center for
BV consolidation. While on consolidation treatment, the International Blood and Marrow Transplant Research [CIB-
patient relapsed again and was initiated on pembrolizumab, MTR] registry),3 pre-HCT salvage,4 and front-line treatment
which resulted in a partial remission lasting approximately setting.5 This shift in practice means that the scenario de-
18 months. At the current progression, the patient is ft, with scribed in the clinical case (above) of a cHL patient with
excellent organ function, and his donor search has identifed double-refractory (refractory to both BV and CPI) disease
a haploidentical sibling. is becoming a common clinical problem. Unlike patients
Prakash Singh Shekhawat
with relapsed non-Hodgkin lymphoma (NHL; median age in

Double-refractory Hodgkin lymphoma | 247


No maintenance Brentuximab maintenance Other maintenance

2020 256 125 33

2019 390 235 95


Year of Transplant

2018 353 316 74

2017 309 305 102

2016 388 233 64

2015 596 106 38

2014 665 20 35

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/247/1851712/247epperla.pdf by guest on 13 December 2021


2013 677 5 34

2012 706 713

0 100 200 300 400 500 600 700 800


Frequency
Figure 1. Increased use of BV as post-autologous transplant consolidation therapy in the US. Red arrow: year of AETHERA trial
publication; 2020 data incomplete. Based on CIBMTR registry data.

the late 60s), the median age of relapsed cHL patients is typ­i­cally sion of sal­vage reg­i­mens is beyond the scope of this review, but
in the 30s.6,7 The young age of these R/R patients means that in Table 1 sum­ma­rizes the com­monly con­sid­ered sal­vage options in
addi­tion to selecting the next sal­vage option (typ­i­cally expected dou­ble-refrac­tory cHL patients.8-17
to pro­vide short-term dis­ease con­trol), the treating team must
also con­sider ther­a­peu­tic modal­i­ties that can pro­vide dura­ble Radiation ther­apy
dis­ease con­trol and hope­fully cure. The lat­ter may not be fea­si­ In relapsed patients with lim­ited treat­ment options, sal­vage radi­
ble for the sub­set of dou­ble-refrac­tory patients with advanced a­tion should be con­sid­ered. Limited ret­ro­spec­tive data show
age, com­pro­mised organ func­tion, and/or poor per­for­mance effec­tive short-term dis­ease con­trol with radi­a­tion ther­apy,
sta­tus. par­tic­u­larly in patients with local­ized nodal dis­ease.18 Although
In this arti­cle we dis­cuss the treat­ment options for dou­ble- pre­clin­i­cal mod­els and case reports dem­on­strate syn­er­gism be-
refrac­tory cHL, begin­ning with poten­tial sal­vage options and tween radi­at­ion and CPIs,19-21 it is unclear if this is a fea­si­ble ap-
followed by opti­mal con­sol­i­da­tion approaches. At the out­set we proach in dou­ble-refrac­tory cHL patients.
acknowl­edge that there are no uni­ver­sally accepted def­i­ni­tions
of BV or CPI refrac­tory dis­ease. In this review, dou­ble-refrac­tory Investigational agents for dou­ble-refrac­tory cHL
dis­ease is loosely defined as cHL that does not achieve at least Camidanlumab tesirine
a par­tial remis­sion or progresses while on BV and CPI (-based Camidanlumab tesirine (cami) is an anti­ body-drug con­ ju­
gate
treat­ments). It is impor­tant to remem­ber that cHL patients pre­ (pyrrolobenzodiazepine) directed against CD25. After impres­
vi­ously achiev­ing remis­sion with either BV or CPI (or reg­i­mens sive anti­tu­mor activ­ity was noted in a phase 1, dose-esca­la­tion,
containing these drugs) and sub­se­quently relaps­ing after a treat­ and dose-expan­sion study of cami in R/R cHL patients (Table
ment-free inter­val may be can­di­dates for rechallenge with these 2),22 a phase 2 study was conducted using 45 µg/kg q3 week
agents. dos­ing for two cycles followed by 30 µg/kg Q3 weeks (in cHL
with prior BV and CPI treat­ments). Cami was active in R/R cHL
Salvage options in dou­ble-refrac­tory cHL patients based on the pre­lim­i­nary results (n  =  51), with an over­all
Conventional cyto­toxic ther­apy options response rate (ORR) of 83% and a com­plete response (CR) rate
Clinical trial enroll­ment, if avail­­able, should be a pri­or­ity in dou­ble- of 38%.23 Grade 3 or higher adverse events were reported in 63%
refrac­tory cHL. Off trial, in a fit patient con­ven­tional multiagent (n  =  32) of cHL patients.23 Of note, there were 3 (6.4%) patients
che­mo­ther­apy as a bridge to allo­ge­neic (allo) HCT is a rea­son­ with Guillain-Barré syn­drome (GBS)/polyradiculopathy (n  =  1 with
able option. In patients who are not can­di­dates for allo-HCT, grade 4 GBS, n  =  1 with grade 2 GBS, and n  =  1 with grade 2 radic-
sin­
gle-agent cyto­ toxic ther­ apy with a pal­ li­
a­tive intent is rea­ ulopathy) that led to an enroll­ment pause. Following a review of
son­able. The choice of che­mo­ther­apy (reg­i­men) is often empir- safety and effi­cacy data (by inde­pen­dent review), the enroll­ment
ic and depends on the patient’s prior treat­ments, mar­row re- pause was lifted. The study recently com­pleted accrual, and final
serve, and organ func­tion. For instance, while both plat­i­num- or results are not yet avail­­able, but the pre­lim­i­nary results with cami
gemcitabine-based ther­a­pies are active, plat­i­num-containing are encour­ag­ing and may pro­vide an addi­tional ther­a­peu­tic op-
reg­i­mens may be less desir­able in patients with chronic kid­ney tion for patients with R/R cHL, par­tic­u­larly those with dual-refrac­
dis­ease, and a gemcitabine-based option could be chal­leng­ tory dis­ease. Strategies to mit­i­gate the auto­im­mune neu­ro­log­i­cal
ing in patients with a poor mar­row reserve. A detailed dis­cus­ tox­ic­ity of this agent will enhance its risk/ben­e­fit pro­file.

248  |  Hematology 2021  |  ASH Education Program


Table 1.  Conventional cyto­toxic ther­apy options in R/R cHL

Study Median age Median


Agent design (range), years Total, N Prior AHCT (n) ORR% (CR%) PFS/EFS (mo) Reference
Gemcitabine based
Gemcitabine II 35 (19-58) 23 0 39 (9) 6.7 Santoro et al8
GDP II 36 (19–57) 23 0 69 (17) NR Baetz et al9
GVD I/II 33 (19–83) 91 40 70 (19) 8.5* Bartlett et al10
IGEV II 30 (17-59) 91 NA 81 (54) 3-y FFP 53% Santoro et al11
Platinum based
ICE II 27 (12-59) 65 NA 88 (26) 58%† Moskowitz et al12

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/247/1851712/247epperla.pdf by guest on 13 December 2021


DHAP II 34 (21-64) 102 NA 89 (21) NR Josting et al13
ESHAP II 34 (18-66) 22 2 73 (40) 3-y DFS 27% Aparicio et al14
Vinca alka­loids
Vinblastine II 31 (23-48) 17 17 59 (12) 8.3 Little et al15
Vinorelbine II NR 24 4 50 (14) 6 Devizzi et al16
Alkylators
Bendamustine II 34 (21–75) 35 27 53 (33) 5.2 Moskowitz et al17
*Median EFS was not reached in the trans­plant-naive group, while it was 8.5 months in patients with prior trans­plant.
†At a median fol­low-up of 43 months, as ana­lyzed by intent to treat, the EFS was 58%.
ASCT, autol­o­gous hema­to­poi­etic cell trans­plan­ta­tion; DFS, dis­ease-free sur­vival; DHAP, dexa­meth­a­sone, high-dose cytarabine, cis­platin; EFS,
event-free sur­vival; ESHAP, etoposide, meth­yl­pred­nis­o­lone, high-dose cytarabine, cis­platin; FFP, free­dom from pro­gres­sion; GDP, gemcitabine,
dexa­meth­a­sone, cis­platin; GVD, gemcitabine, vinorelbine, lipo­so­mal doxo­ru­bi­cin; ICE, ifosfamide, carboplatin, etoposide; IGEV, ifosfamide,
gemcitabine, etoposide, vinorelbine; NA, not appli­ca­ble; NR, not reported; PFS, pro­gres­sion-free sur­vival.

Mammalian tar­get of rapamycin inhib­i­tors While sin­gle-agent HDAC inhib­i­tors have lim­ited activ­ity in R/R
Preclinical stud­ies have shown that the PI3K-Akt-mam­ma­lian tar­ cHL (Table 2),32,33 the com­bi­na­to­rial approach with mTOR inhib­i­
get of rapamycin (mTOR) path­way plays an impor­tant role in the tors seems to be prom­is­ing. In a study that com­bined vorinostat
growth and sur­vival of Reed-Sternberg cells.24 Everolimus is an with mTOR inhib­i­tors (everolimus and sirolimus) in heavily pre-
oral inhib­i­tor of mTOR that is active in R/R cHL (Table 2).25 Recent- treated cHL (n  =  40), the ORR and CR rates were 55% and 33%,
ly, com­bi­na­tion ther­a­pies with everolimus have been explored in respec­tively.34 While the study included patients with prior BV
R/R cHL with encour­ag­ing pre­lim­i­nary activ­ity. In a phase 1/2 fail­ure, none of the patients in the study received CPI.
study of heavily pretreated cHL patients (n  =  15), the com­bi­na­
tion of everolimus and itacitinib (an oral Jak inhib­i­tor) pro­duced Transplant and cel­lu­lar ther­apy options
an ORR and CR rate of 79% and 14%, respec­tively. Of note, 94% Allogeneic HCT
(n  =  14) were dou­ble refrac­tory in the study. The nota­ble grade 3 Adoptive immu­no­ther­apy in the form of an allo-HCT offers a poten­
or higher toxicities included throm­bo­cy­to­pe­nia (43%), neutrope- tially cura­tive option for heavily pretreated R/R cHL patients,
nia (21%), infec­tion (7%), and hyper­ten­sion (7%).26 includ­ing those relaps­ing after a prior auto-HCT, BV, and CPI.6,35,36
Historically, allo-HCT in R/R cHL has been asso­ci­ated with high
Immunomodulatory agents rates of nonrelapse mor­tal­ity (NRM; 35%-45% at 1 year) and dis­ease
Lenalidomide mod­u­lates the immune micro­en­vi­ron­ment in lym­ relapse (up to 50%).37 These rates (with an obvi­ous neg­a­tive im-
phoid malig­nan­cies by interacting with ubiquitin E3 ligase cere- pact on refer­ral pat­terns) and the avail­abil­ity of novel agents in cHL
blon and degrading Ikaros tran­ scrip­
tion fac­
tors.27 Single-agent have led to a decline in the use of allo-HCT in cHL in the US in the
lenalidomide has pro­duced an ORR rang­ing from 13% to 30% last decade (Figure 2A, CIBMTR reg­is­try data). However, in dual-
across stud­ies in R/R cHL (Table 2).28,29 The major tox­ic­ity is hema­ refrac­tory cHL, the role of allo-HCT war­rants reappraisal, espe­cially
to­log­i­cal adverse events, which are slightly higher with con­tin­u­ when focus­ing on mod­ern out­come data from the CIBMTR show­
ous dos­ing.29 Combination approaches have also been stud­ied, ing remark­able improve­ment in sur­vival out­comes (Figure 2B, CIB-
mainly with his­tone deacetylase (HDAC) inhib­i­tors asso­ci­ated with MTR reg­is­try data) for these cHL patients fol­low­ing allografting.
sig­nif­i­cant tox­ic­ity with­out any improve­ment in response rates.30 Unlike the his­tor­i­cal data in which NRM rates were almost 50% and
3-year over­all sur­vival (OS) 25% to 30%,37 in the con­tem­po­rary se-
HDAC inhib­i­tors ries the NRM rates have declined to approx­i­ma­tely 10%, and 3-year
HDAC inhib­i­tors are epi­ge­netic mod­u­la­tors that induce cell death OS is approx­i­ma­tely 60%.7 These improve­ments could be due to
in cHL cell lines by inhibiting STAT6-medi­ated Th-2 cyto­kine and bet­ter con­di­tion­ing approaches,7,38 advances in graft-ver­sus-host
TARC (thy­mus and acti­va­tion-reg­u­lated chemokine) pro­duc­tion.31 dis­ease (GVHD) pre­ven­tion, and over­all advances in sup­port­ive

Prakash Singh Shekhawat


Double-refrac­tory Hodgkin lym­phoma | 249
Table 2.  Investigational ther­a­pies in R/R cHL

Study Median age Median


Agent design (range), years Total (N) Prior AHCT (n) ORR% (CR%) PFS/EFS (mo) Reference
Cami I* 38 (31-53) 57 NR 75 (44) NR Hamadani et al22
Cami II 36 (20-74) 51 31 83 (38) NR Herrera et al23
Everolimus II 32 (19-77) 57 38 46 (9) 8 Johnston et al25
Lenalidomide II 37 (18–74) 15 10 13 NR Kuruvilla et al28
Lenalidomide II 38 (20–83) 42 31 30 8.2 Fehniger et al29
Vorinostat II 42 (20-71) 25 11 4 7.2 Kirschbaum
et al32
Panobinostat II 32 (18-75) 129 129 27 (4) 6.9 Younes et al33

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/247/1851712/247epperla.pdf by guest on 13 December 2021


Idelalisib II 42 (21-80) 25 18 20 (4) 2.3 Gopal et al51
Ibrutinib† Retrosp 35 (26-72) 7 5 57 (43) NA Badar et al52
*Data shown for 30 µg/kg and 45 µg/kg cohorts.
†A clin­i­cal trial is ongo­ing inves­ti­gat­ing the role of ibrutinib in R/R cHL (NCT02824029).
NA, not appli­ca­ble; NR, not reported; Retrosp, ret­ro­spec­tive.

care and trans­plant prac­tices. In addi­tion, donor avail­abil­ity is no Epstein-Barr virus (EBV)-directed cyto­toxic T cells (CTLs)
lon­ger a bar­rier. Several stud­ies looked at the out­comes of alter­na­ directed against LMP 1 and LMP 2 have activ­ity in EBV+ cHL.
tive donor allo-HCT for R/R cHL and reported excel­lent out­comes EBNA1, LMP 1, and LMP 2 are attrac­tive tar­gets and have been
and low NRM (1 year, 10%-15%) among patients who received clin­i­cally tested.45,46 In patients with EBV-asso­ ci­ ated cHL and
posttransplant cyclo­phos­pha­mide (PTCy)-based haploidentical NHL, CTLs with enhanced activ­ity against LMP 1/2 were tested
(haplo) allo-HCT.35,39 with prom­is­ing results, wherein a response to ther­apy was noted
In cur­rent prac­tice, most of the cHL patients under­go­ing allo- in 13 of 21 patients with active dis­ease, includ­ing 11 patients
HCT have CPI expo­sure in the imme­di­ate pre-HCT period. Allo- achiev­ing CR.46 Another small study used LMP-spe­ cific CTLs
HCT in this set­ting (post CPI) may be asso­ci­ated with an increased expressing dom­i­nant-neg­a­tive transforming growth fac­tor β
risk of early posttransplant com­pli­ca­tions, espe­cially severe acute recep­ tor to coun­ ter­act transforming growth fac­ tor β-asso­ ci­
GVHD.40 It has been pos­tu­lated that this phe­nom­e­non is likely ated immune sup­pres­sion in the tumor micro­en­vi­ron­ment with
due to the higher fre­ quency of interferon-γ-pro­ duc­ ing effec­ good clin­i­cal results.47 Currently, a phase 1 study using EBV CTLs
tor T cells and a more pro­nounced T helper 1 dif­fer­en­ti­a­tion in expressing CD30 chi­ me­
ric recep­tor for CD30+ lym­ pho­ mas is
patients exposed to CPI before allo-HCT.41 PTCy has been shown ongo­ing (NCT01192464).
to not only abro­ gate the CPI-induced immune acti­ va­tion but AFM13 is a first-in-class innate cell engager that acti­vates
also pro­mote the vig­or­ous recov­ery of reg­u­la­tory T cells, lead­ the immune sys­tem and tar­gets CD30+ hema­to­logic can­cers.
ing to immune tol­er­ance and thereby suppressing GVHD to mild AFM13 induces the spe­ cific and selec­ tive kill­
ing of CD30+
lev­els.41,42 In a recently published “real-world” anal­ y­
sis of cHL cells by engag­ing CD16 on nat­u­ral killer (NK) cells and CD30
patients who under­ went allo-HCT after CPI treat­ ments, those on the sur­face of the cHL cells (bispecific NK cell engagers,
who received haplo/PTCy had a lower cumulative incidence of or BIKEs). Given the min­ i­
mal sin­
gle-agent activ­ ity of AFM13
relapse (2 year  =  7%) and excel­lent OS (2 year  =  85%). The time in R/R cHL (11.5%),48 it has been com­bined with CPI (AFM13+
from CPI to allo-HCT was an impor­tant pre­dic­tor of GVHD risk, pembrolizumab in CPI-naive R/R cHL) and with cyto­ kine-
wherein patients with a lon­ger time from CPI expo­sure to allo- acti­vated cord blood-derived NK cells.49 The pre­lim­i­nary safety
HCT (>80 days) had a decreased risk of severe acute GVHD.43 and effi­cacy data on first-in-human cord blood-derived NK
Given these improve­ments in recent years, early refer­ral to trans­ BIKEs (CD16A/CD30) in heavily pretreated cHL (n   =  4) are
plant pro­grams must be con­sid­ered for fit cHL patients with dual- pro­voc­a­tive. The ORR/CR rate was 100% and 50%, respec­
refrac­tory dis­ease that responds to avail­­able sal­vage treat­ments. tively, among the 4 patients treated with this approach, with
no evi­dence of cyto­kine release syn­drome, neu­ro­tox­ic­ity,
Cellular ther­a­pies or GVHD noted.50 Currently, the study is enroll­ing patients
Given the remark­able activ­ity of chi­me­ric anti­gen recep­tor mod­i­ (NCT04074746).
fied T (CAR-T) cell ther­apy in R/R B-cell NHL, this approach is un-
der inves­ti­ga­tion in cHL. In a phase 1/2 study of heavily pretreated
cHL (median prior lines  =  7) includ­ing BV, CPI, auto-HCT, and/or
allo-HCT, CD30.CAR-T cell ther­apy was safe and well tol­er­ated. CLINICAL CASE (Con­tin­ued)
The ORR was 72% in those receiv­ing fludarabine-based lympho- The patient in the clin­i­cal case achieved a CR with cami on a
depletion (n  =  31) with a CR rate of 59%,44 but 1-year pro­gres­sion- clin­i­cal trial and went on to receive a reduced-inten­sity con­di­
free sur­vival was only 36%, underscoring the need to improve the tion­ing haplo-HCT from his sib­ling donor. At the last fol­low-up,
plat­form. Currently, a larger phase 2 study is ongo­ing to eval­u­ate the patient was in remis­sion with mild active chronic GVHD.
the effi­cacy of CD30.CAR-T cells in R/R cHL (NCT04268706).

250  |  Hematology 2021  |  ASH Education Program


A 250
196 199 201
Number of Transplants 200 184 186
168 168
157
147
150 136
127
114 106 109
105
100

50

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/247/1851712/247epperla.pdf by guest on 13 December 2021


Year of Transplant

B 100 p<0.001

80
Probability, %

60

40

20
1990-1995 1996-2000 2001-2005
2006-2010 2011-2015 2016-2020
0
0 1 2 3 4 5
Years
Figure 2.  (A) Utilization of allo-HCT for cHL in the US. (B) Overall survival of patients undergoing allo-HCT in the US from 1990 to 2020.
Based on CIBMTR registry data.

Conclusions ceutical Company; Consultancy: Incyte Corporation; ADC Ther-


Although the advent of BV and CPI has changed the ther­a­peu­ apeutics; Pharmacyclics, Omeros, Kite. Speaker’s Bureau: Sanofi
tic land­scape of cHL man­age­ment, these drugs are not cura­tive Genzyme, AstraZeneca, BeiGene.
(in an R/R set­ting). As these agents are moved ear­lier into the
treat­ment lines, many more patients will become dou­ble refrac­ Off-label drug use
tory. Conventional cyto­ toxic and targeted ther­ a­
pies may be Narendranath Epperla: camidanlumab tesirine, AFM13, lenalido-
con­sid­ered in these patients, who are oth­er­wise not eli­gi­ble for mide, everolimus, vorinostat, idelalisib, ibrutinib, panabinostat.
clin­i­cal tri­als or allo-HCT. In patients who are trans­plant eli­gi­ble, Mehdi Hamadani: camidanlumab tesirine, AFM13, lenalidomide,
allo-HCT should be strongly con­ sid­ered given the sig­ nif­i­
cant everolimus, vorinostat, idelalisib, ibrutinib, panabinostat.
improve­ment in out­comes with allo-HCT in the con­tem­po­rary
era. Preliminary results with the cel­lu­lar ther­apy options that in- Correspondence
clude CD30.CAR-Ts, EBV CTLs, and CD30 BIKEs appear prom­is­ Mehdi Hamadani, Blood and Marrow Transplant Program and
ing and are cur­rently in clin­i­cal tri­als. As we con­tinue on our path Cellular Therapy Program, Department of Medicine, Medical Col-
toward a greater under­stand­ing of the biol­ogy of cHL, we will lege of Wisconsin, 9200 W Wisconsin Ave, Ste C5500, Milwau-
be ­able to iden­tify addi­tional ther­a­peu­tic tar­gets with the goal kee, WI 53226; e-mail: mhamadani@mcw​­.edu.
of pro­vid­ing dura­ble responses while min­i­miz­ing tox­ic­ity in R/R
cHL patients. References
1. Johnson P, Federico M, Kirkwood A, et al. Adapted treat­ment guided by
Conflict-of-inter­est dis­clo­sure interim PET-CT scan in advanced Hodgkin’s lym­ phoma. N Engl J Med.
2016;374(25):2419-2429.
Narendranath Epperla: reports Speaker’s Bureau: Verastem and 2. Schmitz N, Pfistner B, Sextro M, et al; Ger­man Hodgkin’s Lymphoma Study
Beigene; Advisory Board: Karyopharm; Honorarium: Genzyme. Group; Lymphoma Working Party of the Euro­pean Group for Blood and
Mehdi Hamadani: research sup­ Prakash Takeda
port/funding: Singh Pharma-
Shekhawat ­Marrow Transplantation. Aggressive con­ven­tional che­mo­ther­apy ­com­pared

Double-refrac­tory Hodgkin lym­phoma | 251


with high-dose che­mo­ther­apy with autol­o­gous haemopoietic stem-​cell 24. Dutton A, Reynolds GM, Dawson CW, Young LS, Murray PG. Constitutive
trans­
­ plan­ta­
tion for relapsed chemosensitive Hodgkin’s dis­ ease: a ran- acti­va­tion of phos­pha­ti­dyl-inositide 3 kinase con­trib­utes to the sur­vival of
domised trial. Lancet. 2002;359(9323):2065-2071. Hodgkin’s lym­phoma cells through a mech­a­nism involv­ing Akt kinase and
3. Moskowitz CH, Nademanee A, Masszi T, et al; AETHERA Study Group. mTOR. J Pathol. 2005;205(4):498-506.
Brentuximab vedotin as con­sol­i­da­tion ther­apy after autol­o­gous stem-cell 25. Johnston PB, Pinter-Brown LC, Warsi G, White K, Ramchandren R. Phase 2
trans­plan­ta­tion in patients with Hodgkin’s lym­phoma at risk of relapse or study of everolimus for relapsed or refrac­tory clas­si­cal Hodgkin lym­phoma.
pro­gres­sion (AETHERA): a randomised, dou­ble-blind, pla­cebo-con­trolled, Exp Hematol Oncol. 2018;7(May):12.
phase 3 trial. Lancet. 2015;385(9980):1853-1862. 26. Svoboda J, Barta SK, Landsburg DJ, et  al. Everolimus plus itacitinib in
4. Advani R, Moskowitz AJ, Bartlett NL, et al. Brentuximab vedotin in com­bi­ relapsed/refrac­tory clas­si­cal Hodgkin lym­phoma: results of a phase I/II
na­tion with nivolumab in relapsed or refrac­tory Hodgkin lym­phoma: 3-year inves­ti­ga­tor ini­ti­ated trial (EVITA study). Blood. 2020;136(suppl 1):20-21.
study results. Blood. 2021;138(6):427-438. 27. Kritharis A, Coyle M, Sharma J, Evens AM. Lenalidomide in non-Hodgkin
5. Connors JM, Jurczak W, Straus DJ, et al; ECHELON-1 Study Group. Brentux- lym­phoma: bio­log­i­cal per­spec­tives and ther­a­peu­tic oppor­tu­ni­ties. Blood.
imab vedotin with che­mo­ther­apy for stage III or IV Hodgkin’s lym­phoma. N 2015;125(16):2471-2476.
Engl J Med. 2018;378(4):331-344. 28. Kuruvilla J, Taylor D, Wang L, Blattler C, Keating A, Crump M. Phase II trial
6. Badar T, Epperla N, Szabo A, et al. Trends in postrelapse sur­vival in clas­sic of lenalidomide in patients with relapsed or refrac­tory Hodgkin lym­phoma.
Hodgkin lym­phoma patients after expe­ri­enc­ing ther­apy fail­ure fol­low­ing Blood. 2008;112(11):3052-3052.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/247/1851712/247epperla.pdf by guest on 13 December 2021


auto-HCT. Blood Adv. 2020;4(1):47-54. 29. Fehniger TA, Larson S, Trinkaus K, et al. A phase 2 mul­ti­cen­ter study of
7. Ahmed S, Ghosh N, Ahn KW, et  al. Impact of type of reduced-inten­ con­tin­u­ous dose lenalidomide in relapsed or refrac­tory clas­si­cal Hodgkin
sity con­di­tion­ing reg­i­men on the out­comes of allo­ge­neic haematopoi- lym­phoma. Blood. 2012;120(21):1623.
etic cell trans­plan­ta­tion in clas­si­cal Hodgkin lym­phoma. Br J Haematol. 30. Maly JJ, Chris­tian BA, Zhu X, et al. A phase I/II trial of panobinostat in com­
2020;190(4):573-582. bi­na­tion with lenalidomide in patients with relapsed or refrac­tory Hodgkin
8. Santoro A, Bredenfeld H, Devizzi L, et al. Gemcitabine in the treat­ment of lym­phoma. Clin Lymphoma Myeloma Leuk. 2017;17(6):347-353.
refrac­tory Hodgkin’s dis­ease: results of a mul­ti­cen­ter phase II study. J Clin 31. Buglio D, Georgakis GV, Hanabuchi S, et al. Vorinostat inhib­its STAT6-medi­
Oncol. 2000;18(13):2615-2619. ated TH2 cyto­kine and TARC pro­duc­tion and induces cell death in Hodgkin
9. Baetz T, Belch A, Couban S, et al. Gemcitabine, dexa­meth­a­sone and cis­ lym­phoma cell lines. Blood. 2008;112(4):1424-1433.
platin is an active and non-toxic che­mo­ther­apy reg­i­men in relapsed or 32. Kirschbaum MH, Goldman BH, Zain JM, et al. A phase 2 study of vorinos-
refrac­tory Hodgkin’s dis­ease: a phase II study by the National Cancer Insti- tat for treat­ment of relapsed or refrac­tory Hodgkin lym­phoma: Southwest
tute of Canada Clinical Trials Group. Ann Oncol. 2003;14(12):1762-1767. Oncology Group Study S0517. Leuk Lymphoma. 2012;53(2):259-262.
10. Bartlett NL, Niedzwiecki D, Johnson JL, et al; Cancer Leukemia Group B. 33. Younes A, Sureda A, Ben-Yehuda D, et  al. Panobinostat in patients with
Gemcitabine, vinorelbine, and pegylated lipo­so­mal doxo­ru­bi­cin (GVD), relapsed/refrac­tory Hodgkin’s lym­phoma after autol­o­gous stem-cell trans­
a sal­vage reg­im­ en in relapsed Hodgkin’s lym­phoma: CALGB 59804. Ann plan­ta­tion: results of a phase II study. J Clin Oncol. 2012;30(18):2197-2203.
Oncol. 2007;18(6):1071-1079. 34. Janku F, Park H, Call SG, et al. Safety and effi­cacy of vorinostat plus siroli-
11. Santoro A, Magagnoli M, Spina M, et  al. Ifosfamide, gemcitabine, and mus or everolimus in patients with relapsed refrac­tory Hodgkin lym­phoma.
vinorelbine: a new induc­tion reg­i­men for refrac­tory and relapsed Hod- Clin Cancer Res. 2020;26(21):5579-5587.
gkin’s lym­phoma. Haematologica. 2007;92(1):35-41. 35. Ahmed S, Kanakry JA, Ahn KW, et al. Lower graft-ver­sus-host dis­ease and
12. Moskowitz CH, Nimer SD, Zelenetz AD, et al. A 2-step com­pre­hen­sive high- relapse risk in post-trans­plant cyclo­phos­pha­mide-based haploidentical ver­
dose chemoradiotherapy sec­ond-line pro­gram for relapsed and refrac­tory sus matched sib­ling donor reduced-inten­sity con­di­tion­ing trans­plant for
Hodgkin dis­ease: anal­y­sis by intent to treat and devel­op­ment of a prog­ Hodgkin lym­phoma. Biol Blood Marrow Transplant. 2019;25(9):1859-1868.
nos­tic model. Blood. 2001;97(3):616-623. 36. Shah NN, Hamadani M. Is there still a role for allo­ge­neic trans­plan­ta­tion in
13. Josting A, Rudolph C, Reiser M, et al; Participating Centers. Time-inten­si­fied the man­age­ment of lym­phoma? J Clin Oncol. 2021;39(5):487-498.
dexa­meth­a­sone/cis­platin/cytarabine: an effec­tive sal­vage ther­apy with 37. Sureda A, Robinson S, Canals C, et al. Reduced-inten­sity con­di­tion­ing com­
low tox­ic­ity in patients with relapsed and refrac­tory Hodgkin’s dis­ease. pared with con­ven­tional allo­ge­neic stem-cell trans­plan­ta­tion in relapsed
Ann Oncol. 2002;13(10):1628-1635. or refrac­tory Hodgkin’s lym­phoma: an anal­y­sis from the Lymphoma Work-
14. Aparicio J, Segura A, Garcerá S, et al. ESHAP is an active reg­i­men for relaps­ ing Party of the Euro­pean Group for Blood and Marrow Transplantation.
ing Hodgkin’s dis­ease. Ann Oncol. 1999;10(5):593-595. J Clin Oncol. 2008;26(3):455-462.
15. Little R, Wittes RE, Longo DL, Wilson WH. Vinblastine for recur­rent Hod- 38. Ghosh N, Ahmed S, Ahn KW, et al. Association of reduced-inten­sity con­di­
gkin’s dis­ease fol­low­ing autol­o­gous bone mar­row trans­plant. J Clin Oncol. tion­ing reg­i­mens with over­all sur­vival among patients with non-Hodgkin
1998;16(2):584-588. lym­phoma under­go­ing allo­ge­neic trans­plant. JAMA Oncol. 2020;6(7):1011-
16. Devizzi L, Santoro A, Bonfante V, et al. Vinorelbine: an active drug for the 1018.
man­age­ment of patients with heavily pretreated Hodgkin’s dis­ease. Ann 39. Martínez C, Gayoso J, Canals C, et al; Lymphoma Working Party of the Euro­
Oncol. 1994;5(9):817-820. pean Group for Blood and Marrow Transplantation. Post-trans­plan­ta­tion
17. Moskowitz AJ, Hamlin PA Jr, Perales MA, et  al. Phase II study of benda- cyclo­phos­pha­mide-based haploidentical trans­plan­ta­tion as alter­na­tive to
mustine in relapsed and refrac­ tory Hodgkin lym­ phoma. J Clin Oncol. matched sib­ling or unre­lated donor trans­plan­ta­tion for Hodgkin lym­phoma:
2013;31(4):456-460. a Registry Study of the Lymphoma Working Party of the Euro­pean Society
18. Goda JS, Massey C, Kuruvilla J, et al. Role of sal­vage radi­at­ion ther­apy for for Blood and Marrow Transplantation. J Clin Oncol. 2017;35(30):3425-
patients with relapsed or refrac­tory Hodgkin lym­phoma who failed autol­o­ 3432.
gous stem cell trans­plant. Int J Radiat Oncol Biol Phys. 2012;84(3):e329-e335. 40. Merryman RW, Kim HT, Zinzani PL, et al. Safety and effi­cacy of allo­ge­neic
19. Deng L, Liang H, Burnette B, et  al. Irradiation and anti-PD-L1 treat­ hema­to­poi­etic stem cell trans­plant after PD-1 block­ade in relapsed/refrac­
ment syn­er­gis­ti­cally pro­mote anti­tu­mor immu­nity in mice. J Clin Invest. tory lym­phoma. Blood. 2017;129(10):1380-1388.
2014;124(2):687-695. 41. Nieto JC, Roldán E, Jiménez I, et al. Posttransplant cyclo­phos­pha­mide after
20. Weichselbaum RR, Liang H, Deng L, Fu YX. Radiotherapy and immu­no­ther­ allo­ge­neic hema­to­poi­etic cell trans­plan­ta­tion mit­i­gates the immune acti­va­
apy: a ben­e­fi­cial liai­son? Nat Rev Clin Oncol. 2017;14(6):365-379. tion induced by pre­vi­ous nivolumab ther­apy. Leukemia. 2020;34(12):3420-
21. Michot JM, Mazeron R, Dercle L, et al. Abscopal effect in a Hodgkin lym­ 3425.
phoma patient treated by an anti-programmed death 1 anti­body. Eur J 42. Ikegawa S, Meguri Y, Mizuhara K, et  al. Pretransplant nivolumab fur­
Cancer. 2016;66(Octo­ber):91-94. ther enhanced Treg expan­ sion after posttransplant cyclo­ phos­
pha­
mide:
22. Hamadani M, Collins G, Caimi P, Samaniego F, Spira A, et  al. Camidan- another aspect for immune tol­er­ance by PTCy after nivolumab. Leukemia.
lumab tesirine in relapsed/refrac­tory lym­phoma: a phase 1, mul­ti­cen­ter, 2021;35(3):929-931.
open-label, dose-esca­la­tion, dose-expan­sion study. Lancet Haematol. 43. Merryman RW, Castagna L, Giordano L, et  al. Allogeneic trans­plan­ta­tion
2021;8(6):e433-e445. after PD-1 block­ade for clas­sic Hodgkin lym­phoma. Leukemia. 2021;35(9):
23. Herrera AF, Carlo-Stella C, Collins GP, et  al. Preliminary results of a phase 2672-2683.
2 study of camidanlumab tesirine (cami), a novel pyrrolobenzodiazepine-​ 44. Ramos CA, Grover NS, Beaven AW, et al. Anti-CD30 CAR-T cell ther­apy in
based anti­body-drug con­ju­gate, in patients with relapsed or refrac­tory Hod- relapsed and refrac­tory Hodgkin lym­phoma. J Clin Oncol. 2020;38(32):3794-
gkin lym­phoma. Blood. 2020;136(suppl 1):21-23. 3804.

252  |  Hematology 2021  |  ASH Education Program


45. Icheva V, Kayser S, Wolff D, et  al. Adoptive trans­fer of Epstein-Barr virus 50. Rezvani K. CAR NK cells: a drive to the future of cell ther­apy. Paper pre-
(EBV) nuclear anti­gen 1-spe­cific T cells as treat­ment for EBV reactivation sented at: AACR Annual Meeting 2021; April 10-15, 2021; vir­ tual online
and lymphoproliferative dis­or­ders after allo­ge­neic stem-cell trans­plan­ta­ meet­ing.
tion. J Clin Oncol. 2013;31(1):39-48. 51. Gopal AK, Fanale MA, Moskowitz CH, et  al. Phase II study of idelalisib, a
46. Bollard CM, Gottschalk S, Torrano V, et al. Sustained com­plete responses selec­tive inhib­i­tor of PI3Kδ, for relapsed/refrac­tory clas­si­cal Hodgkin lym­
in patients with lym­phoma receiv­ing autol­o­gous cyto­toxic T lym­pho­ phoma. Ann Oncol. 2017;28(5):1057-1063.
cytes targeting Epstein-Barr virus latent mem­brane pro­teins. J Clin Oncol. 52. Badar T, Astle J, Kakar IK, Zellner K, Hari PN, Hamadani M. Clinical activ­
2014;32(8):798-808. ity of ibrutinib in clas­si­cal Hodgkin lym­phoma relaps­ing after allo­ge­neic
47. Bollard CM, Tripic T, Cruz CR, et al. Tumor-spe­cific T-cells engineered to stem cell trans­plan­ta­tion is inde­pen­dent of tumor BTK expres­sion. Br J
over­come tumor immune eva­sion induce clin­i­cal responses in patients Haematol. 2020;190(2):e98-e101.
with relapsed Hodgkin lym­phoma. J Clin Oncol. 2018;36(11):1128-1139.
48. Rothe A, Sasse S, Topp MS, et  al. A phase 1 study of the bispecific anti-
CD30/CD16A anti­body con­struct AFM13 in patients with relapsed or refrac­
tory Hodgkin lym­phoma. Blood. 2015;125(26):4024-4031.
49. Bartlett NL, Herrera AF, Domingo-Domenech E, et al. A phase 1b study of
AFM13 in com­bi­na­tion with pembrolizumab in patients with relapsed or © 2021 by The Amer­i­can Society of Hematology

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/247/1851712/247epperla.pdf by guest on 13 December 2021


refrac­tory Hodgkin lym­phoma. Blood. 2020;136(21):2401-2409. DOI 10.1182/hema­tol­ogy.2021000256

Prakash Singh Shekhawat


Double-refrac­tory Hodgkin lym­phoma | 253
HOW CAN WE ENSURE THAT EVERYONE WHO NEEDS A TRANSPLANT CAN GET ONE ?

Allogeneic hematopoietic cell transplantation


for older patients

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/254/1851535/254lin.pdf by guest on 13 December 2021


Richard J. Lin1 and Andrew S. Artz2
Memorial Sloan Kettering Cancer Center, New York, NY; and 2City of Hope National Medical Center, Duarte, CA
1

Hematologic malignances are more common and often higher risk in older patients. Allogeneic hematopoietic cell trans-
plantation (alloHCT) best enables long-term disease control for patients with poor risk or relapsed/refractory hema-
tologic malignancies such as acute myeloid leukemia, myelodysplastic syndromes, or myelofibrosis. Rates of alloHCT
among older patients, while still relatively low compared with younger patients, have risen sharply over the past decade.
Accumulating evidence supports alloHCT for patients ≥60 years of age relative to non-HCT therapies based on improved
overall and disease-free survival. However, a significant proportion of older adults have limitations characterized by geri-
atric assessment. A systematic process to evaluate and optimize older patients may improve decision making, transplant
outcomes, and alloHCT access. We present case-based studies to illustrate a stepwise and rational approach to proper
older patient evaluation, pretransplant optimization, and posttransplant care with attention to important geriatric issues
and quality of life.

LEARNING OBJECTIVES
• Describe access barriers to allogeneic hematopoietic cell transplantation for older adults
• Understand the role of GA, management, and optimization strategies for an older adult throughout the alloHCT
process

Introduction CASE 1
Allogeneic hematopoietic cell transplantation (alloHCT) Mr. RM is a 73­year­old man with coronary artery dis­
remains the best­established curative option for many ease, hypertension, diabetes, and moderate obesity who
patients with advanced hematologic malignancies, par­ resides in a rural town with his wife and children in an
ticularly myeloid neoplasms.1 In recent years, we have active lifestyle. One year ago, he initiated hypomethylat­
witnessed significant advances in reducing transplant­ ing agent therapy through his local oncologist for newly
related mortality, manipulating graft­versus­leukemia diagnosed high­risk, transfusion­requiring myelodysplas­
effect to prevent/treat relapse, and developing alloHCT tic syndrome (MDS) with excess blasts. The MDS evolved
as a platform for novel cellular therapies.2,3 Older age to acute myeloid leukemia (AML) 1 year later, prompting
may have been the most formidable and important bar­ induction with liposomal daunorubicin and cytarabine.
rier, representing the next frontier.4 The demographics His treatment course was complicated by neutropenic
of blood cancer, especially myeloid malignancies, with fever and bacteremia. A follow­up bone marrow biopsy
a median age of onset in the late 60s to early 70s and demonstrated complete remission. Should Mr. RM be
frequently higher risk underscore the need.5 The era of referred for consolidation alloHCT?
alloHCT is upon us; the Center for International Blood
and Marrow Transplantation Research (CIBMTR) reports
that patients aged ≥60 years comprised more than 40% AlloHCT vs chemotherapy in older patients
of adult alloHCT volume in the United States (Figure 1).6 Older patients, especially those in their 70s, face the unique
In this review, we discuss unique challenges facing old­ challenge of finite life expectancy that may be further con­
er patients in alloHCT and strategies to improve their strained by medical comorbidities.7 AlloHCT for older pa­
outcomes. tients with AML poses the dual dangers of complications

254 | Hematology 2021 | ASH Education Program


Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/254/1851535/254lin.pdf by guest on 13 December 2021
Figure 1.  Trends in alloHCT in the United States by increasing recipient age (N = total number of alloHCTs during each calendar year;
Transplant, % reflects the percentage of alloHCT in each age group by calendar year). Data generously provided by the Center for
International Blood and Marrow Transplant Research.

includ­ing death after alloHCT with­out relapse (nonrelapse mor­ ≥60 years with hema­to­logic malig­nan­cies (Figures 1 and 2), fur­
tal­ity) and dis­ease relapse. As such, it is imper­a­tive that phy­si­cians ther stim­u­lated by wider donor avail­abil­ity, includ­ing haploiden­
and patients weigh the ben­e­fits and risks of alloHCT vs nontrans­ tical, for most patients. Rashidi et al15 performed a meta-anal­y­sis
plant approaches, ide­ally early in the treat­ment course. Sever­ of 13 stud­ies of patients with AML 60 years and older who under­
al pop­u­la­tion-based stud­ies have shown that invari­ably, older went alloHCT. The 2-year relapse-free sur­vival and OS were 44%
patients with inter­me­di­ate- or poor-risk AML (which com­prise and 45%, respec­tively, suggesting that alloHCT is a via­ble op­
most newly diag­nosed AMLs in older patients) rarely sur­vive for tion for these patients. Similar find­ings were dem­on­strated for
more than 5 years with­out an alloHCT.8,9 In a study com­par­ing patients with a vari­ety of hema­to­logic malig­nan­cies.16-18 Even
patients with AML aged ≥60 years treated with con­sol­i­da­tion among patients older than 70 years, a recent CIBMTR anal­y­sis
che­ mo­ ther­
apy alone in first com­ plete remis­sion in sev­ eral showed accept­able if not prom­is­ing 2-year pro­gres­sion-free sur­
national coop­er­a­tive tri­als vs sim­i­larly aged patients under­go­ing vival and OS of 32% and 39%, respec­tively, in het­ero­ge­neous
alloHCT in first com­plete remis­sion from the con­tem­po­rary CIB­ dis­eases, donor sources, and reg­i­mens.19 These data rein­force
MTR trans­plant reg­is­try,10 sur­vival was worse for alloHCT in the that chro­no­logic age alone, at least up to 75 years, should not
first 9 months posttransplant rel­a­tive to con­sol­i­da­tion on tri­als. exclude an older patient from alloHCT can­di­dacy. Rather, we
However, after 5 years, alloHCT sig­nif­i­cantly benefited patient pro­pose the patient’s “phys­i­o­logic” age should be eval­u­ated,
over­all sur­vival (OS) at 28.6% vs 13.8% in the chemo-con­sol­i­da­ along with a com­pre­hen­sive assess­ment of the patient’s goals
tion cohort (haz­ard ratio, 0.53; P < .0001). Table 1 high­lights sim­i­ of care, qual­ity of life (QOL), and the eco­sys­tem, includ­ing care­
lar find­ings from sev­eral reg­is­try stud­ies com­par­ing alloHCT with giv­ers, social sup­port sys­tem, finan­cial resources, and liv­ing sit­
nontransplant chemo-con­sol­i­da­tion tri­als for AML.11-13 In addi­tion, u­a­tion (Figure 3).4,20 Although beyond the scope of this review,
3 pro­spec­tive, donor vs no-donor stud­ies for patients with AML even among reduced-inten­sity reg­i­mens, a range of trans­plant
were published in abstract form, which also sup­ports alloHCT in inten­si­ties exist that must be indi­vid­u­al­ized based on patient
this pop­u­la­tion (Table 1). The most recently reported Blood and health and dis­ease risk.19-22 Furthermore, graft-ver­sus-host dis­
Marrow Transplantation Clinical Trial Network (BMT CTN) 1102 ease (GVHD) remains a major cause of mor­bid­ity and func­tional
pro­spec­tively stud­ied bio­log­i­cally assigned, newly diag­nosed impair­ment in this pop­u­la­tion, prompting con­sid­er­ation of lower
high-risk patients with MDS aged 60 to 75 years to alloHCT with GVHD plat­forms (Figure 3).23,24
a matched donor vs hypomethylating ther­apy with­out alloHCT
in the absence of a matched donor; the pres­ence of a matched Transplant access bar­ri­ers for older patients
donor con­ferred a 3-year OS advan­tage of 47.9% vs 26.6%.14 Referral bias and other bar­ri­ers limit access among older pa­
tients to alloHCT. A recent sys­temic review of 26 stud­ies showed
AlloHCT out­comes in older patients that chro­no­logic older age is the sin­gle most impor­tant bar­rier
Associated with many advances in trans­plan­ta­tion, the num­ber to refer patients for alloHCT con­sid­er­ation.25 Specifically, opin­
and pro­por­tion of total alloHCT con­tinue to rise in patients aged ions dif­fer mark­edly among hema­tol­o­gists/oncol­o­gists, trans­
Prakash Singh Shekhawat
Allotransplant in older adults  | 255
Table 1.  Multicenter stud­ies com­par­ing alloHCT to non-HCT con­sol­i­da­tion strat­eg
­ ies in older patients with AML or MDS

AlloHCT donor
Study N Age range Study design dis­ease Comparison groups Survival out­comes Comments
source/con­di­tion­ing
Farag et al. AlloHCT: 94 60-70 Retrospective, mul­ti­cen­ter AlloHCT: CIBMTR Registry Matched: 77% 3-year OS: OS ben­e­fit in all­ cyto­­
(2011)11 Non-HCT: 96 anal­y­sis Non-HCT: CT in CALGB Alternative: 23% AlloHCT: 37% ge­netic risk groups.
AML in CR1 RIC/NMA: 100% Non-HCT: 25%
3-year LFS:
AlloHCT: 32%
Non-HCT: 15%
Kurosawa et al. AlloHCT: 152 50-70 Retrospective, mul­ti­cen­ter AlloHCT: Jap­a­nese Registry Matched: 76% 3-year OS: 183 patients in the CT
(2011)12 Non-HCT: 884 anal­y­sis Non-HCT: Jap­a­nese Registry Alternative: 24% (15% AlloHCT: 62% group even­tu­ally
AML in CR1 cord) Non-HCT: 51% received HCT.
MA: 38% 3-year RFS: OS ben­e­fits in inter­me­
RIC/NMA: 62% AlloHCT: 56% di­ate-risk group.
Non-HCT: 29%
Versluis et al. AlloHCT: 97 ≥60 Retrospective anal­y­sis of AlloHCT: HOVON-SAKK tri­als Matched: 92% 5-year OS: Benefits are seen in
(2015)13 Non-HCT: 177 mul­ti­cen­ter tri­als Non-HCT: HOVON-SAKK tri­als Alternative: 8% AlloHCT: 35% both inter­me­di­ate-
None: 366 AML in CR1 RIC/NMA: 100% Non-HCT: 26% risk and adverse risk
5-year RFS: groups.

256  |  Hematology 2021  |  ASH Education Program


AlloHCT: 32%
Non-HCT: 20%
Ustun et al. AlloHCT: 431 60-77 Retrospective, mul­ti­cen­ter AlloHCT: CIBMTR Registry Matched: 65% 5-year OS: OS/DFS is worse in
(2019)10 Non-HCT: 211 anal­y­sis Non-HCT: CT in coop­er­a­ Alternative: 35% AlloHCT: 28.6% the first 9 months
AML in CR1 tive group tri­als (CALGB, (24% cord) Non-HCT: 13.8% for alloHCT. OS/DFS
SWOG, ECOG) MA: 29% 5-year DFS: ben­e­fits more for
RIC/NMA: 71% AlloHCT: 23.7% poor-risk patients.
Non-HCT: 11.1%
*Niederwieser AlloHCT: 150 (donor) 50-75 Prospective, intent-to-treat, AlloHCT: Donor group Matched: 79% 9-year LFS: Benefits seen across
et al. (2016) Non-HCT: 205 (no donor vs no-donor trial Non-HCT: No donor con­sol­i­ Mismatched: 21% Donor: 25% both inter­me­di­
ASCO donor) AML in CR1 da­tion group RIC/NMA: 100% No donor: 14% ate- and high-risk
Abstract 9-year RI: groups.
e18501 Donor: 42%
No donor: 78%
*Brune et al. AlloHCT: 77 (donor) 50-70 Prospective, intent-to-treat, AlloHCT: Donor group Matched: 100% 3-year OS: 7 patients in con­trol
(2018) ASH Non-HCT: 68 (no donor vs no-donor trial Non-HCT: No donor con­sol­i­ RIC/NMA: 100% Donor: 45% group crossed over.
Abstract 205 donor) AML in CR1 da­tion group No donor: 48% Extremely high relapse
3-year RFS: rate in both arms
Donor: 40% (49% vs 60%)
No donor: 35%
*Foran et al. AlloHCT: 135 (donor) ≥60 Prospective, intent-to-treat, AlloHCT: Donor group Matched: 100% Median OS: 44 patients in the
(2018) EHA Non-HCT: 225 (no donor vs no-donor trial Non-HCT: No donor con­sol­i­ RIC/NMA: 100% Donor: 22.1 months donor group and
Abstract donor) AML in CR1 da­tion group No donor: 13.4 months 25 patients in the
S857 (ECOG (P = .013) no-donor group
E2906) crossed over.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/254/1851535/254lin.pdf by guest on 13 December 2021


Table 1.  Multicenter stud­ies com­par­ing alloHCT to non-HCT con­sol­i­da­tion strat­e­gies in older patients with AML or MDS (Continued)

AlloHCT donor
Study N Age range Study design dis­ease Comparison groups Survival out­comes Comments
source/con­di­tion­ing
Nakamura et al. AlloHCT: 260 (donor) 50-75 Prospective, intent-to-treat, AlloHCT: Donor group Matched: 100% 3-year OS: Similar ben­e­fits in 3-
(2021) (BMT Non-HCT: 124 (no donor vs no-donor trial Non-HCT: No donor con­sol­i­ RIC/NMA: 100% Donor: 47.9% year LFS.
CTN 1102)14 donor) MDS IPSS Intermediate- da­tion group No donor: 26.6% No decrease in QOL.
2/high risk 3-year OS (as treated
anal­y­sis):

Prakash Singh Shekhawat


AlloHCT: 47.4%
Non-HCT: 16%
*Meeting abstract only. ASCO, American Society of Clinical Oncology; ASH, American Society of Hematology; CALGB, Cancer and Leukemia Group B; CR1, first complete remission; CT,
chemotherapy consolidation; DFS, disease-free survival; ECOG, Eastern Cooperative Oncology Group; EHA, European Hematology Association; HOVON-SAKK, Dutch-Belgian Hemato-Oncology
Cooperative Group and the Swiss Group for Clinical Cancer Research; IPSS, International Prognostic Scoring System; LFS, leukemia-free survival; MA, myeloablative conditioning; RFS, relapse
free survival; RI, relapse incidence; RIC/NMA, reduced-intensity/nonmyeloablative conditioning; SWOG, Southwest Oncology Group.

Allotransplant in older adults  | 257


Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/254/1851535/254lin.pdf by guest on 13 December 2021
Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/254/1851535/254lin.pdf by guest on 13 December 2021
Figure 2. Trends in alloHCT in the United States for patients 76 years or older (N = total number of transplants). Data generously
provided by the Center for International Blood and Marrow Transplant Research.

plant phy­si­cians, and trans­plant cen­ters regard­ing the upper age increase in uti­ li­
za­tion. Second, we should explore inno­ va­ tive
limit for alloHCT, likely as a result of indi­vid­ual expe­ri­ence and approaches to incor­po­rate phys­i­o­logic aging eval­u­a­tion by
exper­tise.26,27 Routine aging assess­ment could neu­tral­ize het­ero­ GA in the rou­tine care of older patients, such as an embed­ded
ge­ne­ity in opin­ion; how­ever, the lack of stan­dard­ized geri­at­ric geri­at­ric hema­tol­ogy clinic and telemedicine plat­form.31,32 Last,
assess­ment (GA) tools and resources to accom­plish them chal­ we must invest in greater edu­ca­tional and out­reach efforts to
lenges phys­i­o­logic aging eval­u­a­tion.27 Other noted fac­tors hin­ raise aware­ness of the emerg­ing, prom­is­ing alloHCT out­come
der­ing access included non­white eth­nic ori­gin, insur­ance sta­tus, data, the role of GA, and clin­i­cal trial oppor­tu­ni­ties spe­cif­i­cally
higher comorbidities, and lower socio­eco­nomic sta­tus. Given designed for older patients.26
recent advances in trans­plan­ta­tion using alter­na­tive donors such
as haploidentical and mismatched donors, lack of a matched do­
nor should not be exclu­sion­ary even among older patients.28,29
There are sev­eral poten­tial mit­i­ga­tion strat­e­gies to reduce
access bar­ ri­
ers. First and fore­
most, dis­ ease indi­ ca­
tions for CASE 1 (Continued)
alloHCT should be clearly defined for older patients to sup­ple­ Mr. RM had sev­eral telemedicine vis­its with the trans­plant phy­si­
ment stan­dard alloHCT guide­lines,30 account­ing for worse out­ cian, a clin­i­cal nurse coor­di­na­tor, and a social worker, all­located
comes for AML, MDS, and acute lym­pho­blas­tic leu­ke­mia even in at an aca­demic med­i­cal cen­ter 200 miles away. Cognizant of his
the same dis­ease risk group. Rather than a dichot­o­mous sin­gle comorbid con­di­tions, nec­es­sary eval­u­a­tion, and poten­tial early
deci­sion point of “fit” or “not fit” for trans­plant, we rec­om­mend loss of QOL from alloHCT, Mr. RM and his fam­ily expressed a desire
expe­dited refer­ral for alloHCT eval­u­a­tion in the appro­pri­ate dis­ to pro­ceed. He also com­pleted a remote, video-assisted GA,
ease indi­ca­tions for patients 60 years or older in the pres­ence of which dem­on­strated pre­served self-reported func­tional sta­tus,
ade­quate base­line func­tional sta­tus and with­out severe organ mobil­ity, and cog­ni­tion. In par­al­lel, the unre­lated donor search
comorbidities (Figure 4). We must strive to enroll patients aged proceeded, iden­ ti­
fy­
ing a young matched unre­ lated donor.
>75 years on alloHCT stud­ies; until then, the deci­sion must be During che­mo­ther­apy con­sol­i­da­tion locally, he under­went pre­
indi­vid­u­al­ized in this age group. Figure 2 quantifies the lim­ited transplant test­ing also through his local oncol­o­gist. Four weeks
appli­ca­tion of alloHCT in this cohort but also the sub­ stan­tial later, he began a reduced-inten­sity trans­plant reg­i­men inclu­sive

258  |  Hematology 2021  |  ASH Education Program


Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/254/1851535/254lin.pdf by guest on 13 December 2021
Figure 3.  Solving the puzzle of alloHCT for older patients with hematologic malignancies. MRD, measurable residual disease; PPx,
prophylaxis.

Figure 4.  How we perform alloHCT for an older patient with a hematologic malignancy. HLA, human leukocyte antigen. *Severe
comorbidities: New York Heart Association class 4 heart failure, severe renal dysfunction or end-stage renal disease on dialysis, Child
class C liver cirrhosis, Gold stage 4 chronic obstructive lung disease, metastatic solid tumor, dementia, or any comorbidity signifi­
cantly limiting life expectancy.
Prakash Singh Shekhawat
Allotransplant in older adults  | 259
of posttransplant cyclo­phos­pha­mide for GVHD pro­phy­laxis on addi­tional, prognostically impor­tant domains such as cog­ni­tion,
the BMT CTN 1301 Progress 3 trial (NCT02345850) from a young med­ic ­ a­tion, and frailty scales. These stud­ies are sum­ma­rized in
well-matched unre­lated donor. Table 2.36-44 The ongo­ing BMT CTN 1704 trial (Composite Health
Assessment Risk Model [CHARM]) is a large national study pro­
spec­tively using a stan­dard GA and other mea­sures prior to al­
GA in alloHCT loHCT among patients ≥60 years old, aiming to con­firm these
The shift from fit­ness alone to assessing resilience to dis­ease- find­ings and/or iden­tify addi­tional risk fac­tors (NCT03992352).
related and trans­plant-related stress­ors broad­ens interventional
oppor­tu­ni­ties that may widen access (Figure 3). The term resil­
iency encompasses both the intrin­sic, “phys­i­o­logic” aging pro­
cess and the extrin­sic “eco­sys­tem,” includ­ing care­giver, social CASE 2 (Continued)
sup­port, finance, and resources; GA com­bined with stan­dard
trans­plant psy­cho­so­cial eval­u­a­tion achieves this goal.4 GA is a The trans­ plant team recommended short-term defer­ ral to
mul­ti­dis­ci­plin­ary diag­nos­tic pro­cess that identifies med­i­cal, address GA-defined def­i­cits while con­tinu­ing che­mo­ther­apy

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/254/1851535/254lin.pdf by guest on 13 December 2021


func­tional, and psy­cho­so­cial lim­i­ta­tions of an older per­son and to deepen dis­ease response. Mrs. LK under­went reha­bil­it­ a­tive
place him or her on a con­tin­u­ous spec­trum of fit­ness, vul­ner­a­bil­ ther­apy with phys­i­cal and occu­pa­tional ther­apy supplemented
ity, and frailty and fur­ther informs a mul­ti­dis­ci­plin­ary care plan by home walk­ing and strength­en­ing super­vised by her fam­
to max­i­mize healthy aging, as illus­trated in the fol­low­ing case.33 ily. The res­o­lu­tion of trans­fu­sion-depen­dent ane­mia fur­ther
boosted phys­i­cal recov­ery. The geri­at­rics team man­aged poly­
pharmacy by actively deprescribing non­es­sen­tial med­i­ca­tions
thought to con­trib­ute to the mild cog­ni­tive def­i­cits. Repeated
GA 2 months later dem­on­strated improved func­tional sta­tus
CASE 2 and cog­ni­tion (no lon­ger in the impaired range). Depressive
Mrs. LK is a 70-year-old woman who had stage II early breast symp­toms resolved with more social engage­ment and phys­ic ­ al
can­cer 2 years ago that was treated with sur­gery, radi­a­tion, inde­pen­dence. Based on these results and another informed
and adju­vant che­mo­ther­apy with no evi­dence of dis­ease, mod­ dis­cus­sion, the patient and trans­plant team elected to pro­
er­ate chronic obstruc­tive pul­mo­nary dis­ease, oste­o­ar­thri­tis, ceed. She sub­se­quently under­went reduced-inten­sity con­di­
and atrial fibril­la­tion. She sought treat­ment from her pri­mary tion­ing alloHCT using her 36-year-old haploidentical son with
care phy­si­cian for fatigue and was found to have pan­cy­to­pe­ posttransplant cyclo­phos­pha­mide for pre­ven­tion of GVHD.
nia with periph­eral blasts. A bone mar­row biopsy spec­i­men The patient had a care­giver starting the day before trans­plant
established the diag­ no­sis of AML har­ bor­ing a mono­ somy 7. infu­sion and con­tinu­ing through­out. The trans­plant admis­sion
Due to her comorbidities, low-inten­sity induc­tion com­menced was com­pli­cated by an epi­sode of delir­ium ini­tially rec­og­nized
with azacitidine and venetoclax, which was com­pli­cated only by the care­giver. After exclud­ing organic causes, she received
by ongo­ing cytopenia. Repeat bone mar­row eval­u­at­ion after hal­o­per­i­dol as needed, and occu­pa­tional ther­apy pre­scribed
1 cycle dem­on­strated com­plete remis­sion but with pos­i­tive inten­ sive cog­ ni­
tive exer­ cises. She was discharged on post­
mea­sur­able resid­ual dis­ease by mul­ti­color flow cytom­e­try and transplant day +37 to home with a walker and a home exer­cise
cyto­ge­net­ics. She was referred for trans­plant con­sul­ta­tion. The reg­i­men, avoiding a sub­acute reha­bil­i­ta­tion facil­ity. She con­tin­
GA revealed depen­dence in sev­eral instru­men­tal activ­i­ties of ued “vir­tual” clin­ics vis­its and phys­i­cal face-to-face encoun­ters
daily liv­ing, recently depressed mood, and a screen­ing test and ongo­ing reha­bil­i­ta­tion.
pos­i­tive for mild cog­ni­tive impair­ment. She would like to pur­
sue cura­tive-intent alloHCT con­sol­id ­ a­tion if pos­si­ble. She has a
highly sup­port­ive fam­ily and care­giver who con­cur and under­ Geriatric man­age­ment and opti­mi­za­tion
stand that trans­plant tox­ic­ity may be pro­hib­i­tive, espe­cially While GA may uncover vulnerabilities in older patients con­sid­
con­sid­er­ing the GA-defined def­i­cits and comorbidities. What is er­ing alloHCT, how best to opti­mize patients prior to alloHCT
the appro­pri­ate next step? remains a work in prog­ress. Challenges include short time avail­­
able before alloHCT due to the pace of dis­ease and delayed
refer­ral, nonmodifiable def­i­cits such as comorbidities, and lim­
GA domains affect alloHCT out­comes ited insti­tu­tional resources. Low-inten­sity inter­ven­tions would
Physiologic aging established through GA, cou­pled with antic­i­ be ideal; how­ ever, the BMT CTN conducted a mul­ ti­
cen­
ter,
pated stress­ors of the dis­ease and treat­ments, begins to paint a ran­dom­ized study of struc­tured home exer­cise and a stress man­
pic­ture of phys­i­cal resilience. Serial GA may enrich under­stand­ing age­ment pro­gram prior to trans­plan­ta­tion, find­ing no improve­
of resilience or “bounce back” after treat­ment. In the con­text of ment in phys­i­cal and men­tal func­tion­ing posttransplant.45 While
alloHCT, the GA should address the extrin­sic eco­sys­tem, includ­ not lim­ited to older patients, this accen­tu­ated the need for tar­
ing psy­cho­so­cial sup­port, care­giver sup­port, and resources for geted and/or more inten­sive pretransplant opti­mi­za­tion. Recent­
alloHCT (Figure 3). Artz and col­leagues conducted the ini­tial pi­ ly, Derman, Artz and col­leagues46 conducted the first pilot study
lot study of GA in alloHCT and found sig­nif­i­cant asso­ci­a­tions of apply­ing GA-guided inter­ven­tions in a mul­ti­dis­ci­plin­ary team
pretransplant geri­at­ric impair­ments in func­tion and mobil­ity with clinic (MDC) to opti­mize patients prior to trans­plant. They found
adverse sur­vival out­comes fol­low­ing alloHCT.34,35 Subsequently, that, com­pared to his­tor­i­cal cohorts with sim­i­lar dis­ease and
sev­eral groups inde­pen­dently val­i­dated these find­ings and found trans­plant char­ac­ter­is­tics, the MDC cohort expe­ri­enced fewer

260  |  Hematology 2021  |  ASH Education Program


Table 2.  Studies illus­trat­ing prognostically impor­tant geri­at­ric def­i­cits in older patients under­go­ing alloHCT

Impairment in GA domains with impact on out­comes


Age, median
Study N (range), y Study design dis­ease Comorbidity Function Mobility Cognition Medication Frailty scale
35
Muffly et al. (2014) 203 58 (54-63) Prospective, sin­gle-cen­ter pilot study NRM ↑ NRM ↑ OS ↓
All dis­eases OS ↓ OS ↓
Deschler et al. (2018)36 106 66 (60-78) Prospective, sin­gle-cen­ter trial NRM ↑ OS ↓ OS ↓
Myeloid neo­plasms PFS ↓ PFS ↓
Huang et al. (2020)37 148 62 (50-76) Prospective, sin­gle-cen­ter pilot study OS ↓
All dis­eases PFS ↓
NRM ↑
Toxicities ↑
Pamukcuoglu et al. 52 59 (40-73) Prospective, sin­gle-cen­ter pilot study OS ↓
(2019)40 All dis­eases Toxicities ↑

Prakash Singh Shekhawat


Salas et al. (2021)41 168 58 (19-77) Prospective, sin­gle-cen­ter pilot study NRM ↑ NRM ↑
All dis­eases OS ↓
Olin et al. (2020)38 330 63 (50-77) Multicenter, CIBMTR reg­is­try NRM ↑ NRM ↑
All dis­eases OS ↓
Polverelli et al. (2020)42 228 64 (60-76) Two-cen­ter ret­ro­spec­tive study NRM ↑
OS ↓
Lin et al. (2020)39 457 66 (60-79) Single-cen­ter, ret­ro­spec­tive study NRM ↑ NRM ↑
OS ↓
PFS ↓
Bhargava et al. (2020)43 114 68 (65-75) Single-cen­ter, ret­ro­spec­tive study Toxicities ↑
OS ↓
Sugidono et al. (2021)44 148 62 (50-76) Single-cen­ter, ret­ro­spec­tive study OS ↓
NRM, nonrelapse mor­tal­ity; PFS, pro­gres­sion-free sur­vival.

Allotransplant in older adults  | 261


Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/254/1851535/254lin.pdf by guest on 13 December 2021
inpa­tient deaths, shorter length of stay, fewer dis­charges to a mis­con­cep­tions, novel inter­ven­tions to bol­ster patient resilience,
skilled nurs­ing facil­ity, and improved sur­vival. The crit­i­cal com­ and trans­plant reg­i­mens prom­ises more wide­spread and more
po­nents of the MDC approach likely involve more care­ful patient suc­cess­ful appli­ca­tion of alloHCT for older adults with high-risk
selec­tion, targeted opti­mi­za­tion, and mul­ti­dis­ci­plin­ary col­lab­o­ hema­to­logic malig­nan­cies.
ra­tion.46,47 In addi­tion, early rec­og­ni­tion, espe­cially of eco­sys­tem
bar­ri­ers, through rou­tine eval­u­a­tion best affords oppor­tu­ni­ties to Acknowledgments
opti­mize. Telehealth and a shared care model, for exam­ple, may R.J.L. acknowl­edges the sup­port from the ASH Clinical Research
alle­vi­ate dis­tance bar­ri­ers for rou­tine pre- and posttransplant vis­ Training Institute and the ASH Scholar Award. We acknowl­edge
its, at least when patients can safely reside at home.47 GA-guided edi­to­rial sup­port from Sally Mokhtari. We thank Dr. Sergio Gi­
man­age­ment and inte­gra­tion of geri­at­ric prin­ci­ples of care ralt for com­ments on the con­cept. This work was also support­
should not be lim­ited to pretransplant care. The devel­op­ment of ed in part by the NIH/NCI Cancer Center Support Grant P30
geri­at­ric syn­dromes of func­tional decline, fall, delir­ium, and cog­ CA008748 to Memorial Sloan Kettering Cancer Center and P30
ni­tive impair­ment posttransplant is not uncom­mon, and these CA033572 to City of Hope National Medical Center. The con­tent
syn­dromes are asso­ci­ated with impaired sur­vival and QOL.24,48,49 is solely the respon­si­bil­ity of the authors and does not nec­es­sar­

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/254/1851535/254lin.pdf by guest on 13 December 2021


In addi­tion, dis­charge to a reha­bil­i­ta­tion facil­ity posttransplant ily rep­re­sent the offi­cial views of the National Institutes of Health.
has been shown to be a marker of poor sur­vival.50 These issues Likewise, the views expressed in this arti­cle are those of the au­
require fur­ther pro­spec­tive val­i­da­tion with patient-cen­tric out­ thors and do not reflect the posi­tion of the CIBMTR.
comes of func­tion and QOL.
Conflict-of-inter­est dis­clo­sure
How we per­form alloHCT in an older patient Richard J. Lin: no com­pet­ing finan­cial inter­ests to declare.
We sum­ma­rize our approach to alloHCT in an older patient with Andrew S. Artz: no com­pet­ing finan­cial inter­ests to declare.
hema­to­logic malig­nancy in Figure 4, work­ing toward suc­cess­ful
com­ple­tion of a final check­list. We rec­om­mend that hema­tol­o­
Off-label drug use
gists, patients, and insti­tu­tions first con­sider the “ABCDE” to tri­
Richard J. Lin: nothing to disclose.
age (early) refer­ral. We believe resil­iency mea­sure­ment, through
Andrew S. Artz: nothing to disclose.
GA or equiv­a­lent, is essen­tial in older can­di­dates to sup­ple­ment
stan­dard pre-alloHCT test­ing and the sub­jec­tive “fit­ness” cri­te­
Correspondence
ria. We advo­cate a col­lab­or­ a­tive model partnering the trans­plant
Andrew S. Artz, Department of Hematology and HCT, Center for
team and the dis­ease man­age­ment team (when sep­a­rate) to har­
Cancer and Aging, City of Hope National Medical Center, 1500 E.
mo­nize dis­ease ther­apy with antic­i­pated trans­plant tim­ing, often
Duarte Road, Duarte, CA 91010; e-mail: aartz@coh​­.org.
dic­tated by donor avail­abil­ity. Disease treat­ment may occur dis­
tant from the trans­plant cen­ter, par­tic­u­larly as a range of lower-
inten­sity treat­ments exist for com­mon alloHCT indi­ca­tions. This References
1. Copelan EA. Hematopoietic stem-cell trans­plan­ta­tion. N Engl J Med. 2006;
shared care model ensures more uni­form mes­sag­ing to patients 354(17):1813-1826.
related to alloHCT plans, risks, and ben­e­fits from all­phy­si­cians. 2. Granot N, Storb R. History of hema­ to­poi­etic cell trans­
plan­ta­tion: chal­
Shared care pro­motes the par­al­lel pro­cess of max­i­miz­ing resil­ lenges and prog­ress. Haematologica. 2020;105(12):2716-2729.
ience through GA-targeted inter­ven­tions and prep­a­ra­tion of the 3. Chabannon C, Kuball J, Bondanza A, et al. Hematopoietic stem cell trans­
plan­ta­tion in its 60s: a plat­ form for cel­lu­lar ther­
a­pies. Sci Transl Med.
supporting eco­sys­tem dur­ing dis­ease treat­ment. Alignment of 2018;10(436).
these pro­ cesses facil­i­
tates meet­ ing a “final check­list” before 4. Artz AS. Biologic vs phys­i­o­logic age in the trans­plant can­di­date. Hematol­
alloHCT (Figure 4). We acknowl­edge that not all­older patients ogy Am Soc Hematol Educ Program. 2016;2016(1):99-105.
who embark on this pro­cess will ulti­mately pur­sue alloHCT be­ 5. Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer sta­tis­tics, 2021. CA Cancer
J Clin. 2021;71(1):7-33.
cause of dis­ease relapse, inad­e­quate resilience, and/or changes
6. D’Souza A, Fretham C, Lee SJ, et al. Current use of and trends in hema­to­
in goals of care, underscoring the value of mul­ti­ple touch points poi­etic cell trans­plan­ta­tion in the United States. Biol Blood Marrow Trans­
to dis­cuss patient goals and recalibrate patient expec­ta­tions plant. 2020;26(8):e177-e182.
about the like­li­hood of meet­ing the final check­list. 7. Yourman LC, Lee SJ, Schonberg MA, Widera EW, Smith AK. Prognostic indi­
ces for older adults: a sys­tem­atic review. JAMA. 2012;307(2):182-192.
8. Pulte D, Jansen L, Castro FA, Brenner H. Changes in the sur­vival of older
Conclusion and future direc­tions patients with hema­to­logic malig­nan­cies in the early 21st cen­tury. Cancer.
We rec­om­mend alloHCT as a stan­dard of care option for older 2016;122(13):2031-2040.
patients with high-risk hema­ to­
logic malig­ nan­cies best estab­ 9. Vasu S, Kohlschmidt J, Mrózek K, et al. Ten-year out­come of patients with
lished for AML and MDS. Not only has uti­li­za­tion in older patients acute mye­loid leu­ke­mia not treated with allo­ge­neic trans­plan­ta­tion in first
com­plete remis­sion. Blood Adv. 2018;2(13):1645-1650.
risen mark­edly, but out­comes in older patients also con­tinue
10. Ustun C, Le-Rademacher J, Wang H-L, et al. Allogeneic hema­to­poi­etic cell
to improve due to incor­po­ra­tion of novel trans­plant plat­forms trans­plan­ta­tion com­pared to che­mo­ther­apy con­sol­i­da­tion in older acute
with reduced toxicities, an increased donor pool, and the bet­ter mye­loid leu­ke­mia (AML) patients 60-75 years in first com­plete remis­sion
selec­tion and care of older trans­plant patients. Moreover, we are (CR1): an alli­ance (A151509), SWOG, ECOG-ACRIN, and CIBMTR study. Leu­
begin­ning to appre­ci­ate the impact of aging biol­ogy on trans­ kemia. 2019;33(11):2599-2609.
11. Farag SS, Maharry K, Zhang M-J, et al; Acute Leukemia Committee of the
plant out­comes and to explore mech­a­nism-based, ther­a­peu­tic Center for International Blood and Marrow Transplant Research and Cancer
inter­ven­tions to tar­get aging path­ways.51 The con­ver­gence of and Leukemia Group B. Comparison of reduced-inten­sity hema­to­poi­etic
suc­cess in dis­ease-based ther­a­pies, edu­ca­tion to address age cell trans­plan­ta­tion with che­mo­ther­apy in patients age 60-70 years with

262  |  Hematology 2021  |  ASH Education Program


acute mye­log­e­nous leu­ke­mia in first remis­sion. Biol Blood Marrow Trans­ 32. Wall SA, Knauss B, Compston A, et al. Multidisciplinary telemedicine and
plant. 2011;17(12):1796-1803. the impor­tance of being seen. J Geriatr Oncol. 2020;11(8):1349-1351.
12. Kurosawa S, Yamaguchi T, Uchida N, et al. Comparison of allo­ge­neic hema­ 33. Wildiers H, Heeren P, Puts M, et al. International Society of Geriatric Oncol­
to­poi­etic cell trans­plan­ta­tion and che­mo­ther­apy in elderly patients with ogy con­sen­sus on geri­at­ric assess­ment in older patients with can­cer. J Clin
non-M3 acute mye­log­e­nous leu­ke­mia in first com­plete remis­sion. Biol Oncol. 2014;32(24):2595-2603.
Blood Marrow Transplant. 2011;17(3):401-411. 34. Muffly LS, Boulukos M, Swanson K, et al. Pilot study of com­pre­hen­sive geri­
13. Versluis J, Hazenberg CL, Passweg JR, et al; HOVON and SAKK Leukemia at­ric assess­ment (CGA) in allo­ge­neic trans­plant: CGA cap­tures a high prev­
Groups. Post-remis­sion treat­ment with allo­ge­neic stem cell trans­plan­ta­ a­lence of vulnerabilities in older trans­plant recip­i­ents. Biol Blood Marrow
tion in patients aged 60 years and older with acute mye­loid leu­kae­mia: a Transplant. 2013;19(3):429-434.
time-depen­dent anal­y­sis. Lancet Haematol. 2015;2(10):e427-e436. 35. Muffly LS, Kocherginsky M, Stock W, et al. Geriatric assess­ment to pre­dict
14. Nakamura R, Saber W, Martens MJ, et  al. Biologic assign­ ment trial of sur­vival in older allo­ge­neic hema­to­poi­etic cell trans­plan­ta­tion recip­i­ents.
reduced-inten­sity hema­to­poi­etic cell trans­plan­ta­tion based on donor Haematologica. 2014;99(8):1373-1379.
avail­abil­ity in patients 50-75 years of age with advanced myelodysplastic 36. Deschler B, Ihorst G, Schnitzler S, Bertz H, Finke J. Geriatric assess­ment and
syn­drome [published online 9 June 2021]. J Clin Oncol. qual­ity of life in older patients con­sid­ered for allo­ge­neic hema­to­poi­etic
15. Rashidi A, Ebadi M, Colditz GA, DiPersio JF. Outcomes of allo­ge­neic stem cell trans­plan­ta­tion: a pro­spec­tive risk fac­tor and serial assess­ment anal­y­
cell trans­ plan­ta­
tion in elderly patients with acute mye­ loid leu­
ke­
mia: a sis. Bone Marrow Transplant. 2018;53(5):565-575.
sys­tem­atic review and meta-anal­y­sis. Biol Blood Marrow Transplant. 37. Huang L-W, Sheng Y, Andreadis C, et al. Functional sta­tus as mea­sured by

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/254/1851535/254lin.pdf by guest on 13 December 2021


2016;22(4):651-657. geri­at­ric assess­ment pre­dicts infe­rior sur­vival in older allo­ge­neic hema­
16. Sorror ML, Sandmaier BM, Storer BE, et  al. Long-term out­comes among to­poi­etic cell trans­plan­ta­tion recip­i­ents. Biol Blood Marrow Transplant.
older patients fol­low­ing nonmyeloablative con­di­tion­ing and allo­ge­neic 2020;26(1):189-196.
hema­to­poi­etic cell trans­plan­ta­tion for advanced hema­to­logic malig­nan­ 38. Olin RL, Fretham C, Pasquini MC, et  al. Geriatric assess­ ment in older
cies. JAMA. 2011;306(17):1874-1883. alloHCT recip­i­ents: asso­ci­a­tion of func­tional and cog­ni­tive impair­ment
17. Ringdén O, Boumendil A, Labopin M, et al. Outcome of allo­ge­neic hema­ with out­comes. Blood Adv. 2020;4(12):2810-2820.
to­poi­etic stem cell trans­plan­ta­tion in patients age >69 years with acute 39. Lin RJ, Elko TA, Devlin SM, et al. Impact of geri­at­ric vulnerabilities on allo­
mye­log­e­nous leu­ke­mia: on behalf of the Acute Leukemia Working Party ge­neic hema­to­poi­etic cell trans­plan­ta­tion out­comes in older patients with
of the Euro­pean Society for Blood and Marrow Transplantation. Biol Blood hema­to­logic malig­nan­cies. Bone Marrow Transplant. 2020;55(1):157-164.
Marrow Transplant. 2019;25(10):1975-1983. 40. Pamukcuoglu M, Bhatia S, Weisdorf DJ, et al. Hematopoietic cell trans­plant-
18. Atallah E, Logan B, Chen M, et  al. Comparison of patient age groups in related toxicities and mor­tal­ity in frail recip­i­ents. Biol Blood Marrow Trans­
trans­plan­ta­tion for myelodysplastic syn­drome: the Medi­care cov­er­age plant. 2019;25(12):2454-2460.
with evi­dence devel­op­ment study. JAMA Oncol. 2020;6(4):486-493. 41. Salas MQ , Atenafu EG, Bascom O, et al. Pilot pro­spec­tive study of frailty
19. Muffly L, Pasquini MC, Martens M, et al. Increasing use of allo­ge­neic hema­ and func­tion­al­ity in rou­tine clin­i­cal assess­ment in allo­ge­neic hema­to­poi­
to­poi­etic cell trans­plan­ta­tion in patients aged 70 years and older in the etic cell trans­plan­ta­tion. Bone Marrow Transplant. 2021;56(1):60-69.
United States. Blood. 2017;130(9):1156-1164. 42. Polverelli N, Tura P, Battipaglia G, et al. Multidimensional geri­at­ric assess­
20. Olin RL. Delivering inten­sive ther­a­pies to older adults with hema­to­logic ment for elderly hema­to­log­i­cal patients (≥60 years) sub­mit­ted to allo­ge­
malig­nan­cies: strat­e­gies to per­son­al­ize care. Blood. 2019;134(23):2013-2021. neic stem cell trans­plan­ta­tion: a French-Ital­ian 10-year expe­ri­ence on 228
21. Hourigan CS, Dillon LW, Gui G, et  al. Impact of con­di­tion­ing inten­sity of patients. Bone Marrow Transplant. 2020;55(12):2224-2233.
allo­ge­neic trans­plan­ta­tion for acute mye­loid leu­ke­mia with geno­mic evi­ 43. Bhargava D, Arora M, DeFor TE, et al. Use of poten­tially inap­pro­pri­ate med­
dence of resid­ual dis­ease. J Clin Oncol. 2020;38(12):1273-1283. i­ca­tions in older allo­ge­neic hema­to­poi­etic cell trans­plan­ta­tion recip­i­ents.
22. Dillon LW, Gui G, Logan BR, et  al. Impact of con­di­tion­ing inten­sity and Biol Blood Marrow Transplant. 2020;26(12):2329-2334.
geno­ mics on relapse after allo­ ge­neic trans­ plan­
ta­
tion for patients with 44. Sugidono M, Lo M, Young R, et al. Impact of polypharmacy prior to allo­ge­
myelodysplastic syn­drome. JCO Precis Oncol. 2021;5:265-274. neic hema­to­poi­etic stem cell trans­plan­ta­tion in older adults. Transplant
23. El-Jawahri A, Pidala J, Inamoto Y, et al. Impact of age on qual­ity of life, func­ Cell Ther. 2021;27(4):344.e1-344.e5.
tional sta­tus, and sur­vival in patients with chronic graft-ver­sus-host dis­ 45. Jacobsen PB, Le-Rademacher J, Jim H, et al. Exercise and stress man­age­
ease. Biol Blood Marrow Transplant. 2014;20(9):1341-1348. ment train­ing prior to hema­to­poi­etic cell trans­plan­ta­tion: Blood and Mar­
24. Lin RJ, Baser RE, Elko TA, et al. Geriatric syn­dromes in 2-year, pro­gres­sion- row Transplant Clinical Trials Network (BMT CTN) 0902. Biol Blood Marrow
free sur­vi­vors among older recip­i­ents of allo­ge­neic hema­to­poi­etic cell Transplant. 2014;20(10):1530-1536.
trans­plan­ta­tion. Bone Marrow Transplant. 2021;56(1):289-292. 46. Derman BA, Kordas K, Ridgeway J, et al. Results from a mul­ti­dis­ci­plin­ary
25. Flannelly C, Tan B-E, Tan J-L, et al. Barriers to hema­to­poi­etic cell trans­plan­ clinic guided by geri­at­ric assess­ment before stem cell trans­plan­ta­tion in
ta­tion for adults in the United States: a sys­tem­atic review with a focus on older adults. Blood Adv. 2019;3(22):3488-3498.
age. Biol Blood Marrow Transplant. 2020;26(12):2335-2345. 47. Wildes TM, Artz AS. Characterize, opti­mize, and har­mo­nize: car­ing for older
26. Meyer C, Mau L-W, Mur­phy E-A, et al. Addressing knowl­edge gaps in acute adults with hema­to­logic malig­nan­cies. Am Soc Clin Oncol Educ Book.
mye­loid leu­ke­mia to improve refer­ral for hema­to­poi­etic cell trans­plan­ta­ 2021;41:e266-e274.
tion con­sul­ta­tion. J Natl Compr Canc Netw. 2019;17(12):1473-1481. 48. Fann JR, Alfano CM, Roth-Roemer S, Katon WJ, Syrjala KL. Impact of delir­
27. Mishra A, Preussler JM, Bhatt VR, et al. Breaking the age bar­rier: phy­si­cians’ ium on cog­ni­tion, dis­tress, and health-related qual­ity of life after hema­to­
per­cep­tions of can­di­dacy for allo­ge­neic hema­to­poi­etic cell trans­plan­ta­ poi­etic stem-cell trans­plan­ta­tion. J Clin Oncol. 2007;25(10):1223-1231.
tion in older adults. Transplant Cell Ther. 2021;27(7):617.e1-617.e7. 49. Lin RJ, Hilden PD, Elko TA, et al. Burden and impact of mul­ti­fac­to­rial geri­
28. Kasamon YL, Bolaños-Meade J, Prince GT, et  al. Outcomes of nonmye­ at­ric syn­dromes in allo­ge­neic hema­to­poi­etic cell trans­plan­ta­tion for older
loablative HLA-haploidentical blood or mar­row trans­plan­ta­tion with high- adults. Blood Adv. 2019;3(1):12-20.
dose post-trans­plan­ta­tion cyclo­phos­pha­mide in older adults. J Clin Oncol. 50. Wall SA, Zhao Q , Vasu S, Rosko A. Discharge dis­po­si­tion fol­low­ing hema­
2015;33(28):3152-3161. to­poi­etic cell trans­plan­ta­tion: predicting the need for reha­bil­i­ta­tion and
29. Shaw BE, Jimenez-Jimenez AM, Burns LJ, et al. National mar­row donor pro­ asso­ci­a­tion with sur­vival. Transplant Cell Ther. 2021;27(4):337.e1-337.e7.
gram-spon­sored mul­ti­cen­ter, phase II trial of HLA-mismatched unre­lated 51. Lin RJ, Elias HK, van den Brink MRM. Immune recon­sti­tu­tion in the aging
donor bone mar­row trans­plan­ta­tion using post-trans­plant cyclo­phos­pha­ host: oppor­tu­ni­ties for mech­a­nism-based ther­apy in allo­ge­neic hema­to­
mide. J Clin Oncol. 2021;39(18):1971-1982. poi­etic cell trans­plan­ta­tion. Front Immunol. 2021;12:674093.
30. Kanate AS, Perales M-A, Hamadani M. Eligibility cri­te­ria for patients under­
go­ing allo­ge­neic hema­to­poi­etic cell trans­plan­ta­tion. J Natl Compr Canc
Netw. 2020;18(5):635-643.
31. Goede V, Stauder R. Multidisciplinary care in the hema­tol­ogy clinic: imple­ © 2021 by The Amer­i­can Society of Hematology
mentation of geri­at­ric oncol­ogy. J Geriatr Oncol. 2019;10(3):497-503. DOI 10.1182/hema­tol­ogy.2021000257

Prakash Singh Shekhawat


Allotransplant in older adults  | 263
HOW CAN WE ENSURE THAT EVERYONE WHO NEEDS A TRANSPLANT CAN GET ONE ?

Increasing access to allogeneic hematopoietic


cell transplant: an international perspective

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/264/1851868/264rocha.pdf by guest on 13 December 2021


Vanderson Rocha,1,2,3 Giancarlo Fatobene,1,2,3 and Dietger Niederwieser,4 for the Brazilian Society of Bone
Marrow Transplantation and the Worldwide Network for Blood and Marrow Transplantation
Laboratorio de Investigação Médica (LIM) 31, Serviço de Hematologia e Terapia Celular, Hospital das Clínicas, Faculdade de Medicina, Universidade
1

de São Paulo, São Paulo, Brazil; 2Eurocord, Paris, France; 3Hospital Vila Nova Star - Rede D’Or, São Paulo, Brazil; and 4University of Leipzig, Leipzig,
Germany

Allogeneic hematopoietic cell transplantation (allo-HCT) is a highly complex, costly procedure for patients with onco-
logic, hematologic, genetic, and immunologic diseases. Demographics and socioeconomic status as well as donor avail-
ability and type of health care system are important factors that influence access to and outcomes following allo-HCT.
The last decade has seen an increase in the numbers of allo-HCTs and teams all over the world, with no signs of satura-
tion. More than 80 000 procedures are being performed annually, with 1 million allo-HCTs estimated to take place by the
end of 2024. Many factors have contributed to this, including increased numbers of eligible patients (older adults with or
without comorbidities) and available donors (unrelated and haploidentical), improved supportive care, and decreased
early and late post-HCT mortalities. This increase is also directly linked to macro- and microeconomic indicators that affect
health care both regionally and globally. Despite this global increase in the number of allo-HCTs and transplant centers,
there is an enormous need for increased access to and improved outcomes following allo-HCT in resource-constrained
countries. The reduction of poverty, global economic changes, greater access to information, exchange of technologies,
and use of artificial intelligence, mobile health, and telehealth are certainly creating unprecedented opportunities to
establish collaborations and share experiences and thus increase patient access to allo-HCT. A specific research agenda
to address issues of allo-HCT in resource-constrained settings is urgently warranted.

LEARNING OBJECTIVES
• Understand the panorama of allogeneic hematopoietic cell transplantation (allo-HCT) activity worldwide
• Review factors affecting the access to allo-HCT across the globe
• Discuss possible strategies for expanding allo-HCT activity in resource-constrained areas

Introduction (4) improvements in early and late outcomes due to better


Allogeneic hematopoietic cell transplantation (allo-HCT) is a supportive care. However, allo-HCT is still associated with
highly specialized, technologically sophisticated, resource- significant morbidity and mortality, requiring advanced
intense, and expensive procedure for patients with oth- care consisting of significant infrastructure and a network
erwise incurable hematologic diseases. Numerous factors of specialists from all fields of medicine.
influence whether a patient eligible for allo-HCT actually The World Health Organization (WHO) has recognized
goes on to receive it: donor availability, social issues, eco- the broad idea of the provision of medical products of
nomic status, and health care system.1 The degree to which human origin as an important global medical task and a
these factors, particularly socioeconomic factors, might governmental responsibility at the national level.2,3 Data
influence access to allo-HCT is a question of debate.1 collection and data analysis are also recognized as inte-
In recent decades a worldwide increase in the use of gral parts of the treatment to achieve an efficient and cost-
allo-HCT has been seen due to (1) better donor availabil- effective use of resources. The Worldwide Network for
ity, (2) optimization of indications and rapid evolution of Blood and Marrow Transplantation (WBMT), as a nongov-
molecular diagnostic/prognostic techniques, (3) use of ernmental organization in official relations with the WHO,
novel reduced-intensity conditioning regimens for older has taken up the challenge of collecting global HCT activity
adult patients and/or patients with comorbidities, and data. Information on HCT trends over time provides a sound

264 | Hematology 2021 | ASH Education Program


basis for sci­en­tific soci­e­ties, pol­i­ti­cians, and health care agencies (LA; 3.27), and the Eastern Med­i­ter­ra­nean/Afri­can Region (EMR/
to develop or opti­mize national HCT pro­grams. AFR; 1.98; Figure 2B). Accordingly, allo-HCT/team ratios were
This arti­cle pres­ents a global over­view of the num­bers and higher in EMR/AFR and NA, dem­on­strat­ing a par­tic­u­larly high
types of allo-HCT in dif­fer­ent world regions col­lected by mem­ con­cen­tra­tion of pro­ce­dures in few trans­plant cen­ters in EMR/AFR
bers of the WBMT (the Euro­pean Group for Blood and Mar- (Figure 2B).5
row Trans­plantation, the Center for International Blood and
Marrow Transplant Research, and the Asia-Pacific Blood and Mar- Trends in indi­ca­tions, donor type, and stem cell source
row Transplantation Group) or directly from trans­plant cen­ters The num­ber of trans­plants for all­indi­ca­tions using all­type of
(where no regional reg­is­try is in place) using a uni­form reporting donors (except for plasma cell dis­or­ders and lym­pho­mas in
sheet.4,5 As many patient, dis­ease, donor, and trans­plant-related related allo-HCT; data not shown) increased.5 Leukemia was
fac­tors may improve out­comes, includ­ing those affect­ing access the most fre­quent indi­ca­tion, of which acute mye­loid leu­ke­mia
to allo-HCT, this arti­cle also describes these aspects. (AML) amounted 14 334 HCTs. HCT for myelodysplastic syn­
dromes (MDS) and, to a lower extent, mye­lo­pro­lif­er­a­tive dis­or­
Worldwide allo-HCT activ­ity and over­all trends ders increased steadily over the obser­va­tion period. Increased

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/264/1851868/264rocha.pdf by guest on 13 December 2021


Worldwide, 1 298 897 HCTs (42.9% allo-HCT) pro­ce­dures have allo-HCT activ­i­ties were observed for almost all­indi­ca­tions in
been recorded from the first HCT in 1957 to 2016.5 The annual all­regions except for solid tumors in almost all­regions but
activ­ity increased con­tin­u­ously from 46 563 in 2006 (the foun­ SEAR/WPR and lymphoproliferative dis­or­ders in NA (Figure 3).
da­tion of WBMT and the first global report) to 82 718 in 2016 The highest incre­ments were observed for severe aplastic ane­
(the lat­est com­plete global sur­vey, Table 1),4-6 amounting to a mia and hemo­glo­bin­op­a­thies (Δ2007-2016 > 1000%) and for leu­
global increase of 77.6% since 2006, which was some­what higher ke­mia (Δ2007-2016 > 550%; data not shown).5
in allo-HCT (89.0%) than in auto-HCT (68.9%).5 Allo-HCTs (range, 0-7850) were reported from 76 countries,
includ­ing grafts from unre­lated donors (UDs) and from cord blood
Allo-HCT team activ­ity in 2016 (CB) in 55 and in 41 countries, respec­tively. Absolute UD-HCT num­
In 2016, 38 425 first allo-HCTs, in com­par­i­son to 20 333 in 2006, bers ranged from 0 to 4311, and those of CB ranged from 0 to 1233
were performed world­wide (Figure 1). The increase took place in indi­vid­ual countries. Overall, related-HCT has become more
in all­regions of the world, and in con­trast to 2006, more related fre­quent than UD-HCT, starting in 2014, due to the increased use
(53.6%) than unre­lated HCTs were reported.5 Increases of 20% of related haploidentical (haplo) donors (39.5% of related HCT).
and 50% were noted in the num­ber of allo-HCT teams and allo- Haplo-HCTs (n = 8131) were evenly dis­trib­uted in 62 countries, with
HCTs, respec­tively, while the allo-HCT/team ratio var­ied from abso­lute num­bers rang­ing from 0 to 2554. It is not sur­pris­ing that
15.4 in 2006 to 23.3 in 2016 (Figure 2A). Team den­sity (TD, the more haplo-HCTs were performed in regions with­out or with few
num­ber of teams/10 mil­lion per­sons) was highest in North Amer- UD reg­is­tries (LA, EMR, AFR, and SEAR/WPR).
ica (NA; 23.72), followed by Europe (EUR; 18.53), the South-East Of the 38 425 allo-HCTs, 20% were derived from bone mar­
Asian/Western Pacific Region (SEAR/WPR; 3.91), Latin America row (BM), 73% from periph­eral blood (PB), and 7% from CB.5

Figure 1. Allo-HCT activity according to donor type (related and unrelated), irrespective of source and matching (PBSC, BM, or CB;
matched or mismatched), and region Prakash
comparingSingh
2006 toShekhawat
2016. BM, bone marrow; CB, cord blood; PBSC, peripheral blood stem cell.

Factors affect­ing global access to allotransplant  |  265


A
Number of HCTs

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/264/1851868/264rocha.pdf by guest on 13 December 2021


allo-HCT
B
World Regions

Number of HCTs
HCT/team
Figure 2. (A) Number of allo-HCTs, number of allo-HCT teams, and allo-HCTs per team. (B) Number of allo-HCT teams, number of
allo-HCTs per team, and TD according to region in 2016.

Factors affect­ing access to allo-HCT (Figure 4) 179.45 in South America, 198.28 in NA, and 247.33 in Europe; Joris
Donor avail­abil­ity M., per­sonal com­mu­ni­ca­tion, May 2021). The num­bers of unre­
UD reg­is­tries: impact of eth­nic­ity and eco­nomic issues.  In the lated grafts (BM, PB, and CB cells) expanded >7-fold from 1997
absence of an HLA-iden­ti­cal related donor, hema­to­poi­etic cells to 2020, with increased use of PB (except for the COVID-19 pan­
from HLA-matched or mismatched UD, haplo donors, or CB cells demic period in 2020) and decreased use of BM and CB cells
can be used. Countries with UD reg­is­tries increased from 2 in (Figure 5A). Figure 5B shows the global exchange of prod­ucts
1987 to 57 in 2012 and 119 in 2021. Accordingly, the donor pool has (BM + PB) in 2020. In a National Marrow Donor Program study,
increased in the last 40 years to >40 mil­lion UD, includ­ing CB (May the prob­a­bil­ity of find­ing a UD for black Amer­i­cans is <17%.7 In
2021; https://wmda.info/). Rates of UD per 10 000 inhab­i­tants a recent study of the Brazilian UD reg­is­try (REDOME), this prob­
vary per con­ti­nent (2.98 in Africa, 16.5 in Asia, 61.64 in Oceania, a­bil­ity was <10% for sickle cell dis­ease patients.8 In addi­tion,

266  |  Hematology 2021  |  ASH Education Program


Delta (%)

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/264/1851868/264rocha.pdf by guest on 13 December 2021


Figure 3. Change in % of allo-HCT activity from 2006 and 2016 activity according to diagnosis and region. Adapted with permission
of Haematologica.5

access to searching for and using UD grafts is lim­ited by finan­ cial issues—graft-related costs being an impor­tant fac­tor in low-
cial issues even in dif­ fer­ent areas of high-income countries and mid­dle-income countries (LICs, LMICs).
(HICs). In the US, Paulson et al. found that patients with leu­ke­
mia from poorer areas were less likely to undergo UD allo-HCT Impact of haplo donors on access to allo-HCT.  The use of haplo
com­pared to patients from wealth­ier countries.9 Similar find­ings donors is increas­ing world­wide, but inter­est­ingly, this is more evi­
were also reported in Italy.10 Despite many ret­ro­spec­tive stud­ies dent in LICs and LMICs, prob­a­bly due to the unavail­abil­ity of HLA-
and some pro­spec­tive tri­als reporting sim­i­lar out­comes after UD, matched UD, HCT-asso­ ci­
ated costs, infra­ struc­ture of cen­ ters,
CB, or haplo-HCT, the use of UD over haplo or CB depends on lack of access to drugs, and other fac­tors. According to the last
patient/donor eth­nic­ity, avail­abil­ity of a UD reg­is­try, and finan­ WBMT report, the haplo-HCT trans­plant rate (TR; HCT/10 mil­lion

Figure 4. Factors associated


Prakash Singhwith access to allo-HCT: an international perspective.
Shekhawat
Factors affect­ing global access to allotransplant  |  267
Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/264/1851868/264rocha.pdf by guest on 13 December 2021
Figure 5. (A) Unrelated BM, mobilized PB, and CB grafts shipped from 1997 to 2020. (B) Global exchange of hematopoietic cell
products (BM + PB) in 2020.

pop­u­la­tion) represented 32% and 26% of all­allo-HCTs in SEA/W- quan­tity and qual­ity of rela­tion­ships and the care­giver’s income
PR and LA, respec­ tively, but only 14% in Europe and EMR/ loss may affect the avail­abil­ity of such a per­son.23,24
AFR and 4.9% in NA.5
Economics
Patient demo­graph­ics: age, gen­der, and eth­nic­ity Macro- and micro­eco­nomic fac­tors: impact on access to trans­
Studies inves­ti­gat­ing age as a pre­dic­tor of access to HCT indi­cate plan­ta­tion.  From a global per­spec­tive, access to and rates of allo-
that youn­ger patients are more likely to receive a trans­plant com­ HCT are closely related to the socio­eco­nomic sta­tus of countries
pared to older patients, even in HICs.11,12 Only 5.5% of older adult and regions of the globe. International find­ings show strik­ing dif­
patients with AML (n = 17 555) under­went HCT in the US between fer­ences in abso­lute trans­plant num­bers, TD, and the spread of
2003 and 2012. Other fac­tors asso­ci­ated with a lower like­li­hood allo-HCT, which are affected mainly by a coun­try’s or region’s
of receiv­ing HCT included care at a non­ac­a­demic hos­pi­tal, race macro- or micro­eco­nomic fac­tors related to resources and infra­
other than White, Charlson comorbidity score of ≥1, unin­sured struc­ture.4 For instance, a recent study showed that adults with
sta­tus, and lower edu­ca­tional sta­tus.12 No sig­nif­i­cant gen­der AML treated in São Paulo (Brazil) had infe­rior 5-year over­all sur­
effect has been reported.13 The role of race in turn must be inter- vival (OS) and higher early mor­tal­ity pri­mar­ily due to multiresis-
preted with cau­tion because race is a com­plex social, cul­tural, tant gram-neg­a­tive bac­te­rial and fun­gal infec­tions com­pared to
and polit­i­cal con­struct and not only a bio­log­ic ­ al con­cept. The patients from Oxford (UK). Importantly, Brazilian patients were
avail­abil­ity of a donor for non-Cau­ca­sians and eth­nic minor­i­ties less likely to undergo allo-HCT (28% vs 75%; P < .001) and waited
has improved over the last decades with the use of haplo donors lon­ger for HCT (median, 23.8 vs 7.2 months; P < .001).25
and CB units.14 However, many stud­ ies have linked eco­ nomic The fol­low­ing eco­nomic indi­ca­tors have been asso­ci­ated
issues with race and eth­nic­ity to address access to allo-HCT.9,15 with TR (HCTs/10 mil­lion), other trans­plant indi­ca­tors,4 and out­
comes26-28: (1) human devel­op­ment index (HDI), a com­pos­ite var­
Social fac­tors i­able containing infor­ma­tion about life expec­tancy, edu­ca­tion,
Geographic dis­tance. The dis­tance between a patient and an and gross national income (GNI); (2) health care expen­di­ture
HCT facil­ity appears to be an impor­tant fac­tor to HCT access.16 (HCE) per cap­ita; (3) GNI per cap­ita; (4) World Bank categories:
To ensure proper fol­low-up care, allo-HCT patients with lim­ited HICs, HMICs, LMICs, and LICs.
geo­graphic access must decide whether to make numer­ ous Gratwohl et al4 and Niederwieser et al5 dem­on­strated that
long trips or relo­cate near the HCT facil­ity, either of which can be global num­bers of allo-HCT are still increas­ing. By the end of
a sig­nif­i­cant finan­cial bur­den. There are conflicting results from 2024, almost 1 mil­lion allo-HCT will be performed in 76 of the
published data on the role of dis­tance from the trans­plant cen­ter 194 WHO mem­ber states, yet no HCT was performed in coun-
or the impact of urban/rural areas on out­comes after allo-HCT.9,17 tries with a pop­u­la­tion of <300 000 inhab­i­tants, sur­face area
However, in a recent study, poor access to care, defined as low <700 km2, and GNI < US$1260/per­son. TR was higher in countries
socio­eco­nomic sta­tus and a far dis­tance to the trans­plant cen­ter, with greater gross domes­tic prod­uct, GNI, and HCE per per­
was asso­ci­ated with increased 1-year mor­tal­ity.18 son, higher HDI, more donors, and larger CB banks.4 The asso­
ci­a­tion between trans­plant and mac­ro­eco­nomic fac­tors var­ied
Patient/fam­ily atti­tudes and avail­abil­ity of care­giv­ers. Family sub­stan­tially between donor types, WHO regions, and World
and patient psy­cho­log­i­cal fac­tors may be impor­tant for HCT Bank categories. Macroeconomic resources gen­er­ally showed
access and out­come. To our knowl­edge no study has eval­u­ated higher asso­ci­at­ions with auto-HCT than allo-HCT, with no sub­
how psy­cho­log­ic ­ al fac­tors affect access, yet diag­noses of depres­ stan­tial dif­fer­ences for related- or UD-HCT. Ease of access to
sion, anx­i­ety, or post­trau­matic stress dis­or­der fol­low­ing HCT were HCT as deter­mined by TD gen­er­ally showed a higher asso­ci­a­
asso­ci­ated with sub­op­ti­mal health out­comes and increased mor­ tion for auto- than allo-HCT. Generally, asso­ ci­
a­tions between
tal­ity.19-21 Caregiver avail­abil­ity may also be a bar­rier to patients mac­ ro­
eco­
nomic fac­ tors and TR were higher in the Americas
con­sid­er­ing and pro­ceed­ing to HCT.22 Parents/guard­ians are often and SEAR/WPR Regions. Considering the World Bank catego-
the nat­u­ral care­giv­ers for their chil­dren, but for adult patients, the ries, asso­ci­a­tions were gen­er­ally low, with a stron­ger impact of

268  |  Hematology 2021  |  ASH Education Program


Table 1. Increase in HCT num­bers from 2006 to 2016 according to trans­plant type (allo­ge­neic and autol­o­gous), donor type, and source4-6

Year 2006 2007 2008 2009 2010 2011 2012 2013 2014 2015 2016
Allogeneic HCT 20,333 21,904 23,989 25,539 27,189 29,815 31,926 33,572 35,333 35,897 38,425
Related 11,002 11,611 12,362 12,641 12,945 14,303 15,493 16,305 17,705 18,515 20,594
 Identical n.a. 10,107 10,599 10,624 10,912 11,751 12,322 12,475 12,579 12,511 12,463
 Nonidentical n.a. 1,504 1,763 2,017 2,033 2,552 3,171 3,830 5,126 6,004 8,131
 CB 0 102 92 131 149 143 251 239 298 358 117

Prakash Singh Shekhawat


Unrelated 9,331 10,293 11,627 12,898 14,244 15,512 16,433 17,267 17,628 17,382 17,831
 CB 1,722 2,031 2,266 2,508 2,804 2,927 2,859 2,787 2,568 2,474 2,477
Autologous HCT 26,230 26,805 27,547 30,081 31,730 33,756 36,220 37,464 38,188 41,698 44,293
Total 46,563 48,709 51,536 55,620 58,919 63,571 68,146 71,036 73,521 77,595 82,718
n.a.

Factors affect­ing global access to allotransplant  |  269


Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/264/1851868/264rocha.pdf by guest on 13 December 2021
finan­cial resources on the LMIC cat­e­gory and TD for auto-HCT in health sys­tem (pub­lic vs pri­vate or uni­ver­sal vs non­uni­ver­sal) and
the HMIC cat­e­gory. access to allo-HCT.
Little data are avail­­able on whether eco­nomic fac­tors may HCT is costly and may drain con­sid­er­able resources from pri­
also explain the TR dif­fer­ences found among regions of a par­ mary care, mater­nal and child­hood care, or coun­try­wide vac­ci­na­
tic­u­lar coun­try. This microlevel sce­nario gen­er­ally involves tion pro­grams. On the other hand, car­ing for patients with endemic
the health care sys­tem and eco­nom­ics, while clin­i­cal dif­fer­ dis­eases (eg, hemo­glo­bin­op­a­thies in most emerg­ing countries)
ences should not be expected to cause dif­fer­ent TR among may be costly over time, and HCT may be more cost-effec­tive in
regions within a coun­try with a pub­lic health care sys­tem. the long run.33 Thus, when it comes to stra­te­gic pri­or­it­ies, coun-
An inter­est­ing Span­ish study showed that auto-HCT TR was tries should tai­lor the implementation of ter­tiary care facil­i­ties
asso­ci­ated with only TD, whereas mac­ro­eco­nomic deter­mi­ according to socio­eco­nomic con­di­tions and coun­try-spe­cific dis­
nants exerted a strong influ­ence in the case of allo-TR.29 In eases. It is also crit­i­cal to eval­u­ate the clin­ic
­ al/eco­nomic effec­
par­tic­u­lar, an increase of €1000 in gross domes­tic prod­uct per tive­ness and sus­tain­abil­ity of such an endeavor.34 Unfortunately,
cap­ita would cause an aver­age increase of 1.4 trans­plants per there are scarce data on what type of donor or graft-ver­ sus-
1 ­mil­lion inhab­i­tants.29 Therefore, there may be inequal­ity in host dis­ease (GVHD) pro­phy­laxis could be most cost-effec­tive in

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/264/1851868/264rocha.pdf by guest on 13 December 2021


access to HCT by region due to per cap­ita income, TD, and devel­op­ing econ­o­mies, an issue that urgently war­rants fur­ther
pub­lic hos­pi­tal expen­di­ture. These find­ings may be use­ful to research.
health author­i­ties in mak­ing deci­sions both at the mac­ro­eco­
nomic and local lev­els. Referral of patients to trans­plant and posttransplant care
The com­ plex inter­play of patients, refer­ ring phy­ si­
cians, pay­
Macro- and micro­eco­nomic fac­tors: impact on out­comes of ors, and trans­plant cen­ter-related fac­tors have been pre­vi­ously
allo-HCT.  Studies ana­lyz­ing socio­eco­nomic fac­tors and out­ described.35 These fac­tors need to be com­pre­hen­sively stud­ied
comes after allo-HCT are restricted to patients with suf­fic ­ ient to deliver opti­mal care. Both pay­ors and accred­i­ta­tion agencies
resources to pro­ceed to HCT and might not be rep­re­sen­ta­tive should also attempt to ele­vate the stan­dards of care affect­ing
of all­trans­plant can­di­dates. Hence, although research on out­ trans­plant out­comes after dis­charge from the trans­plant cen­ter.35
comes fol­low­ing allo-HCT in dis­ad­van­taged groups is impor­tant,
it is equally rel­e­vant to ensure that patients in these groups actu­ National and inter­na­tional reg­u­la­tions
ally have access to trans­plan­ta­tion. In many health care set­tings, bench­mark­ing for com­plex pro­
In 2010 we eval­u­ated the asso­ci­a­tion of HDI with rates and ce­dures has become a man­da­tory require­ment by stake­hold­
out­comes of allo-HCT for acute leu­ke­mia in Euro­pean countries. ers to assure clin­i­cal per­for­mance, cost-effec­tive­ness, and
Allo-HCTs performed in countries belong­ing to the upper HDI patient safety.36 Benchmarking has also been inte­grated into
cat­e­gory were asso­ci­ated with higher leu­ke­mia-free sur­vival the Foundation for the Accreditation of Cellular Therapy-Joint
com­ pared to other categories.27 In another Euro­ pean study, Accreditation Committee of the International Society for Cel-
lower cur­rent HCE and HDI were asso­ci­ated with a decreased lular Therapy and the Euro­pean Society for Blood and Mar-
prob­a­bil­ity of OS in allo-HCT recip­i­ents with acute lym­pho­blas­tic row Transplantation qual­ity man­age­ment stan­dards and has
leu­ke­mia.26 An impor­tant Center for International Blood and Mar- been established mainly for devel­oped countries but not for
row Transplant Research study ana­lyz­ing 11 261 allo-HCT recip­ resource-constrained nations.36 Developing mean­ing­ful bench­
i­ents with acute lym­pho­blas­tic leu­ke­mia across 38 countries mark­ing sys­tems for countries with sim­i­lar mac­ro­eco­nomic
showed that trans­plants performed in countries within the low­ fac­tors may help address and cor­rect underperformance.
est HDI quar­tile were asso­ci­ated with an OS infe­rior to countries
in the highest HDI quar­tile.30 Establishing new HCT pro­grams
The asso­ci­a­tion between coun­try-level mac­ro­eco­nomic indi­ Table 2 sum­ ma­rizes impor­ tant steps in establishing new or
ca­tors and out­comes after allo-HCT needs fur­ther eval­u­at­ion. opti­miz­ing established HCT pro­grams. Close coop­er­a­tion with
HCT in lower-income set­tings may be asso­ci­ated with decreased health author­i­ties, pol­i­ti­cians, and phy­si­cians is needed to
avail­­able resources, espe­cially early post HCT.31 In fact, prior work ensure equal­ity of treat­ment around the world. It is essen­tial to
has documented exten­sive var­i­abil­ity in the pro­vi­sion of sup­port­ allo­cate funds for this treat­ment and have sup­port from regional
ive prac­tices across countries.30 It is pos­si­ble that care deliv­ery- and global sci­en­tific soci­e­ties in asso­ci­a­tion with the WHO. Fur-
related fac­tors may have con­trib­uted to the high pro­por­tion of thermore, HCT-spe­ cific min­ i­
mum require­ ments are needed.
deaths from infec­tions seen in HMIC/LMIC/LIC set­tings (eg, lack The Transplant Center and Recipient Issues Standing Commit-
of ade­quate inten­sive care sup­port).25,30,32 Alternatively, worse tee of the WBMT orga­nized a struc­tured review and ana­lyzed,
mac­ro­eco­nomic indi­ca­tors may reflect deficiencies in train­ing, described, and scored (by inde­pen­dent trans­plant phy­si­cians)
expe­ri­ence, or staffing mod­els in cen­ters in resource-constrained min­ i­
mum require­ ments to estab­ lish an HCT pro­ gram.37 This
set­tings. On a patient level, lower socio­eco­nomic sta­tus may be struc­tured set of rec­om­men­da­tions guides the pri­or­i­ti­za­tion to
asso­ci­ated with adverse out­comes owing to chronic stress, low estab­lish a trans­plant pro­gram and set the path for expan­sion
edu­ca­tional level, unaffordability of med­i­ca­tions, and dif­fi­cul­ties and fur­ther devel­op­ment.37
in reaching the trans­plant cen­ter.
How to improve access to allo-HCT
Health care sys­tems Access to allo-HCT is depen­dent on the mul­ti­ple fac­tors reported
Health care is a sec­tor where gov­ern­ments have a lead­ing role. above and sum­ ma­ rized in Figure 6. Improvements in access
Access to and out­comes fol­low­ing allo-HCT depend on how can be obtained by eval­u­at­ing the need for HCT in indi­vid­ual
much a gov­ern­ment invests in gen­eral health and ter­tiary care. countries and the ways in which to improve. The eas­i­est way
To our knowl­edge, there are no data com­par­ing the type of would be to improve the TR in existing cen­ters and estab­lish

270  |  Hematology 2021  |  ASH Education Program


Table 2. How to improve access to HCT

Target Topic Actions


Benchmarking activ­i­ties among Global HCT activ­ity reports Biannual sur­vey since 20064,6,47-50
countries and regions
Starting new pro­grams Alerting health author­i­ties and pol­i­ti­cians about the Organization of WBMT work­shops in coop­er­a­tion with
need for pro­grams in countries with low HCT activ­ity the WHO51
Essential med­i­ca­tion Published pre­vi­ously52
Training of phy­si­cians, nurses, tech­ni­cians, and data Scientific soci­e­ties; accredited trans­plant
man­ag­ers cen­ters
Infrastructure Define essen­tial infra­struc­ture37,53
Site visit from expe­ri­enced phy­si­cians Role of sci­en­tific soci­e­ties

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/264/1851868/264rocha.pdf by guest on 13 December 2021


Financial aspects Optimize treat­ment
Twinning and telemedicine Supervisory telemedicine42
Optimizing existing pro­grams Outcome reg­is­tries Establish out­come reg­is­tries
Analysis of dif­fer­ent tech­niques54
Accreditation Liaise with JACIE/FACT
Utilization of HCT world­wide Analyzing inci­dence (tumor reg­is­tries) and HCT activ­i­ties
for each dis­ease in regions and countries55
Establishing alter­nate donor reg­is­tries Describe chal­lenges in devel­op­ing countries56
Establishing clin­i­cal stud­ies Structures for local reg­is­tries, noninterventional,
interventional stud­ies
FACT, Foundation for the Accreditation of Cellular Therapy; JACIE, Joint Accreditation Committee of the International Society for Cellular Therapy.

new trans­plant cen­ters in regions with low TD. This eval­u­a­tion appli­ca­tions rep­re­sent a unique oppor­tu­nity to nar­row the gap
pro­vi­des com­par­i­sons for bench­mark­ing and some hints on how of access to HCT between devel­oped and devel­op­ing nations.
to cre­ate or opti­mize trans­plant pro­grams. New tech­nol­o­gies Telehealth has been asso­ci­ated with decreas­ing costs, gains
may help in this endeavor. in pro­duc­ tiv­
ity, patient adher­ence, and improved access to
health care.38 Since the 2000s there have been a few reports
Role of new tech­nol­o­gies on the use of telehealth in HCT recip­i­ents,39,40 but the COVID-19
Even in LICs and LMICs, wide­ spread adop­ tion of the mobile pan­demic really caused the field to gain trac­tion world­wide,
phone, invest­ments in elec­tronic med­i­cal records, devel­op­ments includ­ing in the devel­op­ing world. At our pub­lic insti­tu­tion in
in cloud com­ put­
ing, and emer­ gent mobile health (mHealth) Brazil (V.R., G.F.), a recent sur­vey among 232 HCT recip­i­ents

Prakash
Figure 6. Increasing access and Singh Shekhawat
possibly outcomes to allo-HCT using new technologies: an international perspective.

Factors affect­ing global access to allotransplant  |  271


dur­ing the COVID-19 pan­demic showed that one-third of them Conflict-of-inter­est dis­clo­sure
spent >120 min­utes to travel to and from the out­pa­tient clinic, Vanderson Rocha: no com­pet­ing finan­cial inter­ests to declare.
and 42% had a cost >US $10.00 (equiv­a­lent to 5% of the min­i­ Giancarlo Fatobene: no com­pet­ing finan­cial inter­ests to declare.
mum wage/month). Approximately half of them had engaged Dietger Niederwieser: no com­pet­ing finan­cial inter­ests to declare.
in at least 1 pre­vi­ous inter­ac­tion via telehealth with our cen­ter,
and 91% of these patients con­sid­ered it a good expe­ri­ence.41 Off-label drug use
Although not fit for all­clin­i­cal sce­nar­ios, telehealth may be effi­ Vanderson Rocha: no off-label drug use is discussed.
cient and com­ple­men­tary to in-per­son inter­ac­tions with HCT Giancarlo Fatobene: no off-label drug use is discussed.
patients, may allow a cost reduc­tion for patients, and may be Dietger Niederwieser: no off-label drug use is discussed.
espe­cially use­ful in the long-term fol­low-up of HCT sur­vi­vors,40
which often requires insti­tu­tions expe­ri­enced in the man­age­ Correspondence
ment of chronic GVHD and late com­pli­ca­tions. Telementoring is Vanderson Rocha, University of São Paulo, Rua Dr Ovidio Pires de
also a prom­is­ing approach to implementing allo-HCT pro­grams Campos, São Paulo, Brazil 05403-010; e-mail: vanderson​­.rocha​
in that con­text.42 ­@hc​­.fm​­.usp​­.br.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/264/1851868/264rocha.pdf by guest on 13 December 2021


Wearable devices (eg, the Apple Watch) rep­re­sent another
mHealth modal­ ity increas­ ingly assim­ i­
lated in every­ day life. References
These devices can be con­tin­u­ous or inter­mit­tently cap­ture 1. Majhail NS, Omondi NA, Denzen E, Mur­phy EA, Rizzo JD. Access to hema­to­
poi­etic cell trans­plan­ta­tion in the United States. Biol Blood Marrow Trans-
bio­log­ i­
cal data, and recent expe­ ri­ences have been tested
plant. 2010;16(8):1070-1075.
in can­cer and HCT sur­vi­vors.43 Wearable devices along with 2. Warwick RM, Chapman J, Pruett TL, Wang H. Globally con­sis­tent cod­ing
telehealth approaches reduced hos­pi­tal readmissions fol­low­ sys­tems for med­i­cal prod­ucts of human ori­gin. Bull World Health Organ.
ing liver trans­plan­ta­tion.44 Coupling the mas­ sive data gen­ 2013;91(5):314-314A.
er­ated by these tech­nol­o­gies, elec­tronic med­i­cal records, 3. World Health Organization. WHO guid­ing prin­ci­ples on human cell, tis­sue
and organ trans­plan­ta­tion. Accessed 19 Octo­ber 2021. https:​­/​­/www​­.who​
and bio­mark­ers with arti­fi­cial intel­li­gence may poten­tially
­.int​­/transplantation​­/Guiding_PrinciplesTransplantation_WHA63​­.22en​­.pdf.
pre­dict trans­ plant com­ pli­ca­tions such as febrile neutrope- 4. Gratwohl A, Pasquini MC, Aljurf M, et al; Worldwide Network for Blood and
nia and GVHD hours or days in advance.45 In fact, arti­fi­cial Marrow Transplantation. One mil­lion haemopoietic stem-cell trans­plants: a
intel­li­gence has been used to improve the diag­no­sis of birth ret­ro­spec­tive obser­va­tional study. Lancet Haematol. 2015;2(3):e91-100.
5. Niederwieser D, Baldomero H, Bazuaye N, et  al. One and a half mil­lion
asphyxia and dia­betic ret­i­nop­a­thy and pro­vide treat­ment rec­
hema­to­poi­etic stem cell trans­plants: con­tin­u­ous and dif­fer­en­tial improve­
om­men­da­tions in can­cer in dif­fer­ent low-income countries.46 ment in world­wide access with the use of non-iden­ti­cal fam­ily donors. Pub-
With decreas­ing costs, these tech­nol­o­gies could expand out­ lished online 12 August 2021. Haematologica.
pa­tient allo-HCT and allow bet­ter uti­li­za­tion of hos­pi­tal HCT 6. Niederwieser D, Baldomero H, Szer J, et al. Hematopoietic stem cell trans­
beds in regions where trans­plant waiting lists are a real­ity. plan­ta­tion activ­ity world­wide in 2012 and a SWOT anal­y­sis of the World-
wide Network for Blood and Marrow Transplantation Group includ­ing the
Prioritization of trans­plant can­di­dates on waiting lists may also
global sur­vey. Bone Marrow Transplant. 2016;51(6):778-785.
be assisted by intel­li­gent algo­rithms that take into account 7. Gragert L, Eapen M, Williams E, et al. HLA match like­li­hoods for hema­to­poi­
per­sonal and dis­ease char­ac­ter­is­tics, pretransplant workup etic stem-cell grafts in the U.S. reg­is­try. N Engl J Med. 2014;371(4):339-348.
sched­ul­ing, local trans­plant cen­ter per­for­mance, and evi­ 8. Nunes K, Aguiar VRC, Silva M, et al. How ances­try influ­ences the chances
of find­ing unre­lated donors: an inves­ti­ga­tion in admixed Brazilians. Front
dence-based data.
Immunol. 2020;11(6 Novem­ber):584950.
Although tech­no­log­i­cal advances offer unprec­e­dented oppor­ 9. Paulson K, Brazauskas R, Khera N, et al. Inferior access to allo­ge­neic trans­plant
tu­ni­ties to enable access to HCT, the val­i­da­tion of effec­tive solu­ in dis­ad­van­taged pop­u­la­tions: a Center for International Blood and Marrow
tions demands high-qual­ ity data sets, internet con­ nec­tiv­
ity, Transplant Research anal­y­sis. Biol Blood Marrow Transplant. 2019;25(10):
infor­ma­tion tech­nol­ogy infra­struc­ture, and clin­i­cal tri­als, which 2086-2090.
10. Milone G, Sacchi N, Gallina A, et al. Access to alter­na­tive donor hema­to­poi­
are time- and cost-con­sum­ing. Hence, stake­hold­ers must ensure
etic search and trans­plan­ta­tion for acute leu­ke­mia in dif­fer­ent macro-regions
that actions are taken to guide sus­tain­able, equi­ta­ble tech­nol­ of Italy: a GITMO/IBMDR study. Bone Marrow Transplant. 2018;53(3):291-299.
ogy implementation in the HCT realm to ful­fill its prom­ises. 11. Hershenfeld SA, Matelski J, Ling V, Paterson M, Cheung M, Cram P. Utili-
sation and out­comes of allo­ge­neic hema­to­poi­etic cell trans­plan­ta­tion in
Ontario, Canada, and New York State, USA: a pop­u­la­tion-based ret­ro­spec­
Research agenda for resource-constrained countries tive cohort study. BMJ Open. 2020;10(10):e039293.
Most of the cur­rent evi­dence in HCT has been gen­er­ated in HICs 12. Bhatt VR, Chen B, Gyawali B, Lee SJ. Socioeconomic and health sys­tem fac­
tors asso­ci­ated with lower uti­li­za­tion of hema­to­poi­etic cell trans­plan­ta­tion
and may not be appli­ ca­ble to resource-constrained set­ tings.
in older patients with acute mye­loid leu­ke­mia. Bone Marrow Transplant.
Many clin­i­cal chal­lenges faced by devel­op­ing countries (ie, the 2018;53(10):1288-1294.
major­ity of the world’s pop­u­la­tion) are under­rep­re­sented in the 13. Joshua TV, Rizzo JD, Zhang MJ, et  al. Access to hema­to­poi­etic stem cell
HCT lit­er­a­ture (eg, evidenced-based man­age­ment of waiting trans­plan­ta­tion: effect of race and sex. Cancer. 2010;116(14):3469-3476.
lists, peritransplant mul­ti­drug-resis­tant bac­te­ria, trop­i­cal dis­ 14. Barker JN, Boughan K, Dahi PB, et al. Racial disparities in access to HLA-
matched unre­lated donor trans­plants: a pro­spec­tive 1312-patient anal­y­sis.
eases). A research agenda, with a par­tic­u­lar focus on clin­i­cal tri­
Blood Adv. 2019;3(7):939-944.
als, directed to and conducted in resource-constrained countries 15. Bona K, Brazauskas R, He N, et  al. Neighborhood pov­erty and pedi­at­ric
is essen­tial to effec­tively tackle these issues. Initiatives fos­ter­ing allo­ge­neic hema­to­poi­etic cell trans­plan­ta­tion out­comes: a CIBMTR anal­y­
inter­na­tional coop­er­a­tion, funding, and local research capac­ity, sis. Blood. 2021;137(4):556-568.
such as the Amer­i­can Society of Hematology Global Research 16. Delamater PL, Uberti JP. Geographic access to hema­to­poi­etic cell trans­plan­
ta­tion ser­vices in the United States. Bone Marrow Transplant. 2016;51(2):
Award and Clinical Research Training Institutes, should be mul­ 241-248.
ti­plied to advance access to HCT and other cel­lu­lar ther­a­pies 17. Khera N, Gooley T, Flowers MED, et al. Association of dis­tance from trans­
world­wide. plan­ta­tion cen­ter and place of res­i­dence on out­comes after allo­ge­neic

272  |  Hematology 2021  |  ASH Education Program


hema­to­poi­etic cell trans­plan­ta­tion. Biol Blood Marrow Transplant. 2016; the Euro­pean Society for Blood and Marrow Transplantation (EBMT) and
22(7):1319-1323. the Joint Accreditation Committee of ISCT and EBMT (JACIE). Bone Marrow
18. Jamy O, Chen A, Battles K, et al. Impact of access to care on 1-year mor­ Transplant. 2020;55(4):681-694.
tal­ity fol­low­ing allo­ge­neic blood or mar­row trans­plan­ta­tion. Bone Marrow 37. Pasquini MC, Srivastava A, Ahmed SO, et  al. Worldwide Network for
Transplant. 2021;56(6):1364-1372. Blood and Marrow Transplantation (WBMT) rec­om­men­da­tions for estab-
19. Amonoo HL, Barclay ME, El-Jawahri A, Traeger LN, Lee SJ, Huffman JC. lishing a hema­to­poi­etic cell trans­plan­ta­tion pro­gram (part I): min­i­mum
Positive psy­cho­log­i­cal con­structs and health out­comes in hema­to­poi­etic require­ments and beyond. Hematol Oncol Stem Cell Ther. 2020;13(3):131-
stem cell trans­plan­ta­tion patients: a sys­tem­atic review. Biol Blood Marrow 142.
Transplant. 2019;25(1):e5-e16. 38. Snoswell CL, Taylor ML, Comans TA, Smith AC, Gray LC, Caffery LJ. Deter-
20. El-Jawahri A, Traeger L, Greer JA, et al. Effect of inpa­tient pal­li­a­tive care dur­ mining if telehealth can reduce health sys­tem costs: scop­ing review. J Med
ing hema­to­poi­etic stem-cell trans­plant on psy­cho­log­i­cal dis­tress 6 months Inter­net Res. 2020;22(10):e17298.
after trans­plant: results of a ran­dom­ized clin­i­cal trial. J Clin Oncol. 2017; 39. DuHamel KN, Mosher CE, Winkel G, et  al. Randomized clin­ i­
cal trial of
35(32):3714-3721. tele­phone-admin­is­tered cog­ni­tive-behav­ioral ther­apy to reduce post-
21. Ehrlich KB, Miller GE, Scheide T, et al. Pre-trans­plant emo­tional sup­port is trau­matic stress dis­or­der and dis­tress symp­toms after hema­to­poi­etic
asso­ci­ated with lon­ger sur­vival after allo­ge­neic hema­to­poi­etic stem cell stem-cell trans­plan­ta­tion. J Clin Oncol. 2010;28(23):3754-3761.
trans­plan­ta­tion. Bone Marrow Transplant. 2016;51(12):1594-1598. 40. Syrjala KL, Yi JC, Artherholt SB, et al. An online ran­dom­ized con­trolled trial,

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/264/1851868/264rocha.pdf by guest on 13 December 2021


22. Preussler JM, Mau LW, Majhail NS, et al. Caregiver avail­abil­ity and patient with or with­out prob­lem-solv­ing treat­ment, for long-term can­cer sur­vi­vors
access to hema­to­poi­etic cell trans­plan­ta­tion: social worker per­spec­tives after hema­to­poi­etic cell trans­plan­ta­tion. J Cancer Surviv. 2018;12(4):560-
inform prac­tice. Support Care Cancer. 2019;27(11):4253-4264. 570.
23. Frey P, Stinson T, Siston A, et al. Lack of care­giv­ers lim­its use of out­pa­tient 41. Cordeiro A, Fatobene G, Mariano LCB, et al. Telehealth in hema­to­poi­etic
hema­to­poi­etic stem cell trans­plant pro­gram. Bone Marrow Transplant. cell trans­plan­ta­tion: per­spec­tive from patients at a pub­lic hos­pi­tal in Brazil.
2002;30(11):741-748. Blood. 2020;136(suppl 1):26.
24. Sundaramurthi T, Wehrlen L, Friedman E, Thomas S, Bevans M. Hemato- 42. Frutos C, Enciso ME, von Glasenapp A, Quiroz A, Batista J, Niederwieser D.
poietic stem cell trans­plan­ta­tion recip­i­ent and care­giver fac­tors affect­ing Bridging the gap using telemedicine: opti­miz­ing an existing autol­o­gous
length of stay and readmission. Oncol Nurs Forum. 2017;44(5):571-579. hema­to­poi­etic SCT unit into an allo­ge­neic hema­to­poi­etic SCT unit in Para-
25. Silveira DRA, Coelho-Silva JL, Silva WF, et  al. A mul­ti­cen­ter com­par­a­tive guay with the help of the WBMT. Blood Adv. 2019;3(suppl 1):45-47.
acute mye­loid leu­ke­mia study: can we explain the dif­fer­ences in the out­ 43. Mussetti A, Salas MQ , Condom M, et al. Use of telehealth for domi­cil­i­ary
comes in resource-constrained set­tings? Leuk Lymphoma. 2021;62(1):147- fol­low-up after hema­to­poi­etic cell trans­plan­ta­tion dur­ing the COVID-19
157. pan­demic: pro­spec­tive pilot study. JMIR Form Res. 2021;5(3):e26121.
26. Giebel S, Labopin M, Ibatici A, et al. Association of mac­ro­eco­nomic fac­tors 44. Lee TC, Kaiser TE, Alloway R, Woodle ES, Edwards MJ, Shah SA. Telemedi-
with nonrelapse mor­tal­ity after allo­ge­neic hema­to­poi­etic cell trans­plan­ cine based remote home mon­i­tor­ing after liver trans­plan­ta­tion: results of a
ta­tion for adults with acute lym­pho­blas­tic leu­ke­mia: an anal­y­sis from the ran­dom­ized pro­spec­tive trial. Ann Surg. 2019;270(3):564-572.
Acute Leukemia Working Party of the EBMT. Oncologist. 2016;21(3):377- 45. Abbasi J. Wearable dig­i­tal ther­mom­e­ter improves fever detec­tion. JAMA.
383. 2017;318(6):510.
27. Giebel S, Labopin M, Ehninger G, et al; Acute Leukemia Working Party of 46. Owoyemi A, Owoyemi J, Osiyemi A, Boyd A. Artificial intel­ li­
gence for
the Euro­pean Group for Blood and Marrow Transplantation. Association healthcare in Africa. Front Digit Heal. 2020;2(July):6.
of Human Development Index with rates and out­comes of hema­to­poi­ 47. Gratwohl A, Baldomero H, Gratwohl M, et al; Worldwide Network of Blood
etic stem cell trans­ plan­ta­
tion for patients with acute leu­ ke­mia. Blood. and Marrow Transplantation. Quantitative and qual­i­ta­tive dif­fer­ences in use
2010;116(1):122-128. and trends of hema­to­poi­etic stem cell trans­plan­ta­tion: a global obser­va­
28. Frankiewicz A, Peczynski C, Giebel S, et al. Association of coun­try-spe­cific tional study. Haematologica. 2013;98(8):1282-1290.
socio­eco­nomic fac­tors with sur­vival of patients who expe­ri­ence severe 48. Jaimovich G, Martinez Rolon J, Baldomero H, et al. Latin America: the next
clas­sic acute graft-vs.-host dis­ease after allo­ge­neic hema­to­poi­etic cell region for haematopoietic trans­plant prog­ress. Bone Marrow Transplant.
trans­plan­ta­tion: an anal­y­sis from the Transplant Complications Working 2017;52(5):671-677.
Party of the EBMT. Front Immunol. 2020;11(July 23):1537. 49. Baldomero H, Aljurf M, Zaidi SZA, et al; Eastern Med­i­ter­ra­nean Blood and
29. Espigado I, Ortega-Ortega M, Montero-Granados R, Rodriguez-Torres N, Marrow Transplantation Group; Afri­ can Blood and Marrow Transplanta-
Márquez-Malaver FJ. Differences in stem cell trans­plan­ta­tion activ­ity among tion Group; Worldwide Network for Blood and Marrow Transplantation.
regions in Spain: an eco­ nomic expla­ na­
tion. Bone Marrow Transplant. Narrowing the gap for hema­to­poi­etic stem cell trans­plan­ta­tion in the East-
2016;51(11):1537-1539. Med­i­ter­ra­nean/Afri­can region: com­par­i­son with global HSCT indi­ca­tions
30. Wood WA, Brazauskas R, Hu ZH, et al. Country-level mac­ro­eco­nomic indi­ and trends. Bone Marrow Transplant. 2019;54(3):402-417.
ca­tors pre­dict early post-allo­ge­neic hema­to­poi­etic cell trans­plan­ta­tion 50. Gratwohl A, Baldomero H, Aljurf M, et al; Worldwide Network of Blood and
sur­vival in acute lym­pho­blas­tic leu­ke­mia: a CIBMTR anal­y­sis. Biol Blood Marrow Transplantation. Hematopoietic stem cell trans­plan­ta­tion: a global
Marrow Transplant. 2018;24(9):1928-1935. per­spec­tive. JAMA. 2010;303(16):1617-1624.
31. Hong S, Brazauskas R, Hebert KM, et al. Community health sta­tus and out­ 51. Harif M, Weisdorf D, Novitzky N, et al. Special report: sum­mary of the first
comes after allo­ge­neic hema­to­poi­etic cell trans­plan­ta­tion in the United meet­ing of Afri­can Blood and Marrow Transplantation (AfBMT) group, Casa-
States. Cancer. 2021;127(4):609-618. blanca, Morocco, April 19–21, 2018 held under the aus­pices of the World-
32. da Silva WF, da Rosa LI, Seguro FS, et al. Salvage treat­ment for refrac­tory wide Network for Blood and Marrow Transplantation (WBMT). Hematol
or relapsed acute mye­loid leu­ke­mia: a 10-year sin­gle-cen­ter expe­ri­ence. Oncol Stem Cell Ther. 2020;13(4):202-207.
Clinics. 2020;75:1-8. 52. El Fakih R, Greinix H, Koh M, et  al. Worldwide Network for Blood and
33. Arnold SD, Jin Z, Sands S, Bhatia M, Kung AL, Satwani P. Allogeneic hema­to­ Marrow Transplantation (WBMT) rec­om­men­da­tions regard­ing essen­
poi­etic cell trans­plan­ta­tion for chil­dren with sickle cell dis­ease is ben­e­fi­cial tial med­ i­
ca­tions required to estab­ lish an early stage hema­ to­poi­etic
and cost-effec­tive: a sin­gle-cen­ter anal­y­sis. Biol Blood Marrow Transplant. cell trans­plan­ta­tion pro­gram. Transplant Cell Ther. 2021;27(3):267.e1-
2015;21(7):1258-1265. 267.e5.
34. Chaudhri NA, Aljurf M, Almohareb FI, et al. Establishing an autol­o­gous 53. Aljurf M, Weisdorf D, Hashmi SK, et al. Worldwide Network for Blood and
ver­sus allo­ge­neic hema­to­poi­etic cell trans­plant pro­gram in nations with Marrow Transplantation (WBMT) rec­ om­men­
da­tions for establishing a
emerg­ing econ­o­mies. Hematol Oncol Stem Cell Ther. 2017;10(4):173- hema­to­poi­etic stem cell trans­plan­ta­tion pro­gram in countries with lim­ited
177. resources (part II): clin­i­cal, tech­ni­cal and socio-eco­nomic con­sid­er­ations.
35. Majhail NS, Jagasia M. Referral to trans­plant cen­ter for hema­to­poi­etic cell Hematol Oncol Stem Cell Ther. 2020;13(1):7-16.
trans­plan­ta­tion. Hematol Oncol Clin North Am. 2014;28(6):1201-1213. 54. Yoshimi A, Baldomero H, Horowitz M, et al; Worldwide Network of Blood
36. Snowden JA, Saccardi R, Orchard K, et al. Benchmarking of sur­vival out­ and Marrow Transplantation. Global use of periph­eral blood vs bone mar­row
comes fol­low­ing haematopoietic stem cell trans­plan­ta­tion: a review of as source of stem cells for allo­ge­neic trans­plan­ta­tion in patients with bone
existing pro­cesses and the intro­duc­tion of an inter­na­tional sys­tem from mar­row fail­ure. JAMA. 2016;315(2):198-200.

Prakash Singh Shekhawat


Factors affect­ing global access to allotransplant  |  273
55. Cowan AJ, Baldomero H, Atsuta Y, et al. The global state of hema­to­poi­etic ing alter­nate donor reg­is­tries. Bone Marrow Transplant. 2019;54(8):1179-
cell trans­plan­ta­tion for mul­ti­ple mye­loma: an anal­y­sis of the Worldwide 1188.
Network of Blood and Marrow Transplantation data­base and the global
bur­den of dis­ease study. Biol Blood Marrow Transplant. 2020;26(12):2372-
2377.
56. Aljurf M, Weisdorf D, Alfraih F, et  al. Worldwide Network for Blood and © 2021 by The Amer­i­can Society of Hematology
Marrow Transplantation (WBMT) spe­cial arti­cle, chal­lenges fac­ing emerg­ DOI 10.1182/hema­tol­ogy.2021000258

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/264/1851868/264rocha.pdf by guest on 13 December 2021

274  |  Hematology 2021  |  ASH Education Program


HOW CAN WE ENSURE THAT EVERYONE WHO NEEDS A TRANSPLANT CAN GET ONE ?

Increasing access to allotransplants in the


United States: the impact of race, geography,
and socioeconomics

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/275/1852104/275hong.pdf by guest on 13 December 2021


Sanghee Hong1 and Navneet S. Majhail2
Department of Hematology and Oncology, University Hospitals, Case Western Reserve University, Cleveland, OH; and 2Blood and Marrow
1

Transplant Program, Taussig Cancer Center, Cleveland Clinic, Cleveland, OH

Allogeneic hematopoietic cell transplantation (HCT) is particularly susceptible to racial, socioeconomic, and geographic
disparities in access and outcomes given its specialized nature and its availability in select centers in the United States.
Nearly all patients who need HCT have a potential donor in the current era, but racial minority populations are less likely
to have an optimal donor and often rely on alternative donor sources. Furthermore, prevalent health care disparity fac-
tors are further accentuated and can be barriers to access and referral to a transplant center. Research has primarily
focused on defining and quantifying a variety of social determinants of health and their association with access to allo-
geneic HCT, with a focus on race/ethnicity and socioeconomic status. However, research on interventions is lacking and
is an urgent unmet need. We discuss the role of racial, socioeconomic, and geographic disparities in access to allogeneic
HCT, along with policy changes to address and mitigate them and opportunities for future research.

LEARNING OBJECTIVES
• Understand the association of race, geography, and socioeconomic status with access to allogeneic transplanta-
tion in the United States
• Highlight opportunities to evaluate, mitigate, and address social access-related barriers to allogeneic transplantation

be referred to a transplant center in Cleveland, which is


CLINICAL CASE 90 miles away from where she lives.
A 40-year-old African American woman from rural Ohio
has relapsed acute myeloid leukemia (AML). She is hos-
pitalized at a regional hospital close to home to receive Introduction
salvage chemotherapy. She is a single mother with a Although allogeneic HCT is potentially curative for many
10-year-old son and lives in an area that has one of the patients with high-risk hematologic malignancies and oth-
highest rates of poverty in Ohio. She does not have a car er diseases, it is a highly specialized and complex proce-
and uses public transportation. She used to earn an hourly dure that requires comprehensive clinical infrastructure to
wage as a waitress but has been unemployed with no facilitate referral, donor search, transplant hospitalization
health care benefits for the past year since being laid off and supportive care, and posttransplant follow-up. The
at the onset of the COVID epidemic. She had been feel- number of patients receiving allogeneic HCT continues to
ing very fatigued and had noticed spontaneous bruising increase in the US every year with improvements in tech-
for 4 weeks before the diagnosis of relapse; she was con- nology and supportive care, use of less intense condition-
cerned about leukemia recurrence but did not want to ing regimens that allow transplantation in older and frail
see her oncologist given the lack of health insurance and patients, and greater availability of suitable donors.1 How-
concern about paying medical bills. She does not have ever, it is also recognized that many patients who might
any immediate family in the vicinity. Her oncologist has otherwise benefit do not receive allogeneic HCT.2-7 Several
discussed an allogeneic hematopoietic cell transplanta- patient-specific barriers to accessing HCT have been iden-
tion (HCT) for her AML and the fact that she will need to tified. Historically, a lack of suitably HLA-matched donors
Prakash Singh Shekhawat
Access to allogeneic transplantation in the US | 275
Table 1. Sociodemographic fac­tors asso­ci­ated with access to allo­ge­neic HCT

Referencea Population Access var­i­able(s) Key find­ings


Jabo et al3 Age ≥15 years; patients with Age, race/eth­nic­ity, geog­ra­phy, Higher rate of HCT in patients aged ≤40 and in
ALL/AML in California Cancer SES mar­ried patients; women more likely to receive
Registry; 2003-2012 HCT for ALL; lower rates of HCT in His­panic
and non-His­panic Black patients; no asso­ci­a­
tion between dis­tance and HCT uti­li­za­tion; low
neigh­bor­hood quin­tile SES index asso­ci­ated
with less uti­li­za­tion of HCT
Dehn et al10 All ages; donor searches through Race/eth­nic­ity White patients more likely to receive HCT com­
Be the Match reg­is­try; 2016 pared to Black patients
Barker et al12 Age ≤70 years; sin­gle-cen­ter study Race/eth­nic­ity Patients of Euro­pean ances­try more likely to
of patients under­go­ing unre­ receive 8/8 HLA-MUD HCT trans­plant than

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/275/1852104/275hong.pdf by guest on 13 December 2021


lated donor search; 2005-2017 non-Euro­pean ances­try and less likely to have
no MUD or cord-blood grafts
Bhatt et al2 Age 61-75 years; National Cancer Age, race/eth­nic­ity, geog­ra­phy, Lower like­li­hood of receiv­ing HCT in patients
Database, patients with AML; SES, insur­ance cov­er­age who were older, non-White, of lower edu­ca­
2003-2012 tional sta­tus, unin­sured, on Med­ic­aid/
Medi­care, or received care at non­ac­a­demic
facil­ity; no dif­fer­ence in HCT rates among urban
vs rural facil­ity; higher like­li­hood of receiv­ing
HCT in patients who lived ≥37 miles from facil­ity;
no asso­ci­a­tion with median house­hold income
Paulson et al5 Age <66 years; patients with SES, geog­ra­phy Higher county lev­els of pov­erty asso­ci­ated with
AML/ALL/MDS reported to lower trans­plant rates; rural vs urban sta­tus
CIBMTR and SEER; 2000-2010 was not asso­ci­ated with HCT uti­li­za­tion
Delamater and Uberti18 All ages; sev­eral pub­lic data­bases Geography Overall, 66% of US pop­ul­a­tion lives within 60 min­
utes’ travel time and 94% within 3 hours’ travel
time of HCT facil­ity; geo­graphic access to HCT
facil­ity varies by state
Getta et al27 Age ≤70 years; sin­gle-cen­ter study Age Patients ≥65 years were less likely to be referred
of MDS patients; 2008-2015 for HCT eval­u­a­tion; mar­i­tal sta­tus and insur­
ance type were not asso­ci­ated with trans­plant
refer­ral
Table shows rep­re­sen­ta­tive stud­ies in US pop­u­la­tions published since 2015.
a

MDS, myelodysplastic syn­drome.

used to be a bar­rier to HCT, but alter­na­tive donors (eg, hap- In a dis­cus­sion of disparities in access to trans­plan­ta­tion, it is
loidentical, mismatched unre­lated, and umbil­i­cal cord blood) are impor­tant to acknowl­edge the lim­i­ta­tions of the existing lit­er­a­
now used rou­tinely, and nearly all­patients have a suit­able donor ture. Data on patients who receive HCT are robust and cap­tured
for trans­plan­ta­tion. However, age-related, racial, eco­nomic, and well by insti­tu­tional and national reg­is­tries (eg, the Center for
other social disparities con­tinue to limit access to allo­ge­neic International Blood and Marrow Transplant Research [CIBMTR]).
HCT in the US, and many patients who would oth­er­wise ben­efi ­t However, data on patients who are can­ di­
dates for and may
are not referred for and do not receive trans­plan­ta­tion. poten­tially ben­e­fit from HCT are not read­ily avail­­able. National
As high­lighted by the case above, access to HCT is mod­er­ reg­is­tries and sec­ond­ary data­bases (eg, the Surveillance, Epide-
ated by a com­plex inter­play of sev­eral socio­cul­tural, eco­nomic, miology, and End Results Program [SEER] and sin­gle- or multi-
dis­ease, treating pro­vider, hos­pi­tal-related, and health-sys­tem- payer data­bases) often do not include the details required to
related fac­tors. At a patient level, disparities in access can more deter­mine whether HCT was indi­cated for a given patient (eg,
fac­tors that often tend to be closely related (eg, race/eth­nic­ity, dis­ease risk, remis­sion sta­tus, donor avail­abil­ity). Furthermore,
insur­ance sta­tus, edu­ca­tion level, pov­erty, employ­ment sta­tus; sociodemographic bar­ri­ers are a com­plex con­struct, are chal­
Table 1). Studies have established asso­ci­a­tions between age, leng­ing to define, and are not cap­tured reli­ably at an indi­vid­ual
sex, race/eth­nic­ity, insur­ance cov­er­age, and socio­eco­nomic level; hence, most stud­ies focus on pop­ul­a­tion-level indi­ca­tors
sta­ tus (SES) and the uti­ li­
za­
tion of HCT, and less evi­ dence is to define disparities (eg, median house­hold income based on
avail­­able for other fac­tors such as mar­i­tal sta­tus, lan­guage bar­ zip code of res­i­dence). Studies in HCT recip­i­ents have eval­u­ated
ri­ers, dis­tance from trans­plant cen­ter, and care­giver avail­abil­ity.7 com­pos­ite mea­sures that com­bine sev­eral health dis­par­ity fac­
Although the focus of this review is bar­ri­ers to access, the same tors, but these instru­ments need fur­ther val­i­da­tion.8,9 Qualitative
disparities also influ­ence short-term and long-term out­comes stud­ies are also needed to con­tex­tu­al­ize the quan­ti­ta­tive lit­er­
fol­low­ing HCT, and the con­tem­po­rary lit­er­a­ture in this area is a­ture, to deepen our under­stand­ing of access bar­ri­ers, and to
sum­ma­rized in Table 2. iden­tify impactful and timely inter­ven­tions to address them.

276  |  Hematology 2021  |  ASH Education Program


Table 2. Sociodemographic and cen­ter fac­tors asso­ci­ated with out­comes of allo­ge­neic HCT

Referencea Population Variable(s) Key find­ings


Bona et al28 Age ≤18 years; all­diag­noses; SES In chil­dren with malig­nant dis­ease, high neigh­bor­hood
first allo­ge­neic HCT recip­i­ents pov­erty level asso­ci­ated with higher NRM and Med­ic­aid
reported to CIBMTR; 2006-2015 insur­ance sta­tus asso­ci­ated with higher NRM and infe­
rior OS (vs pri­vate insur­ance); no asso­ci­a­tion between
neigh­bor­hood pov­erty and HCT out­comes for non­ma­
lig­nant dis­ease
Hong et al8 Age ≥18 years; all­diag­noses; Several (county-level indi­ca­tors Patients resid­ing in counties with worse com­mu­nity
first allo­ge­neic HCT recip­i­ents of com­mu­nity health) health sta­tus had infe­rior OS; among patients with
reported to CIBMTR; 2014-2016 hema­to­logic malig­nancy, worse com­mu­nity health
sta­tus was asso­ci­ated with infe­rior OS and higher risks
of NRM

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/275/1852104/275hong.pdf by guest on 13 December 2021


Madbouly et al29 All ages; all­ diag­noses; allo­ge­neic Race/eth­nic­ity (ances­try) Higher recip­i­ent-donor Afri­can genetic admix­ture asso­ci­
HCT using 10/10 allele matched ated with lower OS and DFS and higher NRM
MUD reported to CIBMTR; 1995–
2001
Majhail et al16 Adult HCT cen­ters; all­diag­noses; Center vol­ume Higher 100-day and 1-year OS in high-vol­ume (>40 allo­
allo­ge­neic HCT reported to ge­neic HCT/year) vs low-vol­ume cen­ters; pres­ence of
CIBMTR; 2008-2010 and 2012– sur­vi­vor­ship pro­gram asso­ci­ated with higher 1-year OS
2014
Khera et al30 Adult; all­diag­noses; first allo­ge­ Geography (dis­tance from HCT No asso­ci­a­tion of dis­tance and OS, NRM, or relapse;
neic HCT recip­i­ents at sin­gle cen­ter) trend toward higher NRM with increased dis­tance in
cen­ter; 2000-2010 nonmyeloablative HCT recip­i­ents
Bhatt et al31 All ages; hema­to­logic malig­nancy; Time to insur­ance approval Time to insur­ance approval for HCT var­ied between pri­
sin­gle-cen­ter study of first auto vate and pub­lic pay­ers but was not asso­ci­ated with OS
and allo­ge­neic HCT recip­i­ents;
2007-2011
Table shows rep­re­sen­ta­tive stud­ies in US pop­u­la­tions published since 2015.
a

DFS, dis­ease-free sur­vival; NRM, nonrelapse mor­tal­ity; OS, over­all sur­vival.

Racial bar­ri­ers to HCT lower median house­ hold income com­ pared to Whites.14 This
Race and eth­nic­ity are par­tic­u­larly rel­e­vant when con­sid­er­ing dis­par­ity often trans­lates to expo­sure to adverse social deter­
access to allo­ge­neic HCT. First, racial disparities that are rou­ mi­nants of health in the for­mer, includ­ing a greater like­li­hood of
tinely prev­a­lent in health care apply to HCT and in fact may be resid­ing in areas with high pov­erty lev­els, inad­e­quate health care
accen­tu­ated given the com­plex­ity and expense of the pro­ce­ cov­er­age, and lower lev­els of health lit­er­acy. Racial dis­par­ity in
dure along with its restricted avail­abil­ity in select cen­ters in the access to HCT has been well documented, includ­ing a recent
US. Second, there is an ele­ment of donor avail­abil­ity asso­ci­ated study that showed adult His­panic and Black patients with AML
with race and very spe­cific to HCT. Unrelated donor reg­is­tries and acute lym­pho­blas­tic leu­ke­mia hav­ing a lower prob­a­bil­ity of
are over­rep­re­sented by donors of Euro­pean ances­try, and White pro­ceed­ing with HCT.3 The mech­a­nism by which these social
patients have a higher chance of find­ing an HLA-matched unre­ deter­mi­nants of health have an impact on the ulti­mate receipt
lated donor (MUD).10-12 Using data from the National Marrow Do- of allo­ge­neic HCT is com­plex, as dem­on­strated in a study by
nor Program reg­is­try, Gragert et al showed that the like­li­hood of Clay et  al.15 They showed that the rea­sons for not receiv­ing a
find­ing a high-res­o­lu­tion HLA 8/8 allele MUD was 75% for White trans­plant dif­fered by race. Patient deci­sion/treat­ment reluc­
peo­ple of Euro­pean descent and only 16% for Black peo­ple of tance and sta­ble dis­ease sta­tus not severe enough to war­rant
South or Central Amer­i­can ances­try.11 Given that the major­ity of trans­plant were the most impor­tant rea­sons for not pro­ceed­
patients do not have an HLA-iden­ti­cal sib­ling donor, this dis­par­ ing in Euro­pean Amer­i­can patients, whereas comorbidities and
ity in MUD avail­abil­ity by race/eth­nic­ity has sig­nif­i­cant impli­ca­ phy­si­cian deci­sion were the main rea­sons for not pro­ceed­ing in
tions on trans­plant uti­li­za­tion and, ulti­mately, sur­vival and oth- Afri­can Amer­i­can patients. Psychosocial or com­pli­ance con­cerns
er out­comes after HCT. There is a pos­si­bil­ity that this dis­par­ity were iden­ti­fied more often in Afri­can Amer­i­can patients as a rea­
may worsen in the future. In another anal­y­sis that mod­eled the son for not pro­ceed­ing with HCT.
like­li­hood of find­ing HLA-iden­ti­cal sib­lings and unre­lated do-
nors, Besse et al showed that the aver­age num­ber of sib­lings Socioeconomic bar­ri­ers to HCT
and sib­ling match prob­a­bil­ity vary by patient age and race, and In addi­tion to race/eth­nic­ity, the asso­ci­a­tion of SES with access
young minor­ity patients are at greatest risk for not find­ing an to HCT has been well documented, with most stud­ies using
HLA-matched donor.13 US Census tract data to define SES. This is a lim­i­ta­tion of the
In addi­tion to race-related donor issues, minor­ity pop­u­la­tions existing lit­er­a­ture since SES is most accu­rate when it is patient
often have social and eco­nomic bar­ri­ers to refer­ral and donor self-reported. Regardless, using zip codes to esti­mate median
search. In gen­eral, His­panic and Black pop­u­la­tions in the US have house­hold income is a well-val­i­dated method for defin­ing SES in
Prakash Singh Shekhawat
Access to allo­ge­neic trans­plan­ta­tion in the US  |  277
Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/275/1852104/275hong.pdf by guest on 13 December 2021
Figure 1. Framework for investigating interventions to address racial, socioeconomic, and geographic disparities in access to
allogeneic HCT.

health care research. In the con­tem­po­rary lit­er­a­ture, Jabo et al respec­tively.18 There is sig­nif­i­cant var­i­a­tion by state; eg, >70%
have reported an inverse asso­ci­a­tion of neigh­bor­hood SES with of adult res­i­dents in Arizona, Maryland, New Jersey, New York,
HCT uti­li­za­tion; com­pared to the highest-quin­tile SES, patients Rhode Island, and Washington, DC, live within 30 min­utes of an
resid­ing in the low­est quin­tile had a lower like­li­hood of under­go­ HCT, whereas 6% of the US pop­u­la­tion must travel >3 hours to
ing HCT (adjusted rel­a­tive risk, 0.63; 95% CI, 0.47-0.84 for acute access a trans­plant facil­ity.18
lym­pho­blas­tic leu­ke­mia and 0.52; 0.43-0.64, for AML).3 Similar- Since the major­ity of the US pop­u­la­tion live in rea­son­ably
ly, Paulson et al, using data from the CIBMTR, showed that res­ close prox­im­ity to an HCT cen­ter, stud­ies have shown no defin­i­
i­dence in counties with high lev­els of pov­erty was asso­ci­ated tive asso­ci­at­ ion between dis­tance from the trans­plant pro­gram
with a lower prob­a­bil­ity of receiv­ing HCT (in mul­ti­var­i­able anal­ or rural/urban res­i­dence sta­tus and receipt of allo­ge­neic HCT.2,3
y­sis, esti­mated rate ratio was 0.86 per 10% increase in county Interestingly, a recent study has shown that patients a ­ ble to
pop­u­la­tion below the pov­erty line; P  <  .01).5 travel lon­ger dis­tances (≥37 miles) for care were more likely to
Studies that have been ­able to inves­ti­gate the role of social receive trans­plant, pos­si­bly indi­cat­ing bet­ter patient sta­tus or
and eco­ nomic deter­ mi­
nants in greater detail sug­ gest some bet­ter receipt of care in ter­tiary refer­ral hos­pi­tals.2 As illus­trated
mech­a­nisms by which SES influ­ences access to HCT. In a study in the case at the begin­ning of this arti­cle, geo­graphic disparities
using the National Cancer Database, Bhatt et al found that the are likely accen­tu­ated in patients who are socio­eco­nom­i­cally
pri­mary payer for HCT cov­er­age was sig­nif­i­cantly asso­ci­ated under­served to begin with.
with HCT uti­li­za­tion.2 Compared to pri­vate insur­ance, patients Although not spe­cif­i­cally a focus of this review, other social
were less likely to receive HCT if they had Med­ic­aid (odds ratio deter­mi­nants of health can be related to racial, socio­eco­nomic,
[OR], 0.3; 95% CI, 0.3-0.5; P < .0001), Medi­care (OR, 0.7; 0.6-0.8; and geo­graphic disparities and ulti­mately affect access to allo­ge­
P < .0001), unin­sured (OR, 0.2; 0.1-0.5; P  = .0003), and unknown neic HCT. Some exam­ples of such bar­ri­ers include age, sex, patient
insur­ance sta­tus (OR, 0.1; 0.1-0.3; P  <  .0001). pref­er­ence, edu­ca­tional sta­tus, health lit­er­acy level, psy­chi­at­ric
dis­abil­ity, sub­stance abuse, mar­i­tal sta­tus, lan­guage bar­ri­ers, and
Geographic bar­ri­ers to HCT lack of com­pli­ance with med­i­cal care.7
Given its spe­cial­ized and highly reg­u­lated nature, need for expe­
ri­enced per­son­nel, and infrastructural require­ments, HCT is avail­­ Opportunities to address disparities in access to HCT
able through approx­i­ma­tely 200 trans­plant pro­grams in the US. Research to date in the field of HCT has largely focused on
There is a ratio­nale for restricting HCT to select cen­ters since under­stand­ing and defin­ing health care disparities in access
a vol­ume-out­come rela­tion­ship has been dem­on­strated for this to and out­comes of allo­ge­neic HCT, and acknowl­edg­ing and
pro­ce­dure.16,17 However, this does cause a bar­rier to some pa- quan­ ti­
fy­
ing them is an impor­ tant first step. Less work has
tients who need to travel long dis­tances to access a trans­plant been done around inves­ti­gat­ing inter­ven­tions to resolve or
cen­ter. Overall, 48% and 79% of the US adult pop­ul­a­tion and mit­i­gate these disparities. Some rea­sons for this are related to
43% and 72% of the pedi­at­ric pop­u­la­tion have access to an HCT the long-stand­ing sys­temic inequities in health care that need
facil­ity within 30 and 90 min­utes’ travel time from their homes, to be addressed at the soci­e­tal level, the lack of val­i­dated

278  |  Hematology 2021  |  ASH Education Program


health care inter­ven­tions to address these disparities, the dif­ also apply and may in fact be worse given the costs of newer chi­
fi­culty in gen­er­al­iz­ing stud­ies given that social deter­mi­nants me­ric anti­gen recep­tor T-cell ther­a­pies. Ultimately, the ben­e­fit of
may vary at the local and indi­vid­ual lev­els, and the fact that inno­va­tions in HCT and cel­lu­lar ther­apy can be fully real­ized when
such disparities are often out­side the con­trol of what a trans­ all­patients who may ben­e­fit actu­ally receive these pro­ce­dures.
plant cen­ter can real­is­ti­cally influ­ence. Furthermore, the addi­
tional resources and effort required to bring in patients from
dis­ad­van­taged pop­u­la­tions for HCT may not be pri­or­i­tized at
the refer­ring pro­vider and trans­plant cen­ter level. Neverthe- CLINICAL CASE (Con­tin­ued)
less, oppor­tu­ni­ties exist for inter­ven­tions to ensure that pa- The treating hema­tol­o­gist-oncol­o­gist contacted the trans­plant
tients with racial, socio­eco­nomic, and geo­graphic chal­lenges cen­ter early in the patient’s treat­ment course. Local and trans­
receive appro­pri­ate trans­plant-related care (Figure 1). plant cen­ter social work­ers were a ­ ble to enroll the patient in
A major time point for inter­ven­tion to improve access is refer­ a state Med­ic­aid pro­gram and refer her for grants and other
ral from a patient’s oncol­o­gist to the trans­plant cen­ter so that ser­vices (eg, transportation assis­tance). An ini­tial HCT con­sult
indi­ca­tion and can­di­dacy for trans­plan­ta­tion can be deter­mined,

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/275/1852104/275hong.pdf by guest on 13 December 2021


was conducted through telemedicine, and a donor search was
and a donor search can be ini­ti­ated in a timely man­ner.19 Early ini­ti­ated. A close friend stepped in to serve as a ded­i­cated care­
refer­ral in the dis­ease course also allows for the iden­ti­fi­ca­tion of giver dur­ing trans­plant. A MUD was iden­ti­fied, and the patient
psy­cho­so­cial and SES fac­tors that may later hin­der pro­ceed­ing was ­able to suc­cess­fully pro­ceed with an allo­ge­neic trans­plant.
with trans­plan­ta­tion so that they can be addressed early. Social
work­ers and care coor­di­na­tors are an inte­gral part of the trans­
plant team and play an impor­tant role in this early eval­u­a­tion Conflict-of-inter­est dis­clo­sure
and in gath­er­ing appro­pri­ate resources for patients. Some exam­ Sanghee Hong: no com­pet­ing finan­cial inter­ests to declare.
ples of such inter­ven­tions include refer­rals for grants to off­set Navneet S. Majhail: no com­pet­ing finan­cial inter­ests to declare.
out-of-pocket costs, assessing care­giver sup­port, and help­ing
with local hous­ing for patients who must tem­po­rar­ily relo­cate Off-label drug use
to be close to the trans­plant cen­ter.20,21 Particularly, pro­grams Sanghee Hong: none discussed.
often require a ded­i­cated care­giver for HCT recip­i­ents, and inter­ Navneet S. Majhail: none discussed.
ven­tions to iden­tify and sup­port care­giv­ers are needed.21 Addi-
tionally, the devel­op­ment and implementation of tools to assess Correspondence
social chal­lenges can facil­i­tate indi­vid­u­al­ized inter­ven­tions for Navneet S. Majhail, Blood and Marrow Transplant Program,
patients. Some instru­ments that have been eval­u­ated in HCT Taussig Cancer Center, Cleveland Clinic, 9500 Euclid Ave, CA60,
recip­i­ents include the Psychosocial Assessment of Candidates Cleveland, OH 44195; e-mail: majhain@ccf​­.org.
for Transplantation scale, Transplant Evaluation Rating Scale, and
Stanford Integrated Psychosocial Assessment for Transplant.22-25 References
Data are needed for spe­cial pop­u­la­tions (eg, les­bian, gay, bisex­ 1. D’Souza A, Fretham C, Lee SJ, et al. Current use of and trends in hema­to­
poi­etic cell trans­plan­ta­tion in the United States. Biol Blood Marrow Trans-
ual, trans­gen­der, queer patients; health-illit­er­ate indi­vid­u­als; or
plant. 2020;26(8):e177-e182.
non­na­tive English speak­ers) so that appro­pri­ate inter­ven­tions 2. Bhatt VR, Chen B, Gyawali B, Lee SJ. Socioeconomic and health sys­tem fac­
can be designed to help them access HCT. tors asso­ci­ated with lower uti­li­za­tion of hema­to­poi­etic cell trans­plan­ta­tion
In this same con­text, opti­miz­ing health care deliv­ery for allo­ge­ in older patients with acute mye­loid leu­ke­mia. Bone Marrow Transplant.
neic HCT recip­i­ents by improv­ing care coor­di­na­tion among pri­ 2018;53(10):1288-1294.
3. Jabo B, Mor­gan JW, Martinez ME, Ghamsary M, Wieduwilt MJ. Sociodemo-
mary care phy­si­cians, refer­ring hema­tol­o­gists/oncol­o­gists, and graphic disparities in che­mo­ther­apy and hema­to­poi­etic cell trans­plan­ta­tion
trans­plant cen­ters can help mit­i­gate the social bar­ri­ers and chal­ uti­li­za­tion among adult acute lym­pho­blas­tic and acute mye­loid leu­ke­mia
lenges that patients and fam­i­lies face. Khera et al have described patients. PLoS One. 2017;12(4):e0174760.
a patient-cen­tered care frame­work to coor­di­nate allo­ge­neic HCT 4. Majhail NS, Omondi NA, Denzen E, Mur­phy EA, Rizzo JD. Access to hema­to­
poi­etic cell trans­plan­ta­tion in the United States. Biol Blood Marrow Trans-
deliv­ery.26 They lay out var­ i­ous phases in the HCT con­ tin­
uum
plant. 2010;16(8):1070-1075.
and the roles of var­i­ous health care pro­vid­ers. Their frame­work 5. Paulson K, Brazauskas R, Khera N, et al. Inferior access to allo­ge­neic trans­plant
can be expanded to address social dis­par­ity-related fac­tors that in dis­ad­van­taged pop­u­la­tions: a cen­ter for inter­na­tional blood and mar­row
play a role through­out the trans­plant jour­ney. It can also inform trans­plant research anal­y­sis. Biol Blood Marrow Transplant. 2019;25(10):2086-
research on inter­ven­tions since all­stake­hold­ers, includ­ing health 2090.
6. Khera N, Deeg HJ, Kodish E, Rondelli D, Majhail N. Allogeneic hema­
care pro­vid­ers from out­side the trans­plant cen­ter net­work, need to­poi­etic cell trans­plan­ta­tion and other expen­sive cel­lu­lar ther­a­pies: a
to be engaged to address these disparities. A com­mon thread for mir­a­cle for the few but off lim­its to many? J Clin Oncol. 2020;38(12):1268-
patients as they move through dif­fer­ent phases and sites of care 1272.
is their payer, and we need to explore oppor­tu­ni­ties for pro­vider- 7. Flannelly C, Tan BE, Tan JL, et al. Barriers to hema­to­poi­etic cell trans­plan­
ta­tion for adults in the United States: a sys­tem­atic review with a focus on
payer col­lab­o­ra­tions to address bar­ri­ers to trans­plant.
age. Biol Blood Marrow Transplant. 2020;26(12):2335-2345.
There is an urgent need for more research and funding to 8. Hong S, Brazauskas R, Hebert KM, et al. Community health sta­tus and out­
sup­port the inves­ti­ga­tion of inno­va­tive inter­ven­tions to address comes after allo­ge­neic hema­to­poi­etic cell trans­plan­ta­tion in the United
socio­eco­nomic and geo­graphic disparities in access to allo­ge­ States. Cancer. 2021;127(4):609-618.
neic HCT. There is also a soci­e­tal respon­si­bil­ity to address health 9. Hong S, Rybicki LA, Corrigan D, Schold JD, Majhail NS. Community risk
score for eval­u­at­ing health care disparities in hema­to­poi­etic cell trans­plan­
care sys­tem fac­tors that operate at a sys­temic level and ulti­mately ta­tion. Biol Blood Marrow Transplant. 2018;24(4):877-879.
have an impact on access to HCT. Although cel­lu­lar ther­apy was 10. Dehn J, Chitphakdithai P, Shaw BE, et al. Likelihood of pro­ceed­ing to allo­
not the focus of this review, sim­i­lar health care dis­par­ity fac­tors ge­neic hema­to­poi­etic cell trans­plan­ta­tion in the United States after search
Prakash Singh Shekhawat
Access to allo­ge­neic trans­plan­ta­tion in the US  |  279
acti­va­tion in the national reg­is­try: impact of patient age, dis­ease, and 22. Hong S, Rybicki L, Corrigan D, et  al. Psychosocial Assessment of Candi-
search prog­no­sis. Transplant Cell Ther. 2021;27(2):184.e181-184.e113. dates for Transplant (PACT) as a tool for psy­cho­log­i­cal and social eval­u­
11. Gragert L, Eapen M, Williams E, et al. HLA match like­li­hoods for hema­to­ a­tion of allo­ge­neic hema­to­poi­etic cell trans­plan­ta­tion recip­i­ents. Bone
poi­etic stem-cell grafts in the U.S. reg­is­try. N Engl J Med. 2014;371(4):339- Marrow Transplant. 2019;54(9):1443-1452.
348. 23. Harashima S, Yoneda R, Horie T, et al. Psychosocial Assessment of Candidates
12. Barker JN, Boughan K, Dahi PB, et al. Racial disparities in access to HLA- for Transplantation scale (PACT) and sur­vival after allo­ge­neic hema­to­poi­etic
matched unre­lated donor trans­plants: a pro­spec­tive 1312-patient anal­y­sis. stem cell trans­plan­ta­tion. Bone Marrow Transplant. 2019;54(7):1013-1021.
Blood Adv. 2019;3(7):939-944. 24. Solh MM, Speckhart D, Sol­o­mon SR, et al. The Transplant Evaluation Rat-
13. Besse K, Maiers M, Confer D, Albrecht M. On mod­el­ing human leu­ko­cyte ing Scale pre­dicts over­all sur­vival after allo­ge­neic hema­to­poi­etic stem cell
anti­gen-iden­ti­cal sib­ling match prob­a­bil­ity for allo­ge­neic hema­to­poi­etic trans­plan­ta­tion. Blood Adv. 2020;4(19):4812-4821.
cell trans­plan­ta­tion: esti­mat­ing the need for an unre­lated donor source. 25. Hoodin F, Kalbfleisch KR. Factor anal­y­sis and validity of the Transplant Eval-
Biol Blood Marrow Transplant. 2016;22(3):410-417. uation Rating Scale in a large bone mar­row trans­plant sam­ple. J Psycho-
14. Guzman GG. Household income by race and His­panic ori­gin: 2005–2009 and som Res. 2003;54(5):465-473.
2015–2019. US Census Bureau Amer­i­can Community Survey Brief ACSBR-7. 26. Khera N, Martin P, Edsall K, et al. Patient-cen­tered care coor­di­na­tion in hema­
Decem­ber 2020. Accessed 16 Sep­tem­ber 2021. https:​­/​­/www​­.census​­.gov​­ to­poi­etic cell trans­plan­ta­tion. Blood Adv. 2017;1(19):1617-1627.
/content​­/dam​­/Census​­/library​­/publications​­/2020​­/acs​­/acsbr19​­-07​­.pdf. 27. Getta BM, Kishtagari A, Hilden P, et al. Allogeneic hema­to­poi­etic stem cell
15. Clay A, Peoples B, Zhang Y, et  al. Population-based anal­y­sis of hema­to­ trans­plan­ta­tion is underutilized in older patients with myelodysplastic syn­

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/275/1852104/275hong.pdf by guest on 13 December 2021


logic malig­nancy refer­rals to a com­pre­hen­sive can­cer cen­ter, refer­rals for dromes. Biol Blood Marrow Transplant. 2017;23(7):1078-1086.
blood and mar­row trans­plan­ta­tion, and par­tic­i­pa­tion in clin­i­cal trial, sur­ 28. Bona K, Brazauskas R, He N, et  al. Neighborhood pov­erty and pedi­at­ric
vey, and biospecimen research by race. Biol Blood Marrow Transplant. allo­ge­neic hema­to­poi­etic cell trans­plan­ta­tion out­comes: a CIBMTR anal­y­
2015;21(8):1488-1494. sis. Blood. 2021;137(4):556-568.
16. Majhail NS, Mau LW, Chitphakdithai P, et  al. Transplant cen­ter char­ac­ter­is­ 29. Madbouly A, Wang T, Haagenson M, et al. Investigating the asso­ci­a­tion of
tics and sur­vival after allo­ge­neic hema­to­poi­etic cell trans­plan­ta­tion in adults. genetic admix­ture and donor/recip­i­ent genetic dis­par­ity with trans­plant
Bone Marrow Transplant. 2020;55(5):906-917. out­comes. Biol Blood Marrow Transplant. 2017;23(6):1029-1037.
17. Majhail NS, Mau LW, Chitphakdithai P, et  al. National sur­vey of hema­to­ 30. Khera N, Gooley T, Flowers MED, et  al. Association of dis­ tance from
poi­etic cell trans­plan­ta­tion cen­ter per­son­nel, infra­struc­ture, and mod­els of trans­plan­ta­tion cen­ter and place of res­i­dence on out­comes after allo­ge­
care deliv­ery. Biol Blood Marrow Transplant. 2015;21(7):1308-1314. neic hema­to­poi­etic cell trans­plan­ta­tion. Biol Blood Marrow Transplant.
18. Delamater PL, Uberti JP. Geographic access to hema­to­poi­etic cell trans­plan­ 2016;22(7):1319-1323.
ta­tion ser­vices in the United States. Bone Marrow Transplant. 2016;51(2): 31. Bhatt VR, Loberiza FR Jr, Schmit-Pokorny K, Lee SJ. Time to insur­ ance
241-248. approval in pri­ vate and pub­ lic pay­
ers does not influ­ ence sur­ vival in
19. Majhail NS, Jagasia M. Referral to trans­plant cen­ter for hema­to­poi­etic cell patients who undergo hema­to­poi­etic cell trans­plan­ta­tion. Biol Blood Mar-
trans­plan­ta­tion. Hematol Oncol Clin North Am. 2014;28(6):1201-1213. row Transplant. 2016;22(6):1117-1124.
20. Preussler JM, Mau LW, Majhail NS, et al. Patient hous­ing bar­ri­ers to hema­to­
poi­etic cell trans­plan­ta­tion: results from a mixed-meth­ods study of trans­
plant cen­ter social work­ers. Support Care Cancer. 2016;24(3):1167-1174.
21. Preussler JM, Mau LW, Majhail NS, et al. Caregiver avail­abil­ity and patient
access to hema­to­poi­etic cell trans­plan­ta­tion: social worker per­spec­tives © 2021 by The Amer­i­can Society of Hematology
inform prac­tice. Support Care Cancer. 2019;27(11):4253-4264. DOI 10.1182/hema­tol­ogy.2021000259

280  |  Hematology 2021  |  ASH Education Program


IMMUNOLOGY 101: WHAT THE PRACTICING HEMATOLOGIST NEEDS TO KNOW

Immunology 101: fundamental immunology


for the practicing hematologist

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/281/1851987/281carty.pdf by guest on 13 December 2021


Shannon A. Carty
Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI

From an evolutionary perspective, the immune system developed primarily to protect the host from pathogens. In the
continuous balance between killing pathogens and protecting host tissues, selective pressures have shaped the discrim-
inatory functions of the immune system. In addition to protection against microbial pathogens, the immune system also
plays a critical role in antitumor immunity. Immune dysfunction, either under- or overactivity, is found in a wide range of
hematologic disorders. Here we review the fundamental features of the immune system and the key concepts critical to
understanding the impact of immune dysfunction on hematologic disorders.

LEARNING OBJECTIVES
• Describe the cardinal features of the innate vs adaptive arms of the immune system
• Define the mechanisms that result in an inadequate immune response
• Understand the immunotherapeutic approaches to overcome immune dysfunction

age. This response is mediated by germ line­encoded pattern


CLINICAL CASE recognition receptors (PRRs) that can recognize conserved
A 70­year­old man in previously good health presents with 6 features of pathogens or damaged cells, termed pathogen­
weeks of fatigue and drenching night sweats. The physical associated molecular patterns or damage­associated molec­
exam is notable for cervical, axillary, and inguinal lymphade­ ular patterns. Cells of the innate immune system include
nopathy. Laboratory studies are significant for mild anemia granulocytes,monocytes/macrophages,dendriticcells(DCs),
and an elevated lactate dehydrogenase level. Lymph node and natural killer (NK) cells. Through PRRs, innate immune
biopsy reveals CD20+CD10+Bcl6+ large B cells consistent cells recognize broadly conserved structures present in a
with a diffuse large B­cell lymphoma, germinal center sub­ large group of micro­organisms, such as bacterial lipopoly­
type. He is treated with rituximab, cyclophosphamide, doxo­ saccharide, a component of gram­negative bacteria, or
rubicin hydrochloride, vincristine sulfate, and prednisone, pathogen­associated nucleic acids such as double­stranded
also known as R­CHOP, immunochemotherapy for 6 cycles RNA. There are 4 main classes of PRRs: toll­like receptors,
and obtains a complete remission. However, his lymphoma C­type lectin receptors, NOD­like receptors, and RIG­I­like
relapses within 6 months, and he has progressive disease receptors.
despite salvage immunochemotherapy. He then discusses Innate immune cells exert their effector functions through
chimeric antigen receptor T cells (CAR­T) therapy vs a bispe­ the phagocytosis of infected cells and/or extracellular organ­
cific T­cell engager (BiTE) clinical trial with his hematologist. isms, the production of inflammatory cytokines, and the
release of soluble mediators, such as cytokines and chemo­
kines. Different pathogens trigger distinctive innate immune
Introduction responses as distinct effector functions are needed for effec­
tive clearance. For instance, neutrophils are required to pro­
Innate immune system tect against certain fungal pathogens, such as Aspergillus
The immune system is classically divided into 2 primary fumigatus and Candida albicans,1 the putative underlying
arms—the innate and the adaptive (Table 1). The cardinal fea­ mechanism for the increased risk of invasive fungal infections
ture of the innate immune system is the rapid but nonspecific in patients with prolonged neutropenia following high­dose
response to a broad repertoire of pathogens and tissue dam­ chemotherapy or allogeneic hematopoietic stem cell (HSC)
Prakash Singh Shekhawat
Fundamentals of immunology | 281
Table 1. Key char­ac­ter­is­tics of innate vs adap­tive immune cells enhanced with an addi­tional layer of junc­tional diver­sity when
nucle­ot­ides are added or subtracted between dif­fer­ent gene
Innate Adaptive seg­ments dur­ing the recom­bi­na­tion pro­cess. Each resul­tant
Timing of response Minutes to hours Days B or T cell expresses an anti­gen recep­tor with unique spec­i­fic­ity.
Estimates sug­gest that such diver­sity could lead to 1015 to 1018
Self vs non­self No Yes
unique BCR com­bi­na­tions and 1015 to 1020 dis­tinct TCRs; how­ever,
Diversity Limited Extensive unique func­tional pop­u­la­tions are sig­nif­i­cantly smaller due to
Receptors Broad, germ line encoded Highly spe­cific, devel­op­men­tal selec­tion events. Exploiting this fact, poly­mer­ase
somat­i­cally chain reac­tions to detect somat­i­cally rearranged immu­no­glob­u­
rearranged lin or TCR loci are a com­monly used molec­u­lar hematopathology
Cell types Granulocytes, mac­ro­phages, B cells, T cells tech­nique to eval­u­ate for clonality in pathol­ogy spec­i­mens.
DCs, NK cells B cells: B cells develop in the bone mar­row from HSCs, where
they undergo sev­eral pro­gres­sive stages of lin­e­age spec­i­fi­ca­
tion and com­mit­ment. B-cell com­mit­ment and devel­op­ment are

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/281/1851987/281carty.pdf by guest on 13 December 2021


trans­plant.2 Furthermore, some innate immune cells such as DCs reg­u­lated by a coor­di­nated net­work of tran­scrip­tion fac­tors as
and mac­ ro­
phages func­
tion as anti­gen presenting cells (APCs). well as accom­pa­ny­ing epi­ge­netic changes. In the bone mar­row,
Antigen pre­sen­ta­tion is a pro­cess through which small pep­tides devel­op­ing B cells rearrange the heavy-chain and then the light-
are incor­po­rated into the bind­ing groove of the major his­to­com­ chain immu­no­glob­u­lin loci. Following the gen­er­a­tion of an anti­
pat­i­bil­ity com­plex (MHC; also known as human leu­ko­cyte anti­gen) gen recep­tor, devel­op­ing B cells must pass sev­eral check­points
pro­teins. During an infec­tion, small pep­tides derived from path­o­ to pre­vent the pro­duc­tion of self-reac­tive lym­pho­cytes.8 Once
gen pro­teins are presented in the con­text of self-MHC on the sur­ each chain is suc­cess­fully rearranged, a pro­cess called alle­lic
face of APCs to help acti­vate T lym­pho­cytes and thus the adap­tive exclu­sion occurs to pre­vent rearrangement of the other allele,
immune response. which allows each indi­vid­ual cell to pro­duce only a recep­tor of
More recently, immune cells termed “innate lym­phoid cells” sin­gle-anti­gen spec­i­fic­ity. Light-chain loci also undergo isotypic
(ILCs), which have fea­tures of both the innate and adap­tive immune exclu­sion, in which only 1 type of light chain (κ or λ) is expressed
cells, have been rec­og­nized. ILCs respond quickly to infec­tion and by each indi­vid­ual B cell. Once a func­tional BCR forms (as a sur­
do not express anti­gen recep­tors like innate immune cells but pro­ face IgM), the anti­gen recep­tor is tested for self-reac­tiv­ity in a
duce sim­i­lar inflam­ma­tory cyto­kines as T lym­pho­cytes.3 pro­cess known as cen­tral tol­er­ance to elim­i­nate autoreactive
cells. Immature B cells then migrate out of the bone mar­row
Adaptive immune sys­tem to the spleen, where they undergo fur­ther mat­ur­a­tion. In the
In con­trast to the innate immune response, the car­di­nal fea­ periph­ery, B cells that encoun­ter and rec­og­nize self-anti­gen in
tures of the adap­tive immune response include anti­gen spec­i­ the absence of infec­tion will be deleted or become anergic in
fic­ity and the for­ma­tion of immu­no­log­i­cal mem­ory, which is the a pro­cess known as periph­eral tol­er­ance. Failure of these tol­
abil­ity of lym­pho­cytes to respond to a pre­vi­ously encoun­tered er­ance mech­a­nisms can result in B-cell-medi­ated auto­im­mu­nity.
anti­ gen rap­ idly and more effi­ ciently upon reexposure. B and Naive (ie, anti­gen-inex­pe­ri­enced) B cells are acti­vated by sol­u­
T lym­pho­cytes are the pri­mary cells of the adap­tive immune sys­ ble anti­gen through their BCR, which then ini­ti­ates an intra­cel­lu­lar
tem. Both lin­ea ­ ges express anti­gen recep­tors, the B-cell recep­ sig­nal­ing cas­cade. The BCR can also deliver the anti­gen intra­cel­
tor (BCR) and T-cell recep­tor (TCR), respec­tively, which undergo lu­larly for anti­gen processing to allow B cells to pres­ent anti­genic
somatic rearrangement to allow B and T cells to rec­og­nize and pep­tides to T cells. Some anti­gens can acti­vate B cells with­out
respond to spe­cific anti­gens. B cells are the pri­mary medi­at­ors T cell help (T inde­pen­dent) or require T cell help (T depen­dent).
of the humoral (ie, anti­body-medi­ated) immune response, which In T-depen­dent responses, B cells receive addi­tional acti­va­tion
func­tions to pro­tect the host from extra­cel­lu­lar micro-organ­isms sig­nals from CD4+ T fol­lic­u­lar helper cells that rec­og­nizes the anti­
and pre­vent the spread of intra­cel­lu­lar path­o­gens. T cells medi­ate genic pep­tide. Activated B cells can dif­fer­en­ti­ate into sev­eral dif­
the cel­lu­lar immune response, which both sup­ports the humoral fer­ent mature sub­sets,9 includ­ing plasmablasts and lon­ger-lived
immu­nity and acts as the pri­mary effec­tor cell to kill virally infected plasma cells, as well as migrate to the ger­mi­nal cen­ter to undergo
or transformed host cells. fur­ther mat­u­ra­tion, includ­ing somatic hypermutation of the
Lymphocytes of the adap­tive immune sys­tem share com­mon V regions of the immu­no­glob­u­lin genes to gen­er­ate higher-affin­ity
fea­tures of anti­gen spec­i­fic­ity and the for­ma­tion of long-lived antibodies (affin­ity mat­u­ra­tion) and class switching to allow
immune mem­ory. Specificity is achieved by each lym­pho­cyte B cells to pro­duce antibodies with dif­fer­ent effec­tor func­tions
bear­ing an anti­gen recep­tor that rec­og­nizes a sin­gle epi­tope, (IgG, IgE, IgA). Molecular sub­types of dif­fer­ent lym­pho­mas have
which is attained by the somatic rearrangement of sev­eral sets sim­il­ar gene expres­sion sig­na­tures as nor­mal B-cell sub­sets, such as
of gene seg­ments encoding the BCR, which is also a mem­brane- the ger­mi­nal cen­ter B-cell sub­type and the acti­vated B-cell sub­type
bound form of antibodies or immu­no­glob­u­lins (Ig), and TCRs to (or non-ger­mi­nal cen­ter B cell) of dif­fuse large B-cell lym­phoma,10
gen­er­ate recep­tor diver­sity. During B- and T-cell devel­op­ment, which can pre­dict response to cer­tain targeted ther­ap ­ ies.11
the var­i­able (V), diver­sity (D), and join­ing (J) gene seg­ments are T cells: T lym­pho­cytes are one of the few cells of the immune
somat­i­cally rearranged through DNA recom­bi­na­tion medi­ated sys­tem that develop out­side the bone mar­row. T cell pre­cur­
by the recom­bi­na­tion-acti­vat­ing genes (RAG1/2) in a rel­a­tively sors migrate from the bone mar­row to the thy­mus, where they
anal­og ­ ous pro­cess occur­ring at the light and heavy chains of mature into func­tional T cells. Collectively, mature T cells must
the immunoglobin loci or the α and β chains of the TCR locus.4-7 pos­sess a diverse TCR rep­er­toire capa­ble of rec­og­niz­ing the
The com­bi­na­to­rial diver­sity of V(D)J recom­bi­na­tion is fur­ther immense num­ber of for­eign anti­gens that will be encoun­tered

282  |  Hematology 2021  |  ASH Education Program


over the host’s life­time. In con­trast to B cells that respond to sol­ Tregs, naive CD4+ T cells can also be induced to become Tregs
u­ble anti­gen, T cells are stim­u­lated by small anti­genic pep­tides and exert immu­no­sup­pres­sive func­tions in the appro­pri­ate con­
presented on the sur­face of other cells by MHC mol­e­cules. Since text. CD8+ T cells become ter­mi­nally dif­fer­en­ti­ated effec­tor cells
the TCR binds anti­genic pep­tides plus some amino acid res­i­dues or long-lived mem­ory cells fol­low­ing anti­gen stim­u­la­tion.16
of self-MHC pro­tein, it is crit­i­cal that only T cells with a TCR ­able
to rec­og­nize self-MHC, albeit with lim­ited affin­ity, be exported to What con­sti­tutes an effec­tive immune response?
the periph­ery. In addi­tion, it is essen­tial that T cells pos­sess TCRs A suc­cess­ful immune response results in the clear­ance of the path­
that only respond to for­eign anti­gen and that those that pos­ o­gen and the for­ma­tion of immu­no­logic mem­ory while ­pre­vent­ing
sess TCRs rec­og­niz­ing self-pep­tide in the con­text of self-MHC host dam­age. It requires the care­ful and ­inte­grated orches­tra­tion
are elim­i­nated dur­ing devel­op­ment to pre­vent auto­im­mu­nity. of a mul­ti­step pro­cess involv­ing diverse cell types, cyto­kines, and
Similar to B-cell devel­op­ment, devel­op­ing T cells undergo sev­ ana­tomic local­i­za­tion. A ­pro­to­typ­i­cal immune response can be
eral key check­points to ensure the devel­op­ment of func­tional, bro­ken down into 3 main stages: (1) acute inflam­ma­tory response
non-self-reac­tive T cells.12 In the thy­mus, devel­op­ing αβT cells to the invad­ing path­o­gen by the innate immune sys­tem, (2) anti­
undergo 2 fur­ther check­points, known as pos­i­tive and neg­a­tive gen pre­sen­ta­tion, and (3) ­adap­tive immune response involv­ing

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/281/1851987/281carty.pdf by guest on 13 December 2021


selec­tion, to deter­mine TCR fit­ness. Positive selec­tion occurs CD4+ and CD8+ T cells and B ­lym­pho­cytes.
when thy­mo­cytes inter­act with self-anti­gens presented in the Innate acute inflam­ma­tory response: The first line of defense
con­text of class I or class II MHC on cor­ti­cal thy­mic epi­the­lial is typ­i­cally bar­rier sur­faces that seek to pre­vent path­o­gen entry
cells. The thy­mo­cytes that engage anti­gen/MHC with inter­me­ into the host, includ­ ing the epi­ the­ lial sur­
faces and muco­ sal
di­ate affin­ity are protected from apo­pto­sis, whereas those cells ­sur­faces of the respi­ra­tory, gas­tro­in­tes­ti­nal, or uro­gen­i­tal tracts.
that can­not inter­act with self-anti­gen/MHC die by apo­pto­sis. Following entry through these bar­ri­ers, most path­o­gens cause a
The remaining cells then undergo neg­ a­
tive selec­ tion, dur­ing local infec­tion in the tis­sues. Innate immune cells rec­og­nize path­
which cells that react too strongly to self-anti­gen/MHC undergo o­gen-asso­ci­ated molec­u­lar pat­terns and dam­age-asso­ci­ated
apo­pto­sis. This pro­cess helps pro­tect the host from T cells that molec­u­lar pat­terns through their germ line-encoded PRRs,
pos­ sess TCRs with high reac­ tiv­
ity against self-pep­ tides pre­ which ini­ti­ates an intra­cel­lu­lar sig­nal­ing cas­cade to acti­vate their
sented by self-MHC, which would lead to auto­im­mu­nity. Follow­ unique effec­tor func­tions. Cytokine and chemokine pro­duc­tion
ing pos­i­tive and neg­a­tive selec­tion, only remaining thy­mo­cytes pro­motes local inflam­ma­tion, attracting cir­cu­lat­ing innate effec­
with mod­er­ate affin­ity for pep­tide/MHC undergo lin­e­age com­ tor cells such as neu­tro­phils and mono­cytes. If the ini­tial innate
mit­ment to become CD4+ or CD8+ T cells, which then migrate to immune response fails to con­ trol the infec­ tion, the adap­ tive
the periph­ery. Within this con­tin­uum of per­mit­ted reac­tiv­ity, the immune response is trig­gered in local lym­phoid tis­sues by the
devel­op­ing CD4+ thy­mo­cytes that pos­sess the highest affin­ity for pre­sen­ta­tion of path­o­gen-derived anti­gens by APCs.
self-pep­tide/MHC develop into reg­u­la­tory T cells (Tregs), char­ Antigen pre­sen­ta­tion: Following path­o­gen expo­sure, APCs
ac­ter­ized by the expres­sion of the tran­scrip­tion fac­tor FoxP3 and pro­cess for­eign pep­tides and pres­ent them on self-MHC. MHC
the sur­face expres­sion of CD25. Tregs are a spe­cial­ized sub­set of class I and class II mol­e­cules func­tion sim­i­larly to deliver and
CD4+ T cells that sup­press acti­vated CD4+ and CD8+ T cells as well pres­ent short pep­tides on the cell sur­face to allow these pep­
as other immune cells. tides to be rec­og­nized by CD8+ or CD4+ T cells, respec­tively.
Once in the periph­ery, naive T cells cir­cu­late until the TCR The pep­tides presented by the dif­fer­ent clas­ses orig­i­nate from
rec­og­ni­tion of pep­tide-MHC complexes induces an intra­cel­lu­ either intra­cel­lu­lar pro­teins for MHC class I or engulfed extra­cel­
lar bio­chem­i­cal sig­nal­ing cas­cade that acti­vates a pro­gram of lu­lar pro­teins for MHC class II. All cells of the body are at risk of
clonal pro­lif­er­a­tion and dif­fer­en­ti­a­tion (with addi­tional input malig­nant trans­for­ma­tion or infec­tion by intra­cel­lu­lar microbes,
from costimulatory mol­e­cules and cyto­kines), thus transform­ which requires a CD8+ T cell response, and thus all­nucle­ated
ing the naive T cell into an effec­tor T cell. CD4+ and CD8+ T cells cells express MHC class I. Meanwhile, only pro­fes­sional APCs,
undergo anal­o­gous dif­fer­en­ti­a­tion pro­cesses, over the span of such as DCs, mac­ro­phages, and B cells, express MHC class II.
sev­ eral days, to acquire func­ tional matu­ rity but play dis­ tinct Additionally, a sub­set of APCs can pres­ent inter­nal­ized anti­gens
func­tional roles in the adap­tive immune response to path­o­gens. in the con­text of MHC class I mol­e­cules via a mech­a­nism called
Naive cells of both lin­e­ages are acti­vated through their TCRs, cross-pre­sen­ta­tion, thus allowing CD8+ T-cell acti­va­tion against
and their dif­fer­en­ti­a­tion is influ­enced by a com­bi­na­tion of sig­ path­o­gens or tumors that do not directly infect DCs. Inflamma­
nals, includ­ing TCR sig­nal strength, costimulatory ligands, and tory cyto­ kines help acti­ vate APCs and pro­ mote upregulation
the local cyto­kine milieu. The inte­gra­tion of these sig­nals drives costimulatory mol­e­cules, such as B7.1 (CD80) and B7.2 (CD86)
the expres­sion of key tran­scrip­tion fac­tors and effec­tor mol­e­ on the APC cell sur­face. Activated APCs then travel from the site
cules, which endow the acti­vated T cell with its indi­vid­u­al­ized of infec­tion to draining lymph nodes, where they pres­ent anti­
func­tion. Activated CD8+ T cells func­tion to induce death in host gen to naive T cells. T cells that rec­og­nize anti­gen:self-MHC on
cells fol­low­ing acti­va­tion, whereas CD4+ T cells func­tion pri­mar­ an acti­vated APC pro­ceed to sig­nal through the multimeric TCR
ily through cyto­kine pro­duc­tion or via direct cell-to-cell con­tact com­plex. Costimulation by the bind­ing of CD28 on the T-cell sur­
to acti­vate other immune cells. Depending on the sig­nals dur­ing face to a B7 mol­e­cule on the APC is required for T cell acti­va­tion.
dif­fer­en­ti­a­tion, naive CD4+ T cells have the poten­tial to develop A fail­ure to receive both the TCR (sig­nal 1) and costimulation (sig­
into 1 of sev­eral prin­ci­pal effec­tor (or “helper”) sub­sets, includ­ing nal 2) typ­i­cally leads to T-cell anergy and apo­pto­sis. T cell acti­
TH1, TH2, TH17, and T fol­lic­u­lar helper (TFH) cells.13 Different sub­sets va­tion can be fur­ther mod­u­lated by addi­tional costimulatory or
of periph­eral T-cell lym­pho­mas have been described that likely coinhibitory sig­nals.17 Activating sig­nals include those pro­vided
arise from these sub­sets.14,15 In addi­tion to the thymically derived via 4-1BB (CD37), OX40 (CD134), and CD27.

Prakash Singh Shekhawat


Fundamentals of immu­nol­ogy  |  283
Adaptive immune response: Following acti­va­tion by APCs, pres­ sures of an immune response can pro­ mote the loss of
anti­gen-spe­cific T cells and B cells undergo clonal expan­sion anti­ge­nic­ity of the tumor itself through sev­eral poten­tial mech­
and dif­fer­en­ti­a­tion over sev­eral days. B cells pro­duce path­o­gen- a­nisms. Lymphomas, and less com­monly leu­ke­mias, can lose
spe­cific antibodies that can acti­vate com­ple­ments to directly kill MHC class I or class II expres­sion.19-23 Additionally, tumor cells
path­o­gens, opsonize path­o­gens to pro­mote phago­cy­to­sis, and can downregulate costimulatory mol­e­cules to pre­vent T cell
pro­mote anti­body-depen­dent cell-medi­ated cyto­tox­ic­ity. Acti­ acti­va­tion, dysregulation of anti­gen-processing machin­ery can
vated CD4+ T cells release cyto­kines to enhance other immune lead to the loss of anti­gen pre­sen­ta­tion, and selec­tive pres­
cell func­tion, such as mac­ro­phage-medi­ated phago­cy­to­sis. sure could lead to the out­growth of a tumor clone that no lon­
Effector CD8+ T cells release proinflammatory cyto­kines and can ger expresses rec­ og­
nized anti­ gens. Furthermore, the tumor
directly kill infected cells via cytol­y­sis. Following path­o­gen clear­ micro­en­vi­ron­ment in many hema­to­logic malig­nan­cies is immu­
ance, the major­ity of anti­gen-spe­cific lym­pho­cytes undergo cell no­sup­pres­sive, with the upregulation of inhib­i­tory ligands on
death, resulting in clonal con­trac­tion. A small sub­set remains the malig­nant cells or other cells. For instance, cer­tain lym­
as long-lived mem­ory lym­pho­cytes that have the capac­ity to phoma cells may overexpress PD-L1/2, which may be driven
respond to the same path­o­gen more rap­idly upon rechallenge, by viral infec­tion or genetic alter­ations.24-26 Immunosuppressive

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/281/1851987/281carty.pdf by guest on 13 December 2021


lead­ing to less severe or sub­clin­i­cal infec­tion. cells in the tumor micro­en­vi­ron­ment, such as Tregs or mac­ro­
Importantly, the immune sys­tem has sev­eral mech­a­nisms to phages, can also dampen the immune response to leu­ke­mias
pre­serve a care­ful bal­ance between allowing appro­pri­ate acti­ and lym­pho­mas.27
va­tion of the immune sys­tem and pre­vent­ing immune overacti­
vation and sub­se­quent tis­sue dam­age of the host. For exam­ple, Modulating the immune sys­tem to over­come immune
in con­trast to the costimulatory sig­nals outlined above, coinhib­ dys­func­tion
itory sig­nals, such as those medi­ated by ligands to programmed Immunotherapeutic approaches to over­come or bypass immune
cell death 1 pro­tein (PD-1), cyto­toxic T-lym­pho­cyte anti­gen dys­func­tion have had clin­i­cal suc­cess in many hema­to­logic dis­
4 (CTLA-4), and lym­pho­cyte-acti­va­tion gene 3 (LAG3), serve to or­ders. The most long-stand­ing exam­ple of immu­no­ther­apy to
sup­press immune acti­va­tion and act as a bio­log­ic ­ al rheo­stat to treat hema­to­logic malig­nan­cies is the allo­ge­neic HSC trans­plant,
pre­vent tis­sue dam­age from a hyper­ac­tive immune response. first used in humans in 1957.28 Discoveries aris­ing from decades
In addi­tion, Tregs medi­ate their immu­no­sup­pres­sive func­tion in of research have found that the reconstituted donor-derived
trans on other immune cells, includ­ing CD4+ and CD8+ T cells, immune sys­tem, par­tic­u­larly donor lym­pho­cytes, medi­ates a
B cells, DCs, NK cells, and mac­ro­phages, through CTLA-4 expres­ graft-ver­sus-leu­ke­mia/lym­phoma effect but also leads to graft-
sion; pro­duc­tion of the sup­pres­sive cyto­kines IL-10, IL-35, and ver­sus-host dis­ease, dur­ing which acti­vated donor T cells rec­og­
transforming growth fac­ tor β; the con­ ver­sion of extra­ cel­lu­
lar nize the host tis­sue as for­eign, lead­ing to tis­sue dam­age.
aden­o­sine tri­phos­phate to aden­o­sine; and the con­sump­tion of More recent immu­no­ther­a­peu­tic advances have been built
local IL-2. on the rec­og­ni­tion that tumor-spe­cific T cells undergo a pro­
cess termed exhaus­tion, dur­ing which chronic anti­gen stim­u­
Mechanisms that result in an inad­e­quate immune la­tion results in T cells becom­ing increas­ingly less respon­sive,
response lead­ing to decreased cyto­kine pro­duc­tion and the inabil­ity to
The increased risk of malig­nan­cies that develop in patients with pro­mote cytol­y­sis.29 Concomitantly, exhausted cells upregu­
pri­mary (more recently termed inborn errors of immu­nity) or late inhib­i­tory cell sur­face recep­tors. The best stud­ied of these
sec­ond­ary (acquired) immunodeficiencies high­lights the impor­ inhib­i­tory recep­tors is PD-1, which binds its ligands, PD-L1
tance of immune sur­veil­lance to pre­vent can­cer.18 The malig­ and PD-L2, expressed on acti­ vated mac­ ro­phages and other
nant trans­for­ma­tion of host cells pres­ents unique chal­lenges APCs. The engage­ment of PD-1 damp­ens the T-cell response,
to the immune response since the cells are self in ori­gin, and which normally occurs after ini­tial TCR acti­va­tion, likely to pre­
self-tol­er­ance may pre­vent the devel­op­ment of a full immune vent exces­sive responses and is sub­se­quently downregulated.
response. Multiple mech­a­nisms in the tumor micro­en­vi­ron­ment Exhausted T cells, how­ever, con­tinue to express this inhib­i­tory
dampen effec­tive anti­tu­mor responses (Table 2). The selec­tive recep­tor.30 Therapeutic targeting of the PD-1 axis has shown effi­
cacy in solid tumors and some lym­pho­mas, includ­ing clas­si­cal
Hodgkin lym­phoma and pri­mary medi­as­ti­nal B-cell lym­phoma.31-34
Table 2. Mechanisms that dampen immune response in the However, only a sub­ set of clin­ i­
cal responses is long-last­
ing,
tumor micro­en­vi­ron­ment and cor­re­spond­ing immu­no­ther­a­ pos­ si­
bly because PD-1/PD-L1 block­ ade alters sig­
nal­
ing path­
peu­tic approaches ways within the responding T cells but not the epi­ge­netic land­
scape.35,36 Current clin­i­cal tri­als are ongo­ing to exam­ine whether
Mechanisms to evade immune the com­bi­na­tion of epi­ge­netic ther­apy—ie, with hypomethyl­
response Immunotherapeutic approaches ating agents or his­ tone deacetylase inhib­ i­
tors—and immune
Loss of anti­ge­nic­ity of tumor cells CAR-T, BiTE check­point block­ade improves response rates or dura­bil­ity.
(ex: loss of MHC, downregulation Another recent advance in mod­u­lat­ing the immune sys­tem
of costimulatory mol­e­cules)
to fight hema­to­logic malig­nan­cies or viral infec­tion is the use
Increased expres­sion of inhib­i­tory Immune check­point block­ade of redirected autol­ o­gous T cells. The most explored of these
ligands on tumor cells or in approaches is the use of CAR-T to treat relapsed/refrac­tory
micro­en­vi­ron­ment
CD19-expressing B-cell malig­nan­cies.37 This approach modifies
Suppressive immune pop­u­la­tions in CD47 block­ade (exper­i­men­tal) autol­o­gous T cells with the intro­duc­tion of an engineered anti­
tumor micro­en­vi­ron­ment
gen recep­tor to pro­mote an anti­tu­mor response. The engineered

284  |  Hematology 2021  |  ASH Education Program


­ nti­gen recep­tor com­bines an extra­cel­lu­lar domain of a sin­gle-
a 10. Alizadeh AA, Eisen MB, Davis RE, et al. Distinct types of dif­fuse large B-cell
chain var­i­able frag­ment, which is a fusion of the var­i­able regions of lym­phoma iden­ti­fied by gene expres­sion pro­fil­ing. Nature. 2000;403(6769):
503-511.
the heavy and light chains of an immu­no­glob­u­lin rec­og­niz­ing the 11. Wilson WH, Young RM, Schmitz R, et al. Targeting B cell recep­tor sig­nal­
CD19 anti­gen, with intra­cel­lu­lar domains of the CD3ζ chain and ing with ibrutinib in dif­fuse large B cell lym­phoma. Nat Med. 2015;21(8):
the sig­nal­ing domain of a costimulatory mol­e­cule, such as CD28 922-926.
or 4-1BB. The addi­tion of the costimulatory domain improves the 12. Shah DK, Zúñiga-Pflücker JC. An over­view of the intrathymic intri­ca­cies of
T cell devel­op­ment. J Immunol. 2014;192(9):4017-4023.
per­sis­tence and effi­cacy of engineered T cells,38 which mir­rors the
13. Zhu J. T helper cell dif­fer­en­ti­a­tion, het­ero­ge­ne­ity, and plas­tic­ity. Cold Spring
known require­ment of costimulation for nor­mal T cell func­tion. Harb Perspect Biol. 2018;10(10):a030338.
Another method of redirecting autol­o­gous T cells includes the 14. Wang T, Feldman AL, Wada DA, et al. GATA-3 expres­sion identifies a high-
use of BiTEs, which are fusion pro­teins containing 2 anti­body rec­ risk sub­set of PTCL, NOS with dis­ tinct molec­u­lar and clin­ i­cal fea­tures.
og­ni­tion domains.39 One domain rec­og­nizes the CD3 com­plex on Blood. 2014;123(19):3007-3015.
15. de Leval L, Rickman DS, Thielen C, et  al. The gene expres­sion pro­file of
autol­o­gous T cells, and a sec­ond rec­og­nizes a sur­face recep­tor nodal periph­eral T-cell lym­phoma dem­on­strates a molec­u­lar link between
pres­ent on the tumor cells, such as CD19 or CD20 on B-cell malig­ angioimmunoblastic T-cell lym­phoma (AITL) and fol­lic­u­lar helper T (TFH)
nan­cies. BiTEs bring the T cells into the prox­im­ity of the tumor cells. Blood. 2007;109(11):4952-4963.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/281/1851987/281carty.pdf by guest on 13 December 2021


cells, and the engage­ment of both domains pro­motes the acti­va­ 16. Jameson SC, Masopust D. Understanding sub­set diver­sity in T cell mem­ory.
Immunity. 2018;48(2):214-226.
tion of T cells, cyto­kine pro­duc­tion, and cyto­tox­ic­ity inde­pen­dent
17. Chen L, Flies DB. Molecular mech­a­nisms of T cell co-stim­u­la­tion and co-
of the TCR spec­i­fic­ity of the T cell. Currently, only blinatumomab, inhi­bi­tion. Nat Rev Immunol. 2013;13(4):227-242.
a CD3xCD19 BiTE, is approved for B-cell pre­cur­sor acute lym­pho­ 18. Kebudi R, Kiykim A, Sahin MK. Primary immu­no­de­fi­ciency and can­cer in
blas­tic leu­ke­mia, but many oth­ers are cur­rently under­go­ing clin­i­ chil­dren: a review of the lit­er­a­ture. Curr Pediatr Rev. 2019;15(4):245-250.
cal tri­als for B-cell leu­ke­mias and lym­pho­mas. 19. Roemer MG, Advani RH, Redd RA, et al. Classical Hodgkin lym­phoma with
reduced β2M/MHC class I expres­sion is asso­ci­ated with infe­rior out­come
A prom­is­ing, yet underexplored, approach is that of targeting inde­pen­dent of 9p24.1 sta­tus. Cancer Immunol Res. 2016;4(11):910-916.
the immu­no­sup­pres­sive tumor micro­en­vi­ron­ment in hema­to­logic 20. Roemer MGM, Redd RA, Cader FZ, et al. Major his­to­com­pat­i­bil­ity com­plex
malig­nan­cies to pro­mote a more effec­tive T-cell response. One class II and programmed death ligand 1 expres­sion pre­dict out­come after
mech­a­nism to achieve this out­come would be to induce mac­ro­ programmed death 1 block­ade in clas­sic Hodgkin lym­phoma. J Clin Oncol.
2018;36(10):942-950.
phage phago­cy­to­sis of transformed cells and thereby enhance
21. Rimsza LM, Roberts RA, Miller TP, et al. Loss of MHC class II gene and pro­
tumor anti­gen pre­sen­ta­tion to host T lym­pho­cytes. This could be tein expres­sion in dif­fuse large B-cell lym­phoma is related to decreased
achieved by block­ade of CD47 (also known as the “don’t eat me tumor immunosurveillance and poor patient sur­vival regard­less of other
sig­nal”) on tumor cells, which enhances tumor anti­gen pre­sen­ta­ prog­nos­tic fac­tors: a fol­low-up study from the Leukemia and Lymphoma
tion to T cells.40 Clinically, CD47 block­ade in lym­phoma patients Molecular Profiling Project. Blood. 2004;103(11):4251-4258.
22. Roberts RA, Wright G, Rosenwald AR, et al. Loss of major his­to­com­pat­i­bil­
has shown some poten­tially prom­is­ing early-phase clin­i­cal trial ity class II gene and pro­tein expres­sion in pri­mary medi­as­ti­nal large B-cell
results.41,42 lym­ phoma is highly coor­ di­nated and related to poor patient sur­ vival.
Blood. 2006;108(1):311-318.
Conflict-of-inter­est dis­clo­sure 23. Brouwer RE, van der Heiden P, Schreuder GM, et al. Loss or downregula­
Shannon A. Carty: no com­pet­ing finan­cial inter­ests to declare. tion of HLA class I expres­sion at the alle­lic level in acute leu­ke­mia is infre­
quent but func­tion­ally rel­e­vant, and can be restored by inter­feron. Hum
Immunol. 2002;63(3):200-210.
Off-label drug use 24. Green MR, Rodig S, Juszczynski P, et al. Constitutive AP-1 activ­ity and EBV
Shannon A. Carty: nothing to disclose. infec­tion induce PD-L1 in Hodgkin lym­pho­mas and posttransplant lymph­
oproliferative dis­or­ders: impli­ca­tions for targeted ther­apy. Clin Cancer
Correspondence Res. 2012;18(6):1611-1618.
25. Chen BJ, Chapuy B, Ouyang J, et al. PD-L1 expres­sion is char­ac­ter­is­tic of a
Shannon A. Carty, University of Michigan, 109 Zina Pitcher Pl, Rm
sub­set of aggres­sive B-cell lym­pho­mas and virus-asso­ci­ated malig­nan­cies.
1526, Ann Arbor, MI 48109; e-mail: scarty@med​­.umich​­.edu. Clin Cancer Res. 2013;19(13):3462-3473.
26. Roemer MG, Advani RH, Ligon AH, et  al. PD-L1 and PD-L2 genetic alter­
References ations define clas­si­cal Hodgkin lym­phoma and pre­dict out­come. J Clin
1. Romani L, Mencacci A, Cenci E, Del Sero G, Bistoni F, Puccetti P. An immu­ Oncol. 2016;34(23):2690-2697.
no­reg­u­la­tory role for neu­tro­phils in CD4+ T helper sub­set selec­tion in mice 27. Höpken UE, Rehm A. Targeting the tumor micro­en­vi­ron­ment of leu­ke­mia
with can­di­di­a­sis. J Immunol. 1997;158(5):2356-2362. and lym­phoma. Trends Cancer. 2019;5(6):351-364.
2. Lionakis MS, Levitz SM. Host con­trol of fun­gal infec­tions: les­sons from basic 28. Cieri N, Maurer K, Wu CJ. 60 years young: the evolv­ing role of allo­ge­neic
stud­ies and human cohorts. Annu Rev Immunol. 2018;36(April):157-191. hema­to­poi­etic stem cell trans­plan­ta­tion in can­cer immu­no­ther­apy. Cancer
3. Vivier E, Artis D, Colonna M, et al. Innate lym­phoid cells: 10 years on. Cell. Res. 2021;81(17):4373-4384.
2018;174(5):1054-1066. 29. McLane LM, Abdel-Hakeem MS, Wherry EJ. CD8 T cell exhaus­tion dur­ing
4. Brack C, Hirama M, Lenhard-Schuller R, Tonegawa S. A com­plete immu­no­ chronic viral infec­tion and can­cer. Annu Rev Immunol. 2019;37(April):457-495.
glob­u­lin gene is cre­ated by somatic recom­bi­na­tion. Cell. 1978;15(1):1-14. 30. Day CL, Kaufmann DE, Kiepiela P, et  al. PD-1 expres­sion on HIV-spe­cific
5. Seidman JG, Edgell MH, Leder P. Immunoglobulin light-chain struc­tural gene T  cells is asso­ci­ated with T-cell exhaus­tion and dis­ease pro­gres­sion.
sequences cloned in a bac­te­rial plas­mid. Nature. 1978;271(5645):582-585. Nature. 2006;443(7109):350-354.
6. Yanagi Y, Yoshikai Y, Leggett K, Clark SP, Aleksander I, Mak TW. A human T 31. Ansell SM, Lesokhin AM, Borrello I, et al. PD-1 block­ade with nivolumab in
cell-spe­cific cDNA clone encodes a pro­tein hav­ing exten­sive homol­ogy to relapsed or refrac­tory Hodgkin’s lym­phoma. N Engl J Med. 2015;372(4):
immu­no­glob­u­lin chains. Nature. 1984;308(5955):145-149. 311-319.
7. Hedrick SM, Cohen DI, Nielsen EA, Davis MM. Isolation of cDNA clones 32. Younes A, Santoro A, Shipp M, et  al. Nivolumab for clas­si­cal Hodgkin’s
encoding T cell-spe­cific mem­brane-asso­ci­ated pro­teins. Nature. 1984;308 lym­phoma after fail­ure of both autol­o­gous stem-cell trans­plan­ta­tion and
(5955): 149-153. brentuximab vedotin: a multicentre, multicohort, sin­gle-arm phase 2 trial.
8. Melchers F. Checkpoints that con­trol B cell devel­op­ment. J Clin Invest. Lancet Oncol. 2016;17(9):1283-1294.
2015;125(6):2203-2210. 33. Zinzani PL, Ribrag V, Moskowitz CH, et al. Safety and tol­er­a­bil­ity of pem­
9. Cyster JG, Allen CDC. B cell responses: cell inter­ac­tion dynam­ics and deci­ brolizumab in patients with relapsed/refrac­tory pri­mary medi­as­ti­nal large
sions. Cell. 2019;177(3):524-540. B-cell lym­phoma. Blood. 2017;130(3):267-270.
Prakash Singh Shekhawat
Fundamentals of immu­nol­ogy  |  285
34. Armand P, Rodig S, Melnichenko V, et al. Pembrolizumab in relapsed or refrac­ 39. Goebeler ME, Bargou RC. T cell-engag­ing ther­a­pies—BiTEs and beyond.
tory pri­mary medi­as­ti­nal large B-cell lym­phoma. J Clin Oncol. 2019;37(34): Nat Rev Clin Oncol. 2020;17(7):418-434.
3291-3299. 40. Liu X, Pu Y, Cron K, et al. CD47 block­ade trig­gers T cell-medi­ated destruc­
35. Pauken KE, Sammons MA, Odorizzi PM, et al. Epigenetic sta­bil­ity of exhausted tion of immu­no­genic tumors. Nat Med. 2015;21(10):1209-1215.
T cells lim­its dura­bil­ity of reinvigoration by PD-1 block­ade. Science. 2016; 41. Advani R, Flinn I, Popplewell L, et  al. CD47 block­ade by Hu5F9-G4 and
354(6316):1160-1165. rituximab in non-Hodgkin’s lym­phoma. N Engl J Med. 2018;379(18):1711-1721.
36. Ghoneim HE, Fan Y, Moustaki A, et  al. De novo epi­ge­netic pro­grams 42. Ansell SM, Maris MB, Lesokhin AM, et al. Phase I study of the CD47 blocker
inhibit PD-1 block­ade-medi­ated T cell reju­ve­na­tion. Cell. 2017;170(1):142-157. TTI-621 in patients with relapsed or refrac­tory hema­to­logic malig­nan­cies.
e19157e19. Clin Cancer Res. 2021;27(8):2190-2199.
37. June CH, O’Connor RS, Kawalekar OU, Ghassemi S, Milone MC. CAR T cell
immu­no­ther­apy for human can­cer. Science. 2018;359(6382):1361-1365.
38. Milone MC, Fish JD, Carpenito C, et  al. Chimeric recep­ tors containing
CD137 sig­nal trans­duc­tion domains medi­ate enhanced sur­vival of T cells © 2021 by The Amer­i­can Society of Hematology
and increased anti­leu­ke­mic effi­cacy in vivo. Mol Ther. 2009;17(8):1453-1464. DOI 10.1182/hema­tol­ogy.2021000260

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/281/1851987/281carty.pdf by guest on 13 December 2021

286  |  Hematology 2021  |  ASH Education Program


IMMUNOLOGY 101: WHAT THE PRACTICING HEMATOLOGIST NEEDS TO KNOW

How immunodeficiency can lead to malignancy


Sung-Yun Pai,1 Kathryn Lurain,2 and Robert Yarchoan2

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/287/1852069/287pai.pdf by guest on 13 December 2021


Immune Deficiency Cellular Therapy Program, Center for Cancer Research, National Cancer Institute, Bethesda, MD; and 2HIV and AIDS Malignancy
1

Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD

Immunodeficiency, whether acquired in the case of human immunodeficiency virus (HIV) infection or congenital due to
inborn errors of immunity (IEIs), presents clinically with not only infection and immune dysregulation but also increased
risk of malignancy. The range of malignancies seen is relatively limited and attributable to the particular cellular and
molecular defects in each disease. CD4+ T-cell lymphopenia in people living with HIV infection (PLWH) and certain IEIs
drive the predisposition to aggressive B-cell non-Hodgkin lymphomas, including certain rare subtypes rarely seen in
immunocompetent individuals. PLWH and IEI that lead to profound T-cell lymphopenia or dysfunction also are at risk
of cancers related to oncogenic viruses such as Kaposi sarcoma herpesvirus, Epstein-Barr virus, human papillomavirus
(HPV), and Merkel cell polyomavirus. IEIs that affect natural killer cell development and/or function heavily predispose
to HPV-associated epithelial cancers. Defects in DNA repair pathways compromise T- and B-lymphocyte development
during immune receptor rearrangement in addition to affecting hematopoietic and epithelial DNA damage responses,
resulting in both hematologic and nonhematologic cancers. Treatment of cancers in immunodeficient individuals should
be curative in intent and pursued in close consultation with disease experts in immunology and infectious disease.

LEARNING OBJECTIVES
• Understand the immunologic defects in PLWH infection and their relationship to the development of cancer
• Recognize that IEIs affecting lymphocytes and/or DNA repair predispose to cancer

and positive for CD138, CD38, and CD45, as well as both


CLINICAL CASE 1, PART 1 Kaposi sarcoma herpesvirus (KSHV) and Epstein­Barr virus
A 28­year­old cisgender man who immigrated to the (EBV), indicating a diagnosis of primary effusion lym­
United States from Uganda 10 years ago presents to phoma (PEL).
the emergency room with shortness of breath. He also
reports 6 months of weight loss, fatigue, and fevers. On
examination, the patient is in respiratory distress and has Cancers arising in people with HIV/AIDS
raised, violaceous plaques on his trunk and extremities. Cancer and HIV/AIDS have been linked since the first cluster
Chest x­ray reveals a large left­sided pleural effusion. of cases of KS and pneumocystis pneumonia were reported
The patient undergoes testing for human immunodefi­ in 1981. It was soon recognized that people living with HIV
ciency virus (HIV), which is positive. His HIV viral load is (PLWH) were susceptible to other cancers, particularly
150 000 copies/mL, with a CD4+ T­cell count of 110 cells/µL. aggressive B­cell non­Hodgkin lymphomas (NHLs) (Table 1).1
After consultation with an HIV specialist and a pharma­ With the discovery of KSHV (also called human herpesvi­
cist, the patient is started on combination antiretroviral rus 8) in 1994 and its subsequent finding as the causative
therapy (ART) including bictegravir, emtricitabine, and agent of KS, it became apparent that many, but not all, HIV­
tenofovir alafenamide along with sulfamethoxazole/ associated cancers are caused by oncogenic viruses.2
trimethoprim and valacyclovir for opportunistic infection HIV infects CD4+ T cells and certain other cell types, and
prophylaxis. Biopsy specimen of one of the skin lesions is the primary immune defect is CD4+ T­cell lymphopenia. HIV
consistent with Kaposi sarcoma (KS). Preliminary results infection also causes other disruptions of immune function,
from a therapeutic and diagnostic thoracentesis indicate including depletion of other lymphocyte populations, defects
a B­cell lymphoma. Further pathologic workup indicates in lymphocyte function, increased inflammatory cytokines,
the malignant B cells are negative for CD20 and CD19 and chronic inflammation. These immunologic defects impair
Prakash Singh Shekhawat
How immunodeficiency can lead to malignancy | 287
Table 1. Etiology and inci­dence of can­cers among PLWH

Role of SIR in the


Etiologic virus Cancer type immu­no­sup­pres­sion Other risk fac­tors United States3,4 Comment
KSHV Kaposi sar­coma +++ Men who have sex with men 498 Very com­mon in parts of
sub-Saharan Africa
EBV Non-Hodgkin lym­phoma* ++ to ++++ HBV, HCV 12
Hodgkin lym­phoma ++ Male sex 7.7 Mixed cel­lu­lar­ity is most
com­mon sub­type
HPV Anal can­cer + Tobacco, anal warts 19 Incidence increas­ing
Cervical can­cer + Tobacco 3.2 Very com­mon in resource-
lim­ited regions
Oropharyngeal can­cer + Tobacco, alco­hol 1.6-2.2† Incidence increas­ing

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/287/1852069/287pai.pdf by guest on 13 December 2021


Vulvar can­cer + Tobacco 9.4
Penile can­cer + Tobacco, lack of cir­cum­ci­sion 5.3
HBV, HCV Hepatocellular can­cer + Alcohol, cir­rho­sis 3.2
MCPyV Merkel cell car­ci­noma + Sun expo­sure 2.6 At least 15% of cases not
asso­ci­ated with MCPyV
None or Lung can­cer + Tobacco, pul­mo­nary 2 Most com­mon can­cer-related
unknown infec­tions cause of death
Nonmelanoma skin can­cer + Sun expo­sure, fam­ily his­tory 2.1
Conjunctival car­ci­noma + Sun expo­sure 5.5 Rare in the United States;
occurs most often in
sub-Saharan Africa; viral
eti­­ol­ogy suspected
*See Figure 2.

SIR is 1.6 for HPV-related can­cers and 2.2 for HPV-unre­lated can­cers.
SIR, stan­dard­ized inci­dence ratio; +, low; ++, moderate; +++, significant; ++++, very significant role for immunosuppression in pathogenesis.

con­trol of onco­genic viruses, such as KSHV, EBV, human pap­il­lo­ma­ pres­sion depending on the sub­type (Figure 2). Hodgkin lym­
vi­rus (HPV), and Merkel cell polyomavirus (MCPyV), lead­ing to the phoma is also increased but gen­er­ally occurs at pre­served CD4+
devel­op­ment of can­cer (Table 1, Figure 1). KSHV is an essen­tial cause T-cell counts and is asso­ci­ated with use of ART.7 Diffuse large
of KS, and pop­u­la­tions with a higher prev­al­ence of KSHV infec­tion B-cell (DLBCL) is the most com­mon NHL sub­type in PLWH, fol­
expe­ri­ence higher rates of KS, such as men who have sex with men lowed by Bur­kitt lym­phoma and rare sub­types occur­ring almost
and those liv­ing in cer­tain areas of sub-Saharan Africa. PLWH also exclu­sively in PLWH, par­ tic­

larly PEL and plasmablastic lym­
have a higher expo­sure to high-risk types of HPV, increas­ing the phoma. The inci­dence of pri­mary cen­tral ner­vous sys­tem lym­
risk for cer­vi­cal and anal can­cer. Increased rates of tobacco use and phoma, which is asso­ci­ated with pro­found immu­no­sup­pres­sion,
infec­tion with hep­a­ti­tis B virus (HBV) and hep­a­ti­tis C virus (HCV) dras­ti­cally declined with the advent of ART and remains rare.
also con­trib­ute to higher can­cer rates in PLWH (Table 1). Lymphoma often pres­ents at advanced stages in PLWH with
The devel­op­ment of an effec­tive 3-drug ART in the mid-1990s more rapid pro­ gres­sion, fre­quent B symp­ toms, and fre­ quent
allowed for mean­ing­ful res­to­ra­tion of CD4+ T-cell counts, and involve­ ment of extranodal sites and the cen­ tral ner­
vous sys­
AIDS was converted from a death sen­tence to a chronic dis­ease. tem than the gen­eral pop­u­la­tion.8 The depth of the CD4+ nadir,
ART sub­stan­tially reduced the inci­dence of the can­cers asso­ci­ uncon­trolled HIV vire­mia, and inter­rup­tions in ART are all­asso­ci­
ated with pro­found immu­no­de­fi­ciency such as KS and cer­tain ated with increased risk of NHL.9-11 A rel­a­tively large per­cent­age
B-cell lym­pho­mas. However, as PLWH live lon­ger, they have an of HIV-asso­ci­ated lym­pho­mas is asso­ci­ated with EBV, includ­ing
increased cumu­la­tive like­li­hood of devel­op­ing can­cers asso­ci­ almost 100% of pri­mary cen­tral ner­vous sys­tem lym­pho­mas.12
ated with mod­er­ate decreases in CD4+ T-cell counts (Table 1). KSHV is the caus­a­tive agent of PEL, but most are also coinfected
PLWH also remain at risk of can­cers not asso­ci­ated with immu­ with EBV. Chronic B-cell acti­va­tion in PLWH con­trib­utes to lym­
no­de­fi­ciency. In fact, can­cer is now one of the most com­mon or phomagenesis, and coinfection with HBV and/or HCV may also
the most com­mon cause of death in PLWH in regions where ART increase the risk of NHL through this mech­a­nism.13
is read­ily avail­­able. Interestingly, PLWH do not have an increased As exem­pli­fied by case 1, patients with malig­nan­cies that are
inci­dence of cer­tain com­mon can­cers, such as breast, colon, and more fre­quent in HIV/AIDS should be screened for HIV infec­tion,
pros­tate can­cer; how­ever, they may pres­ent at more advanced espe­cially if they are in a high-risk group. Before the devel­op­ment
stages or with more aggres­sive sub­types.5,6 of effec­tive ART, treat­ment with cura­tive-intent che­mo­ther­apy for
NHL is the most com­mon can­cer among PLWH in the United HIV-asso­ci­ated malig­nan­cies was nearly impos­si­ble given the pro­
States.3 NHL is asso­ci­ated with mod­er­ate to severe immu­no­sup­ found immu­no­sup­pres­sion and risk of infec­tious ­com­pli­ca­tions.

288  |  Hematology 2021  |  ASH Education Program


Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/287/1852069/287pai.pdf by guest on 13 December 2021

Figure 1. Key relationships between immune defects and oncogenesis. General relationships are shown in A, and key relationships
specific to the indicated diseases are shown in B-H. Yellow boxes denote key immunologic defects observed in HIV/AIDS and/or IEI;
green boxes denote secondary defects; orange boxes denote infectious agents that contribute to malignancies in ­immunodeficiency;
purple ovals show the principal malignancies associated with the immune defects. DC, dendritic cell; HHV-8, human herpesvirus 8.

Prakash Singh Shekhawat


How immu­no­de­fi­ciency can lead to malig­nancy  |  289
Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/287/1852069/287pai.pdf by guest on 13 December 2021
Figure 2. Pathologic, immune, and viral characteristics of HIV-associated NHL. ABC, activated B cell; GCB, germinal center B cell;
IRF4, interferon regulatory factor 4; MUM1, multiple myeloma 1; PBL, plasmablastic lymphoma. *Indicates MYC gene rearrangements.
This figure was created on biorender​.com.

With ART, it has been shown that most can­cers aris­ing in PLWH
are best treated with the same reg­ i­
mens used to treat HIV- CLINICAL CASE 1, PART 2
neg­a­tive patients.14,15 The National Comprehensive Cancer Net­ For the patient’s PEL, an oncol­ o­
gist pre­scribes cura­ tive-intent
work has published prac­tice guide­lines for can­cer in peo­ple with treat­ment with dose-adjusted etoposide, pred­ni­sone, vin­cris­tine,
HIV, HIV-related NHL, and KS. PLWH should con­tinue ART while cyclo­phos­pha­mide, and doxo­ru­bi­cin (EPOCH) in addi­tion to con­
under­go­ing can­cer ther­apy, and oncol­o­gists should work closely tinu­ing ART. After 6 cycles of EPOCH, the patient’s lym­phoma is in
with HIV spe­cial­ists to ensure opti­mal HIV man­age­ment and infec­ com­plete remis­sion, his CD4+ T cell count is 90 cells/µL, and his
tion pro­phy­laxis. Oncologists should con­sult with phar­ma­cists to HIV viral load is unde­tect­able. Two months later, the patient devel­
avoid drug-drug inter­ac­tions, espe­cially if patients are tak­ing HIV ops increas­ing num­bers of nod­u­lar KS lesions on his lower extrem­
pro­te­ase inhib­i­tors, which are metab­o­lized by the cyto­chrome i­ties. His oncol­o­gist pre­scribes lipo­so­mal doxo­ru­bi­cin to treat the
P450 enzymes. One of the most excit­ing devel­op­ments in can­ KS while the patient’s immune sys­ tem recon­ sti­
tutes fol­low­ing
cer ther­apy is the suc­cess of immune check­point inhib­it­ors tar­ EPOCH. After 4 cycles, there is sig­nif­i­cant flat­ten­ing and light­en­ing
geting PD-1, PD-L1, or CTLA-4. Despite ini­tial tox­ic­ity and effi­cacy of the KS lesions, and lipo­so­mal doxo­ru­bi­cin is discontinued. One
con­cerns, recent stud­ies have shown them to be safe and effec­ year fol­low­ing EPOCH treat­ment, the patient’s PEL is in remis­sion,
tive in expected can­cer types in PLWH with more than 100 CD4+ the cuta­ne­ous KS lesions are barely vis­i­ble, CD4+ T-cell count is
T cells/µL.16 It should be stressed that HIV infec­tion is a chronic, 350 cells/µL, and HIV viral load remains unde­tect­able.
man­ age­
able con­ di­
tion, and US Food and Drug Administration
guid­ance endorses inclu­sion of PLWH with 350 or more CD4+
cells/µL for all­can­cer clin­i­cal tri­als as well as their inclu­sion at
lower CD4+ T cell counts if patients have poten­tially cur­able malig­
nan­cies or if the study agents have been shown to have activ­ity CLINICAL CASE 2
in a given can­cer.17 PLWH treated for can­cer are at increased risk A 25-year-old man with a his­tory of recur­rent respi­ra­tory tract
for sec­ond­ary pri­mary can­cers, fur­ther underscoring the need for infec­tions was referred to an immu­nol­o­gist for eval­u­a­tion. He
vig­i­lance and adher­ence to can­cer screen­ing guide­lines.18 was found to have dysgammaglobulinemia, poor anti­ gen-

290  |  Hematology 2021  |  ASH Education Program


spe­cific responses, hyper-IgM, and IgA defi­ciency, con­sis­tent lymphopenia and fur­ther­more increase the chance of trans­lo­
with a diag­no­sis of com­mon var­i­able immu­no­de­fi­ciency. Sus­ ca­tions asso­ci­ated with lym­phoma. Outside of lym­pho­cytes,
pecting an inborn error of immu­ nity (IEI), the immu­ nol­o­gist muta­tions in these genes and oth­ers involved in homol­o­gous
ordered whole-exome sequenc­ing. While awaiting fur­ther test­ recom­bi­na­tion and the response to DNA dam­age (eg, BLM, the
ing, the patient presented to a local emer­gency room with gene mutated in Bloom syn­drome) ren­der the patients radi­a­tion
cer­vi­cal lymph­ade­nop­a­thy and a medi­as­ti­nal mass and was sen­si­tive. Oncogenesis is com­mon par­tic­u­larly in rap­idly divid­ing
treated with emer­gent cor­ti­co­ste­roids. The biopsy spec­i­men tis­sues, includ­ing skin, gas­tro­in­tes­ti­nal tract, breast, gen­i­to­uri­
was ini­tially read as Hodgkin lym­phoma, but his immu­nol­o­gist nary tract, soft tis­sues, and bone mar­row.
questioned the diag­ no­sis and requested EBV test­ ing of the Defects of hema­to­poi­etic stem cell (HSC) fit­ness, dif­fer­en­ti­
tumor. Rereview of pathol­ogy found that what were thought a­tion, and/or sur­vival con­trib­ute to bone mar­row fail­ure, in the
to be Reed-Sternberg cells were large atyp­i­cal cells that had set­ting of rep­li­ca­tive stress and clonal hema­to­poi­e­sis of a lim­
com­pletely effaced the nodal archi­tec­ture and were pos­i­tive ited pool of HSCs. Accumulation of addi­tional muta­tions pro­
for EBV, CD20, PAX5, and CD30, con­sis­tent with EBV+ DLBCL motes the devel­op­ment of myelodysplasia and ulti­mately acute
­lym­phoma. Whole-exome sequenc­ing showed a het­ero­zy­gous mye­loid leu­ke­mia. In these dis­or­ders (eg, Shwachman-Diamond

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/287/1852069/287pai.pdf by guest on 13 December 2021


E1021K var­i­ant in the PIK3CD gene. After fur­ ther lym­ phoma syn­drome, dyskeratosis congenita, or severe con­gen­i­tal neutro­
treat­ment, he under­went suc­cess­ful allo­ge­neic trans­plan­ta­tion penia), the immu­no­de­fi­ciency is typ­i­cally related to neutropenia
with a matched unre­lated donor. rather than defects of lym­pho­cytes or adap­tive immu­nity.

Individual dis­or­ders with more than 1 mech­a­nism involved


Cancers aris­ing in peo­ple with IEIs Wiskott-Aldrich syn­drome and ded­i­ca­tor of cyto­ki­ne­sis 8
Advances in genet­ics have led to a ver­i­ta­ble explo­sion of defined defi­ciency.  Wiskott-Aldrich syn­drome (WAS) is an X-linked dis­or­
pri­mary immu­no­de­fi­ciency dis­eases, now termed IEIs. The Inter­ der caused by muta­tions in the WAS gene, resulting in a syn­drome
national Union of Immunological Societies (IUIS) cur­rently lists of microthrombocytopenia, eczema, infec­tions, auto­im­mu­nity,
more than 450 dis­eases, cat­e­go­rized broadly into 10 phe­no­typic and pre­dis­po­si­tion to lym­phoma. WAS pro­tein is crit­i­cal for actin
groups (Table 2).19-21 The speed with which these dis­or­ders are cyto­skel­e­tal rearrangement, which in turn is required for for­ma­
being described is evi­dent by the num­ber of dis­or­ders in each tion of the immune syn­apse in response to T, B, and NK cell-cell
bien­nial pub­li­ca­tion (191 in 2011, 430 in 2019) and the fact that the sig­nal­ing, for reg­u­la­tory T-cell func­tion, mono­cyte cyto­kine pro­
2019 meet­ing report, published in Jan­u­ary 2020, was suf­fi­ciently duc­tion, and lym­pho­cyte and den­dritic cell shape and mobil­ity.24
out­dated that a sup­ple­ment adding 26 addi­tional mono­genic Progressive CD8+ T-cell lymphopenia, impaired T cell receptor
gene defects was published in Jan­u­ary 2021. sig­nal­ing and acti­va­tion, poor NK cell func­tion, over­ac­tive B cell
At least 10% of IEIs are stated within the IUIS reports to be receptor sig­nal­ing, and dysregulated mono­cyte acti­va­tion all­
asso­ci­ated with malig­nan­cies. The dis­tri­bu­tion of IEIs asso­ci­ated con­ spire to pro­ mote lymphomagenesis with and with­ out EBV
with malig­nancy is not even with respect to cat­e­gory; very few infec­tion (Figure 1).
fall into the categories of defects in intrin­sic and innate immu­ Dedicator of cyto­ki­ne­sis 8 (DOCK8) defi­ciency is an auto­so­
nity, com­ple­ment defects, and autoinflammatory dis­or­ders.22,23 mal reces­sive dis­or­der that shares fea­tures with WAS, includ­ing
As reviewed below, dis­tinct and overlapping mech­a­nisms are eczema, infec­tions, and auto­im­mu­nity, but also is char­ac­ter­ized
respon­si­ble for a pre­dis­po­si­tion to malig­nancy among patients by her­petic viral skin infec­tions and HPV-driven malig­nancy.25
with IEIs (Figure 1). DOCK8 is also crit­i­cal for actin cyto­skel­e­tal func­tion and func­
tions in a com­ plex with WAS pro­ tein. Immune syn­apses are
General path­o­genic mech­a­nisms poorly formed in DOCK8-defi­cient cells, sim­i­lar to WAS. Several
As in HIV/AIDS, T-cell lymphopenia and dys­func­tion gen­er­ally dis­tinct cel­lu­lar defects in DOCK8-defi­cient patients that may
pre­dis­pose to both poor tumor sur­veil­lance and oppor­tu­nis­tic under­lie HPV-driven can­cers include severe impair­ment of den­
infec­tion with onco­genic viruses. T-cell dys­func­tion may lead to dritic cell motil­ity in skin, decreased num­bers of plasmacytoid
dysregulated pro­lif­er­a­tion of B cells, which may be fur­ther exac­ den­dritic cells that fail to pro­duce inter­feron α, and increased
er­bated by con­di­tions that lead to B-cell lymphopenia or intrin­ death of immune cells, includ­ing skin-res­i­dent CD8+ T cells, due
si­cally aber­rant B-cell sig­nal­ing. to frag­men­ta­tion while attempting to tra­verse through tis­sues
Natural killer (NK) cells are crit­ i­
cal for con­
trol of cer­tain (Figure 1).26
viruses, par­tic­u­larly DNA viruses such as HPV and EBV. Control
of viral infec­tions in skin or tis­sue is often depen­dent on anti­ Common var­i­able immu­no­de­fi­ciency.  Patients with com­mon
gen presenting cells such as mono­ cytes and den­ dritic cells, var­i­able immu­no­de­fi­ciency (CVID) have defi­ciency of anti­body
which must undergo acti­va­tion, changes in shape, and migra­tion pro­duc­tion and humoral immu­nity, with vary­ing degrees of T-
through tis­sues to inter­act with T cells and traf­fic to sec­ond­ary cell dys­func­tion and immune dysregulation (Figure 1). Because
lym­phoid organs. most patients with CVID are not genet­i­cally char­ac­ter­ized, there
Mutations in a host of genes involved in DNA repair pre­dis­ is broad het­ero­ge­ne­ity in dis­ease man­i­fes­ta­tions. Lymphomas
pose to lym­phoid malig­nancy and epi­the­lial malig­nancy, due and lymphoproliferation are com­mon, occur­ring in 17.2% of 1091
to the gen­eral need for faith­ful DNA repair in all­tis­sues. Repair patients with CVID reg­is­tered in the US Immunodeficiency Net­
of dou­ ble-stranded breaks gen­ er­
ated in the T-cell recep­ tor work.27 B-cell lymphopenia is com­mon, and the addi­tional rep­
and immu­no­glob­u­lin loci by RAG1/RAG2 depends on the non­­ li­ca­tive stress on B cells in the con­text of chronic inflam­ma­tion
ho­mol­o­gous end-join­ing path­way. Thus, defects in genes such is a likely con­trib­u­tor. In con­trast to the dis­tri­bu­tion of B-cell
as LIG4, DCLRE1C, XLF1, and NBS1 tend to result in T- and B-cell lym­pho­mas in oth­er­wise healthy adults, there are few fol­lic­u­lar
Prakash Singh Shekhawat
How immu­no­de­fi­ciency can lead to malig­nancy  |  291
Table 2. IEIs asso­ci­ated with malig­nan­cies

Associated malig­nan­cies
Myelodysplastic Gastric ade­
B- or T-cell lym­pho­mas HPV-driven syn­drome and/or no­car­ci­noma
Number IUIS cat­e­gory Examples (ref­er­ence) can­cers mye­loid leu­ke­mia or lym­phoma Other
1 Immunodeficiencies SCID SCID (ADA, RAG1, LIG4, — — — OX40 defi­ciency
affect­ing DCLRE1C, coronin 1a) (KS)
cel­lu­lar and ZAP70 CD40L
humoral immu­nity (cholangio
carcinoma)
2 Combined WAS WAS — — Ataxia-tel­an­gi­ Ataxia-tel­an­gi­ec­
immunodeficiencies Ataxia- DOCK8 defi­ciency ec­ta­sia ta­sia (breast,
with asso­ci­ated or tel­an­gi­ec­ CARD11 auto­so­mal ­ thy­roid, liver,
syndromic fea­tures ta­sia dom­i­nant loss of brain, acute

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/287/1852069/287pai.pdf by guest on 13 December 2021


func­tion lym­pho­blas­tic
Ataxia-tel­an­gi­ec­ta­sia leu­ke­mia)
Nijmegen break­age
syn­drome
Bloom syn­drome
Immunodeficiency
with cen­tro­meric
insta­bil­ity and facial
anom­a­lies
Schimke immuno-
osse­ous dys­pla­sia
Cartilage hair
hypo­pla­sia
DiGeorge syn­drome
CHARGE syn­drome
PNP defi­ciency
3 Predominantly CVID APDS — — CVID —
anti­body CVID Selective IgA
deficiencies Selective IgA defi­ciency
defi­ciency
4 Diseases of immune ALPS ALPS — — — —
dysregulation IPEX X-linked
HLH lymphoproliferative
dis­ease type 1
XMEN
ITK defi­ciency
CD27/CD70 defi­ciency
CTPS1 defi­ciency
IL10R defi­ciency
5 Congenital defects Chronic gran­ — GATA2 GATA2 — —
of phago­cyte u­lo­ma­tous defi­ciency defi­ciency
num­ber or dis­ease Severe
func­tion Severe con­gen­i­tal
con­gen­i­tal neutropenia
neutropenia (ELANE, HAX1,
Leukocyte other)
adhe­sion Shwachman-
defi­ciency Diamond
syn­drome
X-linked
neutropenia
due to gain of
func­tion muta­
tions in WAS
SMARCAD2
6 Defects in intrin­sic IL-12 and WHIM syn­drome Epidermo­­ — — —
and innate inter­feron dysplasia
immu­nity γ path­way verruciformis
defects WHIM syn­drome

292  |  Hematology 2021  |  ASH Education Program


Table 2. (continued)

Associated malig­nan­cies
Myelodysplastic Gastric ade­
B- or T-cell lym­pho­mas HPV-driven syn­drome and/or no­car­ci­noma
Number IUIS cat­e­gory Examples (ref­er­ence) can­cers mye­loid leu­ke­mia or lym­phoma Other
7 Autoinflammatory Familial — — — — —
dis­or­ders Med­i­ter­ra­
nean fever
Aicardi-
Goutieres
syn­drome
8 Complement Hereditary — — — — —
deficiencies angioedema

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/287/1852069/287pai.pdf by guest on 13 December 2021


9 Bone mar­row fail­ure Fanconi Dyskeratosis congenita — Fanconi ane­mia — Fanconi ane­mia
ane­mia Dyskeratosis (breast, skin,
Dyskeratosis congenita head and
congenita Coats plus neck, other)
syn­drome
MIRAGE (SAMD9)
Ataxia
pan­cy­to­pe­
nia syn­drome
(SAMD9L)
10 Phenocopies of IEI Somatic — — — — —
muta­tions
lead­ing to
ALPS
RAS-
asso­ci­ated
auto­im­mune
leukopro-
liferative
dis­ease
CHARGE, coloboma, heart anomaly, choanal atresia, retardation, genital and ear anomalies; HLH, hemophagocytic lymphohistiocytosis; IPEX,
immune dysregulation, polyendocrinopathy, enteropathy, X-linked; MIRAGE, myelodysplasia, infection, restriction of growth, adrenal hypoplasia,
genital phenotypes, and enteropathy; PNP, purine nucleoside phosphorylase; RAS, rat sarcoma; SCID, severe combined immunodeficiency; WHIM,
warts, hypogammaglobulinemia, infections, and myelokathexis.

lym­ pho­ mas reported, skewing instead to extranodal mar­ ginal (PI3K-δ) pro­tein. These muta­tions result in increased PI3K sig­nal­
zone lym­ phoma and mucosa-asso­ ci­
ated lym­ phoid tis­ sue lym­ ing in lym­pho­cytes, where PI3K-δ is expressed.30 The fact that
phoma.28 Gastric can­cers occur in patients with CVID more fre­ muta­ tions in p110δ respon­ si­
ble for acti­vated PI3K-δ syn­drome
quently than the gen­eral pop­u­la­tion and appear to be related to (APDS) are pres­ent as somatic muta­tions in DLBCL, man­tle cell
chronic inflam­ma­tion and cell pro­lif­er­a­tion, pos­si­bly in con­junc­ lym­phoma, and rarely Bur­kitt lym­phoma sup­ports a B-cell intrin­
tion with Helicobacter pylori infec­tion. In an Ital­ian reg­is­try of 455 sic role of over­ac­tive PI3K sig­nal­ing in lymphomagenesis due to
patients with CVID, 25 patients had gas­tric can­cer, with a stan­ uncon­trolled B-cell pro­lif­er­a­tion in the con­text of class switch
dard­ized inci­dence ratio of 6.4, and this was the highest cause recom­bi­na­tion in ger­mi­nal cen­ters. These patients may have lym­
of death. The gas­tric can­cers in the Ital­ian series arose out of a phoma, as in case 2, even before IEI is fully rec­og­nized. In addi­
back­ground of atro­phic gas­tri­tis, intes­ti­nal meta­pla­sia, or dys­pla­ tion, T cells in patients with APDS have impaired func­tion and
sia.29 Lymphoid and/or gran­u­lo­ma­tous infil­tra­tion of the gas­tro­ cyto­tox­ic­ity, have exhausted CD8+ CD57+ T cells, and com­monly
in­tes­ti­nal tract or lungs may fur­ther trig­ger excess immune cell develop T-cell lymphopenia and low naive T-cell counts over time
pro­lif­er­a­tion. Although the inci­dence of malig­nant or pro­lif­er­a­tive (Figure 1).31
dis­or­ders is clearly increased, rou­tine sur­veil­lance is unlikely to be
effec­tive for early detec­tion. Meticulous sup­port­ive care, main­ Ataxia-tel­an­gi­ec­ta­sia.  Biallelic muta­tions in the auto­so­mal gene
taining a high degree of sus­pi­cion for malig­nancy, and aggres­sive ATM result in the syn­drome of ataxia-tel­an­gi­ec­ta­sia, char­ac­ter­ized
workup of symp­toms are key to man­ag­ing patients with CVID. by immu­no­de­fi­ciency, pro­gres­sive cer­e­bel­lar ataxia, oculocuta­
neous tel­an­gi­ec­ta­sia, and can­cer pre­dis­po­si­tion.32 ATM is a kinase
Activated phosphoinositide 3-kinase-δ syn­drome. A sub­set of that has at least 2 crit­i­cal roles in DNA repair. ATM is acti­vated dur­
patients pre­vi­ously diag­nosed with CVID have been shown to ing dou­ble-stranded breaks gen­er­a­tion (includ­ing dur­ing variable-
har­
bor gain-of-func­ tion muta­ tions in the genes encoding the diversity-joining region recom­bi­na­tion) and recruits the MRE11/
cat­a­lytic sub­unit (p110δ) or biallelic loss-of-func­tion muta­tions in RAD51/NBS1 com­plex in a feed-for­ward cycle by phos­phor­
the reg­u­la­tory sub­unit (p85α) of the phosphoinositide 3-kinase-δ y­lat­ing his­tone H2AX. ATM-depen­dent phos­phor­y­la­tion of other
Prakash Singh Shekhawat
How immu­no­de­fi­ciency can lead to malig­nancy  |  293
tar­gets medi­ates cell cycle arrest until DNA dam­age can be re­ Conflict-of-inter­est dis­clo­sure
solved. The broad func­ tion of ATM in DNA repair, within and Sung-Yun Pai has no con­flicts of inter­est to dis­close and is sup­
out­side of lym­pho­cytes, results in mul­ti­ple dif­fer­ent can­cers, ported by Center for Cancer Research, National Cancer Institute.
includ­ing acute leu­ke­mia (both lym­phoid and mye­loid), ­Hodgkin Sung-Yun Pai’s spouse is coin­ven­tor of a pat­ent on com­bi­na­tion
lym­phoma, NHL, and car­ci­no­mas (Figure 1).33 The degree of B-cell ther­ap
­ ies, includ­ing abiraterone to treat pros­tate can­cer.
lymphopenia and anti­body defi­ciency cor­re­lates with can­cer risk. Kathryn Lurain receives research funding from Bristol Myers
Squibb-Celgene Corporation, Merck, EMD Serono, Janssen Re­
GATA2 defi­ciency.  Germline het­ero­zy­gous muta­tions in the search, Lentigen Corp., CTI Biopharma and the National Cancer
GATA2 gene, a tran­scrip­tion fac­tor crit­ic ­ al for HSC devel­op­ment Institute.
and homeo­sta­sis, result in a wide vari­ety of clin­i­cal man­i­fes­ta­ Robert Yarchoan’s research is funded in part by Cooperative
tions, includ­ing atyp­i­cal myco­bac­te­rial infec­tion, DNA viral infec­ Research and Development Agreements (CRADAs) bet­ ween
tions (herpes simplex virus, HPV, EBV, cyto­meg­al­o­vi­rus), lymph­ Celgene Corporation (now Bristol Myers Squibb, Co.) and the
edema, bone mar­ row fail­ure, myelodysplastic syn­ drome, and National Cancer Institute. He has also used drugs for his clin­i­cal
acute mye­loid leu­ke­mia.34 GATA defi­ciency results in mul­ti­ple cel­ and/or lab­o­ra­tory research pro­vided to the NCI by Genentech

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/287/1852069/287pai.pdf by guest on 13 December 2021


lu­lar defects, includ­ing numer­ic ­ al defi­ciency of mono­cytes, den­ Corp., Merck and Co., EMD Serono, Janssen Research, Lentigen
dritic cells, NK cells, and B cells; pro­gres­sive stem cell loss; and Corp., and CTI Biopharma. Robert Yarchoan is a coin­ven­tor on
accu­mu­la­tion of addi­tional cyto­ge­netic abnor­mal­i­ties such as pat­ents on the treat­ment of KSHV-induced lym­phoma using
mono­somy 7 and tri­somy 8. Whether the loss of mature immune immu­no­mod­u­la­tory com­pounds and uses of bio­mark­ers, on
cells is a man­i­fes­ta­tion of HSC loss vs cell intrin­sic defects in sur­ pep­tide vac­cines for HIV and other viral dis­eases, and on inter­
vival or homeo­sta­sis is not clear. Here the immune defect per se, leu­kin (IL)–12 as a treat­ment for Kaposi sar­coma. His spouse,
par­tic­u­larly the lack of NK cells, appears crit­i­cal for the devel­op­ who is also a US gov­ern­ment employee, has pat­ents on KSHV
ment of HPV-driven can­cer.35 EBV+ lym­phoma is far less com­mon viral IL-6, ana­lyz­ing sin­gle-cell epigenomes, induc­ing inter­nal­i­
in GATA2 defi­ciency than EBV vire­mia and is related to immune za­tion of sur­face recep­tors, and vasostatin. It is his under­stand­
defects includ­ing excess pro­lif­er­a­tion of B cells, poor den­dritic ing that for­eign pat­ents have also been or will be filed for these
cell num­ber and func­tion, and thereby poor T-cell response to inven­tions. These inven­tions were all­made as full-time employ­
EBV.36 The pre­dis­po­si­tion to mye­loid leu­ke­mia in con­trast is likely ees of the US gov­ern­ment under 45 Code of Federal Regula­
due to the par­al­lel defect in hema­to­poi­e­sis (Figure 1). tions Part 7. All rights, title, and inter­est to these pat­ents have
been or should by law be assigned to the US Department of
Principles of treat­ment of can­cer in patients with IEIs.  In gen­ Health and Human Services. The gov­ern­ment con­veys a por­tion
eral, patients with IEIs and can­ cer should undergo stan­ dard of the roy­al­ties it receives to its employee-inven­tors under the
treat­ment, with the caveat that radi­a­tion-sen­si­tive patients (eg, Federal Technology Transfer Act of 1986 (P.L. 99-502).
ataxia-tel­an­gi­ec­ta­sia) require dose mod­i­fi­ca­tion or omis­sion alto­
gether of alkylating agents and radi­a­tion. Immunosuppressive Off-label drug use
agents such as cor­ti­co­ste­roids, mono­clo­nal antibodies that tar­ Sung-Yun Pai: Because most IEI present in childhood and very
get B cells, fludarabine, and targeted inhib­i­tors of JAK/STAT or few drugs are formally approved for use in children, off-label
BTK path­ways fur­ther pre­dis­pose to oppor­tu­nis­tic infec­tions. drug use is discussed.
Additional anti­bi­otic pro­phy­laxis and sched­uled replace­ment of Kathryn Lurain: Off-label use of EPOCH for HIV-associated lym­
immu­no­glob­u­lins to main­tain trough lev­els are crit­i­cal com­po­ phomas and primary effusion lymphoma was discussed.
nents of sup­port­ive care.23 Robert Yarchoan: We broadly discuss the use of checkpoint in­
Correction of the under­ly­ing predisposing con­di­tion with allo­ hibitors in PLWH but do not specify tumors for which they are
ge­neic HSC trans­plan­ta­tion con­sol­i­dates can­cer treat­ment and FDA approved.
should be con­sid­ered when fea­si­ble; it is the only ave­nue for long-
term pre­ven­tion of recur­rence and defin­i­tive cure. The rapid rise in
dis­cov­ery of the genes respon­si­ble for IEIs has been paralleled by Correspondence
the devel­op­ment of gene ther­apy approaches, using inte­grat­ing Sung-Yun Pai, National Institutes of Health, Building 10, Room
viral vec­tors to trans­duce autol­o­gous HSCs that are transplanted 1-5142, 10 Center Dr, Bethesda, MD 20892; e-mail: sung-yun​
after appro­pri­ate con­di­tion­ing.37 Gene ther­apy is an excit­ing alter­ ­.pai@nih​­.gov.
na­tive to allo­ge­neic HSC trans­plant that obvi­ates the need for a
matched donor and avoids immu­no­logic com­pli­ca­tions such as References
graft rejec­tion and graft-ver­sus-host dis­ease alto­gether. Newer 1. Yarchoan R, Uldrick TS. HIV-asso­ ci­
ated can­ cers and related dis­ eases.
tech­nol­o­gies based on gene editing also have prom­ise. N Engl J Med. 2018;378(22):2145.
2. Chang Y, Cesarman E, Pessin MS, et al. Identification of her­pes­vi­rus-like DNA
sequences in AIDS-asso­ci­ated Kaposi’s sar­coma. Science. 1994;266(5192):1865-
Acknowledgments  1869.
This work was supported by funding from the Intramural Re­ 3. Hernández-Ramírez RU, Shiels MS, Dubrow R, Engels EA. Cancer risk in
search Program, National Institutes of Health, National Cancer HIV-infected peo­ple in the USA from 1996 to 2012: a pop­u­la­tion-based,
­reg­is­try-link­age study. Lancet HIV. 2017;4(11):e495-e504.
Institute, Center for Cancer Research. The authors acknowl­edge
4. Silverberg MJ, Leyden W, Warton EM, Quesenberry CP Jr, Engels EA, Asgari
the con­tri­bu­tion of Dr Ramya Ramaswami, who cre­ated Figure 2, MM. HIV infec­tion sta­tus, immu­no­de­fic ­ iency, and the inci­dence of non-­
and Dr Luigi Notarangelo, who pro­vided case 2. mel­a­noma skin can­cer. J Natl Cancer Inst. 2013;105(5):350-360.

294  |  Hematology 2021  |  ASH Education Program


5. Coghill AE, Engels EA, Schymura MJ, Mahale P, Shiels MS. Risk of breast, 21. Tangye SG, Al-Herz W, Bousfiha A, et al. The ever-increas­ing array of novel
pros­tate, and colo­rec­tal can­cer diag­noses among HIV-infected indi­vid­u­als inborn errors of immu­nity: an interim update by the IUIS com­mit­tee. J Clin
in the United States. J Natl Cancer Inst. 2018;110(9):959-966. Immunol. 2021;41(3):666-679.
6. Brandão M, Bruzzone M, Franzoi M-A, et  al. Impact of HIV infec­tion on 22. Derpoorter C, Bordon V, Laureys G, Haerynck F, Lammens T. Genes at the
base­line char­ac­ter­is­tics and sur­vival of women with breast can­cer. AIDS. cross­ road of pri­ mary immunodeficiencies and can­ cer. Front Immunol.
2021;35(4):605-618. 2018;9:2544.
7. Biggar RJ, Jaffe ES, Goedert JJ, Chaturvedi A, Pfeiffer R, Engels EA. Hod­ 23. Riaz IB, Faridi W, Patnaik MM, Abra­ham RS. A sys­tem­atic review on pre­
gkin lym­phoma and immu­no­de­fi­ciency in per­sons with HIV/AIDS. Blood. dis­po­si­tion to lym­phoid (B and T cell) neo­pla­sias in patients with pri­mary
2006;108(12):3786-3791. immunodeficiencies and immune dysregulatory dis­or­ders (inborn errors of
8. Noy A. Optimizing treat­ment of HIV-asso­ci­ated lym­phoma. Blood. 2019; immu­nity). Front Immunol. 2019;10:777.
1­34(17):1385-1394. 24. Mastio J, Saeed MB, Wurzer H, Krecke M, Westerberg LS, Thomas C. Higher
9. Bohlius J, Schmidlin K, Costagliola D, et al; Collaboration of Observational inci­dence of B cell malig­nan­cies in pri­mary immunodeficiencies: a com­bi­
HIV Epidemiological Research Europe (COHERE) Study Group. Incidence na­tion of intrin­sic geno­mic insta­bil­ity and exo­cy­to­sis defects at the immu­
and risk fac­tors of HIV-related non-Hodgkin’s lym­phoma in the era of com­ no­log­i­cal syn­apse. Front Immunol. 2020;11:581119.
bi­na­tion antiretroviral ther­apy: a Euro­pean multicohort study. Antivir Ther. 25. Albert MH, Freeman AF. Wiskott-Aldrich Syndrome (WAS) and ded­i­ca­tor of
2009;14(8):1065-1074. cyto­ki­ne­sis 8- (DOCK8) defi­ciency. Front Pediatr. 2019;7:451.
10. Hernández-Ramírez RU, Qin L, Lin H, et al; North Amer­i­can AIDS Cohort 26. Biggs CM, Keles S, Chatila TA. DOCK8 defi­ciency: insights into path­o­phys­

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/287/1852069/287pai.pdf by guest on 13 December 2021


Collaboration on Research and Design of the International Epidemiologic i­­ol­ogy, clin­i­cal fea­tures and man­age­ment. Clin Immunol. 2017;181:75-82.
Databases to Evaluate AIDS. Association of immu­no­sup­pres­sion and HIV 27. Yakaboski E, Fuleihan RL, Sullivan KE, Cunningham-Rundles C, Feuille E.
viraemia with non-Hodgkin lym­phoma risk over­all and by sub­type in peo­ Lymphoproliferative dis­ease in CVID: a report of types and fre­quen­cies
ple liv­ing with HIV in Canada and the USA: a multicentre cohort study. from a US patient reg­is­try. J Clin Immunol. 2020;40(3):524-530.
Lancet HIV. 2019;6(4):e240-e249. 28. Wehr C, Houet L, Unger S, et al. Altered spec­trum of lym­phoid neo­plasms
11. Silverberg MJ, Neuhaus J, Bower M, et al. Risk of can­cers dur­ing interrupted in a sin­gle-cen­ter cohort of com­mon var­i­able immu­no­de­fi­ciency with
antiretroviral ther­apy in the SMART study. AIDS. 2007;21(14):1957-1963. immune dysregulation. J Clin Immunol. 2021;41(6):1250-1265.
12. Shindiapina P, Ahmed EH, Mozhenkova A, Abebe T, Baiocchi RA. Immunol­ 29. Pulvirenti F, Pecoraro A, Cinetto F, et al. Gastric can­cer is the lead­ing cause
ogy of EBV-related lymphoproliferative dis­ease in HIV-pos­i­tive indi­vid­u­als. of death in Ital­ian adult patients with com­mon var­i­able immu­no­de­fi­ciency.
Front Oncol. 2020;10:1723. Front Immunol. 2018;9:2546.
13. Wang Q , De Luca A, Smith C, et al; Hepatitis Coinfection and Non Hodgkin 30. Preite S, Gomez-Rodriguez J, Cannons JL, Schwartzberg PL. T and B-cell
Lymphoma pro­ject team for the Collaboration of Observational HIV Epide­ sig­nal­ing in acti­vated PI3K delta syn­drome: from immu­no­de­fi­ciency to
miological Research Europe (COHERE) in EuroCoord. Chronic hep­a­ti­tis B auto­im­mu­nity. Immunol Rev. 2019;291(1):154-173.
and C virus infec­tion and risk for non-Hodgkin lym­phoma in HIV-infected 31. Durandy A, Kracker S. Increased acti­va­tion of PI3 kinase-δ pre­dis­poses to
patients: a cohort study. Ann Intern Med. 2017;166(1):9-17. B-cell lym­phoma. Blood. 2020;135(9):638-643.
14. Montoto S, Shaw K, Okosun J, et al. HIV sta­tus does not influ­ence out­come 32. Zaki-Dizaji M, Akrami SM, Abolhassani H, Rezaei N, Aghamohammadi A.
in patients with clas­si­cal Hodgkin lym­phoma treated with che­mo­ther­apy Ataxia tel­an­gi­ec­ta­sia syn­drome: moon­light­ing ATM. Expert Rev Clin Immu-
using doxo­ru­bi­cin, bleomycin, vin­blas­tine, and dacarbazine in the highly nol. 2017;13(12):1155-1172.
active antiretroviral ther­apy era. J Clin Oncol. 2012;30(33):4111-4116. 33. Suarez F, Mahlaoui N, Canioni D, et  al. Incidence, pre­ sen­
ta­tion, and
15. Rengan R, Mitra N, Liao K, Armstrong K, Vachani A. Effect of HIV on sur­ prog­no­sis of malig­nan­cies in ataxia-tel­an­gi­ec­ta­sia: a report from the
vival in patients with non-small-cell lung can­cer in the era of highly active French national reg­ is­
try of pri­
mary immune deficiencies. J Clin Oncol.
antiretroviral ther­apy: a pop­u­la­tion-based study. Lancet Oncol. 2012;​ 2015;33(2):202-208.
13(12):​1203-1209. 34. Hsu AP, McReynolds LJ, Holland SM. GATA2 defi­ciency. Curr Opin Allergy
16. Uldrick TS, Gonçalves PH, Abdul-Hay M, et al; Cancer Immunotherapy Tri­ Clin Immunol. 2015;15(1):104-109.
als Network (CITN)-12 Study Team. Assessment of the safety of pembroli­ 35. Moon WY, Powis SJ. Does nat­u­ral killer cell defi­ciency (NKD) increase the
zumab in patients with HIV and advanced can­cer—a phase 1 study. JAMA risk of can­cer? NKD may increase the risk of some virus induced can­cer.
Oncol. 2019;5(9):1332-1339. Front Immunol. 2019;10:1703.
17. Food and Drug Administration (FDA). Cancer Clinical Trial Eligibility Cri- 36. Cohen JI. GATA2 Deficiency and Epstein-Barr virus dis­ease. Front Immunol.
teria: Patients With HIV, Hepatitis B Virus, or Hepatitis C Virus Infections: 2017;8:1869.
Food and Drug Administration Guidance for Industry. Washington, DC: 37. Kohn LA, Kohn DB. Gene ther­a­pies for pri­mary immune deficiencies. Front
FDA; 2020. Immunol. 2021;12:648951.
18. Hessol NA, Whittemore H, Vittinghoff E, et al. Incidence of first and sec­ond
pri­mary can­cers diag­nosed among peo­ple with HIV, 1985-2013: a pop­u­la­
tion-based, reg­is­try link­age study. Lancet HIV. 2018;5(11):e647-e655.
19. Picard C, Bobby Gaspar H, Al-Herz W, et al. International Union of Immu­
nological Societies: 2017 pri­mary immu­no­de­fi­ciency dis­eases com­mit­tee
report on inborn errors of immu­nity. J Clin Immunol. 2018;38(1):96-128.
20. Tangye SG, Al-Herz W, Bousfiha A, et al. Human inborn errors of immu­nity:
2019 update on the clas­si­fi­ca­tion from the International Union of Immuno­
logical Societies expert com­mit­tee. J Clin Immunol. 2020;40(1):24-64. DOI 10.1182/hema­tol­ogy.2021000261

Prakash Singh Shekhawat


How immu­no­de­fi­ciency can lead to malig­nancy  |  295
IMMUNOLOGY 101: WHAT THE PRACTICING HEMATOLOGIST NEEDS TO KNOW

Modified T cells as therapeutic agents


Nathan Singh

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/296/1851965/296singh.pdf by guest on 13 December 2021


Washington University School of Medicine, St Louis, MO

Immunotherapy is now a well-established modality in the treatment of cancer. Although several platforms to redirect the
immune response exist, the use of genetically modified T cells has garnered particular attention in recent years. This is
due, in large part, to their success in the treatment of B-cell malignancies. Adoptively transferred T cells have also dem-
onstrated efficacy in the treatment of systemic viral infections that occur following hematopoietic cell transplantation
prior to immune reconstitution. Here we discuss the techniques that enable redirection of T lymphocytes to treat cancer
or infection and the current indications for these therapies.

LEARNING OBJECTIVES
• Describe the methods that enable production of redirected T cells
• Review the current indications for approved T-cell products

CLINICAL CASE the multicomponent TCR genes are edited to produce


A 56-year-old man seeks treatment from his primary care phy- smaller genes. This rearrangement results in the production
sician for 2 months of progressive fatigue and night sweats. of two recombined genes from more than 1018 potential
Physical examination reveals axillary lymphadenopathy. Labo- rearrangements, endowing each individual T-cell clone
ratory studies reveal only an elevated lactate dehydrogenase with an essentially unique TCR. In addition to this antigen
but are otherwise unremarkable. Positron emission tomogra- specificity, T cells are potent killers of target cells, ensur-
phy (PET) reveals several sites of nodal fluorodeoxyglucose- ing efficient viral eradication. Upon antigen recognition,
uptake. Lymph node biopsy specimen ultimately reveals T cells undergo rapid proliferation to generate a large
CD19+ CD10+ BCL6+ mature B cells with histopathologic and cytotoxic immune response. Finally, antigen-specific
cytogenetic features consistent with germinal center-type T cells are able to differentiate into long-lived memory
large B-cell lymphoma. His Eastern Cooperative Oncology cells, capable of mounting memory responses decades
Group (ECOG) performance status is assessed as 1, and he after the initial infectious insult. Together, these features
initiates chemoimmunotherapy with combination rituximab, not only make T cells highly evolved and efficient con-
cyclophosphamide, doxorubicin, vincristine, and prednisone trollers of infection but also make them very attractive as
(R-CHOP). After 6 cycles, he has evidence of disease progres- anticancer agents.
sion and undergoes salvage chemotherapy with rituximab, Although T cells have great potential to serve as antileu-
ifosfamide, carboplatin, and etoposide (R-ICE). After 2 cycles, kemic agents by recognition of host leukemia as “non-self”
PET reveals persistent progressive disease, demonstrating (this is the primary therapeutic effect of allogeneic hema-
chemotherapy resistance. topoietic cell transplantation [HCT]), they also recognize
healthy host tissues as foreign, resulting in the syndrome of
graft-versus-host disease. Extrication of these 2 processes,
T cells as therapeutic agents graft-versus-leukemia and graft-versus-host, has given rise
Several cellular attributes make T cells extremely effective to the field of T-cell engineering for the purpose of redi-
at their native function of controlling infection. First, T cells recting this potent immune response toward cancer or
are endowed with the ability to differentiate “self” from uncontrolled viral infection.
“non-self” antigens, reflecting exquisite specificity. This
determination is made by the surface membrane antigen Redirecting the T-cell response
receptor called the T-cell receptor (TCR). In a manner sim- The processes of engineering T cells for use as anticancer
ilar to antibody production, the large genes encoding the or antiviral therapeutics are fundamentally distinct and will
TCR undergo somatic recombination, a process in which be addressed separately.

296 | Hematology 2021 | ASH Education Program


Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/296/1851965/296singh.pdf by guest on 13 December 2021
Figure 1. Process of manufacturing CAR T cells.

Cancer ing domain struc­tures are used in the sev­eral CAR T-cell prod­
Redirection of the T-cell response is depen­dent on the type of ucts approved for use by the US Food and Drug Administration
anti­gen they are intended to tar­get. Antigens fall into 2 broad (FDA). Both con­tain the CD3ζ sig­nal­ing domain, which reca­pit­u­
categories—intra­cel­lu­lar and cell sur­face. For a T cell to rec­og­nize lates the clas­si­cal “sig­nal 1” of T-cell stim­u­la­tion through the TCR.
pro­teins that are intra­cel­lu­lar, these pro­teins must be processed These 2 struc­tures dif­fer in the com­po­si­tion of their costimulato-
and presented by major his­to­com­pat­i­bil­ity com­plex (MHC) pro­ ry domains, one employing CD28 and the other 41BB. Correla-
teins for rec­og­ni­tion by TCRs. Introduction of a trans­genic TCR tive data from clin­i­cal tri­als dem­on­strate that CD28-based CARs
that has been designed, or more often iden­ti­fied among nat­ tend to undergo greater and more rapid expan­sion after deliv­ery
u­rally occur­ ring TCRs, to be spe­ cific for a can­cer-asso­ ci­
ated but do not per­sist for long peri­ods. Alternatively, 41BB-based
anti­gen allows for rec­og­ni­tion of intra­cel­lu­lar can­cer anti­gens. CARs do not expand as quickly or to the same degree but can
This pro­cess, which recon­structs all­com­po­nents of the native per­sist for years. Both CAR- and TCR-based approaches rely
TCR com­plex to acti­vate the T-cell response, has thus far been on genetic engi­neer­ing to deliver either of these trans­genic
met with lim­ited suc­cess. Identification of immu­no­genic anti­ recep­tors to T cells. Significant advances in gene ther­apy us-
gens and TCR cog­na­tes is dif­fi­cult. Furthermore, iden­ti­fi­ca­tion of ing viral vec­tors have enabled effi­cient deliv­ery of genes en-
can­cer-spe­cific anti­gens is unusual, and can­cer cells often down- coding these recep­tors to T cells.
regulate sur­face MHC. Although sev­eral stud­ies have eval­u­ated The pro­cess of manufactur­ing autol­o­gous T cells with syn­
the effi­cacy of these agents,1 dis­ease con­trol has been mod­est. thetic anti­ gen recep­ tors (Figure 1) begins with an ini­ tial leu-
For these rea­sons, an approach using syn­thetic anti­gen recep­ kapheresis to iso­ late periph­ eral blood mono­ nu­
clear cells
tors that are not depen­dent on MHC has been devel­oped. The (PBMCs). This PBMC prod­uct undergoes processing to purify
most well devel­oped of these are chi­me­ric anti­gen recep­tors T cells, which are then cul­tured with a stim­u­la­tory agent, often
(CARs). These hybrid recep­tors con­tain an anti­gen rec­og­ni­tion mag­netic beads containing stim­u­la­tory antibodies directed at
domain derived from a mono­clo­nal anti­body. This strat­egy al- CD3 and CD28. This stim­u­la­tion induces T-cell acti­va­tion, DNA
lows for MHC-inde­pen­dent anti­gen rec­og­ni­tion while pre­serv­ rep­li­ca­tion, and pro­lif­er­a­tion. Following stim­u­la­tion, gene ther­
ing much of the native TCR-driven acti­va­tion machin­ery. The apy vec­tors (most often len­ti­vi­rus or ret­ro­vi­rus) are added to the
down­side, of course, is loss of the abil­ity to tar­get intra­cel­lu­lar T-cell cul­tures, which medi­ate trans­fer of the tar­get transgene.
anti­gens. This extra­cel­lu­lar anti­gen-bind­ing domain is linked to These engineered cells are then grown in cul­ture for sev­eral days
intra­cel­lu­lar T-cell acti­vat­ing domains. Currently, only 2 sig­nal­ and finally are cryopreserved for even­tual infu­sion. All of these
Prakash Singh Shekhawat
Modified T cells as ther­a­peu­tic agents  |  297
Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/296/1851965/296singh.pdf by guest on 13 December 2021
Figure 2. Process of manufacturing virus-specific cells.

steps, whether at an aca­demic cen­ter for treat­ment on clin­i­cal Several strat­e­gies exist to iso­late and expand VSTs from a
tri­als or at a com­mer­cial pro­duc­tion facil­ity, are performed under bulk T-cell pop­u­la­tion ex vivo and dif­fer based on the type of
Good Manufacturing Practice guide­lines. At sev­eral time points anti­gen and method of anti­gen pre­sen­ta­tion (Figure 2). The sim­
dur­ing this manufactur­ing pro­cess, qual­ity con­trol mea­sures are plest of these is to deliver a whole viral lysate to anti­gen present-
performed to ensure Good Manufacturing Practice stan­dards. ing cells (APCs), such as den­dritic cells, and cocul­ture these with
Prior to deliv­ery of the T-cell prod­uct, patients receive lympho- T cells. This approach has lim­ited clin­i­cal util­ity, because there
depleting che­mo­ther­apy. Similar to the con­cept in allo­ge­neic is an under­ly­ing risk of trans­fer of a live infec­tious par­ti­cle. An
HCT, this lymphodepletion facil­i­tates suc­cess­ful engraft­ment of alter­na­tive is deliv­er­ing puri­fied or recom­bi­nant viral pro­teins or
the engineered prod­uct. This step can also fur­ther aid the ther­a­ plas­mid DNA encoding viral pro­teins to APCs. The most widely
peu­tic response by reduc­ing dis­ease bur­den, as CAR T cells are used approach employs the use of pools of pep­tide deliv­ered
still most often used for lym­phoid malig­nan­cies. to APCs.10 After selec­tion of the stim­ul­a­tion strat­egy, VSTs are
most often expanded in repeat stim­u­la­tion cul­tures. This pro­cess
Infection results in the pro­duc­tion of a large num­ber of poly­clonal T cells,
The pri­mary indi­ca­tion for adop­tive T-cell immu­no­ther­apy in the but manufactur­ing time is often sev­eral weeks to even months
treat­ment of infec­tious dis­eases is clin­i­cally sig­nif­i­cant viral infec­ long. An alter­na­tive approach is direct iso­la­tion of existing VSTs
tion fol­low­ing HCT. Over the past 2 decades, strat­eg ­ ies to treat from periph­eral blood. Antigen-spe­cific T cells will bind pep­tide-
viruses that are respon­si­ble for sig­nif­i­cant mor­bid­ity and mor­tal­ HLA multimers, and these multimers can be iso­lated along with
ity, such as cyto­meg­a­lo­vi­rus,2 Epstein-Barr virus (EBV),3 ade­no­vi­ bound T cells. This approach can fea­si­bly only be applied in set­
rus,4,5 human her­pes virus 6,6 BK virus,7 var­i­cella zoster virus,8 or tings of high VST fre­quency and as such has lim­ited util­ity.
broad-spec­trum anti­vi­ral treat­ments,9 have been devel­oped. Most
stud­ies have been performed using autol­o­gous cells, derived from Clinical prod­ucts for the treat­ment of can­cer
either the patient them­selves in the set­ting of autol­o­gous trans­ The major­ity of clin­i­cal expe­ri­ence using engi­neer­ing T cells is
plan­ta­tion or the donor in the set­ting of allo­ge­neic trans­plan­ta­ in the treat­ment of B-cell malig­nan­cies. Currently, 5 CAR T-cell
tion. The fun­da­men­tal prin­ci­ple under­ly­ing the gen­er­a­tion of virus- prod­ucts are approved for com­mer­cial use by the FDA; 4 of
spe­cific T cells (VSTs) in both set­tings is to expand a preexisting these tar­get the B-cell anti­gen CD19, and 1 tar­gets the plasma
pop­u­la­tion of anti­gen-spe­cific mem­ory T cells. This requires that cell anti­gen B-cell mat­u­ra­tion anti­gen (BCMA). Although these
the patient or donor has pre­vi­ously been exposed to that virus and prod­ucts dif­fer in sev­eral ways, the pri­mary var­i­ance is derived
an appro­pri­ate method to pres­ent stim­u­la­tory anti­gens to VSTs. from the struc­ture of the recep­tor’s costimulatory domains. In

298  |  Hematology 2021  |  ASH Education Program


Table 1. Pivotal trials of CAR T cells for B cell malignancies

Costimulatory
Characteristic Target domain Patient group No. (evaluable) CR rate (%) FDA approved? Reference
ALL
  Penn/CHOP phase CD19 41BB Children 60 93 Yes 12
1/2a
  ELIANA phase 2 CD19 41BB Children 75 81 Yes 11
(tisagenlecleucel)
  MSKCC phase 1 CD19 CD28 Children 25 75 No 35
  Seattle phase 1/2 CD19 41BB Children 43 93 No 36
  NCI phase 1 CD19 CD28 Children 51 61 No 37

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/296/1851965/296singh.pdf by guest on 13 December 2021


  MSKCC phase 1 CD19 CD28 Adults 53 83 No 38
  Penn phase 1 CD19 41BB Adults 20 90 No 39
  NCI phase 1 CD22 41BB Children 15 73 No 40
  Penn/CHOP phase 1 CD22 41BB Adults and 8 50 No 41
chil­dren
Non-Hodgkin lym­phoma
 Tisagenlecleucel CD19 41BB Adults 93 40 Yes 13
  Axicabtagene CD19 CD28 Adults 101 54 Yes 14, 15
ciloleucel
  Lisocabtagene CD19 41BB Adults 256 53 Yes 23
maraleucel
  Brexucabtagene CD19 CD28 Adults 60 62 Yes 17
autoleucel
Multiple mye­loma
  Idecabtagene BCMA 41BB Adults 128 33 Yes 18
vicleucel
Chronic lym­pho­cytic leu­ke­mia
  Penn phase 2 CD19 41BB Adults 24 25 No 42
Trials iden­ti­fied by loca­tion and/or even­tual com­mer­cial prod­uct name.
CHOP, Children’s Hospital of Philadelphia; MSKCC, Memorial Sloan Kettering Cancer Center; NCI, National Cancer Institute; Penn, University of
Pennsylvania; Seattle, Seattle Children’s Hospital.

addi­tion to these FDA-approved prod­ucts, sev­eral oth­ers have Lisocabtagene maraleucel also con­tains the 41BB costimulatory
been eval­u­ated in clin­i­cal tri­als. Key find­ings from these stud­ies domain but is deliv­ered as 2 indi­vid­ual prod­ucts: one com­posed
are sum­ma­rized in Table 1. of CD4+ and the other of CD8+ T cells.16 It is indi­cated for all­of
the lym­phoma sub­types men­tioned above, as well as grade 3B
Acute lym­pho­blas­tic leu­ke­mia fol­lic­u­lar lym­phoma and DLBCL aris­ing from nonfollicular indo­lent
Currently, the only approved prod­ uct for the man­ age­
ment his­tol­o­gies. Finally, brexucabtagene autoleucel is struc­tur­ally
of acute lym­ pho­blas­
tic leu­
ke­
mia (ALL) is tisagenlecleucel, a iden­ti­cal to axicabtagene ciloleucel but undergoes an addi­tional
CD19-targeted CAR containing the 41BB costimulatory do- B-cell deple­tion step dur­ing prod­uct manufactur­ing and is ap-
main.11,12 It is approved for patients up to 25 years of age who are proved for relapsed or refrac­tory man­tle cell lym­phoma.17
refrac­tory or in sec­ond or later relapse.
Multiple mye­loma
Non-Hodgkin lym­phoma Idecabtagene vicleucel was recently approved for adult patients
Four prod­ucts are approved for the treat­ment of non-Hodgkin with relapsed or refrac­tory mul­ti­ple mye­loma after 4 or more
lym­phoma in adults. Tisagenlecleucel is indi­cated for patients lines of ther­apy, includ­ing an immu­no­mod­u­la­tory agent, a prote-
with dif­fuse large B-cell lym­phoma (DLBCL), high-grade B-cell asome inhib­i­tor, and an anti-CD38 mono­clo­nal anti­body.18 Unlike
lym­phoma, or DLBCL aris­ing from fol­lic­u­lar lym­phoma that is the other approved ther­a­pies, it tar­gets the BCMA on plasma
relapsed or refrac­tory after 2 or more lines of ther­apy.13 Axicabta- cells. Like tisagenlecleucel and listocabtagene maraleucel, it
gene ciloleucel is also directed at CD19 but con­tains a costimula- con­tains the 41BB costimulatory domain.
tory domain derived from CD28.14,15 It is approved for adults with
DLBCL, pri­mary medi­as­ti­nal large B-cell lym­phoma, high-grade Patient selec­tion
B-cell lym­phoma, and DLBCL aris­ing from fol­lic­u­lar lym­phoma Given the avail­abil­ity of CAR T-cell prod­ucts, par­tic­u­larly in the
that is relapsed or refrac­tory after 2 or more lines of ther­apy. man­age­ment of relapsed or refrac­tory non-Hodgkin lym­phoma,
Prakash Singh Shekhawat
Modified T cells as ther­a­peu­tic agents  |  299
Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/296/1851965/296singh.pdf by guest on 13 December 2021
Figure 3. Currently enrolling CAR T cell trials across the globe.

a key clin­i­cal con­sid­er­ation is when to use this ther­apy and which Finally, given the autol­o­gous nature of this ther­apy, prod­
ther­apy to use. At many aca­demic cen­ters, CAR T-cell ther­apy is uct manufactur­ ing is also worth con­ sid­er­
ing. Several stud­ ies
com­monly con­sid­ered for patients with pro­gres­sive lym­phoma have dem­on­strated that both pre­vi­ous expo­sure to che­mo­ther­
after sec­ond-line che­mo­ther­apy or in the set­ting of relapse after apy and the immu­no­sup­pres­sive envi­ron­ment encum­bered by
autol­o­gous trans­plan­ta­tion. Decision mak­ing about using CAR T active malig­nancy can sig­nif­i­cantly impair the effec­tive expan­
cells vs autol­o­gous trans­plan­ta­tion, spe­cif­i­cally in the set­ting of sion of col­lected T cells.24,25 Several cen­ters have explored “early
failed first-line ther­apy, is an active topic of dis­cus­sion and will be bank­ing” pro­ce­dures, in which high-risk patients undergo apher­
assisted by the results of cur­rent ongo­ing ran­dom­ized clin­ic ­ al tri­ e­sis and stor­age of T cells prior to ini­ti­at­ing first-line ther­apy to
als (NCT03391466, NCT03570892, NCT03575351). ensure avail­abil­ity of “fit” T cells should they be needed. This
Incidence and man­age­ment of toxicities is also a rel­e­vant approach remains inves­ti­ga­tional and should only be con­sid­ered
con­sid­er­ation. The most com­mon tox­ic­ity occur­ring fol­low­ in the con­text of a clin­i­cal trial.
ing CAR T-cell ther­apy is cyto­kine release syn­drome (CRS), a The avail­abil­ity of CAR T-cell prod­ucts has increased sig­nif­i­
sys­temic hyperinflammatory syn­drome driven by supraphysi- cantly in the past 5 years. For chil­dren with ALL, ther­apy is almost
ologic pro­duc­tion of inter­leu­kin 6 (IL-6) bystander mye­loid-lin­ always deliv­ered in an aca­demic set­ting, which also facil­i­tates
e­age cells in response to robust T-cell acti­va­tion.19,20 The use avail­abil­ity of tisagenlecleucel. At the time of writ­ing, there were
of IL-6 and IL-6 recep­tor inhib­i­tors, such as tocilizumab, has 324 actively recruiting clin­i­cal tri­als of CAR T-cell prod­ucts. Most
sig­nif­i­cantly improved out­comes for patients. Another highly of these are in the United States (148) and China (150), reflect-
mor­bid tox­ic­ity is immune effec­tor cell-asso­ci­ated neu­ro­tox­ic­ ing the dis­par­ity in access to this plat­form that exists across the
ity syn­drome (ICANS),21 a poorly under­stood neu­ro­psy­chi­at­ric globe (Figure 3).
syn­drome that is com­monly man­aged with cor­ti­co­ste­roids and
anti­ep­i­lep­tics.22 In rare cases, ICANS can prog­ress and lead to
cere­bral edema, a much more seri­ous con­di­tion that is pri­mar­ily Clinical prod­ucts for the treat­ment of infec­tion
man­aged with high-dose ste­roids. Intriguingly, both toxicities Currently, there are no FDA-approved prod­ucts avail­­able for the
appear to be more severe with CD28-based CAR T cells, and for treat­ment of viral infec­tion using T cells. Three phase 3 stud­ies are
this rea­son, many cli­ni­cians will elect to use 41BB-based ther­ cur­rently ongo­ing (NCT04832607, NCT04390113, NCT03394365)
apy, if avail­­able, for patients who are more frail. Clinical tri­als of eval­u­at­ing the effi­cacy of VSTs. One (NCT03394365) is focused on
CAR T prod­ucts only enrolled patients with high-per­for­mance the effi­cacy of a VST prod­uct targeting EBV anti­gens in the man­
sta­tuses (ECOG 0-1). However, real-world data sug­gest that age­ ment of EBV-asso­ ci­
ated posttransplant lymphoproliferative
patients who are not as fit can be suc­cess­fully treated with CAR dis­or­der,26 as well chronic active EBV and EBV-driven hemophago-
ther­apy.23 Indeed, given the man­age­ment options now avail­­ cytic lymphohistiocytosis (HLH). Greater than 100 clin­i­cal tri­als
able for CRS and ICANS, most patients are con­sid­ered eli­gi­ble at all­phases are cur­rently enroll­ing across the globe. An excit­ing
for this ther­apy. Other toxicities include prolonged pan­cy­to­pe­ pros­pect is the devel­op­ment of allo­ge­neic VSTs that can serve as
nia and a dura­ble loss of B cells with asso­ci­ated hypogamma- an “off-the-shelf” treat­ment, elim­i­nat­ing the time delay between
globulinemia. dis­ease onset and treat­ment.27

300  |  Hematology 2021  |  ASH Education Program


3. Heslop HE, Slobod KS, Pule MA, et  al. Long-term out­ come of EBV-
CLINICAL CASE (Con­t in­u ed) spe­cific T-cell infu­sions to pre­vent or treat EBV-related lymphoproliferative
dis­ease in trans­plant recip­i­ents. Blood. 2010;115(5):925-935.
After discussing the dis­ tinc­
tions between autol­ o­
gous trans­ 4. Leen AM, Christin A, Myers GD, et al. Cytotoxic T lym­pho­cyte ther­apy with
plan­ta­tion and CAR T cells, the patient elects to pro­ceed with donor T cells pre­vents and treats ade­no­vi­rus and Epstein-Barr virus infec­
CAR T cells. He undergoes apher­e­sis for planned ther­apy with tions after haploidentical and matched unre­lated stem cell trans­plan­ta­tion.
tisagenlecleucel, with suc­cess­ful prod­uct manufactur­ing. He Blood. 2009;114(19):4283-4292.
5. Zandvliet ML, Falkenburg JH, van Liempt E, et  al. Combined CD8+ and
receives con­di­tion­ing che­mo­ther­apy with fludarabine and
CD4+ ade­no­vi­rus hexon-spe­cific T cells asso­ci­ated with viral clear­ance
cyclo­phos­pha­mide, followed by CAR T-cell infu­sion. He expe­ after stem cell trans­plan­ta­tion as treat­ment for ade­no­vi­rus infec­tion. Hae-
ri­ences chills and rig­ors on day +3, clas­si­fied as grade 1 CRS. matologica. 2010;95(11):1943-1951.
These resolve spon­ta­ne­ously on day +5. After count recov­ery 6. Gerdemann U, Keukens L, Keirnan JM, et al. Immunotherapeutic strat­e­gies
on day +11, he is discharged from the hos­pi­tal with­out incur­ring to pre­vent and treat human her­pes­vi­rus 6 reactivation after allo­ge­neic stem
cell trans­plan­ta­tion. Blood. 2013;121(1):207-218.
any fur­ther tox­ic­ity. Thirty days after treat­ment, PET/com­puted 7. Blyth E, Clancy L, Simms R, et al. BK virus-spe­cific T cells for use in cel­lu­lar
tomog­ra­phy reveals a par­tial response. PET/com­puted tomog­ ther­apy show spec­i­fic­ity to mul­ti­ple anti­gens and polyfunctional cyto­kine
ra­phy at 3 months reveals com­plete remis­sion.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/296/1851965/296singh.pdf by guest on 13 December 2021


responses. Transplantation. 2011;92(10):1077-1084.
8. Blyth E, Gaundar SS, Clancy L, et al. Clinical-grade var­i­cella zoster virus-spe­
cific T cells pro­duced for adop­tive immu­no­ther­apy in hemo­poi­etic stem cell
trans­plant recip­i­ents. Cytotherapy. 2012;14(6):724-732.
Challenges and next-gen­er­a­tion T cells for can­cer 9. Papadopoulou A, Gerdemann U, Katari UL, et  al. Activity of broad-spec­
Although great prog­ ress has been made in the use of engi- trum T cells as treat­ment for AdV, EBV, CMV, BKV, and HHV6 infec­tions after
HSCT. Sci Transl Med. 2014;6(242):242ra83.
neered T cells, par­tic­u­larly with regard to the treat­ment of hema­ 10. Koehne G, Hasan A, Doubrovina E, et al. Immunotherapy with donor T cells
to­logic can­cers, the real­ity is that most patients treated with sen­si­tized with overlapping pentadecapeptides for treat­ment of per­sis­
CD19-directed CAR T cells will not achieve dura­ble remis­sions. tent cyto­meg­a­lo­vi­rus infec­tion or vire­mia. Biol Blood Marrow Transplant.
The 2 fea­tures that have been iden­ti­fied as crit­i­cal bar­ri­ers to the 2015;21(9):1663-1678.
11. Maude SL, Laetsch TW, Buechner J, et al. Tisagenlecleucel in chil­dren and young
broader suc­cess of this ther­apy are (1) ini­tial fail­ure that leads to
adults with B-cell lym­pho­blas­tic leu­ke­mia. N Engl J Med. 2018;378­(5):439-448­.
non­re­sponse and (2) acquired fail­ure that leads to dis­ease re- 12. Maude SL, Frey N, Shaw PA, et  al. Chimeric anti­gen recep­tor T cells for
lapse. The for­mer is more com­mon in lym­phoma, whereas the sustained remis­sions in leu­ke­mia. N Engl J Med. 2014;371(16):1507-1517.
lat­ter is more com­mon in ALL. Several stud­ies have iden­ti­fied 13. Schuster SJ, Bishop MR, Tam CS, et al; JULIET Investigators. Tisagenlecleu-
T cell–intrin­sic fea­tures that are asso­ci­ated with ini­tial fail­ure, cel in adult relapsed or refrac­tory dif­fuse large B-cell lym­phoma. N Engl J
Med. 2019;380(1):45-56.
such as expres­sion of dys­func­tion-asso­ci­ated genes, ter­mi­nal 14. Locke FL, Ghobadi A, Jacobson CA, et al. Long-term safety and activ­ity of
dif­fer­en­ti­a­tion, and impaired T-cell per­sis­tence.25,28-30 New strat­ axicabtagene ciloleucel in refrac­tory large B-cell lym­phoma (ZUMA-1): a
e­gies to enhance T-cell fit­ness using inno­va­tive manufactur­ing sin­gle-arm, multicentre, phase 1-2 trial. Lancet Oncol. 2019;20(1):31-42.
pro­cesses have gen­er­ated great excite­ment and open ave­nues 15. Neelapu SS, Locke FL, Bartlett NL, et  al. Axicabtagene ciloleucel CAR
T-cell ther­ apy in refrac­ tory large B-cell lym­ phoma. N Engl J Med.
to the manufactur­ing of T cells with enhanced func­tion.31,32
2017;377(26):2531-2544.
Several mech­a­nisms have been described that lead to dis­ease 16. Abramson JS, Palomba ML, Gordon LI, et  al. Lisocabtagene maraleu-
relapse, most thor­oughly the phe­nom­e­non of anti­gen escape. In cel for patients with relapsed or refrac­ tory large B-cell lym­ pho­ mas
approx­i­ma­tely 40% of patients with ALL who relapse, the recur­ (TRANSCEND NHL 001): a multicentre seam­ less design study. Lancet.
rent leu­ke­mia lacks detect­able sur­face CD19, enabling dis­ease 2020;396(10254):839-852.
17. Wang M, Munoz J, Goy A, et al. KTE-X19 CAR T-cell ther­apy in relapsed or
out­growth.33,34 To over­ come anti­ gen escape, sev­ eral groups
refrac­tory man­tle-cell lym­phoma. N Engl J Med. 2020;382(14):1331-1342.
have employed dual-anti­ gen targeting to more thor­ oughly 18. Munshi NC, Anderson LD Jr, Shah N, et  al. Idecabtagene vicleucel in
cap­ture malig­nant clones and reduce the like­li­hood of anti­gen relapsed and refrac­tory mul­ti­ple mye­loma. N Engl J Med. 2021;384(8):
loss as a mech­a­nism of relapse (NCT03241940, NCT03330691, 705-716.
19. Singh N, Hofmann TJ, Gershenson Z, et al. Monocyte lin­e­age-derived IL-6
NCT03448393).
does not affect chi­me­ric anti­gen recep­tor T-cell func­tion. Cytotherapy.
2017;19(7):867-880.
Conflict-of-inter­est dis­clo­sure 20. Giavridis T, van der Stegen SJC, Eyquem J, Hamieh M, Piersigilli A, Sadelain M.
CAR T cell-induced cyto­kine release syn­drome is medi­ated by mac­ro­phages
Nathan Singh holds sev­eral pat­ents related to CAR and other
and abated by IL-1 block­ade. Nat Med. 2018;24(6):731-738.
engineered cell ther­a­pies. 21. Gust J, Ponce R, Liles WC, Garden GA, Turtle CJ. Cytokines in CAR T cell-
asso­ci­ated neu­ro­tox­ic­ity. Front Immunol. 2020;11:577027.
22. Maus MV, Alexander S, Bishop MR, et  al. Society for immu­no­ther­apy of
Off-label drug use
can­cer (SITC) clin­i­cal prac­tice guide­line on immune effec­tor cell-related
Nathan Singh: nothing to disclose. adverse events. J Immunother Cancer. 2020;8(2):e001511.
23. Nastoupil LJ, Jain MD, Feng L, et al. Standard-of-care axicabtagene ciloleu-
cel for relapsed or refrac­tory large B-cell lym­phoma: results from the US
Correspondence lym­phoma CAR T con­sor­tium. J Clin Oncol. 2020;38(27):3119-3128.
Nathan Singh, Washington University School of Medicine, 660 S. 24. Das RK, Vernau L, Grupp SA, Barrett DM. Naïve T-cell def­i­cits at diag­no­sis
Euclid Avenue, St Louis, Missouri 63110; e-mail: singh​­.nathan@gmail​ and after che­mo­ther­apy impair cell ther­apy poten­tial in pedi­at­ric can­cers.
­.com. Cancer Discov. 2019;9(4):492-499.
25. Singh N, Perazzelli J, Grupp SA, Barrett DM. Early mem­ory phe­no­types
drive T cell pro­lif­er­a­tion in patients with pedi­at­ric malig­nan­cies. Sci Transl
References Med. 2016;8(320):320ra3.
1. Stadtmauer EA, Fraietta JA, Davis MM, et al. CRISPR-engineered T cells in 26. Prockop S, Doubrovina E, Suser S, et  al. Off-the-shelf EBV-spe­cific T cell
patients with refrac­tory can­cer. Science. 2020;367(6481):eaba7365. immu­no­ther­apy for rituximab-refrac­tory EBV-asso­ci­ated lym­phoma fol­low­
2. Berger C, Turtle CJ, Jensen MC, Riddell SR. Adoptive trans­fer of virus-spe­ ing trans­plan­ta­tion. J Clin Invest. 2020;130(2):733-747.
cific and tumor-spe­cific T cell immu­nity. Curr Opin Immunol. 2009;21(2): 27. Tzannou I, Papadopoulou A, Naik S, et al. Off-the-shelf virus-spe­cific T cells to
224-232. Prakash Singh Shekhawat treat BK virus, human her­pes­vi­rus 6, cyto­meg­a­lo­vi­rus, Epstein-Barr virus, and

Modified T cells as ther­a­peu­tic agents  |  301


ade­no­vi­rus infec­tions after allo­ge­neic hema­to­poi­etic stem-cell trans­plan­ta­ 36. Gardner RA, et al. Intent-to-treat leukemia remission by CD19 CAR T cells
tion. J Clin Oncol. 2017;35(31):3547-3557. of defined formulation and dose in children and young adults. Blood.
28. Deng Q , Han G, Puebla-Osorio N, et al. Characteristics of anti-CD19 CAR T 2017;129:3322-3331.
cell infu­sion prod­ucts asso­ci­ated with effi­cacy and tox­ic­ity in patients with 37. Lee DW, et al. T cells expressing CD19 chimeric antigen receptors for acute lym-
large B cell lym­pho­mas. Nat Med. 2020;26(12):1878-1887. phoblastic leukaemia in children and young adults: a phase 1 dose-escalation
29. Fraietta JA, Lacey SF, Orlando EJ, et al. Determinants of response and resis­ trial. Lancet. 2015;385:517-528.
tance to CD19 chi­me­ric anti­gen recep­tor (CAR) T cell ther­apy of chronic 38. Park JH, et al. Long-term follow-up of CD19 CAR therapy in acute lympho-
lym­pho­cytic leu­ke­mia. Nat Med. 2018;24(5):563-571. blastic leukemia. N Engl J Med. 2018;378:449-459.
30. Singh N, Lee YG, Shestova O, et al. Impaired death recep­tor sig­nal­ing in 39. Frey NV, et al. Optimizing chimeric antigen receptor T-cell therapy for
leu­ke­mia causes anti­gen-inde­pen­dent resis­tance by induc­ing CAR T-cell adults with acute lymphoblastic leukemia. J Clin Oncol. 2020;38:415-422.
dys­func­tion. Cancer Discov. 2020;10(4):552-567. 40. Fry TJ, et al. CD22-targeted CAR T cells induce remission in B-ALL that
31. Lynn RC, Weber EW, Sotillo E, et  al. c-Jun overexpression in CAR T cells is naive or resistant to CD19-targeted CAR immunotherapy. Nat Med.
induces exhaus­tion resis­tance. Nature. 2019;576(7786):293-300. 2018;24:20-28.
32. Weber EW, Parker KR, Sotillo E, et  al. Transient rest restores func­tion­al­ 41. Singh N, et al. Antigen-independent activation enhances the efficacy of
ity in exhausted CAR-T cells through epi­ ge­ netic remodeling. Science. 4-1BB-costimulated CD22 CAR T cells. Nat Med. 2021.
2021;372(6537):eaba1786. 42. Frey NV, et al. Long-term outcomes from a randomized dose optimization

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/296/1851965/296singh.pdf by guest on 13 December 2021


33. Orlando EJ, Han X, Tribouley C, et al. Genetic mech­a­nisms of tar­get anti­ study of chimeric antigen receptor modified T cells in relapsed chronic
gen loss in CAR19 ther­apy of acute lym­pho­blas­tic leu­ke­mia. Nat Med. lymphocytic leukemia. J Clin Oncol. 2020;38:2862-2871.
2018;24(10):1504-1506.
34. Sotillo E, Barrett DM, Black KL, et al. Convergence of acquired muta­tions
and alter­na­tive splic­ing of CD19 enables resis­tance to CART-19 immu­no­
ther­apy. Cancer Discov. 2015;5(12):1282-1295.
35. Curran KJ, et al. Toxicity and response after CD19-specific CAR T-cell therapy
in pediatric/young adult relapsed/refractory B-ALL. Blood. 2019;134:2361– © 2021 by The Amer­i­can Society of Hematology
2368. DOI 10.1182/hema­tol­ogy.2021000262

302  |  Hematology 2021  |  ASH Education Program


INDOLENT LYMPHOMAS

What factors guide treatment selection in


mycosis fungoides and Sezary syndrome?

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/303/1852064/303kim.pdf by guest on 13 December 2021


Youn H. Kim
Department of Dermatology, Stanford University School of Medicine, Palo Alto, CA

Cutaneous T-cell lymphoma (CTCL) comprises a spectrum of T-cell lymphomas with primary skin involvement. Mycosis
fungoides (MF) and Sezary syndrome (SS) are the common subtypes of CTCL in which patients present with widely
diverse profiles of skin involvement and varying extents of extracutaneous disease. Patients with early-stage disease have
an excellent prognosis and are managed primarily with skin-directed therapies; however, those with advanced-stage MF
or SS often require multiple lines and recurrent courses of systemic therapies. Many options are available when consid-
ering systemic agents, and it is often challenging to know how to prioritize therapies to address a patient’s objective
disease and quality of life issues. Appreciating the disease heterogeneity and understanding the patient’s overall disease
profile (eg, skin, lymph nodes, blood, large cell transformation) serve as a useful framework in aligning therapies that
can optimally treat active sites of disease. Tissue or blood biomarkers can be integrated into our process of prioritizing
therapies and personalizing management in MF or SS. Multidisciplinary management and optimizing supportive care are
additional key elements for a favorable outcome. Appropriate patients with high-risk disease should be considered for
allogeneic hematopoietic stem cell transplant.

LEARNING OBJECTIVES
• Understand the clinical heterogeneity of patients with MF and SS
• Appreciate the unique quality of life issues associated with cutaneous disease
• Identify the clinical, pathological, and other biomarkers that help prioritize therapies and personalize clinical man­
agement in MF and SS

Introduction skin electron beam therapy [TSEBT], extracorporeal pho­


Mycosis fungoides (MF) and Sezary syndrome (SS) are the topheresis, bexarotene, vorinostat, romidepsin) and those
most common subtypes of cutaneous T­cell lymphoma originally developed to treat other lymphomas but co­
(CTCL), and although the skin is the primary site of involve­ opted for CTCL (eg, brentuximab vedotin [BV], pralatrex­
ment, the malignant T cells may expand in the lymph node ate). More recently, new systemic therapies have emerged,
(LN), visceral, and blood compartments.1 In the skin lesions built upon rationally selected targets in MF/SS (eg, mog­
of early­stage MF, significant inflammatory infiltrate of amulizumab [MOGA], anti­KIR3DL2 antibody; Table 1).
immune stimulated profile is observed. However, as the Although these newer agents represent a major advance­
disease progresses the malignant T cells acquire a Th2 ment, they have not altered our fundamental treatment
phenotype accompanied by a microenvironment skewed strategy. Only a slim minority of patients will ever be cured of
toward a more immune­suppressive Th2 cytokine profile.2,3 their disease, and most patients will receive numerous lines
A subset of patients with advanced MF or SS undergo a of therapy during their lives. Each failed treatment takes a
transformation into large cell disease (LCT) that is often physical and psychological toll on patients. We have long
associated with more aggressive biological behavior. known that a one­size­fits­all approach cannot work for this
Patients with MF and SS subgroups are often distinct clini­ heterogeneous disease. As our therapeutic armamentarium
cally and biologically but also share similarities, and clinical continues to expand, we are now challenged with how to
features can shift from MF to SS and vice versa in the course best match patients to the treatments most likely to ben­
of a patient’s disease. efit them. Furthermore, it is unclear how best to sequence
The clinical management of MF/SS involves therapies or combine therapies, and given the lack of curative thera­
developed specifically for patients with CTCL (eg, total pies, the ones with fewer cumulative toxicities and durable
Prakash Singh Shekhawat
Personalizing treatment selection in MF/SS | 303
Table 1. Clinical activ­ity of sys­temic ther­a­pies in CTCL* by dis­ease sub­type (clin­i­cal stage, MF vs SS), com­part­ments (skin, LN, blood), ±LCT; tol­er­a­bil­ity

Global/com­pos­ite clin­i­cal activ­ity of all­


Tolerability
com­part­ments*
fac­tors in chronic
Treatment N; sub­type ORR/CR % (n/N) DOR, median PFS, median Skin activ­ity LN activ­ity Blood activ­ity LCT included ther­apy
Standard ther­a­pies
 Bexarotene†,22 56; MF/SS 45% (25/56)/1 CR 42.7 weeks 13.9 weeks Skin RR = pri­mary Limited LN data: Of 17 SS, 8 had N/a Hyperlipidemia
(300 mg/m2 = opti­ IIB-IV; end point: Of 25 skin OR, par­tial Sezary
mal dose) SS (17/56) IIB, 57% (13/23); 7 w/clin­i­cal data:
III, 32% (6/19); LAD - 4/8 PR
IVA, 44% (4/9); 3/7 PR or CR 3/8 SD
IVB, 40% (2/5); 4/7 PD 1/8 PD
SS, 24% (4/17, all­
doses)
 Extracorporeal 51; MF/SS 63% (32/51)/8 CR 22.4 months N/a Compartment-spe­cific data not reported but 16% CR N/a None
photopheresis‡,24 III-IV; SS (39/51) w/clear­ing of blood Sezary dis­ease
 Vorinostat63 74; MF/SS IB-IV; 30% (22/74)/1 CR >185 days TTP median Skin RR = pri­mary LN RR 42% 14/27 SS w/SC Yes Fatigue,
SS (30/74) after 281 days 148 days end point: (10/24) reduc­tion dysgeusia,
IB/IIA, 31% (4/13); >25% ↓plate­lets

304  |  Hematology 2021  |  ASH Education Program


IIB-IV, 30% (18/61);
skin tumors, 23%
(5/22);
SS, 33% (10/30)
 Romidepsin14 96; MF/SS 34% (33/96)/6 CR; 15 months TTP median 8 Skin RR 40% (38/96) LN response Blood RR 38% Yes Fatigue,
IB-IV; SS SS (B1-2), 32% months 35% (13/37) (14/37, 2 dysgeusia,
(37/96) (12/37) by RECIST CR); B2, 46% ↓plate­lets
(6/13, 2 CR)
 Pralatrexate§,37 29; CTCL 45% (13/29)/1 CR Not reached; Not reached; Compartment-spe­cific data not reported, response by Yes Mucositis
(opti­mal CTCL IB-IV; SS (8/29) 73% cont OR 388 days stage:
dose 15 mg/m2 at 6 months >15 mg/m2 MF by stage
weekly 3/4) IB 60% (3/5);
IIB 67% (4/6);
IVA 60% (3/5);
SS 25% (2/8)
 Brentuximab11 48; MF IA-IV; 65% (31/48)/ 15.1 months; 15.9 months; Skin RR 77% (37/48); 2 w/LN+, stage Excluded B2/SS Yes (17/48) PN
CD30 > 10% 5 CR; ORR4 LCT MF/ALCL LCT 15.5 median DOR 20.6 IVA: ORR
(no SS) 65% (11/17) months months, MF/ALCL 100% (2/2,
1 CR)
 Mogamulizumab¶,13 186; MF/SS 28% (52/186)/6 14.1 months; MF 7.7 months Skin RR 42% LN RR 17% Blood RR 68% No (LCT Rash
IB-IV; SS CR; MF 21% 13.1 months; (78/186); median (21/124); (83/122, excluded)
(81/186) (22/105); SS SS 17.3 TTR 3.0 months; median TTR 54 CR);
37% (30/81) months median DOR 20.6 3.3 months; median TTR
months median DOR 1.1 months;
15.5 months median DOR
25.5 months
 Pembrolizumab41 24; MF/SS IIB- 38% (9/24)/2 CR; Not reached; Not reached; Skin RR 38% (9/24); N/a Baseline w/B2, Yes (4/24) irAE (coli­tis, pneu­
IV (23/24); SS 27% (4/15); median PFS at 1 year, 6/24, >90% skin n = 6, 17% (1/6) mo­ni­tis)
SS (15/24) LCT 25% (1/4) fol­low-up 65% clear­ing
58 weeks

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/303/1852064/303kim.pdf by guest on 13 December 2021


Table 1. (continued)

Global/com­pos­ite clin­i­cal activ­ity of all­


Tolerability
com­part­ments*
fac­tors in chronic
Treatment N; sub­type ORR/CR % (n/N) DOR, median PFS, median Skin activ­ity LN activ­ity Blood activ­ity LCT included ther­apy
  Liposomal doxo­ru­bi­ 49; 41% (20/49)/3 CR 6 months 6.2 months; Skin RR 53% (26/49); LN ± vis­ceral Excluded SS Yes
cin§,39 (20 mg/m2, MF IIB-IV (no TTP median best PR/CR dis­ease had
day 1, day 15, SS) 7.4 months lower RR than
28-day cycle) skin-only (22%
vs 52%)
 Gemcitabine§,40 31; MF/SS; SS 65% (20/31)/3 CR; 4.1 months N/a Compartment-spe­cific data not reported but case exam­ Yes (13/31)
(1000 mg/m2, day (11/33) LCT 54% (7/13); ples with activ­ity in LN/blood dis­ease
1, 8, 15) SS 73% (8/11)
  Investigative
ther­a­pies
(w/peer-reviewed
papers)
 Anti-KIR3DL2 anti­ 44; SS (35/44); SS, 43% (15/35); 13.8 months; 8.2 months; SS Of SS: skin RR 51% Of SS: LN RR 11% Of SS: blood RR Yes (6/35 SS, Well tol­er­ated
body (lacutamab), MF IB-IV MF, 13% (1/8); SS 13.8 11.7 months, (18/35, 3 CR) (2/18, 1 CR) 56% (19/34, 5/8 MF)
phase 1/2 study (8/44) LCT no OR months; MF MF 3.9 9 CR)
data (ongo­ing piv­ 6.9 months months
otal trial in SS)25
 Duvelisib64 19; MF/SS; MF 32% no CR; MF n/a; 4.5 months Compartment-spe­cific data not reported Yes (4/19) ↑LFTs
(13/19); SS 38% (5/13); LCT DOR range 0.7-
(5/19) 25% (1/4); SS 10.1 months;
20% (1/5) TTR 2.4

Prakash Singh Shekhawat


months
 Lenalidomide65 32; MF/SS 28% (9/32) no CR 10 months; 8 months Skin RR 53% (10/19) Reports activ­ity, Blood RR 38% N/a Fatigue, skin
IB-IV; TTR 25 mg, MF IB-IV no details (5/13) flares, tran­sient
SS (11/32) 2 months; ↑blood SCs
10 mg,
4 months
*Response assess­ment tools and/or cri­te­ria for clin­i­cal end points may vary across CTCL tri­als.

Time to response is lon­ger than other listed treatments, 300 mg/m2, TTR = 15.7 weeks (180 days), >300 mg/m2, TTR = 8.4 weeks (59 days); poor details on extracutaneous dis­ease sites (4/9
entered as stage IVB had LN+ only); “rate of PD” = 39% (22/56) and 32% (12/38) for 300 mg/m2 and >300 mg/m2; median TTR for other treatment ~2 cycles.

Median time to response 8 months.
§
Caution when used with RT, espe­cially TSEBT.

Caution when used to bridge to allo­ge­neic HSCT (pos­si­ble risk of severe GVHD).
ALCL, ana­plas­tic large cell lym­phoma; DOR, duration of response; irAE, immune-related adverse event; LFT, liver function test; n/a, not appli­ca­ble; OR, odds ratio; PFS, pro­gres­sion-free sur­vival;
PN, peripheral neuropathy; RECIST, response eval­u­a­tion cri­te­ria in solid tumors; RT, radiation therapy; SD, stable disease; TTR, time to response; TTP, time to progression.

Personalizing treat­ment selec­tion in MF/SS  |  305


Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/303/1852064/303kim.pdf by guest on 13 December 2021
responses are pri­or­i­tized. In selected high-risk advanced-stage noted wors­en­ing of his skin dis­ease with recur­rent erythrod­
MF or SS, allo­ge­neic hema­to­poi­etic stem cell trans­plant (HSCT) erma. A skin biopsy con­firmed CTCL with­out evi­dence of LCT.
should be con­sid­ered as a poten­tial cura­tive ther­apy. His blood Sezary flow now showed >10,000 SCs/mm3. Updated
PET/CT find­ings again supported reac­tive or dermatopathic
LAD with no hepatosplenomegaly.

CLINICAL CASE Recognizing dis­ease het­ero­ge­ne­ity within the skin


A 64-year-old male presented with a 2-year his­tory of pro­gres­ and across extracutaneous com­part­ments
sive skin rash that cur­rently affected >80% of the body sur­face MF and SS are unique in their strik­ing het­ero­ge­ne­ity of clin­i­cal,
area with gen­er­al­ized erythroderma asso­ci­ated with severe, bio­log­i­cal, and molec­u­lar fea­tures and the need to indi­vid­u­al­
debil­i­tat­ing itching. Topical and sys­temic ste­roids were min­ ize man­age­ment for an opti­mal out­come. This het­ero­ge­ne­ity is

mally help­ ful. Skin biop­sies showed an atyp­ i­
cal T-cell infil­ fur­ther com­pli­cated by var­i­able clin­i­cal behav­ior across dis­ease
trate. Blood flow dem­on­strated an abnor­mal T-cell pop­u­la­tion com­part­ments (eg, skin, LN, blood). A mean­ing­ful clin­i­cal out­

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/303/1852064/303kim.pdf by guest on 13 December 2021


expressing CD3 and CD4 but lacking CD7 and CD26, a phe­ come of treat­ment in MF/SS con­sists of a dura­ble global (com­
no­type con­sis­tent with Sezary cells (SCs), which represented pos­ite of all­com­part­ments) objec­tive response and improve­ment
68% of lym­pho­cytes with an abso­lute count of 1250/mm3; T-cell of qual­ity of life (QoL) mea­sures,4-7 and achiev­ing these goals
recep­tor next-gen­er­a­tion sequenc­ing (NGS) stud­ies supported requires indi­vid­u­al­i­za­tion of ther­apy. Traditionally, the clin­i­cal
a clonal T-cell pro­cess in the skin and blood. Whole-body pos­ stage has been a pri­mary con­sid­er­ation in treat­ment selec­tion;
i­tron emis­sion tomog­ra­phy/com­puted tomog­ra­phy (PET/CT) how­ever, addi­tional guid­ance is often needed given the het­ero­
imag­ing showed an approx­im ­ a­tely 2 cm axil­lary and ingui­nal ge­ne­ity within stage group­ings. The extent/bur­den of dis­ease
lymph­ade­nop­a­thy (LAD) with a max­i­mum stan­dard­ized uptake or sta­tus of other established prog­nos­tic fac­tors (eg, LCT) in the
value of 3 to 4, con­sis­tent with reac­tive or dermatopathic LNs. skin or other dis­ease com­part­ments fur­ther con­trib­utes to treat­
A diag­no­sis of SS was established, presenting with low-bur­den ment selec­tion.8
SCs and clin­i­cally reac­tive LAD and thus clin­i­cal stage IVA1. The In early-stage MF with patch and/or plaque skin dis­ ease,
patient was treated with oral bexarotene with over­all global skin-directed treat­ment is pre­ferred, and improve­ment of QoL
par­ tial response (75% improve­ ment of his skin dis­ ease and is usu­ally the pri­mary focus of treat­ment. Primary skin-directed
>50% reduc­tion of his SCs), and sup­port­ive care was pro­vided treat­ments include top­i­cal prep­a­ra­tions of ste­roids, mechlore­
with top­ic ­ al ste­roids, emol­lients, and gabapentin for itching. thamine, ret­i­noids, phototherapy (eg, nar­row­band ultra­vi­o­let
Six months later while con­tinu­ing main­te­nance bexarotene, he B), and TSEBT.8 TSEBT is very effec­tive at low doses (eg, 12 Gy)

Thinner skin disease, erythroderma, Tumor-type, thicker skin disease,


blood disease large-cell transformation, LN disease

Mogamulizumab Brentuximab vedotin


Lacutamab Pralatrexate
(investigational)

Sézary cells Romidepsin


Pembrolizumab

Figure 1. Examples of newer systemic agents and their clinical activity in different patient profiles.

306  |  Hematology 2021  |  ASH Education Program


and most appro­pri­ate in patients who have wide­spread thicker ond­ary skin infec­tions as patients often have com­pro­mised skin
or folliculotropic (hair fol­li­cle involve­ment) skin dis­ease.9 If skin- bar­rier func­tion and are com­monly col­o­nized with Staphylococ-
directed treat­ment options fail to man­age a patient’s dis­ease, cus aureus.17 Management with an anti-staph sup­port­ive reg­i­men
then esca­lat­ing to sys­temic agents is appro­pri­ate. has been shown to improve their itching, clear their skin, and
Patients with advanced-stage MF and SS have vary­ing clin­i­ lower infec­tion-asso­ci­ated com­pli­ca­tions.18 The vis­i­ble nature of
cal pre­sen­ta­tions rang­ing from tumor-type skin dis­ease to gen­ CTCL adds a unique neg­a­tive life impact because it con­stantly
er­al­ized erythroderma in which the type and extent of their skin reminds patients of their dis­ease and often pro­hib­its patients
and extracutaneous dis­ease (LN and/or blood dis­ease) drive the from social inter­ac­tion, inten­si­fy­ing the emo­tional bur­den of
ther­ a­
pies that best address a patient’s over­ all dis­
ease. Many their strug­gles.19,20 Thus, in con­trast to other indo­lent lym­pho­mas
stan­dard treat­ment options exist for advanced-stage patients, such as low-grade fol­lic­u­lar lym­phoma, in which watch­ful wait­
includ­ing sys­temic bio­log­i­cal and targeted ther­a­pies or tra­di­ ing can be accept­able, patients with CTCL are often on con­stant
tional cyto­toxic che­mo­ther­apy.8 The ulti­mate selec­tion of the pri­ treat­ment, and QoL ele­ments may lead to a deci­sion to change
mary ther­apy will be based on the assess­ment of the skin and the ther­apy with­out objec­tive dis­ease pro­gres­sion. Optimizing sup­
sever­ity of any extracutaneous dis­ease and on aligning ther­a­pies port­ive care is essen­tial, and clin­i­cal end points such as time to

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/303/1852064/303kim.pdf by guest on 13 December 2021


that address the most symp­tom­atic and threat­en­ing dis­ease. next sig­nif­i­cant ther­apy may be use­ful mea­sures of over­all clin­i­
Systemic treat­ments often exhibit dif­fer­en­tial effi­cacy across cal ben­e­fit.21
skin, blood, and nodal com­part­ments (Table 1 and Figure 1). The
response inter­pre­ta­tion can be fur­ther com­pli­cated by the vary­ Prioritizing treat­ment options in SS
ing response cri­te­ria and clin­i­cal end points used in ear­lier clin­ In patients with SS, the leu­ke­mic sub­type of CTCL, clin­i­cal man­
i­cal tri­als in CTCL; how­ever, more recent tri­als have uti­lized the age­ment is often strat­i­fied by the bur­den of the blood Sezary
inter­na­tional con­sen­sus cri­te­ria that have helped com­pare results dis­ease. Primary sys­temic ther­apy for those with a lower Sezary
across stud­ies.4 There are fre­quent mixed responses in the skin or bur­ den (<5,000 SCs/mm3) includes oral bexarotene,22 meth­
across dif­fer­ent com­part­ments with dis­ease, and ther­a­pies that o­trex­ate,23 and extra­cor­po­real photopheresis.24 In those with
reduce skin dis­ease may not be effec­tive in clear­ing the blood a higher Sezary bur­ den, his­ tone deacetylase inhib­

tors (eg,
or LN dis­ease, and vice versa. For exam­ple, in a patient with MF romidepsin) or newer ther­a­pies, such as MOGA, with more reli­
and wide­spread ulcerating skin tumors (±LCT), we would select able activ­ity in the blood com­part­ment would be appro­pri­ate
treat­ments with known reli­able activ­ity in the skin com­part­ment front­line options (Table 1).14 With MOGA, an anti-CCR4 mono­clo­
(eg, TSEBT, BV).9-12 However, in a patient with SS and high-bur­ nal anti­body in which CCR4 is highly and con­sis­tently expressed
den Sezary dis­ease, we would opt for ther­a­pies that are effi­cient by SCs, objec­tive blood response was observed in 68% (83/122)
at reduc­ing the blood dis­ease (eg, MOGA, romidepsin).13,14 Fur­ of patients with 44% (54/122) achiev­ ing com­plete remis­sion
thermore, some ther­a­pies do not have established, mean­ing­ful (CR) in the piv­otal trial, MAVORIC.13 The median time to response
activ­ity in LCT or bulky dis­ease, whereas some ther­a­pies have in the blood was 1 month, and the median dura­tion of blood
shown activ­ity across all­com­part­ments, includ­ing LCT dis­ease. response was 26 months. The skin response rate was 42% with a
Traditional com­bi­na­tion che­mo­ther­apy reg­i­mens in sys­temic lower nodal response rate of 17% and a global (com­pos­ite of all­
TCLs are often reserved for patients with refrac­tory, advanced com­part­ments) over­all response rate of 28%. However, patients
dis­ease or when allo­ge­neic HSCT is planned for con­sol­i­da­tion.8 with active LCT were excluded in the MOGA piv­otal trial. Given
Lastly, the poten­tial toxicities and tol­er­a­bil­ity of chronic ther­apy the impres­sive clin­i­cal activ­ity in the blood com­bined with a
must be con­sid­ered in pri­or­i­tiz­ing treat­ments and per­son­al­iz­ing favor­able tol­er­a­bil­ity pro­file, MOGA may be a pre­ferred option
man­age­ment. in high-bur­den SS with­out skin LCT or LN dis­ease. Similarly,
the anti-KIR3DL2 anti­body (lacutamab) cur­rently under clin­
Importance of mul­ti­dis­ci­plin­ary care and man­age­ment i­cal devel­op­ment has been shown to be more effec­tive in
of QoL issues: essen­tial role of sup­port­ive ther­apy reduc­ing blood dis­ease com­pared to nodal dis­ease (blood
Ideally, the man­age­ment of patients with CTCL involves a mul­ti­ response rate [RR] 56% vs nodal RR 11%).25 Conversely, BV,
dis­ci­plin­ary approach. It is help­ful to estab­lish col­lab­o­ra­tive col­ which dem­on­strated reli­able effi­cacy in the skin (includ­ing
leagues in hema­tol­ogy/med­i­cal oncol­ogy, radi­a­tion oncol­ogy, LCT) and the LN dis­ease,11 does not have established activ­ity
pathol­ogy, and der­ma­tol­ogy who are inter­ested in cuta­ne­ous in Sezary dis­ease and SS and was excluded in the piv­otal trial
lym­phoma. To address the exten­sive skin dis­ease, spe­cial­ized in CTCL. In SS with exten­sive LN dis­ease (±LCT), romidepsin
radi­a­tion meth­ods such as TSEBT serve as an impor­tant ther­a­ may be pri­or­i­tized over MOGA or other options, as romidepsin
peu­tic alter­na­tive; how­ever, access to cen­ters with TSEBT can has dem­on­strated con­sis­tent clin­i­cal activ­ity across com­part­
be an issue. Low-dose TSEBT can be com­bined with treat­ments ments (Table 1).
that have great effi­cacy in the blood and/or LN com­part­ments
to improve the over­all com­pos­ite response and response dura­ Length of ther­apy and main­te­nance strat­e­gies
tion.15,16 Patients may have exten­sive skin wounds or atyp­i­cal skin To address the chronic course of MF/SS, newer sys­temic ther­a­
infec­tions or develop skin lesions that clin­i­cally and/or path­o­ pies were devel­oped in which clin­i­cal stud­ies allowed patients
log­i­cally mimic CTCL, includ­ing an inflam­ma­tory skin reac­tion to to con­tinue treat­ment with­out a fixed dura­tion until intol­er­ance
lym­phoma ther­apy (eg, MOGA, pembrolizumab). Thus, a close or dis­ease pro­gres­sion occurred. However, for ther­a­pies that have
col­lab­o­ra­tion across spe­cial­ties is essen­tial in pro­vid­ing appro­ dose-cumu­la­tive toxicities such as the periph­eral neu­rop­a­thy
pri­ate clin­i­cal care and opti­mal out­come in CTCL. asso­ci­ated with BV treat­ment, we should con­sider a fixed treat­
Intolerable itching is a com­mon QoL in CTCL, espe­cially in SS. ment course (<6-8 cycles) and re-treat with BV later when
The effec­tive man­age­ment of itching also reduces the risk of sec­ needed or use reduced doses of BV.10,26.27 MOGA can be con­tin­ued
Prakash Singh Shekhawat
Personalizing treat­ment selec­tion in MF/SS  |  307
long-term, but in one-third of patients, it may lead to a rash the phase 3 trial of BV in CD30+ CTCL, the pri­mary end point
(median time to rash, 105 days),13,28-30 and the sever­ ity of the of an objec­tive response rate last­ing >4 months (ORR4) was
rash may affect the length of treat­ment with MOGA. Despite the higher in patients with LCT than with­out LCT (65% vs 39%).11,12
occur­rence of MOGA, patients can be re-treated with MOGA. For Pralatrexate and romidepsin, each as a sin­ gle agent, have
chronic ther­apy in CTCL, treat­ment options or strat­e­gies that also dem­on­strated clin­i­cal activ­ity in MF or SS with LCT,14,37,38
min­im
­ ize immune sup­pres­sion will be pre­ferred given the high- though the data in these sin­gle agents are not as robust as
risk of recur­rent skin infec­tions. in the piv­otal trial of BV. MOGA would not be pri­or­i­tized in
After ini­tial dis­ease reduc­tion, a main­te­nance strat­egy of a sus­ patients with LCT given the lack of established data.13 Older
tain­able treat­ment reg­i­men can be con­sid­ered. This may involve agents such as lipo­so­mal doxo­ru­bi­cin or gemcitabine have
increas­ing the treat­ment inter­val of intra­ve­nous ther­a­pies or transi­ greater tox­ic­ity pro­files but can be used in MF/SS with LCT in
tioning to an oral agent.31 Utilizing a less intense main­te­nance dose refrac­tory set­tings or when newer agents are not avail­­able.39,40
reg­im
­ en of ther­a­pies may lower treat­ment-related toxicities and A recent clin­ i­
cal trial of pembrolizumab in patients with
pro­vide a lon­ger over­all clin­i­cal ben­e­fit. Integrating skin-directed relapsed/refrac­tory MF or SS showed prom­is­ing clin­i­cal activ­
ther­apy is not only essen­tial in com­bi­na­tion with sys­temic agents ity in patients with LCT and can be con­sid­ered in relapsed or

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/303/1852064/303kim.pdf by guest on 13 December 2021


but nec­es­sary when the sys­temic dis­ease is cleared, and patients refrac­tory set­tings.41
then have pri­mar­ily skin-lim­ited dis­ease.
Biomarker-guided selec­tion and inte­gra­tion of ther­a­pies
in MF/SS-CTCL
Currently, numer­ous avail­­able and inves­ti­ga­tional ther­a­pies in
MF/SS-CTCL can tar­get the cell sur­face mol­e­cules of malig­nant
CLINICAL CASE (CON­T IN­UED) T cells, disrupted cellular pathways, and/or the tumor micro­en­vi­
Given the higher bur­den of blood Sezary dis­ease and lack of ron­ment (TME), includ­ing the immune milieu (Table 1). Reliable
LCT in the skin, the patient was treated with MOGA and after bio­mark­ers that enrich the patient sub­sets that can ben­e­fit with
2 cycles expe­ri­enced >90% skin improve­ment and reduc­tion of spe­ cific ther­ a­pies would be ideal, and such bio­ marker infor­
itching. His blood Sezary dis­ease cleared promptly with­out mea­ ma­tion may help select and pri­or­i­tize ther­a­pies among a list of
sur­able abnor­mal cir­cu­lat­ing T cells (B0) by flow cytom­e­try. After options.
4 months of con­tin­ued great response with con­tin­ued blood CR Of the tar­gets on the malig­nant cell sur­face in CTCL, CD30 has
(B0), MOGA was discontinued, and the patient con­tin­ued top­i­cal the most robust data, with a CD30 targeting agent, BV. With the
ther­apy as needed for lim­ited resid­ual skin dis­ease. The patient’s use of more sen­si­tive tools for CD30 detec­tion (eg, mul­ti­spec­
blood Sezary remained clear and his skin near CR off MOGA, but tral imag­ing), sam­ples with nondetectable CD30 expres­sion by
after sev­eral months, he expe­ri­enced dis­ease pro­gres­sion in the rou­tine immu­no­his­to­chem­is­try (IHC) and light micros­copy dem­
skin with rap­idly wors­en­ing, wide­spread thick plaques and nod­ on­strated cell-sur­face CD30 mol­e­cules.10 In MF/SS, the inter- and
u­lar, tumor-type dis­ease. Skin biopsy showed LCT with 10–40% intrapatient (interlesional) CD30 expres­ sion lev­els (by rou­
tine
of neo­plas­tic T cells expressing CD30 by immu­no­his­to­chem­is­try. IHC) can be highly var­i­able in the skin, and BV has dem­on­strated
PET/CT now showed more nota­ble LAD, and a core needle sam­ sig­nif­i­cant clin­i­cal activ­ity in patients with a spec­trum of CD30
pling showed involve­ment with T-cell lym­phoma (CD30 expres­ expres­sion in 2 inde­pen­dent stud­ies in MF/SS.10,36,42 Patients with
sion of 30%). Given the sig­nif­i­cant CD30 expres­sion in the skin >5% CD30max (max­i­mum CD30 level from mul­ti­ple skin sam­ples)
and LN com­part­ments, BV was selected as the next ther­apy. had more reli­able over­all clin­i­cal response com­pared with those
with CD30max <5%; how­ever, mean­ing­ful activ­ity was observed
across all­CD30 lev­els. Further, in the piv­otal study of BV in CD30+
Management of LCT in MF and SS CTCL (ALCANZA), patients with mul­ti­ple skin biop­sies showed a
LCT in MF or SS is rec­og­nized as one of the stron­gest inde­pen­dent nota­ble var­i­abil­ity of CD30 lev­els, rang­ing from nondetectable
adverse prog­nos­tic indi­ca­tors,32,33 and thus knowl­edge of LCT is to >50% expres­sion.11,12 Thus, BV is a treat­ment option even in
a key fac­tor in treat­ment selec­tion (Table 1). However, nota­ble those with low/neg­li­gi­ble lev­els of CD30 expres­sion, espe­cially
inter-rater (pathol­o­gist to pathol­o­gist) var­i­abil­ity is observed in refrac­tory set­tings.
in the inter­pre­ta­tion of LCT as the cri­te­ria is an arbi­trary cut­off Therapies such as MOGA can tar­get the malig­nant cells as
value (>25% of malig­nant T cells are large cells) by his­to­path­o­ well as TME, as the tar­get mol­e­cule CCR4 is expressed not only
logic eval­u­a­tion.34 Accordingly, a sub­set of patients des­ig­nated by the malig­nant T cells but also by the reg­u­la­tory T cells. CCR4 is
as LCT may have a more indo­lent course; there­fore, appro­pri­ate a chemokine recep­tor asso­ci­ated with skin traf­fick­ing, but unlike
clin­ic
­ al judg­ment is essen­tial for opti­mal man­age­ment. In con­ CD30, the mol­ e­
cule is fre­ quently and con­ sis­
tently expressed
trast to sys­temic lym­pho­mas in which anthracycline-based ther­ in the lesional skin (median CCR4, 80%; range of 1%-100% by
apy and autol­o­gous trans­plants are con­sid­ered in the set­ting of IHC) and may not be use­ful as a bio­marker in CTCL.13 However,
transformed dis­ease, LCT in MF/SS is man­aged dif­fer­ently, with some patients with MF/SS have neg­li­gi­ble CCR4 expres­sion, and
greater indi­vid­u­al­ized tai­lor­ing for the extent and biol­ogy of the fur­ther stud­ies will be needed to estab­lish the role of MOGA in
transformed dis­ease. these patients. Further, assess­ment for CCR4 expres­sion is not
CD30 expres­sion is not a require­ment for LCT cri­te­ria, but rou­tinely avail­­able, so a stan­dard­ized pro­to­col for CCR4 assess­
the median CD30 expres­sion is higher in the lesional tis­sue of ment and a bet­ter under­stand­ing of its cor­re­la­tion with clin­i­cal
patients with LCT com­pared to those with­out LCT,35 and thus out­come needs to be established.43 Although both CCR4 and
BV is often a highly effec­tive treat­ment in MF with LCT.10,12,36 In KIR3DL2 are both com­monly expressed in skin dis­ease with LCT,

308  |  Hematology 2021  |  ASH Education Program


nei­ther MOGA nor anti-KIR3DL2 anti­body (lacutamab) has estab­ An NGS study by Ungewickell et al reported that LCT is asso­
lished activ­ity data in patients with LCT.25,44 ci­
ated with a high muta­ tion bur­den and recur­ rent PLCG1
Newer bio­marker plat­forms (eg, action­able NGS pan­els, TME alter­ations.52 Furthermore, recent stud­ies explor­ing the pro­
pro­fil­ing) may help guide treat­ment selec­tion and pri­or­i­tize ther­ grammed cell death 1 (PD-1) and programmed cell death
a­pies in selected set­tings.45,46 The knowl­edge of Th2 skewing 1 ligand 1 (PD-L1) axis show a pos­si­ble link between PD-L1
with dis­ease pro­gres­sion has led to the selec­tion of immune struc­tural var­i­ants and LCT,45 supporting the hypoth­e­sis that
ther­a­pies that shift the immune pro­file from Th2 to Th1, includ­ patients with LCT may have more TME immune alter­ations that
ing inter­ fer­
ons, IL-12, and toll-like recep­tor ago­ nists. Agents allow tumor growth. These patients with PD-L1 alter­ation have
known to deplete reg­u­la­tory T cells in the TME, such as MOGA or expe­ri­enced clin­i­cal ben­e­fit with anti-PD-1 anti­body such as
CD25-targeting agents,47 and to acti­vate mac­ro­phages (increase pembrolizumab.41,53
the “eat-me” sig­nal), such as agents targeting the CD47-SIRPα
axis (eg, SIRPαFc, magrolimab),48,49 may be con­sid­ered as sin­
gle agents or part of com­bi­na­tion reg­i­mens. Patients with geno­
mic data that dem­on­strate path­o­genic alter­ation in the T-cell

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/303/1852064/303kim.pdf by guest on 13 December 2021


immune reg­u­la­tory axis may ben­e­fit from immune ther­a­pies that CLINICAL CASE (Con­tin­ued)
include check­point inhib­i­tors.41 The patient was treated with BV, with great clin­i­cal response
In patients with refrac­tory or high-risk dis­ease, using NGS in the skin and flat­ten­ing of his thicker, tumor-type skin dis­
plat­forms to eval­u­ate for addi­tional prog­nos­tic clues or action­ ease that showed LCT by pathol­ogy. PET/CT obtained after
able path­o­genic genetic var­i­ants can help pri­or­i­tize ther­a­pies. 3 cycles of BV showed clin­i­cal CR of LN dis­ease. The blood
For exam­ple, JAK inhib­i­tors may be con­sid­ered in patients with com­part­ment remained clear of mea­sur­able dis­ease by flow
relapsed/refrac­tory dis­ease with a path­o­genic muta­tion of cytom­e­try. The patient had clin­i­cal fea­tures con­sis­tent with a
JAK1/3.50 Moreover, we can extrap­o­late geno­mic bio­marker data high-risk prog­nos­tic pro­file, includ­ing a his­tory of high blood
from other T-cell lym­pho­mas. For exam­ple, CCR4 gain of func­ SC bur­den, LN dis­ease, and LCT.33 At the ini­ti­a­tion of BV, allo­
tion muta­tions have been asso­ci­ated with supe­rior clin­i­cal out­ ge­neic HSCT was discussed as a poten­tial cura­tive ther­apy in
comes in adult T-cell leu­ke­mia/lym­phoma patients treated with this high-risk set­ting. The patient’s sib­ling was iden­ti­fied as a
MOGA.51 Thus, if such poten­tial path­o­genic var­i­ants of CCR4 are 10/10 matched donor. The patient was 67 years old at this time
noted by NGS data, per­haps MOGA can be pri­or­i­tized over other point, and he agreed to move forth with allo­ge­neic HSCT using
ther­a­pies in high-bur­den Sezary patients or in refrac­tory dis­ease. TSEBT and a total lym­phoid irra­di­a­tion/antithymocyte glob­u­lin
As pre­vi­ously discussed, LCT is a very impor­tant clin­i­cal nonmyeloablative reg­i­men.
and path­o­logic fac­tor to con­sider when selecting ther­apy.

Table 2. Allogeneic HSCT in MF/SS-CTCL

Reference N Intervention Efficacy Transplant-related toxicities


Hosing et al 55
47 6% abla­tive ORR  =  n/a, 2-year PFS 40%, 2-year OS 67% 1-year NRM 10%
94% RIC/NMA 4-year PFS 26%, 4-year OS 51% 2-year NRM 17%
Gr 2–4 aGVHD 40%;
cGVHD 28%
Duarte et al56 60 33% abla­tive ORR  =  n/a, 2-year PFS 34%, 2-year OS 54% 1-year NRM 20%
67% RIC/NMA 5-year PFS 32%, 5-year OS 46% 7-year NRM 22%
7-year PFS 30%, 7-year OS 44%
Paralkar et al57 12 17% abla­tive ORR  =  67%, 2-year PFS 23%, 2-year OS 1-year NRM 25%
83% RIC 56%
De Masson et al58 37 32% abla­tive ORR  =  n/a, 2-year PFS 31%, 2-year OS 1-year NRM 18%
68% RIC/NMA 57% 2-year NRM 18%
Gr 2-4 aGVHD 76%;
cGVHD 44%
Lechowicz 129 36% abla­tive ORR  =  n/a, 1-year PFS 31%, 1-year OS 54% 1-year NRM 19%
et al59 64% RIC/NMA 5-year PFS 17%, 5-year OS 32% 5-year NRM 22%
Gr 2-4 aGVHD 41%; cGVHD 43%
Isufi et al60 16 (n  =  23; 16 MF/SS; RIC except 2 haploidentical CR rate  =  56% 100-day NRM 12%
7 G/D TCL) OS 75% (12 of 16 patients) w/median fol­ Gr 2-4 aGVHD 50%;
low-up 5.5 years cGVHD 56%
Weng et al61 35 100% NMA ORR (CR)  =  80% (57%), 2-year PFS 60%, 1-year NRM 3%
2-year OS 68% 2-year NRM 14%
5-year PFS 41%, 3-year OS 62%, Gr 2-4 aGVHD 16%;
5-year OS 56% cGVHD 32%
aGVHD, acute GVHD; cGVHD, chronic GVHD; Gr, grade; NMA, non-myeloablative; RIC, reduced-inten­sity con­di­tion­ing.
Prakash Singh Shekhawat
Personalizing treat­ment selec­tion in MF/SS  |  309
Allogeneic HSCT as a path to estab­lish dura­ble remis­sion allo­ge­neic HSCT. The unique QoL issues bring unique chal­lenges
in high-risk MF/SS in man­ag­ing patients with CTCL.
Patients with advanced-stage MF and SS have a median sur­ Ultimately, in this highly het­ero­ge­neous dis­ease group we hope
vival of <5 years, are often treated with mul­ti­ple sequen­tial sys­ to estab­lish bio­mark­ers and strat­e­gies that enrich (bet­ter align sub­
temic ther­a­pies, and even­tu­ally become refrac­tory to avail­­able sets) for dura­ble clin­i­cal response and address resis­tance/escape
agents.33 High-dose che­mo­ther­apy followed by autol­o­gous mech­a­nisms while min­i­miz­ing toxicities. More per­son­al­ized and
HSCT has not shown a dura­ble ben­e­fit,54 but allo­ge­neic HSCT tol­er­ab
­ le immunotherapies may be a path­way to achieve a more
that relies on a graft vs lym­phoma effect has led to long-term dura­ ble or sustained clin­ i­
cal response. Advances in bio­ marker-
remis­sions (Table 2).55-61 guided inves­ti­ga­tions in CTCL will enable us to apply evi­dence-
There is some con­tro­versy regard­ing which sub­set of patients based approaches in pri­ or­

tiz­
ing ther­

pies and help develop
should move forth for allo­ge­neic trans­plant in MF/SS and when com­bi­na­tion strat­e­gies that may lead to the clin­i­cal syn­ergy of
in their dis­ease course it is appro­pri­ate. Allogeneic HSCT should com­bined agents.
be con­sid­ered for appro­pri­ate patients with stage IIB through IV
dis­ease who are refrac­tory to mul­ti­ple lines of sys­temic ther­

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/303/1852064/303kim.pdf by guest on 13 December 2021


Conflict-of-inter­est dis­clo­sure
apy, dem­on­strate declin­ing dura­tion of clin­i­cal ben­e­fit, or pre­
Youn H. Kim: Advisory board, Research funding; Innate: Research
s­ent with a very high-risk pro­file. A large inter­na­tional study of
funding; Corvus: Research funding; Galderma: Advisory board,
prog­ nos­ tic fac­
tors in advanced-stage MF and SS (retro-CLIPI)
Research funding; CRISPR therapeutics: Research funding; Secura
iden­ti­fied 3 risk groups with dis­tinct prog­nos­tic out­comes based
Bio: Advisory board; Trillium: Research funding.
on the num­ber of key adverse prog­nos­tic param­e­ters, includ­ing
extracutaneous dis­ease and LCT.33 Patients in the high-risk group
with a 5-year over­all sur­vival (OS) of 28% were clearly appro­pri­ Off-label drug use
ate for allo­ge­neic HSCT. A sig­nif­i­cant pro­por­tion of patients in Youn H. Kim: none.
the inter­me­di­ate-risk (5-year OS, 44%) group may be con­sid­ered
for allo­ge­neic HSCT. The 5-year OS with allo­ge­neic HSCT in the Correspondence
published stud­ies ranged from 32% to 56%,55-62 likely supporting Youn H. Kim, Stanford University School of Medicine, 780 Welch
an improved sur­vival ben­e­fit pro­vided that most patients who Rd, CJ220D, C. J. Huang Bldg, Palo Alto, CA 94304; e-mail:
received allo­ge­neic HSCT included those with inter­me­di­ate- to younkim@stanford​­.edu.
high-risk retro-CLIPI pro­files. The opti­mal tim­ing for allo­ge­neic
HSCT is when the dis­ease is well con­trolled (CR or near CR of all­ References
dis­ease com­part­ments) and before the dis­ease has progressed 1. Willemze R, Cerroni L, Kempf W, et al. The 2018 update of the WHO-EORTC
to a state in which the chance of response or sur­vival with allo­ clas­si­fi­ca­tion for pri­mary cuta­ne­ous lym­pho­mas. Blood. 2019;133(16):1703-
ge­neic HSCT is low. The skin com­part­ment is a fre­quent site of 1714.
2. Kim EJ, Hess S, Richardson SK, et al. Immunopathogenesis and ther­apy of
posttransplant dis­ease relapse, and thus TSEBT is often an impor­ cuta­ne­ous T cell lym­phoma. J Clin Invest. 2005;115(4):798-812.
tant and nec­es­sary com­po­nent of the pre­pa­ra­tory strat­egy in 3. Bobrowicz M, Fassnacht C, Ignatova D, Chang YT, Dimitriou F, Guenova
MF/SS. The median age of patients with advanced MF or SS at E. Pathogenesis and ther­apy of pri­mary cuta­ne­ous T-cell lym­phoma: Col­
diag­no­sis is 60 to 65 years, so an allo­ge­neic reg­i­men with fewer legium Internationale Allergologicum (CIA) update 2020. Int Arch Allergy
trans­plant-related toxicities has also been explored.61 For each Immunol. 2020;181(10):733-745.
4. Olsen EA, Whittaker S, Kim YH, et al. Clinical end points and response cri­
patient, a deci­sion to trans­plant requires care­ful con­sid­er­ation of te­ria in myco­sis fungoides and Sezary syn­drome: a con­sen­sus state­ment
the long-term ben­e­fits and sig­nif­i­cant risks, includ­ing the poten­ of the International Society for Cutaneous Lymphomas, the United States
tial for graft-ver­sus-host dis­ease (GVHD) and dis­ease relapse after Cutaneous Lymphoma Consortium, and the Cutaneous Lymphoma Task
trans­plant. Force of the Euro­pean Organisation for Research and Treatment of Cancer.
J Clin Oncol. 2011;29(18):2598-2607.
In patients with aggres­sive MF or SS, there remains an unmet
5. Kim YH, Demierre MF, Kim EJ, et al. Clinically mean­ing­ful reduc­tion in pru­
need for addi­tional safe and effec­tive ther­a­pies that can bridge ri­tus in patients with cuta­ne­ous T-cell lym­phoma treated with romidepsin.
patients to allo­ge­neic HSCT. And espe­cially for those who are Leuk Lymphoma. 2013;54(2):284-289.
not eli­gi­ble for allo­ge­neic HSCT or have no opti­mal donors, new 6. Dummer R, Prince HM, Whittaker S, et al. Patient-reported qual­ity of life in
and novel immune ther­a­pies and approaches are needed. As patients with relapsed/refrac­tory cuta­ne­ous T-cell lym­phoma: results from
the randomised phase III ALCANZA study. Eur J Cancer. 2020;133(July):120-
these inno­va­tive ther­a­pies are made avail­­able, it is essen­tial to
130.
inte­grate them to man­age patients suc­cess­fully and to con­tinue 7. Porcu P, Hudgens S, Horwitz S, et al. Quality of life effect of the anti-CCR4
explor­ing bio­mark­ers that can help us guide our selec­tion or pri­ mono­clo­nal anti­body mogamulizumab ver­sus vorinostat in patients with
or­i­ti­za­tion of treat­ment options. cuta­ne­ous T-cell lym­phoma. Clin Lymphoma Myeloma Leuk. 2021;21(2):97-
105.
8. Mehta-Shah N, Horwitz SM, Ansell S, et al. NCCN guide­lines insights: pri­
Conclusions and future direc­tions
mary cuta­ne­ous lym­pho­mas, ver­sion 2.2020. J Natl Compr Canc Netw.
The clin­i­cal, path­o­log­i­cal, and bio­log­i­cal fea­tures in MF and SS 2020;18(5):522-536.
are highly het­ero­ge­neous, with sig­nif­i­cant inter- and intra-­patient 9. Hoppe RT, Harrison C, Tavallaee M, et al. Low-dose total skin elec­tron beam
var­i­abil­ity that requires a per­son­al­ized approach to man­age­ ther­apy as an effec­tive modal­ity to reduce dis­ease bur­den in patients with
ment. Available and inves­ti­ga­tive ther­a­pies often have dif­fer­en­ myco­sis fungoides: results of a pooled anal­y­sis from 3 phase-II clin­ic ­ al tri­
als. J Am Acad Dermatol. 2015;72(2):286-292.
tial activ­ity across dis­ease com­part­ments, and thus in addi­tion
10. Kim YH, Tavallaee M, Sundram U, et al. Phase II inves­ti­ga­tor-ini­ti­ated study
to clin­i­cal stage and the pres­ence of LCT, com­part­men­tal activ­ of brentuximab vedotin in myco­sis fungoides and Sézary syn­drome with
ity is an impor­tant ele­ment that guides treat­ment selec­tion. In var­i­able CD30 expres­sion level: a multi-insti­tu­tion col­lab­o­ra­tive pro­ject.
appro­pri­ate high-risk set­tings, patients should be con­sid­ered for J Clin Oncol. 2015;33(32):3750-3758.

310  |  Hematology 2021  |  ASH Education Program


11. Prince HM, Kim YH, Horwitz SM, et al; ALCANZA Study Group. Brentuximab and devel­op­ment of a prog­nos­tic model. J Clin Oncol. 2015;33(32):3766-
vedotin or phy­si­cian’s choice in CD30-pos­i­tive cuta­ne­ous T-cell lym­phoma 3773.
(ALCANZA): an inter­na­tional, open-label, randomised, phase 3, multicentre 34. Gru AA, Kim J, Pulitzer M, et al. The use of cen­tral pathol­ogy review with
trial. Lancet. 2017;390(10094):555-566. dig­i­tal slide scan­ning in advanced-stage myco­sis fungoides and Sézary
12. Kim YH, Prince HM, Whittaker S, et al. Response to brentuximab vedotin syn­drome: a multi-insti­tu­tional and inter­na­tional pathol­ogy study. Am J
ver­sus phy­si­cian’s choice by CD30 expres­sion and large cell trans­for­ma­tion Surg Pathol. 2018;42(6):726-734.
sta­tus in patients with myco­sis fungoides: an ALCANZA sub-anal­y­sis. Eur J 35. Edinger JT, Clark BZ, Pucevich BE, Geskin LJ, Swerdlow SH. CD30 expres­
Cancer. 2021;148(May):411-421. sion and pro­lif­er­a­tive frac­tion in nontransformed myco­sis fungoides. Am J
13. Kim YH, Bagot M, Pinter-Brown L, et al; MAVORIC Investigators. Mogamuli­ Surg Pathol. 2009;33(12):1860-1868.
zumab ver­sus vorinostat in pre­vi­ously treated cuta­ne­ous T-cell lym­phoma 36. Duvic M, Tetzlaff MT, Gangar P, Clos AL, Sui D, Talpur R. Results of a phase
(MAVORIC): an inter­na­tional, open-label, randomised, con­trolled phase 3 II trial of brentuximab vedotin for CD30+ cuta­ne­ous T-cell lym­phoma and
trial. Lancet Oncol. 2018;19(9):1192-1204. lymphomatoid papulosis. J Clin Oncol. 2015;33(32):3759-3765.
14. Whittaker SJ, Demierre MF, Kim EJ, et al. Final results from a mul­ti­cen­ter, 37. Horwitz SM, Kim YH, Foss F, et al. Identification of an active, well-tol­er­ated
inter­na­tional, piv­otal study of romidepsin in refrac­tory cuta­ne­ous T-cell dose of pralatrexate in patients with relapsed or refrac­tory cuta­ne­ous
lym­phoma. J Clin Oncol. 2010;28(29):4485-4491. T-cell lym­phoma. Blood. 2012;119(18):4115-4122.
15. Jothishankar B, Almazan T, Kim Y, et al. Romidepsin and total skin elec­tron 38. O’Connor OA, Pro B, Pinter-Brown L, et  al. Pralatrexate in patients with
beam ther­apy in advanced stage myco­sis fungoides and Sézary syn­drome. relapsed or refrac­tory periph­eral T-cell lym­phoma: results from the piv­otal

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/303/1852064/303kim.pdf by guest on 13 December 2021


Br J Haematol. 2019;186(2):377-379. PROPEL study. J Clin Oncol. 2011;29(9):1182-1189.
16. Fong S, Hong EK, Khodadoust MS, et al. Low-dose total skin elec­tron beam 39. Dummer R, Quaglino P, Becker JC, et al. Prospective inter­na­tional mul­ti­cen­
ther­apy com­bined with mogamulizumab for refrac­tory myco­sis fungoides ter phase II trial of intra­ve­nous pegylated lipo­so­mal doxo­ru­bi­cin monoche­
and Sézary syn­drome. Adv Radiat Oncol. 2021;6(3):100629. motherapy in patients with stage IIB, IVA, or IVB advanced myco­ sis
17. Ahern K, Gilmore ES, Poligone B. Pruritus in cuta­ne­ous T-cell lym­phoma: a fungoides: final results from EORTC 21012. J Clin Oncol. 2012;30(33):4091-
review. J Am Acad Dermatol. 2012;67(4):760-768. 4097.
18. Lindahl LM, Willerslev-Olsen A, Gjerdrum LMR, et  al. Antibiotics inhibit 40. Duvic M, Talpur R, Wen S, Kurzrock R, David CL, Apisarnthanarax N. Phase
tumor and dis­ ease activ­ ity in cuta­ne­ ous T-cell lym­phoma. Blood. II eval­u­a­tion of gemcitabine monotherapy for cuta­ne­ous T-cell lym­phoma.
2019;134(13):1072-1083. Clin Lymphoma Myeloma. 2006;7(1):51-58.
19. Demierre MF, Kim YH, Zackheim HS. Prognosis, clin­i­cal out­comes and qual­ 41. Khodadoust MS, Rook AH, Porcu P, et al. Pembrolizumab in relapsed and
ity of life issues in cuta­ne­ous T-cell lym­phoma. Hematol Oncol Clin North refrac­tory myco­sis fungoides and Sézary syn­drome: a mul­ti­cen­ter phase II
Am. 2003;17(6):1485-1507. study. J Clin Oncol. 2020;38(1):20-28.
20. Jonak C, Porkert S, Oerlemans S, et  al. Health-related qual­ity of life in 42. Rahbar Z, Li S, Tavallaee M, Novoa RA, Kim J, Kim YH. Variability in the
cuta­ne­ous lym­pho­mas: past, pres­ent and future. Acta Derm Venereol. expres­sion of immu­no­his­to­chem­i­cal mark­ers: impli­ca­tions for bio­marker
2019;99(7):640-646. inter­pre­ta­tion in cuta­ne­ous T-cell lym­phoma. J Invest Dermatol. 2018;138(5):
21. Campbell BA, Scarisbrick JJ, Kim YH, Wilcox RA, McCormack C, Prince HM. 1204-1206.
Time to next treat­ment as a mean­ing­ful end­point for tri­als of pri­mary cuta­ 43. Fujii K, Sakamoto Y, Masaki A, et al. Immunohistochemistry for CCR4 C-ter­mi­nus
ne­ous lym­phoma. Cancers (Basel). 2020;12(8):2311. pre­dicts CCR4 muta­tions and mogamulizumab effi­cacy in adult T-cell leu­ke­mia/
22. Duvic M, Hymes K, Heald P, et al; Bexarotene Worldwide Study Group. Bex­ lym­phoma. J Pathol Clin Res. 2021;7(1):52-60.
arotene is effec­tive and safe for treat­ment of refrac­tory advanced-stage 44. Duvic M, Pinter-Brown LC, Foss FM, et  al. Phase ½ study of mogamuli­
cuta­ne­ous T-cell lym­phoma: mul­ti­na­tional phase II-III trial results. J Clin zumab, a defucosylated anti-CCR4 anti­body, in pre­vi­ously treated patients
Oncol. 2001;19(9):2456-2471. with cuta­ne­ous T-cell lym­phoma. Blood. 2015;125(12):1883-1889.
23. Zackheim HS, Epstein EH Jr. Low-dose meth­o­trex­ate for the Sézary syn­ 45. Beygi S, Fernandez-Pol S, Duran G, et al. Pembrolizumab in myco­sis fungoi­
drome. J Am Acad Dermatol. 1989;21(suppl 4, pt 1):757-762. des with PD-L1 struc­tural var­i­ants. Blood Adv. 2021;5(3):771-774.
24. Quaglino P, Knobler R, Fierro MT, et al. Extracorporeal photopheresis for 46. Houlahan KE, Curtis C. A tumor “per­son­al­ity” test to guide ther­a­peu­tic deci­
the treat­ment of erythrodermic cuta­ne­ous T-cell lym­phoma: a sin­gle cen­ sion mak­ing. Cancer Cell. 2021;39(6):747-749.
ter clin­i­cal expe­ri­ence with long-term fol­low-up data and a brief over­view 47. Kawai H, Ando K, Maruyama D, et al. Phase II study of E7777 in Jap­a­nese
of the lit­er­a­ture. Int J Dermatol. 2013;52(11):1308-1318. patients with relapsed/refrac­ tory periph­eral and cuta­ne­ous T-cell lym­
25. Bagot M, Porcu P, Marie-Cardine A, et  al. IPH4102, a first-in-class anti- phoma. Cancer Sci. 2021;112(6):2426-2435.
KIR3DL2 mono­clo­nal anti­body, in patients with relapsed or refrac­tory cuta­ 48. Folkes AS, Feng M, Zain JM, Abdulla F, Rosen ST, Querfeld C. Targeting CD47
ne­ous T-cell lym­phoma: an inter­na­tional, first-in-human, open-label, phase as a can­cer ther­a­peu­tic strat­egy: the cuta­ne­ous T-cell lym­phoma expe­ri­
1 trial. Lancet Oncol. 2019;20(8):1160-1170. ence. Curr Opin Oncol. 2018;30(5):332-337.
26. Corbin ZA, Nguyen-Lin A, Li S, et al. Characterization of the periph­eral neu­ 49. Johnson LDS, Banerjee S, Kruglov O, et al. Targeting CD47 in Sezary syn­
rop­a­thy asso­ci­ated with brentuximab vedotin treat­ment of myco­sis fun­ drome with SIRPalphaFc. Blood Adv. 2019;3(7):1145-1153.
goides and Sézary syn­drome. J Neurooncol. 2017;132(3):439-446. 50. Gomez-Arteaga A, Margolskee E, Wei MT, van Besien K, Inghirami G, Hor­
27. Khan N, Noor S, Geller S, et al. A phase II trial of reduced dose brentuximab witz S. Combined use of tofacitinib (pan-JAK inhib­i­tor) and ruxolitinib (a
vedotin for cuta­ne­ous T-cell lym­phoma. Paper presented at: International JAK1/2 inhib­ i­
tor) for refrac­ tory T-cell prolymphocytic leu­ ke­mia (T-PLL)
Conference on Malignant Lymphoma; June 2021; Lugano, Switzerland. with a JAK3 muta­tion. Leuk Lymphoma. 2019;60(7):1626-1631.
28. Bagot M, Dalle S, Sokol L, et  al. Long-term clin­i­cal ben­e­fit to anti-CCR4 51. Sakamoto Y, Ishida T, Masaki A, et  al. CCR4 muta­tions asso­ci­ated with
mogamulizumab: results from the phase 3 MAVORIC study in pre­vi­ously supe­rior out­come of adult T-cell leu­ke­mia/lym­phoma under mogamuli­
treated cuta­ne­ous T-cell lym­phoma. Blood. 2018;132(suppl 1):2901. zumab treat­ment. Blood. 2018;132(7):758-761.
29. Musiek A, Whittaker S, Horwitz SM, et al. Characterization and out­comes in 52. Ungewickell A, Bhaduri A, Rios E, et al. Genomic anal­y­sis of myco­sis fun­
patients with mogamulizumab-asso­ci­ated skin reac­tions in the MAVORIC goides and Sézary syn­drome identifies recur­rent alter­ations in TNFR2. Nat
trial. Blood. 2020;136(suppl 1):23-24. Genet. 2015;47(9):1056-1060.
30. Hirotsu KE, Neal TM, Khodadoust MS, et  al. Clinical char­ac­ter­iza­tion of 53. Lesokhin AM, Ansell SM, Armand P, et al. Nivolumab in patients with relapsed
mogamulizumab-asso­ci­ated rash dur­ing treat­ment of myco­sis fungoides or refrac­tory hema­to­logic malig­nancy: pre­lim­i­nary results of a phase
or Sézary syn­drome. JAMA Dermatol. 2021;157(6):700-707. Ib study. J Clin Oncol. 2016;34(23):2698-2704.
31. Martinez-Escala ME, Kuzel TM, Kaplan JB, et  al. Durable responses with 54. Wu PA, Kim YH, Lavori PW, Hoppe RT, Stockerl-Goldstein KE. A meta-
main­te­nance dose-spar­ing reg­i­mens of romidepsin in cuta­ne­ous T-cell anal­y­sis of patients receiv­ing allo­ge­neic or autol­o­gous hema­to­poi­etic
lym­phoma. JAMA Oncol. 2016;2(6):790-793. stem cell trans­plant in myco­sis fungoides and Sézary syn­drome. Biol Blood
32. Arulogun SO, Prince HM, Ng J, et al. Long-term out­comes of patients with Marrow Transplant. 2009;15(8):982-990.
advanced-stage cuta­ne­ous T-cell lym­phoma and large cell trans­for­ma­tion. 55. Hosing C, Bassett R, Dabaja B, et al. Allogeneic stem-cell trans­plan­ta­tion in
Blood. 2008;112(8):3082-3087. patients with cuta­ne­ous lym­phoma: updated results from a sin­gle insti­tu­
33. Scarisbrick JJ, Prince HM, Vermeer MH, et al. Cutaneous Lymphoma Interna­ tion. Ann Oncol. 2015;26(12):2490-2495.
tional Consortium study of out­come in advanced stages of myco­sis fungoi­ 56. Duarte RF, Boumendil A, Onida F, et al. Long-term out­come of allo­ge­neic
des and Sézary syn­drome: effect of spe­cific prog­nos­tic mark­ers on sur­vival hema­to­poi­etic cell trans­plan­ta­tion for patients with myco­sis fungoides
Prakash Singh Shekhawat
Personalizing treat­ment selec­tion in MF/SS  |  311
and Sézary syn­drome: a Euro­pean Society for Blood and Marrow Trans­ 62. Mehta-Shah N, Kommalapati A, Teja S, et  al. Successful treat­ment of
plantation Lymphoma Working Party extended anal­ y­
sis. J Clin Oncol. mature T-cell lym­phoma with allo­ge­neic stem cell trans­plan­ta­tion: the
2014;32(29):3347-3348. larg­est mul­ti­cen­ter ret­ro­spec­tive anal­y­sis. Paper presented at: 62nd
57. Paralkar VR, Nasta SD, Morrissey K, et  al. Allogeneic hema­to­poi­etic SCT Amer­i­can Society of Hematology Annual Meeting; 5-8 Decem­ber 2020;
for pri­mary cuta­ne­ous T cell lym­pho­mas. Bone Marrow Transplant. vir­tual meet­ing.
2012;47(7):940-945. 63. Olsen EA, Kim YH, Kuzel TM, et al. Phase IIb mul­ti­cen­ter trial of vorinostat
58. de Masson A, Beylot-Barry M, Bouaziz JD, et al; French Study Group on in patients with per­sis­tent, pro­gres­sive, or treat­ment refrac­tory cuta­ne­ous
Cutaneous Lymphomas and Société Française de Greffe de Moëlle et T-cell lym­phoma. J Clin Oncol. 2007;25(21):3109-3115.
Thérapie Cellulaire. Allogeneic stem cell trans­ plan­ta­
tion for advanced 64. Horwitz SM, Koch R, Porcu P, et al. Activity of the PI3K-δ,γ inhib­i­tor duv­
cuta­ne­ous T-cell lym­pho­mas: a study from the French Society of Bone Mar­ elisib in a phase 1 trial and pre­clin­i­cal mod­els of T-cell lym­phoma. Blood.
row Transplantation and French Study Group on Cutaneous Lymphomas. 2018;131(8):888-898.
Haematologica. 2014;99(3):527-534. 65. Querfeld C, Rosen ST, Guitart J, et al. Results of an open-label mul­ti­cen­ter
59. Lechowicz MJ, Laz­a­rus HM, Carreras J, et al. Allogeneic hema­to­poi­etic cell phase 2 trial of lenalidomide monotherapy in refrac­tory myco­sis fungoides
trans­plan­ta­tion for myco­sis fungoides and Sezary syn­drome. Bone Marrow and Sezary syn­drome. Blood. 2014;123(8):1159-1166.
Transplant. 2014;49(11):1360-1365.
60. Isufi I, Seropian S, Gowda L, et al. Outcomes for allo­ge­neic stem cell trans­
plan­ta­tion in refrac­tory myco­sis fungoides and pri­mary cuta­ne­ous gamma

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/303/1852064/303kim.pdf by guest on 13 December 2021


delta T cell lym­pho­mas. Leuk Lymphoma. 2020;61(12):2955-2961.
61. Weng WK, Arai S, Rezvani A, et al. Nonmyeloablative allo­ge­neic trans­plan­ta­
tion achieves clin­i­cal and molec­u­lar remis­sion in cuta­ne­ous T-cell lym­phoma. © 2021 by The Amer­i­can Society of Hematology
Blood Adv. 2020;4(18):4474-4482. DOI 10.1182/hema­tol­ogy.2021000263

312  |  Hematology 2021  |  ASH Education Program


INDOLENT LYMPHOMAS

Follicular lymphoma: is there an optimal


way to define risk?

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/313/1851762/313casulo.pdf by guest on 13 December 2021


Carla Casulo
University of Rochester, Rochester, NY

Follicular lymphoma (FL) has a long natural history and typically indolent behavior. In the present era, there are a plethora
of prognostic factors combining clinical, biological, and genetic data to determine patient prognosis and help develop
treatment strategies over the course of a patient’s lifetime. The rapid pace of tumor-specific and clinical advances in FL
has created a challenge in the prioritization and implementation of these factors into clinical practice. Developing a com-
prehensive understanding of existing prognostic markers in FL will help select optimal ways of utilization in the clinical
setting and investigate opportunities to define and intervene upon risk at FL diagnosis and disease recurrence.

LEARNING OBJECTIVES
• Describe patient- and tumor-specific factors associated with risk of disease progression, histologic transforma-
tion, and premature death from FL
• Review established prognostic models in FL and their strengths and weaknesses when applied in clinical practice
• Discuss approaches to incorporate the clinical and mutational data into novel prognostic tools at different points
in the patient’s lifetime

Introduction
In American and European adults, follicular lymphoma (FL) CLINICAL CASE
is the second most common non-Hodgkin lymphoma.1 A 52-year-old man presented with painless cervical lymph-
The long natural history and typically indolent behavior adenopathy noticed while shaving. He has a history of
of FL provide opportunities to identify prognostic infor- hypertension and hyperlipidemia. He recently noted being
mation combining clinical, biological, and genetic data to significantly fatigued but denied weight loss or night
determine patient prognosis and help define treatment sweats. Laboratory tests revealed hemoglobin of 10.2g/dL
strategies patients will need over their lifetime. However, (reference range, male: 13.7-17.5g/dL), lactate dehydroge-
the rapid pace of scientific discoveries in FL encompass- nase (LDH) of 270U/dL (upper limit of normal, 225U/dL),
ing both tumor-specific and clinical advances has made and an increased β2 microglobulin level (4.2mg/L; upper
it challenging to prioritize the most clinically relevant limit of normal, 3.0mg/L). No other abnormalities were
and essential prognostic parameters. Importantly, some noted. An excisional lymph node biopsy specimen showed
of the prognostic information currently available often grade 1 to 2 FL comprising small mature centrocytes in a
yields redundant or conflicting results, thereby limiting well-preserved follicular pattern that stained positive for
implementation into practice (Figure 1). A comprehensive CD10, CD20, and BCL2. Flow cytometry detected a clonal
overview of all prognostic markers to assess risk in FL is population of B cells. Fluorescent in situ hybridization
beyond the scope of this review, as it would be inclusive detected t(14;18). Bone marrow biopsy specimen revealed
of genetic, genomic, clinical, and diagnostic imaging- small paratrabecular lymphoid aggregates encompassing
based parameters.2-7 However, through a discussion of a 10% of the bone marrow space. Positron emission tomog-
patient case, this review examines some of the prognostic raphy (PET) showed hypermetabolic lymphadenopathy
factors along the FL continuum, assesses optimal ways of in the bilateral cervical chain, right axillae, retroperito-
utilization in clinical practice, and explores opportunities neum, and right inguinal area with standard uptake value
to define risk in the relapsed setting. of 7 to 11. Lymphadenopathy ranged from 3.2 to 4.7 cm.

Prakash Singh Shekhawat


Defining risk in FL | 313
FLIPI FLIPI2

Variables Variables
Nodal sites Age >60
Elevated LDH Bone marrow involvement
Age >60 Hemoglobin <12 g/dL
Stage III/IV Greatest LN diameter >6 cm
Hemoglobin ≤12 g/dL Serum β2 microglobulin > upper limit of normal

Score Score

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/313/1851762/313casulo.pdf by guest on 13 December 2021


0-1 Low risk 0-1 Low risk
2 Intermediate risk 2 Intermediate risk
3-5 High risk 3-5 High risk

Survival Survival
0-1 5-year OS 91% 0-1 5-year PFS 80%
2 5-year OS 78% 2 5-year PFS 52%
3-5 5-year OS 53% 3-5 5-year PFS 19%

FLEX
PRIMA-PI

Variables Variables
β2 microglobulin >3 mg/L Male sex, SPD in highest quartile,
Bone marrow involvement grade 3A, >2 extranodal sites, ECOG
PS >1, hemoglobin <12 g/dL, β2
microglobulin > normal, peripheral
Score blood natural killer cell count <100/µL,
Low β2 microglobulin <3 mg/L, increased LDH
negative bone marrow
Intermediate β2 microglobulin ≤3 mg/L,
+ bone marrow
High β2 microglobulin >3 mg/L Score
0-2 Low risk
3-9 High risk
Survival
Low 5-year PFS 69%
Survival
Intermediate 5-year PFS 55%
Low 3-year PFS 86%
High 5-year PFS 37%
High 3-year PFS 68%

Figure 1. Clinical prognostic tools and risk scores. ECOG, Eastern Cooperative Oncology Group; LN, lymph node; PS, performance
status.

314  |  Hematology 2021  |  ASH Education Program


The patient ini­ti­ated ther­apy with bendamustine and rituximab An over­ view of the cur­ rently avail­­
able clin­

cal and tumor-
and received 6 cycles. based mark­ers asso­ci­ated with high risk in FL reveals a mul­ti­tude
of var­i­ables (Table 1). Based on this patient’s male sex, pres­ence
of >4 nodal sites, high tumor bur­den, ele­vated LDH, advanced
At pres­ent, there is no agreed-on, uni­form def­i­ni­tion of risk stage, and low hemo­glo­bin, the patient has sev­eral fac­tors asso­
in FL. However, the sur­vival of FL span­ning over 2 decades and ci­ated with higher prob­a­bil­ity of poor out­come. Accordingly,
mul­ti­ple avail­­able novel ther­a­peu­tics with high response rates the patient’s Follicular Lymphoma International Prognosis Index
and long peri­ods of remis­sion pro­vide a rea­son­able bench­mark (FLIPI) score is 4, high risk, and based on this assess­ment, the
of expected patient out­comes at diag­no­sis. The con­cept of high- patient’s over­all sur­vival (OS) is esti­mated to be 70% at 10 years
risk FL has emerged from data dem­on­strat­ing increased prob­a­bil­ for patients in the rituximab era.9,15 The FLIPI incor­po­rates 5 clin­i­
ity of pre­ma­ture death for patients refrac­tory to alkylator-based cal var­i­ables (nodal sites, ele­vated LDH, age >60 years, advanced
ther­apy or anti-CD20 mono­clo­nal antibodies in the first-line set­ stage, and hemo­glo­bin <12 g/dL) into a mul­ti­var­i­ate model to
ting, as well as patients with early dis­ease recur­rence fol­low­ing pre­dict OS. It was constructed to improve sen­si­tiv­ity com­pared

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/313/1851762/313casulo.pdf by guest on 13 December 2021


front­line ther­apy and early his­to­logic trans­for­ma­tion.8-10 For the with the International Prognostic Index for indo­lent lym­phoma
pur­poses of this review, we con­sider patients at increased risk of and based on ret­ro­spec­tive data from patients diag­nosed in the
these adverse out­comes as high risk. 1980s and 1990s treated het­ero­ge­neously at var­i­ous cen­ters.

Table 1. Variables asso­ci­ated with “high-risk FL” (increased risk of death, early recur­rence, trans­for­ma­tion)

Variable Outcome Reference


Clinical/patient based
  Grade 3b vs grades 1-3a Inferior OS Wahlin et al,11 Mercadal et al12
  Increased β2 microglobulin from nor­mal Inferior PFS and OS Press et al,13 Bachy et al14
  Large nodal mass (>7 cm) Inferior PFS Solal-Céligny et al,15 Nooka et al,16 Buske et al17
  Greater than 4 nodal sites Inferior PFS and OS Solal-Céligny et al15
  Bone mar­row involve­ment Inferior PFS and OS Bachy et al14
  Low hemo­glo­bin (under 12 g/dL) Inferior PFS and OS Solal-Céligny et al,15 Nooka et al16
  Advanced stage Inferior PFS and OS Brice et al,18 Buske et al17
  High FL tumor bur­den based on GELF cri­te­ria Inferior PFS and OS Brice et al,18 Buske et al17
  Low lym­pho­cyte to mono­cyte ratio Increased risk of trans­for­ma­tion Gao et al,19 Mohsen et al,20 Mozas et al21
Imaging based
  High base­line SUV max of PET scan Inferior OS and increased risk of early Mir et al,22 Barrington and Meignan23
dis­ease pro­gres­sion (mixed data)
  End of ther­apy PET pos­i­tive (Deauville 4-5) Inferior PFS and OS Trotman et al3
  High total met­a­bolic tumor vol­ume Inferior PFS and OS (mixed data) Barrington and Meignan,23 Skander 2019,50 Meignan et al4
Tumor based
  23-gene GEP sig­na­ture score Inferior PFS Huet et al5
  Genomic alter­ations (gain of chro­mo­some 2p; Inferior PFS Qu et al24
dele­tion 17p, subclonal TP53 muta­tions)
  Low intratumoral immune infil­tra­tion Increased risk of early pro­gres­sion Tobin et al25
  High expres­sion of genes from tumor asso­ci­ Inferior PFS and OS Dave et al,26 Kridel et al27
ated mac­ro­phages
  Increased ctDNA at diag­no­sis and at Inferior PFS and OS Delfau-Larue et al,6 Zohren et al,28 Sarkozy et al29
com­ple­tion of ther­apy
  Select muta­tions in sev­eral genes, includ­ing Enriched in tumors of early progressed Kridel et al30
TP53, SOCS1, B2M, and MYD88 patients
Treatment based
  Disease recur­rence within 24 months of diag­ Inferior OS Casulo et al,31 Jurinovic et al,32 Maurer et al33
no­sis or ther­apy
  Histologic trans­for­ma­tion 18-24 months after Inferior OS Link et al,8 Wagner-Johnson et al,34 Sarkozy et al35
diag­no­sis or ther­apy
  Multiply relapsed dis­ease Inferior PFS Batlevi et al,9 Link et al,36 Rivas-Delgado et al10
  Refractory dis­ease Inferior PFS Batlevi et al,9 Link et al,36 Rivas-Delgado et al10
Prakash Singh Shekhawat
GELF, Groupe d’Etude des Lymphomas Folliculaires; SUV, standard uptake value.

Defining risk in FL  |  315


The FLIPI yields 3 risk clas­si­fi­ca­tions: low (0-1 fac­tor), inter­me­di­ cyclophosphamide, doxorubicin, vincristine, and prednisone or
ate (2-3 fac­tors), and high (4-5 fac­tors) with a dis­tri­bu­tion of low, rituximab, cyclophosphamide, vincristine, and prednisone. More-
inter­me­di­ate, and high risk in 36%, 37%, and 27% of patients, over, for patients with FL treated in the GALLLIUM study, bone
respec­tively. Despite its devel­op­ment in the prerituximab era, the mar­row biopsy was not pre­dic­tive of out­come.38 As with the FLIPI
FLIPI’s prog­nos­tic legit­i­macy has been val­i­dated numer­ous times.16,17 and FLIPI2, the PRIMA-PI model does not inform selec­tion of ther­
This sur­vival esti­mate may seem dis­pro­por­tion­ately low when apy for this patient or mod­i­fi­ca­tion of treat­ment based on his
con­sid­er­ing recent stud­ies eval­u­at­ing prog­no­sis and cause of risk score.
death in FL. An anal­y­sis by Sarkozy et al51 noted that 10-year
OS was approx­i­ma­tely 75% in patients with newly diag­nosed Application of these mod­els in the
FL treated in the rituximab era, but a high-risk FLIPI score did bendamustine/obinutuzumab era
increase the cumu­ la­tive inci­dence of lym­ phoma-related mor­ The FLIPI, FLIPI-2, and PRIMA-PI were devel­ oped in patients
tal­ity (inci­dence of lym­phoma-related mor­tal­ity at 10 years was treated with alkylators but did not include the more fre­quently
4.0% for low-risk FLIPI, 10.0% for inter­me­di­ate-risk FLIPI, and 27% used bendamustine. Although these indi­ces have been val­i­dated

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/313/1851762/313casulo.pdf by guest on 13 December 2021


for high-risk FLIPI, P < .001). The patient’s high-risk FLIPI score and ret­ro­
spec­tively in small series, only the Follicular Lymphoma
high tumor bur­den there­fore inform us of a like­li­hood of adverse Evaluation Index (FLEX) model was pre­dic­tive of out­comes in
out­come and the advan­tages of treat­ment ini­ti­a­tion. patients receiv­ing bendamustine and the novel anti-CD20 anti­
body obinutuzumb.39 This model includes 9 var­i­ables: male sex,
What are other clin­i­cal prog­nos­tic mod­els avail­­able sum of prod­ucts (SPD) in highest quar­tile, grade 3A, >2 extran-
in the rituximab era? odal sites, Eastern Cooperative Oncology Group per­for­mance
As rituximab has dras­ ti­
cally improved patient sur­ vival in the sta­tus >1, hemo­glo­bin <12 g/dL, β2 microglobulin higher than
mod­ern era, it is expected and observed that prog­nos­tic indi­ces nor­mal, periph­eral blood nat­u­ral killer cell count <100/µL, and
have less sen­si­tiv­ity and spec­i­fic­ity for out­comes. The FLIPI2 was increased LDH. A high score (3-9 fac­tors) was pre­dic­tive of poor
devel­oped using 1093 patients with newly diag­nosed FL in need PFS (3-year PFS of 86% for low risk vs 68% for high risk), and sec­
of treat­ment, included nearly 70% of patients treated in the ritux- ond­ary end points also showed poor OS and increased risk of
imab era, and was val­i­dated in an inde­pen­dent cohort of Ital­ian early pro­gres­sion. Given the lack of read­ily avail­­able var­i­ables
patients.37 The FLIPI2 included 5 covariates based on their rel­a­ tested in this model, as part of a sen­si­tiv­ity anal­y­sis, the authors
tive fre­quency and included ele­vated β2 microglobulin, lymph also eval­u­ated miss­ing data. They found that the sur­vival pro­file
node diam­ e­
ter lon­ ger than 6  cm, bone mar­ row involve­ ment, of patients with miss­ing data appeared sim­i­lar to that of com­
low hemo­glo­bin, and age greater than 60 years. This model plete cases, with a PFS haz­ard ratio for miss­ing vs com­plete FLEX
was designed to pre­dict pro­gres­sion-free sur­vival (PFS) and, if data of 0.89 (95% CI, 0.68-1.17; P = .40). In this patient’s case, his
applied to this patient, would sim­i­larly yield a high-risk score of score would be 4 (high risk), based on sex, low hemo­glo­bin, and
3 and pro­vide a 3-year PFS esti­mate of approx­i­ma­tely 39% in the increased LDH and β2 microglobulin. Other param­e­ters included
rituximab era. The validity of the FLIPI 2 was sim­i­larly con­firmed in the FLEX were not avail­­able for eval­ua ­ ­tion. This pre­dicts a
exter­nally in a cohort of 1135 patients from the PRIMA study who 3-year PFS of 86%. The clear dis­ad­van­tage of this prog­nos­tic tool
received che­mo­ther­apy with or with­out main­te­nance rituximab. is the rou­tine lack of sev­eral of the param­e­ters cal­cu­lated. In the
The Primary RItuximab and MAintenance (PRIMA) prog­ nos­ FLEX man­u­script, SPD was used as a sur­ro­gate for tumor bur­den.
tic index (PRIMA-PI) esti­mated disparities in sur­vival based on During model devel­op­ment, SPD had among the highest haz­ard
the num­ber of fac­tors pres­ent in patients treated in the PRIMA ratios for PFS. As SPD requires radio­logic cal­cu­la­tion, bulky dis­
study.14 Using only 2 param­e­ters (bone mar­row involve­ment and ease may be an alter­na­tive.
increased β2 microglobulin), the pri­mary end point was to pre­ Although a Cox pro­por­tional haz­ards anal­y­sis of PFS showed
dict PFS. In this case, the patient’s bone mar­row involve­ment that FLIPI and PRIMA-PI had a lower prog­ nos­tic value across
and increased β2 microglobulin would also result in a high-risk treat­ment reg­i­mens than the FLEX, it is not imme­di­ately appar­
score but pre­dict a dif­fer­ent 5-year PFS of 69% and a 38% risk of ent how this score will become rou­tinely used in clin­i­cal prac­
expe­ri­enc­ing early ther­apy fail­ure (Table 2). Of impor­tance, the tice, and fur­ther study is desir­able in real-world set­tings and in
PRIMA-PI was only pre­dic­tive in patients treated with rituximab, the con­text of non-chemoimmunotherapeutic strat­e­gies such as
lenalidomide and rituximab or rituximab monotherapy.

Table 2. Case patient’s risk assess­ment based on avail­­able Tumor-based, bio­logic, and genetic var­i­ables
clin­i­cal cal­cu­la­tors/data FL cells reside within a rich tumor micro­en­vi­ron­ment (TME) envel-
oped by both non­ma­lig­nant cells and crit­i­cal immune com­po­
Clinical Risk Calculator/ nents that pro­vide sym­bi­otic influ­ences on the FL tumor cell itself
Prognostic Data Survival Outcomes
and back onto the TME. This con­tri­bu­tion to patient out­come
FLIPI score high risk 10-year OS of 70% was first established by Dave et al,26 who dem­on­strated that high
FLIPI2 score high risk 3-year PFS of 39% expres­ sion of genes from FL tumor-asso­ ci­
ated mac­ ro­
phages
was indic­a­tive of poor out­comes. Yet, more con­tem­po­rary stud­ies
PRIMA-PI high risk 5-year PFS of 37%, 38% risk of
hav­ing treat­ment fail­ure within show conflicting results when rituximab is incor­po­rated into treat­
24 months ment reg­i­mens, as dem­on­strated by Kridel et al,27 who showed
FLEX score high risk 3-year PFS of 68%
that high num­bers of CD163+ mac­ro­phages were inde­pen­dent
pre­dic­tors of lon­ger sur­vival in patients receiv­ing anthracycline-
Early relapse 5-year OS 50%
based reg­i­mens. In other work, the mag­ni­tude of intratumoral

316  |  Hematology 2021  |  ASH Education Program


immune infil­tra­tion was found to be an impor­tantcom­po­nent of com­pre­hen­sive col­lec­tion of recur­rent gene muta­tions as well as
poor out­comes, in par­tic­u­lar early treat­ment fail­ure. A study from clin­i­cal risk fac­tors in patients with FL. In the mul­ti­var­i­ate model,
the Princess Alexandra Hospital eval­ u­
ated whether there was high-risk FLIPI score and Eastern Cooperative Oncology Group
an immune infil­tra­tion pro­file in FL asso­ci­ated with early dis­ease >1 car­ried sig­nif­i­cant weight, but EZH2 and ARID1A muta­tions
pro­gres­sion.25 iden­ti­fied a favor­able risk pop­u­la­tion. The m7-FLIPI improved risk
Tobin et al25 performed targeted gene sequenc­ ing using strat­i­fi­ca­tion by reclassifying patients pre­vi­ously clas­si­fied as
NanoString tech­nol­ogy from par­af­fin-embed­ded tis­sue and mul­ high-risk FLIPI into the low-risk m7-FLIPI group, which was a bet­
ti­spec­tral immu­no­flu­o­res­cence on a tis­sue microarray that was ter reflec­tion of their actual FL risk. However in a val­i­da­tion study
applied to 2 groups: a dis­cov­ery cohort of 132 patients from the of patients treated with bendamustine or obinutuzumab in the
Princess Alexandra Hospital with early and advanced stage FL GALLIUM study, although the m7-FLIPI was prog­nos­tic in patients
who received either che­mo­ther­apy or obser­va­tion and 2 inde­ receiv­ing CHOP/CVP and still outperformed the FLIPI, it lost its
pen­dent val­i­da­tion cohorts of 198 patients with advanced stage prog­ nos­ tic sig­
nif­i­
cance in patients receiv­ ing bendamustine or
dis­ease treated with R-CHOP and R-CVP from the Ger­man Low obinutuzumab.43 It is hypoth­e­sized that this inter­ac­tion may be

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/313/1851762/313casulo.pdf by guest on 13 December 2021


Grade Lymphoma Study Group and the Brit­ish Colum­bia Can- driven by EZH2 muta­tions, but this remains to be fur­ther val­i­
cer Agency. They also performed T-cell rep­er­toire anal­y­sis, flow dated.41 In nonchemotherapy-containing reg­i­mens, m7-FLIPI was
cytom­e­try, immu­no­flu­o­res­cence, and next-gen­er­a­tion sequenc­ also not prognostic of out­comes, rais­ing the notion that che­mo­
ing. They showed that immune mol­e­cules from T cells, mac­ro­ ther­apy depen­dence influ­ences the high-risk sig­na­ture score.44
phages, or immune check­points were clus­tered into either high To eval­u­ate the spe­cific end point of POD24, Jurinovic et al32
expres­ sion or low expres­ sion. Low lev­ els of immune mark­ ers reexamined the m7-FLIPI. Using the same patient cohorts and
iden­ti­fied patients enriched for early pro­gres­sion, and PDL2 was muta­tional data, a POD24-spe­cific prog­nos­tic model was devel­
the marker with highest accu­racy to dis­tin­guish groups of low or oped, described as the POD24-PI. It included FLIPI but was
high immune infil­tra­tion. This was val­i­dated in group of uni­formly refined to include muta­ tional sta­tus of only 3 genes (EP300,
treated patients from the Brit­ish Colum­bia Cancer Agency and the FOXO1, and EZH2). Compared with the m7-FLIPI, the new
Ger­man Low Grade Lymphoma Study Group, which showed that POD24-PI model had greater sen­si­tiv­ity to pre­dict POD24 but
tumors with a low immune infil­tra­tion had higher early dis­ease- was not supe­rior to other approaches due to lower spec­i­fic­ity.
related (progression of disease within 24 months of diagnosis/ The Bio-clinical FLIPI (Bio-FLIPI) sim­i­larly inte­grated the FLIPI with
treatment of FL) events. Immune infil­tra­tion by intratumoral T cells genes impli­cated in the TME and iden­ti­fied sev­eral asso­ci­ated
was quan­ti­fied in con­junc­tion with fluorodeoxyglucose-PET in a with early treat­ment fail­ure. Nonetheless, only lack of intrafollic-
study by Nath et al.40 Lymph nodes from patients with high total ular CD4 expres­sion was pre­dic­tive of treat­ment fail­ure.45 As in
met­a­bolic tumor vol­ume had an inverse asso­ci­a­tion with num­bers other exam­ples, assess­ment of muta­tional sta­tus is not stan­dard­
of intratumoral T cells but increased malig­nant B-cell infil­tra­tion. ized and remains part of a research approach.
This sug­gests dis­crep­ant pre­dic­tive util­ity of FDG-PET in patients
with FL depending on the extent of T-cell deple­tion caused by the
Dynamic risk assess­ment through­out the patient’s his­tory
type of chemoimmunotherapy that is admin­is­tered.
Gene expres­sion pro­fil­ing (GEP) stud­ies by Huet et al5 defined
a gene sig­ na­
ture cor­ re­lat­
ing with adverse PFS in FL. They CLINICAL CASE (Con­tin­ued)
defined a model based on the expres­sion of 23 genes char­ac­
ter­is­tic of B-cell centroblasts that was prog­nos­tic inde­pen­dent The patient com­pleted 6 cycles of treat­ment with­ out com­
of the FLIPI score. In a mul­ti­var­i­ate anal­y­sis, those iden­ti­fied as pli­ca­tions. His PET response at the end of ther­apy was com­
high risk by the 23-gene pre­dic­tor had a 5-year PFS of 26% com­ pat­i­ble with a com­plete met­a­bolic response, an achieve­ment
pared with 73% in patients with a low-risk sig­na­ture. This was asso­ci­ated with favor­able out­comes.3,46,47 As part of an ongo­ing
con­firmed in 3 inde­pen­dent val­i­da­tion cohorts. The per­for­mance clin­i­cal trial, eval­u­a­tion of min­i­mal resid­ual dis­ease (MRD) was
of the 23-gene sig­na­ture was eval­u­ated in GALLIUM, in which performed at diag­no­sis, end of ther­apy, and every 6 months for
no prog­nos­tic effect was observed for any of the gene sig­na­ 2 years to assess cor­re­la­tion with dis­ease pro­gres­sion.
tures based on anti­body treat­ment arm. However, che­mo­ther­
apy choice did have a sig­nif­i­cant inter­ac­tion between PFS and
the 23-gene sig­na­ture, in which patients with a high-risk score Several stud­ies have reported the asso­ci­a­tion of MRD assess­
receiv­ing CHOP/CVP had unfa­vor­able out­comes com­pared with ment as prog­nos­tic of out­come in FL. This can be achieved by
those treated with bendamustine, suggesting che­ mo­ther­ apy assess­ment of cir­cu­lat­ing pre­treat­ment cell-free DNA frag­ments
depen­dence of the high-risk sig­na­ture score.41 from apo­pto­tic tumor cells (cir­cu­lat­ing tumor DNA [ctDNA]) and by
At pres­ent, the assess­ment of these immune mark­ers and the assess­ment of BCL2 immunoglobulin heavy chain lev­els quan­ti­fied
capa­bil­ity to per­form GEP stud­ies are not stan­dard­ized at diag­ by poly­mer­ase chain reac­tion (PCR). Identification of the t(14;18)
no­sis, and some tech­niques are not read­ily avail­­able and hence trans­lo­ca­tion can be qual­i­ta­tively and ­quan­ti­ta­tively ­mea­sured in
remain part of a research approach. the bone mar­row and periph­eral blood but is l­im­ited to patients
har­bor­ing the t(14;18) and can also be found in the blood of healthy
Combining clin­i­cal and genetic mod­els patients. Other tech­nol­ogy using next-gen­er­a­tion sequenc­ing
The com­bi­na­tion of key epi­ge­netic muta­tions with a high-risk meth­ods can detect a large spec­trum of genetic alter­ations, such
FLIPI score and patient per­for­mance sta­tus was accom­plished by as the Cancer Personalized Profiling by Deep Sequencing method
Pastore et al42 to yield the m7-FLIPI. Although prior stud­ies empha­ and quan­ti­ta­tion of ctDNA encoding the clonalyl rearranged V(D)
sized the rel­e­vance of sin­gle gene alter­ations, m7-FLIPI included a J Ig recep­tor gene sequence of FL cells.29,48
Prakash Singh Shekhawat
Defining risk in FL  |  317
A pro­spec­tive Ger­
man study ana­ lyzed BCL2 IGH lev­ els Off-label drug use
by PCR in pre- and posttreatment periph­ eral blood in 173 Carla Casulo: nothing to disclose.
patients with FL, find­ing an adverse impact of high pre­treat­
ment lev­els on sub­se­quent PFS.28 As expected, high lev­els of Correspondence
MRD at the con­clu­sion of ther­apy also had poor out­comes. Carla Casulo, University of Rochester, 601 Elmwood Ave, Roches-
The achieve­ment of neg­a­tive or low ctDNA or MRD sim­i­larly ter, NY 14642; e-mail: carla_casulo@urmc​­.rochester​­.edu.
enhances prog­nos­tic infor­ma­tion on patient out­come. Studies
in other lym­pho­mas have dem­on­strated that dynamic ctDNA References
lev­els dur­ing and after ther­apy not only can pre­dict clin­i­cal 1. Teras LR, DeSantis CE, Cerhan JR, Morton LM, Jemal A, Flowers CR. 2016 US
out­comes but may also synergize with other prog­nos­tic fac­ lym­phoid malig­nancy sta­tis­tics by World Health Organization sub­types.
CA Cancer J Clin. 2016;66(6):443-459.
tors such as total met­a­bolic tumor vol­ume.6 The best use of
2. Cottereau AS, Versari A, Luminari S, et al. Prognostic model for high-tumor-
these molec­u­lar assess­ments of response will be dur­ing or bur­den fol­lic­u­lar lym­phoma inte­grat­ing base­line and end-induc­tion PET: a
fol­low­ing treat­ment to facil­i­tate early inter­ven­tion for recur­ LYSA/FIL study. Blood. 2018;131(22):2449-2453.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/313/1851762/313casulo.pdf by guest on 13 December 2021


rence, assess changes in muta­tional pro­file, and be used to 3. Trotman J, Luminari S, Boussetta S, et al. Prognostic value of PET-CT after
guide next ther­apy. first-line ther­apy in patients with fol­lic­u­lar lym­phoma: a pooled anal­y­sis
of cen­ tral scan review in three multicentre stud­ ies. Lancet Haematol.
2014;1(1):e17-e27.
4. Meignan M, Cottereau AS, Versari A, et al. Baseline met­a­bolic tumor vol­ume
pre­dicts out­come in high-tumor-bur­den fol­lic­u­lar lym­phoma: a pooled
CLINICAL CASE (Con­t in­u ed)
anal­y­sis of three mul­ti­cen­ter stud­ies. J Clin Oncol. 2016;34(30):3618-3626.
Approximately 1 year fol­low­ing ther­apy, the patient expe­ri­enced 5. Huet S, Tesson B, Jais J-P, et al. A gene-expres­sion pro­fil­ing score for pre­
dis­ease relapse with high tumor bur­den, low-grade FL. The dic­tion of out­come in patients with fol­lic­u­lar lym­phoma: a ret­ro­spec­tive
train­ing and val­i­da­tion anal­y­sis in three inter­na­tional cohorts. Lancet
adverse prog­nos­tic impact of early dis­ease recur­rence (within
Oncol. 2018;19(4):549-561.
24 months of diag­no­sis) on sur­vival has been well established.31,49 6. Delfau-Larue M-H, van der Gucht A, Dupuis J, et al. Total met­a­bolic tumor
Few prog­nos­tic indi­ces have been tested for use to pre­dict out­ vol­ume, cir­cu­lat­ing tumor cells, cell-free DNA: dis­tinct prog­nos­tic value in
comes at relapse out­side of FLIPI, and this is an unmet need. He fol­lic­u­lar lym­phoma. Blood Adv. 2018;2(7):807-816.
joined a ran­dom­ized phase 2 clin­ic ­ al study ran­dom­iz­ing patients 7. Barrington S TJ, Sahin D, et al. Baseline PET-derived met­a­bolic tumor vol­
ume met­rics did not pre­dict out­comes in fol­lic­u­lar lym­phoma patients
to obinutuzumab with umbralisib, lenalidomide, or CHOP and treated with first-line immuno che­mo­ther­apy and anti­body main­te­nance
was ran­dom­ized to the obinutuzumab with umbralisib arm in the phase III GALLIUM study. Blood. 2018;132(suppl 1):2882.
(NCT03269669). He expe­ ri­
enced a par­ tial met­
a­bolic response 8. Link BK, Maurer MJ, Nowakowski GS, et al. Rates and out­comes of fol­lic­ul­ar
followed by rapid pro­gres­sion of dis­ease. A repeat biopsy con­ lym­phoma trans­for­ma­tion in the immunochemotherapy era: a report from
the University of Iowa/MayoClinic Specialized Program of Research Excel-
firmed low-grade FL. He began ther­apy with axicabtagene cilo-
lence Molecular Epidemiology Resource. J Clin Oncol. 2013;31(26):3272-
leucel on a clin­i­cal study (NCT03105336), now approved by the US 3278.
Food and Drug Administration for relapsed FL in the United States 9. Batlevi CL, Sha F, Alperovich A, et al. Follicular lym­phoma in the mod­ern
(https:​­/​­/www​­.fda​­.gov​­/drugs​­/resources​­-information​­-approved​ era: sur­ vival, treat­
ment out­ comes, and iden­ ti­
fi­
ca­tion of high-risk sub­
­-drugs​­/fda​­-grants​­-accelerated​­-approval​­-axicabtagene​­-ciloleucel​ groups. Blood Cancer J. 2020;10(7):74.
10. Rivas-Delgado A, Magnano L, Moreno-Velázquez M, et al. Response dura­
­-relapsed​­-or​­-refractory​­-follicular​­-lymphoma). tion and sur­vival shorten after each relapse in patients with fol­lic­u­lar lym­
phoma treated in the rituximab era. Br J Haematol. 2019;184(5):753-759.
11. Wahlin BE, Yri OE, Kimby E, et al. Clinical sig­nif­i­cance of the WHO grades of
Conclusions fol­lic­u­lar lym­phoma in a pop­ul­a­tion-based cohort of 505 patients with long
fol­low-up times. Br J Haematol. 2012;156(2):225-233.
A mul­ti­tude of prog­nos­tic var­i­ables are at our dis­posal to under­
12. Mercadal S, Sancho JM, Climent F, et  al. Long-term out­come com­par­ing
stand and pre­dict sur­vival for FL and to iden­tify patients at high his­to­log­i­cal grades of fol­lic­u­lar lym­phoma patients treated with immu-
risk of poor out­come. However, sev­eral of these tools remain nochemotherapy as first-line ther­apy: a ret­ro­spec­tive anal­y­sis from two
inac­ces­ si­
ble in daily prac­tice and have not been ade­ quately insti­tu­tions. Eur J Haematol. 2020;104(3):198-206.
tested to select ther­apy. Unfortunately, none of the risk mod­els 13. Press OW, Unger JM, Rimsza LM, et  al. A com­par­a­tive anal­y­sis of prog­
nos­tic fac­tor mod­els for fol­lic­ul­ar lym­phoma based on a phase III trial of
described here have been val­i­dated as a tool to select or adapt CHOP-rituximab ver­sus CHOP +131iodine—tositumomab. Clin Cancer Res.
ther­apy for patients with FL. Although under­stand­ing prog­no­sis 2013;19(23):6624-6632.
is impor­tant, ulti­mately the goal of any of these tests should be 14. Bachy E, Maurer MJ, Habermann TM, et al. A sim­pli­fied scor­ing sys­tem in
to guide ther­apy. Studies of risk-adapted ther­apy based on MRD de novo fol­lic­ul­ar lym­phoma treated ini­tially with immunochemotherapy.
Blood. 2018;132(1):49-58.
are ongo­ing in dif­fuse large B-cell lym­phoma and begin­ning in
15. Solal-Céligny P, Roy P, Colombat P, et al. Follicular lym­phoma inter­na­tional
FL (Luminari et al52), but this approach remains inves­ti­ga­tional. prog­nos­tic index. Blood. 2004;104(5):1258-1265.
Importantly, assess­ment of risk for the refrac­tory patient or at 16. Nooka AK, Nabhan C, Zhou X, et al. Examination of the fol­lic­u­lar lym­phoma
relapse is an area of ongo­ing inves­ti­ga­tion and may con­trib­ute inter­na­tional prog­nos­tic index (FLIPI) in the National LymphoCare study
to a more pre­cise abil­ity to select the opti­mal treat­ment par­a­ (NLCS): a pro­spec­tive US patient cohort treated pre­dom­i­nantly in com­mu­
nity prac­tices. Ann Oncol. 2013;24(2):441-448.
digm. The ideal way to merge existing clin­i­cal and bio­logic data 17. Buske C, Hoster E, Dreyling M, Hasford J, Unterhalt M, Hiddemann W.
in a sophis­ti­cated and accu­rate model for high-risk FL remains a The Follicular Lymphoma International Prognostic Index (FLIPI) sep­a­rates
work in prog­ress. high-risk from inter­me­di­ate- or low-risk patients with advanced-stage fol­
lic­ul­ar lym­phoma treated front-line with rituximab and the com­bi­na­tion
of cyclo­phos­pha­mide, doxo­ru­bi­cin, vin­cris­tine, and pred­ni­sone (R-CHOP)
Conflict-of-inter­est dis­clo­sure with respect to treat­ment out­come. Blood. 2006;108(5):1504-1508.
Carla Casulo: Research funding from Gilead, Verastem, Genen- 18. Brice P, Bastion Y, Lepage E, et al. Comparison in low-tumor-bur­den fol­
tech, and BMS. lic­u­lar lym­pho­mas between an ini­tial no-treat­ment pol­icy, prednimustine,

318  |  Hematology 2021  |  ASH Education Program


or inter­feron alfa: a ran­dom­ized study from the Groupe d’Etude des Lym- devel­oped by the inter­na­tional fol­lic­u­lar lym­phoma prog­nos­tic fac­tor pro­ject.
phomes Folliculaires. Groupe d’Etude des Lymphomes de l’Adulte. Clinical J Clin Oncol. 2009;27(27):4555-4562.
trial ran­dom­ized con­trolled trial. J Clin Oncol. 1997;15(3):1110-1117. 38. Rutherford SC, Herold M, Hiddemann W, et  al. Impact of bone mar­row
19. Gao F, Hu J, Zhang J, Xu Y. Prognostic value of periph­eral blood lym­pho­ biopsy on response assess­ ment in immunochemotherapy-treated lym­
cyte/mono­cyte ratio in lym­phoma. J Cancer. 2021;12(12):3407-3417. phoma patients in GALLIUM and GOYA. Blood Adv. 2020;4(8):1589-1593.
20. Mohsen A, Taalab M, Abousamra N, Mabed M. Prognostic sig­nif­i­cance of 39. Mir F, Mattiello F, Grigg A, et  al. Follicular Lymphoma Evaluation Index
abso­lute lym­pho­cyte count, abso­lute mono­cyte count, and abso­lute lym­ (FLEX): a new clin­i­cal prog­nos­tic model that is supe­rior to existing risk
pho­cyte count to abso­lute mono­cyte count ratio in fol­lic­u­lar non-Hodgkin scores for predicting pro­gres­sion-free sur­vival and early treat­ment fail­ure
lym­phoma. Clin Lymphoma Myeloma Leuk. 2020;20(9):e606-e615. after front­ line immunochemotherapy. Am J Hematol. 2020;95(12):1503-
21. Mozas P, Rivero A, Rivas-Delgado A, et  al. A low lym­pho­cyte-to-mono­ 1510.
cyte ratio is an inde­pen­dent pre­dic­tor of poorer sur­vival and higher risk 40. Nath K, Law S-C, Sabdia M-B, et  al. Intratumoral T cells have a dif­fer­en­
of his­to­log­i­cal trans­for­ma­tion in fol­lic­u­lar lym­phoma. Leuk Lymphoma. tial impact on FDG-PET param­ e­
ters in fol­ lic­
u­lar lym­
phoma. Blood Adv.
2021;62(1):104-111. 2021;5(12):2644-2649.
22. Mir F, Barrington SF, Brown H, et al. Baseline SUVmax did not pre­dict his­ 41. Bolen CR, Mattiello F, Herold M, et  al. Treatment depen­dence of prog­
to­log­i­cal trans­for­ma­tion in fol­lic­u­lar lym­phoma in the phase 3 GALLIUM nos­tic gene expres­sion sig­na­tures in de novo fol­lic­u­lar lym­phoma. Blood.
study. Blood. 2020;135(15):1214-1218. 2021;137(19):2704-2707.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/313/1851762/313casulo.pdf by guest on 13 December 2021


23. Barrington SF, Meignan M. Time to pre­pare for risk adap­ta­tion in lym­phoma 42. Pastore A, Jurinovic V, Kridel R, et al. Integration of gene muta­tions in risk
by stan­dard­iz­ing mea­sure­ment of met­a­bolic tumor bur­den. J Nucl Med. prog­nos­ti­ca­tion for patients receiv­ing first-line immunochemotherapy for
2019;60(8):1096-1102. fol­lic­u­lar lym­phoma: a ret­ro­spec­tive anal­y­sis of a pro­spec­tive clin­i­cal trial
24. Qu X, Li H, Braziel RM, et al. Genomic alter­ations impor­tant for the prog­ and val­i­da­tion in a pop­u­la­tion-based reg­is­try. Lancet Oncol. 2015;16(9):1111-
no­sis in patients with fol­lic­u­lar lym­phoma treated in SWOG study S0016. 1122.
Blood. 2019;133(1):81-93. 43. Jurinovi V, Passerini V, Oestergaard M, et al. Evaluation of the m7-FLIPI in
25. Tobin JWD, Keane C, Gunawardana J, et al. Progression of dis­ease within patients with fol­lic­u­lar lym­phoma treated within the gal­lium trial: EZH2
24 months in fol­lic­u­lar lym­phoma is asso­ci­ated with reduced intratumoral muta­tion sta­tus may be a pre­dic­tive marker for dif­fer­en­tial effi­cacy of che­
immune infil­tra­tion. J Clin Oncol. 2019;37(34):3300-3309. mo­ther­apy. Blood. 2019;134(suppl 1):122.
26. Dave SS, Wright G, Tan B, et al. Prediction of sur­vival in fol­lic­u­lar lym­phoma 44. Lockmer S, Ren W, Brodtkorb M, et al. M7-FLIPI is not prog­nos­tic in fol­lic­
based on molec­u­lar fea­tures of tumor-infil­trat­ing immune cells. N Engl J u­lar lym­phoma patients with first-line rituximab chemo-free ther­apy. Br J
Med. 2004;351(21):2159-2169. Haematol. 2020;188(2):259-267.
27. Kridel R, Xerri L, Gelas-Dore B, et  al. The prog­nos­tic impact of CD163- 45. Mondello P, Fama A, Larson MC, et al. Intrafollicular CD4+ T-cells as an inde­
pos­i­tive mac­ro­phages in fol­lic­u­lar lym­phoma: a study from the BC can­ pen­dent pre­dic­tor of early clin­i­cal fail­ure in newly diag­nosed fol­lic­u­lar lym­
cer agency and the lym­ phoma study asso­ ci­

tion. Clin Cancer Res. phoma. Blood. 2019;134(suppl 1):121.
2015;21(15):3428-3435. 46. Trotman J, Barrington SF, Belada D, et al; PET inves­ti­ga­tors from the GAL-
28. Zohren F, Bruns I, Pechtel S, et  al. Prognostic value of cir­cu­lat­ing Bcl-2/ LIUM study. Prognostic value of end-of-induc­tion PET response after first-
IgH lev­els in patients with fol­lic­u­lar lym­phoma receiv­ing first-line immu- line immunochemotherapy for fol­lic­u­lar lym­phoma (GALLIUM): sec­ond­ary
nochemotherapy. Blood. 2015;126(12):1407-1414. anal­y­sis of a randomised, phase 3 trial. Lancet Oncol. 2018;19(11):1530-1542.
29. Sarkozy C, Huet S, Carlton VE, et al. The prog­nos­tic value of clonal het­ero­ 47. Pott C, Hoster E, Kehden B, et al. Minimal resid­ual dis­ease response at end
ge­ne­ity and quan­ti­ta­tive assess­ment of plasma cir­cu­lat­ing clonal IG-VDJ of induc­tion and dur­ing main­te­nance cor­re­lates with updated out­come in
sequences at diag­no­sis in patients with fol­lic­u­lar lym­phoma. Oncotarget. the phase III GALLIUM study of obinutuzumab- or rituximab-based immu-
2017;8(5):8765-8774. nochemotherapy in pre­vi­ously untreated fol­lic­u­lar lym­phoma patients.
30. Kridel R, Chan FC, Mottok A, et al. Histological trans­for­ma­tion and pro­gres­ Blood. 2018;132(suppl 1):396.
sion in fol­lic­u­lar lym­phoma: a clonal evo­lu­tion study. PLoS Med. 2016;13(12): 48. Scherer F, Kurtz DM, Newman AM, et al. Distinct bio­log­i­cal sub­types and pat­
e1002197. terns of genome evo­lu­tion in lym­phoma revealed by cir­cu­lat­ing tumor DNA.
31. Casulo C, Byrtek M, Dawson KL, et al. Early relapse of fol­lic­u­lar lym­phoma Sci Transl Med. 2016;8(364):364ra155.
after rituximab plus cyclo­phos­pha­mide, doxo­ru­bi­cin, vin­cris­tine, and pred­ 49. Maurer MJ, Ghesquières H, Link BK, et  al. Diagnosis-to-treat­ment inter­
ni­sone defi­nes patients at high risk for death: an anal­y­sis from the national val is an impor­tant clin­i­cal fac­tor in newly diag­nosed dif­fuse large B-cell
lymphocare study. J Clin Oncol. 2015;33(23):2516-2522. lym­phoma and has impli­ca­tion for bias in clin­i­cal tri­als. J Clin Oncol. 2018;
32. Jurinovic V, Kridel R, Staiger AM, et al. Clinicogenetic risk mod­els pre­dict 36(16):1603-1610.
early pro­gres­sion of fol­lic­u­lar lym­phoma after first-line immunochemother- 50. Skander J, Fredrickson J, Coimbra A, Carano RAD, Christoffer T, El-Galaly C,
apy. Blood. 2016;128(8):1112-1120. Knapp A, Nielsen TG, Sahin D, Bengtsson T, de Crespigny, A. A fully auto-
33. Maurer MJ, Ghesquieres H, Jais JP, et al. Event-free sur­vival at 24 months is mated measurement of total metabolic tumor burden in diffuse large B-cell
a robust end point for dis­ease-related out­come in dif­fuse large B-cell lym­ lymphoma and follicular lymphoma. Blood. 2019;34 (suppl 1):4666.
phoma treated with immunochemotherapy. J Clin Oncol. 2014;32(10):1066- 51. Sarkozy C, Maurer MJ, Link BK, Ghesquieres H, Nicolas E, Thompson CA,
1073. Traverse-Glehen A, Feldman AL, Allmer C, Slager SL, Ansell SM, Habermann
34. Wagner-Johnston ND, Link BK, Taylor M, Bartlett NL. Risk fac­tors for early TM, Bachy E, Cerhan JR, Salles G. Cause of death in follicular lymphoma in
trans­for­ma­tion of fol­lic­u­lar lym­phoma (FL): report from the National Lym- the first decade of the rituximab era: a pooled analysis of French and US
phoCare Study (NLCS). Blood. 2009;114(22):2698. cohorts. J Clin Oncol. 2019 Jan 10;37(2):144-152.
35. Sarkozy C, Trneny M, Xerri L, et al. Risk fac­tors and out­comes for patients with 52. Luminar S, Manni M, Galimberti S, et al. Response-adapted postinduction
fol­lic­u­lar lym­phoma who had his­to­logic trans­for­ma­tion after response to strategy in patients with advanced-stage follicular lymphoma: The FOLL12
first-line immunochemotherapy in the PRIMA trial. J Clin Oncol. 2016;34(22): Study. Journal of Clinical Oncology. 2021 (Oct. 28).
2575-2582.
36. Link BK, Day BM, Zhou X, et al. Second-line and sub­se­quent ther­apy and
out­ comes for fol­ lic­
u­lar lym­
phoma in the United States: data from the
obser­va­tional National LymphoCare Study. Br J Haematol. 2019;184(4):660-
663.
37. Federico M, Bellei M, Marcheselli L, et  al. Follicular Lymphoma Interna- © 2021 by The Amer­i­can Society of Hematology
tional Prognostic Index 2: a new prog­nos­tic index for fol­lic­u­lar lym­phoma DOI 10.1182/hema­tol­ogy.2021000264

Prakash Singh Shekhawat


Defining risk in FL  |  319
INDOLENT LYMPHOMAS

Does MRD have a role in the management


of iNHL?

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/320/1852135/320giudice.pdf by guest on 13 December 2021


Ilaria Del Giudice, Irene Della Starza, and Robin Foà
Hematology, Department of Translational and Precision Medicine, Sapienza University, Rome, Italy

Among indolent non-Hodgkin lymphomas (iNHLs), the analysis of measurable/minimal residual disease (MRD) has been
extensively applied to follicular lymphoma (FL). Treatment combinations have deeply changed over the years, as well as
the techniques to measure MRD, which is currently evaluated only in the setting of clinical trials. Here, we discuss the evi-
dence on the role of molecular monitoring in the management of FL. Mature data support the quantification of molecular
tumor burden at diagnosis as a tool to stratify patients in risk categories and of MRD evaluation at the end of treatment
to predict progression-free survival and overall survival. Moreover, MRD deserves further studies as a tool to refine the
clinical/metabolic response and to modulate treatment intensity/duration. Patients with a higher relapse probability can
be identified, but the relevance of continuous molecular follow-up should be clarified by kinetic models of MRD analysis.
Being the BCL2/heavy chain immunoglobulin gene hybrid rearrangement detectable in about 50% to 60% of advanced FL
and in 30% of positron emission tomography/computed tomography–staged localized FL, technical advancements such
as next-generation sequencing/target locus amplification may allow broadening the FL population carrying a molecular
marker. Droplet digital polymerase chain reaction can better quantify MRD at low levels, and novel sources of DNA, such
as cell-free DNA, may represent a noninvasive tool to monitor MRD. Finally, MRD in other iNHLs, such as lymphoplasmacytic
lymphoma/Waldenström macroglobulinemia and marginal zone lymphoma, is beginning to be explored.

LEARNING OBJECTIVES
• Describe the accumulating data on MRD monitoring in patients with advanced and localized FL in the immuno-
chemotherapy and chemo-free era
• Understand how MRD might be used in the clinical management of patients with FL
• Discuss how new technologies could overcome the current limitations in order to widely apply MRD to patients
with FL and to other indolent NHLs

Introduction some patients not requiring treatment for years, others expe-
Indolent non-Hodgkin lymphomas (iNHLs) often spread to riencing long-lasting remissions after first-line treatment,
the bone marrow (BM) and/or peripheral blood (PB) and and ~20% rapidly relapsing within 24 months from treatment
can be monitored through the identification at diagno- initiation.5 Transformation into an aggressive lymphoma,
sis of a disease marker to be followed during treatment.1 whose frequency has decreased with the use of rituximab
Minimal/measurable residual disease (MRD) analysis has (R), strongly impairs patients’ survival.6 The current prog-
been extensively applied to follicular lymphoma (FL), which nostic scores (follicular lymphoma international prognostic
represents the main focus of this review.2,3 MRD in other index [FLIPI], FLIPI-2, PRIMA-prognostic index [PRIMA-PI],
iNHLs is starting to be explored, and we briefly comment follicular lymphoma evaluation index [FLEX]), based on clin-
on this at the end of the article. ical parameters, fail to predict the clinical course of individ-
ual patients.7,8 Knowledge of FL’s biological heterogeneity,
Follicular lymphoma which relies on the complex interactions between nonma-
FL is the second most frequent non-Hodgkin lymphoma. lignant immune/stromal components and tumor cells, has
Despite the improvement in outcome, mainly due to the not identified clinically applicable predictive biomarkers.9,10
combination of anti-CD20 monoclonal antibodies with che- The molecular hallmark of FL, the t(14;18)(q32;q21) trans-
motherapy, most patients relapse, and the disease remains location, resulting in the hybrid BCL2/heavy chain immu-
uncurable.4 The clinical course of FL is heterogeneous, with noglobulin gene (IGH) rearrangement, is the first necessary

320 | Hematology 2021 | ASH Education Program


but not suf­fi­cient step in lymphomagenesis.11 The BCL2/IGH gene chemoimmunotherapy followed by a con­sol­i­da­tion with R plus
can be used to sup­port FL diag­no­sis and to eval­u­ate treat­ment a ran­dom­ized main­te­nance) and in the FOLL05 trial, ran­domly
response in terms of MRD. The first evi­dence on the value of MRD assigning patients to R-CHOP, R-fludarabine, and mitoxantrone
in FL goes back to 1991.12 Thereafter, treat­ment com­bi­na­tions or R-cyclo­phos­pha­mide, vin­cris­tine, and pred­ni­sone (CVP).17,18 In
have deeply changed, as well as the tech­niques to mea­sure MRD, the for­mer, the molec­u­lar tumor bur­den was cor­re­lated to BM
which is cur­rently eval­u­ated only in the set­ting of clin­i­cal tri­als.7 inva­sion but not with FLIPI.17 In the lat­ter, high BCL2/IGH lev­
Here, we dis­cuss the cur­rent evi­dence on the role of MRD in els at enroll­ment, documented in patients with high FLIPI and
the man­age­ment of FL, starting from 2 clin­i­cal cases. FLIPI2 scores, were sig­nif­i­cantly asso­ci­ated with a lower over­all
response rate and a higher relapse rate and retained a neg­a­tive
impact on 3-year PFS besides high FLIPI and lack of com­plete
response (CR), inde­pen­dently from the ran­dom­i­za­tion arm.18
CLINICAL CASES Likewise, in the NHL1-2003 trial, com­par­ing R-CHOP to R-benda-
Patient 1. A 49-year-old man with FL grade 2, Ann Arbor stage IV, mustine, high pre­treat­ment PB BLC2/IGH lev­els were asso­ci­ated

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/320/1852135/320giudice.pdf by guest on 13 December 2021


high-risk FLIPI, high-tumor bur­den and major breakpoint region with BM involve­ment, stage IV, high tumor bur­den, and increased
(MBR)+ in PB/BM was treated in a clin­i­cal trial from July 2017 with β2-microglobulin, being an inde­pen­dent prog­nos­tic marker for
R-bendamustine, followed by 3 courses of 4-weekly doses of R PFS and fur­ther strat­i­fy­ing patients with inter­me­di­ate/high FLIPI.19
main­te­nance, com­pleted in August 2019. At the end of induc­tion In the Relevance trial, com­ par­
ing the che­ mo­ ther­
apy-free
(EOI), a pos­i­tron emis­sion tomog­ra­phy (PET)−/MRD+ response reg­i­men lenalidomide  +  R (R2) vs R  +  che­mo­ther­apy followed by
was documented. MRD remained pos­i­tive through­out main­te­ main­te­nance, molec­u­lar tumor bur­den at diag­no­sis quan­ti­fied
nance and at the end of treat­ment. In March 2021, 19 months after by drop­let dig­i­tal PCR (ddPCR) was sig­nif­i­cantly asso­ci­ated with
main­te­nance com­ple­tion, he presented with an abdom­i­nal bulky the MRD sta­tus at week 24 (EOI).22
dis­ease recur­rence. The new biopsy spec­i­men showed FL grade In early stage FL, despite a neg­a­tive BM biopsy, BCL2/IGH+
3A; the BM biopsy spec­i­men showed no lym­phoma infil­tra­tion. cells spread­ing from the orig­i­nal lymph node could be detected
Patient 2. Based on the biopsy spec­i­men of a 3-cm ingui­nal in the PB/BM at diag­no­sis in more than 50% of patients.24,26 The
lymph node, in May 2016, an asymp­tom­atic 63-year-old woman 84-month PFS was 90.9% in patients with unde­tect­able/low lev­
was diag­nosed with FL grade 1 to 2, Ann Arbor stage I, MBR+ in els (<1   ×   10−5) of cir­cu­lat­ing BCL2/IGH+ cells quan­ti­fied by ddPCR
PB/BM. She received involved-site radi­a­tion (24 Gy), followed at diag­no­sis vs 38% in those with higher lev­els (P = .015).24
by ofatumumab con­sol­i­da­tion for MRD+ response, as per clin­ Final com­ment. Both in advanced and local­ized FL, quan­ti­
i­cal trial. She became MRD−, which persisted for 1.5 years at a fi­ca­tion of molec­u­lar tumor bur­den at base­line by RQ-PCR or
6-month MRD mon­i­tor­ing. In August 2018, she returned MRD+ in ddPCR pre­dicts PFS, being an inde­pen­dent prog­nos­tic marker.
the BM, shortly followed by a clin­i­cal relapse. Circulating BCL2/IGH lev­els, which may sim­ply reflect not only
tumor bur­ den but also the enhanced lym­ phoma cell migra­
tion and inva­sive­ness, could help to refine our capac­ity to risk-
Is molec­u­lar tumor bur­den at diag­no­sis use­ful strat­ify patients.
to strat­ify ­patients in risk categories?
The tumor bur­den molec­u­lar quan­ti­fi­ca­tion at diag­no­sis pre­dicts Is MRD use­ful to refine the clin­i­cal response?
pro­gres­sion-free sur­vival (PFS) in patients with FL, in both ad- In the pre-R era, sev­eral tri­als showed that autol­o­gous stem cell
vanced and local­ized stages (Table 1). Rambaldi et al13 select- trans­plant induced higher rates of molec­u­lar remis­sion (60%-70%)
ed patients with FL for the pres­ence of BCL2/IGH pos­i­tiv­ity in than stan­dard anthracycline-based che­mo­ther­apy (30%-50%).2,3
the BM and treated them with cyclo­phos­pha­mide, doxo­ru­bi­cin, In the R era, the addi­tion of R induced a con­ver­sion to a MRD−
vin­cris­tine, and pred­ni­sone (CHOP), followed by 4-weekly R in sta­tus in 70% of patients MRD+ after CHOP and in 70% to 84% af-
MRD+ cases, and divided them into low (43%), inter­ me­ di­
ate ter R-fludarabine, mitoxantrone, and dexa­ meth­a­
sone (FND).13,14,17
(34%), and high risk (23%) according to BM BCL2/IGH real-time The com­bi­na­tion of che­mo­ther­apy with R ­front­line showed an
quan­ti­ta­tive (RQ )–poly­mer­ase chain reac­tion (PCR) lev­els at ­increased MRD− response rate at EOI com­pared with che­mo­ther­
diag­no­sis. High BCL2/IGH+ lev­els were sig­nif­i­cantly asso­ci­ated apy alone, with a pref­er­en­tial MRD clear­ance in the PB (Figure 1,
with a reduced event-free sur­vival (EFS) and were an inde­pen­ Table 1).13-15,17-22,24,26-28 In the Gallium trial, the BM MRD− rates at EOI
dent pre­dic­tor of poor clin­i­cal and molec­u­lar response in mul­ti­ in the R-chemo arm were higher than ­pre­vi­ously reported and in-
var­i­ate anal­y­sis (MVA). In con­trast, PB BCL2/IGH lev­els at diag­no­ creased fur­ther in the G-arm, e ­ spe­cially after CHOP/CVP. Indeed,
sis were not pre­dic­tive of EFS. G abro­gated the com­part­ment- and ­chemo-related ­effects ­obser-
In large first-line tri­ als employing R-based chemoimmuno- ved in the R-arm. MRD response rates in the PB were sim­i­lar across
therapy for advanced FL, BCL2/IGH was found in 51% to 66% of all­chemo reg­i­mens (96% G-bendamustine [B], 96% G-CHOP, 94%
enrolled patients.17-19,22 Conversely, in the unpub­lished MRD results G-CVP), as well as in the BM (93% after G-B, 93% after G-CHOP).20,21
from the Gallium trial, in which obinutuzumab (G)–based che­mo­ The impor­tance of MRD was also shown in the Relevance
ther­apy plus G-main­te­nance resulted supe­rior to R-che­mo­ther­apy trial, where MRD was quan­ti­fied for the first time by ddPCR. At
plus R-main­te­nance, a clonal marker was detected in 88% of 1101 EOI, 98% and 78% of patients achieved a com­plete molec­u­lar
patients, higher than in other stud­ies, because both BCL2/IGH response in the PB and BM. A com­plete molec­u­lar response was
and IGH rearrangements were screened by con­sen­sus PCR.20,21 reached more fre­quently with R2 (90%) than with R-chemo (77%)
RQ-PCR strat­i­fied patients with low, inter­me­di­ate, and high (P    =   .022) (Figure 1, Table 1).22
BM tumor bur­den at diag­no­sis with a sig­nif­i­cantly dif­fer­ent PFS, In advanced stage FL, since MRD− sta­tus after treat­ment can be
both in elderly patients treated within the ML17638 trial (short detected in patients in CR or partial response (PR), the molec­u­lar
Prakash Singh Shekhawat
Minimal resid­ual dis­ease in indo­lent NHL  |  321
Table 1. Studies reporting on MRD detec­tion and mon­i­tor­ing in FL

Advanced
Study dis­ease Patients Therapy Tissue Method Marker Tumor bur­den MRD− % Clinical impact Follow-up
Rambaldi BCL2/IGH  + 128 (79 CHOP   ×   6 BM    ±    PB Nested Enrolled PCR  + — End of CHOP: 3-year FFR 17 months
et al13 FL, received R) 4 weeks R PCR patients BM MRD− 36% (43/118) BM MRD− after CHOP (8-39)
untreated in PCR  + (BCL2/IGH PB MRD− 35% (43/121) 52%
patients 100%) After R: MRD− 59  % at BM MRD− after R at
week 12, 74  % at week 28, week 44, 3-year FFR
and 63% at week 44 57% vs 20% MRD+
Rambaldi BCL2/IGH  + 86 (same BM RQ- Enrolled PCR    + At base­line: See above FFR: 64  % MRD− vs 32  % 56 months
et al14 FL, series) PCR patients Low (1 cell in 103–105): MRD  +  (P    <    .006) (40-75)
untreated (BCL2/IGH 43  %;
100%) inter­me­di­ate (1 cell in
102–103): 34  %;
high (>1 cell in 102): 23  %
CR: 71  % in low vs 26  %
in high
EFS: 59  % in

322  |  Hematology 2021  |  ASH Education Program


low/inter­me­di­ate
vs 32% in high
Ladetto High-risk FL 134 Phase 3 BM Nested 73/104 (70  %) — Overall, 65  % (39/60) MRD− PFS MRD− vs MRD  + 51 months
et al15 untreated R-HDS vs PCR BCL2/IGH After CHOP-R: MRD− 44% (P    <    .001), regard­less
<60 years CHOP   ×   6 + (65)  +  IGH (8) After R-HDS: MRD− 80% of treat­ment arm
R   ×   4 (P  <  .002)
(GITMO/IIL)
Bruna High-risk FL 134 Update of the BM Nested — Overall, 65% (39/60) MRD− 13-year OS 13 years
et al16 untreated phase 3 PCR R-HDS 64.5  % vs 68.5  %
<60 years R-HDS vs CHOP-R
CHOP   ×   6  + 13-year OS
R   ×   4 BM MRD− 82.1  % vs
(GITMO/IIL) BM MRD  +   51.9  %
(P    =.030)
20/24 (83  %) patients
with 4-year long-
term MRD− in first CR
Ladetto FL >60 years, 227    ML17638 BM RQ- 116/227 PFS: 80  % in low vs 75  % At EOI: 3-year PFS: at end 42 months
et al17 untreated R-FND ×   4   +   4R PCR (BCL2/IGH in inter­me­di­ate vs MRD− 70  % of con­sol­i­da­tion:
±  4R every 51  %) 66  % in high End of con­sol­i­da­tion: 72  % MRD− vs 39  %
2 months PFS: 61% at month  +  42 MRD− 84  % MRD    +    (16  %) (P   <.007)
in both PCR  +  and Median PFS: 12 months
PCR  − at base­line MRD− (RQ-PCR);
75  % of MRD− free
of PD at 36 months
(P  < .001)
PFS main­te­nance arm:
83  % PCR− vs 60  %
PCR  + (P    = .007)
PFS obser­va­tion arm:
71  % PCR− vs 50  %  
PCR   + (P     <  .001)

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/320/1852135/320giudice.pdf by guest on 13 December 2021


Table 1. (Continued)

Advanced
Study dis­ease Patients Therapy Tissue Method Marker Tumor bur­den MRD− % Clinical impact Follow-up
Galimberti FL untreated 415 Phase 3 BM Nested 220/415 At diag­no­sis: EOI Relapse rate: 33% NA
et al18 FOLL05 trial PCR; (BCL2/IGH 61.9% PCR+ non-CR vs MRD− (qPCR): 109/154 (70.8%) for MRD− vs 41%
R-CHOP, R- RQ- 53%) 38.1% PCR− (P  = .027) (no dif­fer­ence between arms) for MRD+ at EOI
CVP, R-FN PCR RQ-PCR in 105 BM biopsy+ 32% non-CR R-CHOP 39% (P  = .363)
cases vs 21.8% in BM− cases R-FN 36% 3-year PFS 64.3% for
(P  = .021) R-CVP 25% MRD− vs 53.1 for
3-year PFS: 74% in RQ decrease >3 log: MRD+ at EOI (P = .08)
PCR−/BM− at diag­ R-CHOP 42.1% 3-year PFS 66% for
no­sis vs 55% in R-FN 36.8% R-CVP 21.1% 63 MRD− vs 41%
PCR+/BM+ (P  = .04) (P  = .07) for 24 MRD+ at
ORR 38.9% in high RQ vs 12 months from EOI
76.6% in low (P  = .006) (P  = .015)
Relapses: 22% in 3-year PFS 84% for
<1 × 10–4 cop­ies vs 46 MRD− vs 50%
78% in >1 × 10–4 cop­ies for 19 MRD+ at
(P  = .033) at base­line 24 months (P  = .014)
3-year PFS: 80% in low vs
59% in high (P  = .015)
Zohren FL, untreated 114 Phase 3 PB RQ- 114/173 Median PFS: 22 months Overall MRD− 78/92 (85%) 3-year PFS 63% over­all 41 months
et al19 NHL1-2003 trial PCR (BCL2/IGH in high BCL2/IGH After R-B (0-69)
R-CHOP vs R-B 66%) (ratio >1, n  =  28) vs NR MRD− 43/48 (89.6%)
in inter­me­di­ate (ratio After R-CHOP
0.1-1, n  =  24) vs NR in MRD− 35/44 (79.5%)
low (<0.1, n  =  55)
High vs inter­me­di­ate:

Prakash Singh Shekhawat


HR, 4.28; 95% CI,
1.70-10.77 (P  = .002)
High vs low: HR, 3.02;
95% CI, 1.55-5.86
(P  = .001)
Pott FL, untreated 1101 Phase 3 PB   ±   BM RQ- 968/1101 Clonal marker vs no MI PB MRD−: G-vs R-based treat­ 4-year PFS: 80% early 57 months
et al20,21 Gallium trial R PCR (BCL2/IGH marker at base­line: ment (94% vs 89%, P   =.013) respond­ers (PB MI
vs G (CVP, B, Nested +  IGH 88%) stage IV 61% vs 34%; EOI PB   ±   BM MRD−: G- vs R-based MRD−/EOI MRD−)
CHOP)  +  main­ PCR 815 (74%) BM biopsy infil­tra­ treat­ment (92% vs 85%, P   =  .0041) vs 30% PB MI
te­nance suit­able for tion 58.2% vs 14.3%; EOI BM MRD−: G- vs R-based treat­ MRD   +/EOI MRD   +
RQ-PCR high-risk FLIPI 44.5% ment (93% vs 83%, P  =.0014), NS (P   < .0001) vs 60%
vs 30.8%; extranodal in the PB late respond­ers, PB
involve­ment 70% vs EOI MRD− PB  ±  BM: R-CVP 76% vs MI MRD   +/EOI MRD−
40% G-CVP 91.4%; R-CHOP 77.8% vs (P   = .0133)
G-CHOP 91.3%; R-B 89.6% vs EOI MRD− sig­nif­i­
G-B 92.5% cantly affected PFS
EOI PB MRD−: R-CVP 79% vs G-CVP (HR, 0.38; 95% CI,
94%; R-CHOP 93% vs G-CHOP 0.26-0.56; P < .0001)
96%; R-B 96% vs G-B 96% and OS (HR, 0.35;
EOI BM MRD−: R-CHOP 74% vs 95% CI, 0.17-0.72;
G-CHOP 93%; R-B 87% vs G-B P   = .0027)
93%
EOI MRD+: 22/24 (92%) patients
in the G-chemo arm and 36/46
(78%) patients in the R-chemo

Minimal resid­ual dis­ease in indo­lent NHL  |  323


arm achieved MRD neg­a­tiv­ity
early dur­ing main­te­nance
Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/320/1852135/320giudice.pdf by guest on 13 December 2021
Table 1. (Continued)

Advanced
Study dis­ease Patients Therapy Tissue Method Marker Tumor bur­den MRD− % Clinical impact Follow-up
Delfau- FL untreated 440 Phase 3 PB   ±   BM ddPCR 222/440 Tumor bur­den 10 times At EOI (week 24): 3-year PFS: 84% for NA
Larue Relevance trial (BCL2/IGH higher in PB and BM of PB MRD− 98%, BM MRD− 78% MRD− (in PB and/or
et al22 R2 (18 cycles of 50.45%) patients with week 24 R2 arm: MRD− 105/117; 90% BM) vs 55% for
lenalidomide BCL2/IGH+ MRD+, com­pared to vs R-chemo MRD− 70/90; MRD+ (P   = .015)
plus R, patients: patients with week 24 77% (P  =  .022) 3-year PFS: 85% for BM
followed by stage III-IV MRD− (P  = .03 and .02) MRD− vs 54% for BM
R main­te­ dis­ease MRD+ (P   = .011)
nance ther­ (96% vs 90%, MRD+ at EOI: R-chemo
apy every 8 P  =  .004), arm (HR, 3.3; 95%
weeks for FLIPI score >1 CI, 1.2-9.2; P   = .02)
12 cycles) (91% vs 83%, vs R2 arm (HR, 2;
vs R-chemo P  =  .002), 95% CI, 0.6-6.8;
(CHOP) BM involve­ P    = .27)
followed by ment (62% vs
R-main­te­ 48%, P  =  .003)
nance every
8 weeks for

324  |  Hematology 2021  |  ASH Education Program


12 cycles
Pott et al23 R refrac­tory FL Phase 3 PB    ±    BM RQ- 71% (228/319) At MI: PB MRD− 79% (41/52) in G-B PFS (HR, 0.33; P < .0001) 31.8
GADOLIN PCR (63% vs 47% (17/36) in B (P   = .0029) and OS (HR, 0.39; months
trial (and BCL2/IGH+ At EOI, PB or BM MRD− 86% P   = .008) for MRD−
G-B  +  G sub­se­ [MBR, mcr, (54/63) in G-B vs 55% (30/55) in PFS in MRD +3.3
main­te­nance quent 3′MBR] and B (P   = .0002) months in both arms
vs B nested) 72% IGH) in MRD− 8.54 B vs
52% (166/319) 35.71 G-B
suit­able for
RQ-PCR ≤10-4
Localized dis­
Study ease Patients Therapy Tissue Method Marker Tumor bur­den MRD− % Clinical impact Follow-up
Pulsoni Stage I (78%) 67 IFRT (24-30 Gy) PB   ±   BM Nested 72% 84-month PFS: 75% for After RT: MRD −50% Different PFS in MRD+ 82
et al24 Stage II (22%) (+4 weeks R in PCR BCL2/IGH- vs 59% for After R: 16/19 patients post-IFRT: months
untreated MRD+ from RQ- BCL2/IGH+ at base­ (84%) MRD− between untreated (17-196)
FL 2005 in 19 PCR line (P  = .26) patients vs patients
patients) ddPCR 84-month PFS: 90.9% in treated with R after
11patients with MRD 2005 (P   = .049)
<10−5 vs 38% in Different PFS of MRD+
19 patients with MRD patients dur­ing the
≥10−5 by ddPCR at fol­low-up vs per­sis­
base­line (P  = .015) tently MRD− patients
(P   = .038)
Herfarth Stage I (56%) 85 (60 long MIR trial 83 (64 Overall 24/64 57% below 10−4 MRD−: 20/21 (95%) at week 18 15/19 patients MRD− 29.6
et al25 Stage II (44%) term) R  ×  8 cycles  + PB, 13 (38%; PB with no pro­gres­sion; months
untreated IFRT (30-40 BM) 35%, BM 3 patients relapsed,
FL Gy) 46%) in 2 asso­ci­ated or
Stage II predicted by MRD+
> stage I sam­ple
(P  = .0038)
Only tri­als includ­ing anti-CD20 mono­clo­nal antibodies have been included.
FN, fludarabine and mitoxantrone; FND, fludarabine, mitoxantrone, and dexa­meth­a­sone; IFRT, involved field radio­ther­apy; MIR, Mabthera and Involved field Radiotherapy (NCT00509184); NA,
not available; NS, not significant; ORR, over­all response rate; qPCR, quan­ti­ta­tive PCR.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/320/1852135/320giudice.pdf by guest on 13 December 2021


Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/320/1852135/320giudice.pdf by guest on 13 December 2021
Figure 1. Rates of MRD negativity after anti-CD20 based treatments in advanced FL. *Data are expressed as proportion of MRD−
cases at EOI. gDNA, genomic DNA.

and ­clin­i­cal/radio­logic response can be com­bined to refine prog­ Is MRD use­ful to pre­dict PFS?
no­sis. Ladetto et al17 showed that at month +8, 69% of patients The prog­nos­tic impact of MRD in advanced FL has been dem­on­
were CR/nested PCR−, 15% PR/PCR−, and 11% CR/PCR+, with strated across dif­fer­ent treat­ment strat­e­gies. All chemoimmuno-
a 3-year PFS of 77%, 59%, and 45%, respec­tively; the 3 PR/PCR+ therapy tri­als, with or with­out main­te­nance, are asso­ci­ated with
cases relapsed within 23 months. Similar results were shown in a sig­nif­i­cant improve­ment of PFS in molec­u­lar respond­ers, inde­
the FOLL05, where PFS was sig­nif­i­cantly lon­ger in CR-PR/PCR− pen­dent from other prog­nos­tic fac­tors (Table 1). Rambaldi et al13
than in CR-PR/PCR+ patients.18 As in the Relevance study,22 the showed that the achieve­ment of a BM PCR− sta­tus was asso­ci­ated
pre­dic­tive value of MRD was supe­rior to clin­i­cal response in MVA.18 with a higher free­dom-from-recur­rence (FFR) (64% vs 32% for PCR+
Luminari et al,29 in a small sub­group of patients (n = 41) from patients). Interestingly, this study antic­i­pated later obser­va­tions17,18,21
the FOLL05 trial, suggested the com­ple­men­tary role of MRD (ie, most PCR− patients after 6 cycles were already neg­a­tive after 3
and PET at EOI in defin­ing the response to treat­ment, with a cycles, and a delayed MRD clear­ance induced by R was pos­si­ble). In
con­cor­dance of 76%. The 3-year PFS was 78%, 50%, and 27% the phase 3 Gruppo Italiano Trapianto Midollo Osseo/Intergruppo
in 28 PET−/MRD−, 8 PET  −    /MRD  +  , and 5 PET+ cases, respec­ Italiano Linfomi (GITMO/IIL) trial, com­par­ing high-dose sequen­tial
tively (P   =   .015). The con­com­i­tant PET/MRD neg­a­tiv­ity was asso­ che­mo­ther­apy with R and auto­graft (R–high dose sequen­tial che­
ci­ated with a bet­ter out­come: 5-year PFS 75% PET−/MRD− vs mo­ther­apy with autografting [HDS]) to R-CHOP, molec­u­lar remis­
35% PET+ or MRD+ (P = .012).29 The com­ple­men­tary role of PET sion—documented in 44% of R-CHOP–treated patients and 80% of
and MRD was fur­ther supported in 298 patients from the Gallium R-HDS–treated patients—was the stron­gest pre­dic­tor of PFS, EFS,
trial: PET−/MRD+ or PET  +  /MRD− patients had a 2.1 higher risk of and FFR.15 The out­come of patients achiev­ing an MRD− response
pro­gres­sion or death than those with both PET−/MRD−. Never- was sim­i­lar regard­less of the treat­ment received, as was the out­
theless, 15% of PET−/MRD− patients progressed within 2.5 years come of patients remaining MRD+.15 In the phase 3 ML17638 trial,17
from EOI.30 MRD neg­a­tiv­ity (by PCR and RQ-PCR) at the end of con­sol­i­da­tion
Final com­ment. The intro­duc­tion of chemoimmunotherapy with (month +8) was asso­ci­ated with a lon­ger 3-year PFS, representing
R and G has allowed an increase in the rates of MRD neg­a­tiv­ity at an inde­pen­dent pre­dic­tor of out­come, besides clin­i­cal response
EOI up to 70% to 80% and 90%, respec­tively. MRD anal­y­sis is a sen­ and FLIPI.17 Galimberti et al18 showed that the BM MRD sta­tus at 12
si­tive tool to refine clin­i­cal response assess­ment in FL. In addi­tion, and 24 months from EOI, but not at the EOI, predicted the 3-year
the com­bi­na­tion of molec­u­lar and met­a­bolic response assess­ PFS. In a MVA includ­ing FLIPI, BM involve­ment, qual­ity of response,
ment is a prom­is­ing and valu­able tool to be fur­ther explored. and arm of ther­apy, MRD per­sis­tence at month +12 (besides BM
Prakash Singh Shekhawat
Minimal resid­ual dis­ease in indo­lent NHL  |  325
involve­ment) and at month +24 (alone) retained a poor prog­nos­tic ham­pered by the lack of adher­ence to long-term MRD eval­u­a­
role. Zohren et al19 showed that a per­sis­tent PB BCL2/IGH pos­i­tiv­ tions.18,19,21,27 In FOLL05, molec­ul­ar recur­rence pre­ceded clin­i­cal re-
ity (n = 14, 15%) after R-che­mo­ther­apy was asso­ci­ated with a shorter lapse by a median of 5 months in 9 of 10 evaluable cases.18 Zohren
PFS (8.7 months vs not reached, P = .002), despite the 2-log reduc­ et al19 over a 41-month fol­low-up showed that 49 patients remained
tion in BCL2/IGH lev­els and in both treat­ment arms. By MVA, both BCL2/IGH– and 43 converted to BCL2/IGH+. Qualitative molec­u­lar
pre­treat­ment and posttreatment BCL2/IGH lev­els were sig­nif­i­cant relapse was not indic­a­tive of an infe­rior PFS, unless asso­ci­ated with
prog­nos­tic fac­tors, the for­mer being the stron­gest. high BCL2/IGH lev­els. In 20 patients who were rig­or­ously sam­pled,
Recently, 3 stud­ies pro­vided the first evi­dence that a MRD− the inter­val from BCL2/IGH redetection to clin­i­cal relapse was 9.5
achieve­ment fol­low­ing treat­ment is asso­ci­ated also with a pro- months. In the Gallium trial, patients fail­ing to become MRD− had a
longed OS. The long-term fol­low-up of a Brit­ish trial showed the high like­li­hood of expe­ri­enc­ing early pro­gres­sion or death.21
lon­ger sur­vival of MRD− respond­ers after 2 dif­fer­ent che­mo­ther­ More rel­e­vant than the MRD punc­tual eval­u­a­tion is the kinetic
apy reg­i­mens in the pre-R era.31 The updated results of the phase anal­y­sis of molec­u­lar results over time. So far, few data are avail­­
3 GITMO/IIL trial showed a 13-year OS of 82.1% and 51.9% for BM able in FL. The first evi­dence came from Ladetto et al,17 who

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/320/1852135/320giudice.pdf by guest on 13 December 2021


MRD− vs MRD+ patients.16 Moreover, in a sub­group of molec­u­ showed that the accu­mu­la­tion of MRD− results over time reduced
larly mon­i­tored patients for a median of 4 years since treat­ment the like­li­hood of relapse. Kinetic mod­els of MRD anal­y­sis in FL are
com­ple­tion, 20 of 24 (83%) patients alive in CR remained in first awaited.
MRD− response, rais­ing the hypoth­e­sis of a func­tional cure also Final com­ment. Patients with FL with a higher relapse prob­
for FL. In the Gallium trial, at an updated median fol­low-up of a­bil­ity can be iden­ti­fied, but the tim­ing of clin­i­cal relapse is not
57 months, MRD− patients at EOI (n = 564) had a lon­ger PFS than accu­rately predicted by the cur­rent MRD ana­ly­ses. Accumulating
MRD+ (n = 70) (P < .0001) and a bet­ter OS (P = .0027).21 evi­dence sug­gests that a kinetic model of MRD anal­ys­is, more
The Gallium trial also opened the way to novel con­cepts.21 (i) than a sin­gle result, could help in antic­ip ­ at­ing dis­ease recur­rence.
The role of main­te­nance either maintained or increased MRD−
response rates. Indeed, two-thirds of MRD− responses were pre­ Can we use MRD to mod­u­late treat­ment?
served through­out main­te­nance (G, 67.0%; R, 63.2%) with no The first attempts to use MRD as a tool to guide ther­a­peu­tic choic-
dif­fer­ence between the 2 arms in the rate of con­ver­sion to MRD+ es were pro­vided in advanced FL, where R could con­vert MRD+
(6.3% vs 6.1%, respec­tively). Furthermore, within MRD+ patients after CHOP to MRD−, and in local­ized FL after RT.13,24 Two recent
at EOI, 92% of 24 patients in the G-chemo and 78% of 46 in the large pro­spec­tive Ital­ian Fondazione Italiana Linfomi (FIL) tri­als ex-
R-chemo arm became MRD− in the first months of main­te­nance; plored the use of MRD to mod­u­late treat­ment in FL.32,33 FOLL12
the few cases who were never MRD− rap­idly progressed. (ii) was a phase 3 trial designed to prove the fea­si­bil­ity of a postin-
For the first time, MRD was assessed not only at EOI but also duction mod­u­la­tion of the stan­dard 2-year R main­te­nance accord-
at the mid­dle of induc­tion (MI). PFS was sig­nif­i­cantly lon­ger for ing to met­a­bolic response (Deauville score 1-3) and MRD response
PB MRD− vs MRD+ at MI (P < .0001), with early respond­ers (MI at EOI after chemoimmunotherapy in advanced FL. In the exper­i­
MRD−/EOI MRD−) hav­ing the best prog­no­sis. men­tal arm, PET−/MRD− patients (29%) under­went obser­va­tion,
In the Relevance trial, MRD− or MRD+ patients in the PB and/or PET−/MRD+ patients (4%) received repeated weekly R, and PET+
BM at EOI had a 3-year PFS of 84% and 55%, respec­tively, being sig­ patients had 1 dose of ibritumomab tiuxetan followed by stan­
nif­i­cant only in the BM and in the R-chemo but not in the R2 arm.22 dard R main­te­nance. The exper­i­men­tal arm showed a sig­nif­i­cantly
Even in the relapse/refrac­tory set­ting, the GADOLIN trial infe­rior PFS com­pared with the stan­dard 2-year R main­te­nance,
showed that MI MRD in the PB and/or BM was sig­nif­i­cantly dif­fer­ par­tic­u­larly for PET−/MRD− patients.32 It appears that a sin­gle
ent between the G-B and the B arm. In MVA, MRD sta­tus at EOI molec­u­lar response eval­u­a­tion at EOI is prob­a­bly not suf­fi­cient to
(85.7% G-B vs 55% B-arm), treat­ment arm and FLIPI sta­tus were indi­cate dis­ease erad­i­ca­tion and to deintensify treat­ment. Results
strongly pre­dic­tive of PFS and, to a lesser extent, of OS.23 of MRD mon­i­tor­ing will be shown at this meet­ing by Ladetto et al
In local­ized FL, a monocentric expe­ ri­
ence dem­ on­ strated (Abstract submission #146773).
that after treat­ment with local 24 to 30 Gy radiotherapy (RT), The MIRO’ (Molecularly Immuno-Radio-therapy Oriented,
dis­ap­pear­ance of cir­cu­lat­ing BCL2/IGH+ cells in the PB and/or EUDRACT 2012-001676-11) trial for local­ ized FL used MRD to
BM occurred in 50% (n  =  20/40) of patients.24 Additional treat­ guide post-RT treat­ment.33 Thirty per­cent of patients had cir­
ment with R in MRD+ patients after RT or who had molec­u­larly cu­lat­ing BCL2/IGH+ cells,25,33 18 MRD+ patients after RT (60%)
relapsed dur­ing the fol­low-up achieved a molec­u­lar CR in 84% of and 8 MRD+ dur­ing fol­low-up received ofatumumab. With this
cases: the 82-month PFS of MRD+ R-treated patients was sig­nif­i­ strat­egy, 91.7% of patients achieved a molec­ul­ar CR, with a sug-
cantly bet­ter than his­tor­i­cal con­trols, and the relapse prob­a­bil­ity gested clin­i­cal ben­e­fit after 38 months of fol­low-up.
was sig­nif­i­cantly lower in MRD− patients after RT or after R than The main lim­i­ta­tion in the intro­duc­tion of MRD mon­i­tor­ing in
that of patients remaining MRD+.24 the man­age­ment of patients with FL, espe­cially if con­sid­er­ing
Final com­ment. MRD neg­a­tiv­ity is pre­dic­tive of a bet­ter PFS only the BCL2/IGH rearrangement, is represented by the lack of
in all­clin­i­cal tri­als conducted in the past 2 decades, even in a molec­u­lar tar­get in a nota­ble pro­por­tion of cases. It is hoped
relapsed patients, and pos­si­bly of a lon­ger sur­vival in stud­ies that tech­ni­cal advance­ments cur­rently under inves­ti­ga­tion will
with a prolonged fol­low-up. Assessment of MRD at ear­lier time over­come these lim­i­ta­tions (Figure 2, Tables 2-3).34,35-43
points with respect to EOI can also be infor­ma­tive. Final com­ment. R main­te­nance holds and increases the rates
of MRD neg­a­tiv­ity. Two recent tri­als (FOLL12 and MIRO’) explored
Is the prolonged molec­u­lar fol­low-up of clin­i­cal rel­e­vance? MRD-driven mod­u­la­tion of the postinduction ther­apy in FL, with
The prog­nos­tic value of sequen­tial BCL2/IGH assess­ment to pre­ treat­ment inten­si­fi­ca­tion or deintensification in MRD+ and MRD−
dict clin­ic
­ al relapse is supported by lim­ited evi­dences and often patients at the EOI, respec­tively. Final results are awaited.

326  |  Hematology 2021  |  ASH Education Program


Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/320/1852135/320giudice.pdf by guest on 13 December 2021
Figure 2. Standard and new techniques to monitor MRD in FL.

Table 2. Principles of stan­dard and next-gen­er­a­tion tech­nol­o­gies in MRD mon­i­tor­ing

Technology References Potential advan­tages and open issues


RQ-PCR 1,35 The most val­i­dated and stan­dard­ized quan­ti­ta­tive method for MRD detec­tion in FL. Despite remark­able sen­si­tiv­ity and
spec­i­fic­ity, RQ-PCR has nota­ble lim­i­ta­tions due to the lack of BCL2/IGH tar­get in about 40% of advanced FL and 65%
to 70% of local­ized FL, cur­rently not eli­gi­ble for MRD assess­ment, and to the chal­lenge of very low MRD lev­els, where
it is dif­fi­cult to dis­sect if the sig­nal observed by PCR (not quan­ti­fi­able) is due to few resid­ual leu­ke­mic cells or to a
non­spe­cific ampli­fi­ca­tion of nor­mal DNA.
NGS 34,36 Potential deeper sen­si­tiv­ity com­pared with the clas­sic meth­ods.
A sen­si­tiv­ity of 10−6 is achiev­able only when high amounts of DNA are used.
Broad spec­trum of iden­ti­fi­able molec­u­lar tar­gets: a “cap­ture-based” pro­to­col cov­er­ing the cod­ing V, D, J genes of the
IGH loci was capa­ble of detecting clonal rearrangements in 87% (21/24) of lymphoproliferative dis­or­ders.
TLA 37,38 Alternative NGS tool capa­ble of sequenc­ing struc­tural var­i­ants, usu­ally not detected by con­ven­tional PCR approaches,
thanks to the selec­tive ampli­fi­ca­tion and sequenc­ing of entire genes on the basis of the cross-linking of phys­i­cally
prox­i­mal DNA loci.
ddPCR 39,40 A third-gen­er­a­tion quan­ti­ta­tive method based on the par­ti­tion of the tem­plate DNA into water-in-oil drop­lets in which
PCR ampli­fi­ca­tion occurs, allowing the quan­ti­fi­ca­tion of nucleic acid tar­gets with­out the need of the cal­i­bra­tion
curve.
Sensitivity, accu­racy, and repro­duc­ibil­ity at least com­pa­ra­ble to that of RQ-PCR.
A good per­for­mance in the MRD quan­ti­fi­ca­tion in at least 20% to 30% of sam­ples resulting pos­i­tive but not quan­ti­fi­able
by RQ-PCR.
cfDNA 41-43 Plasma is a poten­tially impor­tant source of DNA (ie, cfDNA), poten­tially use­ful to iden­tify dis­tinct bio­log­i­cal sub­types
of lym­pho­mas and to pro­vide insights into the pat­terns of geno­mic evo­lu­tion/resis­tance through­out treat­ment in all­
com­part­ments, not only PB and BM.
A non­in­va­sive tool to mon­i­tor MRD in non-Hodgkin lym­phoma through patient-spe­cific IGH rearrangements, to iden­tify
indi­vid­u­als at an increased risk of relapse and to detect relapse before clin­i­cal evi­dence of dis­ease.
Most cfDNA orig­i­na­tes from leu­ko­cytes, and only a small frac­tion (<10%) is tumor derived, known as ctDNA. ctDNA
con­cen­tra­tion varies among patients and dif­fers according to the type, loca­tion, and stage of can­cer, with some
pro­duc­ing extremely low con­cen­tra­tions. Thus, it is unlikely that cfDNA will reach the sen­si­tiv­ity of MRD anal­y­sis on
cir­cu­lat­ing cells in FL. Before broad clin­i­cal implementation, issues on preanalytical fac­tors must be addressed in
order to achieve con­sis­tent and repro­duc­ible results.
A major stan­dard­i­za­tion effort is under way within the EuroClonality (https:​­/​­/www​­.euroclonalityngs​­.org​­/usr​­/pub​­/pub​­.php) and EuroMRD Consortium
(www​­.euromrd​­.org), to estab­lish guide­lines for NGS and ddPCR MRD anal­y­sis and their future appli­ca­tion in stan­dard clin­i­cal prac­tice.
cfDNA, cell-free DNA; NGS, next-gen­er­a­tion sequenc­ing; TLA, tar­get locus ampli­fi­ca­tion.
Prakash Singh Shekhawat
Minimal resid­ual dis­ease in indo­lent NHL  |  327
Table 3. First appli­ca­tions of next-gen­er­a­tion tech­nol­o­gies to molec­u­lar mon­i­tor­ing in FL

Study Patients Marker/tis­sue/tim­ing Method Potential impact


Genuardi et al38 20 FL with no MBR BCL2/TLA TLA BCL2/TLA in 8 (40%) of “marker-neg­a­tive” cases
or mcr BM The new BCL2/TLA mark­ers were suit­able for RQ-PCR MRD
At diag­no­sis and for MRD anal­y­sis in 4 of 5 cases.
MRD by BCL2-TLA reached good sen­si­tiv­ity lev­els.
Cavalli et al40 67 early stage FL BCL2/IGH ddPCR Concordance between ddPCR and RQ-PCR: 82%
PB, BM ddPCR iden­ti­fied a MBR marker in 8 of 18 (44%) sam­ples that
At diag­no­sis and for MRD resulted in MBR−/mcr− by qual­it­ a­tive nested PCR.
Molecular tumor bur­den at diag­no­sis ≥10−5 sig­nif­i­cantly
predicted PFS only when quan­ti­fied by ddPCR but not by
RQ-PCR.
Higher sen­si­tiv­ity of ddPCR in RQ-PCR PNQ sam­ples.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/320/1852135/320giudice.pdf by guest on 13 December 2021


Sarkozy et al42 34 FL from PRIMA trial IGH NGS 29 (85%) had 1 or more tumor clonotypes in the tumor
Tumor biopsy biopsy spec­i­men.
Plasma 25 (74%) had 1 or more tumor clonotypes in plasma.
At diag­no­sis 18 of 24 (75%) patients with an IGH clonotype had sev­eral
detect­able subclones in the tumor or in the plasma.
13 of 24 (54%) showed a subclone detected in both the
plasma and the tumor.
>50% of cases showed a dif­fer­ent dis­tri­bu­tion of subclones
between tumor and plasma.
High ctDNA lev­els at diag­no­sis predicted short PFS in MVA.
Delfau-Larue et al43 FL PET TMTV (n = 133) ddPCR 23 of 68 cfDNA were BCL2/IGH+ (ctDNA ≤10% cfDNA).
BCL2/IGH High cor­re­la­tion between CTCs and TMTV and between
PB CTC (n = 68) cfDNA and TMTV
PB cfDNA (n = 61) CTCs pre­dic­tive of out­come in uni­var­i­ate anal­y­sis but not in
At diag­no­sis MVA
Total cfDNA lev­els and TMTM are inde­pen­dent pre­dic­tors of
out­come.
CTC, cir­cu­lat­ing tumor cell; PNQ , positive not quantifiable; TMTV, total met­a­bolic tumor vol­ume.

CLINICAL CASES (Con­t in­u ed) na­tive to BM for MYD88L265P detec­tion.44 MYD88L265P detec­tion
Patient 1. The per­sis­tence of MRD pos­it­iv­ity dur­ing the 2-year in the cere­bral spi­nal fluid by ddPCR is also use­ful to diag­nose
treat­ment despite a PET neg­ a­
tiv­
ity sug­ gests a resis­
tance Bing-Neel syn­drome.46
to R-bendamustine treat­ ment. Indeed, the patient quickly Promising results have been pre­lim­i­nar­ily shown in splenic mar­
relapsed. In such patients, a pos­si­ble inten­si­fi­ca­tion/switch of ginal zone lym­phoma, where MRD has been assessed in BM and
treat­ment could be explored in clin­ic ­ al tri­als. PB by ddPCR using IGH allele-spe­cific oli­go­nu­cle­o­tide prim­ers in
Patient 2. Although in this local­ized FL, clin­i­cal relapse was the phase 2 BRISMA/IELSG36 (Bendamustine-rituximab as first-
antic­i­pated by a molec­u­lar con­ver­sion to MRD pos­i­tiv­ity, this is line treatment of splenic marginal zone lymphoma/International
not always the case with the cur­rent MRD mon­i­tor­ing tim­ing and Extranodal Lymphoma Study Group) trial.45
modal­i­ties. The clin­i­cal ben­e­fit of adding an anti-CD20 mono­clo­
nal anti­body to RT in local­ized FL or to treat molec­u­lar relapses Concluding remarks
needs to be addressed in well-designed clin­i­cal tri­als. Despite sev­eral decades of research, MRD anal­y­sis in FL has not
yet entered the day-to-day clin­ i­
cal prac­tice. Indeed, out­
side
of clin­i­cal tri­als, MRD results should not be used to take clin­i­cal
deci­sions in the real-life man­age­ment of patients with FL. The is-
Other iNHLs: lymphoplasmacytic lym­phoma/ sue of BCL2/IGH+ non­ma­lig­nant B cells that can be found in the
Waldenström mac­ro­glob­u­li­ne­mia and mar­ginal PB of healthy indi­vid­u­als47 is mar­ginal in treated patients with FL,
zone lym­phoma because the emer­gence of a BCL2/IGH clone unre­lated to the
Although the role of MRD mon­i­tor­ing in FL is pro­gres­sively dis­ease is very unlikely and rarely requires the need of prov­ing
increas­ing, MRD in other iNHLs, such as lymphoplasmacytic lym­ the sequence iden­tity of the rearrangement.
phoma/Waldenström mac­ro­glob­u­li­ne­mia (WM) and mar­ginal To move MRD in FL from clin­i­cal tri­als to daily prac­tice, fur­
zone lym­ phoma, is only recently starting to be explored.44,45 ther stud­ies are needed to over­come the limit represented by
In WM, MYD88L265P is a diag­nos­tic and pre­dic­tive bio­marker of the lack of a molec­u­lar marker for all­patients; to estab­lish an
ther­apy response. Besides allele-spe­cific RQ-PCR, ddPCR has re- inte­grated risk strat­i­fi­ca­tion; to dem­on­strate whether MRD anal­
cently proved to be a suit­able and sen­si­tive tool for MYD88L265P y­sis can be inte­grated with PET for a refined def­i­ni­tion of “poor
screen­ing and MRD mon­i­tor­ing.44 Both unsorted BM and PB sam­ respond­ers,” can­di­dates to exper­i­men­tal approaches; to explore
ples can be reli­ably tested, as well as cir­cu­lat­ing tumor DNA other MRD-adapted treat­ment modal­i­ties, pos­si­bly with kinetic
(ctDNA), which rep­re­sents an attrac­tive and less inva­sive alter­ mod­els of MRD anal­y­sis; and to eval­ua ­ te if chemo-free com­bi­

328  |  Hematology 2021  |  ASH Education Program


na­tions have an advan­tage in terms of MRD achieve­ment com­ train­ing and val­i­da­tion anal­y­sis in three inter­na­tional cohorts. Lancet
pared with chemoimmunotherapy. Results from new-gen­er­a­tion Oncol. 2018;19(4):549-561.
11. Huet S, Sujobert P, Salles G. From genet­ics to the clinic: a trans­la­tional per­
clin­i­cal tri­als (eg, FIL FOLL12 and Gallium) are eagerly awaited. spec­tive on fol­lic­u­lar lym­phoma. Nat Rev Cancer. 2018;18(4):224-239.
Data are mature to design MRD-driven clin­i­cal tri­als that incor­ 12. Gribben JG, Freedman AS, Neuberg D, et al. Immunologic purg­ing of mar­
po­rate the molec­u­lar mon­i­tor­ing in the man­age­ment of patients row assessed by PCR before autol­o­gous bone mar­row trans­plan­ta­tion for
with FL. B-cell lym­phoma. N Engl J Med. 1991;325(22):1525-1533.
13. Rambaldi A, Lazzari M, Manzoni C, et  al. Monitoring of min­i­mal ­resid­ual
­dis­ease after CHOP and rituximab in pre­vi­ously untreated patients with
Acknowledgments
fol­lic­u­lar lym­phoma. Blood. 2002;99(3):856-862.
Work partly supported by Associazione Italiana per la Ricerca sul 14. Rambaldi A, Carlotti E, Oldani E, et al. Quantitative PCR of bone mar­row
Cancro (AIRC), Metastases 5   x   1000 Special Program, N° 21198, BCL2/IgH+ cells at diag­no­sis pre­dicts treat­ment response and long-term
Milan, Italy (Robin Foà). The authors thank Dr. Marco Ladetto and out­come in fol­lic­u­lar non-Hodgkin lym­phoma. Blood. 2005;105(9):3428-
the FIL MRD net­work. 3433.
15. Ladetto M, De Marco F, Benedetti F, et al; Gruppo Italiano Trapianto di
Midollo Osseo (GITMO); Intergruppo Italiano Linfomi (IIL). Prospective, mul­
Conflict-of-inter­est dis­clo­sure

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/320/1852135/320giudice.pdf by guest on 13 December 2021


ti­cen­ter ran­dom­ized GITMO/IIL trial com­par­ing inten­sive (R-HDS) ver­sus
Ilaria Del Giudice: AstraZeneca, Tolero (advi­sory board). con­ven­tional (CHOP-R) chemoimmunotherapy in high-risk fol­lic­u­lar lym­
Irene Della Starza: no conflicts to disclose. phoma at diag­no­sis: the supe­rior dis­ease con­trol of R-HDS does not trans­
Robin Foà: hon­ o­raria from Janssen, Amgen, Novartis, Incyte, late into an over­all sur­vival advan­tage. Blood. 2008;111(8):4004-4013.
16. Bruna R, Benedetti F, Boccomini C, et al. Prolonged sur­vival in the absence
­Servier, and Sanofi. of dis­ease-recur­rence in advanced-stage fol­lic­u­lar lym­phoma fol­low­ing
chemo-immu­no­ther­apy: 13-year update of the pro­spec­tive, mul­ti­cen­ter
Off-label drug use ran­dom­ized GITMO-IIL trial. Haematologica. 2019;104(11):2241-2248.
Ilaria Del Giudice: nothing to disclose. 17. Ladetto M, Lobetti-Bodoni C, Mantoan B, et al; Fondazione Italiana Linfomi.
Irene Della Starza: nothing to disclose. Persistence of min­i­mal resid­ual dis­ease in bone mar­row pre­dicts out­come
in fol­lic­u­lar lym­pho­mas treated with a rituximab-inten­sive pro­gram. Blood.
Robin Foà: nothing to disclose. 2013;122(23):3759-3766.
18. Galimberti S, Luminari S, Ciabatti E, et  al. Minimal resid­ual dis­ease after
Correspondence con­ven­tional treat­ment sig­nif­i­cantly impacts on pro­gres­sion-free sur­vival
Robin Foà, Hematology, Department of Translational and Pre- of patients with fol­lic­u­lar lym­phoma: the FIL FOLL05 trial. Clin Cancer Res.
cision Medicine, Sapienza University, Via Benevento 6, 00161 2014;20(24):6398-6405.
19. Zohren F, Bruns I, Pechtel S, et  al. Prognostic value of cir­cu­lat­ing Bcl-2/
Rome, Italy; e-mail: rfoa@bce​­.uniroma1​­.it IgH lev­els in patients with fol­lic­u­lar lym­phoma receiv­ing first-line immu-
nochemotherapy. Blood. 2015;126(12):1407-1414.
References 20. Pott C, Hoster E, Kehden B, et al. Minimal resid­ual dis­ease in patients with
1. Pott C, Brüggemann M, Ritgen M, van der Velden VHJ, van Dongen JJM, fol­lic­u­lar lym­phoma treated with obinutuzumab or rituximab as first-line
Kneba M. MRD detec­tion in B-cell non-Hodgkin lym­pho­mas using Ig gene induc­tion immunochemotherapy and main­te­nance in the phase 3 GALLIUM
rearrangements and chro­mo­somal trans­lo­ca­tions as tar­gets for real-time study. Blood. 2016;128(22):613.
quan­ti­ta­tive PCR. Methods Mol Biol. 2019;1956:199-228. 21. Pott C, Hoster E, Kehden B, et al. Minimal resid­ual dis­ease response at end
2. Galimberti S, Genuardi E, Mazziotta F, et al. The min­i­mal resid­ual dis­ease of induc­tion and dur­ing main­te­nance cor­re­lates with updated out­come in
in non-Hodgkin’s lym­pho­mas: from the lab­o­ra­tory to the clin­i­cal prac­tice. the phase III GALLIUM study of obinutuzumab- or rituximab-based immu-
Front Oncol. 2019;9:528. nochemotherapy in pre­vi­ously untreated fol­lic­u­lar lym­phoma patients.
3. Grimaldi D, Genuardi E, Ferrante M, Ferrero S, Ladetto M. Minimal resid­ual Blood. 2018; 132(suppl 1):396.
dis­ease in indo­lent lym­pho­mas: a crit­i­cal assess­ment. Curr Treat Options 22. Delfau-Larue M-H, Boulland M-L, Beldi-Ferchiou A, et  al. Lenalidomide/
Oncol. 2018;19(12):71. rituximab induces high molec­u­lar response in untreated fol­lic­u­lar lym­phoma:
4. Sarkozy C, Maurer MJ, Link BK, et al. Cause of death in fol­lic­u­lar lym­phoma LYSA ancil­lary RELEVANCE study. Blood Adv. 2020;4(14):3217-3223.
in the first decade of the rituximab era: a pooled anal­y­sis of French and US 23. Pott C, Sehn LH, Belada D, et  al. MRD response in relapsed/refrac­tory
cohorts. J Clin Oncol. 2019;37(2):144-152. FL after obinutuzumab plus bendamustine or bendamustine alone in the
5. Casulo C, Byrtek M, Dawson KL, et al. Early relapse of fol­lic­u­lar lym­phoma ­GADOLIN trial. Leukemia. 2020;34(2):522-532.
after rituximab plus cyclo­phos­pha­mide, doxo­ru­bi­cin, vin­cris­tine, and pred­ 24. Pulsoni A, Della Starza I, Cappelli LV, et al. Minimal resid­ual dis­ease monitor-
ni­sone defi­nes patients at high risk for death: an anal­y­sis from the National ing in early stage fol­lic­u­lar lym­phoma can pre­dict prog­no­sis and drive treat­
LymphoCare Study. J Clin Oncol. 2015;33(23):2516-2522. ment with rituximab after radio­ther­apy. Br J Haematol. 2020;188(2):249-258.
6. Federico M, Caballero Barrigón MD, Marcheselli L, et al. Aristotle Consor- 25. Herfarth K, Borchmann P, Schnaidt S, et al. Rituximab with involved field
tium. Rituximab and the risk of trans­for­ma­tion of fol­lic­u­lar lym­phoma: a irra­di­a­tion for early-stage nodal fol­lic­u­lar lym­phoma: results of the MIR
ret­ro­spec­tive pooled anal­y­sis. Lancet Haematol. 2018;5(8):e359-e367. study. HemaSphere. 2018;2(6):e160.
7. Dreyling M, Ghielmini M, Rule S, et al; ESMO Guidelines Committee. Newly 26. Pulsoni A, Starza ID, Frattarelli N, et  al. Stage I/II fol­ lic­
u­lar lym­ phoma:
diag­nosed and relapsed fol­lic­u­lar lym­phoma: ESMO Clinical Practice Guide- spread of BCL2/IGH+ cells in blood and bone mar­row from pri­mary site of
lines for diag­no­sis, treat­ment and fol­low-up. Ann Oncol. 2021;32(3):298- dis­ease and pos­si­bil­ity of clear­ance after involved field radio­ther­apy. Br J
308. Haematol. 2007;137(3):216-220.
8. Mir F, Mattiello F, Grigg A, et  al. Follicular Lymphoma Evaluation Index 27. Hirt C, Schüler F, Kiefer T, et  al. Rapid and sustained clear­ance of cir­cu­
(FLEX): a new clin­i­cal prog­nos­tic model that is supe­rior to existing risk lat­ing lym­phoma cells after che­mo­ther­apy plus rituximab: clin­i­cal sig­nif­i­
scores for predicting pro­gres­sion-free sur­vival and early treat­ment fail­ure cance of quan­ti­ta­tive t(14;18) PCR mon­i­tor­ing in advanced stage fol­lic­u­lar
after front­ line immunochemotherapy. Am J Hematol. 2020;95(12):1503- lym­phoma patients. Br J Haematol. 2008;141(5):631-640.
1510. 28. Boccomini C, Ladetto M, Rigacci L, et al. A brief rituximab, bendamustine,
9. Pastore A, Jurinovic V, Kridel R, et al. Integration of gene muta­tions in risk mitoxantrone (R-BM) induc­ tion followed by rituximab con­ sol­i­
da­
tion in
prog­nos­ti­ca­tion for patients receiv­ing first-line immunochemotherapy for elderly patients with advanced fol­lic­u­lar lym­phoma: a phase II study by
fol­lic­u­lar lym­phoma: a ret­ro­spec­tive anal­y­sis of a pro­spec­tive clin­i­cal trial the Fondazione Italiana Linfomi (FIL). Br J Haematol. 2021;193(2):280-289.
and val­i­da­tion in a pop­u­la­tion-based reg­is­try. Lancet Oncol. 2015;16(9):1111- 29. Luminari S, Galimberti S, Versari A, et  al. Positron emis­sion tomog­ra­phy
1122. response and min­i­mal resid­ual dis­ease impact on pro­gres­sion-free sur­vival
10. Huet S, Tesson B, Jais J-P, et al. A gene-expres­sion pro­fil­ing score for pre­ in patients with fol­lic­u­lar lym­phoma: a sub­set anal­y­sis from the FOLL05 trial
dic­tion of out­come in patients with fol­lic­u­lar lym­phoma: a ret­ro­spec­tive of the Fondazione Italiana Linfomi. Haematologica. 2016;101(2):e66-e68.

Prakash Singh Shekhawat


Minimal resid­ual dis­ease in indo­lent NHL  |  329
30. Pott C, Davies A, Hiddemann W, et al. Metabolic (PET) and MRD response 40. Cavalli M, De Novi LA, Della Starza I, et al. Comparative anal­y­sis between
con­fer reduced risk of pro­gres­sion or death in patients treated within the RQ-PCR and dig­i­tal drop­let PCR of BCL2/IGH gene rearrangement in the
phase III Gallium study. HemaSphere. 2018;2(suppl 1):171-172. periph­eral blood and bone mar­row of early stage fol­lic­ul­ar lym­phoma. Br J
31. Bishton MJ, Rule S, Wilson W, et  al. The UK NCRI study of chlorambucil, Haematol. 2017;177(4):588-596.
mitoxantrone and dexa­meth­as­ one (CMD) ver­sus fludarabine, mitoxantrone 41. Lakhotia R, Roschewski M. Circulating tumour DNA in B-cell lym­pho­mas:
and dexa­meth­a­sone (FMD) for untreated advanced stage fol­lic­u­lar lym­ cur­rent state and future pros­pects. Br J Haematol. 2021;193(5):867-881.
phoma: molec­u­lar response strongly pre­dicts prolonged over­all sur­vival. 42. Sarkozy C, Huet S, Carlton VEH, et  al. The prog­ nos­tic value of clonal
Br J Haematol. 2020;190(4):545-554. ­het­ero­ge­ne­ity and quan­ti­ta­tive assess­ment of plasma cir­cu­lat­ing clonal
32. Luminari S, Galimberti, S, Versari A, et al. Response adapted post induc­tion IG-VDJ sequences at diag­no­sis in patients with fol­lic­u­lar lym­phoma. Onco­
ther­apy in fol­lic­u­lar lym­phoma: updated results of the FOLL12 trial by the target. 2017;8(5):8765-8774.
Fondazione Italiana Linfomi (FIL). Hematol Oncol. 2021;39(suppl 2):133. 43. Delfau-Larue MH, van der Gucht A, Dupuis J, et al. Total met­a­bolic tumor
33. Pulsoni A, Tosti ME, Ferrero S, et  al. Early-stage fol­lic­u­lar lym­phoma: first vol­ume, cir­cu­lat­ing tumor cells, cell-free DNA: dis­tinct prog­nos­tic value in
results of the FIL “Miro” study, a mul­ti­cen­ter phase II trial com­bin­ing local fol­lic­u­lar lym­phoma. Blood Adv. 2018;2(7):807-816.
radio­ther­apy and MRD-driven immu­no­ther­apy. Blood. 2019;134(suppl 1):124. 44. Drandi D, Genuardi E, Dogliotti I, et al. Highly sen­si­tive MYD88L265P muta­
34. Ladetto M, Brüggemann M, Monitillo L, et al. Next-gen­er­a­tion sequenc­ing tion detec­tion by drop­let dig­i­tal poly­mer­ase chain reac­tion in Walden-
and real-time quan­ti­ta­tive PCR for min­i­mal resid­ual dis­ease detec­tion in ström mac­ro­glob­u­li­ne­mia. Haematologica. 2018;103(6):1029-1037.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/320/1852135/320giudice.pdf by guest on 13 December 2021


B-cell dis­or­ders. Leukemia. 2014;28(6):1299-1307. 45. Ferrero S, Ladetto M, Beldjord K, et al. First appli­ca­tion of min­i­mal resid­
35. Della Starza I, Cavalli M, De Novi LA, et al. Minimal resid­ual dis­ease (MRD) in ual dis­ease anal­y­sis in splenic mar­ginal zone lym­phoma tri­als: results from
non-Hodgkin lym­pho­mas: Interlaboratory repro­duc­ibil­ity on mar­row sam­ BRISMA/IELSG36 phase II study. Hematol Oncol. 2019;37(52):224-225.
ples with very low lev­els of dis­ease within the FIL (Fondazione Italiana Lin- 46. Hiemcke-Jiwa LS, Minnema MC, Radersma-van Loon JH, et  al. The use
fomi) MRD Network. Hematol Oncol. 2019;37(4):368-374. of drop­ let dig­i­
tal PCR in liq­uid biop­sies: a highly sen­ si­
tive tech­nique
36. Wren D, Walker BA, Brüggemann M, et  al. Comprehensive trans­ lo­
ca­ for MYD88 p.(L265P) detec­ tion in cere­bro­spi­
nal fluid. Hematol Oncol.
tion and clonality detec­ tion in lymphoproliferative dis­ or­
ders by next- 2018;36(2):429-435.
gen­er­a­tion sequenc­ing. Haematologica. 2017;102:e57-e60. 47. Roulland S, Lebailly P, Lecluse Y, et al. Long-term clonal per­sis­tence and
37. de Vree PJ, de Wit E, Yilmaz M, et al. Targeted sequenc­ing by prox­im­ity evo­ lu­tion of t(14;18)-bear­ ing B cells in healthy indi­ vid­
u­als. Leukemia.
liga­tion for com­pre­hen­sive var­i­ant detec­tion and local haplotyping. Nat 2006;20(1):158-162.
Biotechnol. 2014;32(10):1019-1025.
38. Genuardi E, Klous P, Mantoan B, et  al. Targeted locus ampli­fic ­ a­tion to
detect molec­u­lar mark­ers in man­tle cell and fol­lic­u­lar lym­phoma. Hematol
Oncol. 2021;39(3):293-303.
39. Drandi D, Kubiczkova-Besse L, Ferrero S, et  al. Minimal resid­ual dis­ease
detec­tion by drop­let dig­i­tal PCR in mul­ti­ple mye­loma, man­tle cell lym­
phoma, and fol­lic­u­lar lym­phoma: a com­par­i­son with real-time PCR. J Mol © 2021 by The Amer­i­can Society of Hematology
Diagn. 2015;17(6):652-660. DOI 10.1182/hema­tol­ogy.2021000312

330  |  Hematology 2021  |  ASH Education Program


INHERITED RED CELL DISORDERS BEYOND HEMOGLOBINOPATHIES

Diagnosis and clinical management of red cell


membrane disorders

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/331/1851848/331kalfa.pdf by guest on 13 December 2021


Theodosia A. Kalfa
Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH; and Department of Pediatrics, University of
Cincinnati College of Medicine, Cincinnati, OH

Heterogeneous red blood cell (RBC) membrane disorders and hydration defects often present with the common clinical
findings of hemolytic anemia, but they may require substantially different management, based on their pathophysiology.
An accurate and timely diagnosis is essential to avoid inappropriate interventions and prevent complications. Advances in
genetic testing availability within the last decade, combined with extensive foundational knowledge on RBC membrane
structure and function, now facilitate the correct diagnosis in patients with a variety of hereditary hemolytic anemias
(HHAs). Studies in patient cohorts with well-defined genetic diagnoses have revealed complications such as iron overload
in hereditary xerocytosis, which is amenable to monitoring, prevention, and treatment, and demonstrated that splenec-
tomy is not always an effective or safe treatment for any patient with HHA. However, a multitude of variants of unknown
clinical significance have been discovered by genetic evaluation, requiring interpretation by thorough phenotypic assess-
ment in clinical and/or research laboratories. Here we discuss genotype-phenotype correlations and corresponding clini-
cal management in patients with RBC membranopathies and propose an algorithm for the laboratory workup of patients
presenting with symptoms and signs of hemolytic anemia, with a clinical case that exemplifies such a workup.

LEARNING OBJECTIVES
• Discuss comprehensive evaluation for RBC membrane disorders, including phenotypic and genetic testing
• Review clinical management considerations tailored to the genetic diagnosis and pathophysiology of RBC
membrane disorders
• Elaborate on patient cases of RBC membrane skeleton disorders and hydration defects

the amount of normal α-spectrin produced from the affected


CLINICAL CASE allele. Compound heterozygosity of αLEPRA with a null SPTA1
A European American boy presented with nonimmune mutation in trans is the most common cause of autoso-
hemolyticanemia and hyperbilirubinemia as a neonate, mal recessive (AR) hereditary spherocytosis (HS) due to α-
requiring red blood cell (RBC) transfusion and phototherapy. spectrin deficiency.1-3 In addition, a variant of unknown clini-
He continued to require frequent transfusions every 4 to cal significance (VUCS) was identified in PIEZO1: c.6205G>A
8 weeks. There was no family history of hemolytic anemia. (p.Val2069Met), which, if pathogenic, could cause a compo-
His mother had some spherocytes on her blood smear, but nent of xerocytosis in this patient. Splenectomy in hereditary
she was asymptomatic with no evidence of hemolysis. Since xerocytosis (HX) is associated with life-threatening thrombo-
the patient had been regularly transfused since birth, an RBC philia; therefore, it was important to explore the contribu-
phenotypic evaluation was not feasible to provide a diag- tion of this PIEZO1 VUCS to the patient’s disease.
nosis. Therefore, genetic evaluation on a next-generation Parental studies were performed and confirmed that the
sequencing (NGS) panel of genes associated with hereditary SPTA1 mutations were positioned in trans and were there-
hemolytic anemia (HHA)was performed that showed a non- fore pathogenic, causing HS. The father carried αLEPRA and
sense SPTA1 mutation (c.4295del, p.Leu1432*)and the low- the PIEZO1 p.Val2069Met variant while the mother carried
expression SPTA1 polymorphism c.4339-99C>T (also known the nonsense SPTA1 mutation p.Leu1432*. The patient had
as αLEPRA or low expression PRAgue). αLEPRA, a deep intronic osmotic gradient ektacytometry performed 8 weeks after
variant that affects splicing, causes a significant decrease in a transfusion that showed decreased EImax (maximum
Prakash Singh Shekhawat
Red cell membrane disorders | 331
elon­ga­tion index; ie, the max­im
­ um deformability of the RBCs), car­ry­ing the PIEZO1 VUCS. RBC cat­ion con­tent was eval­u­ated for
com­pat­i­ble with HS but not typ­i­cal since Omin (ie, the hypo­tonic both par­ents and was within nor­mal range, offer­ing addi­tional
osmo­lal­ity in which the elon­ga­tion index is min­i­mal) and Ohyp reas­sur­ance that the PIEZO1 p.Val2069Met var­i­ant was benign.
(ie, the osmo­lal­ity value at which the cells’ aver­age max­i­mum The child con­tin­ued with chronic trans­fu­sions and che­la­tion,
diam­e­ter is half of EImax) were nor­mal because nor­mal donor but his iron over­load appeared to be poorly con­trolled, and there­
RBCs were still pres­ent in his blood (Figure 1A).4 Both par­ents fore a par­tial sple­nec­tomy was performed at 4 years of age, at least
had a nor­mal hemo­glo­bin (Hgb) and retic­ul­o­cyte count as well 2 weeks after pneu­mo­coc­cal and menin­go­coc­cal vac­ci­na­tions
as nor­mal ektacytometry. Each nor­ mal SPTA1 gene pro­ duces were admin­is­tered.5 He remained trans­fu­sion-free for 18 months
α-spectrin well in excess; there­fore, it was not sur­pris­ing that with an Hgb of 8.7 to 11 g/dL, but his ane­mia grad­u­ally wors­ened,
nei­ther par­ent had HS. In addi­tion, the ektacytometry curve of requir­ing fre­quent trans­fu­sions again. A total sple­nec­tomy was
the father’s blood sam­ple showed no evi­dence of HX despite performed at 7 years of age since the remaining splenic tis­sue had

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/331/1851848/331kalfa.pdf by guest on 13 December 2021


Figure 1. HS. (A) Ektacytometry of blood samples from the patient before and after splenectomy and from his parents. The patient had
AR HS due to compound heterozygosity of SPTA1 c.4295del (p.L1432*), shared with the mom, and SPTA1 c.4339-99C>T, shared with
the dad. The dad and the proband were also found to carry the PIEZO1 VUCS c.6205G>A (p.Val2069Met). The parents had a normal
ektacytometry with no evidence of xerocytosis for the father, offering reassurance that the PIEZO1 VUCS is benign, while the patient
after splenectomy had a typical HS curve. Before splenectomy, the patient was chronically transfused. Ektacytometry at that time
was performed at a nadir before the next transfusion and 2 months after a previous one. Transfused RBCs have obviously modified the
curve, giving a falsely normal Omin (ie, the hypotonic osmolality where the elongation index is minimal). The value of Omin provides
information on the initial surface-to-volume ratio of the cell sample. A shift to the right reflects a decrease in the surface area/volume
ratio and corresponds to increased osmotic fragility. (B) Blood smear of the patient about 18 months post partial splenectomy before
starting to require transfusions again, demonstrating significant anisopoikilocytosis and polychromasia as well as many spherocytes,
typical for AR SPTA1-associated HS. (C) Blood smear of the patient 5 years after total splenectomy, stable and without need of transfu-
sions since the time of that surgery. Moderate anisopoikilocytosis and several spherocytes are still noted. Scale bar = 14 mm. Details on
the ektacytometry assay and parameters can be found at https:​/​/www​.cincinnatichildrens​.org​/service​/c​/cancer​-blood​/hcp​/clinical​
-laboratories​/erythrocyte​-diagnostic​-lab​/ektacytometry.

332  |  Hematology 2021  |  ASH Education Program


impres­sively regrown to 435 g, based on the pathol­ogy report. to a hema­tol­o­gist because their Hgb does not improve with iron
The patient has had no fur­ther trans­fu­sion require­ment for the past sup­ple­men­ta­tion—one of the bright exam­ples of why a com­
6 years, with a Hgb now in the 12.7 to 16.7 g/dL range, an abso­lute plete blood count (CBC) for eval­u­a­tion of ane­mia should always
retic­u­lo­cyte count (ARC) of 80 to 150 × 103/µL, and a resolved iron be asso­ci­ated with a retic­u­lo­cyte count.
over­load. Ektacytometry revealed the typ­i­cal HS curve (Figure 1A); Spherocytes are pre­ma­turely destroyed in the spleen as they
a blood smear at 5.5 years of age (before the total sple­nec­tomy) are impeded in pass­ ing through the interendothelial sinu­ soi­
dem­ on­ strated many spherocytes and sig­ nif­i­
cant poikilocytosis dal slits and even­tu­ally phago­cy­tosed by the red pulp mac­ro­
and polychromasia, as expected with α-spectrin defi­ciency (Fig- phages. Therefore, sple­nec­tomy has been used for decades as
ure 1B).6 A recent blood smear, 5 years after the total sple­nec­tomy, the stan­dard of care for HS, with the aim of decreas­ing hemo­ly­
showed an improved RBC mor­phol­ogy, although still with many sis and improv­ing ane­mia. However, increased aware­ness of the
spherocytes and mod­er­ate poikilocytosis (Figure 1C). postsplenectomy increased risks of sep­sis; par­a­sitic infec­tions
of eryth­ro­cytes; and, pos­si­bly, increased thrombophilia and ath­
ero­scle­ro­sis due to the rise in cho­les­terol,15,16 at least par­tially
Hereditary spherocytosis (HS) due to ame­lio­ra­tion of the increased eryth­ro­poi­e­sis, along with

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/331/1851848/331kalfa.pdf by guest on 13 December 2021


Genotype/phe­no­type cor­re­la­tions accu­mu­la­tion of fol­low-up data on the effi­cacy of par­tial sple­nec­
HS, the most com­mon of the RBC mem­brane dis­or­ders, is caused tomy,17,18 has led to a more indi­vid­u­al­ized approach rec­og­niz­ing
by muta­tions in the genes SPTA1, SPTB, ANK1, EPB42, or SLC4A1, the options of total vs par­tial vs no sple­nec­tomy. Therefore, it
lead­ing to an RBC mem­brane skel­e­ton defi­cient in α- or β-spectrin, is cur­rently rec­og­nized that sple­nec­tomy is not needed for all­
ankyrin, pro­tein 4.2, or band 3, respec­tively.6-9 These pro­teins patients with HS. A form of sple­nec­tomy is indi­cated for patients
build the scaf­fold and the ver­ti­cal con­nec­tions of the RBC mem­ with HS who have severe ane­ mia requir­
ing fre­ quent trans­ fu­
brane skel­e­ton with the lipid mem­brane; their defi­ciency allows sions or hav­ing com­pro­mised qual­ity of life; par­tial sple­nec­tomy
mem­ brane loss and, con­ se­
quently, the for­ ma­tion of sphero- should be strongly con­sid­ered as an option when the patient is
cytes.6,9 Patients with auto­ so­mal dom­ i­
nant (AD) HS due to youn­ger than 5 years of age and for AD HS.5
SLC4A1, SPTB, or ANK1 muta­tions or AR EPB42-HS pres­ent with a In the clin­i­cal case above, in which the genetic diag­no­sis con­
range of well-com­pen­sated hemo­ly­sis up to a mod­er­ately severe firmed an SPTA1 null muta­tion in trans to αLEPRA, known to cause
ane­mia (Hb <8 g/dL) with brisk reticulocytosis (>10%).6,10 Severe severe trans­fu­sion-depen­dent spherocytosis, sple­nec­tomy was
AR HS typ­i­cally pres­ents as a trans­fu­sion-depen­dent dis­ease from the expected course of action. However, the addi­tional find­
early infancy and is most fre­quently due to a null SPTA1 var­i­ant in ing of the PIEZO1 VUCS com­pli­cated the deci­sion: if this var­i­
trans to an SPTA1 allele with the αLEPRA poly­mor­phism, allowing for ant was path­o­genic, an addi­tional com­po­nent of HX could be
a small resid­ual expres­sion of nor­mal α-spectrin, as in the case caus­ing intra­vas­cu­lar hemo­ly­sis, lead­ing to an increased risk
above.2,3 Rarely, a sim­i­lar phe­no­type of AR HS may be due to a null of postsplenectomy thrombophilia.19 This is not an uncom­mon
ANK1 muta­tion in trans to a pro­moter or mis­sense var­i­ant of ANK1 sce­nario since PIEZO1 has numer­ous VUCS. Parental stud­ies to
decreas­ing but not oblit­er­at­ing the incor­po­ra­tion of ankyrin into sep­a­rate the var­i­ants and exam­ine the asso­ci­ated phe­no­type by
the mem­brane skel­e­ton.11 With the advent of intra­uter­ine trans­ ektacytometry and RBC cat­ion con­tent are help­ful to guide clin­
fu­sions for severe pre­na­tal ane­mia, we now see the most severe i­cal deci­sions, as in our case. Given the con­cern regard­ing the
HS cases, which used to be embry­o­nal-lethal dur­ing the third tri­ patient’s increas­ing iron over­load despite che­la­tion, the fam­ily
mes­ter of preg­nancy: these are cases due to biallelic SPTA1 null and the pri­mary hema­tol­o­gist decided to pur­sue sple­nec­tomy
var­i­ants or homo­zy­gous SLC4A1 var­i­ants,3,12 which would remain by the time he was 4. Thus, par­tial sple­nec­tomy was performed
trans­fu­sion-depen­dent even after sple­nec­tomy. to mit­i­gate hemo­ly­sis while pre­serv­ing splenic immune func­
tion.5,7,17,18,20 Although par­ tial sple­
nec­tomy (leav­ ing either the
Clinical man­age­ment supe­rior or infe­rior pole of the spleen) in HS is fre­quently a per­
Neonatal hyperbilirubinemia, presenting as early as within the ma­nent solu­tion,5 fol­low-up sur­gery with total sple­nec­tomy may
first 24 hours of life, is fre­quently the presenting sign of HS of any be needed, espe­cially in cases of SPTA1-asso­ci­ated AR HS.
sever­ity. Since fast-ris­ing uncon­ju­gated bil­i­ru­bin poses a risk of Notable excep­ tions of sple­ nec­tomy effi­
cacy are the rare
kernicterus, appro­pri­ate mon­i­tor­ing and treat­ment with photo- cases due to a com­plete defi­ciency of α-spectrin or band 3, who
therapy or, rarely, with exchange trans­fu­sion is needed. A trans­ require either a life­long pro­gram of trans­fu­sions and iron che­la­
fu­sion require­ment in mild or mod­er­ate spherocytosis may not tion or stem cell transplantion.3,12
develop for 2 to 3 weeks after birth, when the nor­mal neo­na­tal Not all­spherocytes are due to HS. The his­tory, blood smear,
hyposplenism resolves. It is dif­fi­cult to pre­dict the phe­no­type of and ektacytometry in non-trans­fu­sion-depen­dent con­gen­i­tal
HS dur­ing the first year of life: trans­fu­sion depen­dency may con­ dyserythropoietic ane­mia type II (CDA-II) may closely resem­
tinue dur­ing the first 9 months of life due to delayed eryth­ro­poi­e­ ble HS (Figure 2).4,21 Suboptimal reticulocytosis and high or
tin response.13 During that time, eval­u­a­tion for the genetic cause high-nor­mal fer­ri­tin val­ues should trig­ger a genetic workup to
of the dis­ease is attrac­tive for phy­si­cians and fam­i­lies, to inform explore this dif­fer­en­tial diag­no­sis. In addi­tion, warm auto­im­
prog­no­sis. Infants with an AD form of HS may ben­e­fit from eryth­ mune hemo­lytic ane­mia fre­quently causes enough RBC mem­
ro­poi­e­tin admin­is­tra­tion to wean off trans­fu­sions faster.14 brane loss to resem­ ble spherocytosis, both in eryth­ ro­
cyte
It is not rare for patients with mild HS who have com­pen­ mor­phol­ogy and ektacytometry. Therefore, the first tests rec-
sated hemo­ly­sis to pres­ent later in life, after a viral infec­tion, such ommended in the workup of a newly presenting hemo­ly­sis are
as with Epstein-Barr virus or par­vo­vi­rus B19; with a hemo­lytic or direct and indi­rect anti­glob­u­lin tests (DAT and IAT) to eval­u­ate
aplastic cri­sis; or with gall­stones or spleno­meg­aly on phys­i­cal for the pos­si­bil­ity of an immune-medi­ated cause of hemo­lytic
exam. Occasionally, tod­dlers are diag­nosed with HS after refer­ral ane­mia since this rad­i­cally alters man­age­ment (Figure 3).
Prakash Singh Shekhawat
Red cell mem­brane dis­or­ders  |  333
incor­po­rate into the mem­brane skel­e­ton and weaken its hor­
i­zon­tal scaf­fold.9,22 These var­i­ants have increased prev­a­lence in
malaria-endemic regions (up to 3% in West Africa), supporting
the hypoth­e­sis that elliptocytosis con­fers a sur­vival advan­tage
to malaria. EPB41 muta­tions are respon­si­ble for fewer than 15%
of HE cases; they lead to altered or defi­cient pro­tein 4.1R, com­
pro­mis­ing the spectrin-4.1R-actin asso­ci­a­tion at the junc­tions.7,23
Very rare cases of mild HE have been described due to homo­
zy­gous non­sense muta­tions in GYPC, caus­ing a com­plete defi­
ciency of glycophorin C and, con­se­quently, a par­tial defi­ciency
of pro­tein 4.1R in the junc­tional com­plex.
HE is char­ac­ter­ized by ellip­ti­cally shaped RBCs and is fre­
quently asymp­tom­atic with min­i­mal or no hemo­ly­sis, nor­mal
Hgb, and a nor­mal or bor­der­line high retic­u­lo­cyte count. Nev-

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/331/1851848/331kalfa.pdf by guest on 13 December 2021


ertheless, patients may pres­ent with exac­er­bated neo­na­tal jaun­
dice or develop epi­sodic hemo­ly­sis with illnesses that cause
hyper­pla­sia of the retic­u­lo­en­do­the­lial sys­tem, eg, viral hep­a­ti­tis,
infec­tious mono­nu­cle­o­sis, bac­te­rial infec­tions, and malaria.7,23
Hereditary pyropoikilocytosis (HPP) is char­ ac­ter­
ized by
in­
creased poikilocytosis, RBC frag­ men­ ta­
tion, and elliptocytes
on the periph­eral smear (Figure 4). The most com­mon form of
HPP is diag­nosed in infants of Afri­can ances­try, who pres­ent with
neo­na­tal jaun­dice and fre­quently a trans­fu­sion require­ment, and
is due to an SPTA1 HE-caus­ing muta­tion in trans to the SPTA1
splic­ing var­ i­
ant c.6531-12C>T, known as αLELY (low expres­ sion
LYon).23,24 αLELY causes a 50% decrease in α-spectrin expres­sion
but is clin­i­cally silent in nor­mal indi­vid­u­als even in the homo­zy­
gous state since α-spectrin is pro­duced well in excess. However,
in trans to an HE-caus­ing SPTA1 muta­tion, αLELY leads to an HPP
phe­no­type because it allows for increased rel­a­tive incor­po­ra­tion
of the abnor­mal spectrin chain into the mem­brane skel­e­ton. Of
note, αLELY has a minor allele fre­quency of up to 25.5% (gnomad​
­.broadinstitute​­.org); there­fore, it is fairly com­mon for an indi­vid­
Figure 2. CDA-II is in the differential diagnosis for HS. A 4-year- ual with non­he­mo­lytic HE to have a child with HPP.
old African American girl presented with DAT-negative, mild HPP can also be caused by homo­zy­gous or com­pound het­ero­
hemolytic anemia (Hgb, 10 g/dL) with a reticulocyte count of zy­gous HE-caus­ing var­i­ants of SPTA1, SPTB, or EPB41 or a com­bi­
2.4%, a normal ARC of 86 × 103/µL, and mild jaundice. She had a na­tion of SPTA1 and SPTB alleles with HE muta­tions.23,24 Hemolysis
history of prolonged neonatal jaundice treated with photother- and poikilocytosis per­sist in those patients (true HPP), while the
apy and no transfusion requirement. Ektacytometry showed a sever­ity of the dis­ease may vary from com­pen­sated hemo­ly­sis
curve that resembled HS, and a blood smear showed sphero- (eg, a case with com­bined SPTA1 p.L155dup and SPTB p.W2024R)
cytes, marked poikilocytosis, and no polychromasia. Inade- up to trans­fu­sion depen­dency unre­spon­sive to sple­nec­tomy (eg,
quate reticulocytosis and a ferritin of 80 ng/mL, at a generous a child with com­pound het­ero­zy­gos­ity for spectrin St. Claude
level for her age with no concurrent inflammation, triggered SPTA1 c.2806-13T>G,24 pro­duc­ing a trun­cated α-spectrin pro­tein
further evaluation with sequencing of an HHA gene panel that and a novel non­sense SPTA1 muta­tion p.R118* in trans).8 An exam­
revealed 2 SEC23B mutations, c.40C>T (p.R14W) and c.367- ple of true HPP due to homo­zy­gous SPTB p.F2014V muta­tion with
3A>G. Follow-up targeted sequencing of her parents con- trans­fu­sion depen­dency that responded well to sple­nec­tomy
firmed that these 2 variants were in trans, causing CDA-II. Scale is detailed in Figure 5, to be contrasted with the exam­ple case
bar = 14 mm. of infan­tile HPP in Figure 4. My lim­ited per­sonal expe­ri­ence with
these rare cases sug­gests that reticulocytopenia while on trans­
fu­sions in severe HHA, such as the reces­sively inherited SPTA1-
Hereditary elliptocytosis and hered­i­tary asso­ci­ated HS or severe true HPP, pre­dicts a poor response to
pyropoikilocytosis (HE/HPP) sple­nec­tomy. A bet­ter-informed ratio­nal approach to treat­ment
Genotype/phe­no­type cor­re­la­tions is being devel­oped, as we are gaining more insights into the spe­
Hereditary elliptocytosis (HE) is caused by het­ero­zy­gous muta­ cific genetic muta­tions caus­ing these dis­eases.
tions in SPTA1, SPTB, and EPB41, the genes encoding α-spectrin,
β-spectrin, and pro­tein 4.1R, respec­tively. HE-caus­ing SPTA1 and Clinical man­age­ment
SPTB var­i­ants are typ­i­cally located in the spectrin tetrameriza- HE is fre­quently diag­nosed inci­den­tally on a blood smear review
tion domain or, less fre­quently, along the chain or close to the or dur­ing inves­ti­ga­tion for gall­stones or spleno­meg­aly or in a
junc­tional com­plex and pro­duce a qual­i­ta­tive rather than quan­ti­ par­
ent of an infant with neo­ na­
tal jaun­
dice diag­
nosed with
ta­tive defect in the spectrin tet­ra­mer; the altered spectrin chains HPP. Infants with the com­mon form of HPP (SPTA1 HE-var­i­ant in

334  |  Hematology 2021  |  ASH Education Program


Hemolysis positive Consider physical agents
DAT and Work-up for autoimmune
(with or without anemia) including
reticulocytes, indirect Coombs or alloimmune causes
MAHA/HUS/aHUS/TTP
unconjugated bilirubin,
haptoglobin
negative
Consider PNH
Globin gene and Consider
Recently or yes
Family h istory HHA/CDA panel or subpanels Wilson disease
chronically
and depending on family history and
transfused patient
blood smear r eview smear review
no
of patient and parents
to assist with diagnosis abnormal
Hemoglobin Globin gene sequencing and
electrophoresis deletion/duplication analysis

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/331/1851848/331kalfa.pdf by guest on 13 December 2021


Suboptimal normal
reticulocytosis,
Ektacytometry abnormal
iron overload,
or o smotic fragility RBC membrane disorder
skeletal
and/or EMA flow cytometry
abnormalities
normal
or

RBC membrane disorders gene panel,


abnormal RBC enzymopathies
CDA gene RBC enzyme activity especially if
panel gene panel splenectomy is contemplated
normal

binuclear or multinuclear erythroblasts Untypable CDA


Bone marrow studies (consider CDA Registries)
normal except erythroid hyperplasia
Figure 3. Proposed algorithm for laboratory workup of a patient presenting with hemolysis with or without anemia. In many cases of
mild HS and most cases of PIEZO1-associated HX, anemia may be well compensated by reticulocytosis. Although our focus here is on
RBC membrane disorders, the differential includes other causes of hemolytic disorders that are mentioned in this algorithm. Evaluation
for autoimmune or, especially in an infant, alloimmune hemolytic anemia with DAT and IAT is the first testing recommended since such
a diagnosis is frequently acute and evolving, requiring immediate action. Of note, warm autoimmune hemolytic anemia in children and
occasionally adults with underlying immune dysregulation may be atypical and conventionally DAT-negative.52-54 Consideration should
also be given to the possibility of MAHA, PNH, and Wilson disease. The cases described in this review, especially that presented in
Figure 6, demonstrate utilization of this algorithm. A blood smear review of the patient and parents and attention to the RBC indices
­including MCV, MCHC, and RDW, along with hemolytic markers (unconjugated bilirubin, lactate dehydrogenase, h ­ aptoglobin—of
note, haptoglobin is reliable after 6 months of life since earlier it may be low due to decreased production by the ­infant’s liver rather
than ­increased consumption) and ferritin and transferrin saturation to consider iron-loading inefficient ­erythropoiesis, can ­provide hints
as to the differential diagnosis. In a non-chronically transfused patient, we suggest phenotypic evaluation considering the ­differential
of globin disorders, followed by RBC membranopathies and enzymopathies. Rare causes of HHA such as unstable Hgb disorders and
CDAs also need to be considered. When suboptimal reticulocytosis or iron overload or skeletal abnormalities are noted in a patient
with hemolytic anemia, the possibility of CDA should be considered and pursued. The combination of blood smear review and osmotic
gradient ektacytometry frequently helps to narrow the differential while alerting clinicians of rare possibilities. Osmotic gradient ekta-
cytometry evaluates the deformability of RBCs as they are subjected to constant shear stress in a medium of increasing osmolality in a
laser diffraction viscometer and is the reference technique for differential diagnosis of erythrocyte membrane and hydration disorders
when a recent transfusion does not interfere with phenotypic evaluation of the patient.4 Flow cytometry with eosin-5′-maleimide bind-
ing of band 3 and Rh-related proteins is a rapid screening test for RBC membrane disorders characterized by membrane loss.55 Osmot-
ic fragility is increased in HS and expected to be decreased in HX (however, it is reported as normal in patients with KCNN4 Arg352His
mutation31). When the patient is recently or chronically transfused, as is typically the case for infants with HHA, genetic evaluation
with clinically available NGS panels or research-based whole-exome sequencing or whole-genome sequencing may provide an accu-
rate diagnosis necessary for appropriate management decisions. Laboratories that offer sequencing on genes or panels associated
with HHAs and red cell membrane disorders can be found by searching the ­Genetic Testing R ­ egistry: https:​/​/www​.ncbi​.nlm​.nih​.gov​/
gtr​/. e.g. https:​/​/www​.ncbi​.nlm​.nih​.gov​/gtr​/all​/tests​/​?term=RBC%20membrane%20disorders. aHUS, atypical ­hemolytic uremic syn-
drome; HUS, hemolytic uremic syndrome; MAHA, microangiopathic hemolytic anemia; PNH, paroxysmal nocturnal hemoglobinuria;
RDW, RBC distribution width; TTP, thrombotic thrombocytopenic purpura.

Prakash Singh Shekhawat


Red cell mem­brane dis­or­ders  |  335
Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/331/1851848/331kalfa.pdf by guest on 13 December 2021
Figure 4. Transient infantile HPP. An African American girl presented within the first day of life after delivery at term with nonimmune
hemolytic anemia and neonatal jaundice. She was treated with triple phototherapy but did not require RBC transfusion. (A) The blood
smear of the patient with HPP showing marked anisocytosis and poikilocytosis with bizarre microcytes and fragmented cells along
with elliptocytes (scale bar = 14 µm). (B) The blood smear of the mother, who had a normal Hgb and reticulocyte count, was notable
for elliptocytes indicating HE. (C) Ektacytometry showed the trapezoid curve characteristic for HE and HPP with decreased deform-
ability for both mother and patient. The patient’s sample was sequenced by NGS on an RBC membrane gene panel, which revealed
the SPTA1 HE-causing variant c.460_463insTTG (p.L155dup) as well as the SPTA1 c.6531-12C>T polymorphism known as αLELY. Targeted
sequencing for these variants revealed that the mother was heterozygous for SPTA1 p.L155dup. The mother did not carry αLELY, which
was presumably inherited by the father. The compound heterozygosity of an HE-causing SPTA1 mutation in trans to αLELY confirmed
the diagnosis of infantile HPP. The child continued to have a mild anemia with reticulocytosis (Hgb, 10.2 g/dL with 3.8% reticulocytes)
and poikilocytosis, with RDW up to 18.7% at her 2-year-old follow-up, but a CBC with reticulocyte count and blood smear at 5 years
of age indicated transition to a nonhemolytic HE phenotype. RDW, RBC distribution width.

trans to αLELY) may have mod­er­ate to severe hemo­lytic ane­mia HX is char­ac­ter­ized by intra­vas­cu­lar hemo­ly­sis and intrahepatic
requir­ing fre­quent trans­fu­sions early in life, but the phe­no­type RBC destruc­tion that per­sists after sple­nec­tomy (there­fore,
improves after the first 1 to 2 years, evolv­ing to typ­i­cal HE.7 HPP splenec­tomy is not only dan­ger­ous but also inef­fec­tive as an
due to biallelic HE-caus­ing muta­tions may pres­ent with mild to HX treat­ment) as well as iron over­load dis­pro­por­tion­ate to the
severe chronic ane­mia with long-term trans­fu­sion depen­dence. trans­fu­sion his­tory.19,33 Significant phe­no­typic var­i­abil­ity exists,
Most but not all­of the cases with severe hemo­ly­sis and a chronic prob­a­bly due to the large num­ber of caus­at­ive var­i­ants, coin-
trans­fu­sion require­ment respond to sple­nec­tomy.8,24 heritance of var­i­ants in other genes affect­ing RBC phe­no­type,34
and mul­ti­ple mech­a­nisms con­trib­ut­ing to the path­o­gen­e­sis.35
Hereditary xerocytosis (HX) HX patients may pres­ent with mild to mod­er­ate hemo­lytic ane­
Genotype/phe­no­type cor­re­la­tions mia, usu­ally due to an aminoterminal PIEZO1 muta­tion or KCNN4
HX, also called dehydrated hered­i­tary stomatocytosis, is an AD var­i­ants, or, with fully com­pen­sated hemo­ly­sis, due to the most
dis­ease caused by muta­tions in PIEZO1 or KCNN4,25-32 cod­ing com­mon carboxyterminal PIEZO1 var­i­ants (Figure 6).19 Despite
respec­ tively for the mechanosensitive cat­ ion chan­ nel PIEZO1 the RBC dehy­dra­tion, mean cel­lu­lar vol­ume (MCV) is high nor­mal
and the cal­ cium ion-acti­ vated potas­ sium (K+) chan­nel known to nor­mal in HX, likely due to the effects of the mutated chan­
as the Gardos chan­nel. HX RBCs have an abnor­mal K+ leak not nels in eryth­ro­poi­e­sis.36 Cases with poly­cy­the­mia despite hemo­
com­pen­sated by a pro­por­tional intra­cel­lu­lar sodium (Na+) gain, ly­sis have also been reported,37 pointing to the hypoth­e­sis that
lead­ing to cel­ lu­
lar dehy­ dra­ tion. Pseudohyperkalemia may be patients with HX may have increased eryth­ro­poi­etic drive for
noted if anal­y­sis of elec­tro­lytes in blood spec­i­mens is delayed. their level of Hgb because of rel­at­ively low 2,3-DPG lev­els and

336  |  Hematology 2021  |  ASH Education Program


Clinical man­age­ment
Genetic coun­sel­ing and close mon­i­tor­ing of preg­nan­cies should
be pro­vided when either par­ent has HX because of the risk of
peri­na­tal edema and non­im­mune hydrops fetalis.19
Splenectomy is contraindicated in HX due to PIEZO1 muta­
tions because it pre­dis­poses patients to life-threat­en­ing venous
and arte­rial throm­bo­em­bolic com­pli­ca­tions, reported to hap­pen
within a month up to 28 years post sple­nec­tomy.19,40,41 Although
no throm­botic com­pli­ca­tions were observed after sple­nec­tomy
in 12 patients with KCNN4-asso­ci­ated HX after fol­low-up for 2 to
44 years,19,31 sple­nec­tomy should be avoided any­way since it pro­vi­
des no ther­a­peu­tic ben­e­fit in either PIEZO1- or KCNN4-asso­ci­ated
HX because intra­vas­cu­lar and intrahepatic RBC destruc­tion per­
sists after sple­nec­tomy.33

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/331/1851848/331kalfa.pdf by guest on 13 December 2021


Management is cur­rently focused on sup­port­ive care for ane­
mia and hemo­ly­sis com­pli­ca­tions and on pre­ven­tive mon­i­tor­ing
to quan­ti­tate and treat iron over­load as needed. Screening with
fer­ri­tin and trans­fer­rin sat­u­ra­tion should be followed (even with a
mod­er­ate increase in fer­ri­tin) by T2* mag­netic res­o­nance imag­
ing of the liver and heart. Iron over­load is much more eas­ily and
effec­tively treated with che­la­tion when diag­nosed early. Phle-
botomy has also been used to address HX-asso­ci­ated iron over­
load. However, based on the fact that hemo­chro­ma­to­sis in HX
is mul­ti­fac­to­rial and includes exces­sive eryth­ro­poi­e­sis caus­ing
increased erythroferrone that suppresses hepcidin as well as the
direct sup­pres­sion of hepcidin,35,42 the effi­cacy of phle­bot­omy to
treat iron over­load in HX needs fur­ther eval­u­a­tion.43
Specific med­i­ca­tions for the treat­ment of HX are not clin­i­cally
avail­­able. A prom­is­ing option may be a spe­cific inhib­i­tor for the
Gardos chan­nel, senicapoc, since KCNN4 is the final com­mon
medi­a­tor of RBC dehy­dra­tion in HX.8,44 Senicapoc was shown to
be well tol­er­ated in a phase 3 trial in sickle cell dis­ease, where
it reduced RBC dehy­dra­tion and hemo­ly­sis and increased Hgb
lev­els.45 A phase 1/2 “proof-of-con­cept study of senicapoc in
patients with famil­ ial dehydrated stomatocytosis caused by
the V282M muta­tion in the Gardos (KCNN4) chan­nel” has been
posted on clinicaltrials​­.gov and is cur­rently recruiting.
Figure 5. Persistent (true) HPP. A boy of Iranian descent present-
ed with severe anemia since infancy. He remained transfusion
Overhydration syn­dromes
dependent; therefore, his erythrocyte phenotype was not eval-
Genotype/phe­no­type cor­re­la­tions
uable. (A) A blood smear of the father (practically identical with
Genetic eti­­ol­ogy in overhydration syn­dromes is var­i­able, along
the blood smear of the mother) and (B) ektacytometry show-
with a phe­no­type of mild to severe hemo­lytic ane­mia.46,47 Blood
ing a trapezoid shape typical for HE for both parental samples
smear is remark­able for abun­dant stomatocytes; RBC phys­i­­ol­ogy
indicated that they both had elliptocytosis. NGS on a panel of
is char­ac­ter­ized by a large net increase in intra­cel­lu­lar Na+ and
RBC membrane disorder genes revealed that the patient was
water not ade­quately com­pen­sated by a decrease in K+. MCV is
homozygous for a novel missense mutation in the SPTB gene
typ­i­cally increased, and mean cor­pus­cu­lar hemo­glo­bin con­cen­
(c.6040T>G, p.F2014V), affecting the spectrin self-association
tra­tion (MCHC) is decreased. Osmotic fra­gil­ity is increased, and
site. Both parents were heterozygous for the same mutation.24
ektacytometry is right-shifted.
The most severe form of overhydrated hered­i­tary stomatocyto-
sis is caused by het­ero­zy­gous mis­sense muta­tions in RHAG, the
con­se­quently high Hb-oxy­gen affin­ity and exac­er­bated tis­sue gene cod­ing for the red cell ammo­nium trans­porter rhesus-
and renal hyp­oxia.38,39 Hepatic and/or myo­car­dial iron over­load asso­ci­ated gly­co­pro­tein; both muta­tions described so far
may be the presenting sign for some patients when not diag­ (p.Phe65Ser and p.Ile61Arg) alter amino acids lin­ing the puta­tive
nosed before adult­hood. Evaluating for hemo­chro­ma­to­sis gene trans­port chan­nel, mak­ing it per­me­able to Na+ ions.48-50
muta­tions is recommended since this may alter clin­i­cal prog­no­ Cryohydrocytosis, Southeast Asian ovalocytosis, and band 3
sis and care. Iron over­load may also be accel­er­ated in cases of Ceinge are caused by cer­tain het­ero­zy­gous muta­tions in SLC4A1,
KCNN4-HX,19 as well as in cases of aminoterminal PIEZO1 path­o­ which alter the band 3 chan­nel to medi­ate cat­ion leak in cold tem­
log­i­cal var­i­ants such as the p.V598M (per­sonal com­mu­ni­ca­tion per­a­tures.46,47 Patients with stomatin-defi­cient ­cryohydrocytosis,
on 2 patients in their early 20s with pref­er­en­tial and severe myo­ caused by muta­tions in SLC2A1 cod­ing for glu­cose trans­porter
car­dial iron over­load). type 1, pres­ent with mod­er­ate hemo­lytic ane­mia, cat­a­racts due
Prakash Singh Shekhawat
Red cell mem­brane dis­or­ders  |  337
Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/331/1851848/331kalfa.pdf by guest on 13 December 2021
Figure 6. HX due to heterozygous PIEZO1 mutation. A European American girl, born at term, developed neonatal hyperbilirubinemia
that prevented discharge from the nursery for 4 days while receiving treatment with phototherapy. Scleral icterus was again noted
at 10 months of age, and a CBC indicated reticulocytosis but a normal Hgb. She continued to have chronic hemolysis and jaundice
and had an extensive workup including negative DAT, normal Hgb electrophoresis, RBC enzyme activity testing showing normal or
increased activity, osmotic fragility testing that was (probably mistakenly) reported as normal rather than as decreased, alpha and
beta globin genetic testing that revealed no mutation or copy number variation, a negative PNH screen by flow cytometry, and bone
marrow studies showing erythroid hyperplasia, no dyserythropoiesis, and markedly increased iron stores. (A) At 10 years of age, she
had osmotic gradient ektacytometry performed that demonstrated a typical HX curve, with left shift due to decreased Omin and
Ohyp (Figure 2A). The CBC at the time indicated an Hgb of 14.2 g/dL, an MCV of 96 fl, an MCH of 36.5 pg, an MCHC of 37.9 g/dL, a
reticulocyte count 16.9%, and an ARC of 643 × 103/µL. (B) A blood smear was significant for polychromasia, macrocytosis, and occa-
sional stomatocytes, target cells, and dense fragmented cells. Of note, sometimes the blood smear may be deceptively normal, with
only a few target cells and no stomatocytes. A determination of the RBC intracellular cation confirmed a reduced K+ content without
a corresponding increase in Na+ content. NGS for an RBC membrane disorder panel revealed 1 of the most common PIEZO1 variants
(c.7367G>A, p.R2456H) causing HX. PNH, paroxysmal nocturnal hemoglobinuria.

to the altered cat­ion per­me­abil­ity of the lens, and neu­ro­log­i­cal ulation should be con­sid­ered in patients who have inad­ver­tently
dis­or­ders caused by disrupted glu­cose trans­port. been splenectomized.

Clinical man­age­ment Conclusions


Management is sup­port­ive for ane­mia and hemo­ly­sis com­pli­ Decades of research on the molec­ u­
lar path­
o­gen­e­
sis of RBC
ca­tions. Splenectomy is usu­ally par­tially effec­tive in reduc­ing mem­brane dis­or­ders have cul­mi­nated,9,51 upon the advent of
hemo­ly­sis, but it car­ries a high risk of throm­bo­em­bolic com­pli­ NGS over the last 10 years, in a high suc­cess rate of genetic diag­
ca­tions and pul­mo­nary hyper­ten­sion.40,41 Prophylactic anticoag- no­sis for patients with such dis­eases. Genetic diag­no­sis is highly

338  |  Hematology 2021  |  ASH Education Program


ben­e­fi­cial for trans­fu­sion-depen­dent patients and likely pru­dent 11. Eber SW, Gonzalez JM, Lux ML, et  al. Ankyrin-1 muta­ tions are a major
to con­sider before pro­ceed­ing to the irre­vers­ible treat­ment of cause of dom­i­nant and reces­sive hered­i­tary spherocytosis. Nat Genet.
1996;13(2):214-218.
sple­nec­tomy, which is not always effec­tive or safe. Increased 12. Kager L, Bruce LJ, Zeitlhofer P, et al. Band 3 nullVIENNA, a novel homo­zy­
infor­ma­tion on geno­type-phe­no­type cor­re­la­tion along with the gous SLC4A1 p.Ser477X var­i­ant caus­ing severe hemo­lytic ane­mia, dyseryth-
cur­rent wider avail­abil­ity of par­tial sple­nec­tomy calls for a new ropoiesis and com­plete dis­tal renal tubu­lar aci­do­sis. Pediatr Blood Cancer.
con­sen­sus state­ment of experts in the field on updated man­age­ 2017;64(3):e26227.
13. Delhommeau F, Cynober T, Schischmanoff PO, et  al. Natural his­tory of
ment guide­lines.
hered­i­tary spherocytosis dur­ing the first year of life. Blood. 2000;95(2):393-
397.
Acknowledgments  14. Tchernia G, Delhommeau F, Perrotta S, et al; Euro­pean Society for Paediat-
This work was supported by National Heart, Lung, and Blood In- ric Haematology and Immunology Working Group on Hemolytic Anemias.
stitute grant R01 HL152099. Recombinant eryth­ro­poi­e­tin ther­apy as an alter­na­tive to blood trans­fu­
sions in infants with hered­i­tary spherocytosis. Hematol J. 2000;1(3):146-
We are grate­ful to the patients, fam­i­lies, and refer­ring phy­ 152.
si­cians par­tici­pat­ing in our diag­nos­tic stud­ies of hered­i­tary 15. Crary SE, Buchanan GR. Vascular com­ pli­
ca­
tions after sple­ nec­tomy for
hemo­lytic ane­mia and to the teams of the Cincinnati Children’s hema­to­logic dis­or­ders. Blood. 2009;114(14):2861-2868.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/331/1851848/331kalfa.pdf by guest on 13 December 2021


Erythrocyte Diagnostic Laboratory and Molecular Genetics Lab- 16. Schilling RF, Gangnon RE, Traver MI. Delayed adverse vas­ cu­
lar events
after sple­nec­tomy in hered­i­tary spherocytosis. J Thromb Haemost. 2008;
oratory for their con­tri­bu­tion to the diag­nos­tic stud­ies of cases
6(8):1289-1295.
presented in the text and fig­ures. 17. Englum BR, Rothman J, Leonard S, et al; Splenectomy in Congenital Hemo-
Clinical out­comes reported in some of these cases are lytic Anemia Consortium. Hematologic out­comes after total sple­nec­tomy
based upon per­sonal com­mu­ni­ca­tion with Drs. Marya Alman- and par­tial sple­nec­tomy for con­gen­i­tal hemo­lytic ane­mia. J Pediatr Surg.
soori, Jeanne Boudreaux, Mor­gan McLemore, Charles Quinn, and 2016;51(1):122-127.
18. Pincez T, Guitton C, Gauthier F, et  al. Long-term fol­low-up of sub­to­tal
Audrey Woerner. sple­nec­tomy for hered­i­tary spherocytosis: a sin­gle-cen­ter study. Blood.
2016;127(12):1616-1618.
Conflict-of-inter­est dis­clo­sure 19. Picard V, Guitton C, Thuret I, et al. Clinical and bio­log­i­cal fea­tures in PIEZO1-
hered­i­tary xerocytosis and Gardos channelopathy: a ret­ro­spec­tive series
Theodosia A. Kalfa: no com­pet­ing finan­cial inter­ests to declare.
of 126 patients. Haematologica. 2019;104(8):1554-1564.
20. Rice HE, Englum BR, Rothman J, et al; Splenectomy in Congenital Hemo-
Off-label drug use lytic Anemia Consortium. Clinical out­comes of sple­nec­tomy in chil­dren:
Theodosia A. Kalfa: nothing to disclose. report of the sple­nec­tomy in con­gen­i­tal hemo­lytic ane­mia reg­is­try. Am J
Hematol. 2015;90(3):187-192.
21. Danise P, Amendola G, Nobili B, et al. Flow-cytometric anal­y­sis of eryth­
Correspondence ro­cytes and retic­u­lo­cytes in con­gen­i­tal dyserythropoietic anae­mia type II
Theodosia A. Kalfa, Cancer and Blood Diseases Institute, Cincin- (CDA II): value in dif­fer­en­tial diag­no­sis with hered­i­tary spherocytosis. Clin
nati Children’s Hospital Medical Center, 3333 Burnet Ave, MLC Lab Haematol. 2001;23(1):7-13.
22. Harper SL, Sriswasdi S, Tang HY, Gaetani M, Gallagher PG, Speicher DW.
7015, Cincinnati, OH 45229-3039; e-mail: theodosia​­.kalfa@cchmc​
The com­mon hered­i­tary elliptocytosis-asso­ci­ated α-spectrin L260P muta­
­.org. tion perturbs eryth­ro­cyte mem­branes by sta­bi­liz­ing spectrin in the closed
dimer con­for­ma­tion. Blood. 2013;122(17):3045-3053.
References 23. Gallagher PG. Hereditary elliptocytosis: spectrin and pro­tein 4.1R. Semin
1. Wichterle H, Hanspal M, Palek J, Jarolim P. Combination of two mutant Hematol. 2004;41(2):142-164.
alpha spectrin alleles under­lies a severe spherocytic hemo­lytic ane­mia. J 24. Niss O, Chonat S, Dagaonkar N, et  al. Genotype-phe­no­type cor­re­la­tions
Clin Invest. 1996;98(10):2300-2307. in hered­i­tary elliptocytosis and hered­i­tary pyropoikilocytosis. Blood Cells
2. Gallagher PG, Maksimova Y, Lezon-Geyda K, et al. Aberrant splic­ing con­ Mol Dis. 2016;61(Octo­ber):4-9.
trib­utes to severe α-spectrin-linked con­gen­i­tal hemo­lytic ane­mia. J Clin 25. Zarychanski R, Schulz VP, Houston BL, et  al. Mutations in the mechano-
Invest. 2019;129(7):2878-2887. transduction pro­tein PIEZO1 are asso­ci­ated with hered­i­tary xerocytosis.
3. Chonat S, Risinger M, Sakthivel H, et  al. Corrigendum: the spec­trum of Blood. 2012;120(9):1908-1915.
SPTA1-asso­ci­ated hered­i­tary spherocytosis. Front Physiol. 2019(18 Octo­ 26. Andolfo I, Alper SL, De Franceschi L, et al. Multiple clin­i­cal forms of dehy-
ber);10:1331. drated hered­i­tary stomatocytosis arise from muta­tions in PIEZO1. Blood.
4. Da Costa L, Suner L, Galimand J, et al; Society of Hematology and Pediatric 2013;121(19):3925-3935, S1-S12.
Immunology Group; French Society of Hematology. Diagnostic tool for red 27. Albuisson J, Murthy SE, Bandell M, et al. Dehydrated hered­i­tary stomatocy-
blood cell mem­brane dis­or­ders: assess­ment of a new gen­er­a­tion ektacy- tosis linked to gain-of-func­tion muta­tions in mechanically acti­vated PIEZO1
tometer. Blood Cells Mol Dis. 2016;56(1):9-22. ion chan­nels. Nat Commun. 2013;4(2013):1884.
5. Rothman JA, Stevens JL, Gray FL, Kalfa TA. How I approach hered­i­tary hemo­ 28. Glogowska E, Lezon-Geyda K, Maksimova Y, Schulz VP, Gallagher PG. Muta-
lytic ane­mia and sple­nec­tomy. Pediatr Blood Cancer. 2020;67(11):e28337. tions in the Gardos chan­nel (KCNN4) are asso­ci­ated with hered­i­tary xero-
6. Eber S, Lux SE. Hereditary spherocytosis—defects in pro­teins that con­nect cytosis. Blood. 2015;126(11):1281-1284.
the mem­brane skel­e­ton to the lipid bila­yer. Semin Hematol. 2004;41(2):118- 29. Rapetti-Mauss R, Lacoste C, Picard V, et al. A muta­tion in the Gardos chan­
141. nel is asso­ci­ated with hered­i­tary xerocytosis. Blood. 2015;126(11):1273-1280.
7. Mohandas N. Inherited hemo­lytic ane­mia: a pos­ses­sive begin­ner’s guide. 30. Andolfo I, Russo R, Manna F, et al. Novel Gardos chan­nel muta­tions linked
Hematology Am Soc Hematol Educ Program. 2018;2018(1):377-381. to dehydrated hered­ i­
tary stomatocytosis (xerocytosis). Am J Hematol.
8. Risinger M, Kalfa TA. Red cell mem­brane dis­or­ders: struc­ture meets func­ 2015;90(10):921-926.
tion. Blood. 2020;136(11):1250-1261. 31. Fermo E, Bogdanova A, Petkova-Kirova P, et al. “Gardos channelopathy”:
9. Lux SE. Disorders of the red cell mem­brane. In: Orkin SH, Fisher DE, Gins- a var­i­ant of hered­i­tary stomatocytosis with com­plex molec­u­lar reg­u­la­tion.
burg D, et al, eds. Nathan and Oski’s Hematology and Oncology of Sci Rep. 2017;7(1):1744.
Infancy and Childhood. Saunders Elsevier; 2015:515-579. 32. Rivera A, Vandorpe DH, Shmukler BE, et  al. Erythrocyte ion con­tent and
10. Kalfa TA, Connor JA, Begtrup AH. EPB42-related hered­i­tary spherocytosis. In: dehy­dra­tion mod­u­late max­i­mal Gardos chan­nel activ­ity in KCNN4 V282M/+
Adam MP, Ardinger HH, Pagon RA, et al, eds. GeneReviews((R)). University of hered­i­tary xerocytosis red cells. Am J Physiol Cell Physiol. 2019;317(2): C287-
Washington; 2016. https://www.ncbi.nlm.nih.gov/books/NBK190102/ C302.

Prakash Singh Shekhawat


Red cell mem­brane dis­or­ders  |  339
33. Platt OS, Lux SE, Nathan DG. Exercise-induced hemo­ly­sis in xerocytosis. dis­ease: a phase III ran­dom­ized, pla­cebo-con­trolled, dou­ble-blind study
Erythrocyte dehy­dra­tion and shear sen­si­tiv­ity. J Clin Invest. 1981;68(3):631- of the Gardos chan­ nel blocker senicapoc (ICA-17043). Br J Haematol.
638. 2011;153(1):92-104.
34. Risinger M, Glogowska E, Chonat S, et al. Hereditary xerocytosis: diag­nos­ 46. Bruce LJ. Hereditary stomatocytosis and cat­ion-leaky red cells—recent
tic con­sid­er­ations. Am J Hematol. 2018;93(3):E67-E69. devel­op­ments. Blood Cells Mol Dis. 2009;42(3):216-222.
35. Jankovsky N, Caulier A, Demagny J, et al. Recent advances in the path­o­ 47. Gallagher PG. Disorders of eryth­ro­cyte hydra­tion. Blood. 2017;130(25):2699-
phys­i­­ol­ogy of PIEZO1-related hered­i­tary xerocytosis. Am J Hematol. 2021; 2708.
96(8):1017-1026. 48. Stewart AK, Shmukler BE, Vandorpe DH, et al. Loss-of-func­tion and gain-of-
36. Caulier A, Jankovsky N, Demont Y, et al. PIEZO1 acti­va­tion delays ery­throid func­tion phe­no­types of stomatocytosis mutant RhAG F65S. Am J Physiol
dif­fer­en­ti­a­tion of nor­mal and hered­i­tary xerocytosis-derived human pro­ Cell Physiol. 2011;301(6):C1325-C1343.
gen­i­tor cells. Haematologica. 2020;105(3):610-622. 49. Genetet S, Ripoche P, Picot J, et  al. Human RhAG ammo­nia chan­nel is
37. Filser M, Giansily-Blaizot M, Grenier M, et al. Increased inci­dence of germ- impaired by the Phe65Ser muta­ tion in overhydrated stomatocytic red
line PIEZO1 muta­tions in indi­vid­u­als with idi­o­pathic erythrocytosis. Blood. cells. Am J Physiol Cell Physiol. 2012;302(2):C419-C428.
2021;137(13):1828-1832. 50. Bruce LJ, Guizouarn H, Burton NM, et  al. The mono­va­lent cat­ion leak in
38. Albala MM, Fortier NL, Glader BE. Physiologic fea­tures of hemo­ly­sis asso­ overhydrated stomatocytic red blood cells results from amino acid sub­sti­
ci­ated with altered cat­ion and 2,3-diphosphoglycerate con­tent. Blood. tu­tions in the Rh-asso­ci­ated gly­co­pro­tein. Blood. 2009;113(6):1350-1357.
1978;52(1):135-141. 51. Lux SE IV. Anatomy of the red cell mem­brane skel­e­ton: unanswered ques­

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/331/1851848/331kalfa.pdf by guest on 13 December 2021


39. Kiger L, Oliveira L, Guitton C, et  al. Piezo1-xerocytosis red cell metabo- tions. Blood. 2016;127(2):187-199.
lome shows impaired gly­col­y­sis and increased hemo­glo­bin oxy­gen affin­ity. 52. Kalfa TA. Warm anti­body auto­im­mune hemo­lytic ane­mia. Hematology Am
Blood Adv. 2021;5(1):84-88. Soc Hematol Educ Program. 2016;2016(1):690-697.
40. Stewart GW, Amess JA, Eber SW, et  al. Thrombo-embolic dis­ease after 53. Voulgaridou A, Kalfa TA. Autoimmune hemo­lytic ane­mia in the pedi­at­ric
sple­nec­tomy for hered­i­tary stomatocytosis. Br J Haematol. 1996;93(2):303- set­ting. J Clin Med. 2021;10(2):216.
310. 54. Kamesaki T, Toyotsuji T, Kajii E. Characterization of direct anti­glob­ul­in test-
41. Iolascon A, Andolfo I, Barcellini W, et al; Working Study Group on Red Cells neg­a­tive auto­im­mune hemo­lytic ane­mia: a study of 154 cases. Am J Hema-
and Iron of the Euro­ pean Hematology Association. Recommendations tol. 2013;88(2):93-96.
regard­ing sple­nec­tomy in hered­i­tary hemo­lytic ane­mias. Haematologica. 55. King MJ, Smythe JS, Mushens R. Eosin-5-maleimide bind­ ing to band 3
2017;102(8):1304-1313. and Rh-related pro­teins forms the basis of a screen­ing test for hered­i­tary
42. Archer NM, Shmukler BE, Andolfo I, et al. Hereditary xerocytosis revisited. spherocytosis. Br J Haematol. 2004;124(1):106-113.
Am J Hematol. 2014;89(12):1142-1146.
43. Kim A, Nemeth E. New insights into iron reg­u­la­tion and eryth­ro­poi­es­is.
Curr Opin Hematol. 2015;22(3):199-205.
44. Rapetti-Mauss R, Picard V, Guitton C, et al. Red blood cell Gardos chan­nel
(KCNN4): the essen­tial deter­mi­nant of eryth­ro­cyte dehy­dra­tion in hered­i­
tary xerocytosis. Haematologica. 2017;102(10):e415-e418.
45. Ataga KI, Reid M, Ballas SK, et al; ICA-17043-10 Study Investigators.
Improvements in haemolysis and indi­ ca­
tors of eryth­
ro­
cyte sur­vival do © 2021 by The Amer­i­can Society of Hematology
not cor­re­late with acute vaso-occlu­sive cri­ses in patients with sickle cell DOI 10.1182/hema­tol­ogy.2021000265

340  |  Hematology 2021  |  ASH Education Program


INHERITED RED CELL DISORDERS BEYOND HEMOGLOBINOPATHIES

Diagnosis and clinical management


of enzymopathies

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/341/1851446/341luzzatto.pdf by guest on 13 December 2021


Lucio Luzzatto
Department of Hematology and Blood Transfusion, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania; and University of
Florence, Firenze, Italy

At least 16 genetically determined conditions qualify as red blood cell enzymopathies. They range in frequency from ultra-
rare to rare, with the exception of glucose-6-phosphate dehydrogenase deficiency, which is very common. Nearly all these
enzymopathies manifest as chronic hemolytic anemias, with an onset often in the neonatal period. The diagnosis can be
quite easy, such as when a child presents with dark urine after eating fava beans, or it can be quite difficult, such as when
an adult presents with mild anemia and gallstones. In general, 4 steps are recommended: (1) recognizing chronic hemolytic
anemia; (2) excluding acquired causes; (3) excluding hemoglobinopathies and membranopathies; (4) pinpointing which
red blood cell enzyme is deficient. Step 4 requires 1 or many enzyme assays; alternatively, DNA testing against an appro-
priate gene panel can combine steps 3 and 4. Most patients with a red blood cell enzymopathy can be managed by good
supportive care, including blood transfusion, iron chelation when necessary, and splenectomy in selected cases; however,
some patients have serious extraerythrocytic manifestations that are difficult to manage. In the absence of these, red blood
cell enzymopathies are in principle amenable to hematopoietic stem cell transplantation and gene therapy/gene editing.

LEARNING OBJECTIVES
• Update clinical, hematologic, biochemical, and molecular approaches to the diagnosis of red blood cell enzymop-
athies
• Pinpoint the role of supportive treatment, targeted treatment, and advanced forms of treatment in the manage-
ment of red blood cell enzymopathies
• Update knowledge about the molecular basis of red blood cell enzymopathies

Introduction essential for the detoxification of hydrogen peroxide (H2O2)


Red blood cell enzymopathies are genetic disorders affecting and of other reactive oxygen species (ROS). ATP is produced
the intraerythrocytic metabolism.1,2 Red blood cells, having by reactions of the “classic” glycolytic pathway; NADPH is
sacrificed their nucleus and other organelles to the unself- produced by the pentose phosphate pathway (Figure 1).3
ish task of providing oxygen to all other cells, are limited in Therefore, it is natural to classify red blood cell enzymopa-
their capacity to withstand metabolic impairment and also thies according to these 2 groups. A third group comprises
limited in how they will be affected: when one important en- enzymes involved in nucleotide metabolism (Table 1).
zyme is deficient, the red blood cell life span is nearly always Features of hemolytic anemias due to enzymopathies
compromised, with consequent hemolysis. Most red blood can be best illustrated through clinical summaries of indi-
cell enzymes are ubiquitously expressed “housekeeping” en- vidual patients.
zymes (Table 1); therefore, there may be pathology in other
tissues as well. Because red blood cells do not have protein
synthesis, however, they are in general more vulnerable.
The main metabolic fuel of the red blood cell is plasma CLINICAL CASE
glucose, and its breakdown within the red blood cell provides A 1-year-old boy born in 1963 from parents not known to
2 key compounds: adenosine triphosphate (ATP), required be consanguineous, although both were from Morcone
mainly for the operation of the cation pump, and nicotinamide (a small town not far from Naples, Italy), was found by his
adenine dinucleotide phosphate, reduced (NADPH), required local pediatrician to have anemia with reticulocytosis. Since
mainly for keeping up the supply of reduced glutathione (GSH) birth he had had moderate jaundice that did not respond
Prakash Singh Shekhawat
Red blood cell enzymopathies | 341
Table 1.  List of red blood cell enzymopathies under­ly­ing CNSHA*

Extraerythrocytic
Chromosomal
Enzyme (Acronym) Gene Clinical Manifesta- Notes
location
tions†
Glycolytic Hexokinase (HK) HK1 10q22 May ben­e­fit from sple­nec­tomy;
path­way36 bone marrow transplantation
(BMT) has been done.
Glucose-6-phos­phate GPI 19q31.1 Rarely myop­a­thy, Ranks sec­ond in fre­quency within
isom­er­ase (GPI) central nervous the gly­co­lytic path­way group
system (CNS) (after PK). May ben­e­fit from
sple­nec­tomy.
Phosphofructokinase (PFK) PFKM 12q13.3 Myopathy, Primarily a mus­cle dis­ease, with
myoglobinuria glycogenosis in mus­cle. CNSHA

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/341/1851446/341luzzatto.pdf by guest on 13 December 2021


mild.7
Aldolase ALDOA 16q22-24 Myopathy, CNS Fever may trig­ger poten­tially fatal
mas­sive rhabdomyolysis.37
Triose phos­phate isom­er­ TPI1 12p13.31 CNS (severe), car­ Disease very rare, but same muta­tion
ase (TPI) dio­my­op­a­thy (p. Glu104Asp) found in sev­eral
cases, suggesting founder effect.
Glyceraldehyde-3- GAPDH 12p13.31- Myopathy Hemolytic ane­mia reported in some
phos­phate dehy­dro­ge­ cases but link with enzyme defi­
nase (GAPD) ciency not clearly established.
Bisphosphoglycerate BPGM 7q33 Nohemo­ly­sis; erythrocytosis can be
mutase (DPGM) attrib­uted to low 2,3-DPG, left-
shifted Hb-O2 dis­so­ci­a­tion curve,
rel­a­tive hyp­oxia.
Phosphoglycerate kinase PGK1 Xq21.1 CNS myop­a­thy May ben­e­fit from sple­nec­tomy;
(PGK) BMT has been done. Early-onset
par­kin­son­ism reported.38
Pyruvate kinase (PK) PKLR 1q22 Iron over­load Ranks first in fre­quency in the gly­co­
lytic enzyme group. May ben­e­fit
from sple­nec­tomy; BMT has been
done.
Redox16,39 Glucose-6-phos­phate dehy­ G6PD Xq28 Very rarely Inalmost all­cases, only AHA from
dro­ge­nase (G6PD) granulocytes exog­e­nous trig­ger‡; CNSHA only
with class I var­i­ants.
Glutathione synthase GSS 20q11.22 CNS May be asso­ci­ated with high
5-oxoproline and met­a­bolic
aci­do­sis.40
Glutathione reduc­tase GSR 8p12 Cataracts AHA from exog­e­nous trig­ger
(fav­ism).
γ-glutamylcysteine synthase GCLC 6p12.1 CNS Mutations affect cat­a­lytic sub­unit.41
Cytochrome b5 reduc­tase CYB5R3 22q13.2 CNS Methemoglobinemia rather than
(CBR) hemo­ly­sis.
Nucleotide Adenylate kinase (AK) AK1 9q34.11 CNS May ben­e­fit from sple­nec­tomy.
metab­o­lism
Pyrimidine 5′-nucle­o­tid­ase NTSC3A 7p14.3 Ranks third in fre­quency among all­
(P5N) red blood cell enzymopathies
(leav­ing aside G6PD). May ben­e­fit
from sple­nec­tomy.
*The molec­u­lar basis of an enzymopathy is in the muta­tion(s) in the respec­tive gene. Most enzymopathies are auto­so­mal reces­sive dis­or­ders.
Therefore, the patient has a muta­tion in each of the 2 alle­lic genes encoding the respec­tive enzyme: the muta­tion may be the same in both alleles
(homo­zy­gos­ity), or there may be 2 dif­fer­ent muta­tions (com­pound het­ero­zy­gos­ity or biallelic muta­tions). In most cases the muta­tion causes insta­
bil­ity of the pro­tein: since mature red blood cells can­not make pro­teins, the enzyme activ­ity with which a retic­u­lo­cyte is endowed will decay as
the red blood cell ages in cir­cu­la­tion.

Since we are deal­ing with ubiq­ui­tously expressed genes, the pres­ence of such man­i­fes­ta­tions is not sur­pris­ing. Whether they are pres­ent or not,
and to what extent, depends mainly on 3 fac­tors: (1) nonerythroid cells may express alter­na­tive forms of a par­tic­u­lar enzyme, from the same gene
or from other genes; (2) if the main mech­a­nism of defi­ciency is enzyme insta­bil­ity, cells other than eryth­ro­cytes may com­pen­sate by increased
bio­syn­the­sis; (3) if the main mech­a­nism of defi­ciency is a qual­i­ta­tive change (eg, in the active site), all­cells expressing the gene will be affected.
The last 2 fac­tors may explain why extraerythrocytic man­i­fes­ta­tions may vary even within the same enzymopathy.

G6PD defi­ciency is wide­spread in malaria-endemic areas, con­se­quent on malaria hav­ing been the selec­tive agent for this poly­mor­phism.42 In areas
endemic for Plasmodium vivax malaria, a test for G6PD defi­ciency should be car­ried out before giv­ing primaquine or tafenoquine, the only drugs
that pre­vent relapse. This has been a strong stim­u­lus for the devel­op­ment of quan­ti­ta­tive point-of-care tests for G6PD defi­ciency.43

342  |  Hematology 2021  |  ASH Education Program


Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/341/1851446/341luzzatto.pdf by guest on 13 December 2021

Figure 1. Red blood cell metabolism. Glycolysis (the Embden-Meyerhof pathway) generates ATP required for cation transport and for
membrane maintenance, while NADH maintains hemoglobin iron in a reduced state. The hexose monophosphate shunt generates
the NADPH that is used to reduce GSH, which protects the red blood cell against oxidant stress (Figure 7); 6-phosphogluconate, after
decarboxylation, can be recycled via pentose sugars to glycolysis. At the side of glycolysis is the Rapoport-Luebering cycle, which
provides 2,3-isphosphoglycerate (2,3-DPG): its level is a critical determinant of the oxygen affinity of hemoglobin. G6PD deficiency is
highly prevalent in many parts of the world (Figure 6); all other enzymopathies are rare to ultrarare. Among them, the order of preva-
lence is PK, followed by P5N, followed by GPI.

Prakash Singh Shekhawat


Red blood cell enzymopathies  | 343
to barbiturates. Over several years his hemoglobin (Hb) level Glycolytic enzymopathies
fluctuated in the range from 7 to 11g/dL, and he received sev- Patient 1 recapitulates the clinical and hematologic features of
eral blood transfusions. At the age of 18, when he was fully reas- patients not just with GPI deficiency but with chronic nonsphero-
sessed (B. Rotoli, MD, in Naples), physical examination showed cytic hemolytic anemia (CNSHA) due to a deficiency of any of
pallor, mild icterus, and splenomegaly. There were no neurolog- the glycolytic enzymes: chronic hemolysis, splenomegaly, gall-
ical signs or muscle weakness. Hb was 10.4 G/dL; mean corpus- stones. What varies greatly is the severity of these features, not
cular volume, 104 fL, reticulocytes, 480×109/L; and unconjugated only from one enzymopathy to another but even from patient to
bilirubin, 5mg/dL; erythrocyte morphology showed anisocytosis patient within the same enzymopathy. This explains why the age
and basophilic stippling in 4% of cells. There were no abnormal of presentation may range from death in utero to adulthood. The
hemoglobins, osmotic fragility was normal, and the autohemolysis anemia is normocytic and normochromic: if it appears to be mac-
test showed increased lysis not corrected by glucose. A 51Cr study rocytic, it is usually on account of marked reticulocytosis. A short-
showed reduction of the red blood cell half-life by about 40%, age of folic acid might also contribute to a high mean corpuscular
with increased radioactive uptake in both the spleen and liver. volume, and since the increased rate of erythropoiesis increases

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/341/1851446/341luzzatto.pdf by guest on 13 December 2021


A panel of red blood cell enzyme assays (A. Zanella, MD, Milan) the requirement of this vitamin, folic acid supplementation is rec-
revealed a marked deficiency of glucose-6-phosphate isomer- ommended. Although there is considerable individual variation in
ase (GPI).4 The patient, who had been placed on daily folic acid tolerating anemia, in all chronic anemias the body tends to adapt.
(5mg/day), continued to require an average of 2.5 blood units The best-known red blood cell intrinsic mechanism of adaptation
per year until, at the age of 25, he developed obstructive jaun- is an increase in bisphosphoglycerate (2,3-DPG) that shifts the
dice due to gallstones. He underwent surgery that included cho- Hb-O2 dissociation curve to the right: this can be expected if the
lecystectomy, choledochoduodenostomy, and splenectomy. The flow of glycolysis is impaired downstream, but not upstream, of the
spleen weighed 500g; microscopic examination showed marked Rapoport-Luebering cycle (Figure 1). Indeed, with phosphofructoki-
congestion, erythrophagocytosis, and small foci of erythropoie- nase deficiency 2,3-DPG decreases,7 but with pyruvate kinase (PK)
sis. A liver biopsy showed evidence of cholestasis. The Hb level deficiency, it increases, and the increased O2 delivery to the tissues
increased and remained for years in the range of 10 to 11g/dL; is of benefit to the patient.8
therefore, regular blood transfusions were no longer needed. The pathophysiology of hemolysis in this group of enzy-
Reticulocytes remained high (in the range of 250-350×109/L). At mopathies consists mainly of the removal of red blood cells by
the age of 27, the patient completed his doctorate in philosophy. macrophages, ie, extravascular hemolysis. That is why splenec-
When the patient was 31 years old, sequencing of the GPI gene tomy is so often beneficial (Figure 2). It cannot be claimed to
revealed that the patient was homozygous for a p.Q343R muta-
tion identical to that independently observed at about the same
time in a patient from Japan.5,6
At the age of 44, the patient developed severe jaundice that
persisted for weeks (the bilirubin peaked at 80mg/dL) and was
admitted to a unit that specialized in liver transplantation (Dr G.
Ramadori, Göttingen). Imaging studies revealed an accessory
spleen. A liver biopsy (reviewed by Dr Tania Roskam, Leuven)
showed massive bilirubin overload, cholestasis, ballooned hepato-
cytes, ceroidosis of the Kupffer cells, and moderate centrolobular
perisinusoidal fibrosis but no evidence of cirrhosis. On supportive
treatment (including transfusion of 10 units of blood), the patient
gradually improved. Quantitative magnetic resonance-imaging
studies (R. Galanello, MD, Cagliari) revealed significant iron over-
load in the liver; deferoxamine was recommended as the iron-
chelating agent least likely to be hepatotoxic.
At the age of 52, after 3 further episodes of severe jaundice,
imaging studies showed stenosis of the choledochoduodenos-
tomy with proximal dilatation of the common duct, numerous
stones, possible cholangitis, and evidence of iron in the acces-
sory spleen and in the liver. The patient underwent surgery
(Dr A.D. Pinna, Bologna) consisting of removing innumerable
stones, including a large one, reconstructing the biliary-intes-
tinal anastomosis, and removing the accessory spleen. Immu- Figure 2. Splenectomy does not cure but does ameliorate CN-
nization against capsulated bacteria was carried out. Since that SHA of PK deficiency. In each of these 10 patients with PK de-
time the patient has been stable clinically and hematologically, ficiency, a significant increase in the steady-state hemoglobin
with an Hb of about 11 g/dL, reticulocytes about 4 times the level was seen after splenectomy (left panel). Whereas in most
upper normal limit, and mildly elevated bilirubin. The patient, types of CNSHA an improvement of anemia, resulting from de-
now 58, is head librarian in an academic institution in Rome; he creased hemolysis, is usually associated with a decrease in the
has recently published a book on the liberalism of the Italian phi- reticulocyte count, in PK deficiency reticulocytes often increase
losopher Benedetto Croce. The last time I had contact with him after splenectomy. This paradoxical phenomenon suggests that
it was to recommend that he receive a COVID-19 vaccination. the spleen selectively removes PK-deficient reticulocytes; the
mechanism is not yet well understood.48 From Zanella et al.8

344 | Hematology 2021 | ASH Education Program


PK deficiency G6PD deficiency
(autosomal) (X-linked)

Homozygous
normal

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/341/1851446/341luzzatto.pdf by guest on 13 December 2021


Heterozygous

Homozygous
deficient

Figure 3. Different phenotypes of heterozygotes for red blood cell enzymopathies. In a heterozygote for PK deficiency, encoded by
an autosomal gene (Table 1), the level of enzyme is about one-half of normal in all red blood cells. Since this level of enzyme is suffi-
cient, there are no clinical consequences—ie, PK deficiency is recessive. In a heterozygote for deficiency of G6PD, encoded by an X-
linked gene, the situation is quite different: X chromosome inactivation generates red blood cell mosaicism, whereby some red blood
cells are entirely normal, and others are G6PD deficient. Therefore, G6PD deficiency is expressed in heterozygotes: it is not recessive.

be a rad­i­cal treat­ment, but if it raises the Hb level suf­fi­ciently The autohemolysis test used in patient 1 was devel­oped
to avoid reg­u­lar blood trans­fu­sion, it will also avoid, in most by J.V Dacie.9 It was based on the obser­va­tion that when red
cases, the need for iron che­la­tion ther­apy, lead­ing to a sub­stan­ blood cells from patients with a wide range of hered­i­tary ane­
tial improve­ment in qual­ity of life. The acces­sory spleen was mias are incu­bated in vitro at 37 °C for 24 to 48 hours, some
an uncalled-for extra hitch in patient 1: over the years, being a frac­tion of them undergo hemo­ly­sis. If the abnor­mal­ity is in the
site of hemo­ly­sis, it may have become larger than it had been mem­brane, sup­ply­ing extra glu­cose usu­ally helps to reduce
orig­i­nally, and per­haps that is why it was missed dur­ing the first hemo­ly­sis; if the abnor­mal­ity is instead in gly­col­y­sis, glu­cose
oper­a­tion. does not help. It was a clever test that gave a strong hint—cor­
Except for phos­pho­glyc­er­ate kinase defi­ciency, which is rect in our case—that the patient had an enzymopathy. The
X-linked, all­other gly­co­lytic enzymopathies are auto­so­mal reces­ test is so eco­nom­i­cal that in prac­tice it is no lon­ger used. The
sive: ie, het­ero­zy­gotes are not affected. This must mean that a sus­pi­cion of enzymopathy must be raised in any patient with
50% reduc­tion in enzyme activ­ity (Figure 3) does not com­pro­mise chronic hemo­lytic ane­mia presenting early in life, or even later,
red blood cell sur­vival. Enzymopathy patients are either homo­zy­ who does not have a hemo­glo­bin­op­a­thy and the mor­phol­ogy
gous for a loss of func­tion muta­tion, or they have 2 dif­fer­ent muta­ char­ac­ter­is­tic of red blood cell membranopathies; there­fore,
tions in the 2 alleles at the same locus (com­pound het­ero­zy­gotes, sound clin­i­cal hema­tol­ogy is still par­a­mount. Once the sus­pi­
also referred to as biallelic). Whenever a patient is homo­zy­gous cion is for­mu­lated, the choice is between a bat­tery of quan­
for a very rare muta­tion, it is likely that the par­ents, even if not ti­ta­tive enzyme assays and a geno­mic approach. Today the
known to be con­san­guin­e­ous, may be ances­trally related. In other lat­ter is prob­a­bly pref­er­a­ble: it con­sists in obtaining from the
genetic dis­or­ders, there are now algo­rithms aiming to esti­mate patient’s DNA the sequence of genes whose muta­tions may
the prob­a­bil­ity that a par­tic­u­lar pre­vi­ously unknown muta­tion is or cause a red blood cell enzymopathy, or con­gen­i­tal hemo­lytic
is not path­o­genic. In the case of enzymopathies, this is not gen­er­ ane­mia in gen­eral.10 This approach has been used suc­cess­fully
ally nec­es­sary because a loss of enzyme activ­ity is in itself a good with a panel of 71 genes,11 at a price (to the Ital­ian National
read­out of path­o­ge­nic­ity. Health Service) of about $950 per patient; sev­eral com­mer­cial
Prakash Singh Shekhawat
Red blood cell enzymopathies  | 345
Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/341/1851446/341luzzatto.pdf by guest on 13 December 2021
Figure 4. Red blood morphology in select enzymopathies. Left panel: in a patient with CNSHA due to PK deficiency, the blood smear
is not diagnostic; however, the presence of “prickle red cells” should raise suspicion. From Mahendra et al.49 Middle panel: “bite cells,”
hemighosts and microspherocytes, characteristic of oxidative hemolysis, are seen in this smear from a child who received a dap-
sone-chlorproguanyl combination for the treatment of acute P. falciparum malaria. From Pamba et al.50 Right panel: red blood cells
with fine basophilic stippling in a patient with P5N deficiency. This enzymopathy is the most common inherited cause of basophilic
stippling, which is also seen with lead poisoning (lead inhibits many enzymes, including P5N, and it can produce a phenocopy of P5N
deficiency). From Rees, Duley, and Marinaki.51

pan­els are now avail­­able. The geno­mic approach is also advan­ dis­abling dis­ease can still hold on and have a qual­ity of life that
ta­geous when the patient is heavily trans­fused, mak­ing a red enables him to con­trib­ute to the life and cul­ture of oth­ers.
blood cell enzyme assay unre­li­able.
Among the gly­co­lytic enzymopathies, the one that has been
best char­ac­ter­ized in terms of man­age­ment is PK defi­ciency (red
blood cell mor­phol­ogy in Figure 4),12 mainly because it is the CLINICAL CASE
most com­mon (although, with an esti­mated fre­quency in the A 3-year-old girl from Egypt was seen in London for an assess­ment
United States of 5 per 100 000, it is still a rare dis­ease); there­fore, of her clin­i­cal state. Her par­ents also requested genetic coun­sel­
the range of the clin­i­cal bur­den is bet­ter known. From the clin­i­ ing. The patient was the cou­ple’s first­born. Her height and weight
cal point of view, it is note­wor­thy that iron over­load is fre­quent, were in the lower 10th per­cen­tile, and she was thin but did not
even in patients who are not trans­fu­sion depen­dent.13 This may appear mal­nour­ished. She had bilat­eral choreoathetosis, invol­un­
be due to hepcidin sup­pres­sion, con­sis­tent with a com­po­nent tary mus­cle move­ments and spasms, and upward eye move­ments;
of inef­fec­tive eryth­ro­poi­e­sis in the ane­mia of PK defi­ciency.14 her speech was lim­ited and dysarthric. Her his­tory was highly sig­
In this respect it is inter­est­ing that, upon diag­nos­tic test­ing of nif­i­cant in that she had been born at 35 weeks’ ges­ta­tion and at
inherited ane­mias by sequenc­ing a gene panel, sev­eral patients birth had had severe jaun­dice, but no hos­pi­tal records were avail­­
who had been diag­ nosed with con­ gen­i­
tal dyserythropoietic able. On one occasion after eating fava beans, she had become
ane­mia had in fact PKLR muta­tions, ie, PK defi­ciency.11 lethargic and had passed dark urine. At the time of her visit, her
Mitapivat, an allo­ste­ric acti­va­tor of PK (Figure 5), pro­duced a blood count, includ­ing retic­u­lo­cytes, was nor­mal: there was no
sig­nif­i­cant increase in hemo­glo­bin level in one-half of 52 patients evi­dence of CNSHA. The 3 mem­bers of the fam­ily were all­Rh+,
with PK defi­ciency in a phase 3 multicentric clin­i­cal trial; this was and there was no ABO incom­pat­i­bil­ity setup. Red blood cell glu­
asso­ci­ated with a decrease in bil­i­ru­bin and in retic­u­lo­cytes.15 cose-6-phos­phate dehy­dro­ge­nase (G6PD) assays yielded val­ues
Mitapivat may be on track to become the first approved drug for of 1.5 in the patient, 7.2 in her mother, and 0.6 in her father (nor­mal
a red blood cell enzymopathy. val­ues, 7-10 IU/g Hb). The most likely diag­no­sis was severe neu­
In the man­age­ment of patient 1, at least 7 senior col­leagues ro­log­i­cal dam­age from kernicterus. We inferred from the G6PD
from 4 dif­fer­ent countries with spe­cial­ized exper­tise in dif­fer­ent results that the father was hemi­zy­gous G6PD defi­cient: sequence
areas have been involved: I found their help invalu­able. At the anal­y­sis of his G6PD gene iden­ti­fied a pre­vi­ously unknown muta­
same time, it must be noted that a per­son with a poten­tially tion, p.N135T, that was hence named G6PD Cairo; the daugh­ter

346  |  Hematology 2021  |  ASH Education Program


Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/341/1851446/341luzzatto.pdf by guest on 13 December 2021
Figure 5. Mitapivat is an allosteric activator of PK. Top panel: the sigmoid-shaped dependence of PK activity on the concentration of
AG-348 (mitapivat) suggests cooperative interaction among the PK subunits. Bottom panel: diagram of the three-dimensional struc-
ture of the PK tetramer with bound mitapivat (gray). Blue: the substrate phosphoenolpyruvate PEP; brown: the physiological activator
fructose-1,6-bisphosphate (FBP). Modified from Kung et al.52

was het­ero­zy­gous and the mother, nor­mal. The mode of inher­ In G6PD-defi­cient per­sons, AHA is a pro­to­type exam­ple of a
i­tance of G6PD defi­ciency and its impli­ca­tions were explained gene-envi­ron­ment inter­ac­tion caus­ing a dis­ease that can be
to the fam­ily. Father and daugh­ter were advised not to eat fava life-threat­en­ing. Favism in chil­dren can be fatal if not promptly
beans. We informed them that the prob­lem of neo­na­tal jaun­dice treated, and rasburicase, poten­tially a life­saver in a patient with
(NNJ) was unlikely to recur if a sub­se­quent baby were male, but an incum­bent tumor lysis syn­drome, can become a seri­ous haz­
unfor­tu­nately, it might recur in a female, although it might be less ard if the patient hap­pens to be G6PD defi­cient (Figure 7B)—a
severe. We empha­sized the impor­tance of hav­ing the baby in a typ­i­cal exam­ple of a pharmacogenetic haz­ard.20
loca­tion where NNJ could be appro­pri­ately man­aged. Patient 2 illus­trates that NNJ, a clin­i­cal man­i­fes­ta­tion of G6PD
defi­ciency known for decades,21,22 is no less impor­tant than
AHA. In most G6PD defi­cient babies, the hyperbilirubinemia is
Redox enzymopathies mild: it over­laps with “phys­i­o­log­i­cal jaun­dice” of the new­born.
The most prominent enzymopathy in this group is indeed G6PD However, in some cases, like this one, it was severe enough to
defi­ciency.16 From an epi­de­mi­o­log­i­cal point of view, whereas the cause kernicterus. In many countries, G6PD defi­ciency is the
gly­co­lytic enzymopathies are all­rare to ultra­rare, here we are most com­mon cause of severe NNJ: in 1 study in Nigeria among
at the other end of the spec­trum: G6PD defi­ciency is pres­ent in babies admit­ted to the hos­pi­tal after show­ing clin­i­cal signs of
an esti­mated 500 mil­lion peo­ple world­wide (Figure 6). From a kernicterus, the fre­quency of G6PD defi­ciency was 78%—com­
clin­i­cal point of view, we are deal­ing with a very dif­fer­ent sit­u­a­ pared to 22% in the gen­eral pop­u­la­tion.23 The under­ly­ing G6PD
tion. Whereas the gly­co­lytic enzymopathies cause life­long CN- var­i­ant was G6PD A−, the same that accounts for many cases
SHA, G6PD defi­ciency in itself does not cause ane­mia and is not of NNJ, includ­ing severe ones, in the United States and one of
even a dis­ease; rather, it is a genetic abnor­mal­ity that in most the rea­sons why neo­na­tal screen­ing for G6PD defi­ciency has
cases remains asymp­tom­atic for a life­time (except for the very been advo­cated.17,24 The rea­sons for sever­ity may be envi­ron­
rare variants that cause CNSHA, clinically similar to that from men­ tal (eg, infec­ tion, expo­sure to naph­ tha­ lene) or genetic
glycolytic enzymopathies16). However, it can man­i­fest at birth (coex­is­tence of 1 or 2 UGT1A1 mutant alleles),25 and they are
through NNJ (as in patient 2) or at any age,17 like a thun­der­bolt in part unknown. The man­age­ment of G6PD-related NNJ is no
out of a blue sky, in the form of acute hemo­lytic ane­mia (AHA), dif­fer­ent from that due to other causes: this child should have
when the per­son is chal­lenged by an agent that causes oxi­da­ had exchange blood trans­fu­sion, but unfor­tu­nately, she did not.
tive dam­age to the red blood cells (Figure 4).18 The agent may G6PD Cairo, the G6PD var­i­ant first iden­ti­fied in patient 2, was
be the inges­tion of fava beans, or infec­tion, or a drug (prob­a­bly sub­se­quently found to be quite com­mon in Egypt and else­
in that order of fre­quency: for the mech­a­nism, see Figure 7).19 where, par­tic­u­larly in patients presenting with acute fav­ism.26

Prakash Singh Shekhawat


Red blood cell enzymopathies  | 347
Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/341/1851446/341luzzatto.pdf by guest on 13 December 2021
Figure 6. Epidemiology of G6PD deficiency throughout the world. Each country on the map is shaded in a color based on the best
estimate of the mean frequency of G6PD deficiency allele(s) in that country (this is the same as the frequency of G6PD-deficient
males). The larger panel gives a color-coded list of 10 common G6PD variants associated with G6PD deficiency: asterisk-shaped sym-
bols in the corresponding colors are shown in the countries where these variants have been observed (for graphic reasons symbols
could not be inserted in all countries where the respective variants are present). Modified from Luzzatto, Ally, and Notaro.16

The impor­tance of G6PD being X-linked can­not be overem- mark­edly defi­cient so that in an indi­vid­ual female the pro­por­
phasized. First, since males have only 1 X chro­mo­some, they tion of G6PD defi­cient red blood cells may be high (even up to
are either hemi­zy­gous G6PD nor­mal or hemi­zy­gous G6PD defi­ 90%). This last point is clin­i­cally rel­e­vant, as it was unfor­tu­nately
cient; females can be homo­zy­gous nor­mal or G6PD defi­cient in our patient 2.
(biallelic, whether homo­zy­gous or com­pound het­ero­zy­gous) Given the high fre­quency of G6PD defi­ciency in many parts
or het­ero­zy­gous for G6PD defi­ciency (ie, with 1 nor­mal allele). of the world (Figure 6), blood donors may be G6PD defi­cient.
Second, in any pop­u­la­tion, according to the Hardy-Weinberg A recent care­ful inves­ti­ga­tion has found that after blood bank
rule, het­ero­zy­gous females are more com­mon than hemi­zy­gous stor­age for 6 weeks the major­ity of G6PD-defi­cient blood units
G6PD-defi­cient males (Table 2). Third, since G6PD is sub­ject to still meet, 24 hours after trans­fu­sion, the in vivo red blood cell
X chro­mo­some inac­ti­va­tion, in het­ero­zy­gous females there is sur­vival tar­get of >75%.27
red blood cell mosa­i­cism, whereby a pro­por­tion of red blood Some ultra­rare enzymopathies in the redox cat­eg ­ ory involve
cells are just as defi­cient in G6PD activ­ity as in a hemi­zy­gous either the bio­syn­the­sis or the func­tion of GSH (Table 1). GSH
male (Figure 3). In het­ero­zy­gotes the expres­sion of G6PD defi­ reduc­tase defi­ciency can man­i­fest as fav­ism,28 confirming the
ciency is highly var­i­able, and since X inac­ti­va­tion can be ran­ key role of GSH in the defense of red blood cells against oxi­da­
domly “skewed,” their enzyme lev­ els range from nor­ mal to tive stress.

348  |  Hematology 2021  |  ASH Education Program


A Divicine
Primaquine
(Fava Beans)

ROS
from Neutrophils

O2- NADP
Glucose 6-Phosphate

Glucose 6-Phosphate
Superoxide Dismutase Dehydrogenase
Rasburicase
GSH 6-Phosphoglucono-
Uric Acid H2O2
δ-Lactone
Glutathione
Catalase Reductase NADPH
Prx2-SH-

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/341/1851446/341luzzatto.pdf by guest on 13 December 2021


6-Phosphogluconolactonase
H2O GSSG
Glutathione 6-Phosphogluconate
Peroxidase
6-Phosphogluconate
Prx2-S-S-
Dehydrogenase

Ribulose 5-Phosphate
B Divicine Primaquine
(Fava Beans)

Oxidative
ROS
Damage
O2-
from Neutrophils
Glucose 6-Phosphate
NADP Glucose 6-Phosphate
Superoxide Dismutase
Dehydrogenase
Rasburicase
GSH 6-Phosphoglucono-δ-Lactone
Uric Acid H2O2
Glutathione 6-Phosphoglucono
Catalase Reductase Lactonase
Prx2-SH-
H2O GSSG NADPH 6-Phosphogluconate
Glutathione
Peroxidase 6-Phosphogluconate
Prx2-S-S- Dehydrogenase

Ribulose 5-phosphate
Figure 7. The role of G6PD in protecting red blood cells from oxidative damage. (A) In G6PD-normal red blood cells, G6PD and
6-phosphogluconate dehydrogenase—2 of the enzymes of the PPP—provide an ample supply of NADPH, which in turn regenerates
GSH when this is oxidized by ROS (eg, superoxide, O2− and H2O2). Thus, when O2− (meant here to represent itself and other ROS) is
produced by pro-oxidant compounds such as primaquine, or the glucosides in fava beans (divicine), or the oxidative burst of neutro-
phils, these ROS are rapidly neutralized; similarly, when rasburicase administered to degrade uric acid produces an equimolar amount
of H2O2, this is rapidly degraded by the combined action of GSH peroxidase, catalase, and Prx2 (peroxiredoxin-2: all 3 mechanisms
are NADPH dependent). (B) In G6PD-deficient red blood cells, where the enzyme activity is reduced, NADPH production is limited,
and it may not be sufficient to cope with the excess ROS generated by pro-oxidant compounds and the consequent excess H2O2.
This diagram also explains why a defect in GSH reductase has very similar consequences to G6PD deficiency. Modified from Luzzatto,
Nannelli, and Notaro.53 PPP, pentose phosphate pathway.

Cytochrome b5 reduc­tase (CBR) defi­ciency is an enzymop-


athy that does not cause hemo­lytic ane­mia: it instead causes
reces­sive con­gen­i­tal met­he­mo­glo­bi­ne­mia,29 often presenting as
*In red blood cells there is also an NADPH-depen­dent enzyme that can
cya­no­sis in infants, that may mis­lead one to sus­pect con­gen­i­tal
reduce met­he­mo­glo­bin back to hemo­glo­bin: it was for­merly called fla­vin
heart dis­ease. This NADH-depen­dent enzyme (for­merly known reduc­tase and is now known to be iden­ti­cal to bil­i­ver­din-IX reduc­tase.30
as diaph­o­rase), capa­ble of reduc­ing met­he­mo­glo­bin (Fe+++) back Once again it is from genetic evi­dence that we know that CBR is the
to hemo­glo­bin (Fe++), is encoded by CYB5R3.* Most mis­sense enzyme respon­si­ble for the phys­i­o­logic main­te­nance of hemo­glo­bin in
muta­tions in this gene cause only met­he­mo­glo­bi­ne­mia (type I the Fe++ state since muta­tions of CYB5R3 cause met­he­mo­glo­bi­ne­mia,
dis­ease), whereas non­sense muta­tions, dele­tions, and some spe­ whereas muta­tions of BLVRB do not.
Prakash Singh Shekhawat
Red blood cell enzymopathies  | 349
Table 2. G6PD defi­ciency in sev­eral sam­ple pop­u­la­tions

Frequency of G6PD defi­ciency


Frequency of G6PD defi­ciency in in females, %
Examples
(hemi­zy­gous) males, %
Heterozygous Homozygous
US 2.5 4.9 0.06
Nigeria 22 34 4.8
Sardinia 12 21 1.4
Thailand 7.3 13.5 0.5
Frequency in US from Chinevere et al44; in Nigeria, from Luzzatto and Allan45; in Sardinia, from Cocco et al46; in Thailand, from Bancone et al.47 The
fre­quency of G6PD defi­ciency in hemi­zy­gous males is iden­ti­cal to the fre­quency of the G6PD mutant allele (or the sum total of the fre­quen­cies if
more than 1 mutant allele is involved). From the male fre­quency, it is easy to cal­cu­late the fre­quen­cies of the female geno­types, on the assump­tion

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/341/1851446/341luzzatto.pdf by guest on 13 December 2021


that the pop­u­la­tion is in Hardy-Weinberg equi­lib­rium. The data in this table show that, as all­text­books say, homo­zy­gous females are few com­
pared to hemi­zy­gous males; on the other hand, het­ero­zy­gous females are almost twice as com­mon as hemi­zy­gous males (which most text­books
fail to say). Because of epi­ge­netic mosa­ic
­ ism con­se­quent on X chro­mo­some inac­ti­va­tion, all­het­ero­zy­gotes have some G6PD defi­cient red blood
cells: on aver­age 50%, but with a wide range of var­ia ­ ­tion. This is why in het­ero­zy­gotes clin­i­cal man­i­fes­ta­tions are, on aver­age, less severe, but not
always so. In every series of patients with fav­ism, there are always het­ero­zy­gous girls.19

cific mis­sense muta­tions cause, in addi­tion, a dev­as­tat­ing neu­ Nucleotide metab­o­lism enzymopathies
ro­log­i­cal syn­drome (type 2 dis­ease, prob­a­bly medi­ated by the P5N defi­ciency, although rare, ranks third in fre­quency among
role of the enzyme in the bio­syn­the­sis of mye­lin phos­pho­lip­ids). enzymopathies (after G6PD defi­ ciency and PK defi­ ciency).32
A point of prac­ti­cal impor­tance is that in patients with met­he­mo­ P5N, encoded by the gene NT5C3A, is par­tic­u­larly impor­tant
glo­bi­ne­mia (whether from CRB defi­ciency or from hemo­glo­bin dur­ing the last stage of ery­throid mat­u­ra­tion. Reticulocytes,
M) pulse oximetry mea­sured with bed­side devices is not reli­able. hav­ ing no RNA syn­ the­
sis, are unable to recy­ cle pyrim­ i­
dine
nucle­o­tides aris­ing from RNA deg­ra­da­tion (as would be the
case in other cells), and these must be hydro­lyzed to nucle­
o­sides before they can exit through the mem­brane. With P5N
CLINICAL CASE
defi­cient, encum­brance by pyrim­i­dine nucle­o­tides seems to
A 12-year-old boy from Izmir, Turkey, the first­born from first-cousin hin­der the abil­ity of retic­u­lo­cytes to get rid of their last ribo­
par­ents with nor­mal blood counts, was referred to an aca­demic somes or of their rem­nants. These become vis­i­ble under the
hema­tol­ogy depart­ment because of hemo­lytic ane­mia diag­nosed shape of baso­philic stip­pling,33 a mor­pho­log­i­cal abnor­mal­ity
at the age of 2. The ane­mia was mod­er­ate most of the time but with char­ac­ter­is­tic of P5N defi­ciency (Figure 4) but not unique to
exac­er­ba­tions, con­com­i­tant to infec­tion, that some­times required it (see patient 1). The hema­to­logic con­se­quences of this en-
blood trans­fu­sion. Physical exam­i­na­tion revealed pal­lor, jaun­dice, zymopathy are illus­trated by patient 3, who has a mod­er­ately
hepa­to­meg­aly (2 cm below the cos­tal bor­der) and spleno­meg­aly severe CNSHA.
(4cm below the cos­tal bor­der). He had ane­mia, with a hemo­glo­ The other (ultra­rare) enzymopathy in this group is adenylate
bin of 7.5 g/dL and a reticulocytosis of 160 × 109/L. White blood cell kinase (AK) defi­ciency. AK, by cat­a­lyz­ing the inter­con­ver­sion
count, plate­lets, and liver enzymes were within the nor­mal range. A of ADP into ATP and adenosine mono-phosphate (AMP), is gen­
direct Coombs test was neg­a­tive. The patient showed an increased er­ally impor­tant in con­trol­ling the level of these com­pounds.3
uncon­ju­gated bil­i­ru­bin of 2.1 mg/dL (upper normal limit 0.8 mg/dL) In red blood cells ATP is cru­cial for the cat­ion pump, and AMP,
and lac­tate dehy­dro­ge­nase 678 IU (upper normal limit 300). Hapto- a pre­cur­sor of the sig­nal­ing mol­e­cule cyclic AMP,3 may be
globin was unde­tect­able. A periph­eral blood smear revealed aniso- impor­tant in maintaining eryth­ro­cyte deformability.34 Indeed,
poikilocytosis, mac­ro­cytes, poly­chro­matic cells, rare elliptocytes in AK defi­ciency there is evi­dence of intra­vas­cu­lar hemo­ly­sis.35
and spherocytes, and baso­philic stip­pling (Figure 4). Hemoglobin For both P5N and AK, it is through their respec­tive genetic
elec­tro­pho­re­sis was nor­mal, and a test for unsta­ble hemo­glo­bin defects that we know they have an essen­tial func­tion in red
was neg­a­tive. Folic acid and vita­min B12 lev­els were nor­mal. An blood cells.
osmotic fra­gil­ity test was nor­mal. A bone mar­row aspi­rate showed
ery­throid hyper­pla­sia; an iron stain did not reveal sideroblasts. An Conclusion
enzyme assay panel yielded nor­mal val­ues of gly­co­lytic enzymes Red blood cell enzymopathies are now under­stood at the bio­
and of G6PD. The ultra­vi­o­let spec­trum of an extract of red blood chem­i­cal and molec­u­lar lev­els, and their clin­i­cal and hema­to­
cell metab­o­lites showed an abnor­mal purine/pyrim­id ­ ine ratio, sug­ logic fea­tures are rea­son­ably well char­ac­ter­ized. G6PD defi­
ges­tive of pyrim­i­dine 5′-nucle­o­tid­ase (P5N) defi­ciency. Sequencing ciency stands out in terms of epi­de­mi­­ol­ogy because it is a
of the NT5C3 gene in the patient’s DNA revealed homo­zy­gos­ity for wide­spread, mostly asymp­tom­atic abnor­mal­ity: it bears wit­
a frame­shift muta­tion (c.393-394del TA, K132Rfs7*). Both par­ents ness to the role of malaria in shap­ing human evo­lu­tion, and its
were het­ero­zy­gous for the same muta­tion.31 clin­i­cal man­i­fes­ta­tions are largely pre­vent­able and man­age­
able. Although rec­og­niz­ing chronic hemo­lytic ane­mia is not dif­
fi­cult, the other enzymopathies are prob­a­bly still underdiag-
Courtesy of Dr Paola Bianchi. nosed; DNA test­ing by gene pan­els should grad­u­ally over­come
this diag­nos­tic gap. In con­trast, the targeted treat­ment of red

350  |  Hematology 2021  |  ASH Education Program


blood cell enzymopathies is nearly uncharted ter­ri­tory. Since novel frame­shift dele­tion in the PKLR gene (p.Arg518fs), and low hepcidin
most of them qual­ify as orphan dis­eases, there are incen­tives to fer­ri­tin ratios. Br J Haematol. 2014;165(4):556-563.
15. Grace RF, Rose C, Layton DM, et al. Safety and effi­cacy of mitapivat in pyru­
to devel­op­ing new ther­a­pies. vate kinase defi­ciency. N Engl J Med. 2019;381(10):933-944.
16. Luzzatto L, Ally M, Notaro R. Glucose-6-phos­phate dehy­dro­ge­nase defi­
Acknowledgments  ciency. Blood. 2020;136(11):1225-1240.
I have been for­tu­nate to work on red blood cell enzymopathies, 17. Kaplan M, Hammerman C. The need for neo­na­tal glu­cose-6-phos­phate
dehy­dro­ge­nase screen­ing: a global per­spec­tive. J Perinatol. 2009;29(suppl
par­tic­u­larly G6PD defi­ciency, in 5 countries. Since I can­not list
1):S46-S52.
all­those to whom I am grate­ful, I will only men­tion Olugbemiro 18. Luzzatto L, Poggi VE. Glucose-6-phos­phate dehy­dro­ge­nase defi­ciency. In:
Sodeinde (Ibadan), the late Graziella Persico (Naples), Tom Vul- O’rkin SH, Fisher DE, Ginsburg D, Look TA, Lux SE, Nathan DG, eds. Nathan
liamy (London), Letizia Longo (New York), Rosario Notaro (Flor- and Oski’s Hematology and Oncology of Infancy and Childhood. Vol. 1.
ence), and Stella Malangahe (Dar es Salaam). Research sup­port Elsevier Saunders; 2015:609-629.
19. Luzzatto L, Arese P. Favism and glu­cose-6-phos­phate dehy­dro­ge­nase defi­
was received from the World Health Organization, National Re-
ciency. N Engl J Med. 2018;378(1):60-71.
search Council (Italy), Medical Research Council (UK), and Na- 20. Zaramella P, De Salvia A, Zaninotto M, et  al. Lethal effect of a sin­gle

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/341/1851446/341luzzatto.pdf by guest on 13 December 2021


tional Institutes of Health (in Nigeria and in the US). I am espe­ dose of rasburicase in a pre­term new­born infant. Pediatrics. 2013;131(1):
cially grate­ful to all­patients with enzymopathies for what I have e309-e312.
learned from them. 21. Doxiadis SA, Fessas P, Valaes T. Glucose-6-phos­ phate dehy­ dro­ge­
nase
defi­ciency. A new aetiological fac­tor of severe neo­na­tal jaun­dice. Lancet.
1961;1(7172):297-301.
Conflict-of-inter­est dis­clo­sure 22. Capps FP, Gilles HM, Jolly H, Worlledge SM. Glucose-6-phos­phate dehy­
Lucio Luzzatto: no com­pet­ing finan­cial inter­ests to declare. dro­ge­nase defi­ciency and neo­na­tal jaun­dice in Nigeria: their rela­tion to the
use of pro­phy­lac­tic vita­min K. Lancet. 1963;2(7304):379-383.
23. Bienzle U, Effiong C, Luzzatto L. Erythrocyte glu­cose 6-phos­phate dehy­
Off-label drug use
dro­ge­nase defi­ciency (G6PD type A−) and neo­na­tal jaun­dice. Acta Paedi-
Lucio Luzzatto: nothing to disclose. atr Scand. 1976;65(6):701-703.
24. Kaplan M, Hammerman C. Glucose-6-phos­ phate dehy­ dro­ge­ nase defi­
Correspondence ciency: a hid­den risk for kernicterus. Semin Perinatol. 2004;28(5):356-364.
Lucio Luzzatto, Department of Hematology and Blood Transfusion, 25. Huang MJ, Kua KE, Teng HC, Tang KS, Weng HW, Huang CS. Risk fac­tors for
severe hyperbilirubinemia in neo­na­tes. Pediatr Res. 2004;56(5):682-689.
Muhimbili University of Health and Allied Sciences, 65001 Dar es Sa- 26. Reading NS, Sirdah MM, Shubair ME, et  al. Favism, the commonest form
laam, Tanzania; e-mail: lluzzatto@blood​­.ac​­.tz. of severe hemo­lytic ane­mia in Palestinian chil­dren, varies in sever­ity with
three dif­fer­ent var­i­ants of G6PD defi­ciency within the same com­mu­nity.
References Blood Cells Mol Dis. 2016;60:58-64.
1. Luzzatto L. Hemolytic ane­mias. In: Jameson JL, Fauci AS, Kasper DL, Hauser 27. Francis RO, D’Alessandro A, Eisenberger A, et al. Donor glu­cose-6-phos­phate
SL, Longo DL, Loscalzo J, eds. Harrison’s Principles of Internal Medicine. dehy­dro­ge­nase defi­ciency decreases blood qual­ity for trans­fu­sion. J Clin
Vol. 1. McGraw-Hill Education; 2018:708-723. Invest. 2020;130(5):2270-2285.
2. Grace RF, Glader B. Red blood cell enzyme dis­or­ders. Pediatr Clin North 28. Kamerbeek NM, van Zwieten R, de Boer M, et al. Molecular basis of glu­ta­thi­
Am. 2018;65(3):579-595. one reduc­tase defi­ciency in human blood cells. Blood. 2007;109(8):3560-
3. Berg JM, Tymoczko JL, Stryer L. Biochemistry. 5th ed. W. H. Freeman and 3566.
Co.; 2002. 29. Percy MJ, Lappin TR. Recessive con­gen­i­tal methaemoglobinaemia: cyto­
4. Alfinito F, Ferraro F, Rocco S, et  al. Glucose phos­phate isom­er­ase (GPI) chrome b(5) reduc­tase defi­ciency. Br J Haematol. 2008;141(3):298-308.
“Morcone”: a new var­i­ant from Italy. Eur J Haematol. 1994;52(5):263-266. 30. Komuro A, Tobe T, Hashimoto K, et al. Molecular clon­ing and expres­sion of
5. Baronciani L, Zanella A, Bianchi P, et  al. Study of the molec­u­lar defects human liver bil­i­ver­din-IX beta reduc­tase. Biol Pharm Bull. 1996;19(6):796-
in glu­cose phos­phate isom­er­ase-defi­cient patients affected by chronic 804.
hemo­lytic ane­mia. Blood. 1996;88(6):2306-2310. 31. Köker SA, Oymak Y, Bianchi P, et  al. A new homo­ zy­gous muta­ tion
6. Kanno H, Fujii H, Hirono A, et al. Molecular anal­y­sis of glu­cose phos­phate (c.393-394del TA/c.393-394del TA) in the NT5C3 gene asso­ ci­
ated with
isom­er­ase defi­ciency asso­ci­ated with hered­i­tary hemo­lytic ane­mia. Blood. pyrim­i­dine-5′-nucle­o­tid­ase defi­ciency: a case report. J Pediatr Hematol
1996;88(6):2321-2325. Oncol. 2019;41(8):e484-e486.
7. Vora S, Davidson M, Seaman C, et al. Heterogeneity of the molec­u­lar lesions 32. Zanella A, Bianchi P, Fermo E, Valentini G. Hereditary pyrim­i­dine 5′-nucle­o­
in inherited phos­pho­fruc­to­ki­nase defi­ciency. J Clin Invest. 1983;72(6):1995- tid­ase defi­ciency: from genet­ics to clin­i­cal man­i­fes­ta­tions. Br J Haematol.
2006. 2006;133(2):113-123.
8. Zanella A, Fermo E, Bianchi P, Chiarelli LR, Valentini G. Pyruvate kinase defi­ 33. Cheson BD, Rom WN, Webber RC. Basophilic stip­pling of red blood cells: a
ciency: the geno­type-phe­no­type asso­ci­a­tion. Blood Rev. 2007;21(4):217- non­spe­cific find­ing of mul­ti­ple eti­­ol­ogy. Am J Ind Med. 1984;5(4):327-334.
231. 34. Semenov AN, Shirshin EA, Muravyov AV, Priezzhev AV. The effects of dif­fer­
9. Dacie JV. Part 1—The con­gen­i­tal anae­mias. In: The Haemolytic Anaemias. ent sig­nal­ing path­ways in adenylyl cyclase stim­u­la­tion on red blood cells
Vol. 1. Churchill; 1960:1-339. deformability. Front Physiol. 2019;10:923.
10. Roy NB, Wilson EA, Henderson S, et  al. A novel 33-gene targeted rese- 35. Dongerdiye R, Kamat P, Jain P, et al. Red cell adenylate kinase defi­ciency in
quencing panel pro­vi­des accu­rate, clin­i­cal-grade diag­no­sis and improves India: iden­ti­fi­ca­tion of two novel mis­sense muta­tions (c.71A>G and c.413G>A).
patient man­ age­ ment for rare inherited anae­ mias. Br J Haematol. J Clin Pathol. 2019;72(6):393-398.
2016;175(2):318-330. 36. van Wijk R, van Solinge WW. The energy-less red blood cell is lost: eryth­
11. Russo R, Andolfo I, Manna F, et al. Multi-gene panel test­ing improves diag­ ro­cyte enzyme abnor­mal­i­ties of gly­col­y­sis. Blood. 2005;106(13):4034-
no­sis and man­age­ment of patients with hered­i­tary ane­mias. Am J Hematol. 4042.
2018;93(5):672-682. 37. Mamoune A, Bahuau M, Hamel Y, et al. A ther­mo­la­bile aldol­ase A mutant
12. Grace RF, Barcellini W. Management of pyru­vate kinase defi­ciency in chil­ causes fever-induced recur­rent rhabdomyolysis with­out hemo­lytic ane­
dren and adults. Blood. 2020;136(11):1241-1249. mia. PLoS Genet. 2014;10(11):e1004711.
13. van Beers EJ, van Straaten S, Morton DH, et al. Prevalence and man­age­ment 38. Sakaue S, Kasai T, Mizuta I, et  al. Early-onset par­kin­son­ism in a ped­i­gree
of iron over­load in pyru­vate kinase defi­ciency: report from the Pyruvate with phos­pho­glyc­er­ate kinase defi­ciency and a het­ero­zy­gous car­rier: do
Kinase Deficiency Natural History Study. Haematologica. 2019;104(2):e51- PGK-1 muta­tions con­trib­ute to vul­ner­a­bil­ity to par­kin­son­ism? NPJ Parkin-
e53. sons Dis. 2017;3:13.
14. Mojzikova R, Koralkova P, Holub D, et al. Iron sta­tus in patients with pyru­ 39. van Zwieten R, Verhoeven AJ, Roos D. Inborn defects in the anti­ox­i­dant
vate kinase defi­ ciency: neo­ na­
tal hyperferritinaemia asso­ ci­
ated with a sys­tems of human red blood cells. Free Radic Biol Med. 2014;67:377-386.
Prakash Singh Shekhawat
Red blood cell enzymopathies  | 351
40. Soylu Ustkoyuncu P, Mutlu FT, Kiraz A, Tag Balkis Z, Yel S. A rare cause of 47. Bancone G, Menard D, Khim N, et al. Molecular char­ac­ter­iza­tion and map­
neo­na­tal hemo­lytic ane­mia: glu­ta­thi­one syn­the­tase defi­ciency. J Pediatr ping of glu­ cose-6-phos­phate dehy­ dro­ ge­
nase (G6PD) muta­ tions in the
Hematol Oncol. 2018;40(1):e45-e49. Greater Mekong Subregion. Malar J. 2019;18(1):20.
41. Mañú Pereira M, Gelbart T, Ristoff E, et al. Chronic non-spherocytic hemo­ 48. van Vuren AJ, van Beers EJ, van Wijk R. A pro­posed con­cept for defec­tive
lytic ane­mia asso­ci­ated with severe neu­ro­log­i­cal dis­ease due to gam- mitophagy lead­ing to late stage inef­fec­tive eryth­ro­poi­e­sis in pyru­vate
ma-glutamylcysteine syn­ the­tase defi­ ciency in a patient of Moroccan kinase defi­ciency. Front Physiol. 2020;11:609103.
ori­gin. Haematologica. 2007;92(11):e102-e105. 49. Mahendra P, Dollery CT, Luzzatto L, Bloom SR. Pyruvate kinase defi­ciency.
42. Luzzatto L. G6PD defi­ciency: a poly­mor­phism bal­anced by het­ero­zy­gote Association with G6PD defi­ciency. BMJ. 1992;305(6856):760-762.
advan­tage against malaria. Lancet Haematol. 2015;2(10):e400-e401. 50. Pamba A, Richardson ND, Carter N, et al. Clinical spec­trum and sever­ity of
43. Pal S, Bansil P, Bancone G, et  al. Evaluation of a novel quan­ti­ta­tive test hemo­lytic ane­mia in glu­cose 6-phos­phate dehy­dro­ge­nase-defi­cient chil­
for glu­cose-6-phos­phate dehy­dro­ge­nase defi­ciency: bring­ing quan­ti­ta­tive dren receiv­ing dap­sone. Blood. 2012;120(20):4123-4133.
test­ing for glu­cose-6-phos­phate dehy­dro­ge­nase defi­ciency closer to the 51. Rees DC, Duley JA, Marinaki AM. Pyrimidine 5′ nucle­o­tid­ase defi­ciency. Br J
patient. Am J Trop Med Hyg. 2019;100(1):213-221. Haematol. 2003;120(3):375-383.
44. Chinevere TD, Murray CK, Grant E Jr, Johnson GA, Duelm F, Hospenthal 52. Kung C, Hixon J, Kosinski PA, et al. AG-348 enhances pyru­vate kinase activ­
DR. Prevalence of glu­cose-6-phos­phate dehy­dro­ge­nase defi­ciency in U.S. ity in red blood cells from patients with pyru­vate kinase defi­ciency. Blood.
Army per­son­nel. Mil Med. 2006;171(9):905-907. 2017;130(11):1347-1356.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/341/1851446/341luzzatto.pdf by guest on 13 December 2021


45. Luzzatto L, Allan NC. Relationship between the genes for glu­ cose-6- 53. Luzzatto L, Nannelli C, Notaro R. Glucose-6-phos­ phate dehy­ dro­
ge­nase
phos­phate dehy­dro­ge­nase and for haemoglobin in a Nigerian pop­u­la­tion. defi­ciency. Hematol Oncol Clin North Am. 2016;30(2):373-393.
Nature. 1968;219(5158):1041-1042.
46. Cocco P, Todde P, Fornera S, Manca MB, Manca P, Sias AR. Mortality in a
cohort of men expressing the glu­cose-6-phos­phate dehy­dro­ge­nase defi­ © 2021 by The Amer­i­can Society of Hematology
ciency. Blood. 1998;91(2):706-709. DOI 10.1182/hema­tol­ogy.2021000266

352  |  Hematology 2021  |  ASH Education Program


INHERITED RED CELL DISORDERS BEYOND HEMOGLOBINOPATHIES

Diamond-Blackfan anemia
Lydie M. Da Costa,1–4 Isabelle Marie,1,5 and Thierry M. Leblanc5

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/353/1851415/353costa.pdf by guest on 13 December 2021


Service d’Hématologie Biologique (Hematology Diagnostic Lab), AP­HP, Hôpital Robert Debré, Paris, France; 2University of Paris, Paris, France;
1

HEMATIM EA4666, Amiens, France; 4Laboratory of Excellence for Red Cells, LABEX GR­Ex, Paris, France; and 5Immuno­Hematology Department,
3

AP­HP, Hôpital Robert­Debré, Paris, France

Diamond-Blackfan anemia (DBA) is an inherited bone marrow failure syndrome, characterized as a rare congenital bone
marrow erythroid hypoplasia (OMIM#105650). Erythroid defect in DBA results in erythroblastopenia in bone marrow as
a consequence of maturation blockade between the burst forming unit–erythroid and colony forming unit–erythroid
developmental stages, leading to moderate to severe usually macrocytic aregenerative (<20×109/L of reticulocytes)
anemia. Congenital malformations localized mostly in the cephalic area and in the extremities (thumbs), as well as short
stature and cardiac and urogenital tract abnormalities, are a feature of 50% of the DBA-affected patients. A significant
increased risk for malignancy has been reported. DBA is due to a defect in the ribosomal RNA (rRNA) maturation as a
consequence of a heterozygous mutation in 1 of the 20 ribosomal protein genes. Besides classical DBA, some DBA-like
diseases have been identified. The relation between the defect in rRNA maturation and the erythroid defect in DBA has
yet to be fully defined. However, recent studies have identified a role for GATA1 either due to a specific defect in its
translation or due to its defective regulation by its chaperone HSP70. In addition, excess free heme-induced reactive oxy-
gen species and apoptosis have been implicated in the DBA erythroid phenotype. Current treatment options are either
regular transfusions with appropriate iron chelation or treatment with corticosteroids starting at 1 year of age. The only
curative treatment for the anemia of DBA to date is bone marrow transplantation. Use of gene therapy as a therapeutic
strategy is currently being explored.

LEARNING OBJECTIVES
• Recognize a case of DBA
• Manage anemia and potential clinical complications of DBA

CLINICAL CASE DBA with different phenotypes (Figure 1). Of note, in the
The family described below is an excellent illustration of 1980s, no DBA gene had been identifed, and steroids were
the various issues that are encountered in the diagnosis started following DBA diagnosis in all instances, which is
and treatment of Diamond­Blackfan anemia (DBA) due to not the recommended treatment currently: steroids should
incomplete penetrance. be initiated only after the frst year of life.1
1. The child, UPN#1447, at age 2.3 years with normal white
and platelet cell counts, was initially diagnosed with
The family is the largest of the 417 DBA families regis­ transient erythroblastopenia of childhood (TEC) on the
tered in the French DBA registry (Observatoire Français basis of an isolated severe normochromic, macrocytic
de l’Anémie de Blackfan­Diamond) and includes 3 gen­ mean corpuscular volume (MCV=91.4 fL) aregenerative
erations of affected patients (Figure 1). In this family, the (7.5×109/L reticulocytes) anemia (hemoglobin (Hb) level
mode of inheritance is familial, as is the case for 45% of the at 79g/L) with erythroblastopenia documented on the
DBA­affected patients registered in the different registries bone marrow smear (1% of total erythroblasts [Eb, nor­
around the world. In 55% of individuals, DBA is the result of mal 5%­30%]). Bone marrow cellularity was normal with
sporadic or de novo inheritance. Interestingly, in the illus­ no signs of dysplasia. Parvovirus B19 serology was nega­
trated family, all 4 children, 2 girls and 2 boys, developed tive for both immunoglobulin M and immunoglobulin G.

Prakash Singh Shekhawat


Diamond­Blackfan anemia, from a clinical case | 353
Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/353/1851415/353costa.pdf by guest on 13 December 2021
Figure 1. Family tree of the described DBA family.

The patient recov­ered with no treat­ment except for a short (60 × 109/L retic­u­lo­cytes). The char­ac­ter­is­tic erythroblastope­
course of ste­roid treat­ment (for a few weeks) with no red cell nia was noted on the bone mar­row smear with only 1% of Eb.
blood trans­fu­sions. At the same time, increased expres­sion UPN#1821 exhibited fea­tures of fetal eryth­ro­poi­e­sis with HbF
of hemo­glo­bin F (HbF) at 9% (nor­mal <2% at 6-24 months of at 18% and a large increase in eADA activ­ity at 5 nmol/min/mg
age) along with a sig­nif­i­cantly ele­vated eryth­ro­cyte aden­o­ Hb. A val­gus foot defor­mity was noted as the only con­gen­i­tal
sine deam­in ­ ase (eADA) activ­ity at 5.95 nmol/min/mg Hb (or malformation. UPN#1821 received a reg­u­lar dose of 2 mg/kg/d
U/g Hb) (nor­mal: 1.50 ± 0.2) has been reported. UPN#1447, ini­ of ste­roid for a month, which allowed for trans­fu­sion inde­pen­
tially diag­nosed with TEC, has been finally diag­nosed with dency, and was sub­se­quently maintained on low-dose ste­roid
DBA after the iden­ti­fi­ca­tion of a RPS19 gene muta­tion. In ret­ ther­apy (0.1  mg/kg/d). However, she became later ste­ roid
ro­spect, the asso­ci­a­tion of pure red blood cell hypo­plas­tic resis­tant, and she is cur­rently being reg­u­larly trans­fused every
ane­mia, in con­junc­tion with an increased per­cent­age of HbF 3 weeks (Table 2).
and eADA activ­ity, and response to ste­roid was in accor­dance 3. UPN#1822 was diag­nosed with DBA at 1 month of age. Surpris­
with this diag­no­sis.1,2 However, it is to be noted some cli­ni­ ingly at this time, the bone mar­row smear did not exhibit the
cians con­sider TEC to be one of the DBA phe­no­types with char­ac­ter­is­tic DBA erythroblastopenia with 31% of ery­throid
a low pen­e­trance.3 TEC and DBA should be dif­fer­en­ti­ated, pre­cur­sors. However, a large increase in eADA activ­ ity of
but it is some­times very dif­fi­cult to dis­crim­i­nate between 4 nmol/min/mg Hb was noted. Posterior hypo­spa­dias was no­
the 2 phe­no­types as only the time course of the evo­lu­tion ticed. He received an ini­tial dose of ste­roid at 2 mg/kg/d, and
of ane­mia can clearly dis­tin­guish between them (see Table 1 the dose was grad­u­ally decreased to 0.2 mg/kg/d, which en­
for dif­fer­en­tial diag­no­sis between TEC and DBA). In order to abled the sta­bi­li­za­tion of the Hb level. The ste­roid treat­ment
be cau­tious, we rec­om­mend a molec­u­lar screen­ing and the lasted for 18 years and 10 months, at which point the patient
care­ful fol­low-up of any child with TEC or inform the par­ents became ste­roid inde­pen­dent and main­tains a rea­son­able Hb
to bring back the child for fur­ther eval­u­a­tion in case of recur­ level with no need for ste­roids (Table 2).
rence of the ane­mia (Table 2). 4. Finally, UPN#1213, the fourth child of this fam­ily, was also a
. UPN#1821 was the sec­ond child of the fam­ily, a girl born 2 years
2 DBA-affected patient. He was diag­nosed at 1 month and 3
later. In con­trast to UPN#1447, UPN#1821 exhibited a 2-month- weeks of age with erythroblastopenia with 4% of Eb in the bone
old, severe ane­mia with a nadir of 20 g/L Hb. The ane­mia was mar­row. Like his sib­lings, his eADA activ­ity was increased to
normochromic and mac­ro­cytic (high MCV of 114.8 fL), which is 4.38 nmol/min/mg Hb. Hypospadias was once again noticed.
clas­si­cal for DBA, and with a mod­est degree of regen­er­a­tion He was treated with an ini­tial dose of 2 mg/kg/d of ste­roid,

354  |  Hematology 2021  |  ASH Education Program


Table 1. Differential diag­no­sis between DBA and tran­sient TEC

Characteristic DBA TEC


Median age at diag­no­sis 2 months >1 year
Inheritance Sporadic (55%) or dom­i­nant (45%) Not inherited
Congenital anom­a­lies In 50% None
Pure red blood cell aplasia (bone mar­row Yes Yes
biopsy) or erythroblastopenia (bone
mar­row aspi­ra­tion)
Hb level Low Low
Reticulocyte count <20 × 109/L <20 × 109/L
MCV Usually high Normal

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/353/1851415/353costa.pdf by guest on 13 December 2021


eADA activ­ity Normal to high Normal
HbF Normal to high Normal
Allelic var­i­a­tion in a RP gene or another 70% to 80% of the patients with No muta­tion found
gene involved in DBA-like cases DBA

which nor­mal­ized the Hb level. He is now 32 years old and i­tor stages.4 The other bio­log­i­cal fea­tures include increased eADA
still under­go­ing ste­roid ther­apy at 10 mg per day. At the last activ­ity, which is ele­vated in 90% of the nontransfused patients with
fol­low-up, he exhibited a mod­er­ate mac­ro­cytic (MCV at 105.9 DBA and the per­sis­tence of fea­tures of fetal eryth­ro­poi­e­sis (high HbF
fL) ane­mia with an Hb level at 108 g/L with 38.6 × 109/L retic­u­lo­ per­cent­age). They should both be mea­sured at diag­no­sis before
cytes, in con­junc­tion with nor­mal white blood cell and plate­let trans­fu­sion or at least 3 months fol­low­ing trans­fu­sion (Table 3). In
counts (Table 2). 50% of the patients with DBA, var­i­ous malformations are reported
mostly in the cephalic area and the extrem­i­ties, with the clas­si­cal
Clinical and bio­log­i­cal pre­sen­ta­tion of DBA: take-home but rare triphalangeal thumbs (Table 4).1,5
mes­sage Going back to the herein reported cases, the molec­ u­
lar
DBA is usu­ally char­ac­ter­ized by a mod­er­ate to severe, mac­ro­cytic screen­ing of the pro­band (UPN#1447) and her sis­ter (UPN#1821)
aregenerative ane­mia. The other cell lin­e­ages are usu­ally nor­mal, and 2 broth­ers (UPN#1822 and UPN#1213) iden­ti­fied a het­ero­zy­
but on occa­ sion, neutropenia, throm­ bo­cy­ to­
pe­
nia, and, in some gous 4–base pair dele­tion near the donor splice site of exon 2 in
instances, even thrombocytosis may be noted at the time of diag­no­ the RPS19 gene (NM_001022.3: c.71 + 3_71 + 6del; p.?). This var­i­ant
sis. The erythroblastopenia (<5% of ery­throid pre­cur­sors) in an oth­ has indeed been found in the puta­tive TEC case (UPN#1447), and
er­wise nor­mal bone mar­row (no dys­pla­sia and nor­mal cel­lu­lar­ity) on it should have been enough to rule out TEC. This alle­lic var­i­a­tion
the bone mar­row smear or pure hypo­plas­tic ane­mia on bone mar­ was found in the nonanemic (Hb 121 g/L; 78.4 × 109/L retic­u­lo­cytes)
row biopsy can con­firm the diag­no­sis. Bone mar­row exam­i­na­tion is mother, who is con­sid­ered a so-called silent phe­no­type, another
man­da­tory to avoid mis­di­ag­no­sis. Erythroblastopenia is the con­se­ dis­tinct fea­ture of DBA. In addi­tion, the mother exhibited a nor­mal
quence of block­ade in ery­throid dif­fer­en­ti­a­tion between the burst MCV (96 fL) and an ele­vated eADA level (4.21 nmol/min/mg Hb).
forming unit–ery­throid and col­ony forming unit–ery­throid pro­gen­ Silent DBA phe­no­type indi­vid­u­als could be either the ­par­ent or

Table 2. Phenotype of the 4 patients with DBA

Characteristic UPN#1447 UPN#1821 UPN#1822 UPN#1213


Age, mo* 27.6 2 1 1.8
Hb, g/L* 79 20 NA NA
MCV, fL* 91.4 114.8 NA NA
Reticulocyte count, × 10 /L*
9
7.5 60 NA NA
eADA, nmol/min/mg Hb* 5.95 5 4 4.38
HbF, %* 9 18 NA NA
Bone mar­row 1% total Eb 1% total Eb 31 4
Congenital abnor­mal­i­ties None Valgus foot Posterior hypo­spa­dias Hypospadias
Hbat the last fol­low-up, g/L 110 90 NA 108
Treatment at the last fol­ Treatment inde­pen­dence Transfusion Treatment inde­pen­dence Steroid
low-up (deceased)
*At pre­sen­ta­tion.
NA, not avail­­able.
Prakash Singh Shekhawat
Diamond-Blackfan ane­mia, from a clin­i­cal case  |  355
Table 3. Diagnosis cri­te­ria of DBA (from the inter­na­tional sys­tolic heart mur­mur due to ane­mia at 120 g/L (nor­mal value
con­sen­sus con­fer­ence1) >140 g/L), which decreased to 75 g/L at day 18 after birth. He
was trans­fused with 70 mL of red blood cells. He has since been
Diagnosis cri­te­ria Age less than 1 year reg­u­larly trans­fused every 3 to 4 weeks. He did not pres­ent any
Macrocytic ane­mia with no other sig­nif­i­cant con­gen­i­tal abnor­mal­i­ties. After 1 year as recommended, ste­
cytopenias roid ther­apy was started, but there was no response. Chelation
ther­apy with deferasirox was started but had to be interrupted
Reticulocytopenia
due to liver tox­ic­ity. Deferoxamine treat­ment (1000 mg/d) by
Normal mar­row cel­lu­lar­ity with a pau­city of nightly infu­sion over 10 hours, 5 out of 7 days, was started to
ery­throid pre­cur­sors
Supporting
cri­te­ria
Table 4. Malformations iden­ti­fied in DBA from Willig et al5
 Major Gene muta­tion described in “clas­si­cal” DBA

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/353/1851415/353costa.pdf by guest on 13 December 2021


Positive fam­ily his­tory Craniofacial Hypertelorism
 Minor Elevated eADA activ­ity Broad, flat nasal bridge
Congenital anom­a­lies described in “clas­si­cal” DBA Cleft pal­ate
Elevated HbF High arched pal­ate
Noevi­dence of another inherited bone mar­row Microcephaly
fail­ure syn­drome
Micrognathia
Microtia
a sib­ling of a DBA pro­band: they are not ane­mic but may exhibit Low-set ears
a mac­ro­cy­to­sis, an ele­vated eADA, and/or a muta­tion in a ribo­
Low hair­line
somal pro­tein (RP) gene. Patients with the “silent phe­no­type”
have a risk of DBA com­pli­ca­tions such as malig­nan­cies and may Epicanthus
trans­mit the dis­ease as dem­on­strated in this fam­ily: the mother Ptosis
has trans­mit­ted the var­i­ant to all 4 of her off­spring, reinforcing the Ophthalmologic Congenital glau­coma
dom­i­nant inher­i­tance in this fam­ily. DBA has been diag­nosed in 2
Strabismus
mem­bers of the third gen­er­at­ion in this fam­ily. UPN#1822 expe­
ri­enced a fetal loss (UPN#1660) in utero at 23 weeks +3 days of Congenital cat­a­ract
a female nondysmorphic fetus exhibiting a severe intra­uter­ine Neck Short neck
growth retar­da­tion (weight: 436.5 g, which is between the 5th
Webbed neck
and the 10th per­cen­tiles; head cir­cum­fer­ence: 18 cm, 5th per­cen­
tile; height [ver­tex/coc­cyx]: 20 cm, 50th per­cen­tile for 23--week Sprengel defor­mity
amen­or­rhea) with oligohydramnios but with­out hydrops fetalis. Klippel-Feil defor­mity
Interestingly, the pla­centa exhibited defects in the ves­sels with Thumbs Triphalangeal
ste­no­ sis, and some villi were hypovascularized. However, no
Duplex or bifid
erythroblastopenia was noted. The same alle­lic var­i­a­tion has been
iden­ti­fied in this fetus. We assume that the major growth retar­ Hypoplastic
da­tion in rela­tion with DBA may be the ori­gin of the fetal death Flat thenar emi­nence
in utero (Table 5). Hydrops fetalis is a fea­ture of DBA, and its fre­
Absent radial artery
quency is likely underestimated. Since our descrip­tion of the first
case of spo­radic fetal loss due a muta­tion in the RPS19 gene,6 we Urogenital Absent kid­ney
sub­se­quently iden­ti­fied addi­tional fetal cases mostly in asso­ci­a­ Horseshoe kid­ney
tion with the RPS19 gene and also in asso­ci­at­ion with the RPL15 Hypospadias
gene.7 These find­ings rein­force, as we stated ear­lier,8 that major
Cardiac Ventricular sep­tal
com­pli­ca­tions can occur dur­ing preg­nancy and the impor­tance of defect
the fol­low-up of the mother and her off­spring dur­ing preg­nancy
Atrial sep­tal defect
with puta­tive aspi­rin treat­ment in order to pre­vent the pla­centa
vas­cu­lar com­pli­ca­tions as noted in our case.8 Coarctation of the
In addi­tion to UPN#1822, UPN#1821 also had 5 off­spring, aorta
among them twins (UPN#779, UNP#1114, UPN#1264, UPN#1573, Complex car­diac
UNP#1574) (Figure 1). Prenatal diag­no­sis was declined, and out anom­a­lies
of the 5 chil­dren, only 1 boy was affected by DBA and car­ried Other mus­cu­lo­skel­e­tal Growth retar­da­tion
the same famil­ial alle­lic var­i­a­tion in the RPS19 gene. UNP#1264 Syndactyly
was born at a nor­mal ges­ta­tional time of 40 weeks with nor­mal
Neuromotor Learning dif­fi­cul­ties
men­su­ra­tion but a con­junc­ti­val and skin pal­lor, tachy­car­dia, and

356  |  Hematology 2021  |  ASH Education Program


Table 5. Mode of DBA rev­e­la­tion take-home mes­sage

Median age at 2 months; neo­na­tes (16% of the cases); hydrops fetalis


Aregenerative usu­ally mac­ro­cytic ane­mia
Erythroblastopenia in an oth­er­wise normocellular bone mar­row
Malformations in par­tic­u­lar cleft pal­ate, thumbs anom­a­lies, heart and uro­gen­i­tal tract anom­a­lies
Short stat­ure, includ­ing intra­uter­ine growth retar­da­tion
eADA ele­va­tion
Fetal eryth­ro­poi­e­sis fea­tures (ele­vated HbF per­cent­age after 6 months)
Complications of preg­nancy
Malignancies

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/353/1851415/353costa.pdf by guest on 13 December 2021


Very rarely aplastic ane­mia

man­age iron over­load. Stem cell trans­plan­ta­tion was con­tem­ ther­apy are prob­a­bly the most prom­is­ing option,19,20 along with
plated, and the sib­lings were tested for the known famil­ial alle­ other targeted ther­a­pies.21 Recently, some inno­va­tive ther­a­pies
lic var­i­a­tion, which was not found in any of them. Unfortunately, have been pro­posed such as cal­mod­u­lin inhib­i­tors16 and met­
none of them was HLA iden­ti­cal. He is cur­rently 6 years old formin,22 and it is likely that other new ther­a­peu­tic options will
and has been recently under­gone trans­plan­ta­tion with a fully emerge from trans­la­tional research (trial NCT03966053 with tri­
matched unre­lated donor. fluo­per­a­zine).23-26

DBA treat­ment: take-home mes­sage Genetics in DBA: take-home mes­sage


DBA treat­ment in 2021 still relies on either chronic blood trans­fu­ An alle­lic var­i­a­tion, always in a het­ero­zy­gous state in a RP, is found
sions (Hb con­cen­tra­tion to be maintained >90 g/L) with opti­mal in approx­i­ma­tely 70% to 80% of the DBA-affected cases.27-30
iron che­la­tion ther­apy started when the fer­ri­tin level is >500 µg/L Mutations in 20 RP genes have been iden­ti­fied (Figure 2). The
or treat­ment with cor­ti­co­ste­roids.9-11 The cor­ti­co­ste­roids (pred­ most fre­quently mutated gene and the one iden­ti­fied in our
ni­sone or pred­nis­o­lone) are to be intro­duced only after the first clin­i­cal case is the RPS19 gene, which accounts for 25% of DBA
year of life in order to enable the max­i­mal growth because short cases glob­ally.31 Large dele­tions in these RP genes have been
stat­ure is part of the malformative syn­drome. The cor­ti­co­ste­ found in approx­i­ma­tely 20% of the DBA cases, mostly in RPS17,
roids are intro­duced at a dose of 2 mg/kg/d, and their effi­cacy RPL35a, and RPS19 genes.32 Thus, at least 70% of the patients
is usu­ally seen starting at 2 weeks fol­low­ing ini­ti­a­tion of ther­ with DBA carry an alle­lic var­i­a­tion, includ­ing a large dele­tion in
apy, reflected by increased retic­u­lo­cyte count and Hb level. If only 8 RP genes, namely, RPS19, RPL5, RPS26, RPL11, RPL35a,
there is no response to ste­roid ther­apy after 1 month, there is no RPS10, RPS24, and RPS17 (Figure 2). Multiple path­o­genic RP
rea­son to con­tinue this ther­apy. In case of effi­cacy, the cor­ti­co­ muta­tions in a patient with DBA have not been reported to
ste­roid should be grad­u­ally decreased to a min­i­mum dose that date. Phenotypic/geno­typic cor­re­la­tion is not read­ily evi­
can main­tain the Hb level above 90 g/L. The max­i­mal con­tin­u­ dent, although it is well established that patients with DBA
ous cor­ti­co­ste­roid dose must be <0.3 mg/kg/d (<0.5 mg/kg/d car­ry­ing a RPS19 gene muta­tion exhibit less malformation
in the countries where access to trans­fu­sions is dif­fi­cult or dan­ com­pared with oth­ers but appear to exhibit a more severe
ger­ous). In case of corticoresistance or corticodependence (>0.3 hema­ to­logic phe­ no­ type and are fre­ quently man­ aged by a
[or >0.5 mg/kg/d]), trans­fu­sions with iron che­la­tion are cur­rently trans­fu­sion pro­gram.33 Neutropenia is more fre­quently asso­
the only alter­na­tive option of treat­ment,9-11 and hema­to­poi­etic ci­ated with RPL35a,34 car­diac anom­a­lies with RPS24,35 cleft
stem cell trans­plan­ta­tion (HSCT) is indi­cated fol­low­ing the avail­ pal­ate with RPL5, and thumbs anom­a­lies with RPL1136 gene
abil­ity of either an HLA-iden­ti­cal sib­ling in whom DBA or the muta­tions. Other RP genes have been found to be mutated
silent phe­no­type has been excluded12-15 or with a fully matched in small sub­sets of DBA-affected patients, each affect­ing <1%
(10/10) unre­lated donor.14,16 HSCT should be performed ide­ally of DBA cohorts (Figure 2). A muta­tion in a RP gene involved in
before the age of 5 years and cer­tainly <10 years to avoid HSCT DBA is asso­ci­ated with defec­tive ribo­somal RNA (rRNA) mat­
com­ pli­
ca­
tions. HSCT is also indi­ cated in patients with clonal u­ra­tion, which is con­sid­ered the sig­na­ture fea­ture of the DBA
evo­lu­tion.12-15 An alter­na­tive source of hema­to­poi­etic stem cells dis­ease, and its doc­u­men­ta­tion con­firms the path­o­ge­nic­ity of
should be con­sid­ered as exper­i­men­tal approaches but may be alle­lic var­i­a­tion of unknown sig­nif­i­cance. DBA-like dis­ease is
indi­cated for patients with clonal evo­lu­tion. HSCT cures ane­mia respon­si­ble for ane­mia with erythroblastopenia and a cer­tain
and pre­vents the risk of myelodysplastic syn­drome (MDS) and degree of ste­roid response but with the absence of a defect in
leu­ke­mia, but patients with DBA still need to be care­fully mon­i­ rRNA mat­u­ra­tion. These DBA-like dis­eases are related to EPO37
tored for the risk of posttransplant solid tumors.17 and GATA-138,39 gene muta­tions. The DBA and DBA-like dis­eases
Last, leu­cine has been found to be effec­tive in small num­ber con­sti­tute the DBA syn­drome. Despite erythroblastopenia and
of patients with DBA.18 New ther­a­peu­tic options such as gene a cer­tain degree of ste­roid response, ADA2 defi­ciency related

Prakash Singh Shekhawat


Diamond-Blackfan ane­mia, from a clin­i­cal case  |  357
Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/353/1851415/353costa.pdf by guest on 13 December 2021
Figure 2. Genes mutated in DBA and DBA-like cases.

to CECR1 or ADA2 gene muta­tions is not con­sid­ered to belong described. Progress is being made in our under­stand­ing of the
to DBA syn­drome (Figure 2).30,40,41 molec­u­lar basis for DBA, path­o­phys­i­­ol­ogy of the dis­ease, and
The DBA fam­ily we described has been affected by malig­nan­ devel­op­ing and pur­su­ing new ther­a­peu­tic options. We antic­i­
cies. The mother, who rep­re­sents a DBA silent phe­no­type, was pate that these advances will enable bet­ter clin­i­cal man­age­ment
diag­nosed with breast can­cer that was rap­idly fatal. The oldest of the patients with DBA in the com­ing years.
off­spring, UNP#1447, car­ry­ing the famil­ial RPS19 alle­lic var­i­a­tion but
never need­ing any treat­ment, was diag­nosed at age 36 years with Acknowledgments 
a rec­tal ade­no­car­ci­noma and passed away fol­low­ing 16 months Our spe­cial thanks go to the affected indi­vid­ua ­ ls, their fam­i­lies,
of treat­ment. These obser­va­tions point to the fact that iden­ti­fy­ and the DBA French patient asso­ci­a­tion (Association Française
ing patients with DBA (and not misdiagnosing them with TEC) is de la Maladie de Blackfan-Diamond [AFMBD], pres­ i­
dent Mar­
impor­tant and that the so-called silent DBA phe­no­types should cel Hibert). We sin­cerely thank our 2 men­tors, Gil Tchernia and
be con­sid­ered in patients with DBA in their fol­low-up, par­tic­u­larly Mohandas Narla, for their sup­port, friend­ship, and many fruit­
for the risk of malig­nan­cies. A recent study established a risk of ful sci­en­tific dis­cus­sions. We also thank all­ our col­lab­o­ra­tors in
5% for malig­nan­cies in nontransplanted patients with DBA in the France and around the world involved in the muta­tional screen­
National Cancer Institute cohort17 and in the Amer­i­can DBA reg­is­ ing anal­y­sis in France (Cécile Masson, Christine Bole, and the
try42,43 with an observed/expected ratio of 4.8 for any malig­nancy, team from the Genomics Core Facility, Institut Imagine-Structure
44.7 for colon car­ci­noma, 9.4 for lung can­cer, 42.4 for oste­o­genic Fédérative de Recherche Necker, and Anaëlle Jaouen, Ludivine
sar­coma, 352 for MDS, and 28.8 for acute mye­loid leu­ke­mia.42,43 David, Julie Galimand, and Hélène Bourdeau, tech­ni­cians from
However, com­pared with the other inherited bone mar­row fail­ the hema­tol­ogy lab in R. Debré hos­pi­tal, Paris). L.M.D.C. is sup­
ure syn­dromes, the observed/expected ratio is lower for DBA.17 ported by #ANR EJPRD/ANR-19-RAR4-0016 (Euro­ pean Union’s
To date, there are no guide­lines for MDS/acute mye­loid leu­ke­mia Horizon 2020 research and inno­va­tion pro­gram under the EJP
and solid tumor screen­ing, except recent pre­lim­i­nary rec­om­men­ RD COFUND-EJP No 825575), the Laboratory of Excellence for
da­tions on colo­rec­tal car­ci­noma,44 but this may be warranted and Red Cells ((LABEX GR-Ex)-ANR Avenir-11-LABX-0005-02), and the
is cur­rently being discussed among DBA coop­er­a­tive groups. French National PHRC OFABD (DBA reg­is­try). T.M.L. and L.M.D.C.
In con­clu­sion, DBA is a fas­ci­nat­ing and com­plex ery­throid are supported by the ANR grant EJPRD DBAGenCure (coor­di­na­
dis­or­der, as illus­
trated from the study of the DBA fam­ ily we tors Juan Ruben and Susana Navarro Ordonez).

358  |  Hematology 2021  |  ASH Education Program


Conflict-of-inter­est dis­clo­sure from the Diamond Blackfan Anemia Registry. Bone Marrow Transplant.
2001;27(4):381-386.
Lydie M. Da Costa: no com­pet­ing finan­cial inter­ests to declare.
16. Taylor AM, Macari ER, Chan IT, et al. Calmodulin inhib­i­tors improve eryth­ro­
Isabelle Marie: no com­pet­ing finan­cial inter­ests to declare. poi­e­sis in Diamond-Blackfan ane­mia. Sci Transl Med. 2020;12(566):eabb5831.
Thierry M. Leblanc: no com­pet­ing finan­cial inter­ests to declare. 17. Alter BP, Giri N, Savage SA, Rosenberg PS. Cancer in the National Cancer
Institute inherited bone mar­row fail­ure syn­drome cohort after fif­teen years
of fol­low-up. Haematologica. 2018;103(1):30-39.
Off-label drug use 18. Vlachos A, Atsidaftos E, Lababidi ML, et al. L-leu­cine improves ane­mia and
Lydie M. Da Costa: nothing to disclose. growth in patients with trans­fu­sion-depen­dent Diamond-Blackfan ane­mia:
Isabelle Marie: nothing to disclose. results from a mul­ti­cen­ter pilot phase I/II study from the Diamond-Blackfan
Thierry M. Leblanc: nothing to disclose. Anemia Registry. Pediatr Blood Cancer. 2020;67(12):e28748.
19. Jaako P, Debnath S, Olsson K, et al. Gene ther­apy cures the ane­mia and
lethal bone mar­row fail­ure in a mouse model of RPS19-defi­cient Diamond-​
Correspondence Blackfan ane­mia. Haematologica. 2014;99(12):1792-1798.
Lydie M. Da Costa, Service d’Hématologie Biologique (Hematol­ 20. Liu Y, Dahl M, Debnath S, et al. Successful gene ther­apy of Diamond-Black­
ogy Diagnostic Lab), AP-HP, Hôpital Robert Debré, 48 bou­le­vard fan ane­mia in a mouse model and human CD34+ cord blood hema­to­poi­etic

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/353/1851415/353costa.pdf by guest on 13 December 2021


Serurier, Paris 75019, France; e-mail: lydie​­.dacosta@rdb​­.aphp​­.fr. stem cells using a clin­i­cally appli­ca­ble lentiviral vec­tor [published online 14
Jan­u­ary 2021]. Haematologica.
21. Siva K, Ek F, Chen J, et al. A phe­no­typic screen­ing assay identifies mod­u­la­
References tors of Diamond Blackfan ane­mia. SLAS Discov. 2019;24(3):304-313.
1. Vlachos A, Ball S, Dahl N, et al; Participants of Sixth Annual Daniella Maria 22. Wilkes MC, Siva K, Varetti G, et  al. Metformin-induced sup­ pres­
sion of
Arturi International Consensus Conference. Diagnosing and treating Dia­ Nemo-like kinase improves eryth­ro­poi­e­sis in pre­clin­i­cal mod­els of Diamond-
mond Blackfan anae­ mia: results of an inter­ na­tional clin­

cal con­sen­sus Blackfan ane­mia through induc­tion of miR-26a. Exp Hematol. 2020;91:65-77.
con­fer­ence. Br J Haematol. 2008;142(6):859-876. 23. Kristiansson A, Gram M, Flygare J, Hansson SR, Åkerström B, Storry JR. The
2. Willig TN, Pérignon JL, Gustavsson P, et  al. High aden­o­sine deam­i­nase role of α1-microglobulin (A1M) in eryth­ro­poi­e­sis and eryth­ro­cyte homeo­
level among healthy pro­bands of Diamond Blackfan ane­mia (DBA) coseg­ sta­sis-ther­a­peu­tic oppor­tu­ni­ties in hemo­lytic con­di­tions. Int J Mol Sci.
regates with the DBA gene region on chro­ mo­ some 19q13. The DBA 2020;21(19):7234.
Working Group of Société d’Immunologie Pédiatrique (SHIP). Blood. 24. Sjögren SE, Flygare J. Progress towards mech­a­nism-based treat­ment for
1998;92(11):4422-4427. Diamond-Blackfan ane­mia. ScientificWorldJournal. 2012;2012:184362.
3. Gustavsson P, Klar J, Matsson H, et al. Familial tran­sient erythroblastopenia 25. Wilkes MC, Siva K, Chen J, et al. Diamond Blackfan ane­mia is medi­ated by
of child­hood is asso­ci­ated with the chro­mo­some 19q13.2 region but not hyper­ac­tive Nemo-like kinase. Nat Commun. 2020;11(1):3344.
caused by muta­tions in cod­ing sequences of the ribo­somal pro­tein S19 26. Doulatov S, Vo LT, Macari ER, et al. Drug dis­cov­ery for Diamond-Blackfan
(RPS19) gene. Br J Haematol. 2002;119(1):261-264. ane­mia using reprogrammed hema­to­poi­etic pro­gen­i­tors. Sci Transl Med.
4. Ohene-Abuakwa Y, Orfali KA, Marius C, Ball SE. Two-phase cul­ture in Dia­ 2017;9(376):eaah5645.
mond Blackfan ane­mia: local­i­za­tion of ery­throid defect. Blood. 2005;105 27. Da Costa L, Leblanc T, Mohandas N. Diamond-Blackfan ane­mia. Blood.
(2):838-846. 2020;136(11):1262-1273.
5. Willig T-N, Niemeyer CM, Leblanc T, et al. Identification of new prog­no­sis 28. Da Costa L, Narla A, Mohandas N. An update on the path­o­gen­e­sis and diag­
fac­tors from the clin­i­cal and epi­de­mi­o­logic anal­y­sis of a reg­is­try of 229 no­sis of Diamond-Blackfan ane­mia. F1000Res. 2018;7:F1000 Faculty Rev-
Diamond-Blackfan ane­mia patients. Pediatr Res. 1999;46(5):553. 1350.
6. Da Costa L, Chanoz-Poulard G, Simansour M, et al. First de novo muta­tion 29. Da Costa L, O’Donohue M-F, van Dooijeweert B, et al. Molecular approaches
in RPS19 gene as the cause of hydrops fetalis in Diamond-Blackfan ane­mia. to diag­nose Diamond-Blackfan ane­mia: the EuroDBA expe­ri­ence. Eur J Med
Am J Hematol. 2013;88(4):340-341. Genet. 2018;61(11):664-673.
7. Wlodarski MW, Da Costa L, O’Donohue M-F, et  al. Recurring muta­tions 30. Ulirsch JC, Verboon JM, Kazerounian S, et al. The genetic land­scape of Dia­
in RPL15 are linked to hydrops fetalis and treat­ment inde­pen­dence in mond-Blackfan ane­mia. Am J Hum Genet. 2018;103(6):930-947.
­Diamond-Blackfan ane­mia. Haematologica. 2018;103(6):949-958. 31. Draptchinskaia N, Gustavsson P, Andersson B, et  al. The gene encoding
8. Faivre L, Meerpohl J, Da Costa L, et  al. High-risk preg­ nan­cies in Dia­ ribo­somal pro­tein S19 is mutated in Diamond-Blackfan anae­mia. Nat Genet.
mond-Blackfan ane­mia: a sur­vey of 64 preg­nan­cies from the French and 1999;21(2):169-175.
Ger­man reg­is­tries. Haematologica. 2006;91(4):530-533. 32. Quarello P, Garelli E, Brusco A, et al. High fre­quency of ribo­somal pro­tein
9. Berdoukas V, Nord A, Carson S, et al. Tissue iron eval­u­a­tion in chron­i­cally gene dele­tions in Ital­ian Diamond-Blackfan ane­mia patients detected by
trans­fused chil­dren shows sig­nif­i­cant lev­els of iron load­ing at a very young mul­ti­plex liga­tion-depen­dent probe ampli­fi­ca­tion assay. Haematologica.
age. Am J Hematol. 2013;88(11):E283-E285. 2012;97(12):1813-1817.
10. Roggero S, Quarello P, Vinciguerra T, Longo F, Piga A, Ramenghi U. Severe 33. Arbiv OA, Cuvelier G, Klaassen RJ, et al. Molecular anal­y­sis and geno­type-
iron over­ load in Blackfan-Diamond ane­ mia: a case-con­ trol study. Am J phe­no­type cor­re­la­tion of Diamond-Blackfan ane­mia. Clin Genet. 2018;​
Hematol. 2009;84(11):729-732. 93(2):​320-328.
11. Vlachos A, Muir E. How I treat Diamond-Blackfan ane­mia. Blood. 2010;116 34. Gianferante MD, Wlodarski MW, Atsidaftos E, et al. Genotype-phe­no­type
(19):3715-3723. asso­ci­a­tion and var­i­ant char­ac­ter­iza­tion in Diamond-Blackfan ane­mia
12. Strahm B, Loewecke F, Niemeyer CM, et al. Favorable out­comes of hema­ caused by path­o­genic var­i­ants in RPL35A. Haematologica. 2021;106(5):​
to­poi­etic stem cell trans­plan­ta­tion in chil­dren and ado­les­cents with 1303-1310.
­Diamond-Blackfan ane­mia. Blood Adv. 2020;4(8):1760-1769. 35. Vlachos A, Osorio DS, Atsidaftos E, et  al. Increased prev­a­lence of con­
13. Fagioli F, Quarello P, Zecca M, et al. Haematopoietic stem cell trans­plan­ gen­i­tal heart dis­ease in chil­dren with Diamond Blackfan ane­mia sug­gests
ta­tion for Diamond Blackfan anae­mia: a report from the Ital­ian Associa­ unrec­og­nized Diamond Blackfan ane­mia as a cause of con­gen­i­tal heart dis­
tion of Paediatric Haematology and Oncology Registry. Br J Haematol. ease in the gen­eral pop­u­la­tion: a report of the Diamond Blackfan ane­mia
2014;165(5):673-681. reg­is­try. Circ Genom Precis Med. 2018;11(5):e002044.
14. Peffault de Latour R, Peters C, Gib­son B, et al; Pediatric Working Party of 36. Gazda HT, Sheen MR, Vlachos A, et  al. Ribosomal pro­ tein L5 and L11
the Euro­pean Group for Blood and Marrow Transplantation; Severe Aplas­ muta­tions are asso­ci­ated with cleft pal­ate and abnor­mal thumbs in
tic Anemia Working Party of the Euro­pean Group for Blood and Marrow ­Diamond-Blackfan ane­mia patients. Am J Hum Genet. 2008;83(6):769-780.
Transplantation. Recommendations on hema­to­poi­etic stem cell trans­plan­ 37. Kim AR, Ulirsch JC, Wilmes S, et al. Functional selec­tiv­ity in cyto­kine sig­nal­
ta­tion for inherited bone mar­row fail­ure syn­dromes. Bone Marrow Trans- ing revealed through a path­o­genic EPO muta­tion. Cell. 2017;168(6):1053-
plant. 2015;50(9):1168-1172. 1064.e151064e15.
15. Vlachos A, Federman N, Reyes-Haley C, Abramson J, Lipton JM. Hemato­ 38. Ludwig LS, Gazda HT, Eng JC, et  al. Altered trans­la­tion of GATA1 in Dia­
poietic stem cell trans­plan­ta­tion for Diamond Blackfan ane­mia: a report mond-Blackfan ane­mia. Nat Med. 2014;20(7):748-753.

Prakash Singh Shekhawat


Diamond-Blackfan ane­mia, from a clin­i­cal case  |  359
39. Sankaran VG, Ghazvinian R, Do R, et al. Exome sequenc­ing identifies GATA1 43. Vlachos A, Rosenberg PS, Atsidaftos E, et al. Increased risk of colon can­
muta­tions resulting in Diamond-Blackfan ane­mia. J Clin Invest. 2012;122 cer and oste­o­genic sar­coma in Diamond-Blackfan ane­mia. Blood. 2018;132
­(7):2439-2443. ­(20):2205-2208.
40. Lee PY. Vasculopathy, immu­no­de­fi­ciency, and bone mar­row fail­ure: the 44. Lipton JM, Molmenti CLS, Hussain M, et al. Colorectal can­cer screen­ing and
intrigu­ing syn­drome caused by defi­ ciency of aden­o­sine deam­ i­
nase 2. sur­veil­lance strat­egy for patients with Diamond Blackfan ane­mia: pre­lim­i­
Front Pediatr. 2018;6:282. nary rec­om­men­da­tions from the Diamond Blackfan Anemia Registry. Pedi-
41. Ulirsch JC, Verboon JM, Kazerounian S, et al. The genetic land­scape of atr Blood Cancer. 2021;68(8):e28984.
­Diamond-Blackfan ane­mia. Am J Hum Genet. 2019;104(2):356.
42. Lipton JM, Federman N, Khabbaze Y, et al; Diamond-Black Anemia Registry.
Osteogenic sar­coma asso­ci­ated with Diamond-Blackfan ane­mia: a report
from the Diamond-Blackfan Anemia Registry. J Pediatr Hematol Oncol. © 2021 by The Amer­i­can Society of Hematology
2001;23(1):39-44. DOI 10.1182/hema­tol­ogy.2021000314

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/353/1851415/353costa.pdf by guest on 13 December 2021

360  |  Hematology 2021  |  ASH Education Program


Contents

ix Editors’ Message
x Reviewers
xi Continuing Medical Education Information

ALL: New Directions for Adult Patients


1 Relapsed ALL: CAR T vs transplant vs novel therapies
NOELLE V. FREY

7 Acute lymphoblastic leukemia in older adults: curtain call for conventional chemotherapy?
MARLISE R. LUSKIN

AML: So Many Options, So Little Time


16 What to use to treat AML: the role of emerging therapies
FELICITAS THOL

24 Whom should we treat with novel agents? Specifc indications for specifc and challenging
populations
LINDSAY WILDE AND MARGARET KASNER

Challenges in Multiple Myeloma Treatment


30 High-risk multiple myeloma: how to treat at diagnosis and relapse?
MARÍA-VICTORIA MATEOS, BORJA PUERTAS MARTÍNEZ, AND VERÓNICA GONZÁLEZ-CALLE

37 Minimal residual disease in multiple myeloma—why, when, where


ANDREW J. YEE AND NOOPUR RAJE

46 Treatment of older adult or frail patients with multiple myeloma


SHAKIRA J. GRANT, CIARA L. FREEMAN, AND ASHLEY E. ROSKO

CLL: Extending Survival


55 Upfront therapy: the case for continuous treatment
CONSTANTINE S. TAM

59 Frontline treatment in CLL: the case for time-limited treatment


VINCENT LÉVY, ALAIN DELMER, AND FLORENCE CYMBALISTA

68 Is there a role for anti-CD20 antibodies in CLL?


HARSH R. SHAH AND DEBORAH M. STEPHENS

Clotting and Bleeding Conundrums


76 Unexplained arterial thrombosis: approach to diagnosis and treatment
JORI E. MAY AND STEPHAN MOLL

85 Cryptogenic oozers and bruisers


KRISTI J. SMOCK AND KAREN A. MOSER

Prakash Singh Shekhawat


Contents | iii
92 Clots in unusual places: lots of stress, lim­ited data, crit­i­cal deci­sions
CAROL MATHEW AND MARC ZUMBERG

100 EVIDENCE-BASED MINIREVIEW


Should war­fa­rin or a direct oral anti­co­ag­u­lant be used in patients presenting with throm­bo­sis in the
splanch­nic or cere­bral veins?
CAROL MATHEW AND MARC ZUMBERG

CML: Success Breeds More Success


106 TKI dis­con­tin­u­at­ ion in CML: how do we make more patients eli­gi­ble? How do we increase the
chances of a suc­cess­ful treat­ment-free remis­sion?
ANDREAS HOCHHAUS AND THOMAS ERNST

113 Lifelong TKI ther­apy: how to man­age car­dio­vas­cu­lar and other risks
MICHAEL J. MAURO

122 Blast and accel­er­ated phase CML: room for improve­ment


JOAN HOW, VINAYAK VENKATARAMAN, AND GABRIELA SORIANO HOBBS

Coagulation Laboratory Potpourri


129 Direct oral anti­co­ag­u­lant (DOAC) inter­fer­ence in hemo­sta­sis assays
KAREN A. MOSER AND KRISTI J. SMOCK

What Do Aplastic Anemia, PNH, and “Hypoplastic” Leukemia Have in Common?


134 The clin­i­cal and lab­o­ra­tory eval­u­a­tion of patients with suspected hypocellular mar­row fail­ure
SIOBÁN KEEL AND AMY GEDDIS

143 When does a PNH clone have clin­i­cal sig­nif­i­cance?


DARIA V. BABUSHOK

153 When to worry about inherited bone mar­row fail­ure and mye­loid malig­nancy pre­dis­po­si­tion
syn­dromes in the set­ting of a hypocellular mar­row
ANUPAMA NARLA

Defeating Diffuse, Double-Hit, and Dogged Non-Hodgkin Lymphoma


157 Double-hit lym­phoma: opti­miz­ing ther­apy
KIERON DUNLEAVY

164 Relapsed dis­ease: off-the-shelf immunotherapies vs cus­tom­ized engineered prod­ucts


REEM KARMALI

Emerging Therapies and Considerations for Sickle Cell Disease


174 Gene ther­apy for sickle cell dis­ease: where we are now?
JULIE KANTER AND COREY FALCON

181 Hematopoietic cell trans­plan­ta­tion for sickle cell dis­ease: updates and future direc­tions
LAKSHMANAN KRISHNAMURTI

190 EVIDENCE-BASED MINIREVIEW


In young chil­dren with severe sickle cell dis­ease, do the ben­e­fits of HLA-iden­ti­cal sib­ling donor HCT
out­weigh the risks?
NIKETA SHAH AND LAKSHMANAN KRISHNAMURTI

196 Clinical trial con­sid­er­ations in sickle cell dis­ease: patient-reported out­comes, data ele­ments,
and the stake­holder engage­ment frame­work
SHERIF M. BADAWY

iv  |  Hematology 2021  |  ASH Education Program


Hemophilia Update: Our Cup Runneth Over
206 How do we opti­mally uti­lize fac­tor con­cen­trates in per­sons with hemo­philia?
MING Y. LIM

215 EVIDENCE-BASED MINIREVIEW


For over­weight or obese per­sons with hemo­philia A, should fac­tor VIII dos­ing be based on ideal or
actual body weight?
NICOLETTA MACHIN AND MING Y. LIM

219 Factor-mimetic and rebalancing ther­a­pies in hemo­philia A and B: the end of fac­tor con­cen­trates?
PATRICK ELLSWORTH AND ALICE MA

226 Hemophilia gene ther­apy: ush­er­ing in a new treat­ment par­a­digm?


LINDSEY A. GEORGE

Hodgkin Lymphoma: Cure and Optimal Survivorship


234 Controversies in the man­age­ment of early-stage Hodgkin lym­phoma
KRISTIE A. BLUM

240 How to choose first sal­vage ther­apy in Hodgkin lym­phoma: tra­di­tional che­mo­ther­apy vs novel agents
JULIA DRIESSEN, SANNE H. TONINO, ALISON J. MOSKOWITZ, AND MARIE JOSÉ KERSTEN

247 Double-refrac­tory Hodgkin lym­phoma: tack­ling relapse after brentuximab vedotin and check­point
inhib­i­tors
NARENDRANATH EPPERLA AND MEHDI HAMADANI

How Can We Ensure That Everyone Who Needs a Transplant Can Get One?
254 Allogeneic hema­to­poi­etic cell trans­plan­ta­tion for older patients
RICHARD J. LIN AND ANDREW S. ARTZ

264 Increasing access to allo­ge­neic hema­to­poi­etic cell trans­plant: an inter­na­tional per­spec­tive


VANDERSON ROCHA, GIANCARLO FATOBENE, AND DIETGER NIEDERWIESER, FOR THE BRAZILIAN SOCIETY OF BONE
MARROW TRANSPLANTATION AND THE WORLDWIDE NETWORK FOR BLOOD AND MARROW TRANSPLANTATION

275 Increasing access to allotransplants in the United States: the impact of race, geog­ra­phy, and
socio­eco­nom­ics
SANGHEE HONG AND NAVNEET S. MAJHAIL

Immunology 101: What the Practicing Hematologist Needs to Know


281 Immunology 101: fun­da­men­tal immu­nol­ogy for the prac­tic­ing hema­tol­o­gist
SHANNON A. CARTY

287 How immu­no­de­fi­ciency can lead to malig­nancy


SUNG-YUN PAI, KATHRYN LURAIN, AND ROBERT YARCHOAN

296 Modified T cells as ther­a­peu­tic agents


NATHAN SINGH

Indolent Lymphomas
303 What fac­tors guide treat­ment selec­tion in myco­sis fungoides and Sezary syn­drome?
YOUN H. KIM

313 Follicular lym­phoma: is there an opti­mal way to define risk?


CARLA CASULO

320 Does MRD have a role in the man­age­ment of iNHL?


ILARIA DEL GIUDICE, IRENE DELLA STARZA, AND ROBIN FOÀ

Inherited Red Cell Disorders Beyond Hemoglobinopathies


331 Diagnosis and clin­i­cal man­age­ment of red cell mem­brane dis­or­ders
THEODOSIA A. KALFA

Prakash Singh Shekhawat


Contents | v
341 Diagnosis and clin­i­cal man­age­ment of enzymopathies
LUCIO LUZZATTO

353 Diamond-Blackfan ane­mia


LYDIE M. DA COSTA, ISABELLE MARIE, AND THIERRY M. LEBLANC

It Takes a Village: Maximizing Supportive Care and Minimizing Toxicity During Childhood Leukemia
Therapy
361 Fungal diag­nos­tic test­ing and ther­apy: nav­i­gat­ing the neutropenic period in chil­dren with high-
risk leu­ke­mia
BRIAN T. FISHER

368 Minimizing car­diac tox­ic­ity in chil­dren with acute mye­loid leu­ke­mia


HARI K. NARAYAN, KELLY D. GETZ, AND KASEY J. LEGER

376 Managing ther­apy-asso­ci­ated neu­ro­tox­ic­ity in chil­dren with ALL


DEEPA BHOJWANI, RAVI BANSAL, AND ALAN S. WAYNE

Let the CHIP(s) Fall Where They May? Burdens and Benefits of Diagnosing Clonal Hematopoiesis
384 CHIP: is clonal hema­to­poi­e­sis a sur­ro­gate for aging and other dis­ease?
LUKASZ P. GONDEK

390 Mechanisms of somatic trans­for­ma­tion in inherited bone mar­row fail­ure syn­dromes


HARUNA BATZORIG CHOIJILSUREN, YEJI PARK, AND MOONJUNG JUNG

399 When are idi­o­pathic and clonal cytopenias of unknown sig­nif­i­cance (ICUS or CCUS)?
AFAF E. W. G. OSMAN

Management Strategies for Sickle Cell Disease


405 Optimizing man­age­ment of sickle cell dis­ease in patients under­go­ing sur­gery
CHARITY I. OYEDEJI AND IAN J. WELSBY

411 Treatment dilem­mas: strat­eg ­ ies for priapism, chronic leg ulcer dis­ease, and pul­mo­nary
hyper­ten­sion in sickle cell dis­ease
ROBERTA C.G. AZBELL AND PAYAL CHANDARANA DESAI

MDS: Beyond a One-Size-Fits-All Approach


418 Have we reached a molec­ul­ar era in myelodysplastic syn­dromes?
MARIA TERESA VOSO AND CARMELO GURNARI

428 Lower risk but high risk


AMY E. DeZERN

435 EVIDENCE-BASED MINIREVIEW


Molecular pre­ci­sion and clin­i­cal uncer­tainty: should molec­u­lar pro­fil­ing be rou­tinely used to guide
risk strat­i­fi­ca­tion in MDS?
DAN­IEL R. RICHARDSON AND AMY E. DeZERN

439 Oral hypomethylating agents: beyond con­ve­nience in MDS


ELIZABETH A. GRIFFITHS

MPN: New Directions


710 MPN and throm­bo­sis was hard enough . . . ​now there’s COVID-19 throm­bo­sis too
ANNA FALANGA

448 EVIDENCE-BASED MINIREVIEW


Are DOACs an alter­na­tive to vita­min K antag­on
­ ists for treat­ment of venous throm­bo­em­bo­lism in
patients with MPN?
FRANCESCA SCHIEPPATI AND ANNA FALANGA

453 Novel ther­a­pies vs hema­to­poi­etic cell trans­plan­ta­tion in mye­lo­fi­bro­sis: who, when, how?
JAMES ENGLAND AND VIKAS GUPTA

463 Running inter­feron inter­fer­ence in treating PV/ET: meet­ing unmet needs


ELIZABETH A. TRAXLER AND ELIZABETH O. HEXNER

vi  |  Hematology 2021  |  ASH Education Program


Multidisciplinary Hematologic Disorders: What Is the Hematologist’s Role?
469 Hereditary hem­or­rhagic tel­an­gi­ec­ta­sia (HHT): a prac­ti­cal guide to man­age­ment
ADRIENNE M. HAMMILL, KATIE WUSIK, AND RAJ S. KASTHURI

478 Chronic throm­bo­em­bolic pul­mo­nary hyper­ten­sion: anticoagulation and beyond


KARLYN A. MARTIN AND MICHAEL J. CUTTICA

485 Rebalanced hemo­sta­sis in liver dis­ease: a mis­un­der­stood coagulopathy


LARA N. ROBERTS

Neutropenia: Doing More with Less


492 Diagnosis and ther­a­peu­tic deci­sion-mak­ing for the neutropenic patient
JAMES A. CONNELLY AND KELLY WALKOVICH

504 Understanding neutropenia sec­ond­ary to intrin­sic or iat­ro­genic immune dysregulation


KELLY WALKOVICH AND JAMES A. CONNELLY

514 Impaired myelopoiesis in con­gen­i­tal neutropenia: insights into clonal and malig­nant hema­to­poi­e­sis
JULIA T. WARREN AND DAN­IEL C. LINK

Perioperative Consultative Hematology: Can You Clear My Patient for Surgery?


521 Perioperative con­sul­ta­tive hema­tol­ogy: can you clear my patient for a pro­ce­dure?
ALLISON ELAINE BURNETT, BISHOY RAGHEB, AND SCOTT KAATZ

529 How to man­age bleed­ing dis­or­ders in aging patients need­ing sur­gery


MOUHAMED YAZAN ABOU-ISMAIL AND NATHAN T. CONNELL

536 Heparin-induced throm­bo­cy­to­pe­nia and car­dio­vas­cu­lar sur­gery


ALLYSON M. PISHKO AND ADAM CUKER

Pregnancy in Special Populations: Challenges and Solutions


545 How to eval­u­ate and treat the spec­trum of TMA syn­dromes in preg­nancy
MARIE SCULLY

552 Pregnancy in spe­cial pop­u­la­tions: chal­lenges and solu­tions prac­ti­cal aspects of man­ag­ing von
Willebrand dis­ease in preg­nancy
OZLEM TURAN AND REZAN ABDUL KADIR

559 Prevention and man­age­ment of venous throm­bo­em­bo­lism in preg­nancy: cut­ting through the
prac­tice var­i­a­tion
LESLIE SKEITH

Survivorship After Allogeneic Hematopoietic Cell Transplant


570 Psychosocial and finan­cial issues after hema­to­poi­etic cell trans­plan­ta­tion
DAVID BUCHBINDER AND NANDITA KHERA

578 Noninfectious com­pli­ca­tions of hema­to­poi­etic cell trans­plan­ta­tion


KIRSTEN M. WILLIAMS

587 Infectious com­pli­ca­tions and vac­cines


PER LJUNGMAN

The Changing Landscape of α- and β-Thalassemia Diagnosis and Treatment


592 Advances in the man­age­ment of α-thal­as­se­mia major: rea­sons to be opti­mis­tic
PAULINA HORVEI, TIPPI MACKENZIE, AND SANDHYA KHARBANDA

600 β-Thalassemia: evolv­ing treat­ment options beyond trans­fu­sion and iron che­la­tion
ARIELLE L. LANGER AND ERICA B. ESRICK

607 Optimal strat­e­gies for car­rier screen­ing and pre­na­tal diag­no­sis of α- and β-thal­as­se­mia
CHERYL MENSAH AND SUJIT SHETH
Prakash Singh Shekhawat
Contents | vii
The COVID Crash: Lessons Learned from a World on Pause
614 How to rec­og­nize and man­age COVID-19-asso­ci­ated coagulopathy
GLO­RIA F. GERBER AND SHRUTI CHATURVEDI

621 COVID-19 and throm­bo­sis: searching for evi­dence


BRIGHT THILAGAR, MOHAMMAD BEIDOUN, RUBEN RHOADES, AND SCOTT KAATZ

628 Passive immune ther­a­pies: another tool against COVID-19


LISE J. ESTCOURT

Update in Graft-versus-Host Disease


642 Acute GVHD: think before you treat
LAURA F. NEWELL AND SHERNAN G. HOLTAN

648 Updates in chronic graft-ver­sus-host dis­ease


BETTY K. HAMILTON

655 What else do I need to worry about when treating graft-ver­sus-host dis­ease?
AREEJ EL-JAWAHRI

What’s New in Plasma Cell Disorders?


662 Advances in MGUS diag­no­sis, risk strat­i­fi­ca­tion, and man­age­ment: intro­duc­ing myeloma-defining
genomic events
OLA LANDGREN

673 Smoldering mul­ti­ple mye­loma: evolv­ing diag­nos­tic cri­te­ria and treat­ment strat­e­gies
ALISSA VISRAM, JOSELLE COOK, AND RAHMA WARSAME

682 AL amy­loid­osis: untangling new ther­a­pies


SUSAN BAL AND HEATHER LANDAU

What’s New in the Transfusion Management of Sickle Cell Disease?


689 How to avoid the prob­lem of eryth­ro­cyte alloimmunization in sickle cell dis­ease
FRANCE PIRENNE, A­LINE FLOCH, AND ANOOSHA HABIBI

696 Indications for transfusion in the management of sickle cell disease


HYOJEONG HAN, LISA HENSCH, AND VENÉE N. TUBMAN

704 Management of hemo­lytic trans­fu­sion reac­tions


JEANNE E. HENDRICKSON AND ROSS M. FASANO

viii  |  Hematology 2021  |  ASH Education Program


IT TAKES A VILLAGE: MAXIMIZING SUPPORTIVE CARE AND MINIMIZING TOXICITY DURING CHILDHOOD LEUKEMIA THERAPY

Fungal diagnostic testing and therapy:


navigating the neutropenic period
in children with high-risk leukemia

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/361/1851411/361fisher.pdf by guest on 13 December 2021


Brian T. Fisher
Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA; and Department of Biostatistics, Epidemiology and Informatics,
Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA

Children, adolescents, and young adults receiving intensive chemotherapy for acute myeloid leukemia or high-risk or
relapsed acute lymphoblastic leukemia sustain prolonged periods of neutropenia that predispose them to invasive fun-
gal disease (IFD). For many decades the standard of care for these patients was to initiate empirical antifungal therapy
after a period of prolonged fever and neutropenia. Recent publications have yielded important evidence on the utility of
different diagnostic and therapeutic approaches aimed at reducing the impact of IFD among these patients during these
vulnerable periods. This case-based review highlights and interprets the published data to provide context for the IFD
diagnostic and therapeutic recommendations proposed in multiple published guidelines. Personalized approaches are
offered at points where evidence is lacking. Time points where specific knowledge gaps exist are identified along the
clinical trajectory of the prolonged neutropenic period to illustrate areas for future investigation.

LEARNING OBJECTIVES
• Learn the evidence supporting prophylaxis, preemptive, and empirical antifungal therapy during periods of pro-
longed neutropenia secondary to intensive chemotherapy for leukemia
• Understand the utility and limitations of existing fungal biomarkers at specifc clinical time points during pro-
longed periods of neutropenia

Introduction
CLINICAL CASE Invasive fungal diseases (IFDs) have long been identifed as
An 11-year-old girl with recently diagnosed acute myeloid important opportunistic infections in children, adolescents,
leukemia (AML) is admitted to the hospital for induction and young adults receiving chemotherapy for AML and for
2 chemotherapy. She receives cyarabine, daunorubicin, high-risk or relapsed acute lymphoblastic leukemia (ALL).
etoposide, and gemtuzumab ozogamicin in induction 1 and The epidemiology of IFDs in these patient populations is
is scheduled to receive cytarabine, daunorubicin, and etopo- displayed in Table 1. The interpretation of IFD incidence
side for induction 2. What prophylactic antifungal regimen across each study needs to consider the IFD defnition used,
and fungal surveillance testing should be employed once she whether antifungal prophylaxis was administered, and the
develops neutropenia? Approximately 2 weeks into her neu- study design. Early reports from pediatric AML chemother-
tropenic period (absolute neutrophil count <200 cells/µL), apy clinical trials estimated an IFD incidence of 14% to 23%
she develops fevers without any localizing signs or symp- per cycle of chemotherapy.1 More recent investigations
toms. Blood cultures are drawn, and cefepime is initiated for have found a lower but still substantial incidence of IFD,
empirical antibiotic therapy. The blood cultures are negative ranging from 3% to 7%.2,3 The decline in IFD rates among
and she remains stable, but fevers persist for more than 96 patients with AML is likely multifactorial, including the use of
hours. What adjustments should be made to her antifungal published research criteria for defning probable or proven
regimen, if any, and what additional diagnostic testing, if any, IFDs that are more restrictive,3 the decreased use of ste-
should be performed? roids in this population, and the optimization of supportive
care measures. The rates of IFD in ALL are less well defned,
Prakash Singh Shekhawat
Fungal management in leukemia | 361
Table 1. Epidemiology of IFDs from select pedi­at­ric leu­ke­mia cohorts

Author, year Study design Patient type Antifungal pro­phy­laxis IFD def­i­ni­tion IFD inci­dence
AML
Sung et al1 Summary of adverse Newly diag­nosed AML Not rou­tinely used Not spec­i­fied 18% to 23% inci­dence per
event data from che­mo­ther­apy course
che­mo­ther­apy RCT
Fisher et al2 RCT of 2 anti­fun­gal De novo, sec­ond­ary, Fluconazole or 2008 EORTC/MSG cri­te­ria33 3.1% in caspofungin
pro­phy­laxis reg­i­mens or relapsed AML caspofungin sub­jects*;
7.2% in fluconazole sub­jects*
ALL
Rosen et al4 Retrospective sin­gle- Any type of newly Not rou­tinely used Not spec­i­fied 10%†
cen­ter obser­va­tional diag­nosed ALL
cohort

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/361/1851411/361fisher.pdf by guest on 13 December 2021


Wang et al7 Retrospective mul­ti­cen­ter Reported by ALL type Varied by health care 2008 EORTC/MSG cri­te­ria33 6.2% over­all†;
obser­va­tional cohort and risk strata cen­ter 5.8% SR ALL†;
8.7% HR ALL†;
3.8% nonrelapsed ALL†;
12.9% relapsed refrac­tory ALL†
Castagnola Retrospective sin­gle- Any type of newly Not rou­tinely used 2008 EORTC/MSG cri­te­ria33 5.4% over­all†;
et al6 cen­ter obser­va­tional diag­nosed ALL 15.2% HR ALL†
cohort
*5-month cumu­la­tive inci­dence.
†Cumulative inci­dence for dura­tion of che­mo­ther­apy.
CTCAE, com­mon ter­mi­nol­ogy cri­te­ria for adverse events; EORTC/MSG, Euro­pean Organization for Research and Treatment of Cancer/Mycoses Study
Group; IFD, invasive fungal disease; RCT, ran­dom­ized con­trolled trial.

but avail­­able data sug­gest the sub­stan­tial pres­ence of IFD. Initial of recent ste­roid expo­sure in esca­lat­ing the risk of IFD in leu­ke­
obser­va­tional stud­ies across all­types of pedi­at­ric ALL reported mia patients should not be overlooked.13,14 The declin­ing reli­ance
an IFD rate of 10%, with IFD rates nearing 20% in relapsed ALL on ste­roids in con­tem­po­rary AML che­mo­ther­apy reg­i­mens and
patients.4,5 However, these stud­ies did not employ the published increas­ing reli­ance on dexa­meth­a­sone in cer­tain ALL reg­i­mens
proven or prob­a­ble IFD cri­te­ria and thus may have overestimated might explain some of the reported decline in IFD inci­dence in
the true rate of IFD. Contemporary stud­ies sug­gest that proven the for­mer and increase in IFD inci­dence in the lat­ter.15,16
or prob­a­ble IFD rates in stan­dard-risk ALL patients have declined Results from a small ran­ dom­ized trial performed by Pizzo
to 5.8% but remain high at 12.9% for relapsed ALL patients and at et al in the early 1980s supported the util­ity of empir­i­cal anti­
8.7% to 15.2% for high-risk ALL patients.6,7 fun­gal ther­apy in patients with per­sis­tent fever and neutrope-
Although rates of IFD have var­ied by time period and by leu­ nia despite broad-spec­trum empir­i­cal anti­bi­otic ther­apy.17 This
ke­mia type, their per­sis­tence as oppor­tu­nis­tic infec­tions in these approach quickly became the stan­dard of care and has remained
patient pop­u­la­tions remains a con­cern. IFDs con­tinue to be a as such for the bet­ter part of 4 decades. In recent years there has
feared com­pli­ca­tion of leu­ke­mia treat­ment because of the poten­ been increas­ing focus on advanc­ing best prac­tices for reduc­ing
tial for sig­nif­i­cant mor­bid­ity and mor­tal­ity asso­ci­ated with these IFD mor­bid­ity and mor­tal­ity in pedi­at­ric AML and ALL patients.
path­ o­gens. Invasive can­ di­di­
as­is is the most com­ mon form of Several impor­tant inves­ti­ga­tions and inter­na­tional guide­lines
proven or prob­a­ble IFD and has been asso­ci­ated with a 10% attrib­ have been published that pro­ vide guid­ ance on the util­ ity of
ut­­able mor­tal­ity in chil­dren.8 Proven or prob­ab ­ le inva­sive mold pro­phy­lac­tic and pre­emp­tive anti­fun­gal ther­apy approaches as
dis­eases, such as asper­gil­lo­sis and mucormycosis, are less com­ well as fun­gal diag­nos­tic tools for these patient pop­u­la­tions. In
mon but have esti­mated case fatal­ity rates in excess of 30%.9,10 this review we use the pro­vided clin­i­cal case to illus­trate how
Multiple fac­tors pre­dis­pose chil­dren with leu­ke­mia to IFD. They published evi­ dence can sup­ port impor­ tant IFD man­ age­ment
include neutropenia, expo­sure to high-dose ste­roids, hyper­gly­ deci­sions at var­i­ous time points dur­ing a period of prolonged
ce­mia, and prolonged broad-spec­trum anti­bi­ot­ics.11 Prolonged neutropenia after leu­ke­mia che­mo­ther­apy. Knowledge gaps in
neutropenia is by far the most documented of these fac­tors. need of future inves­ti­ga­tion are noted.
The dura­tion of neutropenia at which the risk for IFD increases
sig­nif­i­cantly is not clearly defined and likely varies by patient. Should anti­fun­gal pro­phy­laxis be admin­is­tered dur­ing
However, in gen­eral the risk for IFD is esti­mated to increase sub­ prolonged neutropenia peri­ods?
stan­tially in patients with neutropenia last­ing for more than 10 In the first decade of the cur­rent cen­tury, the approach to anti­
days. The dura­tion of neutropenia will vary depending on the fun­gal pro­phy­laxis for neutropenia after receipt of inten­sive che­
che­mo­ther­apy reg­i­men admin­is­tered, but most con­tem­po­rary mo­ther­apy for leu­ke­mia var­ied sig­nif­i­cantly. An inter­na­tional
che­mo­ther­apy reg­i­mens for AML and high-risk or relapsed ALL sur­vey performed by Sung et al found that 77% and 91% of Chil-
will result in mul­ti­ple epi­sodes of sustained abso­lute neu­tro­phil dren’s Oncology Group Centers and Berlin-Frankfurt-Muen­ster
counts below 200 cells/µL that last for 2 to 3 weeks.11,12 While Group Centers, respec­ tively, were using 5 dif­ fer­ ent anti­
fun­
gal
neutropenia is the dom­i­nant risk fac­tor for IFD, the impor­tance agents to admin­is­ter anti­fun­gal pro­phy­laxis to chil­dren with AML.18

362  |  Hematology 2021  |  ASH Education Program


This var­i­a­tion in prac­tice was not par­tic­u­larly sur­pris­ing because a mul­ti­dis­ci­plin­ary qual­ity improve­ment ini­tia­tive inclu­sive of
of the lim­ited data from pedi­at­ric obser­va­tional or ran­dom­ized oncol­ogy, infec­tious dis­eases, and anti­mi­cro­bial stew­ard­ship
stud­ies com­par­ing the effec­tive­ness of dif­fer­ent anti­fun­gal pro­ experts could be help­ful to guide deci­sions on local fun­gal pre­
phy­laxis approaches. Adult neutropenia man­age­ment guide­lines ven­tion efforts.
did exist at that time and recommended anti­fun­gal pro­phy­laxis My cur­ rent per­ sonal pref­
er­ ence is to use an echinocandin
dur­ing peri­ods of neutropenia resulting from inten­sive che­mo­ as the anti­fun­gal pro­phy­laxis agent. This is because echinocan-
ther­apy. This included a spe­cific rec­om­men­da­tion of posacon- dins have a rel­a­tively lim­ited side effect pro­file with less con­cern
azole pro­ phy­laxis for patients older than 13 years receiv­ ing about alter­ing the phar­ma­co­ki­net­ics of other admin­is­tered med­
che­mo­ther­apy for AML, which was based on ran­dom­ized con­ i­ca­tions. Additionally, our patients with antic­i­pated prolonged
trolled trial data reveal­ing the effi­cacy of posaconazole com­ neutropenia after inten­sive che­mo­ther­apy are often mon­i­tored
pared to fluconazole or itraconazole in an adult AML pop­u­la­tion.19 in the hos­pi­tal for their peri­ods of neutropenia, so a once-a-day
Fortunately, larger pedi­at­ric com­par­a­tive effec­tive­ness stud­ies intra­ve­nous med­i­ca­tion is fea­si­ble. Finally, chal­lenges in achiev­ing
soon followed. A ret­ro­spec­tive obser­va­tional cohort study lev­er­ the appro­pri­ate antimold azole dose for each child is a lim­it­ing
aged inpa­tient hos­pi­tal admin­is­tra­tive data and var­i­a­tion in anti­ com­po­nent of this class of anti­fun­gal agents. However, as out­pa­

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/361/1851411/361fisher.pdf by guest on 13 December 2021


fun­gal pro­phy­laxis uti­li­za­tion to reveal that anti­fun­gal pro­phy­laxis tient obser­va­tion of chil­dren dur­ing peri­ods of prolonged neutro-
in chil­dren, ado­les­cents, and young adults with AML did reduce penia is increas­ingly uti­lized, the ben­e­fits of enteral pro­phy­laxis
mor­tal­ity and resource uti­li­za­tion com­pared to no pro­phy­laxis.20 will increase, which may favor antimold azoles. Knowledge gaps
This study con­firmed the pre­sumed ben­e­fit of anti­fun­gal pro­ still exist that if filled would improve the abil­ity to uti­lize azoles in
phy­laxis in pedi­at­ric patients but could not deter­mine whether this set­ting. These gaps include ideal dos­ing for chil­dren, spe­cif­i­
the pro­phy­lac­tic agent needed to include antimold activ­ity. A cally for posaconazole, and tar­get troughs for pro­phy­laxis.
sub­se­quent ran­dom­ized con­trolled trial performed within the
Children’s Oncology Group (ACCL0933) helped to answer this Is sur­veil­lance test­ing with fun­gal bio­mark­ers warranted
ques­tion by com­par­ing the effi­cacy of caspofungin pro­phy­laxis dur­ing prolonged neutropenia?
with fluconazole pro­phy­laxis.2 In this trial caspofungin pro­phy­ An increas­ing num­ber of com­mer­cially avail­­able non-cul­ture-
laxis sig­nif­i­cantly lowered the risk of proven or prob­a­ble IFD, based fun­gal bio­mark­ers offer the poten­tial for sur­veil­lance test­
spe­cif­i­cally reduc­ing the fre­quency of inva­sive asper­gil­lo­sis. ing to detect the pres­ence of an IFD at early onset. The most
As these and other pedi­at­ric stud­ies began to pop­u­late the lit­ well stud­ied of these bio­mark­ers are the Aspergillus galactoman-
er­a­ture, an inter­na­tional sup­port­ive care com­mit­tee endeav­ored nan (GM) enzyme immu­no­as­say (EIA; Platelia™) and the beta-D-
to col­late and inter­pret the data in a pedi­at­ric-spe­cific, sys­temic glu­can (BDG) assay (Fungitell®). The GM EIA is designed to detect
anti­fun­gal pro­phy­laxis guide­line.21 This guide­line included a sys­ GM, which is a cell wall com­po­nent of Aspergillus spe­cies, while
tem­atic review of stud­ies com­par­ing dif­fer­ent anti­fun­gal pro­phy­ the BDG assay aims to detect BDGs found in the cell walls of var­
laxis reg­i­mens. These data informed a strong rec­om­men­da­tion i­ous path­o­genic fungi, includ­ing Aspergillus and Candida spp.
for antimold pro­phy­laxis in pedi­at­ric AML patients and a weak Results from stud­ies assessing the util­ity of these 2 bio­mark­ers as
rec­om­men­da­tion for antimold pro­phy­laxis after inten­sive che­ a sur­veil­lance tool for early IFD dur­ing peri­ods of prolonged neu-
mo­ther­apy for high-risk or relapsed ALL. The lat­ter was a weak tropenia in adults were received with much opti­mism. Maertens
rec­om­men­da­tion because of lim­ited IFD epi­de­mi­­ol­ogy and com­ et al assessed the GM EIA’s abil­ity to detect inva­sive asper­gil­lo­sis
par­a­tive effec­tive­ness data spe­cific to this pop­u­la­tion. This weak dur­ing 362 prolonged neutropenic peri­ods in adults receiv­ing
rec­om­men­da­tion cou­pled with chal­lenges in admin­is­ter­ing anti­ che­mo­ther­apy for hema­to­logic malig­nancy or con­di­tion­ing for a
fun­gal pro­phy­laxis to chil­dren with ALL (eg, greater poten­tial for hema­to­poi­etic cell trans­plan­ta­tion (HCT).22 They found the oper­
drug-drug inter­ac­tions and man­age­ment mostly in the out­pa­tient at­ing char­ac­ter­is­tics of the GM EIA for sur­veil­lance test­ing to be
set­ting) has likely resulted in less rou­tine use of anti­fun­gal pro­phy­ rea­son­able (sen­si­tiv­ity, spec­i­fic­ity, and pos­i­tive pre­dic­tive val­ues
laxis in patients with ALL com­pared to those with AML. This could, (PPV) and neg­a­tive pre­dic­tive val­ues (NPV) were 72.9%, 99.1%,
in part, explain the pre­vi­ously cited increas­ing rates of IFD in ALL 93.1%, and 70.8%, respec­tively). In a sim­i­larly designed study,
patients. Randomized con­trolled tri­als assessing the ideal pro­phy­ Odabasi et al assessed the BDG assay as a sur­veil­lance tool for
laxis approach in ALL-spe­cific patient pop­u­la­tions are warranted. proven or prob­a­ble IFD in adults with neutropenia after AML che­
Notably, the guide­line remained flex­i­ble on which antimold mo­ther­apy. They found the BDG assay oper­at­ing char­ac­ter­is­tics
agent to use for pro­phy­laxis in either AML or ALL patients, sug- to also be rea­son­able (sen­si­tiv­ity, spec­i­fic­ity, PPV, and NPV were
gesting an echinocandin or an azole with antimold activ­ity. The 100%, 90%, 43%, and 100%, respec­tively).23 These 2 pro­spec­
choice of anti­fun­gal agent depends on fac­tors such as the age tive obser­va­tional cohorts led to US Food and Drug Adminis-
of the child, inpa­tient vs out­pa­tient set­ting of neutropenia man­ tration approval for these bio­mark­ers, and many pedi­at­ric and
age­ment, and con­cern for drug-drug inter­ac­tions. The guide­line adult cen­ters adopted an IFD sur­veil­lance test­ing approach (ie,
did rec­om­mend against using a sys­temic amphotericin for­mu­la­ weekly or twice-weekly GM EIA and BDG assays) dur­ing peri­ods
tion for pro­phy­laxis because of a documented increased risk of of prolonged neutropenia but prior to the onset of any signs or
adverse effects. symp­toms of IFD.
Before adapting guide­line rec­om­men­da­tions to their patients, The trans­port­abil­ity of these adult data to chil­dren was appro­
cli­ni­cians need to assess the epi­de­mi­­ol­ogy of IFD across their pri­ately questioned. First, an assess­ ment of the BDG assay in
local leu­ke­mia pop­u­la­tion. A cen­ter could have dif­fer­ing rates oth­er­wise healthy chil­dren suggested higher base­line lev­els of
of proven or prob­a­ble IFD and/or a dif­fer­ent fun­gal path­o­gen BDG in chil­dren and ado­les­cents, which could lead to fre­quent
dis­tri­bu­tion that under­mines the gen­er­al­iz­abil­ity of results from false-pos­i­tive results.24 Subsequently, a series of smaller stud­
published stud­ ies to their patient pop­ u­
la­
tion. In this set­ ting, ies were published assessing the GM EIA and the BDG assay in
Prakash Singh Shekhawat
Fungal man­age­ment in leu­ke­mia  |  363
Table 2. Operating char­ac­ter­is­tics of the Aspergillus GM EIA and BDG assay for sur­veil­lance test­ing in pedi­at­ric patients with
can­cer or under­go­ing HCT

Study cohort Study’s IFD


Author, year size inci­dence rate Sensitivity Specificity PPV NPV
Aspergillus GM EIA
Hovi et al34 98 8.0% 93% 50% 14% 99%
Steinbach et al35 64 0% 87% 0% 0% 98%
Hayden et al36 56 30.4% 87% 65% 69% 85%
Badiee et al37 62 16.1% 92% 90% 82% 96%
Koltze et al38
34 17.6% 100% 83% 100% 97%
Fisher et al26 425 0.7% 0% 98.8% 0% 99.9%

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/361/1851411/361fisher.pdf by guest on 13 December 2021


BDG assay
Badiee et al37 62 16.1% 46% 50% 26% 71%
Koltze et al38
34 17.6% 29% 83% 20% 89%
Fisher et al26 425 1.4% 0% 94.7% 0% 99.8%
Data in this table adapted from Lehrnbecher et al, Table 3.25

pedi­at­ric patients with prolonged neutropenia after che­mo­ther­apy broad-spec­trum anti­bi­otic ther­apy, were ran­dom­ized to receive
or HCT con­di­tion­ing reg­i­mens. These stud­ies were sys­tem­at­i­cally or not receive sys­temic con­ven­tional ampho­ter­i­cin B ther­apy.17
reviewed by Lehrnbecher et al,25 and the results are sum­ma­rized Those sub­jects ran­dom­ized to anti­fun­gal ther­apy were observed
in Table 2. The oper­at­ing char­ac­ter­is­tics var­ied widely for both to have a faster res­ol­u­tion of fever. This approach was labeled
assays across stud­ies included in the review. The authors con­ “empir­i­cal anti­fun­gal ther­apy for prolonged fever and neutrope-
cluded that lower rates (ie, lower pre­test prob­a­bil­ity) of proven or nia.” Multiple stud­ies assessing the util­ity of empir­i­cal anti­fun­gal
prob­ab­ le IFD in the more recent cohorts increased the like­li­hood ther­apy were sub­se­quently performed, and col­lec­tively, these
of false-pos­i­tive results, which lim­ited the util­ity of sur­veil­lance stud­ies con­firmed the ben­e­fits of empir­i­cal anti­fun­gal ther­apy.28
test­ing. A more recent large obser­va­tional cohort of pedi­at­ric AML Based on these data, the 2017 update of the inter­na­tional pedi­at­
patients con­firmed the lim­ited util­ity of the GM EIA and the BDG ric fever and neutropenia guide­lines recommended that patients
assays for sur­veil­lance test­ing (Table 2).26 Prior to these pub­li­ca­ at high risk for IFD and with fever and neutropenia persisting
tions, our cen­ter had been employing weekly sur­veil­lance test­ing lon­ger than 96 hours receive empir­i­cal anti­fun­gal ther­apy with
with the GM EIA. After reviewing the data from these pedi­at­ric either an echinocandin or lipo­so­mal ampho­ter­i­cin B.29
stud­ies, we have elected to stop sur­veil­lance test­ing. However, this rec­om­men­da­tion existed prior to the recently
Other nonculture diag­ nos­
tic plat­forms besides the GM EIA published rec­om­men­da­tion to admin­is­ter antimold pro­phy­laxis
and BDG assays may be appro­ pri­ate for sur­ veil­
lance test­ing. to patients with antic­i­pated prolonged neutropenia after inten­
These would include mold-spe­ cific poly­ mer­ase chain reac­ tion sive leu­ke­mia che­mo­ther­apy. This rec­om­men­da­tion to admin­is­ter
(PCR) assays, spe­cif­i­cally for Aspergillus spp. and path­o­gens of antimold pro­phy­laxis at the start of neutropenia raises the ques­
the Mucorales order, and plasma micro­bial cell-free DNA (cfDNA) tion of whether empir­ic ­ al anti­fun­gal ther­apy is still needed. As the
sequenc­ ing plat­forms. Assessments of mold-spe­ cific PCRs in exam­ple case illus­trates, some pedi­at­ric patients will develop pro-
pedi­at­ric cohorts have focused on sce­nar­ios in which the patient longed fever and neutropenia despite receiv­ing both pro­phy­lac­tic
is displaying clin­i­cal symp­toms concerning for IFD (ie, prolonged antimold ther­apy and empir­i­cal broad-spec­trum anti­bi­ot­ics at the
fever and neutropenia) but they have not been assessed under a onset of fever. In the ran­dom­ized trial of caspofungin vs fluconazole
sur­veil­lance pro­to­col.25 A recent inves­ti­ga­tion reported the expe­ pro­phy­laxis in pedi­at­ric AML, many patients devel­oped prolonged
ri­ence of monthly cfDNA sequenc­ing for sur­veil­lance of IFD in 40 fever and neutropenia—even those ran­dom­ized to the caspofungin
at-risk pedi­at­ric patients with can­cer or under­go­ing HCT.27 While treat­ment arm.2 Unfortunately, there are lim­ ited data to guide
a sub­set of patients had cfDNA sequenc­ing that revealed the pres­ whether a patient like the one in our clin­i­cal case should con­tinue
ence of a fun­gal path­o­gen, it is not clear that sur­veil­lance cfDNA the antimold pro­phy­lac­tic agent started at the begin­ning of neu-
test­ing improved the clin­i­cal man­age­ment of these patients. Addi- tropenia or if the prolonged fever and neutropenia should prompt
tionally, on some of the cfDNA sequenc­ing results mul­ti­ple path­o­ a tran­si­tion to a dif­fer­ent antimold agent to serve as empir­i­cal anti­
gens of unclear sig­nif­i­cance were iden­ti­fied. Future inves­ti­ga­tions fun­gal ther­apy for the remain­der of the neutropenic period. This is
in larger pedi­at­ric cohorts are nec­es­sary before con­sid­er­ing the an impor­tant knowl­edge gap deserv­ing fur­ther inves­ti­ga­tion.
com­mit­ment of health care resources toward sur­veil­lance test­ing Until data are avail­­able, cli­ni­cians will need to dis­cuss locally
with either PCR or cfDNA sequenc­ing diag­nos­tic plat­forms. how they want to approach such patients. Notably, the break­
through proven or prob­a­ble IFD rate for sub­jects receiv­ing caspo-
Empirical anti­fun­gal ther­apy fungin pro­phy­laxis in the ran­dom­ized trial was 3.1%,2 which may
As noted above, Pizzo et al published their land­mark ran­dom­ be too high for some cli­ni­cians not to change to a dif­fer­ent empir­
ized trial in 1982. A total of 34 chil­dren, ado­les­cent, and young i­cal antimold ther­a­peu­tic. However, a true expan­sion in anti­fun­gal
adult patients with per­sis­tent fever and neutropenia, despite cov­er­age from pro­phy­lac­tic caspofungin would likely neces­si­

364  |  Hematology 2021  |  ASH Education Program


An 11-year-old female with AML
develops neutropenia aer
inducon II chemotherapy

Surveillance fungal biomarker tesng


Start an-mold prophylaxis therapy
• Do NOT perform surveillance biomarker
• Suggest an echinocandin as first-line therapy
tesng if on an-mold prophylaxis
• If unable to administer echinocandin
• Posive biomarker test result more likely to
consider an-mold azole
be a false posive

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/361/1851411/361fisher.pdf by guest on 13 December 2021


On echinocandin prophylaxis she
develops new fever. Despite empirical
anbiocs her fever and neutropenia
persist for >96 hours

What are next steps?


Comprehensive Clinical Exam

If no focal or systemic signs besides fever: If any focal or systemic signs concerning for IFD:
• Connue an-mold agent started as • Broaden an-mold anfungal therapy
prophylaxis and monitor closely • Consult with immunocompromised infecous
• Alternavely, could pursue a pre-empve diseases specialist to guide addional
diagnosc approach (see text) diagnosc tesng

Figure 1. Conceptual model for antifungal diagnostic and therapeutic decisions during neutropenia after chemotherapy for AML.

tate a tran­si­tion to a lipid ampho­ter­i­cin B for­mu­la­tion, which can was noninferior for sur­vival and resulted in a sig­nif­i­cant reduc­tion
have a sig­nif­i­cant tox­ic­ity risk. Therefore, in this sit­u­a­tion I elect in anti­fun­gal expo­sure.30 However, sig­nif­i­cantly more sub­jects in
to review the his­tory and cur­rent clin­i­cal state of each patient the pre­emp­tive arm sustained a prob­a­ble or proven IFD. Santolaya
before decid­ing whether to broaden beyond the pro­phy­lac­tic et al performed a sim­i­lar ran­dom­ized trial in 149 pedi­at­ric patients
anti­fun­gal agent. If a patient has no symp­toms beyond fever and with per­sis­tent fever and neutropenia.31 Patients in the pre­emp­
neutropenia and the patient’s med­i­cal his­tory and expo­sure his­ tive arm had a sig­nif­i­cant reduc­tion in anti­fun­gal expo­sure and
tory do not increase my con­cern for IFD, I will rec­om­mend main- mor­tal­ity rates sim­i­lar to those started on empir­i­cal anti­fun­gal
taining ther­apy with the same antimold pro­phy­laxis agent. ther­apy. The 8th Euro­pean Conference on Infections in Leukaemia
ref­er­enced the lat­ter study in stat­ing their grade B rec­om­men­
Preemptive anti­fun­gal ther­apy for prolonged fever da­tion that pre­emp­tive ther­apy may be con­sid­ered as an alter­
and neutropenia na­tive approach to empir­i­cal ther­apy in pedi­at­ric patients.32 The
Preemptive anti­ fun­
gal ther­apy is a con­ cept gar­ner­
ing increas­ chal­lenge with a pre­emp­tive approach in pedi­at­ric patients is
ing inter­est. Evidence from stud­ies assessing this approach may that it often relies on fun­gal bio­mark­ers to guide final deci­sions.
even­tu­ally solve the clin­i­cal conun­drum of whether a patient with Pediatric-spe­cific data on the util­ity of these bio­mark­ers to either
prolonged fever and neutropenia should remain on anti­ fun­gal detect or exclude IFD at the time point of prolonged fever and
pro­phy­laxis vs transitioning to empir­i­cal anti­fun­gal ther­apy. A pre­ neutropenia are not read­ily avail­­able. As such it is dif­fi­cult to rely
emp­tive ther­apy approach relies on the uti­li­za­tion of results from on these bio­mark­ers for this pre­emp­tive vs empir­i­cal clin­i­cal deci­
a clin­i­cal exam­i­na­tion and from diag­nos­tic stud­ies that inform the sion. Hopefully, future inves­ti­ga­tions will define the util­ity of exist-
deci­sion to alter ther­apy for a given patient. In 2009 Cordonnier ing and novel bio­mark­ers for this clin­i­cal sce­nario.
et al com­pared a pre­emp­tive anti­fun­gal approach to an empir­i­cal
one in 293 pedi­at­ric and adult patients with prolonged or recur­ Summary
rent fever and neutropenia. The pre­emp­tive approach included The diag­nos­tic and ther­a­peu­tic approaches to IFD in pedi­at­
clin­i­cal exam­i­na­tions, GM EIA test­ing, and radio­graphic imag­ing. ric patients with AML and high-risk or relapsed ALL con­tinue to
Any signs or symp­toms of IFD from those stud­ies prompted the evolve. In the past decade, increas­ingly avail­­able pedi­at­ric-spe­
ini­ti­a­tion or broad­en­ing of anti­fun­gal ther­apy. Preemptive ther­apy cific evi­dence has informed pedi­at­ric-spe­cific guide­lines. Figure 1
Prakash Singh Shekhawat
Fungal man­age­ment in leu­ke­mia  |  365
pro­vi­des a con­cep­tual model of my inter­pre­ta­tion and appli­ca­ 13. Johnston DL, Lewis V, Yanofsky R, et al. Invasive fun­gal infec­tions in pae­di­
tion of some of the existing data and guide­lines detailed above at­ric acute mye­loid leu­kae­mia. Mycoses. 2013;56(4):482-487.
14. Lai HP, Chen YC, Chang LY, et al. Invasive fun­gal infec­tion in chil­dren with
using the pro­posed clin­i­cal case. Antimold ther­apy for chil­dren per­sis­tent febrile neutropenia. J Formos Med Assoc. 2005;104(3):174-179.
with antic­i­pated prolonged neutropenia after AML or ALL che­ 15. Kavcic M, Fisher BT, Li Y, et al. Induction mor­tal­ity and resource uti­li­za­tion
mo­ther­apy should be con­sid­ered the stan­dard of care. While GM in chil­dren treated for acute mye­loid leu­ke­mia at free-stand­ing pedi­at­ric
EIA or BDG assays should be dis­cour­aged for IFD sur­veil­lance, hos­pi­tals in the United States. Cancer. 2013;119(10):1916-1923.
16. Teachey DT, O’Connor D. How I treat newly diag­nosed T-cell acute lym­
more data are needed on using these and other novel bio­mark­
pho­blas­tic leu­ke­mia and T-cell lym­pho­blas­tic lym­phoma in chil­dren. Blood.
ers at other clin­i­cal time points. Finally, for patients who develop 2020;135(3):159-166.
prolonged fever and neutropenia while on antimold pro­phy­laxis, 17. Pizzo PA, Robichaud KJ, Gill FA, Witebsky FG. Empiric anti­bi­otic and anti­
it is not always nec­es­sary to tran­si­tion to a dif­fer­ent empir­i­cal fun­gal ther­apy for can­cer patients with prolonged fever and granulocyto-
anti­fun­gal agent. More data are needed to deter­mine the ideal penia. Am J Med. 1982;72(1):101-111.
18. Lehrnbecher T, Ethier MC, Zaoutis T, et al. International var­i­a­tions in infec­
approach to this lat­ter clin­i­cal sce­nario. tion sup­port­ive care prac­tices for pae­di­at­ric patients with acute mye­loid
leu­kae­mia. Br J Haematol. 2009;147(1):125-128.
Conflict-of-inter­est dis­clo­sure

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/361/1851411/361fisher.pdf by guest on 13 December 2021


19. Cornely OA, Maertens J, Winston DJ, et  al. Posaconazole vs. fluconazole
Brian T. Fisher: research funding: Pfizer, Merck; chair: Safety Mon- or itraconazole pro­phy­laxis in patients with neutropenia. N Engl J Med.
2007;356(4):348-359.
itoring Board, Astellas inves­ti­ga­tion of isavuconazole.
20. Fisher BT, Kavcic M, Li Y, et  al. Antifungal pro­ phy­
laxis asso­ ci­
ated with
decreased induc­tion mor­tal­ity rates and resources uti­lized in chil­dren with
Off-label drug use new-onset acute mye­loid leu­ke­mia. Clin Infect Dis. 2014;58(4):502-508.
Brian T. Fisher: nothing to disclose. 21. Lehrnbecher T, Fisher BT, Phil­ lips B, et  al. Clinical prac­ tice guide­
line
for sys­temic anti­fun­gal pro­phy­laxis in pedi­at­ric patients with can­cer
Correspondence and hema­to­poi­etic stem-cell trans­plan­ta­tion recip­i­ents. J Clin Oncol.
Brian T. Fisher, Division of Infectious Diseases, Children’s Hospital 2020;38(27):3205-3216.
22. Maertens J, Verhaegen J, Lagrou K, Van Eldere J, Boogaerts M. Screening
of Philadelphia, Roberts Pediatric Research Center, 2716 South
for cir­cu­lat­ing galactomannan as a non­in­va­sive diag­nos­tic tool for inva­sive
Str, Rm 10-362, Philadelphia, PA 19146; e-mail: fisherbria@chop​ asper­gil­lo­sis in prolonged neutropenic patients and stem cell trans­plan­ta­
­.edu. tion recip­i­ents: a pro­spec­tive val­i­da­tion. Blood. 2001;97(6):1604-1610.
23. Odabasi Z, Mattiuzzi G, Estey E, et al. Beta-D-glu­can as a diag­nos­tic adjunct
References for inva­sive fun­gal infec­tions: val­i­da­tion, cut­off devel­op­ment, and per­for­
1. Sung L, Lange BJ, Gerbing RB, Alonzo TA, Feusner J. Microbiologically mance in patients with acute mye­log­e­nous leu­ke­mia and myelodysplastic
documented infec­ tions and infec­ tion-related mor­tal­
ity in chil­dren with syn­drome. Clin Infect Dis. 2004;39(2):199-205.
acute mye­loid leu­ke­mia. Blood. 2007;110(10):3532-3539. 24. Smith PB, Ben­ja­min DK Jr, Alexander BD, Johnson MD, Finkelman MA, Stein-
2. Fisher BT, Zaoutis T, Dvorak CC, et  al. Effect of caspofungin vs fluco- bach WJ. Quantification of 1,3-beta-D-glu­can lev­els in chil­dren: pre­lim­i­nary
nazole pro­phy­laxis on inva­sive fun­gal dis­ease among chil­dren and young data for diag­nos­tic use of the beta-glu­can assay in a pedi­at­ric set­ting. Clin
adults with acute mye­loid leu­ke­mia: a ran­dom­ized clin­i­cal trial. JAMA. Vaccine Immunol. 2007;14(7):924-925.
2019;322(17):1673-1681. 25. Lehrnbecher T, Robinson PD, Fisher BT, et al. Galactomannan, β-D-glu­can,
3. Donnelly JP, Chen SC, Kauffman CA, et  al. Revision and update of the and poly­mer­ase chain reac­tion-based assays for the diag­no­sis of inva­
con­sen­sus def­i­ni­tions of inva­sive fun­gal dis­ease from the Euro­pean Orga- sive fun­ gal dis­ ease in pedi­ at­
ric can­ cer and hema­ to­
poi­ etic stem cell
nization for Research and Treatment of Cancer and the Mycoses Study trans­plan­ta­tion: a sys­tem­atic review and meta-anal­y­sis. Clin Infect Dis.
Group Education and Research Consortium. Clin Infect Dis. 2020;71(6): 2016;63(10):1340-1348.
1367-1376. 26. Fisher BT, Westling T, Boge CLK, et al. Prospective eval­ua ­ ­tion of galacto-
4. Rosen GP, Nielsen K, Glenn S, Abelson J, Deville J, Moore TB. Invasive fun­ mannan and (1 → 3) β-d-glu­can assays as diag­nos­tic tools for inva­sive fun­
gal infec­tions in pedi­at­ric oncol­ogy patients: 11-year expe­ri­ence at a sin­gle gal dis­ease in chil­dren, ado­les­cents, and young adults with acute mye­loid
insti­tu­tion. J Pediatr Hematol Oncol. 2005;27(3):135-140. leu­ke­mia receiv­ing fun­gal pro­phy­laxis. J Pediatric Infect Dis Soc. 2021;
5. Leahey AM, Bunin NJ, Belasco JB, Meek R, Scher C, Lange BJ. Novel multi- 10(8):864-871.
agent che­mo­ther­apy for bone mar­row relapse of pedi­at­ric acute lym­pho­ 27. Armstrong AE, Rossoff J, Hollemon D, Hong DK, Muller WJ, Chaudhury
blas­tic leu­ke­mia. Med Pediatr Oncol. 2000;34(5):313-318. S. Cell-free DNA next-gen­er­a­tion sequenc­ing suc­cess­fully detects infec­
6. Castagnola E, Palmisani E, Mesini A, Saffioti C, Micalizzi C, Dufour C. Com- tious path­o­gens in pedi­at­ric oncol­ogy and hema­to­poi­etic stem cell trans­
ment on: inva­sive fun­gal infec­tions in chil­dren with acute lym­pho­blas­tic plant patients at risk for inva­sive fun­gal dis­ease. Pediatr Blood Cancer.
leu­ke­mia. Pediatr Blood Cancer. 2020;67(1):e28035. 2019;66(7):e27734.
7. Wang SS, Kotecha RS, Bernard A, et al. Invasive fun­gal infec­tions in chil­dren 28. Goldberg E, Gafter-Gvili A, Robenshtok E, Leibovici L, Paul M. Empirical
with acute lym­pho­blas­tic leu­kae­mia: results from four Aus­tra­lian cen­tres, anti­fun­gal ther­apy for patients with neutropenia and per­sis­tent fever: sys­
2003-2013. Pediatr Blood Cancer. 2019;66(10):e27915. tem­atic review and meta-anal­y­sis. Eur J Cancer. 2008;44(15):2192-2203.
8. Zaoutis TE, Argon J, Chu J, Berlin JA, Walsh TJ, Feudtner C. The epi­de­mi­­ 29. Lehrnbecher T, Robinson P, Fisher B, et al. Guideline for the man­age­ment of
ol­ogy and attrib­ut­­able out­comes of candidemia in adults and chil­dren fever and neutropenia in chil­dren with can­cer and hema­to­poi­etic stem-cell
hos­pi­tal­ized in the United States: a pro­pen­sity anal­y­sis. Clin Infect Dis. trans­plan­ta­tion recip­i­ents: 2017 update. J Clin Oncol. 2017;35(18):2082-2094.
2005;41(9):1232-1239. 30. Cordonnier C, Pautas C, Maury S, et al. Empirical ver­sus pre­emp­tive anti­
9. Burgos A, Zaoutis TE, Dvorak CC, et al. Pediatric inva­sive asper­gil­lo­sis: a fun­gal ther­apy for high-risk, febrile, neutropenic patients: a ran­dom­ized,
mul­ti­cen­ter ret­ro­spec­tive anal­y­sis of 139 con­tem­po­rary cases. Pediatrics. con­trolled trial. Clin Infect Dis. 2009;48(8):1042-1051.
2008;121(5):e1286-e1294. 31. Santolaya ME, Alvarez AM, Acuña M, et al. Efficacy of pre-emptive ver­sus
10. Wattier RL, Dvorak CC, Hoffman JA, et  al. A pro­ spec­ tive, inter­
na­
tional empir­i­cal anti­fun­gal ther­apy in chil­dren with can­cer and high-risk febrile
cohort study of inva­sive mold infec­tions in chil­dren. J Pediatric Infect Dis neutropenia: a ran­dom­ized clin­i­cal trial. J Antimicrob Chemother. 2018;
Soc. 2015;4(4):313-322. 73(10):2860-2866.
11. Fisher BT, Robinson PD, Lehrnbecher T, et al. Risk fac­tors for inva­sive fun­gal 32. Groll AH, Pana D, Lanternier F, et al; 8th Euro­pean Conference on Infections
dis­ease in pedi­at­ric can­cer and hema­to­poi­etic stem cell trans­plan­ta­tion: a in Leukaemia. 8th Euro­pean Conference on Infections in Leukaemia: 2020
sys­tem­atic review. J Pediatric Infect Dis Soc. 2018;7(3):191-198. guide­lines for the diag­no­sis, pre­ven­tion, and treat­ment of inva­sive fun­gal
12. Alexander S, Fisher BT, Gaur AH, et al; Children’s Oncology Group. Effect of dis­eases in pae­di­at­ric patients with can­cer or post-haematopoietic cell
levofloxacin pro­phy­laxis on bac­ter­emia in chil­dren with acute leu­ke­mia or trans­plan­ta­tion. Lancet Oncol. 2021;22(6):e254-e269.
under­go­ing hema­to­poi­etic stem cell trans­plan­ta­tion: a ran­dom­ized clin­i­cal 33. De Pauw B, Walsh TJ, Donnelly JP, et al; Euro­pean Organization for Research
trial. JAMA. 2018;320(10):995-1004. and Treatment of Cancer/Invasive Fungal Infections Cooperative Group;

366  |  Hematology 2021  |  ASH Education Program


National Institute of Allergy and Infectious Diseases Mycoses Study Group 36. Hayden R, Pounds S, Knapp K, et al. Galactomannan antigenemia in pedi­
Consensus Group. Revised def­i­ni­tions of inva­sive fun­gal dis­ease from the at­ric oncol­ogy patients with inva­sive asper­gil­lo­sis. Pediatr Infect Dis J.
Euro­pean Organization for Research and Treatment of Cancer/Invasive 2008;27(9):815-819.
Fungal Infections Cooperative Group and the National Institute of Allergy 37. Badiee P, Alborzi A, Karimi M, et  al. Diagnostic poten­tial of nested PCR,
and Infectious Diseases Mycoses Study Group (EORTC/MSG) Consensus galactomannan EIA, and beta-D-glu­can for inva­sive asper­gil­lo­sis in pedi­at­
Group. Clin Infect Dis. 2008;46(12):1813-1821. ric patients. J Infect Dev Ctries. 2012;6(4):352-357.
34. Hovi L, Saxen H, Saarinen-Pihkala UM, Vettenranta K, Meri T, Richardson 38. Koltze A, Rath P, Schöning S, et al. β-D-glu­can screen­ing for detec­tion of
M. Prevention and mon­ i­
tor­
ing of inva­sive fun­
gal infec­
tions in pedi­ at­
ric inva­sive fun­gal dis­ease in chil­dren under­go­ing allo­ge­neic hema­to­poi­etic
patients with can­cer and hema­to­logic dis­or­ders. Pediatr Blood Cancer. stem cell trans­plan­ta­tion. J Clin Microbiol. 2015;53(8):2605-2610.
2007;48(1):28-34.
35. Steinbach WJ, Addison RM, McLaughlin L, et  al. Prospective Aspergillus
galactomannan anti­gen test­ing in pedi­at­ric hema­to­poi­etic stem cell trans­ © 2021 by The Amer­i­can Society of Hematology
plant recip­i­ents. Pediatr Infect Dis J. 2007;26(7):558-564. DOI 10.1182/hema­tol­ogy.2021000267

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/361/1851411/361fisher.pdf by guest on 13 December 2021

Prakash Singh Shekhawat


Fungal man­age­ment in leu­ke­mia  |  367
IT TAKES A VILLAGE: MAXIMIZING SUPPORTIVE CARE AND MINIMIZING TOXICITY DURING CHILDHOOD LEUKEMIA THERAPY

Minimizing cardiac toxicity in children


with acute myeloid leukemia

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/368/1851685/368narayan.pdf by guest on 13 December 2021


Hari K. Narayan,1 Kelly D. Getz,2 and Kasey J. Leger3
1
Department of Pediatrics, University of California San Diego, La Jolla, CA; 2Departments of Biostatistics, Epidemiology & Informatics and Pediatrics,
Perelman School of Medicine, University of Pennsylvania; Division of Oncology, The Children’s Hospital of Philadelphia, Philadelphia, PA; and
3
Department of Pediatrics, University of Washington, Seattle Children’s Hospital, Seattle, WA

Anthracycline chemotherapy remains an integral component of modern pediatric acute myeloid leukemia (AML) regimens
and is often delivered at high doses to maximize cancer survival. Unfortunately, high-dose anthracyclines are associated with
a significant risk of cardiotoxicity, which may result in early and/or long-term left ventricular systolic dysfunction and heart
failure. Moreover, the development of cardiotoxicity during pediatric AML therapy is associated with lower event-free and
overall survival, which may be partially attributable to incomplete anthracycline delivery. A combined strategy of primary
cardioprotection and close cardiac monitoring can maximize chemotherapy delivery while reducing the toxicity of inten-
sive AML therapy. Primary cardioprotection using dexrazoxane reduces short-term cardiotoxicity without compromising
cancer survival. Liposomal anthracycline formulations, which are under active investigation, have the potential to mitigate
cardiotoxicity while also improving antitumor efficacy. Primary cardioprotective strategies may reduce but not eliminate
the risk of cardiotoxicity; therefore, close cardiac monitoring is also needed. Standard cardiac monitoring consists of serial
echocardiographic assessments for left ventricular ejection fraction decline. Global longitudinal strain has prognostic utility
in cancer therapy-related cardiotoxicity and may be used as an adjunct assessment. Additional cardioprotective measures
should be considered in response to significant cardiotoxicity; these include cardiac remodeling medications to support
cardiac recovery and anthracycline dose interruption and/or regimen modifications. However, the withholding of anthracy-
clines should be limited to avoid compromising cancer survival. A careful approach to cardioprotection during AML therapy
is critical to maximize the efficacy of leukemia treatment while minimizing the short- and long-term risks of cardiotoxicity.

LEARNING OBJECTIVES
• Recognize the impact of anthracycline-associated cardiotoxicity on cardiac and leukemia outcomes in the treat-
ment of pediatric acute myeloid leukemia
• Understand the role of primary cardioprotection during anthracycline-containing chemotherapy for pediatric
acute myeloid leukemia
• Understand the strengths and limitations of cardiac imaging modalities and the role of echocardiography during
pediatric acute myeloid leukemia therapy

cycles including 300 mg/m2 daunorubicin and 48 mg/m2


CLINICAL CASE mitoxantrone. On day 14 of intensification 1 (following
A 15-year-old girl was diagnosed with acute myeloid leu- 300 mg/m2 daunorubicin), she was admitted to the inten-
kemia (AML) after presenting with fatigue, menorrhagia, sive care unit with klebsiella septic shock. After pressor
and fever. Cytogenetic analyses demonstrated a t(9;11) initiation for fluid-refractory hypotension, echocardiogra-
KMT2A-MLLT3 fusion. The patient commenced induc- phy demonstrated a significant decline in left ventricular
tion therapy with cytarabine, daunorubicin with dexra- ejection fraction (LVEF) from 60% at baseline to 35%.
zoxane, and gemtuzumab ozogamicin. After induction,
there was no evidence of residual leukemia, and she was
stratified to low-risk AML treatment per the standard arm Introduction
of the Children’s Oncology Group (COG) trial AAML1831 While clinical outcomes of pediatric AML have improved
(NCT04293562) with planned receipt of 5 chemotherapy over time with intensification of therapy, the risk of leukemia

368 | Hematology 2021 | ASH Education Program


recur­rence and ther­apy-related mor­bid­ity and mor­tal­ity remains
high.1 Anthracyclines are crit­i­cal for opti­mal sur­vival,2 and deliv­
ered doses in AML sub­sets treated with­out consolidative trans­
plant may exceed 600 mg/m2 of doxo­ru­bi­cin equiv­a­lents after
account­ing for the recently rec­og­nized 10:1 cardiotoxic dose
equiv­a­lence of mitoxantrone in com­par­i­son with doxo­ru­bi­cin.3
As a result, anthracycline-asso­ci­ated cardiotoxicity is com­mon,
with late car­dio­my­op­a­thy rates of up to 27%.4 Early LV sys­tolic
dys­func­tion (LVSD) dur­ing AML ther­apy is also com­mon and has
impor­tant impli­ca­tions for car­dio­vas­cu­lar out­comes and sur­vival.
The inci­dence and impact of early LVSD were eval­u­ated in 2 re-
cent ran­dom­ized phase 3 COG tri­als in de novo pedi­at­ric AML.5,6
In AAML0531, 12% of chil­dren with non-FLT3-mutant AML devel­

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/368/1851685/368narayan.pdf by guest on 13 December 2021


oped grade 2 + LVSD dur­ing or fol­low­ing ther­apy (n =  1022; 5-
year fol­low-up).6 In AAML1031, with more com­pre­hen­sive ascer­
tain­ment due to man­dated site reporting of car­diac func­tional
param­e­ters, the rate of grade 2 + LVSD was 21% (n =  1014; median
3.5-year fol­low-up).5 In fur­ther anal­y­sis of AAML0531, those who
devel­oped LVSD on ther­apy were much more likely to have per­
sis­tent or recur­rent LVSD dur­ing off-pro­to­col fol­low-up (haz­ard
ratio [HR], 12.1; 95% CI, 4.2-34.8). Moreover, on-ther­apy grade
2 + LVSD was asso­ci­ated with a >15% reduc­tion in 5-year event-
free and over­all sur­vival (Figure 1).6 Given that this pro­to­col re-
quired dis­con­tin­u­a­tion of anthracyclines after the devel­op­ment
of LVSD, infe­ rior sur­ vival was likely driven in part by incom­
plete anthracycline deliv­ery com­pro­mis­ing che­mo­ther­a­peu­tic
effi­cacy. Thus, devel­op­ing an effec­tive strat­egy to reduce the
cardiotoxicity of AML ther­apy while maintaining leu­ke­mia-free
sur­vival is crit­i­cal. A com­bined approach of pri­mary cardiopro-
tection and close car­diac mon­i­tor­ing to guide tox­ic­ity-respon­
sive cardioprotective mea­sures dur­ing che­mo­ther­apy can help
bal­ance the some­what com­pet­ing goals of max­i­mal ther­apy
Figure 1. Event-free (A) and Overall (B) survival according to
deliv­ery and cardiotoxicity reduc­tion to opti­mize both leu­ke­mia
the occurrence of Grade 2 + LVSD, in the presence or absence
and car­dio­vas­cu­lar out­comes.
of associated bloodstream infection, on COG trial AAML0531.
Grade 2 + LVSD was defined as LVEF <50% or LVFS <24% based
Primary cardioprotection
on Common Terminology Criteria for Adverse Events V3.0.
Primary cardioprotection involves the con­cur­rent use of cardi-
Adapted with permission from Getz et al.6
oprotective med­ i­
ca­
tions dur­
ing anthracycline ther­ apy or the
use of less cardiotoxic agents to reduce cardiotoxicity risk. Two
impor­tant pri­mary cardioprotective strat­e­gies per­ti­nent to pedi­
at­ric AML ther­apy are dexrazoxane and lipo­so­mal anthracyclines. equiv­a­lence ratio to doxo­ru­bi­cin, wherein dexrazoxane is dosed
at 5-10:1 with dau­no­ru­bi­cin, 40:1 with mitoxantrone, and 50:1
Dexrazoxane with idarubicin.10 However, given evolv­ing dose equiv­a­lence ra-
Dexrazoxane is the only US Food and Drug Administration-ap- tios as new data sets emerge,3 dexrazoxane dos­ing should also
proved drug for pre­vent­ing anthracycline-induced cardiotoxici- take into con­sid­er­ation dem­on­strated phar­ma­co­ki­net­ics, safety,
ty. Its approval is restricted to met­a­static breast can­cer patients and effi­cacy. Studies of dexrazoxane in com­bi­na­tion with agents
receiv­ing >300 mg/m2 of doxo­ru­bi­cin based on data across mul­ com­monly used in AML (ie, dau­no­ru­bi­cin, mitoxantrone, idaru-
ti­ple ran­dom­ized phase 3 tri­als dem­on­strat­ing lower rates of car­ bicin) have shown safety and/or effi­cacy in dose ranges/ratios
diac events and/or heart fail­ure with con­cur­rent dexrazoxane.7 sim­i­lar to those pro­posed above.7,10
In 2014 dexrazoxane was des­ig­nated as an orphan drug for the Dexrazoxane has been shown to sig­nif­i­cantly reduce short-
“pre­ven­tion of car­dio­my­op­a­thy for chil­dren and ado­les­cents 0 term LVSD in anthracycline-treated chil­dren with acute lym­pho­
through 16 years of age treated with anthracyclines,” allowing blas­tic leu­ke­mia, non-Hodgkin lym­phoma, and Ewing sar­coma.11-13
for its use in pedi­at­ric AML. Dexrazoxane mit­i­gates anthracy- Evaluations of dexrazoxane use in pedi­at­ric AML, though prom­is­
cline-induced cardiomyocyte injury by stim­u­lat­ing the selec­tive ing, have his­tor­i­cally been lim­ited to small sin­gle-insti­tu­tion stud­
deg­ra­da­tion of topoisomerase IIβ, a molec­u­lar tar­get of anthra- ies.14,15 More recently, the short-term cardioprotective effects of
cyclines, thereby reduc­ing DNA dam­age and oxi­da­tive stress.8 dexrazoxane were ana­lyzed in the COG trial for de novo AML,
Dexrazoxane is admin­is­tered as a bolus infu­sion over 15 min­utes AAML1031.5 Dexrazoxane use was nonrandomized and deter­
imme­di­ately prior to anthracycline admin­is­tra­tion at a dose ratio mined by insti­ tu­tional prac­tice; 96 (9.5%) patients received
of 10:1 with doxo­ru­bi­cin.9 Dosing with alter­na­tive anthracycline dexrazoxane and 918 (90.5%) did not. The 4-year risk for grade
agents has his­tor­i­cally been based on their cardiotoxicity dose 2 + LVSD was 45% lower with dexrazoxane than with­out (26.5%
Prakash Singh Shekhawat
Minimizing cardiotoxicity in pedi­at­ric AML  |  369
Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/368/1851685/368narayan.pdf by guest on 13 December 2021
Figure 2. Temporal trends in LVEF by dexrazoxane exposure.
Standard errors of the mean LVEF measurements are presented Figure 3. Cumulative incidence of relapse and overall mortali-
as vertical bars. The (*) denotes treatment courses that include ty in the combined COG randomized trials of DRZ. Cumulative
an anthracycline. Adapted with permission from Getz et al.5 incidences at 10 years were not significantly different by DRZ
status for either outcome. DRZ, dexrazoxane; DRZ+, exposed to
DRZ; DRZ−, not exposed to DRZ. Adapted with permission from
Chow et al.17
vs 42.2%; HR =  0.55; 95% CI, 0.36-0.86; P =  .009), with greater
reduc­tions in risk for higher-grade cardiotoxicity. While dexra-
zoxane did not pro­vide com­plete pro­tec­tion against LVSD, its should be noted that dexrazoxane reduces but does not elim­
use was asso­ci­ated with smaller declines in LVEF (Figure 2), less i­nate short-term cardiotoxicity. In addi­tion, fur­ther research is
wors­en­ing over time, and a trend toward greater like­li­hood of needed to assess non­car­diac toxicities such as typhlitis rates,
LVSD res­o­lu­tion. Although label­ing warns that dexrazoxane may which were qual­i­ta­tively higher in Hodgkin lym­phoma patients
increase the myelosuppressive effects of che­mo­ther­apy, dura­ receiv­ing dexrazoxane in P9425 (8.4% vs 2.8%) and to deter­mine
tions of neutropenia, inten­sive care unit require­ments, and rates the long-term effects of dexrazoxane on car­dio­vas­cu­lar mor­bid­
of mucositis and blood­stream infec­tion did not dif­fer with dexra- ity and mor­tal­ity.18
zoxane expo­sure. Additionally, 5-year event-free (49.0% vs 45.1%;
P =  .534) and over­all sur­vival esti­ma­tes (65.0% vs 61.9%; P  =  .613) Liposomal anthracyclines
were com­pa­ra­ble between those who did and did not receive Liposomal deliv­ery sys­tems for anthracyclines are a prom­is­ing
dexrazoxane. However, sig­ nif­i­
cantly lower treat­ ment-related strat­egy to mit­i­gate cardiotoxicity while enhanc­ing anti­tu­mor
mor­tal­ity with dexrazoxane (4.6% vs 12.6%; P =  .024) sug­gests effi­cacy.19-21 Liposomal drug encap­ su­
la­
tion reduces its pen­ e­
that it may pre­vent acute car­diac events that con­trib­ute to early trance into the tight cap­il­lary junc­tions of the heart while allow-
patient deaths. ing sustained sys­temic cir­cu­la­tion and pref­er­en­tial accu­mu­la­tion
Despite com­pel­ling evi­dence of an early cardioprotective ben­ in the tumor.22 Meta-ana­ly­ses of adult tri­als com­par­ing lipo­so­mal
e­fit, <10% of AML patients receive dexrazoxane, likely due to early doxo­ru­bi­cin to free drug dem­on­strate a >50% reduc­tion in clin­
con­cerns for increased sec­ond­ary malig­nancy risk.5 This asso­ci­a­tion i­cal and sub­clin­i­cal car­diac dys­func­tion with lipo­so­mal for­mu­la­
was ini­tially suggested in an anal­y­sis of two Hodgkin lym­phoma tions.23,24 Intensification of che­mo­ther­apy with lipo­so­mal encap­
tri­als that included the ran­dom­ized admin­is­tra­tion of dexrazoxane su­lated dau­no­ru­bi­cin, DaunoXome® (DNX), has proven effec­tive
in the con­text of mod­er­ate-dose anthracycline, etoposide, and and tol­er­a­ble in the con­text of pedi­at­ric AML stud­ies conducted
alkylator ther­apy, P9425 (n =  216) and P9426 (n =  255).16 However, in by the International Berlin-Frankfurt-Münster Study Group. In
sub­se­quent ana­ly­ses of these tri­als and the COG T-cell acute lym­ the relapse set­ting, DNX plus fludarabine and cytarabine (FLAG)
pho­blas­tic leu­ke­mia/lym­phoma trial P9404 (n =  537) with lon­ger dem­on­strated enhanced leu­ke­mic effi­cacy and com­pa­ra­ble car-
fol­low-up (median, 12.6 years),17 dexrazoxane did not sig­nif­i­cantly diotoxicity com­pared to FLAG alone.25 Children with de novo
increase the 10-year relapse risk (16.1% with dexrazoxane vs 19.1% AML expe­ri­enced sim­i­lar rates of cardiotoxicity and less over­
with­out; HR =  0.81; 95% CI, 0.60-1.08), over­all mor­tal­ity (12.8% vs all treat­ment-related tox­ic­ity with DNX, given at a higher than
12.2%; HR =  1.03; 95% CI, 0.73-1.45), or deaths due to sec­ond­ary equiv­a­lent dose vs idarubicin, dur­ing induc­tion.26 DNX is no lon­
can­cers (2.0% vs 1.6%; HR =  1.24; 95% CI, 0.49-3.15; Figure 3). More- ger being manufactured and thus is not avail­­able for use in AML.
over, these find­ings remained sim­il­ar in long-term ana­ly­ses of the CPX-351 is a lipo­so­mal for­mu­la­tion of dau­no­ru­bi­cin plus cytar-
indi­vid­ual Hodgkin lym­phoma tri­als. Likewise, sec­ond­ary malig­ abine recently approved by the US Food and Drug Administration
nan­cies were rare over the fol­low-up on AAML1031 (n =  4), with only in adults and chil­dren with ther­apy-related AML or AML/myelo-
one occur­ring after dexrazoxane ther­apy.5 dysplastic syn­drome, given its favor­able effi­cacy and tol­er­a­ble
Overall, suf­fi­
cient data exist to sup­ port the use of dexra- safety pro­file.27,28 The cardioprotective effects of CPX-351 are not
zoxane in chil­dren with AML to mit­i­gate short-term LVSD and established; how­ever, clin­i­cal cardiotoxicity has been described
improve the poten­tial for tar­get anthracycline dose deliv­ery. It in early tri­als. A ran­dom­ized study of CPX-351 vs dau­no­ru­bi­cin

370  |  Hematology 2021  |  ASH Education Program


and cytarabine in 309 older adults with sec­ond­ary AML dem­on­ can­cer sur­vi­vor pop­u­la­tions.33,35 However, lim­i­ta­tions include the
strated sim­i­lar rates of cardiotoxicity despite nearly dou­ble the need for spe­cial­ized equip­ment, soft­ware, and train­ing and a
rate of postremission consolidative anthracycline deliv­ery on the pau­city of data supporting its use in youn­ger chil­dren with can­
CPX-351 arm.28 In 38 heavily anthracycline pretreated chil­dren cer. When avail­­able, 3D LVEF is the pre­ferred screen­ing method
with relapsed AML who received CPX-351 followed by FLAG on in ado­les­cents and young adults.32
the COG phase 1/2 study, grade 2 + LVSD occurred in 7 (18%).27 Although valid and impor­tant heart fail­ure screen­ing mea­
There are a pau­city of data to inform how this com­pares with sures, LVEF and LVFS declines may be late find­ings in the pro­
tra­di­tional anthracycline or nonanthracycline sal­vage reg­i­mens. gres­sion of anthracycline-related car­dio­my­op­a­thy.36 Strain,
CPX-351 is cur­rently being stud­ied in the up-front set­ting by COG spe­cif­i­cally GLS, has been pro­posed as an alter­na­tive mea­sure to
in a phase 3 ran­dom­ized study in de novo AML (NCT04293562). facil­i­tate ear­lier, more sen­si­tive cardiotoxicity detec­tion. Strain
If found to mit­i­gate cardiotoxicity while enhanc­ing or even main- is the frac­tional change in myo­car­dial fiber length, or defor­ma­
taining leu­ke­mia-free sur­vival, lipo­so­mal anthracyclines could tion, dur­ing the car­diac cycle. GLS rep­re­sents strain along the LV
sub­stan­tially improve ther­a­peu­tic options for AML. long axis, aver­aged across the LV. GLS declines pre­cede and are

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/368/1851685/368narayan.pdf by guest on 13 December 2021


prog­nos­tic of LVEF declines in adults dur­ing and after anthracy-
Cardiac mon­i­tor­ing and tox­ic­ity-respon­sive cline ther­apy.37 GLS abnor­mal­i­ties are also prev­a­lent in child­hood
cardioprotection can­cer sur­vi­vors in the pres­ence of nor­mal LVEF and are asso­
While pri­mary cardioprotection may reduce the risk for cardio- ci­ated with established car­dio­vas­cu­lar risk fac­tors, suggesting
toxicity, the above data dem­ on­strate that these approaches that they are clin­i­cally rel­e­vant.38 In recent years GLS has been
have not elim­i­nated car­diac risk. Thus, close monitoring of car­ incor­po­rated in adult cardio-oncol­ogy imag­ing guide­lines, and
diac func­tion dur­ing and after AML ther­apy remains crit­i­cal to its use is becom­ing more wide­spread.32 However, 1-year results
clin­i­cal man­age­ment and is used to guide tox­ic­ity-respon­sive from the only ran­dom­ized trial com­par­ing the effi­cacy of a GLS-
cardioprotective inter­ven­tions. vs LVEF-guided approach to cardioprotective ther­apy ini­ti­a­tion
in adults receiv­ing anthracyclines failed to dem­on­strate a ben­e­fit
Cardiac mon­i­tor­ing of GLS.39 In addi­tion, pedi­at­ric data attesting to the util­ity of GLS
Cardiac mon­i­tor­ing dur­ing pedi­at­ric AML ther­apy typ­i­cally con­ dur­ing anthracycline ther­apy are lim­ited. Based on the cur­rent
sists of serial precycle echo­ car­di­
og­ra­
phy to assess for LVSD, evi­dence, GLS may serve as an adjunct mea­sure, but AML treat­
manifested as declines in LV frac­tional short­en­ing (LVFS), LVEF, ment deci­sions should not be altered based on GLS alone.40
and/or global lon­gi­tu­di­nal strain (GLS). Each of these meth­ods
has strengths and lim­i­ta­tions that are rel­e­vant to clin­i­cal man­ Toxicity-respon­sive cardioprotection dur­ing che­mo­ther­apy
age­ment (Table 1). Cardiac mon­i­tor­ing pro­vi­des the oppor­tu­nity for addi­tional cardi-
LVFS is the frac­tional cav­ity diam­e­ter change between dias­ oprotective inter­ven­tions in response to cardiotoxicity detec­tion,
tole and sys­tole. It is assessed with two-dimen­sional (2D) or with the goal of reduc­ing fur­ther car­diac injury and supporting
M-mode echo­car­di­og­ra­phy via a lin­ear inter­ro­ga­tion through the car­diac recov­ery. The two main strat­e­gies of cardioprotection in
cen­ter of the LV. LVFS has a long his­tor­i­cal pre­ce­dent and is easy response to LVSD dur­ing che­mo­ther­apy include with­hold­ing an-
to acquire and mea­sure.29 However, it is sub­op­ti­mal in screen­ thracyclines (ide­ally tem­po­rar­ily) and using car­diac remodeling
ing given its sig­nif­i­cant angle depen­dence and poor reli­abil­ity med­i­ca­tions. There are lim­ited data to guide cli­ni­cians regard­ing
between acqui­si­tions.30 when to insti­tute these inter­ven­tions; acknowl­edg­ing this pau­
LVEF is the frac­tional cav­ity vol­ume change between dias­ city, our pro­posed man­age­ment strat­egy is depicted in Figure 4.
tole and sys­tole. Cardiac mag­netic res­o­nance imag­ing (CMRI) Dose inter­rup­tions or reduc­tions in anthracycline deliv­ery should
is the opti­mal modal­ity for LVEF der­i­va­tion given its high-res­ be min­i­mized given the poten­tial to reduce che­mo­ther­a­peu­tic
o­lu­tion, direct LV mea­sure­ment in 3 dimen­sions with­out geo­ effi­cacy. Brief delays may be con­sid­ered in the set­ting of LVSD,
met­ric assump­tions.31 However, CMRI is cur­rently reserved for with the goal of func­tional recov­ery and resump­tion of anthracy-
sec­ond­ary eval­u­a­tion, pri­mar­ily related to its lesser avail­abil­ity.32 cline deliv­ery. Withholding anthracycline should be con­sid­ered
Echocardiography, the stan­ dard pri­ mary screen­ ing modal­ ity, in cases of sig­nif­i­cant dys­func­tion; in these set­tings we sug­gest
can be used to derive LVEF using mul­ti­ple meth­ods. The Teich- replacing it with an inten­sive non-anthracycline-containing che­
holz for­mula can be used to cal­cu­late LVEF from lin­ear dimen­ mo­ther­apy block such as high-dose cytarabine and asparaginase
sions; how­ ever, this method is not recommended due to its (Capizzi II). The pres­ence of per­sis­tent and/or sig­nif­i­cant LVSD
sig­nif­i­cant geo­met­ric assump­tions and resulting inaccuracy.33,34 should prompt car­di­­ol­ogy con­sul­ta­tion, fur­ther car­diac test­ing,
Alternate 2D LVEF der­i­va­tions include the five/sixths area-length and con­sid­er­ation of car­diac remodeling med­i­ca­tions such as
(ie, “bul­let”) method and the biplane Simpson’s method; pref­er­ angio­ten­sin converting enzyme inhib­i­tor or beta-blocker ther­
ence among these is gen­er­ally lab­o­ra­tory depen­dent. Although apy; fur­ther pedi­at­ric data are needed to deter­mine the ben­
supe­rior to Teichholz, these meth­ods also have lim­i­ta­tions due e­fit of these med­i­ca­tions in this set­ting. Recovery of func­tion
to chal­lenges with angle depen­dence, mea­sure­ment repro­duc­ after these tox­ic­ity-respon­sive inter­ven­tions may allow for the
ibil­ity, and geo­met­ric assump­tions such that their sen­si­tiv­ity to resump­tion or con­tin­u­a­tion of anthracycline deliv­ery.
detect abnor­mal LVEF com­pared to CMRI in child­hood can­cer
sur­vi­vor pop­u­la­tions is only approx­i­ma­tely 50%.33,35 Summary
Three-dimen­sional (3D) echo­car­di­og­ra­phy allows com­plete The goals of care dur­ ing pedi­at­
ric AML ther­ apy are to max­ i­
LV visu­al­i­za­tion, yield­ing a more repro­duc­ible and accu­rate LVEF mize cure while min­i­miz­ing short- and long-term cardiotoxici-
mea­sure­ment in com­par­i­son with 2D meth­ods; the sen­si­tiv­ity to ty. To pro­mote the safety and effi­cacy of che­mo­ther­apy, treat­
detect LVSD in com­par­i­son with MRI is 53% to 68% in child­hood ment reg­i­mens should include pri­mary cardioprotection in the
Prakash Singh Shekhawat
Minimizing cardiotoxicity in pedi­at­ric AML  |  371
Table 1. Indices used to assess for LVSD

Threshold for sig­


Measure Strengths Limitations nif­i­cant LVSD Recommendation for use

LVFS

• Long his­tor­i­cal pre­ce­dent •  Highly angle depen­dent <24%a Adjunct mea­sure only except
• Easy to acquire and mea­sure • Poor reli­abil­ity (~7% error in the set­ting of inad­e­quate
when in the nor­mal range)30 win­dows for alter­nate mea­
sures

LVEF

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/368/1851685/368narayan.pdf by guest on 13 December 2021


Teichholz for­mula • Easy to acquire and mea­sure • Significant geo­met­ric <50%a Not recommended
(from lin­ear dimen­ assump­tions
sions) • Inaccurate

2D echo • Standard across most lab­o­ •  Geometric assump­tions May be used as pri­mary
(5/6 area length or ra­to­ries •  Measurement error screen­ing
biplane Simpson’s • Established util­ity in heart • Only ~50% sen­si­tiv­ity to
method) fail­ure detect LVSD in com­par­i­son
with CMRI (in sur­vi­vor data)

3D echo •  Better repro­duc­ibil­ity • Requires spe­cial­ized train­ing Preferred for pri­mary screen­
• 53%–68% sen­si­tiv­ity to and equip­ment ing in ado­les­cents and young
detect LVSD in com­par­i­son • Limited data in youn­ger adults, when avail­­able
with CMRI (in sur­vi­vor data) chil­dren

CMRI • Optimal LVEF repro­duc­ibil­ity • Requires spe­cial­ized train­ing Secondary eval­u­a­tion as indi­
and accu­racy and equip­ment catedb
• Can mea­sure other poten­ •  Less avail­­ableb
tially impor­tant car­diac • Compliance chal­lenges in
fea­tures (eg, pre­cise mea­ youn­ger chil­drenb
sure­ment of mass; edema, • An improve­ment in out­
fibro­sis) comes has not been dem­on­
strated in adult cardio-oncol­
ogy pop­u­la­tions
• Limited pedi­at­ric cardio-
oncol­ogy data

GLS

• More sen­si­tive mea­sure of • An improve­ment in out­ >−16% (approx­i­ Adjunct mea­sure only (fur­ther
car­diac func­tion comes has not been dem­on­ mate)42 or ≥15% study needed)
• Prognostic of sub­se­quent strated in adult cardio-oncol­ wors­en­ing from
LVEF declines37 ogy pop­u­la­tions39 base­line valuec
• Limited pedi­at­ric cardio-
oncol­ogy data

The table depicts the der­iv ­ a­tions of imag­ing-based mea­sures of left ven­tric­ul­ar sys­tolic func­tion. LVFS is the frac­tional change in cav­ity diam­e­ter dur­ing the car­diac cycle. LVEF
is the frac­tional change in cav­ity vol­ume dur­ing the car­diac cycle. Strain is the frac­tional change in myo­car­dial fiber length, or defor­ma­tion, dur­ing the car­diac cycle, typ­i­cally
derived from speckle-track­ing anal­y­sis of 2D echo­car­dio­graphic images. GLS is the aver­age defor­ma­tion in the lon­gi­tu­di­nal dimen­sion. 2D or 3D echo­car­dio­graphic der­i­va­tions
of LVEF should be the pri­mary screen­ing mea­sure used to assess for LVSD dur­ing pedi­at­ric AML ther­apy. Strengths, lim­it­ a­tions, and rec­om­men­da­tions for use are based on gen­
eral car­dio­vas­cu­lar-imag­ing quan­ti­fi­ca­tion guide­lines and adult cardio-oncol­ogy guide­lines, with addi­tional sup­port­ive ref­er­ences listed within the table and foot­notes.29,32,34,40
a
LVFS and LVEF cut­offs are based on grade 2 sever­ity events for the terms “left ven­tric­u­lar sys­tolic dys­func­tion” and “ejec­tion frac­tion decreased” in the National Cancer
Institute Common Terminology Criteria for Adverse Events (Versions 3.0 and 5.0, respec­tively). Precise cut­offs may vary among treat­ment pro­to­cols, and cli­ni­cians should
con­sult the spe­cific pro­to­col for fur­ther guid­ance.
b
Some indi­ca­tions for CMRI in sec­ond­ary eval­u­a­tion are poor echo­car­dio­graphic win­dows resulting in inad­e­quate assess­ment of func­tion, bor­der­line/inde­ter­mi­nate echo­
car­dio­graphic results in which a more pre­cise LVEF esti­mate would change clin­i­cal man­age­ment, or base­line or sig­nif­i­cant LVSD to eval­u­ate for under­ly­ing car­dio­my­op­a­
thy.31,32 CMRI does not employ ion­iz­ing radi­a­tion, and assess­ment of LV vol­umes, mass, and LVEF does not require intra­ve­nous con­trast. However, youn­ger chil­dren require
seda­tion/anes­the­sia to com­plete CMRI, which is an impor­tant con­sid­er­ation.43 Increasing avail­abil­ity cou­pled with ongo­ing tech­no­log­i­cal improve­ments that reduce motion
arti­facts/patient com­pli­ance demands, scan­ning time, and postprocessing time may result in increased CMRI use in pedi­at­ric can­cer pop­u­la­tions in the future.
c
GLS val­ues are typ­i­cally expressed as a neg­a­tive per­cent­age; less neg­a­tive val­ues reflect worse sys­tolic func­tion (eg, −16% is worse than −18%). The limit of nor­mal has not
been pre­cisely established in pedi­at­rics; the reported limit of −16% is an impre­cise esti­mate based on prior pedi­at­ric stud­ies.42 The wors­en­ing of 15% from the base­line value
is based on adult cardio-oncol­ogy guide­lines and refers to the per­cent wors­en­ing from the base­line value,32 not the abso­lute wors­en­ing in GLS (eg, a change from −20% to
−17% is a 15% wors­en­ing). LVEDD, left ven­tric­u­lar end-dia­stolic diam­e­ter; LVEDV, left ven­tric­u­lar end-dia­stolic vol­ume; LVESD, left ven­tric­u­lar end-sys­tolic diam­e­ter; LVESV,
left ven­tric­u­lar end-sys­tolic vol­ume.

372  |  Hematology 2021  |  ASH Education Program


Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/368/1851685/368narayan.pdf by guest on 13 December 2021
Figure 4. Proposed management of cardiotoxicity during pediatric AML therapy. This figure depicts our proposed algorithm to
manage cardiotoxicity during AML therapy, with the goal of balancing cardiac safety with effective delivery of AML-directed che-
motherapy. LVEF cutoffs are based on National Cancer Institute Common Terminology Criteria for Adverse Events (CTCAE) V3.0 for
LVSD: grade 2 LVSD is defined as asymptomatic LVEF decline of 40% to 49%. Grade 3 LVSD is defined as LVEF decline of 20% to 39%,
or symptomatic heart failure. These LVEF thresholds are consistent with grade 2/3 “ejection fraction decreased” in CTCAE V5.0.
*Additional testing may include biomarkers (eg, N-terminal-pro B-type natriuretic peptide), CMRI, or alternate testing as indicated.
Biomarkers currently have an established role in secondary/further evaluation but are under investigation as a primary screening
modality. ^Cardiac remodeling medications may include angiotensin converting enzyme inhibitors (eg, enalapril) and beta-blockers
(eg, carvedilol). #When omission of anthracyclines is necessary due to persistent LVSD, efforts should be made to maintain the inten-
sity of chemotherapy, if appropriate. Thus, we suggest replacing with an intensive non-anthracycline-containing chemotherapy block,
such as high-dose cytarabine and asparaginase (Capizzi II).

form of con­cur­rent dexrazoxane and car­diac mon­i­tor­ing with Acknowledgments


addi­tional tox­ic­ity-respon­sive cardioprotective inter­ven­tions as Hari K. Narayan is supported by Padres Pedal the Cause/RADY
indi­cated. Given the del­i­cate bal­ance needed to man­age com­ no. #PTC2020 and UC San Diego Moore Cancer Center, Special-
pet­ing onco­logic and car­dio­vas­cu­lar risks, a close col­lab­o­ra­tion ized Cancer Control Support Grant NIH/NCI P30CA023100. Kelly
between oncol­o­gists and car­di­­ol­o­gists is crit­i­cal to opti­miz­ing Getz is supported by an National Heart, Lung, and Blood Insti-
patient care. This col­lab­o­ra­tion should begin dur­ing che­mo­ther­ tute Career Development Award (1K01HL143153-01).
apy and con­tinue in sur­vi­vor­ship, with long-term mon­i­tor­ing and
the poten­ tial implementation of sec­ ond­ary and ter­ tiary heart Conflict-of-inter­est dis­clo­sure
fail­ure pre­ven­tion mea­sures.41 As onco­logic ther­a­pies con­tinue Hari K. Narayan: nothing to disclose.
to rap­idly evolve, the devel­op­ment of ded­i­cated, inter­dis­ci­plin­ Kelly Getz: nothing to disclose.
ary pedi­at­ric cardio-oncol­ogy pro­grams may serve to meet this Kasey J. Leger: research funding: Abbott Diagnostics; advi­sory
impor­tant need. board: Jazz Pharmaceuticals.

Off-label drug use


Hari K. Narayan: Off-label use of several drugs is discussed:
angiotensin converting enzyme inhibitors (ex. enalapril); beta
CLINICAL CASE (Con­t in­u ed) blockers (ex. Carvedilol); asparaginase dexrazoxane (given under
The patient was diag­nosed with grade 3 LVSD in the set­ting FDA orphan drug designation for pediatric cancers, but not tech-
of sep­sis. With inten­sive sup­port­ive care, the patient recov­ nically a labeled indication).
ered and was discharged on cycle day 34. LVEF improved to Kelly Getz: Off-label use of several drugs is discussed: angioten-
48% on echo­car­di­og­ra­phy at dis­charge, with fur­ther recov­ery sin converting enzyme inhibitors (ex. enalapril); beta blockers
to 55% 1.5 weeks later. The patient was admit­ted for inten­si­ (ex. Carvedilol); asparaginase dexrazoxane (given under FDA or-
fi­ca­tion 2, includ­ing full-dose mitoxantrone with dexrazoxane, phan drug designation for pediatric cancers, but not technically
and her che­mo­ther­apy course was com­pleted with­out fur­ther a labeled indication).
com­pli­ca­tions. The patient is cur­rently 1 year fol­low­ing ther­apy Kasey J. Leger: Off-label use of several drugs is discussed: angio-
and is being mon­i­tored with annual echo­car­dio­grams due to tensin converting enzyme inhibitors (ex. enalapril); beta blockers
high-dose anthracycline expo­sure and a his­tory of on-ther­apy (ex. Carvedilol); asparaginase dexrazoxane (given under FDA or-
LVSD. phan drug designation for pediatric cancers, but not technically
a labeled indication).
Prakash Singh Shekhawat
Minimizing cardiotoxicity in pedi­at­ric AML  |  373
Correspondence bi­cin (DaunoXome) in pae­di­at­ric patients with relapsed or resis­tant solid
Kasey J. Leger, Department of Hematology/Oncology, Cancer tumours. Br J Cancer. 2006;95(5):571-580.
21. Bellott R, Auvrignon A, Leblanc T, et  al. Pharmacokinetics of lipo­so­mal
and Blood Disorders Center, Seattle Children’s Hospital, 4800 dau­ no­ru­bi­
cin (DaunoXome) dur­ ing a phase I-II study in chil­ dren with
Sand Point Way NE, M/S MB8.501, Seattle, WA 98105; e-mail: relapsed acute lym­pho­blas­tic leu­kae­mia. Cancer Chemother Pharmacol.
kasey​­.leger@seattlechildrens​­.org. 2001;47(1):15-21.
22. Waterhouse DN, Tardi PG, Mayer LD, Bally MB. A com­par­i­son of lipo­so­mal
for­mu­la­tions of doxo­ru­bi­cin with drug admin­is­tered in free form: chang­ing
References tox­ic­ity pro­files. Drug Saf. 2001;24(12):903-920.
1. Aplenc R, Meshinchi S, Sung L, et al. Bortezomib with stan­dard che­mo­ther­ 23. Rafiyath SM, Rasul M, Lee B, Wei G, Lamba G, Liu D. Comparison of safety
apy for chil­dren with acute mye­loid leu­ke­mia does not improve treat­ment and tox­ic­ity of lipo­so­mal doxo­ru­bi­cin vs. con­ven­tional anthracyclines: a
out­comes: a report from the Children’s Oncology Group. Haematologica. meta-anal­y­sis. Exp Hematol Oncol. 2012;1(1):10.
2020;105(7):1879-1886. 24. van Dalen EC, Michiels EM, Caron HN, Kremer LC. Different anthracycline
2. Fernandez HF, Sun Z, Yao X, et  al. Anthracycline dose inten­si­fi­ca­tion in der­i­vates for reduc­ing cardiotoxicity in can­cer patients. Cochrane Data-
acute mye­loid leu­ke­mia. N Engl J Med. 2009;361(13):1249-1259. base Syst Rev. 2010;2010(5):CD005006.
3. Feijen EAM, Leisenring WM, Stratton KL, et al. Derivation of anthracycline 25. Kaspers GJ, Zimmermann M, Reinhardt D, et al. Improved out­come in pedi­

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/368/1851685/368narayan.pdf by guest on 13 December 2021


and anthra­qui­none equiv­al­ence ratios to doxo­ru­bi­cin for late-onset cardio- at­ric relapsed acute mye­loid leu­ke­mia: results of a ran­dom­ized trial on lipo­
toxicity. JAMA Oncol. 2019;5(6):864-871. so­mal dau­no­ru­bi­cin by the International BFM Study Group. J Clin Oncol.
4. Barlogis V, Auquier P, Bertrand Y, et al. Late car­dio­my­op­a­thy in child­hood 2013;31(5):599-607.
acute mye­loid leu­ke­mia sur­vi­vors: a study from the L.E.A. pro­gram. Hae- 26. Creutzig U, Zimmermann M, Bourquin JP, et al. Randomized trial com­par­
matologica. 2015;100(5):e186-e189. ing lipo­so­mal dau­no­ru­bi­cin with idarubicin as induc­tion for pedi­at­ric acute
5. Getz KD, Sung L, Alonzo TA, et al. Effect of dexrazoxane on left ven­tric­u­ mye­loid leu­ke­mia: results from Study AML-BFM 2004. Blood. 2013;122(1):
lar sys­tolic func­tion and treat­ment out­comes in patients with acute mye­ 37-43.
loid leu­ke­mia: a report from the Children’s Oncology Group. J Clin Oncol. 27. Lancet JE, Uy GL, Cortes JE, et al. CPX-351 (cytarabine and dau­no­ru­bi­cin)
2020;38(21):2398-2406. lipo­some for injec­tion ver­sus con­ven­tional cytarabine plus dau­no­ru­bi­cin in
6. Getz KD, Sung L, Ky B, et al. Occurrence of treat­ment-related cardiotoxic- older patients with newly diag­nosed sec­ond­ary acute mye­loid leu­ke­mia.
ity and its impact on out­comes among chil­dren treated in the AAML0531 J Clin Oncol. 2018;36(26):2684-2692.
clin­i­cal trial: a report from the Children’s Oncology Group. J Clin Oncol. 28. Cooper TM, Absalon MJ, Alonzo TA, et al. Phase I/II study of CPX-351 fol-
2019;37(1):12-21. lowed by fludarabine, cytarabine, and granulocyte-col­ony stim­u­lat­ing fac­
7. Cvetković RS, Scott LJ. Dexrazoxane: a review of its use for cardioprotec- tor for chil­dren with relapsed acute mye­loid leu­ke­mia: a report from the
tion dur­ing anthracycline che­mo­ther­apy. Drugs. 2005;65(7):1005-1024. chil­dren’s oncol­ogy group. J Clin Oncol. 2020;38(19):2170-2177.
8. Lyu YL, Kerrigan JE, Lin CP, et al. Topoisomerase IIβ-medi­ated DNA dou­ble- 29. Lopez L, Colan SD, Frommelt PC, et al. Recommendations for quan­ti­fi­ca­tion
strand breaks: impli­ca­tions in doxo­ru­bi­cin cardiotoxicity and pre­ven­tion by meth­ods dur­ing the per­for­mance of a pedi­at­ric echo­car­dio­gram: a report
dexrazoxane. Cancer Res. 2007;67(18):8839-8846. from the Pediatric Measurements Writing Group of the Amer­i­can Society
9. Schuchter LM, Hensley ML, Meropol NJ, Winer EP; Amer­i­can Society of Clin- of Echocardiography Pediatric and Congenital Heart Disease Council.
ical Oncology Chemotherapy and Radiotherapy Expert Panel. 2002 update J Am Soc Echocardiogr. 2010;23(5):465-495, quiz, 576-577.
of rec­om­men­da­tions for the use of che­mo­ther­apy and radio­ther­apy pro­tec­ 30. Selamet Tierney ES, Hollenbeck-Pringle D, Lee CK, et al; Pediatric Heart
tants: clin­i­cal prac­tice guide­lines of the Amer­i­can Society of Clinical Oncol- Network Investigators. Reproducibility of left ven­tric­u­lar dimen­sion ver­sus
ogy. J Clin Oncol. 2002;20(12):2895-2903. area ver­sus vol­ume mea­sure­ments in pedi­at­ric patients with dilated car­
10. Vachhani P, Shin S, Baron J, Thompson JE, Wetzler M, Griffiths EA, Ontiveros dio­my­op­a­thy. Circ Cardiovasc Imaging. 2017;10(11):e006007.
EP, Spangenthal EJ, Wang ES. Dexrazoxane for cardioprotection in older 31. Leiner T, Bogaert J, Friedrich MG, et al. SCMR Position Paper (2020) on clin­
adults with acute myeloid leukemia. Leuk Res Rep. 2017 Apr 14;7:36-39. i­cal indi­ca­tions for car­dio­vas­cu­lar mag­netic res­o­nance. J Cardiovasc Magn
11. Harake D, Franco VI, Henkel JM, Miller TL, Lipshultz SE. Cardiotoxicity in Reson. 2020;22(1):76.
child­hood can­cer sur­vi­vors: strat­e­gies for pre­ven­tion and man­age­ment. 32. Plana JC, Galderisi M, Barac A, et  al. Expert con­sen­sus for multimodality
Future Cardiol. 2012;8(4):647-670. imag­ing eval­ua ­ ­tion of adult patients dur­ing and after can­cer ther­apy: a
12. Lipshultz SE, Sambatakos P, Maguire M, et al. Cardiotoxicity and cardiopro- report from the Amer­ i­
can Society of Echocardiography and the Euro­
tection in child­hood can­cer. Acta Haematol. 2014;132(3-4):391-399. pean Association of Cardiovascular Imaging. J Am Soc Echocardiogr.
13. Wexler LH. Ameliorating anthracycline cardiotoxicity in children with cancer: 2014;27(9):911-939.
clinical trials with dexrazoxane. Semin Oncol. 1998 Aug;25(4 Suppl 10):86-92. 33. Armstrong GT, Plana JC, Zhang N, et al. Screening adult sur­vi­vors of child­
14. Sánchez-Medina J, Gonzalez-Ramella O, Gallegos-Castorena S. The effect hood can­cer for car­dio­my­op­a­thy: com­par­i­son of echo­car­di­og­ra­phy and
of dexrazoxane for clin­i­cal and sub­clin­i­cal cardiotoxicity in chil­dren with car­diac mag­netic res­o­nance imag­ing. J Clin Oncol. 2012;30(23):2876-2884.
acute mye­loid leu­ke­mia. J Pediatr Hematol Oncol. 2010;32(4):294-297. 34. Lang RM, Badano LP, Mor-Avi V, et al. Recommendations for car­diac cham­
15. Schloemer NJ, Brickler M, Hoffmann R, et al. Administration of dexrazoxane ber quan­ti­fi­ca­tion by echo­car­di­og­ra­phy in adults: an update from the
improves car­diac indi­ces in chil­dren and young adults with acute mye­loid Amer­i­can Society of Echocardiography and the Euro­pean Association of
leu­ke­mia (AML) while maintaining sur­vival out­comes. J Pediatr Hematol Cardiovascular Imaging. J Am Soc Echocardiogr. 2015;28(1):1-39.e14.
Oncol. 2017;39(5):e254–e258. 35. Toro-Salazar OH, Ferranti J, Lorenzoni R, et  al. Feasibility of echo­car­dio­
16. Tebbi CK, London WB, Friedman D, et al. Dexrazoxane-asso­ci­ated risk for graphic tech­niques to detect sub­clin­i­cal can­cer ther­a­peu­tics-related car­
acute mye­loid leu­ke­mia/myelodysplastic syn­drome and other sec­ond­ary diac dys­func­tion among high-dose patients when com­pared with car­diac
malig­nan­cies in pedi­at­ric Hodgkin’s dis­ease. J Clin Oncol. 2007;25(5):493– mag­netic res­o­nance imag­ing. J Am Soc Echocardiogr. 2016;29(2):119-131.
500. 36. Lipshultz SE, Lipsitz SR, Sallan SE, et al. Long-term enalapril ther­apy for left
17. Chow EJ, Asselin BL, Schwartz CL, et al. Late mor­tal­ity after dexrazoxane ven­tric­u­lar dys­func­tion in doxo­ru­bi­cin-treated sur­vi­vors of child­hood can­
treat­ ment: a report from the chil­ dren’s oncol­
ogy group. J Clin Oncol. cer. J Clin Oncol. 2002;20(23):4517-4522.
2015;33(24):2639-2645. 37. Oikonomou EK, Kokkinidis DG, Kampaktsis PN, et al. Assessment of prog­
18. Schwartz CL, Constine LS, Villaluna D, et  al. A risk-adapted, response- nos­tic value of left ven­tric­u­lar global lon­gi­tu­di­nal strain for early pre­dic­tion
based approach using ABVE-PC for chil­dren and ado­les­cents with inter­ of che­mo­ther­apy-induced cardiotoxicity: a sys­tem­atic review and meta-
me­di­ate- and high-risk Hodgkin lym­phoma: the results of P9425. Blood. anal­y­sis. JAMA Cardiol. 2019;4(10):1007-1018.
2009;114(10):2051-2059. 38. Armstrong GT, Joshi VM, Ness KK, et al. Comprehensive echo­car­dio­graphic
19. Forssen EA, Coulter DM, Proffitt RT. Selective in vivo local­i­za­tion of dau­no­ru­ detec­tion of treat­ment-related car­diac dys­func­tion in adult sur­vi­vors of
bi­cin small unilamellar ves­ic­ les in solid tumors. Cancer Res. 1992;52(12):3255– child­hood can­cer: results from the St. Jude Lifetime Cohort Study. J Am
3261. Coll Cardiol. 2015;65(23):2511-2522.
20. Lowis S, Lewis I, Elsworth A, et al; United Kingdom Children’s Cancer 39. Thavendiranathan P, Negishi T, Somerset E, et al; SUCCOUR Investigators.
Study Group New Agents; Société Française d’Oncologie Pédiatrique Strain-guided man­age­ment of poten­tially cardiotoxic can­cer ther­apy. J Am
Pharmacology Group. A phase I study of intra­ve­nous lipo­so­mal dau­no­ru­ Coll Cardiol. 2021;77(4):392-401.

374  |  Hematology 2021  |  ASH Education Program


40. Zamorano JL, Lancellotti P, Rodriguez Muñoz D, et  al. 2016 ESC Position 43. Fratz S, Chung T, Greil GF, et al. Guidelines and pro­to­cols for car­dio­vas­cu­lar
Paper on can­cer treat­ments and car­dio­vas­cu­lar tox­ic­ity devel­oped under mag­netic res­o­nance in chil­dren and adults with con­gen­i­tal heart dis­ease:
the aus­pices of the ESC Committee for Practice Guidelines: the Task Force SCMR expert con­sen­sus group on con­gen­i­tal heart dis­ease. J Cardiovasc
for can­cer treat­ments and car­dio­vas­cu­lar tox­ic­ity of the Euro­pean Society Magn Reson. 2013;15(1):51.
of Cardiology (ESC). Eur Heart J. 2016;37(36):2768-2801.
41. Arme­ nian SH, Gelehrter SK, Chow EJ. Strategies to pre­ vent anthracy-
cline-related con­ges­tive heart fail­ure in sur­vi­vors of child­hood can­cer. Car-
diol Res Pract. 2012;2012(1):713294.
42. Levy PT, Machefsky A, Sanchez AA, et al. Reference ranges of left ven­tric­u­lar
strain mea­sures by two-dimen­sional speckle-track­ing echo­car­di­og­ra­phy in
chil­dren: a sys­tem­atic review and meta-anal­y­sis. J Am Soc Echocardiogr. © 2021 by The Amer­i­can Society of Hematology
2016;29(3):209-225.e6. DOI 10.1182/hema­tol­ogy.2021000268

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/368/1851685/368narayan.pdf by guest on 13 December 2021

Prakash Singh Shekhawat


Minimizing cardiotoxicity in pedi­at­ric AML  |  375
IT TAKES A VILLAGE: MAXIMIZING SUPPORTIVE CARE AND MINIMIZING TOXICITY DURING CHILDHOOD LEUKEMIA THERAPY

Managing therapy-associated neurotoxicity


in children with ALL

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/376/1851679/376bhojwani.pdf by guest on 13 December 2021


Deepa Bhojwani,1 Ravi Bansal,2 and Alan S. Wayne1
Division of Hematology-Oncology, Children’s Hospital Los Angeles, Norris Comprehensive Cancer Center, Keck School of Medicine,
1

University of Southern California, Los Angeles, CA; and 2Division of Research on Children, Youth and Families, Children’s Hospital Los Angeles, Keck
School of Medicine, University of Southern California, Los Angeles, CA

Several chemotherapeutic agents and novel immunotherapies provide excellent control of systemic and central nervous
system (CNS) leukemia but can be highly neurotoxic. The manifestations of subacute methotrexate neurotoxicity are
diverse and require vigilant management; nonetheless, symptoms are transient in almost all patients. As methotrexate is
a crucial drug to prevent CNS relapse, it is important to aim to resume it after full neurologic recovery. Most children tol-
erate methotrexate rechallenge without significant delays or prophylactic medications. Neurotoxicity is more frequent
with newer immunotherapies such as CD19– chimeric antigen receptor T (CAR T) cells and blinatumomab. A uniform
grading system for immune effector cell–associated neurotoxicity syndrome (ICANS) and algorithms for management
based on severity have been developed. Low-grade ICANS usually resolves within a few days with supportive measures,
but severe ICANS requires multispecialty care in the intensive care unit for life-threatening seizures and cerebral edema.
Pharmacologic interventions include anticonvulsants for seizure control and glucocorticoids to reduce neuroinflamma-
tion. Anticytokine therapies targeted to the pathophysiology of ICANS are in development. By using illustrative patient
cases, we discuss the management of neurotoxicity from methotrexate, CAR T cells, and blinatumomab in this review.

LEARNING OBJECTIVES
• Discuss management of methotrexate neurotoxicity and rechallenge strategy
• Review assessment and management of ICANS associated with CAR T-cell therapy and blinatumomab

Introduction predictors and mediators of immune effector cell-associated


Optimization of chemotherapeutic regimens has enabled neurotoxicity syndrome (ICANS), with the goal of identify-
substitution of cranial irradiation with systemic and intrathe- ing those at high risk and developing interventions to miti-
cal agents for prophylaxis of central nervous system (CNS) gate its severe manifestations. In this review, we discuss the
disease in childhood acute lymphoblastic leukemia (ALL).1 management of neurotoxicity associated with methotrexate,
However, even radiation-free contemporary regimens can CAR T-cell therapy, and blinatumomab in children with ALL.
cause severe neurotoxicity, leading to morbidity and mor-
tality, therapy delays, and fear in patients and families. The
incidence of symptomatic neurotoxicity during frontline ALL
therapy is approximately 10% to 12%, and its manifestations CLINICAL CASE 1
are varied, as are the inciting agents.2,3 Methotrexate-related A 14-year-old Hispanic girl with high-risk B-ALL without CNS
stroke-like syndrome (SLS), posterior reversible encepha- involvement was receiving standard-of-care chemother-
lopathy syndrome, cerebral sinus venous thrombosis, and apy according to the Children’s Oncology Group proto-
seizures are the common CNS events that result from stan- col AALL0232. She had tolerated chemotherapy relatively
dard ALL chemotherapeutic agents.4 With increasing use well, including 6 doses of intrathecal methotrexate in the
of immunotherapies for patients with ALL, neurotoxicity is induction and consolidation phases. The first interim main-
being reported more frequently, with incidence of up to tenance phase was initiated with 15mg of intrathecal meth-
50% in patients treated with CD19- chimeric antigen recep- otrexate followed by 5g/m2 of intravenous methotrexate
tor T (CAR T)-cell therapy.5 Multiple studies are investigating over 24 hours. Methotrexate cleared appropriately, and she

376 | Hematology 2021 | ASH Education Program


21 days of intra­ve­nous or intra­the­cal meth­o­trex­ate with spec­i­fied
clin­i­cal and radio­graphic fea­tures, while exclud­ing other iden­ti­fi­
able causes. Risk fac­tors for meth­o­trex­ate neu­ro­tox­ic­ity ­iden­ti­fied
in mul­ti­var­i­able ana­ly­ses are age >10 years, His­panic eth­nic­ity,
and ele­vated serum aspar­tate trans­am­i­nase dur­ing induc­tion/
con­sol­i­da­tion.8,11 Coadministration of intra­the­cal meth­o­trex­ate in
sys­temic cytarabine- and cyclo­phos­pha­mide-containing treat­
ment blocks has been noted as a period of increased risk.8,9
Leukoencephalopathy or sub­cor­ti­cal white mat­ter changes
are the char­ac­ter­is­tic MRI find­ings of meth­o­trex­ate neu­ro­tox­
ic­
ity. Diffusion-weighted imag­ ing (DWI) is the most sen­ si­ tive
modal­ity dur­ing the acute phase and shows abnor­mally high
dif­fu­sion of water in brain areas that cor­re­spond with clin­i­cally

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/376/1851679/376bhojwani.pdf by guest on 13 December 2021


appar­ent neu­ro­logic def­i­cits.12 Because changes noted on DWI
can be tran­sient, a neg­a­tive MRI does not exclude a diag­no­sis
of meth­o­trex­ate neu­ro­tox­ic­ity. On the other hand, white mat­ter
hyperintensities in T2-weighted and fluid-attenuated inversion
recovery MRIs can per­sist and evolve over time; there­fore, await-
ing com­plete nor­mal­i­za­tion of MRI find­ings prior to rechallenge
with meth­o­trex­ate is not prac­ti­cal.
The path­o­phys­i­­ol­ogy of sub­acute meth­o­trex­ate neu­ro­tox­ic­
Figure 1. Diffusion-weighted MRI of patient 1 four hours after ity is not com­pletely under­stood. Accumulation of homocyste-
onset of symptoms. DWI demonstrated abnormal diffusion in bi- ine and its metab­o­lites in plasma and cere­bro­spi­nal fluid (CSF) is
lateral centrum semiovale (red arrows), greater on the left side. toxic to vas­cu­lar endo­the­lium. Release of aden­o­sine and neu­ro­
These findings indicate increased diffusion of water in the affect- nal injury are also pro­posed mech­a­nisms.13
ed regions suggestive of cytotoxic edema.
Acute man­age­ment of meth­o­trex­ate neu­ro­tox­ic­ity
Supportive care and close mon­i­tor­ing are the main­stays of man­
received 3 doses of leucovorin 15 mg/m2 at hours 42, 48, and 54. age­ment dur­ing the acute epi­sode. A sin­gle, brief sei­zure will
Nine days later, she presented to the emer­gency depart­ment not require phar­ma­co­logic inter­ven­tion, but anti­con­vul­sants are
with acute-onset right-sided leg weak­ness, which progressed to admin­is­tered for prolonged or repeated sei­zures. Symptoms can
the right arm and face over a 2-hour period. In addi­tion to right wax and wane; there­fore, fre­quent neu­ro­logic exam­i­na­tions
paraparesis and facial droop, she was noted to have apha­sia and guide sub­se­quent man­age­ment. Oral feeds should be held until
dys­ar­thria. Magnetic res­o­nance imag­ing (MRI) performed 4 hours swallowing is deemed safe. Because apha­sia and dys­ar­thria are
after the onset of symp­toms showed restricted dif­fu­sion bilat­er­ distressing to patients and fam­i­lies, psy­cho­so­cial sup­port, reas­
ally in the cen­trum semiovale, greater on the left side (Figure 1). sur­ance, and pro­vid­ing alter­nate meth­ods of com­mu­ni­ca­tion may
She was admit­ted to the inten­sive care unit for close obser­va­ lessen anx­i­ety. Computerized tomog­ra­phy (CT) is not gen­er­ally
tion. During the next 24 hours, her speech wors­ened, and she help­ful but may be needed if symp­toms are sug­ges­tive of CNS
became more con­fused. There was mod­er­ate improve­ment in bleed. CSF exam­i­na­tion is only indi­cated if the patient is febrile
right-sided weak­ness, but she devel­oped weak­ness in the left arm and there are con­cerns for CNS infec­tion. As detailed above, MRI
and leg. With con­tin­ued sup­port­ive care, begin­ning day 3 of hos­ with DWI can sup­port the diag­no­sis of meth­o­trex­ate neu­ro­tox­
pi­tal­i­za­tion, her symp­toms grad­u­ally improved, facial asymmetry ic­ity, but its util­ity in cases of mild, tran­sient, and clas­si­cal symp­
resolved, her speech became clearer, and she was a ­ ble to swal­ toms is lim­ited. Input from a neu­rol­o­gist is valu­able in most cases,
low. She required phys­i­cal ther­apy for 2 more days to regain her par­tic­u­larly with prolonged symp­toms. Severe SLS is best man­
base­line strength and ambu­late inde­pen­dently. aged in a crit­i­cal care set­ting, as patients may require intu­ba­tion
with mechan­i­cal ven­ti­la­tion for air­way pro­tec­tion.
Based on the pre­sumed mech­a­nisms of meth­o­trex­ate neu­ro­
Subacute meth­o­trex­ate neu­ro­tox­ic­ity tox­ic­ity and evi­dence of homocysteine caus­ing excit­atory effects
Approximately 3% to 7% of chil­dren treated with con­tem­po­rary on N-methyl-D-aspar­tate recep­tors, the use of the N-methyl-D-
front­line ALL reg­i­mens develop sub­acute meth­o­trex­ate neu­ro­ aspar­ tate antag­ o­nist dextromethorphan has been reported in
tox­ic­ity.6-8 Seizures, enceph­a­lop­a­thy, apha­sia, and SLS are com­ the acute set­ting and for sec­ond­ary pro­phy­laxis.14 To com­bat
mon pre­sen­ta­tions.7-9 Symptoms typ­i­cally begin 3 to 11 days after vaso­di­la­ta­tion from aden­o­sine, ami­noph­yl­line use has also been
intra­the­cal or intra­ve­nous meth­o­trex­ate, can wax and wane over described, as it dis­places aden­o­sine from its recep­tor on endo­
sev­eral hours or days, and are char­ac­ter­is­ti­cally tran­sient and the­lial cells.15 However, supporting such approaches is dif­fi­cult
resolve in 1 to 4 days. Most patients make a full recov­ery, although due to a lack of con­trols, par­tic­u­larly in the set­ting of the tran­sient
recov­ery may require sev­eral months of inten­sive reha­bil­i­ta­tion in nat­u­ral course of meth­o­trex­ate neu­ro­tox­ic­ity and rapid res­o­lu­
a small sub­set of patients with severe SLS.9 The ­defin­ing char­ tion of symp­toms with­out phar­ma­ceu­ti­cal inter­ven­tion in most
ac­ter­is­tics of meth­o­trex­ate-SLS are listed in Table 1.10 In brief, patients. In addi­ tion, dextromethorphan itself can cause CNS
­meth­o­trex­ate-SLS is defined as neu­ro­tox­ic­ity that occurs within effects such as diz­zi­ness and rest­less­ness, whereas ami­noph­yl­line

Prakash Singh Shekhawat


Neurotoxicity in chil­dren with ALL  |  377
Table 1. Defining char­ac­ter­is­tics of meth­o­trex­ate-SLS (reproduced with per­mis­sion10)

Neurotoxicity occur­ring within 21 days of intra­ve­nous or intra­the­cal meth­o­trex­ate with 3 char­ac­ter­is­tics that all­need to be fulfilled:
  1. New onset of one or more of pare­sis or paral­y­sis; move­ment dis­or­der or bilat­eral weak­ness; apha­sia or dys­ar­thria; altered men­tal sta­tus includ­ing
con­scious­ness (e.g., som­no­lence, con­fu­sion, dis­ori­en­ta­tion, and emo­tional labil­ity); and/or sei­zures with at least one of the other symp­toms.
  2. Either char­ac­ter­is­tic, but often tran­sient, white mat­ter changes indi­cat­ing leukoencephalopathy on MRI or a char­ac­ter­is­tic clin­i­cal course with
wax­ing and wan­ing symp­toms usu­ally lead­ing to com­plete (some­times par­tial) res­o­lu­tion within a week.
  3.  No other iden­ti­fi­able cause.
Characteristic oval-shaped lesions of the sub­cor­ti­cal white mat­ter (mostly fron­tal or pari­e­tal) on MRI are best seen on dif­fu­sion-weighted (hyper­in­
tense) or appar­ent dif­fu­sion coef­fi­cient (hypointense) images. Can be graded 1-5 according to CTCAEv4.03 for enceph­a­lop­a­thy.
This con­sen­sus def­i­ni­tion was devel­oped by the Ponte de Legno inter­na­tional child­hood ALL group to assist cli­ni­cians in dif­fer­en­ti­at­ing meth­o­trex­ate
-SLS from sim­i­lar neurotoxicities (eg, pos­te­rior revers­ible enceph­a­lop­a­thy syn­drome) and to enable reli­able com­par­i­sons of inci­dence of meth­o­trex­ate
-SLS between dif­fer­ent tri­als.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/376/1851679/376bhojwani.pdf by guest on 13 December 2021


can cause tachy­car­dia, anx­i­ety, head­ache, and sei­zures.16 Current
gaps in knowl­edge regard­ing the util­ity of dextromethorphan CLINICAL CASE 1 (Con­tin­ued)
and ami­noph­yl­line for the treat­ment and pro­phy­laxis of meth­o­ This patient made a full recov­ery 5 days after the onset of symp­
trex­ate neu­ro­tox­ic­ity pre­clude spe­cific rec­om­men­da­tions. toms. Her sec­ond course of intra­ve­nous high-dose meth­o­trex­
ate was delayed by 2 weeks, dur­ing which she con­tin­ued to
Rechallenge with meth­o­trex­ate after the first epi­sode receive mer­cap­to­pu­rine. Leucovorin res­cue was ini­ti­ated early
of neu­ro­tox­ic­ity at hour 30 and given every 6 hours for 5 doses dur­ing courses
Multiple stud­ies have val­i­dated the safety of rechallenge with 2 and 3. As she did not develop recur­rent neu­ro­tox­ic­ity, the last
meth­o­trex­ate after the first epi­sode of neu­ro­tox­ic­ity, as 82% to course of high-dose meth­o­trex­ate was admin­is­tered with the
92% of patients did not develop recur­rent symp­toms.6-9 Impor- stan­dard leucovorin res­cue. She com­pleted all­planned ther­apy
tantly, omis­sion of meth­o­trex­ate for CNS-directed ther­apy after a for ALL with­out fur­ther com­pli­ca­tions.
neu­ro­toxic epi­sode increased the risk of CNS relapse, as dem­on­
strated in a large Aus­tra­lian cohort of 1251 pedi­at­ric patients with
ALL, includ­ing 95 who expe­ri­enced meth­o­trex­ate neu­ro­tox­ic­ity.8 Recurrence of meth­o­trex­ate neu­ro­tox­ic­ity
The 5-year CNS relapse-free sur­vival was 89% when intra­the­cal Due to the rar­ity of recur­rences, there are no avail­­able guide­
meth­o­trex­ate was per­ma­nently discontinued and replaced with lines for a sec­ond rechallenge. Patients with recur­rent brief sei­
alter­nate intra­the­cal ther­apy after a symp­tom­atic neu­ro­tox­ic­ity zures may tol­er­ate sub­se­quent meth­o­trex­ate under the cover of
epi­sode, com­pared with 95% if patients con­tin­ued to receive appro­pri­ate anti­con­vul­sants for the remain­der of ther­apy. Very
pro­to­col-pre­scribed doses of intra­the­cal meth­o­trex­ate (P = .047). lim­ited data are avail­­able to assess out­comes of sec­ond rechal-
These data high­light the impor­tance for the con­tin­u­a­tion of opti­ lenge after recur­rent SLS. In the UKALL 2003 report, 4 patients
mal ther­apy to max­i­mize cure. with a sec­ond epi­sode of SLS con­tin­ued to receive meth­o­trex­ate
Rechallenge with meth­o­trex­ate should be delayed until full after com­plete neu­ro­logic recov­ery.9 Of these, 3 patients tol­er­
neu­ro­logic recov­ery, which is typ­i­cally less than 7 days. For some ated meth­o­trex­ate well, and 1 devel­oped per­sis­tent neu­ro­logic
patients, this may result in the omis­sion or delay of meth­o­trex­ate def­i­cits, suggesting that a sub­set of patients may con­tinue to
ther­apy by 1 to 2 weeks. Successful resump­tion of stan­dard ther­ receive meth­o­trex­ate safely even after 2 epi­sodes of neu­ro­tox­
apy is pos­si­ble in most patients, with­out the need for exces­sive ic­ity. However, cau­tion is warranted as there are case reports of
delays, sub­sti­tu­tions, or pro­phy­lac­tic med­i­ca­tions. Substitution of patients with poor neu­ro­logic out­comes and rare fatal­i­ties after
intra­the­cal meth­o­trex­ate by cytarabine with or with­out hydro­cor­ repeated epi­sodes of meth­o­trex­ate neu­ro­tox­ic­ity.9,20 In these
ti­sone can be con­sid­ered tem­po­rar­ily for 1 or 2 doses, with the uncom­mon sce­nar­ios, par­tic­u­larly when patients do not make
resump­tion of intra­the­cal meth­o­trex­ate as soon as pos­si­ble. Leu- a full recov­ery in the expected time frame or have recur­rent
covorin res­cue of 2 sys­temic doses (5 mg/m2/dose) at hours 24 severe meth­o­trex­ate neu­ro­tox­ic­ity, com­ple­tion of CNS-directed
and 30 after intra­the­cal meth­o­trex­ate rechallenge is a rea­son­able ther­apy with alter­nate agents would require an indi­vid­u­al­ized
strat­egy to attempt to min­i­mize recur­rent epi­sodes. For sys­temic approach based on the patient’s risk of CNS relapse.
meth­ot­rex­ate rechallenge, early leucovorin res­cue at hour 30 or
36 after the start of the 24-hour infu­sion of high-dose meth­o­trex­
ate could be con­sid­ered. Although there are no ran­dom­ized stud­
ies for this sec­ond­ary pro­phy­laxis, leucovorin is the only drug with
strong clin­ic­ al evi­dence for reduc­ing the inci­dence of meth­o­trex­ CLINICAL CASE 2
ate neu­ro­tox­ic­ity.17-19 The the­o­ret­i­cal con­cern of “res­cue” of intra­ The patient is a 16-year-old Cau­ca­sian male with B-ALL in sec­ond
the­cal meth­o­trex­ate by leucovorin and con­se­quent reduc­tion of relapse with high bone mar­row leu­ke­mia bur­den. He received
its anti­leu­ke­mic effect is off­set by the abil­ity to con­tinue admin­is­ levetiracetam for sei­zure pro­phy­laxis and fludarabine and cyclo­
tra­tion of this impor­tant drug to reduce risk of relapse. Additional phos­pha­mide for lymphodepletion prior to infu­sion of autol­o­
leucovorin res­cue may not be needed indef­i­nitely, espe­cially if gous anti-CD19 CAR T cells. On postinfusion day 3, he devel­oped
the patient tol­er­ates the sub­se­quent rechallenge well. grade 2 cyto­kine release syn­drome (CRS) manifested by fevers,

378  |  Hematology 2021  |  ASH Education Program


hyp­oxia, and hypo­ten­sion.21 Initial man­ age­ment included oxy­ stud­ies (Figure 2).21 ICANS is defined as “a dis­or­der char­ac­ter­
gen sup­ple­men­ta­tion, intra­ve­nous fluid bolus, tocilizumab, and ized by a path­o­logic pro­cess involv­ing the CNS fol­low­ing any
meth­yl­pred­nis­o­lone. After tran­sient improve­ment, grade 2 CRS immune ther­apy that results in the acti­va­tion or engage­ment
recurred with wors­en­ing hypo­ten­sion on day 5, requir­ing dopa­ of endog­e­nous or infused T cells and/or other immune effec­tor
mine infu­sion. A sec­ond dose of tocilizumab was admin­is­tered for cells.”21 With CD19 CAR T ther­apy, neu­ro­tox­ic­ity of all­grades
CRS. On day 6, he was lethar­gic, with expres­sive dys­pha­sia and has been reported in 28% to 64% of patients and grades ≥3
an immune effec­tor cell–asso­ci­ated enceph­a­lop­a­thy (ICE) score in 13% to 50%.5 Fatal ICANS was reported in approx­i­ma­tely 3%
of 5, meet­ing cri­te­ria for grade 2 ICANS.21 Dexamethasone was ini­ of adult patients.22 ICANS begins in the first week postinfusion
ti­ated for ICANS, and CT of the head was performed, which was (median 4-6 days) in 90% of patients, soon after the onset of
nor­mal. However, his neu­ro­logic sta­tus rap­idly declined through­ CRS or as CRS symp­toms sub­side. Cases of ICANS with­out CRS
out the day, and he devel­oped a gen­er­al­ized tonic-clonic sei­zure are uncom­mon and have less severe man­i­fes­ta­tions. Typically,
that abated after 2 doses of lorazepam. He was given a load­ing neu­ro­logic symp­toms are tran­sient and resolve in 5 to 11 days
dose of levetiracetam and trans­ferred to the inten­sive care unit. but can be noted for up to 8 weeks.5,23 Delayed-onset ICANS

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/376/1851679/376bhojwani.pdf by guest on 13 December 2021


He was stu­por­ous on exam­i­na­tion and required intu­ba­tion for at 1 month after infu­sion has also been described.24 Early signs
apneic epi­sodes. ICANS had progressed to grade 4. Aggressive of ICANS include expres­sive dys­pha­sia, tremor, and dysgraphia
neuroprotective mea­sures were insti­tuted with ele­va­tion of head and can prog­ress to sei­zures, depressed level of con­scious­
of bed and sup­port­ive inter­ven­tions to main­tain normotension ness, enceph­a­lop­a­thy, and coma. Cerebral edema is a dreaded
and nor­mo­ther­mia. Anakinra and high-dose meth­yl­pred­nis­o­lone con­se­quence of ICANS. Baseline risk fac­tors for ICANS include
were ini­ti­ated. high bone mar­row dis­ease bur­den and preexisting neu­ro­logic
def­i­cits, whereas postinfusion risk fac­tors include higher-grade
CRS, inflam­ma­tory mark­ers such as fever and increased fer­ri­
ICANS tin, and high serum lev­els of spe­cific cyto­kines (eg, inter­leu­
Accurate assess­ments of the inci­dence and sever­ity of neu­ro­ kin [IL]–2, IL-6, IL-10, IL-15).22,23,25 Although mech­a­nisms lead­ing
tox­ic­ity were lim­ited by dif­fer­ing def­i­ni­tions and grad­ing cri­te­ria to ICANS are unclear, exper­i­men­tal mod­els sug­gest cyto­kine-
used in ear­lier CAR T stud­ies. Therefore, the Amer­i­can Society induced neuroinflammation and endo­the­lial acti­va­tion, with
of Transplantation and Cellular Therapy (ASTCT) published con­ sub­se­ quent dis­ rup­ tion of the blood-brain bar­ rier and direct
sen­ sus def­i­
ni­
tions and grad­ ing in 2019 for uni­ for­
mity across neu­ro­nal injury.22,25

Figure 2. ASTCT consensus grading for ICANS. Adapted from Lee et al21 and Traube et al44 with permission. ICANS grading (blue box)
has 5 elements. The first element assesses encephalopathy and uses a scoring tool: ICE (green box) for patients ≥12 years and CAPD
(orange box) for patients <12 years of age. CAPD, Cornell Assessment of Pediatric Delirium; ICE, immune effector cell–associated
encephalopathy; ICP, increased intracranial pressure.
Prakash Singh Shekhawat
Neurotoxicity in chil­dren with ALL  |  379
Acute man­age­ment of ICANS orrhages have also been observed with severe ICANS.22 In an
Commercially avail­­able CAR T-cell prod­ucts in the United States study of adult patients, EEG showed dif­fuse slowing in 76% of
are governed under the Food and Drug Administration Risk Eval- patients dur­ing acute ICANS and was also help­ful in detecting
uation and Mitigation Strategy drug safety pro­gram to ensure sub­clin­i­cal sei­zures.22 Characteristic CSF find­ings for ICANS are
that health care pro­vid­ers and rel­e­vant staff are trained in the mod­er­ate leu­ko­cy­to­sis with lym­pho­cyte pre­dom­i­nance and
rec­og­ni­tion and man­age­ment of toxicities asso­ci­ated with CAR T mark­edly ele­vated pro­tein.22
ther­apy, includ­ing ICANS. With broader expe­ri­ence gained in the For grade 2 ICANS, glu­co­cor­ti­coids (dexa­meth­a­sone 0.2 mg/
man­age­ment of ICANS, sev­eral clin­i­cal man­age­ment guide­lines kg/dose, max­i­mum 10 mg every 6 hours, or meth­yl­pred­nis­o­lone
have been published in the past 4 years.26-28 General rec­om­men­ equiv­al­ents) should be strongly con­sid­ered as brief courses do
da­tions are sum­ma­rized in Figure 3. not neg­at­ively affect the effi­cacy of the CAR T cells and may
Although pro­phy­lac­tic anti­con­vul­sants are not used uni­ shorten the dura­ tion of symp­ toms.29 For higher-grade ICANS
formly due to lim­ited evi­dence for pre­ven­tion of ICANS-related 3 or 4, along with con­tin­ued sup­port­ive care in col­lab­o­ra­tion
sei­zures, they are recommended in patients with a his­tory of with intensivists, neu­rol­o­gists, and neu­ro­sur­geons, the dose of

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/376/1851679/376bhojwani.pdf by guest on 13 December 2021


sei­zures or other neu­ro­logic comorbidities. Close clin­i­cal mon­ glu­co­cor­ti­coids should be increased (meth­yl­pred­nis­o­lone 10 or

tor­ing and atten­ tive sup­port­
ive care are often suf­fi­
cient for 20 mg/kg/dose, max­i­mum 500 or 1000 mg every 12 hours) until
grade 1 ICANS. Tocilizumab (anti–IL-6 recep­tor anti­body) is only improve­ment, followed by rapid taper. In addi­tion, mul­ti­ple anti­
indi­cated for con­cur­rent CRS, not for treat­ment of ICANS. Due con­vul­sants for opti­mal sei­zure con­trol, air­way pro­tec­tion by
to its large size (molec­u­lar weight 149 kDA), tocilizumab does intu­ba­tion and mechan­i­cal ven­ti­la­tion, and decreas­ing intra­cra­
not cross the blood-brain bar­rier and may also facil­i­tate a par­a­ nial pres­sure with hyperosmolar ther­apy or neu­ro­sur­gi­cal inter­
dox­ic­ al shift of IL-6 from the blood to the brain.22 Neuroimaging ven­tion may be warranted.
pref­er­a­bly with MRI, elec­tro­en­ceph­a­lo­gram (EEG), and lum­bar There are very lim­ited clin­i­cal data on other phar­ma­co­logic
punc­ture with open­ing pres­sure can sup­port the diag­no­sis of inter­ven­tions targeted to the path­o­phys­i­­ol­ogy of ICANS. Siltux-
ICANS and rule out other causes of neu­ro­logic symp­toms such imab binds to IL-6 (unlike tocilizumab that binds to the IL-6 recep­
as infec­tion, CNS leu­ke­mia. or hem­or­rhage. MRI changes of focal tor) and facil­i­tates removal of IL-6 from the cir­cu­la­tion, leav­ing
or dif­fuse cere­bral edema guide ICANS grad­ing and man­age­ less IL-6 to enter the brain.27 Anakinra, the IL-1 recep­tor antag­o­
ment. Leptomeningeal enhance­ment and mul­ti­fo­cal microhem- nist, may also be of ben­e­fit as mono­cyte-derived IL-1 can pre­cede

Figure 3. General guidelines for management of ICANS. Frequent neurologic assessments, multidisciplinary supportive care, and glu-
cocorticoids form the backbone of ICANS management. In addition, several novel strategies are being studied for refractory ICANS,
as described in the text. ATG, anti–thymocyte globulin; CRP, C-reactive protein; EVD, external ventricular drain; ICU, intensive care
unit; LP, lumbar puncture; NPO, nothing by mouth.

380  |  Hematology 2021  |  ASH Education Program


and induce IL-6 secre­tion.30,31 The molec­u­lar weight of anakinra
is 17.3 kDA, and ther­a­peu­tic con­cen­tra­tions can be achieved in CLINICAL CASE 3
the CSF.32 Another potential approach to reduce neuroinflamma- A 9-year old His­panic girl with B-ALL in first relapse had min­i­mal
tory cyto­kine pro­duc­tion by mono­cytes is the use of lenzilumab, resid­ual dis­ease 0.2% after reinduction che­mo­ther­apy. Blinatu-
the anti–granulocyte-mac­ro­phage col­ony-stim­u­lat­ing fac­tor momab con­tin­u­ous infu­sion 15 µg/kg/d was ini­ti­ated, and she
mono­clo­nal anti­body. Lenzilumab reduced neuroinflammation was mon­i­tored inpa­tient. On day 4, she complained of head­ache
after CAR T-cells in pre­clin­i­cal mod­els.33 Clinical tri­als of these that improved with acet­amin­o­phen. Three hours later, she devel­
agents as pro­ phy­lac­
tic strat­e­
gies for ICANS are ongo­ ing oped acute-onset con­fu­sion and delir­ium. She was agi­tated and
(NCT04359784, NCT04150913, NCT04432506, NCT04148430, moaning, she could not fol­low com­mands, and her speech was
NCT04205838, NCT04314843). Dasatinib, a tyro­ sine kinase inco­her­ent. She was assessed to have grade 2 enceph­a­lop­a­thy.
inhib­i­tor, reduces CAR T-cell sig­nal­ing revers­ibly by inter­fer­ing Blinatumomab infu­sion was imme­di­ately stopped, and dexa­
with lym­pho­cyte-spe­cific pro­tein tyro­sine kinase.34 In a murine meth­a­sone was admin­is­tered. Within an hour of these inter­ven­
model, the dose of dasatinib was titrated to achieve par­tial or tions, her symp­toms began to improve, and 6 hours later, her

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/376/1851679/376bhojwani.pdf by guest on 13 December 2021


com­plete inhi­bi­tion of CAR T-cell func­tion.34 This strat­egy of mod­ neu­ro­logic sta­tus was back to nor­mal. Because symp­toms did
u­la­tion of CAR T-cell func­tion with dasatinib is being stud­ied as not recur, blinatumomab infu­sion was restarted after 3 days at
an inter­ven­tion for CRS and ICANS in an ongo­ing clin­i­cal trial a lower dose of 5 µg/kg/d, along with dexa­meth­a­sone premed-
(NCT04603872). The use of intra­the­cal hydro­cor­ti­sone, meth­ ication. The dose of blinatumomab was esca­lated to 15 µg/kg/d
o­trex­ate, and cytarabine in ste­roid-refrac­tory ICANS has been a week later, and she tol­er­ated the rest of the course well.
described in case reports.35,36 Additional stud­ies are needed to
inves­ti­gate if such attempts to reduce neuroinflammation by
intra­the­cal ther­apy and elim­i­nate immune effec­tor cells in the Neurotoxicity after blinatumomab
CSF with cyto­toxic agents will be ben­e­fi­cial in the man­age­ment Similar prin­ci­ples of grad­ing and man­age­ment of ICANS from
of ICANS with­out neg­a­tively affect­ing CNS leu­ke­mia con­trol by CAR T apply to neu­ro­tox­ic­ity from blinatumomab. All clin­i­cal tri­
CAR T cells. However, in severe life-threat­ en­
ing ICANS, elim­ i­ als to date have reported grades of indi­vid­ual neu­ro­toxic events
na­tion of the CAR T cells is delib­er­ate, for exam­ple, with anti– based on the Common Terminology Criteria for Adverse Events
thy­mo­cyte glob­u­lin36 or cyclo­phos­pha­mide or by acti­vat­ing CAR (CTCAE), but future stud­ies will likely fol­low the ASTCT ICANS
T-cell sui­cide approaches designed into spe­cific con­structs.37 guide­lines. The inci­dence of blinatumomab-asso­ci­ated neu­ro­
tox­ic­ity was 54% in adult patients with relapsed/refrac­tory ALL38
and 24% in a sim­i­lar pedi­at­ric cohort.39 Of all­neu­ro­logic events,
10% to 17% were sei­zures. In adult patients, symp­toms began at
CLINICAL CASE 2 (Con­t in­u ed) a median of 9 days and also included tremor, som­no­lence, diz­zi­
With con­tin­ued aggres­sive neurocritical care and mul­ti­modal ness, con­fu­sion, and enceph­a­lop­a­thy.38 Median time to onset of
ther­apy, his clin­i­cal con­di­tion began to improve on day 9, evi- neu­ro­tox­ic­ity may be ear­lier in pedi­at­ric patients.39
denced by spon­ta­ne­ous move­ments and with­drawal to pain. Blinatumomab has a very short elim­i­na­tion half-life (1.25
No fur­ther sei­zure activ­ity was noted on EEG. Methylpredniso- hours), and most toxicities resolve after interrupting the infu­sion
lone was tapered day 10 onward, and he was extubated on day and ini­ti­at­ing dexa­meth­a­sone.38,39 Blinatumomab infu­sion should
11. He con­tin­ued to have resid­ual apha­sia that grad­u­ally recov­ be stopped for any sei­zure and for grade 3 or 4 nonseizure neu­ro­
ered to base­line by day 15. tox­ic­ity.40 Prompt ini­ti­a­tion of dexa­meth­a­sone (0.2-0.4 mg/kg/d,
max­i­mum 24 mg, in 3 divided doses for 1-3 days) is recommended

Table 2. Summary of man­i­fes­ta­tions and man­age­ment of neu­ro­tox­ic­ity from meth­o­trex­ate, CAR T cells, and blinatumomab

Characteristic Methotrexate CAR T cells Blinatumomab


Frequency of neu­ro­tox­ic­ity 3%-7% 28%-64% 24%-54%
Risk fac­tors Older age, His­panic eth­nic­ity, High dis­ease bur­den, high-grade Older age, non­white race, prior
his­tory of ele­vated trans­am­i­nases CRS, preexisting neu­ro­logic neu­ro­logic events, >2 prior
def­i­cits sal­vage ther­a­pies
Time of onset from ther­apy 3-11 days 4-6 days Median 9 days in adult patients,
ini­ti­a­tion may be ear­lier in pedi­at­ric
patients
Common signs/symp­toms Seizures, enceph­a­lop­a­thy, SLS Seizures, dys­pha­sia, enceph­a­lop­a­thy, Seizures, tremor, som­no­lence,
cere­bral edema enceph­a­lop­a­thy
Average time to res­o­lu­tion 1-4 days 5-11 days 1-5 days
MRI find­ings Leukoencephalopathy Cerebral edema, leptomeningeal Insufficient data avail­­able
enhance­ment, mul­ti­fo­cal
microhemorrhages
Management Supportive care Neurocritical care, ste­roids, anti­con­ Steroids, inter­rup­tion of
vul­sants, anticytokine ther­a­pies blinatumomab infu­sion
Prakash Singh Shekhawat
Neurotoxicity in chil­dren with ALL  |  381
for any severe neu­ro­tox­ic­ity. Blinatumomab is per­ma­nently dis- References
continued if the patient devel­ops more than 1 sei­zure, any grade 4 1. Vora A, Andreano A, Pui C-H, et  al. Influence of cra­nial radio­ther­apy on
out­come in chil­dren with acute lym­pho­blas­tic leu­ke­mia treated with con­
neu­ro­tox­ic­ity, or if neu­ro­logic recov­ery is delayed > 7 days. For all­
tem­po­rary ther­apy. J Clin Oncol. 2016;34(9):919-926.
other patients, blinatumomab can be restarted at one-third of the 2. Parasole R, Petruzziello F, Menna G, et al. Central ner­vous sys­tem com­pli­
dose (ie, 5 µg/kg, max­i­mum 9 µg) with dexa­meth­a­sone premedi- ca­tions dur­ing treat­ment of acute lym­pho­blas­tic leu­ke­mia in a sin­gle pedi­
cation (5 mg/m2, max­i­mum 20 mg) after improve­ment to grade ≤ 1 at­ric insti­tu­tion. Leuk Lymphoma. 2010;51(6):1063-1071.
and a min­im ­ um of 3 days. If neu­ro­tox­ic­ity does not recur, blinatu- 3. Banerjee J, Niinimäki R, Lähteenmäki P, et al. The spec­trum of acute cen­tral
ner­vous sys­tem symp­toms dur­ing the treat­ment of child­hood acute lym­
momab can be esca­lated to full dose after 7 days. Secondary sei­
pho­blas­tic leu­kae­mia. Pediatr Blood Cancer. 2020;67(2):e27999.
zure pro­phy­laxis with anti­con­vul­sants may be con­sid­ered for the 4. Orgel E, Bhojwani D. Medical sup­port­ive care for treat­ment-related tox­ic­ity
remain­der of the cycle and for sub­se­quent cycles. Rechallenge of in child­hood ALL. In: Vora A, ed. Childhood Acute Lymphoblastic Leuke-
blinatumomab is not recommended after recur­rence of the ini­tial mia. Springer International Publishing; 2017:299-321.
5. Gust J, Ceppi F, Turtle C. Neurotoxicities after CART T-cell immu­no­ther­
neu­ro­toxic event.
apy. In: Lee D, Shah N, eds. Chimeric Antigen Receptor T-Cell Therapies for
Cancer. Elsevier; 2020:83-105.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/376/1851679/376bhojwani.pdf by guest on 13 December 2021


Conclusions 6. Badke C, Flem­ing A, Iqbal A, et al. Rechallenging with intra­the­cal meth­o­trex­
Table 2 sum­ma­rizes the man­i­fes­ta­tions and man­age­ment of neu­ ate after devel­op­ing sub­acute neu­ro­tox­ic­ity in chil­dren with hema­to­logic
ro­tox­ic­ity from meth­o­trex­ate, CAR T cells, and blinatumomab. A malig­nan­cies. Pediatr Blood Cancer. 2016;63(4):723-726.
7. Bhojwani D, Sabin ND, Pei D, et al. Methotrexate-induced neu­ro­tox­ic­ity and
detailed under­stand­ing of the mech­an ­ isms of these neurotoxici-
leukoencephalopathy in child­hood acute lym­pho­blas­tic leu­ke­mia. J Clin
ties will lead to the devel­op­ment of targeted approaches for pre­ Oncol. 2014;32(9):949-959.
ven­tion and treat­ment. Until then, close mon­i­tor­ing and atten­tive, 8. Mateos MK, Marshall GM, Barbaro PM, et al. Methotrexate-related cen­tral
mul­ti­modal sup­port­ive care form the back­bone of man­ag­ing clin­ neu­ro­tox­ic­ity: clin­i­cal char­ac­ter­is­tics, risk fac­tors and genome-wide asso­
i­cal man­i­fes­ta­tions. In addi­tion, glu­co­cor­ti­coids and anticytokine ci­a­tion study in chil­dren treated for acute lym­pho­blas­tic leu­ke­mia [pub-
lished online 11 Feb­ru­ary 2021]. Haematologica.
ther­a­pies dampen the neuroinflammatory cas­cade in ICANS. Neu-
9. Bond J, Hough R, Moppett J, Vora A, Mitchell C, Goulden N. Stroke-like syn­
rologic symp­toms are usu­ally tran­sient, and most patients appear drome caused by intra­the­cal meth­o­trex­ate in patients treated dur­ing the
to make a full recov­ery. However, screen­ing MRIs detect leuko- UKALL 2003 trial. Leukemia. 2013;27(4):954-956.
encephalopathy in 20% of chil­dren receiv­ing meth­o­trex­ate dur­ 10. Schmiegelow K, Attarbaschi A, Barzilai S, et al; Ponte di Legno tox­ic­ity
work­ing group. Consensus def­i­ni­tions of 14 severe acute toxic effects for
ing front­line ALL ther­apy,7 and even asymp­tom­atic patients with
child­hood lym­pho­blas­tic leu­kae­mia treat­ment: a Delphi con­sen­sus. Lancet
leukoencephalopathy are at high risk for long-term adverse neu- Oncol. 2016;17(6):e231-e239.
robehavioral prob­lems.41 Survivors of child­hood ALL treated with 11. Taylor OA, Brown AL, Brackett J, et al. Disparities in neu­ro­tox­ic­ity risk and
che­mo­ther­apy alone con­tinue to have infe­rior neurocognitive out­comes among pedi­at­ric acute lym­pho­blas­tic leu­ke­mia patients. Clin
out­comes, par­tic­u­larly in domains of exec­u­tive func­tion, atten­ Cancer Res. 2018;24(20):5012-5017.
12. Inaba H, Khan RB, Laningham FH, Crews KR, Pui C-H, Daw N-C. Clinical
tion, and mem­ory.42 Similar pedi­at­ric data are not avail­­able after
and radio­log­i­cal char­ac­ter­is­tics of meth­o­trex­ate-induced acute enceph­
immu­no­ther­apy, but a recent study noted clin­i­cally sig­nif­i­cant a­lop­a­thy in pedi­at­ric patients with can­cer. Ann Oncol. 2008;19(1):178-
neu­ro­psy­chi­at­ric prob­lems in 50% of adult patients 1 to 5 years 184.
after CD19 CAR T-cell ther­apy.43 Therefore, in addi­tion to devel­ 13. Schmiegelow K. Advances in indi­vid­ual pre­dic­tion of meth­o­trex­ate tox­ic­
ity: a review. Br J Haematol. 2009;146(5):489-503.
op­ing strat­e­gies to decrease the fre­quency and mor­bid­ity of
14. Afshar M, Birnbaum D, Golden C. Review of dextromethorphan admin­is­
acute neu­ro­tox­ic­ity from ALL ther­apy, it is also imper­a­tive to tra­tion in 18 patients with sub­acute meth­o­trex­ate cen­tral ner­vous sys­tem
devise approaches to reduce its con­se­quences on long-term tox­ic­ity. Pediatr Neurol. 2014;50(6):625-629.
out­comes. 15. Razi W, Haque AU, Sadiq H, Ullah R, Jabbar N, Mirza S. Safety and effi­cacy of
ami­noph­yl­line in intra­the­cal meth­o­trex­ate-related neu­ro­log­i­cal tox­ic­ity in
large pedi­at­ric oncol­ogy cen­tre. J Coll Physicians Surg Pak. 2021;30(4):481-
Conflict-of-inter­est dis­clo­sure
484.
Deepa Bhojwani: no com­pet­ing finan­cial inter­ests to declare. 16. Lexicomp. Riverwoods, IL: Wolters Kluwer Health, Inc. Accessed 15 June
Ravi Bansal: no com­pet­ing finan­cial inter­ests to declare. 2021. http:​­/​­/online​­.lexi​­.com
Alan S. Wayne: receives research funding from Kite Pharma (a Gilead 17. Mahoney DH Jr, Shuster JJ, Nitschke R, et al. Acute neu­ro­tox­ic­ity in chil­dren
com­pany) and Institut de Recherches Internationales Servier. with B-pre­cur­sor acute lym­phoid leu­ke­mia: an asso­ci­a­tion with inter­me­di­ate-
dose intra­ve­nous meth­o­trex­ate and intra­the­cal tri­ple ther­apy—a Pediatric
Oncology Group study. J Clin Oncol. 1998;16(5):1712-1722.
Off-label drug use 18. Winick NJ, Bowman WP, Kamen BA, et al. Unexpected acute neu­ro­logic
Deepa Bhojwani: multiple off-label drugs are discussed in the tox­ic­ity in the treat­ment of chil­dren with acute lym­pho­blas­tic leu­ke­mia. J
article: dextromethorphan, aminophylline, siltuximab, anakinra, Natl Cancer Inst. 1992;84(4):252-256.
lenzilumab, and dasatinib. 19. Cohen IJ. Neurotoxicity after high-dose meth­o­trex­ate (MTX) is ade­quately
explained by insuf­fi­cient folinic acid res­cue. Cancer Chemother Pharma-
Ravi Bansal: multiple off-label drugs are discussed in the article: col. 2017;79(6):1057-1065.
dextromethorphan, aminophylline, siltuximab, anakinra, lenzi- 20. Dabagh S, David H, Young S, Doan A, Bhojwani D. Severe, fatal meth­o­trex­ate-
lumab, and dasatinib. related neu­ro­tox­ic­ity in 2 ado­les­cent patients with ALL. J Pediatr Hematol
Alan S. Wayne: multiple off-label drugs are discussed in the article: Oncol. 2020;42(8):e839-e844.
21. Lee DW, Santomasso BD, Locke FL, et  al. ASTCT con­ sen­
sus grad­ing
dextromethorphan, aminophylline, siltuximab, anakinra, lenzi-
for cyto­kine release syn­drome and neu­ro­logic tox­ic­ity asso­ci­ated with
lumab, and dasatinib. immune effec­tor cells. Biol Blood Marrow Transplant. 2019;25(4):625-
638.
Correspondence 22. Gust J, Hay KA, Hanafi L-A, et al. Endothelial acti­va­tion and blood-brain bar­
rier dis­rup­tion in neu­ro­tox­ic­ity after adop­tive immu­no­ther­apy with CD19
Deepa Bhojwani, MD, Division of Hematology-Oncology,
CAR-T cells. Cancer Discov. 2017;7(12):1404-1419.
­Children’s Hospital Los Angeles, 4650 Sunset Blvd., MS 54, Los 23. Gofshteyn JS, Shaw PA, Teachey DT, et al. Neurotoxicity after CTL019 in a
Angeles, CA 90027; email: dbhojwani@chla.usc.edu pedi­at­ric and young adult cohort. Ann Neurol. 2018;84(4):537-546.

382  |  Hematology 2021  |  ASH Education Program


24. Tallantyre EC, Evans NA, Parry-Jones J, Mor­gan MPG, Jones CH, Ingram W. 36. Wang M, Jain P, Chi TL, et al. Management of a patient with man­tle cell lym­
Neurological updates: neu­ro­log­i­cal com­pli­ca­tions of CAR-T ther­apy. J Neu- phoma who devel­oped severe neu­ro­tox­ic­ity after chi­me­ric anti­gen recep­
rol. 2021;268(4):1544-1554. tor T-cell ther­apy in ZUMA-2. J Immunother Cancer. 2020;8(2):e001114.
25. Morris EC, Neelapu SS, Giavridis T, Sadelain M. Cytokine release syn­drome 37. Caulier B, Enserink JM, Wälchli S. Pharmacologic con­trol of CAR T cells. Int
and asso­ci­ated neu­ro­tox­ic­ity in can­cer immu­no­ther­apy [published online J Mol Sci. 2021;22(9):4320.
17 May 2021]. Nat Rev Immunol. 2021. 38. Stein AS, Schiller G, Ben­ja­min R, et al. Neurologic adverse events in patients
26. Neelapu SS, Tummala S, Kebriaei P, et al. Chimeric anti­gen recep­tor T-cell with relapsed/refrac­tory acute lym­pho­blas­tic leu­ke­mia treated with blina-
ther­apy—assess­ment and man­age­ment of toxicities. Nat Rev Clin Oncol. tumomab: man­age­ment and miti­gat­ing fac­tors. Ann Hematol. 2019;98(1):
2018;15(1):47-62. 159-167.
27. Maus MV, Alexander S, Bishop MR, et  al. Society for Immunotherapy of 39. von Stackelberg A, Locatelli F, Zugmaier G, et al. Phase I/phase II study of
Cancer (SITC) clin­i­cal prac­tice guide­line on immune effec­tor cell-related blinatumomab in pedi­at­ric patients with relapsed/refrac­tory acute lym­
adverse events. J Immunother Cancer. 2020;8(2):e001511. pho­blas­tic leu­ke­mia. J Clin Oncol. 2016;34(36):4381-4389.
28. Cornillon J, Hadhoum N, Roth-Guepin G, et  al. Management of CAR-T 40. BLINCYTO (blinatumomab) for injec­tion. Vol. 2021. Accessed 15 June 2021.
cell-related enceph­a­lop­a­thy syn­drome in adult and pedi­at­ric patients: rec­ https:​­/​­/www​­.accessdata​­.fda​­.gov​­/drugsatfda_docs​­/label​­/2017​­/125557​
om­men­da­tions of the French Society of Bone Marrow trans­plan­ta­tion and s008lbl​­.pdf
cel­lu­lar ther­apy (SFGM-TC) [in French]. Bull Cancer. 2020;107(1S):S12-S17. 41. Cheung YT, Sabin ND, Reddick WE, et al. Leukoencephalopathy and long-

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/376/1851679/376bhojwani.pdf by guest on 13 December 2021


29. Karschnia P, Jordan JT, Forst DA, et al. Clinical pre­sen­ta­tion, man­age­ment, term neurobehavioural, neurocognitive, and brain imag­ing out­comes in
and bio­mark­ers of neu­ro­tox­ic­ity after adop­tive immu­no­ther­apy with CAR T sur­vi­vors of child­hood acute lym­pho­blas­tic leu­kae­mia treated with che­mo­
cells. Blood. 2019;133(20):2212-2221. ther­apy: a lon­gi­tu­di­nal anal­y­sis. Lancet Haematol. 2016;3(10):e456-e466.
30. Strati P, Ahmed S, Kebriaei P, et al. Clinical effi­cacy of anakinra to mit­i­gate 42. Cheung YT, Krull KR. Neurocognitive out­ comes in long-term sur­ vi­vors of
CAR T-cell ther­apy-asso­ci­ated tox­ic­ity in large B-cell lym­phoma. Blood child­hood acute lym­pho­blas­tic leu­ke­mia treated on con­tem­po­rary treat­ment
Adv. 2020;4(13):3123-3127. pro­to­cols: a sys­tem­atic review. Neurosci Biobehav Rev. 2015;53:108-120.
31. Norelli M, Camisa B, Barbiera G, et al. Monocyte-derived IL-1 and IL-6 are 43. Ruark J, Mullane E, Cleary N, et al. Patient-reported neu­ro­psy­chi­at­ric out­
dif­fer­en­tially required for cyto­kine-release syn­drome and neu­ro­tox­ic­ity comes of long-term sur­vi­vors after chi­me­ric anti­gen recep­tor T cell ther­
due to CAR T cells. Nat Med. 2018;24(6):739-748. apy. Biol Blood Marrow Transplant. 2020;26(1):34-43.
32. Galea J, Ogungbenro K, Hulme S, et al. Intravenous anakinra can achieve 44. Traube C, Silver G, Kearney J, et al. Cornell assess­ment of pedi­at­ric delir­
exper­i­men­tally effec­tive con­cen­tra­tions in the cen­tral ner­vous sys­tem ium: a valid, rapid, obser­va­tional tool for screen­ing delir­ium in the PICU.
within a ther­a­peu­tic time win­dow: results of a dose-rang­ing study. J Cereb Crit Care Med. 2014;42(3):656-663.
Blood Flow Metab. 2011;31(2):439-447.
33. Sterner RM, Sakemura R, Cox MJ, et al. GM-CSF inhi­bi­tion reduces cyto­kine
release syn­drome and neuroinflammation but enhances CAR-T cell func­
tion in xeno­grafts. Blood. 2019;133(7):697-709.
34. Mestermann K, Giavridis T, Weber J, et  al. The tyro­sine kinase inhib­i­tor
dasatinib acts as a phar­ma­co­logic on/off switch for CAR T cells. Sci Transl
Med. 2019;11(499):eaau5907.
35. Shah NN, Johnson BD, Fenske TS, Raj RV, Hari P. Intrathecal che­mo­ther­apy
for man­age­ment of ste­roid-refrac­tory CAR T-cell-asso­ci­ated neu­ro­tox­ic­ity © 2021 by The Amer­i­can Society of Hematology
syn­drome. Blood Adv. 2020;4(10):2119-2122. DOI 10.1182/hema­tol­ogy.2021000269

Prakash Singh Shekhawat


Neurotoxicity in chil­dren with ALL  |  383
LET THE CHIP(S) FALL WHERE THEY MAY ? BURDENS AND BENEFITS OF DIAGNOSING CLONAL HEMATOPOIESIS

CHIP: is clonal hematopoiesis a surrogate for aging


and other disease?

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/384/1851752/384gondek.pdf by guest on 13 December 2021


Lukasz P. Gondek
The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD

Somatic mutations are an unavoidable consequence of aging tissues. Even though most mutations are functionally silent,
some may affect genes critical to proper tissue self-renewal and differentiation, resulting in the outgrowth of affected
cells, also known as clonal expansion. In hematopoietic tissue such clonal dominance is known as clonal hematopoiesis
(CH). Sporadic CH is frequent in aging and affects over 10% of individuals beyond the fifth decade of life. It has been
associated with an increased risk of hematologic malignancies and cardiovascular disease. In addition to aging, CH has
been observed in other hematologic conditions and confers an adaptation of hematopoietic stem cells (HSCs) to vari-
ous environmental stressors and cell-intrinsic defects. In the presence of extrinsic stressors such as genotoxic therapies,
T-cell-mediated immune attack, or inflammation, somatic mutations may result in augmentation of HSC fitness. Such
attuned HSCs can evade the environmental insults and outcompete their unadapted counterparts. Similarly, in inherited
bone marrow failures, somatic mutations in HSCs frequently lead to the reversion of inherited defects. This may occur
via the direct correction of germline mutations or indirect compensatory mechanisms. Occasionally, such adaptation
may involve oncogenes or tumor suppressors, resulting in malignant transformation. In this brief article, we focus on the
mechanisms of clonal dominance in various clinical and biological contexts.

LEARNING OBJECTIVES
• Review the etiology and nomenclature of CH
• Discuss the mechanism of clonal expansion and stem cell ftness in various clinical and biological contexts

Hemogram (normal range) Additional studies (normal range)


CLINICAL CASE
Absolute reticulocyte count
The patient is a 69-year-old man who is a computer program-
25 (24.1-87.7 K/µL)
mer and has a medical history of hypertension, hyperlipid-
emia, coronary artery disease, and rheumatoid arthritis. He RDW: 15 (11%-14%)
was recently found to have mild normocytic normochromic Hgb, hemoglobin; MCV, mean corpuscular volume; PLT, platelet;
anemia during his annual physical exam. His initial workup RBC, red blood cell; RDW, red cell distribution width; TIBC, total
iron-binding capacity; WBC, white blood cell.
is shown in the table below (abnormal values are in bold).

A bone marrow biopsy revealed normocellular marrow


for age and trilineage hematopoiesis with no dysplasia
Hemogram (normal range) Additional studies (normal range)
and no increased blasts. Normal iron stores and no ringed
WBC: 6.2 (4.5-11 K/µL) Serum iron: 50 (60-150µg/dL) sideroblasts were noted on iron stain. Cytogenetics stud-
RBC: 4.0 (4.5-5.9M/µL) Ferritin: 520 (30-400ng/mL) ies showed a normal male karyotype. A next-generation
sequencing (NGS) targeted myeloid panel revealed a
PLT: 189 (150-350 K/µL) Transferrin 102 (200-400mg/dL)
DNMT3A R882C mutation at a variant allele frequency
Hgb: 12.2 (13-16.3g/dL) TIBC: 250 (300-360µg/dL) (VAF) of 2.5%.
MCV: 90 (80-100 fL) B12 and folate within normal limits

384 | Hematology 2021 | ASH Education Program


Introduction The eti­­ol­ogy of clonal dom­i­nance
The term “clonal hema­to­poi­e­sis” (CH) describes the dom­i­nance of Aging
a hema­to­poi­etic clone aris­ing from a sin­gle hema­to­poi­etic stem Despite the human body’s very effec­tive DNA repair machin­ery,
cell (HSC) rel­a­tive to the rest of the hema­to­poi­etic cells. Since somatic muta­tions are inev­i­ta­ble, and their num­ber increases
the intro­duc­tion of NGS tech­nol­ogy in hema­to­poi­e­sis research, with time. In fact, every tis­ sue accu­mu­ lates somatic muta­
an asso­ci­a­tion between CH and var­i­ous hema­to­logic and nonhe- tions dur­ing devel­op­ment and aging, resulting in the gen­er­a­
matologic con­di­tions has been reported. Thus, a num­ber of new tion of cells with dif­fer­ent geno­types, a phe­nom­e­non known
terms and acro­nyms comprising CH have been pro­posed. as somatic mosa­i­cism.5,6 This is par­tic­u­larly prev­a­lent in highly
For the pur­pose of this review, CH of inde­ter­mi­nate poten­tial pro­lif­er­a­tive tis­sues such as the hema­to­poi­etic sys­tem, and
(CHIP) and age-related clonal hema­to­poi­e­sis (ARCH) will be con­ somatic muta­tions are pres­ent in the blood of nearly all­older
sid­ered syn­on­y­mous and will denote CH char­ac­ter­ized by the adults.7,8 Fortunately, because of their pre­dom­i­nantly sto­chas­
pres­ence of 1 or more somatic muta­tions in genes asso­ci­ated tic nature, most muta­tions affect noncoding regions and thus
with hema­to­logic malig­nan­cies in oth­er­wise healthy indi­vid­u­als remain func­tion­ally silent. Assuming that approx­i­ma­tely 1000

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/384/1851752/384gondek.pdf by guest on 13 December 2021


with­out obvi­ous hema­to­logic con­di­tions. Moreover, the muta­ HSCs are active and con­ trib­ute equally to human hema­ to­
tions must be pres­ent at a VAF ≥2%, although con­sen­sus on the poi­e­sis, somatic mosa­i­cism would be expected at a level of
bio­log­i­cally and clin­i­cally mean­ing­ful VAF cut­off is unknown and 0.001 in the absence of clonal advan­tage of an indi­vid­ual HSC.
has been a sub­ject of ongo­ing research.1-3 In fact, anal­y­sis of the periph­eral blood of patients in the fifth
Clonal cytopenia of unde­ ter­
mined sig­ nif­i­
cance (CCUS) decade of life using ultradeep sequenc­ing showed the pres­
describes the pres­ence of CH in indi­vid­u­als with per­sis­tent uni- ence of somatic mosa­i­cism with a median VAF of 0.002 in 95%
or multilineage cytopenias that can­not be explained by other of indi­vid­u­als. Interestingly, most clones remained sta­ble over
con­di­tions.3 Since cytopenia in CCUS is thought to be due to time.8 Thus, somatic mosa­ i­
cism is fre­
quent with aging but
the under­ly­ing clonal pro­cess, this con­di­tion often rep­re­sents an rarely results in clonal advan­tage and sub­se­quent CH. The rel­
early mye­loid neo­plasm.2 A detailed descrip­tion and the clin­i­cal a­tive expan­sion of a sin­gle clone can arise either due to the
con­se­quences of CCUS are pro­vided in an accom­pa­ny­ing review increased fit­ ness and growth advan­ tage of an HSC (large
by Dr. Osman. numer­a­tor) or as a result of the HSC’s pool con­trac­tion (small
Idiopathic cytopenia of unde­ter­mined significance (ICUS) denom­i­na­tor). Pathogenic muta­tions, often in can­cer-asso­
denotes per­sis­tent uni- or multilineage cytopenia unex­plained ci­ated genes, fre­quently result in the expan­sion of HSCs and
by other con­di­tions and hav­ing no evi­dence of CH.4 clonal dom­i­nance. Nonfunctional var­i­ants, on the other hand,
We pro­pose that the term CH be used for any clonal pro­cess serv­ing only as “clonal mark­ers,” may become detect­able in the
that does not ful­fill the cri­te­ria for the above-men­tioned con­di­ case of HSC pool attri­tion and when ultradeep or wide breadth
tions (Table 1). of cov­er­age NGS is applied. Consistent with this notion, CH was
found to be sig­nif­i­cantly more prev­a­lent when whole-genome
sequenc­ing meth­ods were applied.7 What then is the under­
ly­ing mech­a­nism of hema­to­poi­etic clonal expan­sion in aging
Table 1. Definition of clin­i­cal enti­ties asso­ci­ated with CH
hema­ to­poi­ e­
sis? Undoubtedly, the HSC pool decreases with
age; how­ever, sto­chas­tic HSC attri­tion does not appear to be
Clonal hema­to­poi­e­sis of inde­ter­mi­nate poten­tial (CHIP)
the major means behind clin­i­cally con­se­quen­tial CH (Figure 1)
  •  Presence of somatic muta­tion(s) in gene(s) asso­ci­ated with hema­to­ CH seems to be pre­dom­i­nantly driven by a cell auton­o­mous
logic malig­nan­cies
increase in HSC self-renewal and pos­i­tive selec­tion rather than
  •  Normal hemogram genetic drift.9 This is mostly due to qual­i­ta­tive and/or quan­ti­
  •  No clin­i­cal of path­o­log­i­cal evi­dence of hema­to­logic malig­nancy ta­tive changes in pro­teins encoded by so-called driver genes.
Age-related clonal hema­to­poi­e­sis (ARCH)
DNMT3A and TET2 are among the most com­monly mutated
genes in ARCH and belong to the class of epi­ge­netic mod­i­fi­ers.
  •  Any CH asso­ci­ated with aging
Since both are essen­tial in self-renewal and dif­fer­en­ti­a­tion, they
Clonal cytopenia of unde­ter­mined sig­nif­i­cance (CCUS) impose their effects in a cell auton­o­mous man­ner.
  •  Presence of somatic muta­tion(s) in gene(s) asso­ci­ated with hema­to­ The DNMT3A gene encodes a methyltransferase respon­si­ble
logic malig­nan­cies for de novo DNA meth­yl­a­tion that is crit­i­cal for tran­si­tion from
  •  Presence of clin­i­cally rel­e­vant and per­sis­tent cytopenia in 1 or more one cell state to another and thus is required for proper tis­sue
periph­eral blood cell lin­e­ages devel­op­ment and stem cell main­te­nance. Animal stud­ies have
  •  Diagnostic cri­te­ria of mye­loid neo­plasm not fulfilled
shown that the loss of DNMT3A aug­ments HSC self-renewal with
only a mod­est impact on dif­fer­en­ti­a­tion, lead­ing to the grad­ual
  •  No other causes of cytopenia and molec­u­lar aber­ra­tion expan­sion of mutated HSC and clonal dom­i­nance.10
Idiopathic cytopenia of unde­ter­mined sig­nif­i­cance (ICUS) TET2 is the sec­ond most fre­quently mutated gene in CHIP.
  •  Presence of clin­i­cally rel­e­vant and per­sis­tent cytopenia in 1 or more The prod­uct of this gene belongs to the fam­ily of dioxygen-
periph­eral blood cell lin­e­ages ases. TET2 medi­ates DNA demethylation by cat­a­lyz­ing the oxi­
  •  Not explained by any other dis­ease da­ tion of 5-methylcytosine to 5-hydroxymethylcytosine that
remains unmethylated dur­ing sub­se­quent cell divi­sion.11 Simi-
  •  Diagnostic cri­te­ria of mye­loid neo­plasm not fulfilled
larly to DNMT3A loss, ani­mal mod­els of TET2 knock­out exhibit

Prakash Singh Shekhawat


Mechanism of clonal hema­to­poi­e­sis  |  385
Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/384/1851752/384gondek.pdf by guest on 13 December 2021
Figure 1. Context-dependent mechanism of clonal expansion.

increased HSC self-renewal and skewed hema­to­poi­etic dif­fer­ Immune attack.  The immune destruc­tion of hema­to­poi­etic cells
en­ti­a­tion favor­ing myelomonocytic lin­e­age.12 Similar gain of is a hall­mark of acquired aplastic ane­mia (AA). CH is pres­ent in up
self-renewal has been observed with ASXL1 muta­tions.13 The to 50% of AA patients.19 Perhaps the best exam­ple of clonal im-
impact of less com­mon CH muta­tions on clonal growth advan­ mune escape is the clonal advan­tage of the par­ox­ys­mal noc­tur­
tage, par­tic­ul­arly in genes involved in mRNA splic­ing, is not well nal hemo­glo­bin­uria (PNH) clone aris­ing from phosphatidylinosi-
under­stood. tol gly­can class A-defi­cient HSC. The PNH clone can be found at
low lev­els in most healthy indi­vid­u­als, but the cells lack a com­
Cell-extrin­sic fac­tors pet­i­tive advan­tage under homeo­static con­di­tions. Under im-
A very mod­est increase in the fit­ness of an indi­vid­ual HSC often mune pres­sure, gly­co­syl-phosphatidylinositol-defi­cient cells are
requires years or decades to gain dom­i­nance over unmutated less sus­cep­ti­ble to T-cell-medi­ated kill­ing and out­com­pete their
coun­ter­parts. This mostly indo­lent pro­cess may be fur­ther accel­ unmutated coun­ter­parts.20 This obser­va­tion raises the pos­si­bil­ity
er­ated by a pleth­ora of extra­cel­lu­lar stress­ors, such as increased that the gly­co­syl-phosphatidylinositol-anchored pro­tein might
pro­lif­er­a­tive pres­sure, inflam­ma­tion, and genotoxic agents. The be a crit­i­cal tar­get rec­og­nized by T cells. Inactivating muta­tions
most pro­found stress that can be imposed on the hema­to­poi­etic or loss-of-het­ero­zy­gos­ity of HLA class 1 alleles, allowing for resis­
sys­tem is related to hema­to­poi­etic recon­sti­tu­tion after allo­ge­ tance against autoreactive T cells, is another exam­ple of HSC
neic bone mar­row trans­plan­ta­tion. Recent stud­ies have dem­on­ adap­ta­tion to immune-medi­ated stress.21 The role of other muta­
strated that CH clones pres­ent in allo­ge­neic stem cell donors tions in increased HSC fit­ness is less clear. DNMT3A and ASXL1
undergo accel­er­ated expan­sion in the recip­i­ents while remain- are seen in up to 15% of patients with AA.19 Also, muta­tions in
ing rel­at­ ively dor­mant in the donors.14-16 Inflammation is another BCOR and BCORL1 (tran­scrip­tional core­pres­sors) are fre­quently
pre­sump­tive mod­i­fier of CH and has been fre­quently asso­ci­ated asso­ci­ated with AA. Unlike clones with DNMT3A and ASXL1 muta­
with tis­sue aging. Mutated HSC may gain a selec­tive growth tions that may expand over time and lead to malig­nant trans­
advan­tage when exposed to proinflammatory sig­nals such as IL-6 for­ma­tion, clones car­ry­ing BCOR and BCORL1 muta­ tions re-
or IFN-gamma.17,18 main sta­ble and sel­dom result in pro­gres­sion to acute mye­loid

386  |  Hematology 2021  |  ASH Education Program


leu­ke­mia/myelodysplastic syn­drome.19 The exact mech­a­nism of Cell-intrin­sic defects
the afore­men­tioned muta­tions to aug­ment HSC fit­ness in the In addi­tion to spo­radic CH asso­ci­ated with aging or shaped by
pres­ence of T-cell-medi­ated attack is unclear. extra­cel­lu­lar stress­ors, CH can also rep­re­sent an adap­ta­tion to
intrin­
sic HSC defects under­ ly­
ing inherited bone mar­ row fail­
Genotoxic ther­a­pies. Unlike already primed HSCs with muta­ ure syn­dromes (iBMFs). iBMFs occur as a result of germline HSC
tions resulting in improved fit­ness, some hema­to­poi­etic clones muta­tions and include short telo­mere syn­dromes (STS), impaired
lack the clonal advan­tage under homeo­static con­di­tions but are ribo­some bio­gen­e­sis (Shwachman-Diamond syn­drome; SDS),
capa­ble of evad­ing extra­cel­lu­lar insults or intra­cel­lu­lar defects an increase in DNA dam­age (Fanconi ane­mia; FA), and SAMD9/
(Figure 1). Patients treated with genotoxic che­ mo­ ther­apy or SAMD9L muta­tions (MIRAGE syn­drome). Several com­pen­sa­
radio­ther­apy are at risk for a ther­apy-related mye­loid neo­plasm. tory mech­a­nisms may lead to an increase in HSC self-renewal
It is now well rec­og­nized that min­is­cule hema­to­poi­etic clones in iBMFs. Randomly occur­ring muta­tions in HSCs may result in
car­ry­ing TP53 or PPM1D muta­tions fre­quently not detect­able by somatic rever­sion of the genetic defect, allowing the corrected
stan­dard NGS tech­niques are pres­ent long before treat­ment with clone to “res­cue” hema­to­poi­etic out­put. This may be achieved

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/384/1851752/384gondek.pdf by guest on 13 December 2021


genotoxic agents.22 It is likely that func­tion­ally incon­se­quen­tial via the direct cor­rec­tion of germline muta­tion or an indi­rect com­
somatic muta­tions in TP53 and PPM1D, under homeo­static con­di­ pen­sa­tory mech­a­nism (Table 1).25 Somatic rever­sion of DKC1, a
tions, are a­ ble to resist genotoxic stress. Given that these muta­ gene fre­quently mutated in STS, or gain-of-func­tion muta­tion in
tions likely con­fer an inad­e­quate response to DNA dam­age, it is the TERT pro­moter lead­ing to increased mRNA expres­sion have
not sur­pris­ing that while exposed to genotoxic ther­apy they may been observed in STS. Direct cor­rec­tion of dam­ag­ing muta­tions
cir­cum­vent apo­pto­sis and acquire addi­tional DNA defects. The in FA patients and the full or par­tial func­tion res­to­ra­tion of sev­eral
com­mon exam­ple is the loss of the wild-type TP53 allele result- genes in the FANC com­plex have also been observed. Since
ing in a com­plete loss of func­tion and sub­se­quent accu­mu­la­tion bone mar­row fail­ure in FA is mainly medi­ated by p53-induced
of addi­tional genetic defects and leu­ke­mic trans­for­ma­tion.23,24 apo­pto­sis, the emer­gence of CH with TP53 loss-of-func­tion

Table 2. CH char­ac­ter­is­tics in var­i­ous clin­i­cal con­texts and selec­tive pres­sures

Selective pres­sure Clinical con­text CH char­ac­ter­is­tics Frequency (%)


Extrinsic/intrin­sic Aging DNMT3A 8
TET2 2
ASXL1 2
Extrinsic AA DNMT3A 9
ASXL1 13
BCOR 6
PNH TET2 8
SUZ12 3
U2AF1 3
ASXL1 3
JAK2 5
Chemotherapy TP53 3
PPM1D 15
Intrinsic DC Backmutation 11
TERT pro­moter 5
FA Gene con­ver­sion 9
FANC point muta­tion Case reports
FANC frame­shift muta­tion Case reports
SDS Isochromosome 7q 44
Monosomy 7 13
Deletion 20q 22
TP53 48
SCN CSF3R 12
MIRAGE syn­drome Deletion 7q 18
UPD7q 20
DC, dyskeratosis congenita; SCN, severe con­gen­i­tal neutropenia.

Prakash Singh Shekhawat


Mechanism of clonal hema­to­poi­e­sis  |  387
may pro­vide bet­ter sur­vival and improve­ment in hema­to­poi­ Off-label drug use
etic out­put. The lat­ter is an exam­ple of the indi­rect cor­rec­ Lukasz P. Gondek: nothing to disclose.
tion of a germline defect. Similarly, TP53 muta­tions may res­cue
bone mar­row fail­ure in SDS patients in whom abnor­mal ribo­ Correspondence
some bio­gen­e­sis trig­gers p53-medi­ated apo­pto­sis. The cleav­ Lukasz P. Gondek, The Sidney Kimmel Comprehensive Cancer
age of EIF6 by SBDS pro­tein is essen­tial for proper ribo­some Center, Johns Hopkins University School of Medicine, 1650 Or-
assem­bly. Thus, either muta­tions in EIF6 or chro­mo­some 20q leans St, CRB1-290, Baltimore, MD 21287; e-mail: lgondek1@jhmi​
dele­tion resulting in EIF6 haploinsufficiency con­fers the indi­ ­.edu.
rect cor­rec­tion of SBDS muta­tions.26 Finally, the dupli­ca­tion of
the hypo­mor­phic SBDS allele, through the emer­gence of CH References
with iso­ chro­mo­ some 7q, may par­ tially reverse the dis­ ease 1. Steensma DP, Bejar R, Jaiswal S, et al. Clonal hema­to­poi­e­sis of inde­ter­mi­
phe­no­type.27 In con­trast, in SAMD9/SAMD9L-medi­ated bone nate poten­tial and its dis­tinc­tion from myelodysplastic syn­dromes. Blood.
mar­row fail­ure, somatic rever­sion may be accom­plished by the 2015;126(1):9-16.
2. Gondek LP, DeZern AE. Assessing clonal haematopoiesis: clin­i­cal bur­dens

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/384/1851752/384gondek.pdf by guest on 13 December 2021


loss of the mutant allele located on chro­mo­some 7q (Table 2).28 and ben­e­fits of diag­nos­ing myelodysplastic syn­drome pre­cur­sor states.
The role of CH in iBMFs is discussed in detailed by Dr. Jung in Lancet Haematol. 2020;7(1):e73-e81.
an accom­pa­ny­ing arti­cle. 3. DeZern AE, Malcovati L, Ebert BL. CHIP, CCUS, and other acro­nyms: def­i­ni­
CH is com­mon in BMFs and fre­quently rep­re­sents an adap­ta­ tion, impli­ca­tions, and impact on prac­tice. Am Soc Clin Oncol Educ Book.
2019;39(Jan­u­ary):400-410.
tion of HSCs to extra­cel­lu­lar or cell-intrin­sic stress­ors, improv­ing
4. Valent P, Bain BJ, Bennett JM, et  al. Idiopathic cytopenia of unde­ter­
stem cell fit­ness and periph­eral blood count. However, such cor­ mined sig­nif­i­cance (ICUS) and idi­o­pathic dys­pla­sia of uncer­tain sig­nif­
rec­tion may result in a higher inci­dence of hema­to­logic malig­ i­cance (IDUS), and their dis­tinc­tion from low risk MDS. Leuk Res. 2012;
nan­cies, espe­cially when involv­ing puta­tive onco­genes or tumor 36(1):1-5.
sup­pres­sors. 5. De S. Somatic mosa­i­cism in healthy human tis­sues. Trends Genet. 2011;27(6):
217-223.
6. Risques RA, Kennedy SR. Aging and the rise of somatic can­cer-asso­ci­ated
muta­tions in nor­mal tis­sues. PLoS Genet. 2018;14(1):e1007108.
7. Zink F, Stacey SN, Norddahl GL, et  al. Clonal hema­to­poi­e­sis, with and
with­ out can­ di­date driver muta­ tions, is com­mon in the elderly. Blood.
CLINICAL CASE (Con­t in­u ed) 2017;130(6):742-752.
The patient had a sin­gle hot spot muta­tion in DNMT3A with a 8. Young AL, Challen GA, Birmann BM, Druley TE. Clonal haematopoiesis
harbouring AML-asso­ci­ated muta­tions is ubiq­ui­tous in healthy adults. Nat
VAF of 2.5%. This likely rep­re­sents spo­radic CH asso­ci­ated with
Commun. 2016;7(August):12484.
aging. The patient’s normocytic normochromic and hypopro- 9. Watson CJ, Papula AL, Poon GYP, et  al. The evo­lu­tion­ary dynam­ics and
liferative ane­mia is likely related to chronic inflam­ma­tion given fit­ness land­scape of clonal hema­to­poi­e­sis. Science. 2020;367(6485):1449-
the patient’s his­tory of rheu­ma­toid arthri­tis and the ele­va­tion of 1454.
inflam­ma­tory mark­ers (fer­ri­tin). The hemogram and iron stud­ies 10. Challen GA, Goodell MA. Clonal hema­to­poi­e­sis: mech­a­nisms driv­ing dom­
i­nance of stem cell clones. Blood. 2020;136(14):1590-1598.
are highly sug­ges­tive of ane­mia of inflam­ma­tion (AI), par­tic­u­ 11. Ko M, Huang Y, Jankowska AM, et al. Impaired hydrox­yl­ation of 5-methylcy-
larly in the absence of nutri­tional deficiencies or other appar­ tosine in mye­loid can­cers with mutant TET2. Nature. 2010;468(7325):839-
ent abnor­mal­i­ties, includ­ing an unre­mark­able bone mar­row 843.
exam­i­na­tion. It appears that the ane­mia is not caused solely by 12. Li Z, Cai X, Cai CL, et  al. Deletion of Tet2 in mice leads to dysregulated
hema­to­poi­etic stem cells and sub­se­quent devel­op­ment of mye­loid malig­
an under­ly­ing CH. Additionally, the pres­ence of a minor clone
nan­cies. Blood. 2011;118(17):4509-4518.
account­ing for only 5% of the total hema­to­poi­etic out­put (het­ 13. Abdel-Wahab O, Gao J, Adli M, et al. Deletion of Asxl1 results in myelodyspla-
ero­zy­gous muta­tion with a VAF of 2.5%) would not be suf­fi­cient sia and severe devel­op­men­tal defects in vivo. J Exp Med. 2013;210(12):2641-
to have a sig­nif­i­cant impact on the hema­to­poi­etic out­put from 2659.
95% nonclonal coun­ter­parts in a cell-auton­o­mous man­ner. How- 14. Gondek LP, Zheng G, Ghiaur G, et al. Donor cell leu­ke­mia aris­ing from clonal
hema­to­poi­e­sis after bone mar­row trans­plan­ta­tion. Leukemia. 2016;30(9):
ever, the exact VAF cut­off asso­ci­ated with cytopenias is not yet 1916-1920.
established, and the val­ues are likely muta­tion-spe­cific. Of note, 15. Wong WH, Bhatt S, Trinkaus K, et al. Engraftment of rare, path­o­genic donor
DNMT3A muta­tions seem to be more fre­quent in older patients hema­to­poi­etic muta­tions in unre­lated hema­to­poi­etic stem cell trans­plan­
with AI, but the pres­ence of small clones (<5% VAF) has no ta­tion. Sci Transl Med. 2020;12(526).
16. Frick M, Chan W, Arends CM, et  al. Role of donor clonal hema­to­poi­e­
impact on clin­i­cal out­come.29 Even though the patient may be
sis in allo­ge­neic hema­to­poi­etic stem-cell trans­plan­ta­tion. J Clin Oncol.
at risk for pro­gres­sion to myelodysplastic syn­drome or acute 2019;37(5):375-385.
mye­loid leu­ke­mia, the prob­ab ­ il­ity appears to be small given the 17. Meisel M, Hinterleitner R, Pacis A, et al. Microbial sig­nals drive pre-leukae-
lack of high-risk CH fea­tures such as mul­ti­ple muta­tions, spli- mic myeloproliferation in a Tet2-defi­cient host. Nature. 2018;557(7706):580-
ceosome, IDH1/2 or TP53 muta­tions, and high VAF.30-32 Since 584.
18. Hormaechea-Agulla D, Matatall KA, Le DT, et  al. Chronic infec­tion drives
the pres­ence of CHIP is an impor­tant risk fac­tor for a major car­ Dnmt3a-loss-of-func­tion clonal hema­to­poi­e­sis via IFNγ sig­nal­ing. Cell Stem
dio­vas­cu­lar event, the patient should be coun­seled on life­style Cell. 2021;28(8):1428-1442.e61442e6.
mod­i­fi­ca­tion to address mod­i­fi­able car­dio­vas­cu­lar risk fac­tors. 19. Yoshizato T, Dumitriu B, Hosokawa K, et al. Somatic muta­tions and clonal
hema­to­poi­e­sis in aplastic ane­mia. N Engl J Med. 2015;373(1):35-47.
20. Cooper JN, Young NS. Clonality in con­text: hema­to­poi­etic clones in their
mar­row envi­ron­ment. Blood. 2017;130(22):2363-2372.
21. Babushok DV, Perdigones N, Perin JC, et al. Emergence of clonal hema­to­
Conflict-of-inter­est dis­clo­sure poi­e­sis in the major­ity of patients with acquired aplastic ane­mia. Cancer
Lukasz P. Gondek: no com­pet­ing finan­cial inter­ests to declare. Genet. 2015;208(4):115-128.

388  |  Hematology 2021  |  ASH Education Program


22. Gib­son CJ, Lindsley RC, Tchekmedyian V, et al. Clonal hema­to­poi­e­sis asso­ 28. Pastor VB, Sahoo SS, Boklan J, et  al. Constitutional SAMD9L muta­tions
ci­ated with adverse out­comes after autol­o­gous stem-cell trans­plan­ta­tion cause famil­ial myelodysplastic syn­drome and tran­sient mono­somy 7. Hae-
for lym­phoma. J Clin Oncol. 2017;35(14):1598-1605. matologica. 2018;103(3):427-437.
23. Bernard E, Nannya Y, Hasserjian RP, et al. Implications of TP53 alle­lic state 29. van Zeventer IA, de Graaf AO, Wouters HJCM, et al. Mutational spec­trum
for genome sta­bil­ity, clin­i­cal pre­sen­ta­tion and out­comes in myelodysplas- and dynam­ics of clonal hema­to­poi­e­sis in ane­mia of older indi­vid­u­als.
tic syn­dromes. Nat Med. 2020;26(10):1549-1556. Blood. 2020;135(14):1161-1170.
24. Jasek M, Gondek LP, Bejanyan N, et al. TP53 muta­tions in mye­loid malig­nan­ 30. Abelson S, Collord G, Ng SWK, et al. Prediction of acute mye­loid leu­kae­mia
cies are either homo­zy­gous or hemi­zy­gous due to copy num­ber-neu­tral risk in healthy indi­vid­u­als. Nature. 2018;559(7714):400-404.
loss of het­ero­zy­gos­ity or dele­tion of 17p. Leukemia. 2010;24(1):216-219. 31. Desai P, Mencia-Trinchant N, Savenkov O, et al. Somatic muta­tions pre­cede
25. Pasca S, Gondek LP. Clonal hema­to­poi­e­sis and bone mar­row fail­ure syn­ acute mye­loid leu­ke­mia years before diag­no­sis. Nat Med. 2018;24(7):1015-
dromes. Best Pract Res Clin Haematol. 2021;34(2):101273. 1023.
26. Kennedy AL, Myers KC, Bowman J, et al. Distinct genetic path­ways define 32. Rossi M, Meggendorfer M, Zampini M, et  al. Clinical rel­e­vance of clonal
pre-malig­nant ver­sus com­pen­sa­tory clonal hema­to­poi­e­sis in Shwachman- hema­to­poi­e­sis in the oldest-old pop­u­la­tion. Published online 14 June 2021.
Diamond syn­drome. Nat Commun. 2021;12(1):1334. Blood.
27. Minelli A, Maserati E, Nicolis E, et al. The iso­chro­mo­some i(7)(q10) car­ry­ing
c.258 + 2t>c muta­tion of the SBDS gene does not pro­mote devel­op­ment of

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/384/1851752/384gondek.pdf by guest on 13 December 2021


mye­loid malig­nan­cies in patients with Shwachman syn­drome. Leukemia. © 2021 by The Amer­i­can Society of Hematology
2009;23(4):708-711. DOI 10.1182/hema­tol­ogy.2021000270

Prakash Singh Shekhawat


Mechanism of clonal hema­to­poi­e­sis  |  389
LET THE CHIP(S) FALL WHERE THEY MAY ? BURDENS AND BENEFITS OF DIAGNOSING CLONAL HEMATOPOIESIS

Mechanisms of somatic transformation in


inherited bone marrow failure syndromes

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/390/1852164/390choijilsuren.pdf by guest on 13 December 2021


Haruna Batzorig Choijilsuren,1,2,* Yeji Park,1,* and Moonjung Jung1
1
Division of Hematology, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD; and 2Department of Molecular and
Cellular Biology, Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore, MD
*These authors contributed equally.

Inherited bone marrow failure syndromes (IBMFS) cause hematopoietic stem progenitor cell (HSPC) failure due to ger-
mline mutations. Germline mutations influence the number and fitness of HSPC by various mechanisms, for example,
abnormal ribosome biogenesis in Shwachman-Diamond syndrome and Diamond-Blackfan anemia, unresolved DNA cross-
links in Fanconi anemia, neutrophil maturation arrest in severe congenital neutropenia, and telomere shortening in short
telomere syndrome. To compensate for HSPC attrition, HSPCs are under increased replication stress to meet the need for
mature blood cells. Somatic alterations that provide full or partial recovery of functional deficit implicated in IBMFS can
confer a growth advantage. This review discusses results of recent genomic studies and illustrates our new understand-
ing of mechanisms of clonal evolution in IBMFS.

LEARNING OBJECTIVES
• Learn recent advances in our understanding of clonal hematopoiesis in IBMFS
• Recognize early signs of malignant transformation in IBMFS

Introduction ideal timing for transplant. Although there are no consen­


Inherited bone marrow failure syndromes (IBMFS) manifest as sus surveillance guidelines with a high level of evidence
ineffective and stressed hematopoiesis due to germline muta­ due to a lack of randomized controlled trials, experts gen­
tions that cause hematopoietic stem progenitor cell (HSPC) erally agree with serial monitoring of blood counts and
failure. The causes of HSPC failure are different depending on bone marrow (BM). Risk­based surveillance recommenda­
disease mechanisms: unresolved DNA cross­links in Fanconi tions are summarized in the Figure 1.
anemia (FA), telomere shortening in dyskeratosis congenita Clonal hematopoiesis (CH) is frequently seen in older
(DC) (also known as short telomere syndromes [STSs]), and individuals. Ten percent to 20% of individuals older than 70
ribosomopathy in Shwachman­Diamond syndrome (SDS) years carry somatic mutations in blood, most frequently in
and Diamond­Blackfan anemia (DBA).1 However, clinical con­ genes involved in epigenetic regulation (DNMT3A, TET2,
sequences are similar in that patients experience ineffec­ and ASXL1), which is associated with about 1% per year risk
tive hematopoiesis, leading to replicative stress and HSPC for progression to myeloid malignancies.4 Clonal hema­
exhaustion, and frequently develop malignant transformation topoiesis in IBMFS has a couple of important differences
(Table 1). compared with CH observed in older, otherwise healthy
Successful hematopoietic stem cell transplant (HSCT) individuals; it occurs more frequently and early in life. The
can cure bone marrow failure (BMF) and prevent leuke­ mutational spectrum is also quite different. This is likely
mic transformation; however, transplant outcome is poor because preexisting molecular and cellular defects due
in patients who have already developed malignant clones to germline mutations cause stem cell exhaustion at base­
before HSCT.2,3 Besides, HSCT comes with a price of poten­ line, and somatic mutations that can compensate under­
tial end­organ damage and accelerated solid tumorigene­ lying defects confer a growth advantage.5 It is currently
sis. Therefore, the major goal of surveillance is to detect not established whether the detection of clones with par­
early signs of malignant transformation and determine the ticular mutations or progressive expansion of such clones

390 | Hematology 2021 | ASH Education Program


Table 1. Inherited BMF syn­dromes

Causative genes Frequent chro­mo­ Frequent somatic


Syndrome ­(inher­i­tance) Etiology Clinical man­i­fes­ta­tions somal abnor­mal­i­ties muta­tions
SDS SBDS (AR)—m/c Ribosomopathy Hematopoietic/onco­logic: 20q– (inter­sti­tial EIF6—improve ribo­
DNAJC21 (AR) neutropenia, ane­mia, throm­ dele­tion q11.21- some bio­gen­e­sis
SRP54 (AD) bo­cy­to­pe­nia, aplastic ane­ q13.32)—loss of and pro­tein trans­la­
ELF1 (AR) mia, MDS, AML EIF6 tion effi­ciency
Nonhematologic: exo­crine pan­ i(7)(q10)—dupli­ca­tion
cre­atic insuf­fi­ciency, fail­ure of hypo­mor­phic High risk:
to thrive, mal­ab­sorp­tion, SBDS allele Biallelic TP53—
short stat­ure, skel­e­tal abnor­ impaired cell cycle
mal­i­ties (chondrodysplasia or High risk: check­point and
con­gen­i­tal tho­racic dys­ –7/7q– apo­pto­sis
Complex

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/390/1852164/390choijilsuren.pdf by guest on 13 December 2021


tro­phy), endo­crine abnor­
mal­i­ties, liver dys­func­tion,
cog­ni­tive impair­ment
FA FANCA (AR)—m/c Inability to repair Hematologic/onco­logic: 1q+ Somatic rever­sion
FANCB (XR) DNA inter­strand throm­bo­cy­to­pe­nia, ane­mia, of FANC genes
FANCC (AR) cross-links neutropenia, aplastic ane­mia, High risk: by back muta­
FANCD1/BRCA2 (AR) MDS, leu­ke­mia, squa­mous 3q+ tion, intra­genic
FANCD2 (AR) cell car­ci­noma (head and –7/7q– recom­bi­na­tion,
FANCE (AR) neck, anogenital, esoph­ Complex sec­ond site muta­
FANCF (AR) a­geal), embry­o­nal tumors Cryptic RUNX1 trans­ tions suppressing
FANCG (AR) (medul­lo­blas­toma, neu­ro­ lo­ca­tion the effect of the
FANCI (AR) blas­toma, Wilms tumor in orig­i­nal muta­tion,
FANCJ/BRIP1/BACH1 patients with biallelic BRCA2 frame-restor­ing
(AR) muta­tions) sec­ond site inser­
FANCL (AR) Nonhematopoietic: short stat­ tions and dele­tions
FANCM (AR) ure, low birth weight, fail­ure
FANCN/PALB2 (AR) to thrive, micro­ceph­aly, High risk:
FANCO/RAD51C (AR) microphthalmia, hear­ing loss, RUNX1
FANCP/SLX4 (AR) tri­an­gu­lar face, micrognathia, RAS path­way (KRAS,
FANCQ/ERCC4 (AR) car­diac abnor­mal­i­ties (pat­ent PTPN11)—(small
FANCR/RAD51 (AD) duc­tus arte­ri­o­sus, atrial sep­ num­ber of cases)
FANCS/BRCA1 (AR) tal defect, ven­tric­u­lar sep­tal
FANCT/UBE2T (AR) defect), tracheoesophageal
FANCU/XRCC2 (AR) fis­tula, esoph­a­geal atre­sia,
FANCV/REV7 (AR) horse­shoe or ectopic kid­neys,
FANCW/RFWD3 (AR) thumb/radius abnor­mal­i­ties,
hypo­plas­tic the­nar emi­nence,
clinodactyly, café-au-lait
spots, hypo/hyper­pig­men­ta­
tion of the skin
SCN ELANE (AD)—m/c Neutrophil mat­u­ Hematologic/onco­logic: severe High risk: CSF3R—enhanced
HAX1 (AR) ra­tion arrest and neutropenia at birth, recur­ –7/7q– and prolonged
GFI1 (AD) apo­pto­sis rent infec­tions, MDS, AML Complex response to G-CSF
G6PC3 (AR)
JAGN1 (AR) High risk:
TCIRG1 (AD) RUNX1
WAS (XR)
CSF3R (AD, AR)
Short telo­mere TERT (AD, AR)—m/c Telomere short­en­ing Hematologic/onco­logic: High risk: Somatic rever­sion—
syn­drome TERC (AD) throm­bo­cy­to­pe­nia, ane­mia, –7/7q– back muta­tion
DKC1 (XR) neutropenia, pan­cy­to­pe­ Complex of DKC1, TERT
NOLA3/NOP10 (AR) nia, MDS, AML, immu­no­ pro­moter GOF
NOLA2/NHP2 (AR) de­fi­ciency, squa­mous cell muta­tion, mitotic
TINF2 (AD) car­ci­noma recom­bi­na­tion
WRAP53/TCAB1 (AR) Nonhematopoietic: the muco­
CTC1 (AR) cu­ta­ne­ous triad (retic­u­lated High risk:
RTEL1 (AD, AR) skin ­pig­men­ta­tion, nail dys­ TP53a
ACD/TPP1 (AD, AR) tro­phy, oral leukoplakia), short
PARN (AD, AR) stat­ure, low birth weight,
NAF1 (AD) fail­ure to thrive, hear­ing loss,
STN1 (AD) ret­i­nop­a­thy, muco­sal stric­
NPM1 (AD) tures, pul­mo­nary fibro­sis, liver
ZCCHC8 (AD) fibro­sis

Prakash Singh Shekhawat


Clonal hema­to­poi­e­sis in IBMFS  |  391
Table 1. Inherited BMF syndromes (Continued)

Causative genes Frequent chro­mo­ Frequent somatic


Syndrome ­(inher­i­tance) Etiology Clinical man­i­fes­ta­tions somal abnor­mal­i­ties muta­tions
DBA RPS19 (AD)—m/c Ribosomopathy; red Hematologic/onco­logic: Abnormal cyto­ge­net­ TP53,a PPM1D, ASXL1
RPL3 (AD) blood cell aplasia Anemia, reticulocytopenia, ics infre­quent (small num­ber of
RPL5 (AD) MDS, AML, solid tumors cases)b
RPL7 (AD) (oste­o­genic sar­coma, lung,
RPL11 (AD) colon and cer­vix)
RPL14 (AD) Nonhematologic: low birth
RPL15 (AD) weight, growth retar­da­tion,
RPL18 (AD) devel­op­men­tal delay, short
RPL19 (AD) stat­ure, micro­ceph­aly, other
RPL23 (AD) cra­nio­fa­cial malformation,
RPL26 (AD) con­gen­i­tal glau­coma or cat­

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/390/1852164/390choijilsuren.pdf by guest on 13 December 2021


RPL27 (AD) a­ract, stra­bis­mus, short neck,
RPL31 (AD) thumb abnor­mal­i­ties, horse­
RPL35a (AD) shoe kid­ney, hypo­spa­dias,
RPL36 (AD) car­diac abnor­mal­i­ties
PRS7 (AD)
RPS8 (AD)
RPS10 (AD)
RPS15 (AD)
RPS17 (AD)
RPS24 (AD)
RPS26 (AD)
RPS27 (AD)
RPS27A (AD)
RPS28 (AD)
RPS29 (AD)
GATA1 (XR)
TSR2 (XR)
SAMD9/SAMD9L SAMD9 (AD) Growth inhi­bi­tion MIRAGE syn­drome –7/7q– Somatic rever­sion
dis­or­ders SAMD9L (AD) Hematologic/onco­logic: 5q– by LOF SAMD9 or
Thrombocytopenia, ane­mia, SAMD9L muta­tions
MDS with mono­somy 7, ETV6, RUNX1, SETBP1
recur­rent infec­tions, (small num­ber of
bleed­ing cases)
Nonhematologic: intra­uter­ine
growth restric­tion, devel­
op­men­tal delay, adre­nal
hypo­pla­sia, chronic diar­rhea,
gen­i­tal anom­a­lies
Ataxia-pan­cy­to­pe­nia syn­drome
Hematologic/onco­logic:
Pancytopenia, ane­mia, MDS
with mono­somy 7/del(7q),
AML, recur­rent infec­tions
Nonhematologic: ataxia, cer­
e­bel­lar atro­phy, ret­i­nal dys­
func­tion, behav­ioral abnor­
mal­i­ties, alve­o­lar proteinosis
The dif­fer­en­tial effects of biallelic vs het­ero­zy­gous TP53 muta­tions to leu­ke­mic trans­for­ma­tion have not been stud­ied in IBMFS other than SDS.
a

Whether somatic muta­tions con­trib­ute to malig­nant trans­for­ma­tion in DBA has not been fully inves­ti­gated.
b

m/c, most com­mon; AD, auto­so­mal dom­i­nant; AR, auto­so­mal reces­sive; GOF, gain of func­tion; XR, X-linked reces­sive.

war­rants empir­ic
­ al treat­ments before frank cytopenias or myel­ recov­ery of the affected pro­tein. It has been reported in many
odysplastic syn­ dromes (MDS)/acute mye­ loid leu­ke­
mia (AML) inherited genetic dis­ or­
ders, and poten­ tial mech­ an­isms have
arise. For exam­ple, CSF3R muta­tions in severe con­gen­i­tal neutro­ been reported as back muta­tion, intra­genic recom­bi­na­tion, sec­
penia (SCN) or het­ero­zy­gous TP53 muta­tions in SDS can per­sist ond site muta­tions suppressing the effect of the orig­in ­ al muta­
for years with­out pro­gres­sion to malig­nancy, even in the set­ting tion, frame-restor­ing sec­ond site inser­tions, and dele­tions.6 It
of mul­ti­ple muta­tions in a given patient. Determination of ideal can con­trib­ute to CH by con­fer­ring sur­vival ben­e­fit to reverted
tim­ing for HSCT in the set­ting of CH in IBMFS requires fur­ther clones, but the expan­sion of hema­to­poi­etic clones is not always
stud­ies. asso­ci­ated with higher pro­pen­sity for leu­ke­mic trans­for­ma­
Somatic rever­ sion is the acquired alter­ ation of inherited tion. This may be due to the over­all improved fit­ness of HSPC
path­o­genic muta­tion, resulting in the par­tial or full func­tional by correcting under­ly­ing func­tional defects while keep­ing the

392  |  Hematology 2021  |  ASH Education Program


At diagnosis
- CBC
- BM with cytogenetics/FISH, NGS myeloid panel
- germline genetic testing
- chromosome breakage test for FA
- telomere length (flow-FISH) for STS
- red cell adenosine deaminase for DBA
- pancreatic isoamlyase and/or trypsinogen for SDS
- HLA typing

Risk Stratification for Surveillance

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/390/1852164/390choijilsuren.pdf by guest on 13 December 2021


Low-risk patients Intermediate-risk patients High-risk patients
- stable and mild cytopenias - mild or moderate cytopenias, but stable - falling or increasing counts
- transfusion independent - transfusion independent - severe cytopenias
- no BM dysplasia - no BM dysplasia - transfusion dependent
- no cytogenetic abnormalities - non-high-risk cytogenetic abnormalities - BM dysplasia or increased blasts
- high-risk cytogenetics/FISH (del7q/-7, complex;
RUNX1 translocation or +3q in FA)
- biallelic TP53 # or RUNX1 mutations

CBC every 3-6 months CBC every 1-3 months


BM every 1 year BM every 3-6 months
NGS panel every 1-3 years* NGS panel every 1 year*
Refer to HSCT for advanced planning CBC every 1-2 months
Immediate BM and repeat every 3-6 months
HSCT with the best available donor

Figure 1. Surveillance recommendations for patients with IBMFS based on the risk of malignant transformation. Detailed diagnostic
workup for BMF is described by DeZern and Churpek.7 Once diagnosed, patients are followed by CBC with differential, BM aspiration
and biopsy with cytogenetics, FISH, and NGS myeloid panel at regular intervals. The frequency of follow-up depends on the disease
and mutation types, baseline counts and BM findings, and the presence of interval changes on subsequent visits. Although the NGS
myeloid panel is not a part of standard practice for the management of IBMFS, it is becoming more widely used with advancement of
our understanding in CH in both IBMFS and the general population. NGS myeloid panel results are particularly useful to select patients
who are at high risk of malignant transformation without overt clinical signs of disease progression. We can increase the frequency
of surveillance and initiate discussion and planning for early intervention for the best possible outcome in these high-risk patients.
*The optimal frequency and the best genomic source (PB vs BM) of NGS myeloid panel testing have not been fully established.
#
Although biallelic TP53 mutations are known high-risk features, conventional NGS techniques do not readily provide allelic status of a
given mutation. The severity of BMF is defined as the following: mild—ANC <1500/µL, hemoglobin ≥8 g/dL, or platelets 50-150 K/µL;
moderate—ANC <1000/µL, hemoglobin <8 g/dL, or platelets <50 K/µL; severe—ANC <500/µL, hemoglobin <8 g/dL, or platelets
<30 K/µL. ANC, absolute neutrophil count; CBC, complete blood count; FISH, fluorescence in situ hybridization; PB, peripheral blood.

cell cycle check­point intact, most impor­tantly p53, which can in the mye­ loid lin­
e­age, but no frank dys­ pla­
sia is seen and
actively sup­press tumor­i­gen­e­sis. blasts are not increased. Cytogenetics showed del(20q) in 2 of
In this review, the term CH will be used to describe a phe­nom­ 20 cells. Peripheral blood mye­loid neo­plasm panel sequenc­ing
e­non where any HSPC clone with somatic geno­mic alter­ations— result reveals 2 muta­tions in the EIF6 gene: D112N (var­i­ant alle­lic
includ­ing sin­gle-nucle­o­tide var­i­ants, indels, large dele­tions, or fre­quency [VAF] 5%) and N106S (VAF 2%) and 2 muta­tions in the
gross chro­mo­somal abnor­mal­i­ties—con­trib­utes to a larger pool TP53 gene: R248W (VAF 1%) and R110L (VAF 0.4%). The hema­
of mature blood cells than what is expected for any sin­gle HSPC tol­o­gist decides to mon­i­tor his counts every 3 months, as well
clone under homeo­sta­sis. as BM every 3 to 6 months, and repeat blood mye­loid neo­plasm
panel sequenc­ing in 1 year to mon­i­tor the TP53 mutant clones.

CLINICAL CASE Shwachman-Diamond syn­drome


A 21-year-old man with SDS presented to clinic for a 6-month SDS is char­ac­ter­ized by con­gen­i­tal anom­a­lies; BMF, with neutro­
fol­low-up. The patient has mild neutropenia and mild throm­ penia being the most prominent fea­ture; and exo­crine pan­cre­
bo­cy­to­pe­nia, and blood counts have been sta­ble in the past atic insuf­fi­ciency.8 Biallelic muta­tions in the SBDS gene are most
6 months. The BM is hypocellular with mild dysmorphologies com­mon (>90%). Less fre­quently, muta­tions in DNAJC21, SRP54,

Prakash Singh Shekhawat


Clonal hema­to­poi­e­sis in IBMFS  |  393
Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/390/1852164/390choijilsuren.pdf by guest on 13 December 2021
Figure 2. Clonal hematopoiesis in IBMFS. Underlying genetic mutations lead to ineffective and stressed hematopoiesis, and u ­ ltimately
HSPC attrition. Genetic variations occurred during lifetime of HSPC may confer a growth advantage if it corrected underlying genetic
and/or cellular defects by somatic reversion. Alternatively, bypassing cell cycle check point by acquiring high risk genetic features,
such as loss of p53, may confer a growth advantage. Clones that reverted underlying defects may have competitive advantage and
higher fitness, resulting in improved counts and stable hematologic course. Whereas clones with high risk somatic alterations (eg,
TP53 mutations or monosomy 7) may result in clonal expansion without functional recovery of underlying defects. These clones are
more prone to acquire additional secondary mutations and/or progress to MDS/AML.

and EFL1 pres­ent with clin­i­cal fea­tures of SDS. About 20% to 30% abnor­mal­ity was also del(20q) (47%), which is the loca­tion for
of patients develop MDS/AML, which is fre­quently accom­pa­nied the EIF6 gene. In another study, CH was observed in 59% of
by com­plex kar­yo­type.2,9-11 patients, and TP53 muta­tions were the most fre­quent (48%) (of
A recent sequenc­ing study shed light on our under­stand­ing note, EIF6 was not included in the gene panel in this study).13 The
of the mech­a­nisms of clonal evo­lu­tion in SDS. Clonal hema­to­poi­ prev­a­lence of TP53 muta­tions increased with age; about 80%
e­sis was com­mon in patients with SDS (72% among those with­ of patients older than 10 years had at least one TP53 muta­tion,
out frank MDS/AML).12 Interestingly, mutated genes in SDS were and fre­quently two or more TP53 mutations were dis­cov­ered in
dif­fer­ent from those of the gen­eral pop­u­la­tion. Mutations in EIF6 a given patient. The pres­ence of TP53 muta­tions among patients
were the most com­mon (59%), followed by TP53 (39.8%) and with SDS is not an impending sign of leu­ke­mic trans­for­ma­tion,
CSNK1A1 (7.2%), whereas muta­tions in DNMT3A, TET2, or ASXL1 even though TP53 muta­ tions are fre­
quently encoun­ tered in
were less fre­quent. The most com­monly observed chro­mo­somal MDS/AML and por­tend a poor prog­no­sis.

394  |  Hematology 2021  |  ASH Education Program


The SBDS pro­tein coop­er­ates with the GTPase EFL1 to release is also highly char­ac­ter­is­tic in FA and fre­quently pre­cedes mono­
EIF6 from the nascent 60S ribo­somal sub­unit in the cyto­sol, so somy 7/del(7q).24-26 RUNX1 abnor­mal­i­ties, includ­ing cryp­tic trans­
that the 60S sub­unit is free to asso­ci­ate with the 40S sub­unit and lo­ca­tion, also indi­cate high-risk of trans­for­ma­tion.23
form the mature 80S ribo­some.14 Therefore, the loss-of-func­tion Single-nucle­o­tide poly­mor­phism array anal­y­sis is one of the
(LOF) EIF6 var­i­ants would result in increased avail­abil­ity of the 60S non­in­va­sive mon­i­tor­ing meth­ods that can iden­tify chro­mo­somal
sub­unit for mat­u­ra­tion and com­pen­sate for the defec­tive SBDS. abnor­mal­i­ties from blood sam­ples. From 130 patients with FA
In fact, EIF6 var­i­ants were fre­quently found in patients with SDS, (mean age, 14.5 years; range, 0-50 years), sin­gle-nucle­o­tide poly­
which was asso­ci­ated with clonal expan­sion due to improved mor­phism array detected chro­mo­somal mosaic events (CMEs) in
trans­ la­
tional effi­
ciency and stem cell fit­ ness. While func­ tional 16 patients (12.3%).27 Nine of 16 patients car­ried mul­ti­ple CMEs.
com­pen­sa­tion for pri­mary ribo­somal defects improved sur­vival of The pres­ence of CMEs was asso­ci­ated with higher inci­dence of
HSPC in SDS, it did not nec­es­sar­ily increase the risk of malig­nant hema­to­logic and solid can­cers, as well as increased risk of death.
trans­for­ma­tion. On the other hand, biallelic TP53 muta­tions were Large-scale next gen­er­a­tion sequenc­ing (NGS) stud­ies have
almost always observed in patients with leu­ke­mic trans­for­ma­ not been published in FA; there­fore, it is dif­fi­cult to spec­u­late the

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/390/1852164/390choijilsuren.pdf by guest on 13 December 2021


tion. P53 is a tran­scrip­tion fac­tor that medi­ates cell cycle arrest con­tri­bu­tion of somatic muta­tions to clonal evo­lu­tion. Prior stud­
and apo­pto­sis in response to cel­lu­lar stress sig­nals. Biallelic LOF ies using Sanger sequenc­ ing of selected onco­ genes showed
muta­tions in TP53 allow HSPCs to prog­ress through cell cycle largely neg­a­tive results, but that may be due to the small num­
and escape apo­pto­sis even in the pres­ence of crit­i­cal cel­lu­lar ber of genes included in the panel and Sanger sequenc­ing not
stresses posed by ribosomopathy, thereby con­fer­ring a growth being sen­si­tive enough.23
advan­tage. Single-cell DNA sequenc­ing on serial sam­ples from More recently, targeted mye­loid panel sequenc­ing performed
patients who devel­oped AML revealed that patients can have on 16 patients with FA with MDS/AML revealed 10 patients had
mul­ti­ple inde­pen­dent TP53 mutant clones for a long time, and detect­able somatic muta­tions, most fre­quently involv­ing RUNX1,
the par­tic­u­lar clone car­ry­ing biallelic muta­tions can give rise to and the RAS path­ way genes (KRAS and PTPN11).28 Of these
leu­ke­mia over sev­eral years.12 The pres­ence of biallelic muta­tions 16 patients, 12 patients had either chro­mo­some 3 or 7 abnor­
usu­ally can­not be assessed in the con­ven­tional bulk sequenc­ing, mal­i­ties and/or RUNX1 muta­tions, confirming the prior reports.
which only informs VAF but not alle­lic sta­tus of a given muta­tion Another NGS study of 7 patients with FA (includ­ing 1 with AML)
in cells. This raises a ques­tion whether performing sin­gle-cell DNA also con­firmed fre­quent CH from a young age, vir­tu­ally observed
sequenc­ing reg­u­larly on patients with known TP53 mutant clones in all­patients tested, regard­less of the pres­ence of periph­eral
can help select patients with biallelic TP53 muta­tions who are at blood cytopenia or leu­ke­mia.29 One AML sam­ple in that study
risk of leu­ke­mic trans­for­ma­tion. Of course, this expen­sive mon­i­ car­ried mul­ti­ple MDS/AML muta­tions, includ­ing TP53. Although
tor­ing approach will need to be care­fully assessed in clin­i­cal tri­als a clear pat­tern of clonal evo­lu­tion can­not be deter­mined from
to con­firm effi­cacy and cost-effec­tive­ness. these stud­ ies due to small sam­ ple sizes, a rel­ a­
tive lack of
DBA is another inherited ribosomopathy due to het­ero­zy­gous DNMT3A, TET2, and SF3B1 was observed in both stud­ies, sug­
muta­tions in the com­po­nents of either the small 40S or large 60S gesting the dif­fer­ent somatic muta­tional spec­trum in FA.
ribo­somal sub­units.8 It is char­ac­ter­ized by red blood cell aplasia Approximately 15% to 18% of patients with FA are reported
and con­gen­i­tal anom­aly. The risk of MDS/AML is rel­a­tively low to have somatic rever­sion. It fre­quently results in CH, because
com­pared with other IBMFS discussed here, with a cumu­la­tive recov­ery of the pro­tein func­tion often con­fers a growth advan­
inci­dence of AML of 5% by the age of 46 years.15 The muta­tional tage. It can result in improved counts and sta­ble hema­to­logic
spec­trum lead­ing to MDS/AML has not been sys­tem­at­i­cally or course, with lon­ger sur­vival, lower risk of MDS/AML, and less like­
func­ tion­ally stud­ied in DBA. Somatic muta­ tions in TP53 and li­hood to require HSCT.30-32 However, malig­nant trans­for­ma­tion
PPM1D in MDS have been reported in spo­radic case reports,16-18 in coexisting nonrevertant FA cells may still occur, requir­ing reg­
which raises a pos­ si­
bil­
ity that sim­ i­
lar mech­ a­
nisms may drive u­lar monitoring.30
malig­nant trans­for­ma­tion in DBA and SDS.
Severe con­gen­i­tal neutropenia
Fanconi ane­mia SCN is char­ac­ter­ized by severe neutropenia from birth, com­pli­
FA is the most com­mon IBMFS. At least 22 genes have been iden­ cated by recur­rent infec­tions. Treatment with granulocyte colony-
ti­fied (FANCA-FANCW).19,20 The under­ly­ing cel­lu­lar defect is the stimulating factor (G-CSF) can increase the cir­cu­lat­ing neu­tro­phil
inabil­ity to repair DNA cross-links, lead­ing to apo­pto­sis via the counts and is effec­tive for pre­ven­tion of recur­rent infec­tions. The
p53 path­way, or trans­for­ma­tion if p53 is inactivated.21 Patients most com­mon cause is the ELANE gene muta­tions (∼50%), fol­
have con­gen­i­tal anom­a­lies, BMF, and can­cer pre­dis­po­si­tion, lowed by HAX1 and G6PC3 (10%-20%).13
most nota­bly squa­mous cell car­ci­noma of the head and neck.20 About 20% of patients tak­ing G-CSF develop MDS/AML in
Patients have a 30% to 40% cumu­la­tive risk of mye­loid malig­ their life­time.33,34 Somatic muta­ tions in CSF3R (encoding the
nan­cies by age 40 years.9,22 Myeloid malig­nan­cies often arise G-CSF recep­tor) and RUNX1 have been fre­quently observed in
from clones with abnor­mal kar­yo­types, char­ac­ter­is­ti­cally unbal­ those who devel­oped MDS/AML.35 From a targeted sequenc­ing
anced trans­lo­ca­tions lead­ing to gains or losses of chro­mo­somes. study of 40 patients, CH was fre­quently observed (62%; mean
Gain of chro­mo­some 1q is the most com­mon kar­yo­typic abnor­ age, 16.6 years).13 As expected, the most fre­quently observed
mal­ity in FA, which can be seen with­out MDS/AML and does not muta­tion was the CSF3R muta­tion (40%).13 All CSF3R var­i­ants
indi­cate impending trans­for­ma­tion.23 Conversely, mono­ somy were non­sense muta­tions resulting in trun­ca­tion of cyto­plas­mic
7/del(7q) are signs of impending trans­ for­
ma­tion, if not trans­ domain of the G-CSF recep­tor, lead­ing to prolonged and hyper­
formed already, which requires urgent HSCT. Gain of chro­mo­ ac­tive sig­nal­ing in response to G-CSF. Therefore, HSPCs car­ry­ing
some 3q, which is asso­ci­ated with increased expres­sion of EVI1, CSF3R muta­tions have a growth advan­tage in the pres­ence of
Prakash Singh Shekhawat
Clonal hema­to­poi­e­sis in IBMFS  |  395
G-CSF. Independent CSF3R-mutant clones can coex­ist, expand paralogs on the chro­mo­some 7q21.2. Both SAMD9 and SAMD9L
over many years, or some­times dis­ap­pear. pro­teins are endosome fusion facil­i­ta­tors, and they play a role
Another very fre­quently observed muta­tion in MDS/AML was in deg­ra­da­tion of growth fac­tor recep­tors by inter­nal­i­za­tion.
the RUNX1 muta­tion (64.5%), and in fact, approx­i­ma­tely 80% of Gain-of-func­tion muta­tions in SAMD9 or SAMD9L have a growth-
them also had the con­cur­rent CSF3R muta­tions.33 Serial eval­u­ inhib­i­tory effect by reduc­ing Raf/MEK/ERK sig­nal­ing, and loss
a­tion of a patient who devel­oped AML with CSF3R and RUNX1 of mutated allele con­ fers growth advan­ tage. Mutant allele is
muta­tions sug­gests that the CSF3R muta­tions were likely ini­ti­ lost fre­quently by acquired chro­mo­some 7 loss (mono­somy 7 or
at­ing events con­fer­ring a pro­lif­er­a­tive advan­tage, and an addi­ del(7q)) or con­com­i­tant somatic LOF muta­tions.47,48
tional RUNX1 muta­tion later in the dis­ease pro­gres­sion led to a
block in dif­fer­en­ti­a­tion and fueled leu­ke­mo­gen­e­sis. Conclusion
Clonal hema­ to­
poi­e­sis is com­
monly observed in patients with
Short telo­mere syn­drome IBMFS at a much youn­ger age than in the gen­eral pop­u­la­tion. The
Telomere length is reg­ u­lated by coop­ er­

tion of telomerases muta­tional spec­trum is also dif­fer­ent in IBMFS: epi­ge­netic reg­u­la­

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/390/1852164/390choijilsuren.pdf by guest on 13 December 2021


and telomeric pro­ tein complexes (shelterin and CST [CTC1- tors, such as DNMT3A, TET2, and ASXL1, the most fre­quently mu­
STN1-TEN1]).36 Germline muta­tions in telo­mere com­po­nents may tated genes in the aging pop­u­la­tion, are rel­a­tively rare—except
result in short telo­mere length, which is asso­ci­ated with BMF, in the older patients with STS. Instead, somatic geno­mic alter­
hepatic and pul­mo­nary fibro­sis, and can­cer pre­dis­po­si­tion.37 ations that can restore the orig­i­nal func­tion of pro­tein (eg, somat­
The telo­meres are short­ened on each cell divi­sion due to an ic rever­sion of the caus­a­tive muta­tion or com­pen­sa­tory sec­ond
“end-rep­li­ca­tion prob­lem,” where 5′ to 3′ DNA poly­mer­ases can muta­tions) or that allow path­o­logic adap­ta­tion for sur­vival (eg,
only extend the DNA strand in one direc­tion in the pres­ence of a by inac­ti­va­tion of p53 or loss of mutant allele) are pref­er­en­tially
tem­plate sequence, leav­ing the extreme 5′ end of the chro­mo­ selected for clonal expan­sion (Figure 2). Reduced HSPC num­
some as a gap in rep­li­ca­tion.38 Critically short telo­mere lengths, bers and fit­ness at base­line also cre­ate an envi­ron­ment in which
below the first per­cen­tile for age-matched con­trols, cause cel­ clones with such genetic alter­ations out­com­pete other HSPCs
lu­lar senes­cence and pre­ma­ture cell death, as well as a clin­i­cal and quickly estab­lish clonal dom­i­nance. However, based on cur­
syn­drome known as STS (also known as DC or telo­mere biol­ogy rently avail­­able evi­dence, CH itself, unless accom­pa­nied by high-
dis­or­der). Patients exhibit a wide spec­trum of dis­ease man­i­fes­ta­ risk cyto­ge­netic abnor­mal­i­ties, pro­gres­sive cytopenias, and/or
tions, from clas­si­cal DC with a muco­cu­ta­ne­ous triad (oral leuko­ BM mor­pho­logic changes, does not require imme­di­ate defin­i­tive
plakia, lacy retic­u­lar skin hyper­pig­men­ta­tion, and nail dys­tro­phy) treat­ments even in the set­ting of IBMFS (Table 1). Future stud­
and early onset BMF to adult-onset MDS/AML, pul­mo­nary fibro­ ies should address whether pre­emp­tive inter­ven­tions based on
sis, or liver dis­ease with­out appar­ent BMF.39 high-risk CH fea­tures improve patient out­come in clin­i­cal tri­als.
All MDS/AML cases in STS were asso­ci­ated with abnor­mal
kar­yo­types, most com­monly mono­somy 7 (56%).40 The somatic
muta­ tional land­scapes in these patients (n  = 
14; median age
CLINICAL CASE (Con­tin­ued)
53 years) were sim­i­lar to that of MDS/AML in the gen­eral pop­
u­la­tion. In patients with­out MDS/AML (median age, 59 years), This patient car­ries the 2 most com­mon somatic muta­tions in
6 of 20 eval­ua ­ ted patients had CH (30%) with the mean VAF of SDS, EIF6 and TP53, and these muta­tions are likely aris­ing from
8.2% (range, 2.2%-32.9%). Mutations in TP53 (n = 3) were most inde­pen­dent clones. Deletion 20q is also a com­mon cyto­ge­
com­monly observed, followed by TET2 (n = 2).40 On the other netic find­ing that results in the loss of EIF6. The pres­ence of TP53
hand, 2 other stud­ies with slightly youn­ger indi­vid­u­als reported muta­tions is a high-risk fea­ture; how­ever, given the small clone
that somatic muta­tions asso­ci­ated with MDS/AML were rarely sizes and sta­ble blood counts and BM mor­phol­ogy, impending
observed, whereas CH was com­mon.41,42 Whether the pres­ence leu­ke­mic trans­for­ma­tion is unlikely. Although there is no avail­­able
of somatic muta­tions dis­cov­ered by NGS is asso­ci­ated with an con­sen­sus guide­line, more fre­quent fol­low-up with blood counts
increased risk of malig­nant trans­for­ma­tion, with­out other coex­ and BM exam­i­na­tion along with serial NGS panel sequenc­ing is
isting high-risk clinical features, is not known at this time. desir­able for early detec­tion of dis­ease pro­gres­sion.
Somatic rever­sion of the telomerase activ­ity in the periph­eral
blood has been reported. Examples include acquired gain-of-
func­tion muta­tions in the TERT pro­moter region in patients with Acknowledgments 
the TERT or PARN het­ero­zy­gous muta­tions, lead­ing to overex­ This work was supported in part by National Heart Lung and
pression of the wild-type allele.43 A back muta­tion in DKC142 and Blood Institute grant K99 HL150628 (M.J.). Authors thank Dr. Robert
mitotic recom­bi­na­tion of the TERC gene resulting in isodisomy Brodsky and Dr. Amy DeZern for their constructive feedback.
of the wild-type allele in patients with germline het­ero­zy­gous
TERC muta­tions have also been reported.44 These somatic rever­ Conflict-of-inter­est dis­clo­sure
sion events can sta­bi­lize the telo­mere lengths and allow clonal Haruna Batzorig Choijilsuren: no con­flicts to dis­close.
expan­sion of the reverted clones. Yeji Park: no con­flicts to dis­close.
Moonjung Jung: no con­flicts to dis­close.
SAMD9/SAMD9L dis­or­ders
Germline muta­tions in SAMD9 and SAMD9L cause MIRAGE (myel­ Off-label drug use
odysplasia, infec­tion, restric­tion of growth, adre­nal hypo­pla­sia, Haruna Batzorig Choijilsuren: nothing to disclose.
gen­i­tal phe­no­types, and enter­op­a­thy) syn­drome and ataxia pan­ Yeji Park: nothing to disclose.
cy­to­pe­nia syn­drome, respec­tively.45,46 SAMD9 and SAMD9L are Moonjung Jung: nothing to disclose.

396  |  Hematology 2021  |  ASH Education Program


Correspondence 22. Kutler DI, Singh B, Satagopan J, et  al. A 20-year per­spec­tive on the
Moonjung Jung, Division of Hematology, Department of Medi­ International Fanconi Anemia Registry (IFAR). Blood. 2003;101(4):1249-
1256.
cine, School of Medicine, Johns Hopkins University, 720 Rutland 23. Quentin S, Cuccuini W, Ceccaldi R, et al. Myelodysplasia and leu­ke­mia of
Ave, Ross Research Building Room 1032B, Baltimore, MD 21205; Fanconi ane­mia are asso­ci­ated with a spe­cific pat­tern of geno­mic abnor­
e-mail: mjung@jhmi​­.edu. mal­i­ties that includes cryp­tic RUNX1/AML1 lesions. Blood. 2011;117(15):
e161–e170.
24. Tönnies H, Huber S, Kuhl J-S, Gerlach A, Ebell W, Neitzel H. Clonal chro­mo­
References somal aber­ra­tions in bone mar­row cells of Fanconi ane­mia patients: gains
1. Shimamura A. Aplastic ane­mia and clonal evo­lu­tion: germ line and somatic of the chro­mo­somal seg­ment 3q26q29 as an adverse risk fac­tor. Blood.
genet­ics. Hematology Am Soc Hematol Educ Program. 2016;2016(1): 2003;101(10):3872-3874.
74-82. 25. Cioc AM, Wagner JE, MacMillan ML, DeFor T, Hirsch B. Diagnosis of myel­
2. Myers KC, Furutani E, Weller E, et  al. Clinical fea­tures and out­comes of odysplastic syn­drome among a cohort of 119 patients with Fanconi ane­
patients with Shwachman-Diamond syn­drome and myelodysplastic syn­ mia: mor­pho­logic and cyto­ge­netic char­ac­ter­is­tics. Am J Clin Pathol.
drome or acute mye­loid leu­kae­mia: a multicentre, ret­ro­spec­tive, cohort 2010;133(1):92-100.
study. Lancet Haematol. 2020;7(3):e238–e246. 26. Meyer S, Bristow C, Wappett M, et al. Fanconi ane­mia (FA)-asso­ci­ated 3q

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/390/1852164/390choijilsuren.pdf by guest on 13 December 2021


3. Giardino S, de Latour RP, Aljurf M, et al; Severe Aplastic Anemia and Chronic gains in leu­ke­mic trans­for­ma­tion con­sis­tently tar­get EVI1, but do not affect
Malignancies Working Parties of Euro­pean Blood and Marrow Transplanta­ low TERC expres­sion in FA. Blood. 2011;117(22):6047-6050.
tion group. Outcome of patients with Fanconi ane­mia devel­op­ing myel­ 27. Reina-Castillón J, Pujol R, López-Sánchez M, et al. Detectable clonal mosa­
odysplasia and acute leu­ke­mia who received allo­ge­neic hema­to­poi­etic i­cism in blood as a bio­marker of can­cer risk in Fanconi ane­mia. Blood Adv.
stem cell trans­plan­ta­tion: a ret­ro­spec­tive anal­y­sis on behalf of EBMT group. 2017;1(5):319-329.
Am J Hematol. 2020;95(7):809-816. 28. Chao MM, Thomay K, Goehring G, et al. Mutational spec­trum of Fanconi
4. Jaiswal S, Ebert BL. Clonal hema­to­poi­e­sis in human aging and dis­ease. Sci- ane­mia asso­ci­ated mye­loid neo­plasms. Klin Padiatr. 2017;229(6):329-
ence. 2019;366(6465):eaan4673. 334.
5. Tsai FD, Lindsley RC. Clonal hema­to­poi­e­sis in the inherited bone mar­row 29. Heidemann S, Bursic B, Zandi S, et al. Cellular and molec­u­lar archi­tec­ture
fail­ure syn­dromes. Blood. 2020;136(14):1615-1622. of hema­to­poi­etic stem cells and pro­gen­i­tors in genetic mod­els of bone
6. Hirschhorn R. In vivo rever­sion to nor­mal of inherited muta­tions in humans. mar­row fail­ure. JCI Insight. 2020;5(4):e131018.
J Med Genet. 2003;40(10):721-728. 30. Gregory JJ Jr, Wagner JE, Verlander PC, et al. Somatic mosa­i­cism in Fanconi
7. DeZern AE, Churpek JE. Approach to the diagnosis of aplastic anemia. ane­mia: evi­dence of geno­typic rever­sion in lymphohematopoietic stem
Blood Adv. 2021;5(12):2660-2671. cells. Proc Natl Acad Sci U S A. 2001;98(5):2532-2537.
8. Shimamura A, Alter BP. Pathophysiology and man­age­ment of inherited 31. Soulier J, Leblanc T, Larghero J, et al. Detection of somatic mosa­i­cism and
bone mar­row fail­ure syn­dromes. Blood Rev. 2010;24(3):101-122. clas­si­fi­ca­tion of Fanconi ane­mia patients by anal­y­sis of the FA/BRCA path­
9. Alter BP, Giri N, Savage SA, Rosenberg PS. Cancer in the National Cancer way. Blood. 2005;105(3):1329-1336.
Institute inherited bone mar­row fail­ure syn­drome cohort after fif­teen years 32. Ramírez MJ, Pujol R, Trujillo-Quintero JP, et  al. Natural gene ther­apy by
of fol­low-up. Haematologica. 2018;103(1):30-39. reverse mosa­ i­
cism leads to improved hema­ tol­
ogy in Fanconi ane­ mia
10. Cada M, Segbefia CI, Klaassen R, et al. The impact of cat­e­gory, cytopathol­ patients. Am J Hematol. 2021;96(8):989-999.
ogy and cyto­ge­net­ics on devel­op­ment and pro­gres­sion of clonal and malig­ 33. Skokowa J, Steinemann D, Katsman-Kuipers JE, et  al. Cooperativity
nant mye­loid trans­for­ma­tion in inherited bone mar­row fail­ure syn­dromes. of RUNX1 and CSF3R muta­tions in severe con­gen­i­tal neutropenia: a
Haematologica. 2015;100(5):633-642. unique path­way in mye­loid leu­ke­mo­gen­e­sis. Blood. 2014;123(14):2229-
11. Donadieu J, Fenneteau O, Beaupain B, et al; Associated inves­ti­ga­tors of 2237.
the French Severe Chronic Neutropenia Registry. Classification of and 34. Rosenberg PS, Zeidler C, Bolyard AA, et al. Stable long-term risk of leu­kae­mia
risk fac­ tors for hema­ to­
logic com­ pli­
ca­tions in a French national cohort in patients with severe con­gen­i­tal neutropenia maintained on G-CSF ther­
of 102 patients with Shwachman-Diamond syn­ drome. Haematologica. apy. Br J Haematol. 2010;150(2):196-199.
2012;97(9):1312-1319. 35. Link DC. Mechanisms of leu­ke­mic trans­for­ma­tion in con­gen­i­tal neutrope­
12. Kennedy AL, Myers KC, Bowman J, et al. Distinct genetic path­ways define nia. Curr Opin Hematol. 2019;26(1):34-40.
pre-malig­nant ver­sus com­pen­sa­tory clonal hema­to­poi­e­sis in Shwachman- 36. Lim CJ, Cech TR. Shaping human telo­meres: from shelterin and CST com­
Diamond syn­drome. Nat Commun. 2021;12(1):1334. plexes to telomeric chro­ma­tin orga­ni­za­tion. Nat Rev Mol Cell Biol. 2021;​
13. Xia J, Miller CA, Baty J, et al. Somatic muta­tions and clonal hema­to­poi­e­sis 22(4):283-298.
in con­gen­i­tal neutropenia. Blood. 2018;131(4):408-416. 37. Calado RT, Young NS. Telomere dis­eases. N Engl J Med. 2009;361(24):2353–
14. Warren AJ. Molecular basis of the human ribosomopathy Shwachman- 2365.
Diamond syn­drome. Adv Biol Regul. 2018;67:109-127. 38. Lingner J, Cooper JP, Cech TR. Telomerase and DNA end rep­li­ca­tion: no
15. Vlachos A, Rosenberg PS, Atsidaftos E, Alter BP, Lipton JM. Incidence of lon­ger a lag­ging strand prob­lem? Science. 1995;269(5230):1533-1534.
neo­pla­sia in Diamond Blackfan ane­mia: a report from the Diamond Blackfan 39. Schratz KE. Extrahematopoietic man­ i­
fes­
ta­
tions of the short telo­ mere
Anemia Registry. Blood. 2012;119(16):3815-3819. syn­dromes. Hematology Am Soc Hematol Educ Program. 2020;2020(1):
16. Lindsley RC, Saber W, Mar BG, et  al. Prognostic muta­tions in myelodys­ 115-122.
plastic syn­drome after stem-cell trans­plan­ta­tion. N Engl J Med. 2017;376(6): 40. Schratz KE, Haley L, Danoff SK, et al. Cancer spec­trum and out­comes in the
536-547. Men­de­lian short telo­mere syn­dromes. Blood. 2020;135(22):1946-1956.
17. Simkins A, Bannon SA, Khoury JD, et al. Diamond-blackfan ane­mia predis­ 41. Kirschner M, Maurer A, Wlodarski MW, et  al. Recurrent somatic muta­
posing to myelodysplastic syn­drome in early adult­hood. JCO Precis Oncol. tions are rare in patients with cryp­tic dyskeratosis congenita. Leukemia.
2017;22(1):1-5. 2018;32(8):1762-1767.
18. Schaefer EJ, Lindsley RC. Significance of clonal muta­tions in bone mar­ 42. Perdigones N, Perin JC, Schiano I, et al. Clonal hema­to­poi­e­sis in patients
row fail­ure and inherited myelodysplastic syn­drome/acute mye­loid leu­ke­ with dyskeratosis congenita. Am J Hematol. 2016;91(12):1227-1233.
mia pre­dis­po­si­tion syn­dromes. Hematol Oncol Clin North Am. 2018;32(4): 43. Maryoung L, Yue Y, Young A, et al. Somatic muta­tions in telomerase pro­
643-655. moter coun­ter­bal­ance germline loss-of-func­tion muta­tions. J Clin Invest.
19. Knies K, Inano S, Ramírez MJ, et al. Biallelic muta­tions in the ubiquitin ligase 2017;127(3):982-986.
RFWD3 cause Fanconi ane­mia. J Clin Invest. 2017;127(8):3013-3027. 44. Jongmans MC, Verwiel ET, Heijdra Y, et  al. Revertant somatic mosa­i­cism
20. Nalepa G, Clapp DW. Fanconi anae­mia and can­cer: an intri­cate rela­tion­ by mitotic recom­bi­na­tion in dyskeratosis congenita. Am J Hum Genet.
ship. Nat Rev Cancer. 2018;18(3):168-185. 2012;90(3):426-433.
21. Ceccaldi R, Parmar K, Mouly E, et al. Bone mar­row fail­ure in Fanconi ane­mia 45. Narumi S, Amano N, Ishii T, et al. SAMD9 muta­tions cause a novel mul­ti­sys­
is trig­gered by an exac­er­bated p53/p21 DNA dam­age response that impairs tem dis­or­der, MIRAGE syn­drome, and are asso­ci­ated with loss of chro­mo­
hema­to­poi­etic stem and pro­gen­i­tor cells. Cell Stem Cell. 2012;11(1):36-49. some 7. Nat Genet. 2016;48(7):792-797.

Prakash Singh Shekhawat


Clonal hema­to­poi­e­sis in IBMFS  |  397
46. Nagamachi A, Matsui H, Asou H, et al. Haploinsufficiency of SAMD9L, an 48. Nagata Y, Narumi S, Guan Y, et  al. Germline loss-of-func­tion SAMD9 and
endosome fusion facil­i­ta­tor, causes mye­loid malig­nan­cies in mice mim­ SAMD9L alter­ations in adult myelodysplastic syn­ dromes. Blood. 2018;​
ick­ing human dis­eases with mono­somy 7. Cancer Cell. 2013;24(3):305- 132(21):2309-2313.
317.
47. Wong JC, Bryant V, Lamprecht T, et al. Germline SAMD9 and SAMD9L muta­
tions are asso­ci­ated with exten­sive genetic evo­lu­tion and diverse hema­to­ © 2021 by The Amer­i­can Society of Hematology
logic out­comes. JCI Insight. 2018;3(14):e121086. DOI 10.1182/hema­tol­ogy.2021000271

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/390/1852164/390choijilsuren.pdf by guest on 13 December 2021

398  |  Hematology 2021  |  ASH Education Program


LET THE CHIP(S) FALL WHERE THEY MAY ? BURDENS AND BENEFITS OF DIAGNOSING CLONAL HEMATOPOIESIS

When are idiopathic and clonal cytopenias


of unknown signifcance (ICUS or CCUS)?

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/399/1852160/399osman.pdf by guest on 13 December 2021


Afaf E. W. G. Osman
Division of Hematology and Hematologic Malignancies, University of Utah, Salt Lake City, UT

Rapid advances in sequencing technology have led to the identification of somatic mutations that predispose a signifi-
cant subset of the aging population to myeloid malignancies. Recently recognized myeloid precursor conditions include
clonal hematopoiesis of indeterminate potential (CHIP) and clonal cytopenia of unknown significance (CCUS). These
conditions can present diagnostic challenges and produce unwarranted anxiety in some instances. While the risk of pro-
gression to myeloid malignancies is very low in CHIP, true CCUS confers an exponential increase in risk. Idiopathic cyto-
penia of unknown significance (IDUS) lacks the predisposing genetic mutations and has a variable course. In this review
we define the early myeloid precursor conditions and their risk of progression. We present our diagnostic approach to
patients with unexplained cytopenias and discuss the clinical consequences of CHIP and CCUS.

LEARNING OBJECTIVES
• Identify myeloid precursor conditions (CCUS and CHIP) and their clinical consequences
• Understand the diagnostic approach to patients with unexplained cytopenias
• Understand the role of next­generation sequencing in the workup of unexplained cytopenias

Introduction CLINICAL CASE


Cancer precursor states were frst recognized in solid tu­ An 80­year­old man with pancytopenia was referred
mors and led to a better understanding of the multistep to our clinic. He had a past medical history of chronic
processes underlying oncogenesis. In hematology, mono­ obstructive pulmonary disease and recurrent deep vein
clonal gammopathy of undetermined signifcance and thrombosis. His medications included an oral anticoagu­
monoclonal B­cell lymphocytosis are well­characterized lant and antihypertensive therapy. He did not report any
cancer precursor states. These conditions are defned by fatigue, recurrent infections, fever, or night sweats. Initial
the presence of clonally expanded hematopoietic cells, the testing showed the following:
absence of overt malignancy, and increased risk of devel­
oping lymphoid malignancies. In parallel, clonal hema­ Hemogram (normal range) Additional studies (normal range)
topoiesis and clonal cytopenias are emerging as newly
WBC: 3.16 (4.5-11 K/µL) Serum iron: 65 (60-150 mcg/dL)
recognized precursor states that increase the risk of devel­
oping myeloid malignancies, in particular myelodysplastic RBC: 3.14 (4.5-5.9M/µL) Ferritin: 266 (30-400ng/mL)
syndrome (MDS) and acute myeloid leukemia (AML). Rap­ PLT: 86 (150-350 K/µL) RDW: 20.7 (11%-14%)
id advancements in genomic technology have resulted Hgb: 10.2 (13-16.3g/dL) Absolute reticulocyte count: 43.2
in the detection of cancer­associated mutations without (47-152 K/µL)
the morphologic changes characteristic of MDS and AML. MCV: 96 (80-100 fL) TSH, B12, folate, zinc, copper, and lac­
This has led to the identifcation of new precursor states tate dehydrogenase within normal
with increased predisposition to myeloid malignancies. limits
In this brief review, we discuss the defnition, clinical con­ Antinuclear antibody negative
sequences, and management of early myeloid precursor MCV, mean corpuscular volume; RBC, red blood cell; WBC, white
states. blood cell.

Prakash Singh Shekhawat


CHIP, CCUS, and ICUS: defning myeloid precursors | 399
Morphologic exam­i­na­tion of the bone mar­row showed a nor­ of devel­op­ing a hema­to­logic malig­nancy.1-3 The iden­tity of the
mocellular mar­row with trilineage hema­to­poi­e­sis and no evi­ mutated genes affects the risk of pro­gres­sion. While genes that
dence of dys­pla­sia or increased blasts. Flow cytometry did not code for epi­ge­netic mod­i­fi­ers (DNMT3A, TET2, and ASXL1) are the
show abnor­mal cells. Conventional karyotyping showed 2 abnor­ most com­monly mutated in CHIP, they do not con­fer the highest
mal cell lines: 46,XY,del(12)(q24.1),add(22)(q13)[3]/46,XY,add(10) risk of pro­gres­sion. Two stud­ies exam­ined CHIP in blood sam­ples
(q22)[2]/46,XY[18]. Next-gen­er­a­tion sequenc­ing (NGS) from his taken years before patients devel­oped AML and showed a higher
bone mar­row revealed the pres­ence of 3 muta­tions: prev­a­lence of CHIP in these preleukemic sam­ples.5,6 Mutations in
splic­ing-fac­tor genes (U2AF1, SRSF2, and SF3B1) as well as IDH1,
1 .  TP53 c.743G>A, p.Arg248Gln (NM_000546.5)
IDH2, and TP53 con­ferred a higher risk of pro­gres­sion.5,6
  Variant allele fre­quency: 18.6%
The risk of pro­gres­sion is also influ­enced by the num­ber of
2.  U2AF1 c.471G>T, p.Gln157His (NM_006758.2)
muta­tions in an indi­vid­ual with CHIP. Multiple driver muta­tions in
  Variant allele fre­quency: 19.7%
the same indi­vid­ual are thought to reflect clonal evo­lu­tion and
3.  DNMT3A c.2173 + 1G>A, p.? (NM_175629.2)
the acqui­si­tion of pas­sen­ger muta­tions in the path­way toward
 Variant allele frequency: 19.2% Clonal hematopoiesis of inde­

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/399/1852160/399osman.pdf by guest on 13 December 2021


frank malig­nancy.
terminate potential
Blood counts are nor­mal by def­i­ni­tion in CHIP, but a higher
red cell dis­tri­bu­tion width (RDW) seems to pre­dict a higher risk
In 2014, 3 groups of research­ers simul­ta­neously dis­cov­ered the of pro­gres­sion.5 A higher RDW is thought to reflect under­ly­ing
pres­ence of can­cer-asso­ci­ated muta­tions in the periph­eral blood inef­fec­tive hema­to­poi­e­sis and often her­alds fall­ing counts and
of seem­ingly nor­mal older indi­vid­u­als.1-3 The term clonal hema­ malig­nant trans­for­ma­tion.
to­poi­e­sis of inde­ter­mi­nate poten­tial (CHIP) was coined and re­
fers to the pres­ence of muta­tions asso­ci­ated with hema­to­logic Clinical con­se­quences and man­age­ment of CHIP
malig­nan­cies in indi­vid­u­als with nor­mal blood counts and no evi­ Currently, there are no clear clin­i­cal indi­ca­tions to screen for
dence of hema­to­logic malig­nan­cies (Table 1).4 These muta­tions CHIP. Nevertheless, due to expanding indi­ca­tions for genetic
rep­re­sent somatic events that lead to the clonal expan­sion of sequenc­ing, CHIP is increas­ingly diag­nosed in the clin­i­cal set­
hema­ to­poi­etic stem cells. ARCH (age-related clonal hema­ to­ ting. Current indi­ca­tions for genetic sequenc­ing include germ­
poi­es­ is) is another acro­nym used to describe this phe­nom­en ­ on. line sequenc­ing for the eval­u­a­tion of can­cer pre­dis­po­si­tion, for
The bio­log­i­cal mech­a­nisms under­ly­ing CHIP are addressed in an guid­ance on ther­apy choices in solid tumors, for detecting cir­cu­
accom­pa­ny­ing review by L. Gondek. lat­ing tumor DNA, and in the workup of unex­plained cytopenias.
CHIP is rel­a­tively com­mon in the aging pop­u­la­tion, with ini­ In patients with solid tumors and their rel­a­tives, test­ing for
tial stud­ies esti­mat­ing a prev­a­lence of roughly 10% among indi­ germline pre­ dis­
po­
si­
tion is rou­ tinely done on DNA extracted
vid­u­als aged 70 to 80. In those stud­ies, CHIP con­fers a 10-fold from the periph­eral blood. Germline pre­dis­po­si­tion genetic pan­
increase in the risk of devel­op­ing a hema­to­logic malig­nancy.1-3 els can include genes involved in CHIP, such as TP53. Not infre­
This trans­lates to a rate of pro­gres­sion of 0.5% to 1% per year, sim­ quently, these pan­els will return results in which muta­tions are
i­lar to the rate of pro­gres­sion from mono­clo­nal gammopathy of detected at a VAF below the usual het­ero­zy­gos­ity rates (VAFs
unde­ter­mined sig­nif­i­cance and mono­clo­nal B-cell lym­pho­cy­to­sis below 40%). These clones could rep­re­sent CHIP, and addi­tional
to lym­phoid malig­nan­cies.1-3 Accordingly, most older indi­vid­u­als tis­sue test­ing may be needed to clar­ify this.7,8 Similarly, test­ing
with CHIP have nor­mal blood counts, no fea­tures of hema­to­logic for cir­cu­lat­ing tumor DNA is done using blood sam­ples and could
malig­nan­cies, and a very low risk of pro­gres­sion. However, the uncover CHIP clones.9 CHIP clones can also be uncov­ered when
risk of pro­gres­sion from CHIP to mye­loid malig­nan­cies can vary sequenc­ing is conducted using solid-tumor tis­sue sam­ples due
and depends on sev­eral fac­tors. The size of the CHIP clone cor­re­ to con­tam­i­na­tion with blood cells.10
lates with the mag­ni­tude of risk. A var­i­ant allele fre­quency (VAF) With the increase in CHIP diag­noses, sev­eral large cen­ters,
cut­off of 1% to 2% was ini­tially used to define CHIP. This cut­off was includ­ing ours, have started ded­i­cated CHIP clin­ics to eval­u­ate
based on the lower limit of detec­tion of whole exome sequenc­ and fol­low these patients.11 A diag­no­sis of CHIP in older indi­vid­u­
ing but may not be ideal for cur­rent clin­ic ­ al prac­tice. Clones with als could pro­duce unwar­ranted anx­i­ety. The risk of hema­to­logic
a VAF of 10% or more dem­on­strate a 50-fold increase in the risk malig­nan­cies for car­ri­ers of CHIP who lack high-risk fea­tures is
minor. It is impor­tant to reas­sure these indi­vid­u­als that most of
them will not develop hema­to­logic malig­nan­cies and do not
require clin­i­cal inter­ven­tion.12 On the other hand, clones with
Table 1.  The most com­monly mutated genes in CHIP/CCUS
larger VAFs (>10%) and clones with muta­tions in high-risk genes
and their respec­tive func­tions
or with muta­tions in more than 1 driver gene war­rant closer
mon­i­tor­ing with serial blood counts. A bone mar­row exam­i­na­
Genes Function/path­way
tion should be pur­sued if these patients develop cytopenias. The
DNMT3A, TET2, ASXL1, IDH1, IDH2 Epigenetic reg­u­la­tors workup for cytopenias is fur­ther detailed below.
SF3B1, SRSF2, U2AF1 RNA splic­ing Certain CHIP clones can carry sig­nif­i­cant risk to patients with
TP53, PPM1D DNA repair/cell cycle
solid tumors who will undergo treat­ment with cyto­toxic che­
mo­ther­apy and/or radi­a­tion. Cytotoxic che­mo­ther­apy and ion­
JAK2, CBL, GNAS, GNB1 Signaling
iz­ing radi­a­tion are likely to intro­duce a selec­tive pres­sure that
BCOR, BCORL1 Transcription reg­u­la­tion leads to the expan­sion of CHIP clones with muta­tions in the DNA
Mutations in DNMT3A, TET2, and ASXL1 are the most fre­quent muta­tions repair path­ways (TP53, CHEK2, and PPM1D). The pres­ence of
in CHIP and con­fer a lower risk of pro­gres­sion in sol­i­tary. CHIP muta­tions increases the risk of treat­ment-related mye­loid

400  |  Hematology 2021  |  ASH Education Program


Persistent cytopenias:
hemoglobin <13.0 g/dL [males], <12.0 g/dL [females]; absolute neutrophil count
<1·8 × 109/L; platelets <150 × 109/L

Rule out nutritional, autoimmune,


infectious, toxic, and neoplastic
processes

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/399/1852160/399osman.pdf by guest on 13 December 2021


Unexplained cytopenias
Dysplasia or
Bone marrow aspiration and increased
biopsy myeloblasts

MDS/AML
MDS
Cytogenetic evaluation of the defining
bone marrow cytogenetic
abnormalities

SOMATIC
NGS panel including genes mutation CCUS
associated with myeloid VAF>2%
malignancies

No evidence of clonality

ICUS

Figure 1. The diagnostic workup for cytopenias starts with ruling out underlying etiologies, including nutritional, autoimmune,
infectious, drug-mediated, and neoplastic processes. NGS plays an important role in the workup of unexplained cytopenias to
differentiate between CCUS and ICUS.

neo­plasms.13 This find­ing has impor­tant clin­i­cal impli­ca­tions for after aor­tic valve implan­ta­tion pro­ce­dures.12-14 In our CHIP clinic,
patients with solid tumors under­ go­
ing adju­vant/neoadjuvant we assess the pres­ence of clas­sic risk fac­tors of car­dio­vas­cu­lar
che­mo­ther­apy and/or radi­a­tion. Currently, no clin­i­cal guide­lines dis­ease, such as hyper­lip­id­emia, dia­be­tes mellitus, hyper­ten­sion,
call for screen­ing for CHIP in this pop­u­la­tion. and smok­ing. We refer patients who require inter­ven­tions to our
Special atten­tion should also be paid to the car­dio­vas­cu­lar risk cardio-oncol­o­gist for fur­ther man­age­ment.
asso­ci­ated with CHIP. The increase in all­-cause mor­tal­ity detected
in ear­lier CHIP stud­ies was mostly attrib­ut­­able to an increase in The diag­nos­tic approach to unex­plained cytopenias
fatal car­dio­vas­cu­lar events, includ­ing ische­mic stroke, cor­o­nary Unexplained cytopenias are defined as per­sis­tent cytopenias in 1
heart dis­ease, and early-onset myo­car­dial infarc­tion.1,2,14 Mutations or more cell lin­e­ages with­out a clear under­ly­ing eti­­ol­ogy (hemo­
in the 2 most com­mon CHIP genes, TET2 and DNMT3A, are asso­ glo­bin <13.0 g/dL [males], <12.0 g/dL [fe­males]; abso­lute neu­tro­
ci­ated with an increased risk of car­dio­vas­cu­lar dis­ease through phil count <1.8 × 109/L; plate­lets <150 × 109/L).16 The workup for
proinflammatory mech­a­nisms.1,14 In addi­tion, JAK2 V617F-mutated cytopenias starts with rul­ing out nutri­tional, auto­im­mune, infec­
CHIP is asso­ci­ated with venous throm­bo­sis, includ­ing deep vein tious, toxic, and neo­plas­tic causes.17 The term “unex­plained cy­
throm­bo­sis and pul­mo­nary embolism.15 Carriers of CHIP were also topenias” is used when the ori­gin of cytopenias is not explained
found to have worse heart fail­ure out­comes and worse out­comes by these causes or by con­com­i­tant illnesses.
Prakash Singh Shekhawat
CHIP, CCUS, and ICUS: defin­ing mye­loid pre­cur­sors | 401
Table 2.  The unique clin­i­cal fea­tures dif­fer­en­ti­at­ing ICUS, CHIP, CCUS, and MDS

ICUS CHIP CCUS MDS


Cytopenias + − + +
Somatic muta­tions − + + +
Morphologic dys­pla­sia − − − +
Increased blasts − − − ±
Risk of trans­for­ma­tion to AML Very low Very low Low Low to very high
Suggested man­age­ment and - CBC with dif­fer­en­tial, his­ - Routine health main­te­ - CBC with dif­fer­en­tial every 3-6 mo Treatment per stage-
fol­low-up* tory, and phys­ic­ al annu­ nance. - History and phys­i­cal annu­ally or spe­cific guide­lines
ally or semi­an­nu­ally - Consider annual CBC with semi­an­nu­ally
dif­fer­en­tial - Repeat bone mar­row biopsy if

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/399/1852160/399osman.pdf by guest on 13 December 2021


- Assess car­dio­vas­cu­lar risk counts worsen
- Assess car­dio­vas­cu­lar risk
+, pres­ent; −, absent.
*Evidence-based guide­lines for man­age­ment of CHIP, CCUS, and idi­o­pathic dys­pla­sia of unknown sig­nif­i­cance are cur­rently lacking. Follow-up should
be tai­lored to each patient’s indi­vid­ual risk and needs.

MDS is one of the most com­mon hema­to­logic malig­nan­cies and ment are dif­fer­ent (Table 2). ICUS is defined as unex­plained cyto­
fre­quently pres­ents with unex­plained cytopenias.17 The workup of penia that does not meet the min­i­mal cri­te­ria for MDS. CCUS is
unex­plained cytopenias starts with rul­ing out MDS (Figure 1). MDS defined as ICUS with one or more somatic muta­tions typ­i­cally
is a group of het­ero­ge­neous clonal hema­to­poi­etic malig­nan­cies found in patients with mye­loid malig­nan­cies. Idiopathic dys­pla­
char­ac­ter­ized by inef­fec­tive hema­to­poi­e­sis and increased risk sia of unknown sig­nif­i­cance is a con­di­tion with­out cytopenia that
of AML. The diag­no­sis of MDS requires mor­pho­log­i­cal stud­ies of usu­ally rep­re­sents a reac­tive pro­cess.24
periph­eral blood and bone mar­row and cyto­ge­net­ics to iden­tify
non­ran­dom chro­mo­somal abnor­mal­i­ties. Diagnostic cri­te­ria for The clin­i­cal sig­nif­i­cance of CCUS
MDS include dys­pla­sia in at least 10% of cells in any hema­to­poi­ The cur­rent model of pro­gres­sion from CHIP to a mye­loid malig­
etic lin­e­age, increased mye­lo­blasts, or MDS-defin­ing cyto­ge­netic nancy includes a step­wise pro­gres­sion through a num­ber of dis­
abnor­mal­i­ties detected through karyotyping.18 It is impor­tant to ease states. CHIP is the ear­li­est iden­ti­fi­able entity in this model;
note that mor­pho­logic cri­te­ria for MDS suf­fer from great interpro­ by def­i­ni­tion, it requires the pres­ence of nor­mal blood counts.
vider var­i­abil­ity in terms of detecting dys­pla­sia.19 The devel­op­ment of cytopenia rep­re­sents the next man­i­fes­ta­tion
NGS plays an impor­tant role in the workup of unex­plained of a dys­func­tional hema­to­poi­etic sys­tem progressing toward a
cytopenias. Although not included in the cur­rent diag­nos­tic cri­ frank mye­loid malig­nancy. Cytopenia in CCUS is a result of the
te­ria for MDS, NGS pan­els from the periph­eral blood can aid in clonal hema­to­poi­etic pro­cess lead­ing to inef­fec­tive hema­to­poi­
the diag­no­sis of MDS and other hema­to­logic malig­nan­cies.20,21 e­sis. It is impor­tant to rule out other causes of cytopenia before
The absence of can­ cer-asso­ci­
ated somatic muta­ tions in the diag­nos­ing CCUS. For exam­ple, a patient with iso­lated throm­
periph­ eral blood has a high neg­a­ tive pre­ dic­tive value for an bo­cy­to­pe­nia and a low-level DNMT3A muta­tion could have idi­
under­ly­ing mye­loid malig­nancy.22 On the other hand, the pres­ o­pathic throm­bo­cy­to­pe­nic pur­pura (ITP). If the throm­bo­cy­to­pe­
ence of these muta­tions in the periph­eral blood raises the sus­ nia responds to treat­ment with ste­roids, the patient is unlikely to
pi­cion of an under­ly­ing hema­to­logic malig­nancy. The pres­ence have CCUS. The DNMT3A muta­tion in this sit­u­a­tion is less sig­nif­i­
of larger clones with VAFs of ≥10% and clones with 2 or more cant because DNMT3A-mutated CHIP is com­mon in healthy older
muta­tions in the periph­eral blood has a pos­i­tive pre­dic­tive value indi­vid­u­als.1-3
of 0.86 and 0.88 for the diag­no­sis of a mye­loid neo­plasm in cyto­ CCUS con­fers a much higher risk of pro­gres­sion to a mye­
penic patients. Conversely, smaller clones involv­ing only 1 muta­ loid malig­nancy than ICUS, with a 10-year cumu­la­tive prob­ab ­ il­
tion in the com­mon epi­ge­netic reg­u­la­tors (DNM3TA, TET2, and ity of pro­gres­sion of 82% in CCUS vs 9% in ICUS. Furthermore,
ASXL1) have a low pre­dic­tive value unless a sec­ond muta­tion is CCUS clones with a VAF >20% are asso­ci­ated with a more than
pres­ent in the same sam­ple. Mutations in spliceosome genes 95% risk of pro­gres­sion to a mye­loid malig­nancy in a 10-year
(SF3B1, SRSF2, and U2AF1), JAK2, and RUNX1 have the highest period.23 Consequently, a VAF of 20% is likely a bet­ter cut­off to
pre­dic­tive value for mye­loid neo­plasms irrespective of co-occur­ define CCUS than the 2% used for defin­ing CHIP. The pres­ence
ring muta­tions.23 of muta­tions in spe­cific genes (such as U2AF1, ZRSR2, SRSF2,
In the absence of diag­nos­tic cri­te­ria for MDS, unex­plained JAK2, or RUNX1) and the pres­ence of mul­ti­ple muta­tions also her­
cytopenia could rep­re­sent one of two enti­ties depending on ald a high risk of pro­gres­sion of up to 80% to 90% in 5 years.23
the pres­ ence of clonal genetic abnor­ mal­ i­
ties: clonal cytope­ Although the term “CCUS” is now accepted by experts in the
nia of unknown sig­nif­i­cance (CCUS) or idi­o­pathic cytopenia of field, I would argue that this is a mis­no­mer because of the high
unknown sig­nif­i­cance (ICUS).24-26 It is impor­tant to dif­fer­en­ti­ate risk of pro­gres­sion to mye­loid malig­nancy in true CCUS. Larger
between CCUS and ICUS because the prog­no­sis and man­age­ clonal events can also be seen in CCUS and are asso­ci­ated with

402  |  Hematology 2021  |  ASH Education Program


a risk of pro­gres­sion. Trisomy 8 detected by fluo­res­cence in situ 4. Steensma DP, Bejar R, Jaiswal S, et al. Clonal hema­to­poi­e­sis of inde­ter­mi­
hybrid­iza­tion sig­naled the pres­ence of a low-level clonal pro­cess nate poten­tial and its dis­tinc­tion from myelodysplastic syn­dromes. Blood.
2015;126(1):9-16.
and increased risk of sub­se­quent MDS in a group of patients with 5. Abelson S, Collord G, Ng SWK, et al. Prediction of acute mye­loid leu­kae­mia
unex­plained cytopenias.27 Focal copy num­ber aber­ra­tions and risk in healthy indi­vid­u­als. Nature. 2018;559(7714):400-404.
copy-neu­tral loss of het­ero­zy­gos­ity detected by sin­gle-nucle­o­ 6. Desai P, Mencia-Trinchant N, Savenkov O, et al. Somatic muta­tions pre­cede
tide poly­mor­phism assays are asso­ci­ated with worse sur­vival in acute mye­loid leu­ke­mia years before diag­no­sis. Nat Med. 2018;24(7):1015-
1023.
patients with CCUS.28
7. Slavin TP, Coffee B, Bernhisel R, et  al. Prevalence and char­ac­ter­is­tics of
On the other hand, the clin­i­cal course for ICUS is more indo­ likely-somatic var­i­ants in can­cer sus­cep­ti­bil­ity genes among indi­vid­u­als
lent. Patients with ICUS and no evi­dence of clonality can be mon­ who had hered­i­tary pan-can­cer panel test­ing. Cancer Genet. 2019;235-
i­tored with peri­odic blood counts. Patients with CCUS require 236(June):31-38.
closer mon­i­tor­ing of their blood counts and repeat bone mar­row 8. Weitzel JN, Chao EC, Nehoray B, et  al. Somatic TP53 var­i­ants fre­quently
con­found germ-line test­ing results. Genet Med. 2018;20(8):809-816.
exam­i­na­tion if counts worsen (Table 2). 9. Jensen K, Konnick EQ , Schweizer MT, et  al. Association of clonal hema­
The clone size and the num­ber of muta­tions in CCUS tend to­poi­e­sis in DNA repair genes with pros­tate can­cer plasma cell-free DNA

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/399/1852160/399osman.pdf by guest on 13 December 2021


to be much higher than in CHIP. The pat­tern of muta­tions in test­ing inter­fer­ence. JAMA Oncol. 2021;7(1):107-110.
CCUS tends to resem­ble muta­tions found in lower-risk MDS.29,30 10. Bolton KL, Gillis NK, Coombs CC, et al. Managing clonal hema­to­poi­e­sis in
patients with solid tumors. J Clin Oncol. 2019;37(1):7-11.
Furthermore, patients with high-risk CCUS had sim­i­lar sur­vival
11. Bolton KL, Zehir A, Ptashkin RN, et al. The clin­i­cal man­age­ment of clonal
to those with mye­ loid neo­plasms.23 High-risk CCUS lies in a hema­to­poi­e­sis: cre­a­tion of a clonal hema­to­poi­e­sis clinic. Hematol Oncol
close con­tin­uum to lower-risk MDS, and the line between these Clin North Am. 2020;34(2):357-367.
2 enti­ties con­tin­ues to blur. Patients with CCUS can also have 12. Gondek LP, DeZern AE. Assessing clonal haematopoiesis: clin­i­cal bur­dens
low-level dys­ pla­
sia, and some patients may develop trans­ fu­ and ben­e­fits of diag­nos­ing myelodysplastic syn­drome pre­cur­sor states.
Lancet Haematol. 2020;7(1):e73-e81.
sion-depen­dent ane­mia.29 While no large ran­dom­ized tri­als exist 13. Bolton KL, Ptashkin RN, Gao T, et  al. Cancer ther­apy shapes the fit­ness
in this space, a sin­gle-cen­ter study exam­in­ing the treat­ment of land­scape of clonal hema­to­poi­e­sis. Nat Genet. 2020;52(11):1219-1226.
trans­fu­sion-depen­dent CCUS showed some suc­cess with MDS- 14. Jaiswal S, Natarajan P, Silver AJ, et  al. Clonal hema­to­poi­e­sis and risk of
type ther­a­pies, includ­ing growth fac­tors and hypomethylating ath­ero­scle­rotic car­dio­vas­cu­lar dis­ease. N Engl J Med. 2017;377(2):111-121.
15. Edelmann B, Gupta N, Schnoeder TM, et al. JAK2-V617F pro­motes venous
agents.31 Patients with high-risk CCUS who develop trans­fu­sion
throm­bo­sis through β1/β2 integrin acti­va­tion. J Clin Invest. 2018;128(10):​
depen­dency or signs and symp­toms of bone mar­row fail­ure may 4359-4371.
ben­e­fit from treat­ment algo­rithms used in low-risk MDS. Consen­ 16. Greenberg PL, Tuechler H, Schanz J, et  al. Cytopenia lev­ els for aiding
sus treat­ment and mon­i­tor­ing guide­lines for CCUS are cur­rently estab­ lish­ment of the diag­ no­
sis of myelodysplastic syn­ dromes. Blood.
lacking. 2016;128(16):2096-2097.
17. Malcovati L, Hellström-Lindberg E, Bowen D, et al; Euro­pean Leukemi­
Our patient was diag­nosed with high-risk CCUS. Although aNet. Diagnosis and treat­ment of pri­mary myelodysplastic syn­dromes in
his periph­eral counts were highly sus­pi­cious for MDS, he did not adults: rec­om­men­da­tions from the Euro­pean LeukemiaNet. Blood. 2013;​
have dys­pla­sia, increased mye­lo­blasts, or MDS-defin­ing cyto­ge­ 122(17):2943-2964.
netic abnor­mal­i­ties. The patient was coun­seled about his con­di­ 18. Arber DA, Orazi A, Hasserjian R, et al. The 2016 revi­sion to the World Health
Organization clas­si­fi­ca­tion of mye­loid neo­plasms and acute leu­ke­mia.
tion and is being mon­i­tored with monthly hemograms due to the
Blood. 2016;127(20):2391-2405.
high-risk nature of his muta­tions. A repeat bone mar­row exam­ 19. Parmentier S, Schetelig J, Lorenz K, et al. Assessment of dys­plas­tic hema­
i­na­tion will be pur­sued if his counts worsen or if he devel­ops to­poi­ e­ sis: les­
sons from healthy bone mar­ row donors. Haematologica.
symp­toms of bone mar­row fail­ure. 2012;97(5):723-730.
20. Haferlach T, Nagata Y, Grossmann V, et al. Landscape of genetic lesions in
944 patients with myelodysplastic syn­dromes. Leukemia. 2014;28(2):241-
Conflict-of-inter­est dis­clo­sure 247.
Afaf E. W. G. Osman: no com­pet­ing finan­cial inter­ests to declare. 21. Zheng G, Chen P, Pallavajjalla A, et  al. The diag­nos­tic util­ity of targeted
gene panel sequenc­ing in dis­crim­i­nat­ing eti­­ol­o­gies of cytopenia. Am J
Hematol. 2019;94(10):1141-1148.
Off-label drug use 22. Steensma DP. How I use molec­u­lar genetic tests to eval­u­ate patients who
Afaf E. W. G. Osman: use of hypomethylating agents and growth have or may have myelodysplastic syn­dromes. Blood. 2018;132(16):1657-
factors in CCUS is briefly discussed in this article. 1663.
23. Malcovati L, Gallì A, Travaglino E, et al. Clinical sig­nif­i­cance of somatic
muta­ tion in unex­ plained blood cytopenia. Blood. 2017;129(25):3371-
Correspondence 3378.
Afaf E. W. G. Osman, Division of Hematology and Hematologic 24. Valent P, Bain BJ, Bennett JM, et al. Idiopathic cytopenia of unde­ter­mined
Malignancies, University of Utah, 2000 Circle of Hope Dr, Salt sig­nif­i­cance (ICUS) and idi­o­pathic dys­pla­sia of uncer­tain sig­nif­i­cance (IDUS),
Lake City, UT 84103; e-mail: afaf​­.osman@hsc​­.utah​­.edu. and their dis­tinc­tion from low risk MDS. Leuk Res. 2012;36(1):1-5.
25. Valent P, Orazi A, Steensma DP, et al. Proposed min­i­mal diag­nos­tic cri­te­ria
for myelodysplastic syn­dromes (MDS) and poten­tial pre-MDS con­di­tions.
References Oncotarget. 2017;8(43):73483-73500.
1. Jaiswal S, Fontanillas P, Flannick J, et al. Age-related clonal hema­to­poi­e­ 26. Valent P, Horny HP, Bennett JM, et  al. Definitions and stan­dards in the
sis asso­ci­ated with adverse out­comes. N Engl J Med. 2014;371(26):2488- diag­no­sis and treat­ment of the myelodysplastic syn­dromes: con­sen­sus
2498. state­ments and report from a work­ing con­fer­ence. Leuk Res. 2007;31(6):
2. Genovese G, Kähler AK, Handsaker RE, et  al. Clonal hema­to­poi­e­sis and 727-736.
blood-can­cer risk inferred from blood DNA sequence. N Engl J Med. 2014;​ 27. Petrova-Drus K, Hasserjian R, Pozdnyakova O, et  al. Clinicopathologic
371(26):​2477-2487. eval­u­a­tion of cytopenic patients with iso­lated tri­somy 8: a detailed
3. Xie M, Lu C, Wang J, et al. Age-related muta­tions asso­ci­ated with clonal com­par­i­son between idi­o­pathic cytopenia of unknown sig­nif­i­cance and
hema­to­poi­etic expan­sion and malig­nan­cies. Nat Med. 2014;20(12):1472- low-grade myelodysplastic syn­drome. Leuk Lymphoma. 2017;58(3):569-
1478. 577.

Prakash Singh Shekhawat


CHIP, CCUS, and ICUS: defin­ing mye­loid pre­cur­sors | 403
28. Mikkelsen SU, Safavi S, Dimopoulos K, et al. Structural aber­ra­tions are asso­ 31. Xie Z, Nanaa A, Saliba AN, et al. Treatment out­come of clonal cytopenias of
ci­ated with poor sur­vival in patients with clonal cytopenia of unde­ter­mined unde­ter­mined sig­nif­i­cance: a sin­gle-insti­tu­tion ret­ro­spec­tive study. Blood
sig­nif­i­cance. Haematologica. 2021;106(6):1762-1766. Cancer J. 2021;11(3):43.
29. Kwok B, Hall JM, Witte JS, et  al. MDS-asso­ci­ated somatic muta­tions and
clonal hema­to­poi­e­sis are com­mon in idi­o­pathic cytopenias of unde­ter­
mined sig­nif­i­cance. Blood. 2015;126(21):2355-2361.
30. Cargo CA, Rowbotham N, Evans PA, et al. Targeted sequenc­ing identifies
patients with pre­clin­i­cal MDS at high risk of dis­ease pro­gres­sion. Blood. © 2021 by The Amer­i­can Society of Hematology
2015;​126(21):2362-2365. DOI 10.1182/hema­tol­ogy.2021000272

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/399/1852160/399osman.pdf by guest on 13 December 2021

404  |  Hematology 2021  |  ASH Education Program


MANAGEMENT STRATEGIES FOR SICKLE CELL DISEASE

Optimizing management of sickle cell disease


in patients undergoing surgery

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/405/1851532/405oyedeji.pdf by guest on 13 December 2021


Charity I. Oyedeji1 and Ian J. Welsby1,2
1
Division of Hematology, Department of Medicine, Duke University School of Medicine, Durham, NC; 2Department of Anesthesiology, Duke University
Medical Center, Durham, NC

Individuals with sickle cell disease (SCD) are likely to be referred for surgery at some point in their lifetime due to a
high incidence of musculoskeletal and intrabdominal complications such as avascular necrosis and gallbladder disease.
Preoperative optimization is a multidisciplinary process that involves a hematologist with SCD expertise, an anesthe-
siologist, and the surgical team. The type and risk classification of the surgery, disease severity, medications, baseline
hemoglobin, transfusion history, and history of prior surgical complications are often documented. Clinicians should
consider perioperative risk assessment that includes determining the patient’s functional status and cardiovascular risk
and screening for obstructive sleep apnea. Many patients will require preoperative transfusion to reduce the risk of
postoperative complications such as acute chest syndrome and vaso-occlusive pain crises. The hematologist should
consider the patient’s preoperative transfusion requirements and ensure that the surgical team has an appropriate plan
for postoperative observation and management. This often includes follow-up laboratory studies, a postoperative pain
management plan, and venous thromboembolism prophylaxis. The transfusion plan should be patient-specific and take
into account the SCD genotype, baseline hemoglobin, disease severity, risk classification of the surgery, and history of
prior surgical complications. In the intraoperative and postoperative period, dehydration, hypothermia, hypotension,
hypoxia, and acidosis should be avoided, and incentive spirometry should be utilized to minimize complications such as
acute chest syndrome. In this review we discuss preoperative, intraoperative, and postoperative strategies to optimize
patients with SCD undergoing surgery.

LEARNING OBJECTIVES
• Understand strategies for optimizing patients with sickle cell disease undergoing surgery
• Review indications for preoperative transfusion for patients with sickle cell disease
• Understand options for postoperative pain management and venous thromboembolism prophylaxis in patients
with sickle cell disease

vative therapies including nonsteroidal anti­inflammatory


CLINICAL CASE drugs, a corticosteroid injection, and physical therapy. Her
A 46­year­old African American woman with hemoglobin pain and mobility continued to worsen. A repeat MRI of the
(Hb) sickle­beta plus thalassemia (HbSβ+) and right hip hips 2 years later showed bilateral AVN that was worse in
avascular necrosis (AVN) was referred to a sickle cell clinic the right hip than the left. Due to the failure of conservative
for optimization prior to right total hip replacement. management, orthopedics recommended a right total hip
She reported progressive bilateral hip pain that had replacement.
been worse on the right for the past 10 years. She denied Her medical history included sickle cell disease (SCD)
having a precipitating injury. The pain worsened with pro­ retinopathy, anxiety, depression, hypersomnia, and gas­
longed sitting and standing or walking short distances. troesophageal reflux. She had no history of stroke, venous
She was most concerned that the hip pain made it diffi­ thromboembolism (VTE), or chronic cardiopulmonary com­
cult for her to play with her grandchildren. Three years ear­ plications. She had contracted pneumonia 8 years prior,
lier, magnetic resonance imaging (MRI) of the right hip had and her last transfusion had been 30 years earlier. Her med­
shown a small area of AVN of the femoral head. She was ications included methadone, oxycodone, pantoprazole,
referred to orthopedics and was managed with conser­ aripiprazole, buspirone, folic acid, and dronabinol. She was
Prakash Singh Shekhawat
Perioperative management of sickle cell disease | 405
not on hydroxy­urea due to an intol­er­ance. She had no prior sur­ or pul­mo­nary hyper­ten­sion; non-SCD comorbidities; recent hos­
ger­ies and no aller­gies to med­i­ca­tions. She lived alone but had 2 pi­tal­i­za­tions; recent infec­tions; base­line Hb; trans­fu­sion his­tory
sup­port­ive adult chil­dren. She smoked 1 cig­a­rette per day, drank with a his­tory of trans­fu­sion reac­tions and ease of crossmatch;
alco­hol occa­sion­ally, and had no illicit drug use. and prior sur­gi­cal com­pli­ca­tions.7,8 The hema­tol­o­gist should
On phys­i­cal exam, her blood pres­sure was 117/78 mmHg; heart con­sider trans­fu­sion require­ments and tar­get sickle Hb per­cent
rate, 102/min; res­pi­ra­tion rate, 18/min; tem­per­a­ture, 36.7 °C; (%HbS) based on the patient’s geno­type, phe­no­type, and Hb.
pain score, 8/10; and weight, 73 kg. The mus­cu­lo­skel­e­tal exam Medications must be reviewed to avoid drug-drug inter­ac­tions
showed dif­fi­culty mount­ing the exam table and pain on inter­nal and neph­ro­toxic med­i­ca­tions in the set­ting of perioperative fluid
and exter­nal hip rota­tion, with the right hip worse than the left. shifts. The perioperative team should con­sider an appro­pri­ate
Her Hb level was 8.7 g/dL; hemat­o­crit level, 34.6%; white blood acu­ity of post­op­er­a­tive mon­i­tor­ing, includ­ing lab­o­ra­tory test­ing,
cell count, 5.7 × 109/L; plate­let count; 198 000; and abso­lute a pain man­age­ment plan, and VTE pro­phy­laxis.
retic­u­lo­cyte count, 93.3  ×  109/L, with 2.68% retic­u­lo­cytes. Her
Hb elec­tro­pho­re­sis showed a sickle hemo­glo­bin [HbS] level of Preoperative risk assess­ment

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/405/1851532/405oyedeji.pdf by guest on 13 December 2021


67.0%; HbA2, 4.6%; HbA, 17.7%; fetal hemo­glo­bin [HbF], 10.7%. The anes­the­si­­ol­o­gist typ­i­cally assesses the patient’s perioperative
Her com­plete met­a­bolic pro­file, lac­tate dehy­dro­ge­nase, fer­ risk and fit­ness for sur­gery and plans anes­the­sia accord­ingly. Sur­
ri­tin, and urine microalbumin were all­within nor­mal. Coro­ geries are strat­i­fied into risk categories based on their poten­tial for
navirus was not detected on SARS-CoV-2 poly­mer­ase chain intraoperative blood loss and post­op­er­a­tive com­pli­ca­tions (ie, low,
reac­tion. mod­er­ate, or high risk; Table 2). The patient’s his­tory of strokes,
acute chest syn­drome, obstruc­tive sleep apnea (OSA), adverse
reac­tions to seda­tion, or recur­rent VTE is documented since these
Introduction increase the patient’s risk of perioperative com­pli­ca­tions.
SCD is one of the most com­mon inherited dis­or­ders in the world.1 Functional capac­ity is often mea­sured by the abil­ity to per­
Patients with SCD expe­ri­ence red blood cell sick­ling, lead­ing to form met­a­bolic equiv­a­lent tasks.9 Patients unable to per­form ≥4
micro­vas­cu­lar occlu­sion that results in com­pli­ca­tions such as met­ab ­ olic equiv­al­ent tasks (ie, climbing a flight of stairs) have an
acute vaso-occlu­sive pain cri­ses, acute chest syn­drome, chronic increased risk of car­diac events.9,10 Consider OSA screen­ing since
organ dam­age, and mus­cu­lo­skel­e­tal com­pli­ca­tions.2 Surgical the con­di­tion increases a patient’s risk of perioperative hyp­ox­
com­pli­ca­tions are more com­mon in patients with SCD com­pared emia.11,12 The risk of car­dio­vas­cu­lar events is often assessed using
to the gen­eral pop­u­la­tion due to their increased risk of post­op­ val­i­dated mod­els such as the revised car­diac risk index. Patients
er­a­tive acute chest syn­drome, infec­tions, vaso-occlu­sive pain at high risk for car­diac events and with a known his­tory of car­
cri­ses, and 30-day sur­gi­cal mor­tal­ity of 1.1%.3,4 Failure to appro­ dio­pul­mo­nary dis­ease such as pul­mo­nary hyper­ten­sion, car­diac
pri­ately opti­mize a patient with SCD perioperatively can lead to symp­toms, or poor func­tional sta­tus should con­sider addi­tional
com­pli­ca­tions such as acute chest syn­drome, which is asso­ci­ated test­ing with echo­car­dio­gram, stress test­ing, and/or a car­di­­ol­
with an increased risk of death.5,6 With appro­pri­ate plan­ning, ogy con­sul­ta­tion.10
man­age­ment, and post­op­er­a­tive mon­i­tor­ing, health care pro­vid­
ers can increase the like­li­hood of opti­mal sur­gi­cal out­comes. Preoperative trans­fu­sion
Preoperative opti­mi­za­tion for patients with SCD often includes
Determine if the sur­gery is really nec­es­sary sim­ple trans­fu­sion or red cell exchange (RCE). The pri­mary goal
Given the high risk of both sur­gi­cal and post­op­er­a­tive com­pli­ of pre­op­er­a­tive trans­fu­sion is to reduce the risk of post­op­er­a­tive
ca­tions in patients with SCD, it is imper­a­tive to first deter­mine com­pli­ca­tions by increas­ing the Hb and reduc­ing the %HbS.13
whether con­ser­va­tive man­age­ment strat­e­gies have truly failed. Patients with SCD are at increased risk of post­op­er­at­ ive mor­bid­
Consider the risks asso­ci­ated with sur­gery vs con­tinu­ing with ity pri­mar­ily related to acute chest syn­drome and pain cri­ses and
con­ser­va­tive man­age­ment, includ­ing refer­ral to a spe­cial­ist cen­ have a higher risk of mor­tal­ity com­pared to the gen­eral pop­u­la­
ter offer­ing less inva­sive options. Ascertain a patient’s per­sonal tion. The evi­dence on pre­op­er­a­tive trans­fu­sion in both pedi­at­ric
moti­va­tions for and expected out­comes of hav­ing sur­gery to and adult pop­u­la­tions shows var­i­able out­comes in pre­vent­ing
deter­mine whether sur­gery will allow them to achieve their per­ adverse out­comes and mor­tal­ity.13,14
sonal goals. This pro­cess should be a shared deci­sion between The Transfusion Alternatives Preoperatively in Sickle Cell Dis­
the patient and health care pro­vid­ers. ease (TAPS) study, which was a mul­ti­cen­ter ran­dom­ized con­trolled
trial (RCT) com­par­ing no trans­fu­sion with trans­fu­sion within 10
Preoperative man­age­ment days of low- or mod­er­ate-risk sur­gery for patients with geno­type
Once the team has planned to pro­ceed with sur­gery, the next hemoglobin SS disease (HbSS) or sickle-beta zero thalassemia
step is to eval­u­ate the patient’s risk of perioperative com­pli­ca­ (HbSβ0), showed that patients who received pre­op­er­a­tive trans­fu­
tions. This is typ­i­cally performed by an anes­the­si­­ol­o­gist and a sion had a lower risk of post­op­er­a­tive acute chest syn­drome and
hema­tol­o­gist with exper­tise in SCD either in sep­a­rate or mul­ti­ other clin­i­cally impor­tant com­pli­ca­tions (15% vs 39%; P = .023).15
dis­ci­plin­ary out­pa­tient vis­its or by vir­tual meet­ing. Consider the Despite this, there was no dif­fer­ence in post­op­er­a­tive pain cri­sis,
sur­gi­cal indi­ca­tion and the type of sur­gi­cal pro­ce­dure the patient hos­pi­tal length of stay, or readmission rates between patients
is under­go­ing and any spe­cial con­sid­er­ations (Table 1). For chil­ who received pre­op­er­a­tive trans­fu­sion com­pared with no trans­
dren, ensure they are up to date on their SCD-spe­cific health fu­sion.13,15 A mul­ti­cen­ter RCT com­par­ing pre­op­er­a­tive con­ser­va­tive
screen­ing, which includes transcranial Dopp­ler to assess their trans­fu­sion (increas­ing Hb to 10  g/dL) to aggres­sive trans­fu­sion
stroke risk. Document the risk of the sur­gery; SCD sever­ity based (using RCE to decrease %HbS to <30%) found no sig­nif­i­cant dif­
on their geno­type; com­pli­ca­tions such as stroke, renal dis­ease, fer­ence in the fre­quency of seri­ous com­pli­ca­tions.5 Ten per­cent of

406  |  Hematology 2021  |  ASH Education Program


Table 1. Common indi­ca­tions for sur­gery in sickle cell dis­ease and con­sid­er­ations

Surgical indi­ca­tion Surgery Surgical con­sid­er­ations


Severe avas­cu­lar necro­sis Total hip replace­ment27,28 - High risk of periprosthetic and wound infec­tion and respi­ra­tory
com­pli­ca­tions
- Monitor tem­per­a­ture, fluid bal­ance, and Hb
- Postoperative pro­phy­lac­tic anticoagulation for a min­i­mum of 10-14 days
and up to 35 days; LMWH is recommended
- Early mobi­li­za­tion
- Chest phys­io­ther­apy with incen­tive spi­rom­e­try
Tonsillar hyper­tro­phy, pedi­at­ric ENT sur­gery such as ton­sil­lec­tomy - At increased risk of noc­tur­nal hyp­oxia pre- and post­op­er­a­tively
OSA, and/or recur­rent ton­sil­li­tis and adenoidectomy14,29 - Monitor for dehy­dra­tion and post­op­er­a­tive oral fluid intake
Cholelithiasis Cholecystectomy30 - Use least inva­sive tech­nique (lap­a­ro­scopic)
- Monitor for pul­mo­nary com­pli­ca­tions (atel­ec­ta­sis, acute chest syn­

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/405/1851532/405oyedeji.pdf by guest on 13 December 2021


drome) and vaso-occlu­sive cri­sis of the liver
Splenic seques­tra­tion Splenectomy31,32 - Use least inva­sive tech­nique (lap­a­ro­scopic)
- At risk for postsplenectomy infec­tion; immu­nize with pneu­mo­coc­cal,
menin­go­coc­cal, Haemophilus influ­enza at least 10-14 days before sur­
gery or post­op­er­a­tively if emer­gency sur­gery is required
- Consider pro­phy­lac­tic anticoagulation to pre­vent postsplenectomy
por­tal vein sys­tem throm­bo­sis
Priapism Surgical shunt or penile pros­the­sis33 - At risk for post­op­er­a­tive erec­tile dys­func­tion, penile gan­grene, and
per­i­neal abscess
Pregnancy com­pli­ca­tions (eg, Cae­sar­ian sec­tion26,34 - Hydrate and main­tain spO2 > 94%
pre­eclamp­sia) - Consider trans­fu­sion to Hb 10 g/dL
- Monitor fluid bal­ance to avoid vol­ume over­load
- Postpartum pro­phy­lac­tic anticoagulation for both vag­i­nal and Cae­
sar­ean deliv­er­ies for 6 weeks starting 6-12 hours after all­deliv­ery (no
sooner than 4 hours after epi­du­ral cath­e­ter removal). LMWH and war­fa­
rin are not contraindicated with breastfeeding.
Moyamoya dis­ease Cerebral revas­cu­lar­i­za­tion35 - At risk for reduced cere­bral blood flow due to gen­eral anes­the­sia
- Aggressive fluid hydra­tion prior to sur­gery
- Utilize intraoperative EEG
Valvular dis­ease Cardiac sur­gery36 - At risk for hem­or­rhage, stroke, renal fail­ure, and acute chest syn­drome
- For valve replace­ment, at risk of hemo­ly­sis, throm­bo­sis, and infec­tion
- Use incen­tive spi­rom­e­try, anticoagulation, and pro­phy­lac­tic anti­bi­ot­ics
- Maintain nor­mo­ther­mia to min­i­mize vaso­con­stric­tion and sick­ling
EEG, elec­tro­en­ceph­a­lo­gram; ENT, ear, nose, and throat; spO2, oxy­gen sat­u­ra­tion.

patients in each group devel­oped acute chest syn­drome.5 Based recommended with a post­trans­fu­sion goal of a Hb level no greater
on avail­­able evi­dence, some experts have con­ cluded that the than 11 g/dL in order to avoid hyper­vis­cos­ity. For patients with Hb
ben­e­fit of pre­op­er­a­tive trans­fu­sion is mostly related to increas­ing >9 g/dL under­go­ing mod­er­ate-risk sur­gery, RCE is recommended.
Hb rather than reduc­ing %HbS.13 Although the cur­rent evi­dence For patients with Hb >9g/dL with­out a severe phe­no­type under­
supporting pre­op­er­a­tive trans­fu­sion is low qual­ity, the ben­e­fits go­ing low-risk sur­gery, con­sider pro­ceed­ing with­out trans­fu­sion.13
out­weigh the harms; there­fore, pre­op­er­a­tive trans­fu­sion is still Patients under­go­ing emer­gency sur­gery may receive sim­ple trans­
recommended.13 fu­sion to increase Hb to 10 g/dL, if indi­cated. Sometimes, pre­op­
There is a lack of con­sen­sus on the best prac­tices for pre­op­er­ er­a­tive trans­fu­sion may not be accept­able due to the poten­tial for
a­tive and peripartum trans­fu­sions. High-qual­ity data are needed delaying a life-sav­ing sur­gery; there­fore, intraoperative or post­op­
in the era of hydroxy­ urea and other new dis­ ease-mod­ i­
fy­
ing er­a­tive trans­fu­sion may be con­sid­ered.13 See the sum­mary of trans­
ther­ a­
pies. We advise that cli­ ni­
cians fol­
low national and local fu­sion rec­om­men­da­tions in Table 3.
guide­ lines. According to the Amer­ i­
can Society of Hematology
trans­fu­sion guide­lines, in some sit­u­a­tions RCE may reduce the risk Alloimmunization
of acute chest syn­drome and pain cri­sis in patients with geno­type The inci­dence of alloimmunization in SCD ranges from 7% to 58%
HbSS/HbSβ0, Hb <9 g/dL, or a severe phenotype (characterized depending on age, num­ber of pre­vi­ous trans­fu­sions, and use of
as hav­ing a his­tory of stroke, recur­rent acute chest syn­drome, red cell phe­no­typic matching.16,17 The TAPS study noted no dif­fer­
or prior severe post­op­er­a­tive com­pli­ca­tions) or any per­son with ence in alloimmunization between patients receiv­ing pre­op­er­a­
SCD under­go­ing high-risk sur­gery. Patients with a severe phe­no­ tive trans­fu­sion com­pared to no trans­fu­sion likely due to the use
type are most likely to ben­e­fit from achiev­ing a post-RCE goal of of extended red cell phe­no­typic matching, and par­tic­i­pants had
%HbS <30%. For patients with Hb <9 g/dL with­out a severe phe­ pre­vi­ously received fewer trans­fu­sions com­pared to other stud­
no­type under­go­ing mod­er­ate-risk sur­gery, sim­ple trans­fu­sion is ies.15 When com­par­ing aggres­sive trans­fu­sion to con­ser­va­tive
Prakash Singh Shekhawat
Perioperative man­age­ment of sickle cell dis­ease  |  407
Table 2. Surgical risk clas­si­fi­ca­tion

Low risk Moderate risk High risk


Dental extrac­tions Head and neck sur­gery Cardiac sur­gery (eg, valve replace­ment)
Ophthalmologic pro­ce­dures (ie, laser sur­gery, Orthopedic sur­gery (eg, total hip replace­ment) Intrathoracic sur­gery
cat­a­ract sur­gery)
Hernia repair Urologic sur­gery Brain sur­gery
Dilation and curet­tage Cholecystectomy Transplant sur­gery (heart, lung, liver, kid­ney)
Wound debride­ment Splenectomy Major vas­cu­lar sur­gery (eg, aorta repair)
Endoscopy Hysterectomy Major spine sur­gery
Superficial tis­sue biopsy (eg, breast biopsy) Uterine abla­tion Surgeries requir­ing prolonged gen­eral
anes­the­sia >4 hours (eg, Whipple pro­ce­dure)

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/405/1851532/405oyedeji.pdf by guest on 13 December 2021


Cae­sar­ian sec­tion
Appendectomy

trans­fu­sion, an RCT showed an increased risk of alloimmuniza­ sim­i­lar to a patient’s usual inpa­tient acute pain plan and includ­ing
tion with aggres­sive trans­fu­sion; there­fore, many cen­ters pre­fer patient-con­trolled anal­ge­sia (PCA).7 The deci­sion to include con­
sim­ple trans­fu­sion over RCE when appro­pri­ate.5 Patients with a tin­u­ous basal opi­oid infu­sion vs on-demand dos­ing only should
his­tory of mul­ti­ple alloantibodies, delayed hemo­lytic trans­fu­sion be patient-spe­cific and deter­mined by the patient’s level of post­
reac­tion (DHTR), and/or hyperhemolysis have an increased risk op­er­a­tive pain and opi­oid tol­er­ance.19 Referral to the local acute
of adverse out­comes related to trans­fu­sion; there­fore, care­ful pain team is advised to ensure appro­pri­ate opi­ate dos­ing and
con­sid­er­ation should be given prior to trans­fu­sion. The risk of nonopiate adjuncts, such as a short course of non­ste­roi­dal anti-
trans­fu­sion-related com­pli­ca­tions should be discussed and doc­ inflam­ma­tory drugs (5-7 days if no renal risk), intra­ve­nous (IV) ket­
umented, and in some sit­ua ­ ­tions, trans­fu­sion is not pos­si­ble. In amine or lido­caine, regional blocks, and acet­amin­o­phen.19
the event an emer­gent trans­fu­sion is required, con­sider hav­ing
crossmatched blood avail­­able on-site even if there is no plan Intraoperative period and imme­di­ately after sur­gery
for trans­fu­sion. The blood should be anti­gen-neg­a­tive for any The most impor­ tant fac­tor to con­ sider intraoperatively is to
pos­i­tive antibodies and pre­vi­ous antibodies even if those anti­ avoid imbal­ances in vol­ume sta­tus, tem­per­a­ture, acid-base bal­
bodies are no lon­ger detect­able in order to min­i­mize the risk of ance, blood pres­ sure, and oxy­ gen­
a­tion since derange­ ments
DHTR. For life-threat­en­ing ane­mia, con­sider immu­no­sup­pres­sive increase red blood cell sick­ling, which can result in acute organ
ther­apy if trans­fu­sion is abso­lutely required.8,13,18 In patients with injury. It can also man­i­fest as a vaso-occlu­sive pain cri­sis, as acute
reli­gious objec­tions to trans­fu­sion or if trans­fu­sion is not pos­si­ chest syn­drome, as an acute kid­ney injury, or even as an ische­mic
ble due to severe alloimmunization/pre­vi­ous DHTR, it is impor­ stroke. Practical strat­e­gies to main­tain euvolemia include avoid­
tant to opti­mize the Hb in advance with med­i­ca­tions such as ing prolonged fasting prior to sur­gery with­out IV flu­ids, mon­i­
hydroxy­urea, voxelotor, and ery­throid-stim­u­lat­ing agents. tor­ing fluid intake and out­put, and decreas­ing IV flu­ids as soon
as patients are a ­ ble to main­tain ade­quate oral fluid intake. Avoid
Pain man­age­ment plan extremely cold or hot ambi­ent tem­per­a­tures pre­op­er­a­tively, in
The patient’s cur­rent pain reg­im­ en should be reviewed, and a the oper­at­ing room, and in the recov­ery space while using fluid
post­op­er­a­tive pain man­age­ment plan should be devel­oped, often and con­vec­tive warming tech­nol­ogy aggres­sively.

Table 3. Transfusion rec­om­men­da­tions based on sickle cell geno­type, hemo­glo­bin, and sur­gi­cal risk clas­si­fi­ca­tion

Hemoglobin
Sickle cell geno­type (g/dL) Surgical risk Transfusion rec­om­men­da­tion
HbSS or HbSβ - thal­as­se­mia
0
<9 Low or mod­er­ate Simple trans­fu­sion, par­tial exchange, or RCE
HbSS or HbSβ0- thal­as­se­mia >9 Low or mod­er­ate Partial exchange or RCE
HbSC, HbSβ -thal­as­se­mia, and HbSS on hydroxy­urea and
+
>9 Low No trans­fu­sion
ele­vated HbF with­out severe phe­no­type*
HbSC or HbSβ+- thal­as­se­mia >9 Moderate Partial exchange or RCE
All geno­types — High RCE
*Severe phe­no­type is defined as hav­ing a his­tory of stroke, recur­rent acute chest syn­drome, or prior severe sur­gi­cal com­pli­ca­tions.
HbSC, hemoglobin SC disease.

408  |  Hematology 2021  |  ASH Education Program


Postoperative period i­ta­tion 3 days a week. On fol­low-up in the hema­tol­ogy clinic, she
All patients with SCD should use an incen­tive spi­rom­e­ter post­ reported sig­nif­i­cant improve­ments in mobil­ity, range of motion,
op­er­a­tively since this has been shown to reduce the inci­dence and pain. She no lon­ger required the use of an assist device to
of atel­ec­ta­sis and acute chest syn­drome in hos­pi­tal­ized patients ambu­late and was a ­ ble to meet her goal of being a ­ ble to play
with SCD.20,21 Monitor for post­op­er­a­tive infec­tion, and if a patient’s with her grandchildren.
tem­per­a­ture is ≥38 °C, inves­ti­gate for a source of the fever with
blood and urine cul­tures, wound inspec­tion, and chest x-ray, as
indi­cated.8 Minimize the risk of hyp­oxia with the judi­cious use of Conclusion
anal­ge­sics to avoid respi­ra­tory depres­sion. If oxy­gen sat­u­ra­tion Preoperative opti­mi­za­tion is an essen­tial part of care for indi­vid­
falls by ≥2% below the patient’s base­line or is ≤94%, pro­vide sup­ u­als with SCD under­go­ing sur­gery. People with SCD are more
ple­men­tal oxy­gen and eval­u­ate the patient with con­sid­er­ation likely to have sur­gi­cal com­pli­ca­tions com­pared to the gen­eral
for post­op­er­a­tive com­pli­ca­tions such as acute chest syn­drome pop­u­la­tion, so it is imper­a­tive that con­sul­ta­tion with an SCD spe­
and pul­mo­nary embolism. The eval­u­a­tion often includes phys­i­ cial­ist be included in the opti­mi­za­tion plan. Appropriate opti­mi­

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/405/1851532/405oyedeji.pdf by guest on 13 December 2021


cal exam­i­na­tion, arte­rial blood gas, and chest imag­ing. Consider za­tion and man­age­ment prior to, dur­ing, and after sur­gery can
checking the met­a­bolic pro­file after sur­gery to assess for acute reduce the risk of adverse out­comes in indi­vid­u­als with SCD.
kid­ney injury and elec­tro­lyte derange­ments. More research is needed to improve perioperative man­age­ment
guide­lines for SCD.
Anticoagulation man­age­ment
SCD causes hyper­co­ag­u­la­bil­ity due to mul­ti­ple alter­ations in
Acknowledgments
hemo­sta­sis, includ­ing increased plate­let acti­va­tion, acti­va­tion
Charity I. Oyedeji rec­og­nizes sup­port from the Amer­i­can Soci­
of the clot­ting cas­cade, impaired fibri­no­ly­sis, and intra­vas­cu­lar
ety of Hematology Research Training Award for Fellows and the
hemo­ly­sis.22,23 The inci­dence of VTE in indi­vid­u­als with SCD is
Duke Center for Research to Advance Healthcare Equity, which
as high as 12% before the age of 40.24 Hospitalized adults with
is supported by the National Institute on Minority Health and
SCD should receive VTE pro­phy­laxis if there is no con­tra­in­di­ca­
Health Disparities under award num­ber U54MD012530.
tion since VTE in SCD is asso­ci­ated with increased mor­tal­ity.25
There is insuf­fi­cient evi­dence on post­op­er­a­tive VTE pro­phy­laxis
in chil­dren. For both pedi­at­ric and adult pop­u­la­tions, we sug­gest
Conflict-of-inter­est dis­clo­sure
Charity I. Oyedeji: no com­pet­ing finan­cial inter­ests to declare.
pre­scrib­ing VTE pro­phy­laxis based on national or local guide­
Ian J. Welsby: no com­pet­ing finan­cial inter­ests to declare.
lines. Most inpa­tient teams use low-molec­u­lar-weight hep­a­rin
(LMWH) or low-dose unfractionated hep­a­rin. Patients under­go­
ing sur­ger­ies that require prolonged immo­bi­li­za­tion, such as hip Off-label drug use
sur­gery, require extended VTE pro­phy­laxis (Table 1). Perioper­ Charity I. Oyedeji: nothing to disclose.
ative teams should con­sider the use of inter­mit­tent pneu­matic Ian J. Welsby: nothing to disclose.
com­pres­sion dur­ing the hos­pi­tal stay and encour­age early mobi­
li­za­tion. LMWH or war­fa­rin is recommended for post­par­tum VTE Correspondence
pro­phy­laxis in all­indi­vid­u­als with SCD who plan to breastfeed Charity I. Oyedeji, Division of Hematology, Department of Med­
since DOACs are contraindicated (Table 1).26 icine, Duke University School of Medicine, 315 Trent Dr, Hanes
House, Ste 261, DUMC Box 3939, Dur­ham, NC 27710; e-mail: char
­ity​­.oyedeji@duke​­.edu.

CLINICAL CASE (Con­tin­ued) References


Our patient’s perioperative risk scores were documented by 1. Modell B, Darlison M. Global epi­de­mi­­ol­ogy of haemoglobin dis­or­ders and
derived ser­vice indi­ca­tors. Bull World Health Organ. 2008;86(6):480-
the anes­the­si­­ol­o­gist as low risk, with phys­i­cal sta­tus con­sis­tent
487.
with mild sys­temic dis­ease with­out sub­stan­tial lim­i­ta­tions and 2. Kato GJ, Piel FB, Reid CD, et al. Sickle cell dis­ease. Nat Rev Dis Primers.
a low risk of hav­ing OSA. She received RCE 4 days before sur­ 2018;4(March):18010.
gery. Post-RCE Hb improved to 11.4 g/dL and HbS was 24.8%. 3. Buck J, Davies SC. Surgery in sickle cell dis­ease. Hematol Oncol Clin North
She under­went total right hip replace­ment under spi­nal anes­the­ Am. 2005;19(5):897-902, vii.
4. Koshy M, Weiner SJ, Miller ST, et al. Surgery and anes­the­sia in sickle cell dis­
sia with­out com­pli­ca­tions. Her post­op­er­a­tive Hb was 9.6 g/dL. ease. Cooperative Study of Sickle Cell Diseases. Blood. 1995;86(10):3676-3684.
An anes­the­sia acute pain ser­vice man­aged her post­op­er­a­tive Accessed 27 Sep­tem­ber 2021.
pain. A con­tin­u­ous infu­sion of ropivicane via a cath­e­ter inserted 5. Vichinsky EP, Haberkern CM, Neumayr L, et al; Preoperative Transfusion in
under ultra­sound guid­ance into the adduc­tor canal was supple­ Sickle Cell Disease Study Group. A com­par­i­son of con­ser­va­tive and aggres­
sive trans­fu­sion reg­i­mens in the perioperative man­age­ment of sickle cell
mented by IV hydromorphone PCA and a low-dose ketamine
dis­ease. N Engl J Med. 1995;333(4):206-213.
infu­sion. Her home meth­a­done was started once she was tol­ 6. Allareddy V, Roy A, Lee MK, et al. Outcomes of acute chest syn­drome in
er­at­ing a diet, and the PCA and ketamine were discontinued on adult patients with sickle cell dis­ease: pre­dic­tors of mor­tal­ity. PLoS One.
post­op­er­a­tive day (POD) 1. Cyclobenzaprine and ketorolac were 2014;9(4):e94387.
started once renal func­tion was deter­mined to be sta­ble. 7. Adjepong KO, Otegbeye F, Adjepong YA. Perioperative man­age­ment of
sickle cell dis­ease. Mediterr J Hematol Infect Dis. 2018;10(1):e2018032.
For post­op­er­a­tive pro­phy­laxis for deep vein throm­bo­sis, she 8. Walker I, Trompeter S, Howard J, et  al. Guideline on the peri-oper­a­tive
received 30 mg of LMWH twice a day for 30 days, which started man­age­ment of patients with sickle cell dis­ease: guide­line from the Asso­
on POD 1. She was discharged on POD 2 with out­pa­tient reha­bil­ ciation of Anaesthetists. Anaesthesia. 2021;76(6):805-817.

Prakash Singh Shekhawat


Perioperative man­age­ment of sickle cell dis­ease  |  409
9. Weinstein AS, Sigurdsson MI, Bader AM. Comparison of pre­ op­er­
a­tive 22. Kelley D, Jones LT, Wu J, Bohm N. Evaluating the safety and effec­tive­ness of
assess­ ment of patient’s met­ a­
bolic equiv­a­lents (METs) esti­ mated from venous throm­bo­em­bo­lism pro­phy­laxis in patients with sickle cell dis­ease.
his­tory ver­sus mea­sured by exer­cise car­diac stress test­ing. Anesthesiol Res J Thromb Thrombolysis. 2017;43(4):463-468.
Pract. 2018;2018(Sep­tem­ber):5912726. 23. Naik RP, Streiff MB, Lanzkron S. Sickle cell dis­ease and venous throm­bo­em­
10. Fleisher LA, Fleischmann KE, Auerbach AD, et al. 2014 ACC/AHA guide­line bo­lism: what the anticoagulation expert needs to know. J Thromb Throm-
on perioperative car­dio­vas­cu­lar eval­u­a­tion and man­age­ment of patients bolysis. 2013;35(3):352-358.
under­ go­ing non­ car­
diac sur­ gery: a report of the Amer­ i­
can College of 24. Brunson A, Lei A, Rosenberg AS, White RH, Keegan T, Wun T. Increased
Cardiology/Amer­i­can Heart Association Task Force on Practice Guidelines. inci­dence of VTE in sickle cell dis­ease patients: risk fac­tors, recur­rence and
Circulation. 2014;130(24):e278-e333. impact on mor­tal­ity. Br J Haematol. 2017;178(2):319-326.
11. Katz T, Schatz J, Roberts CW. Comorbid obstruc­ tive sleep apnea and 25. Novelli EM, Huynh C, Gladwin MT, Moore CG, Ragni MV. Pulmonary
increased risk for sickle cell dis­ease mor­bid­ity. Sleep Breath. 2018;22(3):797- embolism in sickle cell dis­ease: a case-con­trol study. J Thromb Haemost.
804. 2012;10(5):760-766.
12. Chung F, Abdullah HR, Liao P. STOP-Bang Questionnaire: a prac­ ti­
cal 26. Bates SM, Middeldorp S, Rodger M, James AH, Greer I. Guidance for the treat­
approach to screen for obstruc­tive sleep apnea. Chest. 2016;149(3):631- ment and pre­ven­tion of obstet­ric-asso­ci­ated venous throm­bo­em­bo­lism.
638. J Thromb Thrombolysis. 2016;41(1):92-128.
13. Chou ST, Alsawas M, Fasano RM, et  al. Amer­i­can Society of Hematology 27. Kamath AF, McGraw MH, Isra­el­ite CL. Surgical man­age­ment of osteonecro­

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/405/1851532/405oyedeji.pdf by guest on 13 December 2021


2020 guide­lines for sickle cell dis­ease: trans­fu­sion sup­port. Blood Adv. sis of the fem­o­ral head in patients with sickle cell dis­ease. World J Orthop.
2020;4(2):327-355. 2015;6(10):776-782.
14. Waldron P, Pegelow C, Neumayr L, et al. Tonsillectomy, adenoidectomy, 28. Falck-Ytter Y, Francis CW, Johanson NA, et al. Prevention of VTE in ortho­pe­
and myringotomy in sickle cell dis­ease: perioperative mor­bid­ity. Preop­ dic sur­gery patients: Antithrombotic Therapy and Prevention of Thrombosis,
erative Transfusion in Sickle Cell Disease Study Group. J Pediatr Hematol 9th ed: Amer­i­can College of Chest Physicians Evidence-Based Clinical
Oncol. 1999;21(2):129-135. Practice Guidelines. Chest. 2012;141(suppl 2):e278S-e325S.
15. Howard J, Malfroy M, Llewelyn C, et  al. The Transfusion Alternatives Pre- 29. Warrier R, Chauhan A, Athale U. Tonsillectomy and adenoidectomy for
operatively in Sickle Cell Disease (TAPS) study: a randomised, con­trolled, obstruc­tive sleep apnea in sickle cell ane­mia. Indian J Pediatr. 2010;77(6):669-
multicentre clin­i­cal trial. Lancet. 2013;381(9870):930-938. 672.
16. Chou ST, Jackson T, Vege S, Smith-Whitley K, Friedman DF, Westhoff 30. Martins RA, Soares RS, Vito FB, et al. Cholelithiasis and its com­pli­ca­tions
CM. High prev­ a­
lence of red blood cell alloimmunization in sickle cell in sickle cell dis­ease in a uni­ver­sity hos­pi­tal. Rev Bras Hematol Hemoter.
dis­ease despite trans­ fu­sion from Rh-matched minor­ ity donors. Blood. 2017;39(1):28-31.
2013;122(6):1062-1071. 31. Al-Salem AH. Splenic com­pli­ca­tions of sickle cell ane­mia and the role of sple­nec­
17. Lasalle-Williams M, Nuss R, Le T, et  al. Extended red blood cell anti­gen tomy. ISRN Hematol. 2011;2011(October 2010):864257.
matching for trans­fu­sions in sickle cell dis­ease: a review of a 14-year expe­ 32. Zhang N, Yao Y, Xue W, Wu S. Early pro­phy­lac­tic anticoagulation for por­tal
ri­ence from a sin­gle cen­ter (CME). Transfusion. 2011;51(8):1732-1739. vein sys­tem throm­bo­sis after sple­nec­tomy: a sys­tem­atic review and meta-
18. Noizat-Pirenne F, Habibi A, Mekontso-Dessap A, et al. The use of rituximab anal­y­sis. Biomed Rep. 2016;5(4):483-490.
to pre­vent severe delayed haemolytic trans­fu­sion reac­tion in immu­nized 33. Olujohungbe A, Burnett AL. How I man­age priapism due to sickle cell dis­
patients with sickle cell dis­ease. Vox Sang. 2015;108(3):262-267. ease. Br J Haematol. 2013;160(6):754-765.
19. Brandow AM, Carroll CP, Creary S, et  al. Amer­i­can Society of Hematology 34. Jain D, Atmapoojya P, Colah R, Lodha P. Sickle cell dis­ease and preg­nancy.
2020 guide­lines for sickle cell dis­ease: man­age­ment of acute and chronic Mediterr J Hematol Infect Dis. 2019;11(1):e2019040.
pain. Blood Adv. 2020;4(12):2656-2701. 35. Smith ER, Scott RM. Surgical man­age­ment of moyamoya syn­drome. Skull
20. Niss O, Cole-Jenkins C, Davis B, et al. Prevention of acute chest syn­drome Base. 2005;15(1):15-26.
by implementing a stan­dard­ized pro­cess to improve incen­tive spi­rom­e­try 36. Crawford TC, Carter MV, Patel RK, et al. Management of sickle cell dis­ease
use in hos­pi­tal­ized patients with sickle cell dis­ease. Blood. 2017;130(suppl in patients under­go­ing car­diac sur­gery. J Card Surg. 2017;32(2):80-84.
1):132.
21. Bellet PS, Kalinyak KA, Shukla R, Gelfand MJ, Rucknagel DL. Incentive spi­
rom­e­try to pre­vent acute pul­mo­nary com­pli­ca­tions in sickle cell dis­eases. © 2021 by The Amer­i­can Society of Hematology
N Engl J Med. 1995;333(11):699-703. DOI 10.1182/hema­tol­ogy.2021000274

410  |  Hematology 2021  |  ASH Education Program


MANAGEMENT STRATEGIES FOR SICKLE CELL DISEASE

Treatment dilemmas: strategies for priapism,


chronic leg ulcer disease, and pulmonary
hypertension in sickle cell disease

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/411/1851512/411azbell.pdf by guest on 13 December 2021


Roberta C.G. Azbell1,2 and Payal Chandarana Desai1,3
The Ohio State University Wexner Medical Center, Department of Internal Medicine, Columbus, OH; 2Division of Hospital Medicine, Columbus, OH;
1

and 3Division of Hematology and Oncology, Columbus, OH

Sickle cell disease is a disorder characterized by chronic hemolytic anemia and multiorgan disease complications.
Although vaso-occlusive episodes, acute chest syndrome, and neurovascular disease frequently result in complication
and have well-documented guidelines for management, the management of chronic hemolytic and vascular-related com-
plications, such as priapism, leg ulcers, and pulmonary hypertension, is not as well recognized despite their increasing
reported prevalence and association with morbidity and mortality. This chapter therefore reviews the current updates
on diagnosis and management of priapism, leg ulcers, and pulmonary hypertension.

LEARNING OBJECTIVES
• Differentiate the types of priapism presenting in patients with SCD and understand current therapeutic and pre­
ventative regimens
• Understand phenotypic presentation of and current therapeutic options for chronic leg ulcerations
• Be familiar with the current screening, diagnostic, and therapeutic recommendations for pulmonary hypertension
in SCD

Background tologist. He followed regularly with his pediatric hematol­


Sickle cell disease (SCD) is a β­hemoglobinopathy that is ogist and is currently taking hydroxyurea, folic acid, and
hallmarked by red blood cell (RBC) sickling and chronic cholecalciferol. His SCD­related complications include
hemolytic anemia. Point mutations in both β­globin genes elevated transcranial Doppler velocity, 3 episodes of ACS,
lead to an unstable hemoglobin molecule (HbS),1 which and once­yearly VOE hospitalizations. At today’s visit, he
polymerizes into chains to form a “sickled” shape. This reports new episodes of painful, intermittent erections that
ultimately causes vaso­occlusion, intravascular hemolysis, occur about 4 nights per week and last about 3 to 4 hours.
and endothelial dysfunction. Although the complications They awaken him from sleep and sometimes resolve with
of SCD are broad, there are recognized overlapping “sub­ rigorous exercise.
phenotypes” of the disease. The high hemoglobin/high
viscosity subtype is associated with increased episodes of
acute chest syndrome (ACS) and vaso­occlusive episodes Priapism
(VOEs),2 whereas the hyperhemolytic subtype is associated Definition, types, prevalence, and pathophysiology
with increased vascular complications such as priapism, Priapism is defined as an erection lasting greater than 4
leg ulceration, and pulmonary hypertension.2­4 This chapterhours. Priapism can occur in patients with SCD of all ages
reviews the definitions, pathophysiology, diagnosis, and but may peak between 20 and 25 years of age.5 In SCD,
management of those vascular complications. increased RBC sickling occurs within the corpus cavernosa,
possibly as a result of abnormal endothelial adherence, rel­
ative acidosis during sleep and during erection, and defi­
cient erection control mechanisms involving an impaired
CLINICAL CASE nitric oxide–cyclic GMP pathway.6 Priapism is characterized
A 24­year­old man with SCD (genotype hemoglobin SS) as low flow (ischemic) or high flow (nonischemic/arterial).
Prakash
presents to a clinic to establish care Singh
with an adult Shekhawat
hema­ Nonischemic priapism is caused by trauma and damage to

Treatment priapism, ulcers, pulmonary hypertension | 411


the cavernosal artery and is not a uro­logic emer­gency as there is on priapism has not been well stud­ied. There are, how­ever, case
no last­ing dam­age to penile tis­sue. Ischemic priapism, how­ever, reports suggesting hydroxy­urea and trans­fu­sion are ben­efi ­ ­cial in
can cause last­ing tis­sue dam­age and requires prompt uro­logic pre­vent­ing recur­rent epi­sodes of stuttering priapism.16,17
eval­u­at­ion. When ische­mic priapism lasts less than 4 hours and
recurs, it is referred to as stuttering. Both stuttering and ische­ Novel ther­a­pies
mic priapism are described as pain­ful, which can help dif­fer­en­ti­ A phase 2 sin­gle-arm trial is cur­rently inves­ti­gat­ing the effi­cacy
ate them from nonischemic priapism.7 and safety of crizanlizumab in the man­age­ment of SCD-related
priapism (ClinicalTrials​­
.gov Identifier NCT03938454). Crizanli­
Management zumab is a mono­clo­nal anti­body targeting the leu­ko­cyte adhe­
Although increased hydra­ tion and pain man­ age­ ment can be sion mod­u­la­tor P-selectin and was approved by the US Food and
attempted for priapism last­ing less than 4 hours,8 first-line ther­ Drug Administration in 2019 to reduce VOEs in SCD.2
apy for acute ische­mic priapism is decom­pres­sion via aspi­ra­tion,
followed by intracavernous injec­tion (ICI) of sym­pa­tho­mi­metic
agents.7,9 Penile blood is then sent for blood gas test­ing to con­

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/411/1851512/411azbell.pdf by guest on 13 December 2021


firm eti­­ol­ogy, although in patients with SCD, it is often ische­mic.
CLINICAL CASE (Con­tin­ued)
Sympathomimetics work via α-medi­ated vaso­con­stric­tion within
the cor­pus cavernosa, and uro­logic data sug­gest that aspi­ra­tion The patient is diag­nosed with stuttering priapism and is started
plus ICI is more effec­tive than aspi­ra­tion alone.7 These data were on daily baclofen for pre­ven­tion. The fre­quency of epi­sodes
supported in a pro­spec­tive study of 15 patients with sickle cell decreases over 2 years, and he remains clin­ic­ ally sta­ble for the
ane­mia.8 next 10 years. However, he now reports a new, recur­rent wound
RBC exchange trans­fu­sions have also been shown to be safe on his inner right ankle.
and effec­tive in both acute and pre­ven­ta­tive man­age­ment of pri­
apism.10 There have been pre­vi­ously described adverse effects
char­ac­ter­ized by cere­bral vas­cu­lar events (   Association of SCD, Leg ulcers
priapism, exchange transfusion, and neurological events syn­ Definition, types, prev­a­lence, and path­o­phys­i­­ol­ogy
drome) in this set­ting,11,12 but it is thought to be more related to The prev­a­lence of sickle cell leg ulcers (SCLUs) is unknown, but
post­trans­fu­sion hemo­glo­bin and asso­ci­ated increased vis­cos­ity, many patients report their first ulcer by the sec­ond decade of
rather than the actual trans­fu­sion itself. life. The pro­posed path­o­phys­i­­ol­ogy in SCD involves predispos­
Although no other ther­a­pies have been shown to be effec­tive ing vasculopathy, local skin insult, pain-medi­ated neu­ro­genic
in large tri­als for the acute man­age­ment of priapism, case reports inflam­ma­tion, and low ten­sile strength of scar tis­sue.3 SCLUs are
sug­gest some ther­a­pies may be effec­tive in the pre­ven­ta­tive set­ often located in the lower extrem­i­ties, spe­cif­i­cally where there
ting. A 2017 Cochrane review ana­lyz­ing the data on these ther­ is poor cir­cu­la­tion, thin skin, and less sub­cu­ta­ne­ous fat (eg, peri­
a­pies, which included ephed­ rine, sildenafil, stilboestrol, and malleolar).3,4 Ulcers can be sin­gu­lar, stuttering, or chronic and fre­
etilefrine, found no dif­fer­ence in effi­cacy between treat­ment and quently recur at the site of pre­vi­ous ulcer­a­tions. A distinguishing
pla­cebo in the pre­ven­tion of recur­rent priapism.13 Oral sym­pa­ fea­ture from other venous ulcers is the pres­ence of pain.3 Prior
tho­mi­met­ics such as ephed­rine, pseudoephedrine, and etilefrine trauma is a poten­tial predisposing fac­tor.3,18 Other asso­ci­ated
work sys­tem­i­cally via the same mech­a­nism as sym­pa­tho­mi­metic fac­tors include HbSS geno­type,3,19 male sex, age older than
ICI. Other pro­posed ther­a­pies dis­rupt the abnor­mal nitric oxide– 20 years, and reduced hemo­glo­bin F (HbF) lev­els.19 There has
cGMP path­way and include phosphodiesterase 5 inhib­i­tors such been prior debate in the lit­er­a­ture as to the role of hydroxy­urea
as sildenafil and tadalfil. Although some case reports sup­port and pre­dis­po­si­tion to SCLUs. However, sev­eral large reviews
their use for ­reduc­tion in recur­rence with long-term dos­ing,13,14 have dis­ puted this. Of the 505 patients being screened for
the data on the use of sildenafil in pul­mo­nary hyper­ten­sion (PH) pul­mo­nary hyper­ten­sion, 39% were tak­ing hydroxy­urea. There
raise con­cern for increased VOEs with its use.15 Hormonal ther­a­ was no sta­tis­ti­cal dif­fer­ence in prev­a­lence of leg ulcers among
pies sup­press andro­genic effects on penile erec­tion9 and include patients with and with­out hydroxy­urea.20
GNrH a ­go­ nists, estro­gens, antiandrogens, and ketoconazole.
Despite reported suc­ cess in pre­ vent­
ing recur­ rent priapism Management
events, sig­nif­i­cant adverse effects include erec­tile dys­func­tion, There are var­i­ous pro­posed inter­ven­tions for SCLUs, ­includ­ing
mood changes, and hot flashes and poten­tial for car­dio­vas­cu­ pharmaceutical treat­ments (vas­cu­lar, anti­ox­i­dants, growth fac­tors,
lar com­pli­ca­tions.9,14 Baclofen, a selec­tive γ-aminobutyric acid HbF syn­the­sis stim­u­la­tors), top­i­cal treat­ments (wound care, anti­
recep­tor ago­nist, was effec­tive in treat­ment for sleep-related bi­ot­ics, growth fac­tors, ste­roids), and ­sur­gi­cal/nonpharmaceutical
priapism in a UK cohort study but was not effec­tive in the SCD agents. A 2020 Cochrane review found 6 ran­dom­ized con­trol
cohort.14 ­tri­als (RCTs) inves­ti­gat­ing these ther­a­pies.4 One study inves­ti­gat­
Although none of these ther­a­pies have been inves­ti­gated for ing isoxsuprine, a vas­cu­lar agent, had mixed results when eval­
detu­mes­cence in acute priapism epi­sodes, they have been anec­ u­at­ing com­plete ulcer clo­sure. Of the 30 patients, only 23.9%
dot­ally used with some suc­cess. The com­bi­na­tion of pseudo­ healed (of those, 63.6% treat­ment vs 36.4% pla­cebo), 36.9% had
ephedrine and baclofen has been par­tic­u­larly effec­tive in some improved ulcers (47% treat­ment vs 53% pla­cebo), 15.2% had no
patients within our prac­tice and can be trialed while awaiting change, and 23.9% had dete­ri­o­ra­tion of ulcers.4,21 Arginine buty­
(but should not delay) urol­ogy con­sul­ta­tion (Figure 1). Regarding rate, an HbF stim­u­la­tor, dem­on­strated 30% com­plete clo­sure
the main­stays of SCD ther­a­pies, the direct effect of hydroxy­urea com­pared with 8% in the stan­dard local care arm.4,22

412  |  Hematology 2021  |  ASH Education Program


3RVVLEOH EHQHILWVRI+8
WUDQVIXVLRQ WKHUDS\ ORQJWHUP
3UHYHQWLRQ
RUDOV\PSDWKRPLPHWLFV
KRUPRQDO WKHUDSLHV

/DVWLQJKRXUV
VWXWWHULQJ

,QFUHDVHG RUDORU,9K\GUDWLRQ
$FXWH 3DLQPDQDJHPHQW
3VHXGRHSKHGULQH EDFORIHQ

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/411/1851512/411azbell.pdf by guest on 13 December 2021


3ULDSLVP

(While awaiting urology can trial


acute stuttering therapies)

8URORJLFFRQVXOWDWLRQ IRU
,VFKHPLF LQWUDFDYHUQRVDO DVSLUDWLRQ 
V\PSDWKRPLPHWLF LQMHFWLRQ
/DVWLQJKRXUV

$UWHULDO 1RLQWHUYHQWLRQ QHHGHG

Figure 1. How I treat priapism. For episodes lasting greater than 4 hours, urgent urologic consultation for prompt aspiration +/- sym­
pathomimetic injection is recommended. For stuttering episodes, it is reasonable to attempt increased hydration and pain manage­
ment. Long-term oral sympathomimetics may be beneficial in the prevention of recurrent episodes, as well as therapy with hydroxy­
urea (HU) or chronic transfusions. Hu, hydroxyurea; IV, intravenous.

Wound care. Local wound care is cru­cial to treating SCLUs used to eval­u­ate for response, and if the response is pos­i­tive,
and involves debride­ment, infec­tion con­trol, and main­te­nance trans­fu­sions are con­tin­ued until com­plete ulcer clo­sure.18
of a moist wound envi­ron­ment.19 Appropriate com­pres­sive ban­
dages and gar­ments (eg, stock­ings) and Unna boots are impor­ Novel ther­a­pies. A phase 2 tri­ple-blind RCT assessing the tol­
tant in maintaining a moist envi­ron­ment for healing (Figure 2). er­a­bil­
ity and effi­ cacy of top­ i­
cal sodium nitrate is under way
Topical anti­bi­ot­ics are not rou­tinely recommended due to risk (­ClinicalTrials​­.gov Identifier NCT02863068), after the 2014 phase 1
of con­tact sen­si­ti­za­tion, bac­te­rial resis­tance, and lack of mois­ cohort trial results were prom­is­ing and dem­on­strated a significant
ture bal­ance.19 increase in periwound blood flow, as well as dose-depen­dent
decreases in SCLU size.23
Pain man­age­ment. SCLUs cause sig­nif­i­cant pain and mor­bid­ In addi­tion, a dou­ble-blind RCT is eval­u­at­ing the safety and
ity, and man­age­ment should include ade­quate anal­ge­sic ther­ effi­cacy of intra­der­mal deferoxamine patches (ClinicalTrials​­.gov
apy. Topical anes­thet­ics may be used, but severe pain fre­quently Identifier NCT04058197).
war­ rants opi­ oid ther­ apy. Neuropathic pain may respond to
selec­tive sero­to­nin recep­tor inhib­i­tors, pregabalin, gabapentin,
or tri­cy­clic anti­de­pres­sants. Given the chro­nic­ity of some ulcers,
they may require chronic, not just acute, reg­i­mens.19 CLINICAL CASE (Con­tin­ued)
Our patient’s ulcer com­pletely resolved with wound care and a
Transfusions.  Chronic trans­fu­sions decrease HbS per­cent­age, short trans­fu­sion pro­to­col. He required a 6-month dose increase
RBC sick­ling, and hemo­ly­sis-related pathol­ogy. Transfusions in his chronic pain reg­ i­
men and did not have any fur­ ther
have been used in both acute and pre­ven­ta­tive set­tings19 for recur­rences over the past 6 years. At his office visit today, he
SCLUs and as perioperative sup­port for patients who require notes wors­en­ing dyspnea with reg­u­lar activ­i­ties and increas­
sur­gery.3 There are no RCTs to deter­mine goal hemo­glo­bin, HbS ing fatigue.
­per­cent­age, or dura­tion of ther­apy. Generally, a 6-month trial is

Prakash Singh Shekhawat


Treatment priapism, ulcers, pul­mo­nary hyper­ten­sion  |  413
&RPSUHVVLYH EDQGDJHV
&RPSUHVVLRQJDUPHQWV
/RFDOGHEULGHPHQW
Wound Care 0RLVWZRXQGEHGPDLQWHQDQFH
7UHDWPHQW RIVXSHULPSRVHG
LQIHFWLRQV

7RSLFDODQHVWKHWLFV
16$,'VRSLRLGVGHSHQGLQJRQ
Analgesia VHYHULW\ RISDLQ
$GHTXDWHLQFUHDVHG DQDOJHVLFV

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/411/1851512/411azbell.pdf by guest on 13 December 2021


IRUGUHVVLQJFKDQJHV

$YRLGLOOILWWLQJ VKRHV
$YRLGWUDXPD
Prevention 7UHDWPHQW RIXQGHUO\LQJ
GLVHDVH 6&'WKHUDS\
RSWLPL]DWLRQ 

Figure 2. Multimodal approach to SCLUs. The treatment of leg ulcers requires comprehensive wound care, adequate analgesics, and
preventative measures. NSAID, nonsteroidal anti-inflammatory drug.

Pulmonary hyper­ten­sion to the lack of evi­dence dem­on­strat­ing treat­ment ben­e­fit.29


Presentation and diag­no­sis The Amer­ i­
can Thoracic Society published guide­ lines in 2014
Progressive dyspnea on exer­tion or exer­cise lim­i­ta­tions, espe­ regard­ing risk strat­i­fi­ca­tion, screen­ing, and man­age­ment of
cially with exertional hyp­oxia, should prompt fur­ther inves­ti­ga­ PH in patients with SCD. Based on evi­dence that suggested
tion for PH.24 On exam­i­na­tion, patients may have signs of right 13% of patients with a nor­mal echo­car­dio­gram develop an ele­
heart fail­ure, includ­ing ele­vated jug­u­lar venous pul­sa­tion, accen­ vated TRV after 3 years of fol­low-up,30 the Amer­i­can Thoracic
tu­ated or fixed S2 split­ting, tri­cus­pid regur­gi­ta­tion (TR) mur­mur, Society recommended a screen­ing echo­car­dio­gram every 1 to
and/or periph­eral edema. 3 years in all­patients with SCD.31 However, fol­low-up stud­ies sug­
Although right heart cath­e­ter­i­za­tion (RHC) is the gold stan­ gest that the rate of pro­gres­sion to PH may actu­ally be slower.32
dard of diag­no­sis, its inva­sive­ness has led to the devel­op­ment of The most recent National Institutes of Health and Amer­ i­
can
more prac­ti­cal prog­nos­tic tools to pre­dict the pres­ence of PH. TR Society of Hematology guide­lines rec­om­mend against rou­tine
max­i­mum veloc­ity on echo­car­dio­gram can esti­mate pul­mo­nary screen­ing in asymp­tom­atic patients with SCD33,34 and rec­om­
arte­rial sys­tolic pres­sure. A TR veloc­ity (TRV) more than 2.5 m/s mend obtaining a diag­nos­tic echo­car­dio­gram only for symp­
was dem­on­strated to pre­dict the pres­ence of PH,24 and recent tom­atic patients (Figure 3).
stud­ies have shown that greater than 50% of patients with a TRV
of 2.9 m/s are diag­nosed with PH on RHC.25 Elevated brain natri­ Definition, types (groups), prev­a­lence, and path­o­phys­i­­ol­ogy
uretic peptide (BNP) lev­els are also asso­ci­ated with the pres­ence PH is defined as a rest­ ing mean pul­ mo­ nary arte­rial pres­
sure
of PH, and ele­vated N-terminal-pro-BNP lev­els are an inde­pen­ (mPAP) more than 25 mm Hg by RHC.24 In SCD, recur­rent hemo­ly­
dent risk fac­tor for mor­tal­ity in patients with SCD.24,26,27 A 2011 sis and its dis­rup­tion of the vas­cu­lar endo­the­lium ulti­mately leads
pro­spec­tive study in France found that in patients with a TRV to smooth mus­cle pro­lif­er­a­tion and adven­ti­tial fibro­blast accu­
from 2.5 to 2.8 m/s, NT-pro-BNP more than 164, and a 6-min­ute mu­la­tion.35 This, in turn, leads to increased pul­mo­nary vas­cu­lar
walk dis­tance less than 333 m, the pos­i­tive pre­dic­tive value of resis­tance, right ven­tric­u­lar (RV) dys­func­tion, and decreased
diag­nos­ing PH on RHC was 62% (false-pos­it­ ive rate of 7%).28 car­diac out­put.36 The prev­al­ence of PH is thought to be 6% to
10% in SCD36 and can be fur­ther char­ac­ter­ized by the loca­tion of
Screening hyper­ten­sion rel­a­tive to the cap­il­lary sys­tem.
Despite the asso­ci­a­tion of PH with increased mor­tal­ity, there Precapillary or pul­mo­nary arte­rial hyper­ten­sion (PAH) is defined
is no con­sen­sus regard­ing screen­ing guide­lines, par­tially due by a mPAP greater than 25 mm Hg and nor­mal left ven­tric­u­lar end-

414  |  Hematology 2021  |  ASH Education Program


Signs and Symptoms
• '\VSQHD DWUHVWRUZLWKH[HUWLRQWKDWLVRXWRI
SURSRUWLRQWRNQRZQFRQGLWLRQ
• +\SR[HPLDDWUHVWRUZLWKH[HUWLRQ
• &KHVW SDLQDWUHVWRUZLWKH[HUWLRQ
• ,QFUHDVHLQH[HUFLVHOLPLWDWLRQ
• +LVWRU\RIUHFXUUHQWK\SR[HPLDDWUHVWRUZLWKH[HUWLRQ
• (YLGHQFHRIVOHHSGLVRUGHUHGEUHDWKLQJ
K\SR[HPLD
• +LVWRU\RIV\QFRSHRUSUHV\QFRSH
• 6ZLWKSUHGRPLQDQW3RUQHZXQH[SHFWHGPXUPXU 759PV
• 6LJQVRIKHDUWIDLOXUHDQGRUIOXLGRYHUORDG
And one or both of the following:

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/411/1851512/411azbell.pdf by guest on 13 December 2021


0:'P
2EWDLQD %13!SJP/
5HIHUWR
GLDJQRVWLF
3+VSHFLDOLVW
HFKRFDUGLRJUDP

759PV
Comorbid conditions
• &RQQHFWLYHWLVVXH GLVHDVH
• 8QGHUO\LQJSXOPRQDU\GLVHDVH DVWKPD
*Assist with SCD-specific periprocedural planning:
Disease complications associated with PH • &RQVLGHUWUDQVIXVLRQV
• /HJXOFHUV • +ROGDQWLFRDJXODWLRQLIDSSOLFDEOH
• 3ULDSLVP • (QVXUHZDUPEODQNHWDQGSRVWSURFHGXUH,9IOXLGV
• $OEXPLQXULD DUHUHDGLO\DYDLODEOH

Figure 3. How I approach PH screening. Every patient with SCD should be screened for “red-flag” signs and symptoms of PH at
routine visits (when there is no acute illness present). Any red-flag sign/symptom, or the presence of certain comorbidities or dis­
ease-specific complications, should be further evaluated with a diagnostic echocardiogram. A moderately elevated TRV, or a mild
elevation with other prognostic factors, should warrant further referral to a specialist. IV, intravenous; 6MWD, 6-minute walk distance.

dia­stolic pres­sure less than 15 mm Hg and falls under PH group I. were fur­ther supported by the find­ings of Minniti et al38 in 2009.
Postcapillary or pul­mo­nary venous hyper­ten­sion is defined by an In that cohort study, patients with RHC-con­firmed PH receiv­ing
mPAP greater than 25 mm Hg with an ele­vated left ven­tric­u­lar bosentan or ambrisentan had reduced BNP lev­els, reduced TRVs,
end-dia­stolic pres­sure higher than 15 mm Hg,24 and it can be fur­ and improved 6-minute walk test.
ther grouped into PH World Health Organization groups II to V
based on eti­­ol­ogy. Pre- and postcapillary are dis­trib­uted evenly PDE-5 inhib­i­tors. Although sildenafil has been shown to de­
in patients with SCD, and patients often have fea­tures of both.25,35 crease TRV and BNP,39 a 2011 mul­ti­cen­ter trial of sildenafil was
stopped pre­ma­turely due to increased hos­pi­tal­i­za­tion for VOEs.
Management Potential con­tri­bu­tions to these events include lack of SCD ther­
PAH-spe­cific ther­a­pies include the fol­low­ing. apy opti­mi­za­tion prior to the inter­ven­tion and sildenafil-spe­cific
mech­a­nisms that may cause myal­gias and inflam­ma­tory pain.15
Endothelin-recep­tor antag­o­nists. Endothelin 1 is a potent vaso­
con­stric­tor that is ele­vated in SCD. The Randomized, Placebo- Other PAH ther­a­pies. Prostanoids (epoprostenol, treprostinil,
Controlled, Double-Blind, Multicenter, Parallel Group Study to As­ iloprost, beraprost) have not spe­cif­i­cally been stud­ied in SCD
sess the Efficacy, Safety and Tolerability of Bosentan in Patients but have shown ben­e­fit in non-SCD patients with PH.40 In addi­
With Symptomatic Pulmonary Arterial Hypertension Associated tion, 1 case study dem­on­strated decreased pul­mo­nary arte­rial
With Sickle Cell Disease (ASSET)-1 and -2 tri­als in 2009 were de­ sys­tolic pres­sure in 10 patients treated with 5 days of L-argi­nine.41
signed to inves­ti­gate the safety, effi­cacy, and tol­er­a­bil­ity of bosen­
tan, an endothelin 1 recep­tor antag­o­nist, in patients with PAH and PH ther­a­pies
PH, respec­tively. Unfortunately, both tri­als were ter­mi­nated early Intensification of SCD-spe­cific ther­a­pies. The direct ben­e­fit of
due to slow enroll­ment. However, the lim­ited data obtained dem­ hydroxy­urea on PH has not been stud­ied, but it reduces epi­
on­strated tol­er­a­bil­ity and increased exer­cise tol­er­ance,37 which sodes of VOEs and ACS, both of which are asso­ci­ated with an

Prakash Singh Shekhawat


Treatment priapism, ulcers, pul­mo­nary hyper­ten­sion  |  415
increase in PA pres­sures. Hydroxyurea should there­fore indi­rectly 3. Minniti CP, Kato GJ. How we treat sickle cell patients with leg ulcers clin­i­cal
decrease the risk of acute right-sided heart fail­ure and death in cases 40th anni­ver­sary issue. Am J Hematol. 2016;91(1):22-30.
4. Martí-Carvajal AJ, Knight-Madden JM, Martinez-Zapata MJ. Interventions for
patients with under­ly­ing PH.24 The effect of chronic trans­fu­sions treating leg ulcers in peo­ple with sickle cell dis­ease. Cochrane Database
on PH has also not been for­mally stud­ied; how­ever, because it Syst Rev. 2014;2014(12):CD008394.
reduces com­pli­ca­tions of SCD sim­i­lar to hydroxy­urea, it should 5. Emond AM, Holman R, Hayes RJ, Serjeant GR. Priapism and impo­tence in
the­o­ret­i­cally improve out­comes in patients with under­ly­ing PH. homo­zy­gous sickle cell dis­ease. Arch Intern Med. 1980;140(11):1434-1437.
6. Bivalacqua TJ, Burnett AL. Priapism: new con­cepts in the path­o­phys­i­­ol­ogy
and new treat­ment strat­e­gies. Curr Urol Rep. 2006;7(6):497-502.
Treatment of comorbidities.  Patients who have con­cur­rent right 7. Mon­ta­gue DK, Jarow J, Broderick GA, et al; Members of the Erectile Dys­
heart fail­ure or albu­min­uria can be treated with diuret­ics but cau­ function Guideline Update Panel; Amer­i­can Urological Association. Amer­
tiously, given the pos­si­bil­ity of vol­ume deple­tion and increased i­can Urological Association guide­line on the man­age­ment of priapism. J
RBC sick­ling.24,31 In addi­tion, patients with under­ly­ing sleep ap­ Urol. 2003;170(4, pt 1):1318-1324.
8. Mantadakis E, Ewalt DH, Cavender JD, Rogers ZR, Buchanan GR. Outpatient
nea and throm­botic dis­ease should be man­aged accord­ingly. penile aspi­ra­tion and epi­neph­rine irri­ga­tion for young patients with sickle
cell ane­mia and prolonged priapism. Blood. 2000;95(1):78-82.
Novel ther­a­pies

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/411/1851512/411azbell.pdf by guest on 13 December 2021


9. Broderick GA. Priapism and sickle–cell ane­mia: diag­no­sis and non­sur­gi­cal
A phase 2 dou­ble-blind RCT is eval­u­at­ing the safety, tol­er­a­bil­ ther­apy. J Sex Med. 2012;9(1):88-103.
ity, and effi­cacy of riociguat (ClinicalTrials​­.gov NCT02633397) in 10. Ballas SK, Lyon D. Safety and effi­cacy of blood exchange trans­fu­sion for
priapism com­pli­cat­ing sickle cell dis­ease. J Clin Apher. 2016;31(1):5-10.
patients with SCD. Several of the sec­ond­ary out­come mea­sures 11. Siegel JF, Rich MA, Brock WA. Association of sickle cell dis­ease, priapism,
spe­cif­i­cally relate to mark­ers of PH. exchange trans­fu­sion and neu­ro­log­i­cal events: ASPEN syn­drome. J Urol.
In addi­tion, a phase 3 ran­dom­ized par­al­lel-arm trial is eval­u­at­ 1993;150(5, pt 1):1480-1482.
ing the effect of RBC exchange ther­apy com­pared with stan­dard 12. Rackoff WR, Ohene-Frempong K, Month S, Scott JP, Neahring B, Cohen AR.
Neurologic events after par­tial exchange trans­fu­sion for priapism in sickle
of care on car­dio­vas­cu­lar risk (ClinicalTrials​­.gov NCT04084080).
cell dis­ease. J Pediatr. 1992;120(6):882-885.
Secondary out­comes spe­cif­i­cally eval­u­ate the effect on mark­ers 13. Chinegwundoh FI, Smith S, Anie KA. Treatments for priapism in boys
of PH. and men with sickle cell dis­ease. Cochrane Database Syst Rev. 2017;9(9):
CD004198.
Conflict-of-inter­est dis­clo­sure 14. Johnson MJ, McNeillis V, Chiriaco G, Ralph DJ. Rare dis­or­ders of pain­ful
erec­tion: a cohort study of the inves­ti­ga­tion and man­age­ment of stuttering
Roberta C.G. Azbell: no conflicts to disclose.
priapism and sleep-related pain­ful erec­tion. J Sex Med. 2021;18(2):376-384.
Payal Chandarana Desai: con­sul­tant for GBT for grant review; 15. Machado RF, Barst RJ, Yovetich NA, et al; walk-PHaSST Investigators
advi­sory board for Forma; funding from the National Institutes of and Patients. Hospitalization for pain in patients with sickle cell dis­ease
Health, University of Tennessee, and University of Pittsburgh; and treated with sildenafil for ele­vated TRV and low exer­cise capac­ity. Blood.
speaker for Novartis. 2011;118(4):855-864.
16. Anele UA, Mack AK, Resar LMS, Burnett AL. Hydroxyurea ther­apy for pria­
pism pre­ven­tion and erec­tile func­tion recov­ery in sickle cell dis­ease: a case
Off-label drug use report and review of the lit­er­a­ture. Int Urol Nephrol. 2014;46(9):1733-1736.
Roberta C.G. Azbell: During the discussion of prevention of recur­ 17. Hassan A, Jam’a A, Al Dabbous IA. Hydroxyurea in the treat­ment of sickle
rent priapism, we discuss the off-label use of oral sympathomimet­ cell asso­ci­ated priapism. J Urol. 1998;159(5):1642.
ics, baclofen, phosphodiesterase 5 inhibitors, hormonal therapies 18. Trent JT, Kirsner RS. Leg ulcers in sickle cell dis­ease. Adv Skin Wound Care.
2004;17(8):410-416.
(GnRH agonists, estrogens, antiandrogens), ketoconazole, hydroxy­ 19. Ladizinski B, Bazakas A, Mistry N, Alavi A, Sibbald RG, Salcido R. Sickle cell
urea, and exchange transfusion. With regard to management dis­ease and leg ulcers. Adv Skin Wound Care. 2012;25(9):420-428.
of SCLUs, we discuss the off-label use of exchange transfusions, 20. Minniti CP, Taylor JG VI, Hildesheim M, et  al. Laboratory and echo­car­di­
hydroxyurea, topical sodium nitratre, and intradermal deferoxamine og­ra­phy mark­ers in sickle cell patients with leg ulcers. Am J Hematol.
2011;86(8):705-708.
patches. We discuss the off-label use of hydroxyurea in pulmonary
21. Serjeant GR, Howard C. Isoxsuprine hydro­chlo­ride in the ther­apy of sickle
hypertension. cell leg ulcer­a­tion. West Indian Med J. 1977;26(3):164-166.
Payal Chandarana Desai: During the discussion of prevention of 22. McMahon L, Tamary H, Askin M, et al. A ran­dom­ized phase II trial of argi­nine
recurrent priapism, we discuss the off-label use of oral sympatho­ buty­rate with stan­dard local ther­apy in refrac­tory sickle cell leg ulcers. Br J
mimetics, baclofen, phosphodiesterase 5 inhibitors, hormonal ther­ Haematol. 2010;151(5):516-524.
23. Minniti CP, Gorbach AM, Xu D, et al. Topical sodium nitrite for chronic leg
apies (GnRH agonists, estrogens, antiandrogens), ketoconazole, ulcers in patients with sickle cell anae­mia: a phase 1 dose-find­ing safety
hydroxyurea, and exchange transfusion. With regard to manage­ and tol­er­a­bil­ity trial. Lancet Haematol. 2014;1(3):e95-e103.
ment of SCLUs, we discuss the off-label use of exchange transfusions, 24. Ataga KI, Klings ES. Pulmonary hyper­ten­sion in sickle cell dis­ease: diag­
hydroxyurea, topical sodium nitratre, and intradermal deferoxamine no­sis and man­age­ment. Hematology Am Soc Hematol Educ Program.
2014;2014(1):425-431.
patches. We discuss the off-label use of hydroxyurea in pulmonary
25. Gordeuk VR, Castro OL, Machado RF. Pathophysiology and treat­ment of
hypertension. pul­mo­nary hyper­ten­sion in sickle cell dis­ease. Blood. 2016;127(7):820-828.
26. Machado RF, Anthi A, Steinberg MH, et al. Risk of death in sickle cell dis­
Correspondence ease. Vasc Med. 2006;296(3):310-318.
Payal Chandarana Desai, The Ohio State University, 1800 Cannon 27. Machado RF, Hildesheim M, Mendelsohn L, Remaley AT, Kato GJ, Gladwin
MT. NT-pro brain natri­uretic pep­tide lev­els and the risk of death in the coop­
Drive, 1100D Lincoln Tower, Columbus, OH 43210. e-mail: payal​
er­at­ ive study of sickle cell dis­ease. Br J Haematol. 2011;154(4):512-520.
­.desai@osumc​­.edu. 28. Parent F, Bachir D, Inamo J, et  al. A hemo­dy­namic study of pul­mo­nary
hyper­ten­sion in sickle cell dis­ease. N Engl J Med. 2011;365(1):44-53.
References 29. Yawn BP, Buchanan GR, Afenyi-Annan AN, et al. Management of sickle cell
1. Rees DC, Williams TN, Gladwin MT. Sickle-cell dis­ ease. Lancet. 2010; dis­ease: sum­ mary of the 2014 evi­ dence-based report by expert panel
376(9757):2018-2031. mem­bers. JAMA. 2014;312(10):1033-1048.
2. Ofori-Acquah SF. Sickle cell dis­
ease as a vas­
cu­
lar dis­
or­
der. Expert Rev 30. Ataga KI, Moore CG, Jones S, et al. Pulmonary hyper­ten­sion in patients with
Hematol. 2020;13(6):645-653. sickle cell dis­ease: a lon­gi­tu­di­nal study. Br J Haematol. 2006;134(1):109-115.

416  |  Hematology 2021  |  ASH Education Program


31. Klings ES, Machado RF, Barst RJ, et al; Amer­i­can Thoracic Society Ad Hoc 37. Barst RJ, Mubarak KK, Machado RF, et al; ASSET study group. Exercise
Committee on Pulmonary Hypertension of Sickle Cell Disease. An offi­cial capac­ity and haemodynamics in patients with sickle cell dis­ease with pul­
Amer­i­can Thoracic Society clin­i­cal prac­tice guide­line: diag­no­sis, risk strat­ mo­nary hyper­ten­sion treated with bosentan: results of the ASSET stud­ies.
i­fi­ca­tion, and man­age­ment of pul­mo­nary hyper­ten­sion of sickle cell dis­ Br J Haematol. 2010;149(3):426-435.
ease. Am J Respir Crit Care Med. 2014;189(6):727-740. 38. Minniti CP, Machado RF, Coles WA, Sachdev V, Gladwin MT, Kato GJ. Endo­
32. Desai PC, May RC, Jones SK, et al. Longitudinal study of echo­car­di­og­ra­phy- thelin recep­tor antag­o­nists for pul­mo­nary hyper­ten­sion in adult patients
derived tri­cus­pid regurgitant jet veloc­ity in sickle cell dis­ease. Br J Haema- with sickle cell dis­ease. Br J Haematol. 2009;147(5):737-743.
tol. 2013;162(6):836-841. 39. Machado RF, Martyr S, Kato GJ, et al. Sildenafil ther­apy in patients with sickle
33. Liem RI, Lanzkron S, D Coates T, et  al. Amer­i­can Society of Hematology cell dis­ease and pul­mo­nary hyper­ten­sion. Br J Haematol. 2005;130(3):445-
2019 guide­lines for sickle cell dis­ease: car­dio­pul­mo­nary and kid­ney dis­ 453.
ease. Blood Adv. 2019;3(23):3867-3897. 40. Machado RF, Gladwin MT. Chronic sickle cell lung dis­ease: new insights
34. National Heart, Lung, and Blood Institute. Evidence-based man­age­ment into the diag­no­sis, path­o­gen­e­sis and treat­ment of pul­mo­nary hyper­ten­
of sickle cell dis­ease: expert panel, 2014. Accessed 26 May 2019. https:­/​/​­ sion. Br J Haematol. 2005;129(4):449-464.
­​­www​­.nhlbi​­.nih​­.gov​­/sites​­/default​­/files​­/media​­/docs​­/sickle​­-cell​­-disease​ 41. Morris CR, Morris SM Jr, Hagar W, et al. Arginine ther­apy: a new treat­ment
­-report 020816_0.pdf for pul­mo­nary hyper­ten­sion in sickle cell dis­ease? Am J Respir Crit Care
35. Wood KC, Gladwin MT, Straub AC. Sickle cell dis­ease: at the cross­roads of Med. 2003;168(1):63-69.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/411/1851512/411azbell.pdf by guest on 13 December 2021


pul­mo­nary hyper­ten­sion and dia­stolic heart fail­ure. Heart. 2020;106(8):562-
568.
36. Potoka KP, Gladwin MT. Vasculopathy and pul­mo­nary hyper­ten­sion in sickle © 2021 by The Amer­i­can Society of Hematology
cell dis­ease. Am J Physiol Lung Cell Mol Physiol. 2015;308(4):L314-L324. DOI 10.1182/hema­tol­ogy.2021000275

Prakash Singh Shekhawat


Treatment priapism, ulcers, pul­mo­nary hyper­ten­sion  |  417
MDS: BEYOND A ONE - SIZE - FITS -ALL APPROACH

Have we reached a molecular era


in myelodysplastic syndromes?

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/418/1851957/418voso.pdf by guest on 13 December 2021


Maria Teresa Voso1,2 and Carmelo Gurnari1,3
Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy; 2Santa Lucia Foundation, IRCCS, Neuro­Oncohematology,
1

Rome, Italy; and 3Translational Hematology and Oncology Research Department, Taussig Cancer Center, Cleveland Clinic, OH

Myelodysplastic syndromes (MDS) are characterized by heterogeneous biological and clinical characteristics, leading
to variable outcomes. The availability of sophisticated platforms of genome sequencing allowed the discovery of recur-
rently mutated genes, which have led to a new era in MDS. This is reflected by the 2016 update of the World Health
Organization classification, in which the criteria to define MDS with ringed sideroblasts include the presence of SF3B1
mutations. Further, the detection of somatic mutations in myeloid genes at high variant allele frequency guides the diag-
nostic algorithm in cases with cytopenias, unclear dysplastic changes, and normal karyotypes, supporting MDS over
alternative diagnoses. SF3B1 mutations have been shown to play a positive prognostic role, while mutations in ASXL1,
EZH2, RUNX1, and TP53 have been associated with a dismal prognosis. This is particularly relevant in lower- and interme-
diate-risk disease, in which a higher number of mutations and/or the presence of “unfavorable” somatic mutations may
support the use of disease-modifying treatments. In the near future, the incorporation of mutation profiles in currently
used prognostication systems, also taking into consideration the classical patient clinical variables (including age and
comorbidities), will support a more precise disease stratification, eg, the assignment to targeted treatment approaches
or to allogeneic stem cell transplantation in younger patients.

LEARNING OBJECTIVES
• Discuss the impact of somatic mutations in MDS and how to integrate molecular data within current diagnostic
and prognostic classifications
• Review the clinical management of patients with MDS according to the disease mutational status and identify
actionable targets

CLINICAL CASE 1 findings, a myelodysplastic syndrome (MDS) with RS and


A 78­year­old woman with an unremarkable medical his­ single­lineage dysplasia was diagnosed.
tory presented to our clinic with isolated macrocytic
anemia (hemoglobin [Hb], 9 g/dL; mean corpuscular vol­
ume, 107 fL). Metabolic as well as nutritional deficien­ Introduction
cies and other common causes of anemia were ruled out MDS is a highly heterogeneous group of disorders defined
after an initial diagnostic workup, while the endogenous by the presence of ineffective hematopoiesis with periph­
erythropoietin (EPO) level was 358 mU/mL. The bone eral blood (PB) cytopenias, dysplastic changes in ≥10% of
marrow (BM) evaluation revealed expansion of erythro­ cells of one or more myeloid lineages, and a variable risk
poiesis (70% of marrow cellularity) with marked dysplas­ of progression to acute myeloid leukemia (AML).1 Clinical
tic changes in 20% of erythroid cells and the presence of presentations of MDS reflect its biological heterogeneity.
1% blasts and 12% ring sideroblasts (RS) by iron staining Based on the major risk­prognostication systems, the In­
(Perls’ Prussian blue; Figure 1). The karyotype was nor­ ternational Prognostic Scoring System (IPSS) and its revi­
mal, while next­generation sequencing (NGS) mutational sion (IPSS­R),2, 3 two­thirds of patients will present with
analysis identified a canonical SF3B1K700E mutation at a lower­risk disease (LR­MDS) and are generally treated with
variant allelic frequency (VAF) of 21%. Based on these supportive care. In these cases the goal of treatment is the

418 | Hematology 2021 | ASH Education Program


mye­ loid gene muta­ tions on sur­vival in patients with MDS.8-10
The num­ber of muta­tions itself has been shown to play a sig­nif­
i­cant prog­nos­tic role by most stud­ies, with patients car­ri­ers of
over 3 driver muta­tions char­ac­ter­ized by reduced sur­vival and a
higher prob­a­bil­ity of leu­ke­mia pro­gres­sion.11-13 This is par­tic­u­larly
rel­e­vant in lower- or inter­me­di­ate-risk MDS, in which a higher
num­ber of somatic muta­tions, or the pres­ence of “unfa­vor­able”
muta­tions (ie, TP53), may refine the prog­no­sis and set the indi­
ca­tion for DMT or HSCT.14 In a sur­vey of 104 genes, Haferlach et
al showed by uni­var­i­ate anal­y­sis that among the 25 genes found
to affect sur­vival, only 5 (ASXL1, KRAS, PRPF8, RUNX1, and SF3B1)
retained sig­nif­i­cance after correcting for confounding fac­tors.8
These muta­tions were incor­po­rated into a novel hybrid geno­
mic-clin­i­cal model that more accu­rately predicted MDS patients’

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/418/1851957/418voso.pdf by guest on 13 December 2021


prog­no­sis when com­pared to both the IPSS-R and the “gene-
only” model.8 Another study from the Cleveland Clinic iden­ti­fied
one favor­able (SF3B1) and 5 unfa­vor­able gene muta­tions (ASXL1,
EZH2, NPM1, RUNX1, and TP53) in a cohort of 508 MDS patients.10
Figure 1. Perls’ Prussian blue stain of the patient’s BM smear
These data led to the design of a score, includ­ing age, IPSS-R,
showing RS. Classical type 3 RS are shown (according to Mufti
and the muta­tional sta­tus of EZH2, SF3B1, and TP53, that out­
et al57), with multiple siderotic granules in a perinuclear position
performed the clas­ si­cal MDS risk-pre­ dic­tion mod­ els (Table 1)
surrounding the nucleus (lower-right cell) or encompassing at
and could pre­dict lon­gi­tu­di­nal dis­ease evo­lu­tion regard­less of
least one-third of the nuclear circumference (middle cell).
treat­ment.10 Of note, these scores unveiled discrepancies in risk
assign­ment for a not neg­li­gi­ble frac­tion of patients. For instance,
if tak­ing into con­sid­er­ation the IPSS-R, the hybrid “powered”
ame­lio­ra­tion of cytopenias, represented by ane­mia in about 70% model upstaged 58% of patients pre­vi­ously cat­e­go­rized as inter­
of cases, through trans­fu­sion sup­port, eryth­ro­poi­e­tin admin­is­tra­ me­di­ate risk to high as well as 26% from lower to high risk.9
tion, or lenalidomide treat­ment in MDS with del(5q). Conversely, More recently, by apply­ing Bayes­ian net­works and Dirichlet
higher-risk MDS patients, defined by inter­me­di­ate to very high pro­cesses in a study enroll­ing 2043 patients anno­tated for the
IPSS-R,4 require dis­ease-mod­i­fy­ing treat­ments (DMT) such as hy­ pres­ence of muta­tions in 47 fre­quently mutated genes, Bersanelli
pomethylating agents (HMA) and a timely eval­u­a­tion for allo­ge­ et al iden­ti­fied 8 clus­ters, each char­ac­ter­ized by dif­fer­ent genetic
neic hema­to­poi­etic stem cell trans­plan­ta­tion (HSCT), the most lesions, clin­i­cal phe­no­types, and prog­no­ses.15 By using a ran­dom-
effec­tive treat­ment and the only poten­tially cura­tive option.5 effects Cox mul­ti­state model, the authors com­bined 63 clin­i­cal
and geno­mic var­i­ables to pro­file indi­vid­ual patients’ prog­no­ses,
Integration of molec­u­lar data into clas­si­fi­ca­tion yield­ing a C-index of 0.75 and 0.74 when cross-val­i­dated inter­
and prog­nos­ti­ca­tion sys­tems nally and in an inde­pen­dent cohort, respec­tively.15 This sem­i­nal
Paralleling the var­i­able clin­i­cal pre­sen­ta­tion, MDS biol­ogy is fas­ study paves the way for an MDS prog­nos­ti­ca­tion sys­tem not only
ci­nat­ingly com­plex. In the last decade, the avail­abil­ity of sophis­ account­ing for tra­di­tional clin­i­cal and mor­pho­log­i­cal var­i­ables
ti­cated genome-sequenc­ing plat­forms led to the iden­ti­fi­ca­tion but also incor­po­rat­ing recent advances in MDS biol­ogy.
of recur­rently mutated genes, shed­ding light on new aspects of
the dis­ease patho­bi­­ol­ogy. Accordingly, del(5q) and SF3B1 muta­ Limitations to the appli­ca­tion of geno­mics
tions have been incor­po­rated in the most recent World Health to MDS diag­nos­tics
Organization (WHO) clas­si­fi­ca­tion and define spe­cific MDS sub­ If the idea of a hybrid geno­mic-clin­i­cal model sounds appeal­ing,
types.1 However, the com­mon risk-prog­nos­ti­ca­tion tools (ie, the sev­eral fac­tors still play a role in deter­min­ing the actual fea­si­
IPSS and IPSS-R), used world­wide for clin­i­cal deci­sion-mak­ing bil­ity and repro­duc­ibil­ity of such an approach. There is no gen­
and trial eli­gi­bil­ity, consider only clin­i­cal/cyto­ge­netic var­i­ables. eral con­sen­sus yet on sequenc­ing tech­niques and data anal­y­sis
Briefly, the IPSS score groups patients in a 4-tier model (low, across lab­o­ra­to­ries. The clin­i­cal impact of some var­i­ants on pa­
inter­me­di­ate-1, inter­me­di­ate-2, and high),2 while the more recent tients’ out­comes remains unknown, and of utmost impor­tance,
IPSS-R includes 5 risk groups (very low, low, inter­me­di­ate, high, bio­log­i­cal dif­fer­ences in muta­tion char­ac­ter­is­tics such as VAF,
and very high), improv­ing strat­i­fi­ca­tion of kar­yo­type and of the type (mis­sense vs trun­cat­ing), and loca­tion (func­tional domain
pro­ por­tion of blasts and tak­ ing into account the sever­ ity of vs other posi­tions across the gene) are still not homo­ge­neously
cytopenias (Table 1).3 The IPSS-R, ini­tially devel­oped in patients con­sid­ered. For instance, TP53 muta­tions are clas­si­cally con­sid­
receiv­ing sup­port­ive treat­ment, has since been val­i­dated in ered prognostically unfa­vor­able. However, a recent coop­er­a­tive
patients treated with DMT and ther­apy-related MDS.6,7 study dem­on­strated that their clin­i­cal impact is beyond the mere
The advent of NGS and the grow­ing body of evi­dence con­ binary pres­ence/absence.16 Indeed, Bernard et al con­firmed that
cerning the prog­nos­tic impact of somatic muta­tions in MDS have besides VAF, the TP53 alle­lic state is a strong pre­dic­tive fac­tor,
posed new chal­lenges, such as the incor­po­ra­tion of this infor­ show­ing that patients with monoallelic TP53 muta­tions and VAF
ma­tion into the established risk-strat­i­fi­ca­tion mod­els. To this ≤22% were char­ac­ter­ized by sur­vival and response to ther­apy
end, sev­eral stud­ies have dem­on­strated the impact of somatic sim­i­lar to wild-type coun­ter­parts.16

Prakash Singh Shekhawat


Toward a tai­lored molec­u­lar approach in MDS | 419
Table 1.  Comparison of existing prog­nos­ti­ca­tion mod­els in MDS

Model Variables C index Reference


International Prognostic Scoring System (IPSS)2 Karyotype 0.65 2
BM blasts %
Number of cytopenias
Revised International Prognostic Scoring System Karyotype 0.67 3
(IPSS-R)3 BM blasts %
Degree of cytopenias
MD Anderson Cancer Center (MDACC)55 Karyotype 0.65 49
BM blasts %
Degree of cytopenias
Age
Performance sta­tus

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/418/1851957/418voso.pdf by guest on 13 December 2021


Prior trans­fu­sion
World Health Organization-based Prognostic Scoring Karyotype 0.65 50
System (WPSS)56 WHO cat­e­gory
Transfusion require­ment
Genoclinical model according to Haferlach et al8 Gender NA 8
Age
Degree of cytopenias (PLT, Hb)
BM blasts %
Karyotype
ASXL1, CBL, ETV6, EZH2, KRAS, LAMB4, NCOR2,
NF1, NPM1, NRAS, PRPF8, RUNX1, TET2, TP53
Genoclinical model according to Nazha et al10 Karyotype 0.71 10
BM blasts %
Degree of cytopenias
Age
WHO cat­e­gory
Disease ontog­eny (sec­ond­ary vs de novo)
TP53, EZH2, and SF3B1
Genoclinical model according to Bersanelli et al15 63 clin­i­cal and molec­u­lar var­i­ables 0.75 14
PLT, plate­lets; NA, not avail­­able.

In addi­tion, the costs of genome-scan­ning approaches (despite Key clin­i­cal point


drop­ping dra­mat­i­cally in the last years) still rep­re­sent an obsta­cle In Case 1, the results of NGS were deter­ mi­ nant in assigning
for a world­wide hybrid geno­mic-clin­i­cal appli­ca­tion used on a the patient to the cor­rect WHO-defined cat­e­gory (MDS-RS), in
day-by-day basis, espe­cially in devel­op­ing countries. defin­ing the prog­no­sis, and in guid­ing the clin­i­cal man­age­ment
toward targeted treat­ment with luspatercept.

From biol­ogy to clas­si­fi­ca­tion and tai­lored treat­ment:


CLINICAL CASE 1 (Con­t in­u ed) the case of SF3B1 muta­tions
Our 78-year-old woman had an LR-MDS (IPSS score, 0; IPSS-R, As show­cased by our patient, SF3B1-mutant MDS is an emblem
2). According to the com­ mon guide­ lines, the patient was of geno­type/phe­no­type asso­ci­a­tion, char­ac­ter­ized by a spe­cific
started on alpha EPO (40 000 UI sub­cu­ta­ne­ously once weekly, prog­nos­tic out­come and the avail­abil­ity of a tai­lored treat­ment.
increased to 80 000 UI weekly after 12 weeks). However, after SF3B1 is the most com­monly mutated splic­ing-fac­tor gene and
6 months the hemo­glo­bin lev­els remained unchanged, and she defi­nes a well-char­ac­ter­ized group of MDS with RS, lead­ing to its
even­ tu­
ally became trans­ fu­
sion depen­ dent (1 red blood cell def­i­ni­tion as “the lord of the rings” of MDS.19-21 The asso­ci­a­tion is
unit [RBC]/week). At this point, con­sid­er­ing the diag­no­sis of so strong that if the muta­tion is pres­ent, the thresh­old of RS pro­
SF3B1-mutant MDS with RS and sin­gle-lin­e­age dys­pla­sia and por­tion in the BM to sat­isfy the WHO MDS-RS cri­te­ria low­ers from
the erythropoiesis-stimulating agent fail­ure, the patient was 15% to 5% (as in our patient).1 In a recent study, the International
enrolled in the MEDALIST trial and started on luspatercept.17 Working Group argues for the rec­og­ni­tion of SF3B1-mutant MDS
A pro­ gres­sive increase in hemo­ glo­bin lev­els and a reduc­ as a dis­tinct noso­logic entity and pro­poses spe­cific diag­nos­tic
tion of trans­fu­sion needs were observed after 8 weeks, with cri­te­ria (Table 2).22 This plea is supported by the strong geno­
achieve­ment of a minor hema­to­logic improve­ment—ery­throid type/phe­no­type asso­ci­a­tion char­ac­ter­ized by the prev­a­lence of
response (1 RBC unit/3 weeks), according to the revised Inter­ female sex, a higher degree of ane­mia and lower BM blast per­
national Working Group 2018 hema­to­log­i­cal response cri­te­ria— cent­ ages, a favor­ able dis­ease course, the pres­ ence of SF3B1
still persisting after 27 months on treat­ment.18 muta­tions as a founding event in the major­ity of cases, and the
recent avail­abil­ity of a spe­cific treat­ment.17,22

420  |  Hematology 2021  |  ASH Education Program


Table 2.  Proposed diag­nos­tic cri­te­ria for the subentity of MDS with mutated SF3B1

1. Cytopenia defined by stan­dard hema­to­logic ref­er­ence val­ues


2. Presence of a somatic SF3B1 muta­tion
3. Isolated ery­throid or multilineage dys­pla­sia, with or with­out RS
4. BM blasts <5% and PB blasts <1%
5. WHO cri­te­ria not fulfilling any other cat­e­gory
6. Normal kar­yo­type or any cyto­ge­netic abnor­mal­ity other than del(5q), mono­somy 7, inv(3) or abnor­mal 3q26, com­plex (≥3)
7. Presence of any addi­tional somatic gene muta­tion other than RUNX1 and/or EZH2*
*Additional JAK2V617F, CALR, or MPL muta­tions strongly sup­port the diag­no­sis of myelodysplastic syndrome/myeloproliferative neoplasm with ring
sideroblasts and thrombocytosis (MDS/MPN-RS-T). Adapted from Malcovati et al.22

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/418/1851957/418voso.pdf by guest on 13 December 2021


SF3B1, together with del(5q), rep­re­sents an ideal exem­plum gene mye­loid panel iden­ti­fied somatic muta­tions in 4 genes
of how progresses in dis­ease patho­bi­­ol­ogy can not only improve (DNMT3A, RUNX1, SRSF2, and TET2) at a median VAF of 12%
dis­ease clas­si­fi­ca­tion but also open pos­si­bil­i­ties for tai­lored treat­ (range, 1.5%-12.6%). Based on the pres­ence of severe multilin­
ments. The haploinsufficiency of CSNK1A1 and RPS14 rep­re­sents ear cytopenias and uni­lin­ear dys­pla­sia and the evi­dence of <5%
the molec­u­lar lesions under­ly­ing the exqui­site sen­si­tiv­ity of del(5q) BM blasts, a diag­no­sis of MDS unclas­si­fi­able (MDS-U) with sin­
cases to lenalidomide.23,24 The pres­ence of SF3B1 muta­tions gen­er­ gle-lin­e­age dys­pla­sia was made. The patient’s IPSS score was
ates aber­rant splic­ing events due to misrecognition of 3′ splice site 0.5 (inter­me­di­ate-1), while IPSS-R was 4 (inter­me­di­ate). Taking
with deg­ra­da­tion of about 50% of the aber­rant mRNAs via non­ into con­sid­er­ation the young age at pre­sen­ta­tion, the sever­ity
sense-medi­ated mRNA decay.25 Moreover, the ABCB7 and PPOX of cytopenias lead­ing to trans­fu­sion depen­dency of RBCs and
genes, involved in mito­ chon­ drial iron metab­ o­lism, have been plate­lets, and the muta­tional pro­file, a deci­sion to pro­ceed to
found to be sig­nif­i­cantly downregulated in SF3B1-mutant MDS and upfront HSCT was made. She had no major com­pli­ca­tions from
are thought to be asso­ci­ated with the pres­ence of RS.22,26 These the trans­plan­ta­tion pro­ce­dure and is pres­ently in com­plete
molec­u­lar dis­tur­bances gen­er­ate a higher degree of inef­fec­tive remis­sion (CR) at 24 months from HSCT.
eryth­ro­poi­e­sis and, there­fore, of ane­mia. Luspatercept is a recom­
bi­nant inhib­i­tor of transforming growth fac­tor β a ­ ble to reduce
SMAD2/3 sig­nal­ing, over­com­ing the impair­ment imprinted by the Myeloid gene muta­tion anal­y­sis as a help­ful diag­nos­tic
genetic lesion and enabling late-stage ery­throid dif­fer­en­ti­a­tion. In tool for MDS
the sin­gle-arm phase 2 trial (PACE-MDS) enroll­ing ane­mic LR-MDS As shown by the patient’s pre­sen­ta­tion, the num­ber and type of
patients, luspatercept induced hema­to­logic improve­ment in 69% muta­tional events iden­ti­fied by NGS were help­ful not only in sup­
of RS-pos­i­tive cases vs 43% of RS-neg­a­tive cases. When looking at porting the diag­no­sis of MDS but also for decid­ing to pro­ceed
SF3B1 sta­tus, hema­to­logic improve­ment was enriched in mutated to HSCT. As men­tioned before, somatic muta­tions (except SF3B1)
patients (77% vs 40% in neg­a­tive cases).27 These results were later are not part of cur­rent clas­si­fi­ca­tion schemes of MDS, whose hall­
con­firmed in patients with MDS-RS by the ran­dom­ized phase 3 mark remains the pres­ence of BM dys­pla­sia, and there is no sin­gle
MEDALIST trial, which showed that luspatercept (given at a dose muta­tion that can be con­sid­ered 100% patho­gno­monic of the dis­
of 1-1.75 mg/kg sub­cu­ta­ne­ously every 21 days) induced trans­fu­ ease. However, the assess­ment of dys­pla­sia in hypocellular cases
sion inde­pen­dence in 38% of patients at 8 weeks vs 13% in the with low blast counts and nor­mal kar­yo­types poses chal­lenges
pla­cebo arm.17 This trial led to the approval of luspatercept by the even well-expe­ri­enced mor­phol­o­gists. In these cases, NGS may
US Food and Drug Administration and the Euro­pean Medicines add impor­tant infor­ma­tion since over 90% of patients with mye­
Agency for patients with LR-MDS with RS and trans­fu­sion-depen­ loid dis­eases have been shown to pres­ent somatic muta­tions.11,12
dent ane­mia, who have an unsat­is­fac­tory response to or are inel­i­ Indeed, when using a well-constructed gene panel, the absence
gi­ble for eryth­ro­poi­e­tin-based ther­apy. of any molec­u­lar lesion in a patient with iso­lated cytopenia with­
out BM dys­pla­sia and a nor­mal kar­yo­type has a high neg­a­tive pre­
dic­tive value and should prompt the con­sid­er­ation of alter­na­tive
diag­noses (eg, auto­im­mune cytopenias), espe­cially in youn­ger
CLINICAL CASE 2 patients.28 Conversely, the iden­ ti­
fi­
ca­
tion of somatic muta­
tions
A 55-year-old woman was admit­ted to our hos­pi­tal after notic­ may under­pin a diag­no­sis of MDS, as in our patient, or direct phy­
ing the appear­ance of pete­chiae over her arms and legs and si­cians toward alter­na­tive diag­noses (ie, large gran­u­lar lym­pho­
after a find­ing of pan­cy­to­pe­nia (Hb, 9 g/dL; abso­lute neu­tro­ cytic leu­ke­mia in the case of STAT3 muta­tions).29 When apply­ing
phil count, 0.8 × 109/L; plate­lets, 13 109/L) at a com­plete blood NGS to rou­tine diag­nos­tics, clonal hema­to­poi­e­sis of indetermined
count eval­u­a­tion. Her BM aspi­ra­tion was hypocellular, and the poten­tial (CHIP) must be taken into account, since it is a very fre­
kar­yo­type was nor­mal. A core biopsy con­firmed the BM hypo­ quent find­ing in older adult indi­vid­u­als (up to 60% over the age
cellularity and showed <5% CD34+ cells with the pres­ence of of 80) and is char­ac­ter­ized by muta­tions usu­ally affect­ing one
rare micromegakaryocytes and a nor­mal reticulin-staining pat­ iso­lated gene (fre­quently, DNMT3A, TET2, and ASXL1), at a 2% to
tern. The patient was started on EPO 40 000 UI/week, but no 30% VAF.14,30-32 In the con­text of CHIP, a high num­ber of muta­tions
changes in Hb lev­els were observed. NGS anal­y­sis using a 30- and increased alle­lic bur­den are indi­ca­tions for fre­quent hema­to­
Prakash Singh Shekhawat
Toward a tai­lored molec­u­lar approach in MDS | 421
Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/418/1851957/418voso.pdf by guest on 13 December 2021
Figure 2.  Model of disease evolution in MDS. Upper panel: risk factors commonly associated with the development of myeloid disor­
ders (germline variants, smoking, aging, and cancer treatment exposure such as chemo/radiotherapy). Middle panel: a hypothetical
model of clonal evolution in which the founding event (or germline predisposition lesion) leads to subsequent acquisition/loss of new
somatic mutations in a linear/branching fashion. Lower panel: the typical acquisition of methylation during MDS progression, which
leads to the silencing of genes such as tumor suppressor and the disruption of many cell pathways (DNA repair, apoptosis, cell cycle,
cell adherence).

log­i­cal fol­low-up since these muta­tions may be the pro­dromes Key clin­i­cal points
for the onset of MDS.14,30,31 It is worth­while to under­line here that In Case 2 the deci­sion to pro­ceed to HSCT upfront was mostly
the pres­ence of cytopenias and BM dys­pla­sia remains an essen­tial based on clin­i­cal con­sid­er­ations, includ­ing the PB tri­lin­ear cyto­
require­ment for the diag­no­sis of MDS, while CHIP refers to a con­ penia lead­ing to RBC and plate­let trans­fu­sion depen­dency. The
di­tion in which only somatic muta­tional events are pres­ent.1 Other avail­abil­ity of the muta­tion pro­file, which showed the pres­ence
four-letters acronyms are used to define the case of patients with of 4 muta­tions also affect­ing RUNX1 and SRSF2 genes, was fur­
somatic mutations and cytopenias in the absence of BM dysplasia ther sup­port­ive of the diag­no­sis of MDS and of the high prob­a­bil­
(CCUS, clonal cytopenia of uncertain significance), BM dysplasia ity of rapid dis­ease pro­gres­sion.
without cytopenia and clonal events (IDUS, idiopathic dysplasia of
unknown significance), and cytopenia without dysplasia and Considerations on allo­ge­neic HSCT in patients with MDS
mutations (ICUS, idiopathic cytopenias of uncertain significance).14 in the molec­u­lar era
The increas­ ing num­ber of somatic muta­ tions also mir­ rors Although allo­ge­neic HSCT rep­re­sents the only cura­tive option
dis­ease pro­gres­sion, which is an intrin­sic char­ac­ter­is­tic of MDS in MDS, only 10% to 15% of patients will even­tu­ally undergo this
(Figure 2). This implies not only lin­ear and branching clonal evo­ pro­ce­dure. Indeed, the demo­graph­ics of the dis­ease (ie, most
lu­tion, defined by the acqui­si­tion or loss of somatic muta­tions, patients are diag­nosed with MDS at ages >70) con­sti­tute a major
respec­tively, but also increased DNA meth­yl­a­tion lev­els.33,34 For fac­tor lim­it­ing HSCT acces­si­bil­ity. However, the risk of relapse
these rea­sons, reassessment of the muta­tional pro­file at the time remains the major out­ come deter­ mi­
nant even after HSCT. In
of dis­ease pro­gres­sion is indi­cated to ensure treat­ment opti­mi­ this con­text, Della Porta et al iden­ti­fied lesions in ASXL1, RUNX1,
za­tion, espe­cially in youn­ger patients. and TP53 genes as inde­pen­dent pre­dic­tors of dis­mal out­come.35

422  |  Hematology 2021  |  ASH Education Program


Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/418/1851957/418voso.pdf by guest on 13 December 2021
Figure 3. Time course of the identification of genes involved in myeloid neoplasms with germline predisposition. Time line de­
picting the discovery of the most important genes involved in the development of myeloid disorders with germline predisposition.
Different colors represent the associated peculiar features (eg, platelet disorders or organ dysfunction). For gene groups (eg, Fanconi
anemia), the year of discovery of the first gene associated with the disease is indicated.

More impor­tantly, the role of these somatic events was inde­pen­ go­ing allo­ge­neic HSCT, patients older than 40 years har­bor­ing
dent of the IPSS-R risk score, empha­siz­ing once more the need to muta­tions in TP53 and RAS path­way genes had sig­nif­i­cantly
inte­grate clin­i­cal and molec­u­lar fac­tors. Indeed, the com­bi­na­tion shorter sur­vival and a higher risk of relapse than their wild-type
of IPSS-R plus muta­tional char­ac­ter­is­tics changed the pre­dic­tion coun­ter­parts.38 While RAS path­way muta­tions were unfa­vor­able
of posttransplant out­comes for 34% of cases. Further, in another only for patients receiv­ing reduced-inten­sity con­di­tion­ing, TP53
study new somatic events were found to be acquired at relapse, muta­tions were asso­ci­ated with a dis­mal prog­no­sis irrespec­
with the pres­ence of dis­ease-related muta­tions at 30 days from tive of the inten­sity of the con­di­tion­ing reg­i­men. Likewise, TP53
HSCT asso­ci­ated with a high risk of pro­gres­sion at a median of muta­tions had an impact on the out­come of HSCT when asso­
67 days from muta­tion detec­tion.36 ci­ated with com­plex kar­yo­type, while RAS path­way muta­tions
In this line, Heuser et al iden­ti­fied muta­tions of NRAS, U2AF1, were prognostically unfa­vor­able in the con­text of MDS/mye­lo­
IDH2, and TP53 and/or the pres­ence of a com­plex kar­yo­type as pro­lif­er­a­tive neo­plasms in another study.39
adverse mark­ers of sur­vival in MDS patients under­go­ing HSCT.37
Although dif­fer­ent stud­ies found diverse pat­terns of gene muta­ Germline muta­tions
tions influ­enc­ing out­comes, TP53 muta­tions remain among the The sit­u­a­tion becomes even haz­ier when con­sid­er­ing genes
major deter­mi­nants of poor prog­no­sis post HSCT, being con­ mutated in mye­loid neo­plasms with germline pre­dis­po­si­tion,
sis­tently asso­ci­ated across stud­ies with reduced sur­vival and a which have been increas­ingly iden­ti­fied in the last few years
high risk of relapse. In a large cohort of 1514 MDS patients under­ (Figure 3). The pre­cise rec­og­ni­tion of such var­i­ants is impor­tant
Prakash Singh Shekhawat
Toward a tai­lored molec­u­lar approach in MDS | 423
Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/418/1851957/418voso.pdf by guest on 13 December 2021
Figure 4.  Exemplificative list of genes recurrently mutated in MDS, with impact on clinical features and on treatment options.
Shown are exemplary gene mutations, their prognostic impact, the associated recurrent clinical features, and possible therapeutic
interventions.

for a cor­rect patient man­age­ment (espe­cially for genes with Implications of molec­u­lar data for treat­ment strat­egy
other organ involve­ment, such as Fanconi ane­mia cases and Several emerg­ing targeted ther­a­pies are cur­rently under eval­
oth­ers), for genetic coun­sel­ing, and ulti­mately for donor selec­ u­ a­
tion for patients with MDS and recur­ rent gene muta­ tions
tion in patients eli­gi­ble for HSCT (unre­lated vs related donors).40 (Figure 4). For instance, vita­min C is ­able to restore the met­a­
In these cases the var­ia ­ nts must be con­firmed on a nonhemato­ bolic impair­ment imprinted by TET2 muta­tions (NCT03682029,
poietic tis­sue (eg, fibro­blasts, hair fol­li­cles, or nails), and genet­ NCT03999723) while prom­is­ing results have been obtained in
ic coun­sel­ing must be required. Generally, these patients pres­ vitro with splic­ing inhib­i­tors (eg, H3B-8800),43 but unfor­tu­nately,
ent with youn­ger age, a pos­it­ive fam­ily his­tory, and in some they have not been con­firmed by the phase 1, first-in-human clin­
instances with extrahematologic fea­tures (such as in Emberger i­cal study (NCT02841540).
syn­drome, or MonoMAC in case of GATA2 defects).41 However, APR-246 is a small mol­e­cule ­able to reestablish p53 func­tions
a low pen­e­trance may result in an absent fam­ily his­tory, and by restor­ing its con­for­ma­tion and has shown encour­ag­ing clin­
late age at MDS diag­no­sis (eg, in DDX41-pos­i­tive cases) may i­cal results in vivo in com­bi­na­tion with HMA.44 A recent phase
chal­lenge the rec­og­ni­tion.42 As dem­on­strated by a sem­i­nal 1b/2 study of its com­bi­na­tion with azacitidine (AZA) in patients
study, these issues play a sig­nif­i­cant role in the HSCT con­text, with TP53-mutant MDS or AML with 20% to 30% BM blasts
where donor selection becomes of utmost impor­tance. Indeed, showed a 73% over­all response rate (ORR), with 50% of patients
the top mutated genes in youn­ger patients (<40 years) includ­ achiev­ing CR and 58%, a cyto­ge­netic response.45 The effi­cacy of
ed GATA2, PIGA, and SBDS, which are recur­rently mutated in this com­bi­na­tion was also underlined by the reduc­tion of muta­
mye­loid neo­plasms with germline pre­dis­po­si­tion, while muta­ tional bur­den and of p53 expres­sion by immu­no­his­to­chem­is­try.45
tions in TET2, DNMT3A, SRSF2, and SF3B1 were sig­nif­i­cantly less However, the phase 3 of this trial (NCT03745716) missed the pri­
com­mon.38 mary end point, as the dif­fer­ence in CR between the 2 arms did

424  |  Hematology 2021  |  ASH Education Program


Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/418/1851957/418voso.pdf by guest on 13 December 2021
Figure 5. Clinicobiological characterization of MDS and tailored treatment. MDS patients represent a heterogeneous multitude
characterized by different clinical, karyotypic, morphologic, and molecular features. In particular, the lower panel demonstrates how
the incorporation of molecular information into currently available prognostication schemes will enable in the near future better
prognostication and tailored treatments.

not meet the predefined thresh­old for sta­tis­ti­cal sig­nif­i­cance col­leagues recently showed that ivosidenib in com­bi­na­tion with
(33.3% vs 22.4% in the exper­i­men­tal vs AZA-monotherapy group, AZA induced responses in 78% of newly diag­nosed patients with
respec­tively; P = .13).46 These data high­light the time­less need to IDH1-mutant AML inel­i­gi­ble for inten­sive ther­apy and IDH1 muta­
con­firm phase 1 and 2 results with larger phase 3 stud­ies even in tion clear­ance in 71% of those achiev­ing CR.50 Another study
patients with molec­u­larly defined dis­eases, which dis­play wide com­bin­ing ivosidenib plus venetoclax with or with­out AZA in the
clin­i­cal het­ero­ge­ne­ity. same patient group (NCT03471260) is cur­rently ongo­ing, and the
Beyond spe­cific TP53 acti­va­tors, magrolimab, a first-in-class pre­lim­i­nary results have dem­on­strated the tol­er­a­bil­ity and effec­
anti-CD47 anti­body, has shown par­tic­u­lar activ­ity in TP53-mu­ tive­ness of this com­bi­na­tion (ORR, 89%).53
tated MDS/AML, as revealed by the results of a phase 1b trial in
com­bi­na­tion with AZA in this set­ting.47 These 2 drugs act syn­er­ Conclusions
gis­ti­cally, induc­ing “eat me” sig­nals on leu­ke­mic stem cells and MDS is a puz­zling dis­or­der with a bio­log­i­cal intri­cacy reflected
restor­ing mac­ro­phage-medi­ated phago­cy­to­sis. by the dif­fi­cul­ties and lim­i­ta­tions of existing clas­si­fi­ca­tions and
The recent results of the IDH1/2 inhib­ i­
tors ivosidenib and prog­nos­ti­ca­tion sys­tems. The abun­dance of molec­u­lar infor­ma­
enasidenib in AML have also opened new sce­nar­ios for patients tion opens new sce­nar­ios in the clin­i­cal set­ting, indeed inau­gu­
with MDS.48-50 In a phase 2 study, enasidenib in com­bi­na­tion with rat­ing a new era in MDS and delin­eat­ing a more pre­cise, objec­
AZA showed an ORR of 67% in newly diag­nosed IDH2-mutant tive, and per­son­al­ized path (Figure 5).
high-risk-MDS patients, and a 50% ORR was obtained when enas­ The bioinformatic inter­pre­ta­tion of the clin­i­cal role of somatic
idenib is used as a sin­gle agent in HMA-treated patients. The muta­ tions is still prob­lem­ atic. Nevertheless, machine-learn­ ing
pre­lim­i­nary anal­y­sis of another study (NCT02074839) cur­rently approaches may in the near future rep­re­sent a valu­able tool to
eval­u­at­ing ivosidenib in IDH1-mutant cases dem­on­strated a 42% solve the short­com­ings of cur­rent diag­nos­tic schemes, pos­si­
ORR in relapsed/refrac­tory MDS.51,52 Furthermore, DiNardo and bly unveiling fur­ther prog­nos­tic impli­ca­tions. By com­bin­ing

Prakash Singh Shekhawat


Toward a tai­lored molec­u­lar approach in MDS | 425
mor­pho­logic and molec­u­lar data, these approaches will enable 12. Papaemmanuil E, Gerstung M, Malcovati L, et al; Chronic Myeloid Disorders
the iden­ti­fi­ca­tion of non­ran­dom geno­typic/mor­pho­logic rela­tion­ Working Group of the International Cancer Genome Consortium. Clinical
and bio­log­i­cal impli­ca­tions of driver muta­tions in myelodysplastic syn­
ships, bet­ter defin­ing clin­i­cally rel­e­vant phe­no­types.54 However, dromes. Blood. 2013;122(22):3616-3627, quiz 3699.
a lim­i­ta­tion of these stud­ies is dem­on­strated by the sta­tis­ti­cal 13. Falconi G, Fabiani E, Piciocchi A, et  al. Somatic muta­tions as mark­ers of
power of the sam­ple size, which does not account for the clin­i­cal out­come after azacitidine and allo­ge­neic stem cell trans­plan­ta­tion in high­
and prog­nos­tic weight of less fre­quently mutated genes. Larger er-risk myelodysplastic syn­dromes. Leukemia. 2019;33(3):785-790.
14. Fenaux P, Haase D, Santini V, et al; Euro­pean Society for Medical Oncol­
cohorts of patients will help unravel the com­plex­ity of MDS biol­
ogy Guidelines Committee. Myelodysplastic syn­ dromes: ESMO Clinical
ogy with the intent of gen­er­at­ing molec­u­larly ori­ented clas­si­fi­ Practice Guidelines for diag­no­sis, treat­ment and fol­low-up†☆. Ann Oncol.
ca­tion and prog­nos­ti­ca­tion sys­tems with rel­e­vant clin­i­cal util­ity. 2021;32(2):142-156.
15. Bersanelli M, Travaglino E, Meggendorfer M, et al. Classification and per­
Acknowledgments son­al­ized prog­nos­tic assess­ment on the basis of clin­i­cal and geno­mic fea­
We thank the Amer­ i­
can Ital­
ian Cancer Foundation Fellowship tures in myelodysplastic syn­dromes. J Clin Oncol. 2021;39(11):1223-1233.
16. Bernard E, Nannya Y, Hasserjian RP, et al. Implications of TP53 alle­lic state
Program (to C. G.), AIRC 5 × 1000 call “Metastatic dis­ease: the key for genome sta­bil­ity, clin­i­cal pre­sen­ta­tion and out­comes in myelodysplas­
unmet need in oncol­ogy to MYNERVA” pro­ject, #21267 (MYeloid tic syn­dromes. Nat Med. 2020;26(10):1549-1556.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/418/1851957/418voso.pdf by guest on 13 December 2021


NEoplasms Research Venture AIRC), and MIUR grant N. 2017WX­ 17. Fenaux P, Platzbecker U, Mufti GJ, et al. Luspatercept in patients with low­
R7ZT (to M. T. V.). er-risk myelodysplastic syn­dromes. N Engl J Med. 2020;382(2):140-151.
18. Platzbecker U, Fenaux P, Adès L, et  al. Proposals for revised IWG 2018
We would also like to acknowl­edge Stefan Hohaus for help­ful
hema­to­log­i­cal response cri­te­ria in patients with MDS included in clin­i­cal
com­ments on the man­u­script. tri­als. Blood. 2019;133(10):1020-1030.
19. Papaemmanuil E, Cazzola M, Boultwood J, et al; Chronic Myeloid Disorders
Conflict-of-inter­est dis­clo­sure Working Group of the International Cancer Genome Consortium. Somatic
Maria Teresa Voso: no com­pet­ing finan­cial inter­ests to declare. SF3B1 muta­tion in myelodysplasia with ring sideroblasts. N Engl J Med.
Carmelo Gurnari: no com­pet­ing finan­cial inter­ests to declare. 2011;365(15):1384-1395.
20. Yoshida K, Sanada M, Shiraishi Y, et al. Frequent path­way muta­tions of splic­ing
machin­ery in myelodysplasia. Nature. 2011;478(7367):64-69.
Off-label drug use 21. Palomo L, Solé F. SF3B1: the lord of the rings in MDS. Blood. 2020;136(2):149-
Maria Teresa Voso: nothing to disclose. 151.
Carmelo Gurnari: nothing to disclose. 22. Malcovati L, Stevenson K, Papaemmanuil E, et al. SF3B1-mutant MDS as a
dis­tinct dis­ease sub­type: a pro­posal from the International Working Group
Correspondence for the Prognosis of MDS. Blood. 2020;136(2):157-170.
Maria Teresa Voso, Department of Biomedicine and Prevention, 23. Ebert BL, Pretz J, Bosco J, et al. Identification of RPS14 as a 5q− syn­drome
gene by RNA inter­fer­ence screen. Nature. 2008;451(7176):335-339.
University of Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; e- 24. Schneider RK, Ademà V, Heckl D, et  al. Role of casein kinase 1A1 in the
mail: voso@med​­.uniroma2​­.it. biol­ogy and targeted ther­apy of del(5q) MDS. Cancer Cell. 2014;26(4):509-
520.
References 25. Shiozawa Y, Malcovati L, Gallì A, et al. Aberrant splic­ing and defec­tive
1. Arber DA, Orazi A, Hasserjian R, et al. The 2016 revi­sion to the World Health mRNA pro­duc­tion induced by somatic spliceosome muta­tions in myel­
Organization clas­si­fi­ca­tion of mye­loid neo­plasms and acute leu­ke­mia. odysplasia. Nat Commun. 2018;9(1):3649.
Blood. 2016;127(20):2391-2405. 26. Nikpour M, Scharenberg C, Liu A, et al. The trans­porter ABCB7 is a medi­a­
2. Greenberg P, Cox C, LeBeau MM, et al. International scor­ing sys­tem for eval­ tor of the phe­no­type of acquired refrac­tory ane­mia with ring sideroblasts.
u­at­ing prog­no­sis in myelodysplastic syn­dromes. Blood. 1997;89(6):2079- Leukemia. 2013;27(4):889-896.
2088. 27. Platzbecker U, Germing U, Götze KS, et al. Luspatercept for the treat­ment
3. Greenberg PL, Tuechler H, Schanz J, et al. Revised inter­na­tional prog­nos­tic of anae­mia in patients with lower-risk myelodysplastic syn­dromes (PACE-
scor­ing sys­tem for myelodysplastic syn­dromes. Blood. 2012;120(12):2454- MDS): a multicentre, open-label phase 2 dose-find­ing study with long-term
2465. exten­sion study. Lancet Oncol. 2017;18(10):1338-1347.
4. Pfeilstöcker M, Tuechler H, Sanz G, et al. Time-depen­dent changes in mor­ 28. Steensma DP. How I use molec­u­lar genetic tests to eval­u­ate patients who
tal­ity and trans­for­ma­tion risk in MDS. Blood. 2016;128(7):902-910. have or may have myelodysplastic syn­dromes. Blood. 2018;132(16):1657-
5. Platzbecker U. Treatment of MDS. Blood. 2019;133(10):1096-1107. 1663.
6. Voso MT, Fenu S, Latagliata R, et  al. Revised International Prognostic 29. Koskela HL, Eldfors S, Ellonen P, et  al. Somatic STAT3 muta­tions in large
Scoring System (IPSS) pre­dicts sur­vival and leu­ke­mic evo­lu­tion of myelo­ gran­u­lar lym­pho­cytic leu­ke­mia. N Engl J Med. 2012;366(20):1905-1913.
dysplastic syn­dromes sig­nif­i­cantly bet­ter than IPSS and WHO Prognostic 30. Jaiswal S, Fontanillas P, Flannick J, et al. Age-related clonal hema­to­poi­e­sis
Scoring System: val­i­da­tion by the Gruppo Romano Mielodisplasie Ital­ian asso­ci­ated with adverse out­comes. N Engl J Med. 2014;371(26):2488-2498.
Regional Database. J Clin Oncol. 2013;31(21):2671-2677. 31. Genovese G, Kähler AK, Handsaker RE, et  al. Clonal hema­to­poi­e­sis and
7. Kuendgen A, Nomdedeu M, Tuechler H, et  al. Therapy-related myelo­ blood-can­ cer risk inferred from blood DNA sequence. N Engl J Med.
dysplastic syn­dromes deserve spe­cific diag­nos­tic sub-clas­si­fi­ca­tion and 2014;371(26):2477-2487.
risk-strat­i­fi­ca­tion—an approach to clas­si­fi­ca­tion of patients with t-MDS. 32. van Zeventer IA, Salzbrunn JB, de Graaf AO, et al. Prevalence, pre­dic­tors,
Leukemia. 2021;35(3):835-849. and out­ comes of clonal hema­ to­poi­e­sis in indi­
vid­
ua­ls aged ≥80 years.
8. Haferlach T, Nagata Y, Grossmann V, et al. Landscape of genetic lesions in Blood Adv. 2021;5(8):2115-2122.
944 patients with myelodysplastic syn­dromes. Leukemia. 2014;28(2):241- 33. da Silva-Coelho P, Kroeze LI, Yoshida K, et al. Clonal evo­lu­tion in myelodys­
247. plastic syn­dromes. Nat Commun. 2017;8(21 April):15099.
9. Nazha A, Al-Issa K, Hamilton BK, et al. Adding molec­u­lar data to prog­nos­tic 34. Figueroa ME, Skrabanek L, Li Y, et  al. MDS and sec­ond­ary AML dis­play
mod­els can improve pre­dic­tive power in treated patients withmyelodys­ unique pat­ terns and abun­ dance of aber­ rant DNA meth­ yl­

tion. Blood.
plastic syn­dromes. Leukemia. 2017;31(12):2848-2850. 2009;114(16):3448-3458.
10. Nazha A, Narkhede M, Radivoyevitch T, et  al. Incorporation of molec­ul­ar 35. Della Porta MG, Gallì A, Bacigalupo A, et al. Clinical effects of driver somatic
data into the Revised International Prognostic Scoring System in treated muta­tions on the out­comes of patients with myelodysplastic syn­dromes
patients with myelodysplastic syn­ dromes. Leukemia. 2016;30(11):2214- treated with allo­ge­neic hema­to­poi­etic stem-cell trans­plan­ta­tion. J Clin
2220. Oncol. 2016;34(30):3627-3637.
11. Bejar R, Stevenson K, Abdel-Wahab O, et al. Clinical effect of point muta­ 36. Duncavage EJ, Jacoby MA, Chang GS, et al. Mutation clear­ance after trans­
tions in myelodysplastic syn­ dromes. N Engl J Med. 2011;364(26):2496- plan­ta­tion for myelodysplastic syn­drome. N Engl J Med. 2018;379(11):1028-
2506. 1041.

426  |  Hematology 2021  |  ASH Education Program


37. Heuser M, Gabdoulline R, Löffeld P, et al. Individual out­come pre­dic­tion for 49. Stein EM, DiNardo CD, Pollyea DA, et al. Enasidenib in mutant IDH2 relapsed
myelodysplastic syn­drome (MDS) and sec­ond­ary acute mye­loid leu­ke­mia or refrac­tory acute mye­loid leu­ke­mia. Blood. 2017;130(6):722-731.
from MDS after allo­ge­neic hema­to­poi­etic cell trans­plan­ta­tion. Ann Hematol. 50. DiNardo CD, Stein AS, Stein EM, et al. Mutant isocitrate dehy­dro­ge­nase 1
2017;96(8):1361-1372. inhib­i­tor ivosidenib in com­bi­na­tion with azacitidine for newly diag­nosed
38. Lindsley RC, Saber W, Mar BG, et al. Prognostic muta­tions in myelodysplas­ acute mye­loid leu­ke­mia. J Clin Oncol. 2021;39(1):57-65.
tic syn­drome after stem-cell trans­plan­ta­tion. N Engl J Med. 2017;376(6):536- 51. Richard-Carpentier G, DeZern AE, Takahashi K, et  al. Preliminary results
547. from the phase II study of the IDH2-inhib­ i­
tor enasidenib in patients
39. Yoshizato T, Nannya Y, Atsuta Y, et al. Genetic abnor­mal­i­ties in myelodys­ with high-risk IDH2-mutated myelodysplastic syn­ dromes (MDS). Blood.
plasia and sec­ond­ary acute mye­loid leu­ke­mia: impact on out­come of stem 2019;134(suppl 1):678-678.
cell trans­plan­ta­tion. Blood. 2017;129(17):2347-2358. 52. Foran JM, DiNardo CD, Watts JM, et al. Ivosidenib (AG-120) in patients with
40. University of Chicago Hematopoietic Malignancies Cancer Risk Team. How IDH1-mutant relapsed/refrac­ tory myelodysplastic syn­ drome: updated
I diag­nose and man­age indi­vid­u­als at risk for inherited mye­loid malig­nan­ enroll­ ment of a phase 1 dose esca­ la­
tion and expan­ sion study. Blood.
cies. Blood. 2016;128(14):1800-1813. 2019;134(suppl 1):4254.
41. Wlodarski MW, Hirabayashi S, Pastor V, et al; Euro­pean Working Groups of 53. Lachowiez CA, Borthakur G, Loghavi S, et  al. Phase Ib/II study of the
Myelodysplastic Syndromes. Prevalence, clin­i­cal char­ac­ter­is­tics, and prog­ IDH1-mutant inhib­i­tor ivosidenib with the BCL2 inhib­i­tor venetoclax +/-
no­sis of GATA2-related myelodysplastic syn­dromes in chil­dren and ado­les­ azacitidine in IDH1-mutated hema­to­logic malig­nan­cies. J Clin Oncol.
cents. Blood. 2016;127(11):1387-1397, quiz 1518. 2020;38(suppl 15):7500.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/418/1851957/418voso.pdf by guest on 13 December 2021


42. Polprasert C, Schulze I, Sekeres MA, et al. Inherited and somatic defects in 54. Nagata Y, Zhao R, Awada H, et  al. Machine learn­ing dem­on­strates that
DDX41 in mye­loid neo­plasms. Cancer Cell. 2015;27(5):658-670. somatic muta­tions imprint invari­ant mor­pho­logic fea­tures in myelodys­
43. Cimmino L, Dolgalev I, Wang Y, et al. Restoration of TET2 func­tion blocks plastic syn­dromes. Blood. 2020;136(20):2249-2262.
aber­rant self-renewal and leu­ke­mia pro­gres­sion. Cell. 2017;170(6):1079- 55. Kantarjian H, O’Brien S, Ravandi F, et al. Proposal for a new risk model in
1095.e201095e20. myelodysplastic syn­drome that accounts for events not con­sid­ered in the
44. Maslah N, Salomao N, Drevon L, et  al. Synergistic effects of PRIMA-1Met orig­i­nal International Prognostic Scoring System. Cancer. 2008;113(6):1351-
(APR-246) and 5-azacitidine in TP53-mutated myelodysplastic syn­dromes 1361.
and acute mye­loid leu­ke­mia. Haematologica. 2020;105(6):1539-1551. 56. Malcovati L, Germing U, Kuendgen A, et  al. Time-depen­dent prog­nos­tic
45. Sallman DA, DeZern AE, Garcia-Manero G, et al. Eprenetapopt (APR-246) scor­ing sys­tem for predicting sur­vival and leu­ke­mic evo­lu­tion in myelodys­
and azacitidine in TP53-mutant myelodysplastic syn­dromes. J Clin Oncol. plastic syn­dromes. J Clin Oncol. 2007;25(23):3503-3510.
2021;39(14):1584-1594. 57. Mufti GJ, Bennett JM, Goasguen J, et al; International Working Group on
46. Aprea Therapeutics. Aprea Therapeutics announces results of pri­ mary Morphology of Myelodysplastic Syndrome. Diagnosis and clas­si­fi­ca­tion of
end­point from phase 3 trial of eprenetapopt in TP53 mutant myelodys­ myelodysplastic syn­drome: International Working Group on Morphology
plastic syn­dromes (MDS). Accessed 6 Sep­tem­ber 2021. https:​­/​­/ir​­.aprea​ of myelodysplastic syn­drome (IWGM-MDS) con­sen­sus pro­pos­als for the
­.com​­/news​­-releases​­/news​­-release​­-details​­/aprea​­-therapeutics​­-announces​ def­i­ni­tion and enu­mer­a­tion of mye­lo­blasts and ring sideroblasts. Haema-
­-results​­-primary​­-endpoint​­-phase​­-3. tologica. 2008;93(11):1712-1717.
47. Sallman DA, Malki MA, Asch AS, et al. Tolerability and effi­cacy of the first-in-
class anti-CD47 anti­body magrolimab com­bined with azacitidine in MDS and
AML patients: phase Ib results. J Clin Oncol. 2020;38(suppl 15):7507-7507.
48. DiNardo CD, Stein EM, de Botton S, et  al. Durable remis­sions with ivos­
idenib in IDH1-mutated relapsed or refrac­tory AML. N Engl J Med. 2018;​ © 2021 by The Amer­i­can Society of Hematology
378(25):​2386-2398. DOI 10.1182/hema­tol­ogy.2021000276

Prakash Singh Shekhawat


Toward a tai­lored molec­u­lar approach in MDS | 427
MDS: BEYOND A ONE - SIZE - FITS -ALL APPROACH

Lower risk but high risk


Amy E. DeZern

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/428/1851953/428dezern.pdf by guest on 13 December 2021


Division of Hematologic Malignancies, Sidney Kimmel Cancer Center at Johns Hopkins, Baltimore, MD

Risk stratification is crucial to the appropriate management of most cancers, but in patients with myelodysplastic syn-
dromes (MDS), for whom expected survival can vary from a few months to more than a decade, accurate disease prog-
nostication is especially important. Currently, patients with MDS are often grouped into higher-risk (HR) vs lower-risk (LR)
disease using clinical prognostic scoring systems, but these systems have limitations. Factors such as molecular genetic
information or disease characteristics not captured in the International Prognostic Scoring System–Revised (IPSS-R) can
alter risk stratification and identify a subset of patients with LR-MDS who actually behave more like those with HR-MDS.
This review describes the current identification and management of patients with LR-MDS whose condition is likely to
behave in a less favorable manner than predicted by the IPSS-R.

LEARNING OBJECTIVES
• Describe limitations of currently available prognostic scoring systems for patients with MDS
• Discuss the current status of biologic upstaging, specifcally with respect to the lower­risk patient who may require
treatments usually administered to higher­risk patients
• Highlight the heterogeneity in WHO­defned treatment­related MDS
• Review which lower­risk patients by the IPSS­R may beneft from early consideration of allogeneic transplant

Introduction of 0.82×109/L. Bone marrow biopsy specimen exhibits hyper­


In myelodysplastic syndromes (MDS), an optimized under­ cellularity, trilineage dysplasia and 2% blasts, karyotype with
standing of a patient’s risk (even outside of the prognosti­ 8 of 20 metaphases with chromosome 21 loss, and next­
cation systems) is critical as therapeutic strategies can vary generation sequencing (NGS) revealing a RUNX1 mutation
from observation of mild cytopenias or supportive treat­ with variant allele frequency (VAF) of 26% and a monoallelic
ments with transfusions through active chemotherapy and TP53 mutation with a VAF of 11%. IPSS­R score is low risk at
even early allogeneic hematopoietic stem transplantation 3 with 2 points for intermediate cytogenetics and 1 point for
(BMT). Here I consider lower­risk (LR) disease to encompass Hgb. She received a transfusion for anemia at diagnosis.
very low­, low­, and intermediate­risk categories assigned
by the International Prognostic Scoring System–Revised
(IPSS­R).1 The intermediate­risk group in particular is hetero­ CLINICAL CASE 2
geneous with a substantial proportion behaving like higher
A 69­year­old woman also was treated for breast can­
risk (HR) with early disease progression and risk of death.
cer with radiation therapy at 50 Gy. She had new ane­
Treatment­related MDS (tMDS) is another prognostic chal­
mia (Hgb, 9.9 g/dL; PLTs, 359 × 109/L; and ANC, 1.6 × 109/L).
lenge in the LR arena.3
A bone marrow biopsy specimen was 30% cellular with
dysgranulopoiesis and erythroid dysplasia. Blasts were
1%. Karyotype had 17 of 20 cell lines with trisomy 8 and a
DNMT3A mutation with a VAF of 41%. IPSS­R score was low
CLINICAL CASE 1 risk at 2, with 2 points for intermediate cytogenetics only.
A 68­year­old woman was treated 15 years ago for breast
adenocarcinoma including adjuvant chemotherapy. Labora­
tory values now reveal a platelet (PLT) count of 102×109/L, Current prognostic strategies are imperfect
hemoglobin (Hgb) of 8.2g/dL, and white blood cell (WBC) The use of prognostic scoring systems is common in many
count of 2.65×109/L with an absolute neutrophil count (ANC) malignant diseases and important for predicting the sur­

428 | Hematology 2021 | ASH Education Program


Table 1. Currently avail­­able prog­nos­tic scor­ing sys­tems in MDS

Blood Marrow Patient


System Comments
Hgb PLTs ANC Blasts Cyto Age Txn PS
IPSS + + + + + + Has been revised now (2012) using the same
prog­nos­tic param­e­ters (num­ber and depth or
cytopenias, mar­row blast per­cent­age [more gran­
u­lar] and kar­yo­type), was a
­ ble to reclassify nearly
25% of lower-risk MDS patients into a higher-risk
cat­e­gory
WPSS + + + Time depen­dent which is use­ful over the course of
patient’s dis­ease
MDS LR + + + + + + May be applied spe­cif­i­cally to lower-risk dis­ease

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/428/1851953/428dezern.pdf by guest on 13 December 2021


with more var­i­ables
MDAPSS + + + + + + + Contains more clin­i­cal var­i­ables, espe­cially age and
trans­fu­sion bur­den, which could upstage a patient
FPSS + + + Higher-risk dis­ease and includes azacytidine-
treated patients, likely is not use­ful in lower-risk
patients, even for upstaging
IPSS-R + + + + + + Used most com­monly at diag­no­sis and for clin­i­cal
trial eli­gi­bil­ity, lim­i­ta­tions discussed in text
The prog­nos­tic scor­ing sys­tems are listed in order of pub­li­ca­tion with anno­ta­tions on the var­i­ables included and com­ments on their use in lower
risk dis­ease.
Cyto, cyto­ge­net­ics; FPSS, French Prognostic Scoring System; MDAPSS, MD Anderson Prognostic Scoring System; MDS LR, scor­ing sys­tem for
patients with lower-risk MDS; PS, per­for­mance sta­tus; Txn, trans­fu­sions; WPSS, WHO Classification-Based Prognostic Scoring System.

vival of indi­vid­u­als, but these sys­tems are often based on ret­ro­ incor­po­rated into IPSS-R, would have altered the risk cat­e­gory.5
spec­tive patient data with het­er­og­e­nous treat­ments and often Although I do know that a chro­mo­somal alter­ation iden­ti­fied
incor­po­rate only a few salient clin­i­cal or bio­log­i­cal fea­tures that only by WGS has the same prog­nos­tic mean­ing as that iden­ti­fied
were sta­tis­ti­cally sig­nif­i­cant in a given model. Although the IPSS by meta­phase karyotyping, I believe this sug­gests fur­ther need
and its IPSS-R1 (among oth­ers) are use­ful tools for clin­i­cal deci­ for effi­cient incor­po­ra­tion of addi­tional cyto­ge­netic and molec­u­
sion mak­ing (Table 1), these scor­ing sys­tems have draw­backs lar data into prog­nos­tic scor­ing sys­tems.
and may fail to cap­ture impor­tant prog­nos­tic infor­ma­tion at the
indi­vid­ual level.
The key lim­i­ta­tion to the prog­nos­ti­ca­tion sys­tems is their
inabil­ity to cap­ture all­ rel­e­vant biol­ogy. For exam­ple, cyto­ge­net­ics
is the only bio­log­i­cal param­e­ter included in the IPSS-R, and unfor­ CLINICAL CASES 2 (continued)
tu­nately, although an IPSS revi­sion incor­po­rat­ing NGS molec­u­lar Patient 1 went on to receive 14 cycles of azacytidine. Transplant
data is in devel­op­ment, this has yet to be reported. At the 2015 options were pur­ sued but no avail­­ able donor located. She
meeting of the American Society of Hematology, Bejar et al2 from remained red blood cell trans­fu­sion depen­dent (RBC-TD) with
the International Working Group for Prognosis in MDS–Molecular Hgb rou­tinely below 8 g. Repeat mar­row assess­ment con­tin­ued
Committee reviewed ana­ly­ses that showed somatic muta­tions to show LR fea­tures; ther­apy was not altered. She ulti­mately
in MDS are asso­ci­ated with clin­i­cal fea­tures and pre­dict prog­no­ progressed to AML 19 months after pre­sen­ta­tion. Patient 2 has
sis inde­pen­dent of the IPSS-R. Bersanelli et al4 recently showed been followed with every 3-monthly blood counts and bien­nial
that inte­gra­tion of clin­i­cal data with NGS pro­fil­ing improves the mar­row assess­ments for 6 years with near-com­plete sta­bil­ity.
accu­racy of cur­rently avail­­able prog­nos­tic scores. The authors Patients 1 and 2 high­light fur­ther sig­nif­i­cant clin­i­cal lim­i­ta­
pro­vided evi­dence from another large, inter­na­tional data­base tion in the IPSS-R: how to cat­e­go­rize tMDS. About 15% of MDS
that MDS could be clas­si­fied into 8 dis­tinct sub­types according are tMDS and gen­er­ally con­sid­ered at HR. These dis­or­ders are
to spe­cific geno­mic fea­tures.4 These sub­groups do not cor­re­ of clin­i­
cal impor­tance for patients and of grow­ ing aca­ demic
late with mor­pho­logic categories defined by the cur­rent World inter­est, espe­cially as patients live lon­ger due to more effec­
Health Organization (WHO) clas­si­fi­ca­tion and displayed sig­nif­i­ tive ther­a­pies. With closer fol­low-up, this cat­e­gory is likely to
cantly dif­fer­ent clin­i­cal phe­no­types and out­come.4 be the larg­est increase in patients with “higher-risk” low-risk
The IPSS-R was based only on stan­dard G-banded meta­phase MDS. The IPSS-R cohort did not con­tain any patients with tMDS,
karyotyping. A recent study exam­ined whole-genome sequenc­ and the prog­no­sis of these patients is com­pli­cated by sev­eral
ing (WGS) of patients with MDS or acute mye­loid leu­ke­mia (AML) non-MDS fac­tors, includ­ing prior toxicities of ther­apy (trans­fu­
for fea­si­bil­ity assess­ment.5 Among 42 patients with MDS in this sions, alloantibodies, organ lim­i­ta­tions), as well as other med­i­
study, the con­ven­tional results (trans­lo­ca­tions and copy num­ber cal comorbidities. There is the com­mon per­cep­tion that patients
var­i­a­tion) were con­firmed in all­cases by WGS; nearly 25% had with tMDS have high-risk dis­ease, regard­less of IPSS-R score, and
addi­tional chro­mo­somal abnor­mal­i­ties iden­ti­fied by WGS that, if there­fore war­rant aggres­sive ther­apy. Patient 2’s course would
Prakash Singh Shekhawat
Lower risk but high risk | 429
Table 2. Features in lower-risk MDS that sug­gest higher-risk behav­ior

MDS char­ac­ter­is­tic Feature asso­ci­ated with lesser prog­no­sis


Etiology of MDS Treatment related, can behave in a het­er­og­e­nous fash­ion
Fibrosis in core biopsy Grade 2 or higher
Cytopenia Symptomatic neutropenia
Decrease in PLTs >25%
Ongoing RBC trans­fu­sion depen­dence
Anemia or throm­bo­cy­to­pe­nia refrac­tory to trans­fu­sions
Karyotype Clonal emer­gence of unfa­vor­able kar­yo­type
Somatic muta­tions Multiple muta­tions (≥3 somatic muta­tions)

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/428/1851953/428dezern.pdf by guest on 13 December 2021


T53, RUNX1, ASXL1 muta­tions
Absence of SF3B1 muta­tion (espe­cially in MDS with ring sideroblasts)
Multiple somatic muta­tions
Inherited pre­dis­po­si­tion Patients with known germline var­i­ant in their dis­ease may be less likely to
respond to tra­di­tional ther­a­pies and require stem cell ther­apy sooner
Treatment response Primary treat­ment fail­ure vs sec­ond­ary fail­ure

argue against this. According to the cur­rent def­i­ni­tion of the WBC count was 2.4 × 109/L, ANC was 1.36 × 109/L, Hgb was
WHO, tMDS is defined by the his­tory of receipt of che­mo­ther­apy 7.9 g/dL, and PLTs were 99 × 109/L. Marrow biopsy spec­i­men was
and/or radio­ther­apy for nonmyeloid malig­nancy or med­i­cal con­ 95% cel­lu­lar with ery­throid pre­dom­i­nance, trilineage dyspoi­
di­tion.6 This can­not always allow for dis­ease that may not truly esis, and 3% blasts. There were 25% to 30% ring sideroblasts
be attrib­uted to the pre­vi­ous ther­apy (per­haps in patient 2) as with a dif­fuse increase in retic­u­lar fibro­sis. His final diag­no­sis
the dis­ease behav­ior is more akin to de novo MDS. Nonetheless, was MDS with ring sideroblasts and multilineage dys­pla­sia. His
the IPPS-R has been shown to be prognostically inad­eq ­ uate in kar­yo­type was 46,XY, and NGS showed SF3B1 R625H with a VAF
tMDS, as in patient 1.7 The US MDS Clinical research con­sor­tium of 38% with­out comutations to sug­gest mye­lo­fi­bro­sis. IPSS-R
reported shorter sur­vival in tMDS than in de novo MDS, includ­ing score was inter­me­di­ate-risk dis­ease at 3.5 with 1 for cyto­ge­net­
in LR patients.8 Zeidan et al8 ana­lyzed out­comes in 370 patients ics, 1 for 3% blasts, 1 for Hgb less than 8 g, and 0.5 for PLTs less
with tMDS to under­stand the prog­nos­tic util­ity of cur­rent risk than 100 × 109/L.
strat­i­fi­ca­tion tools in tMDS. The over­all sur­vival (OS) of patients Clinical lim­i­ta­tions in use of prog­nos­tic scor­ing sys­tems also
with tMDS was sig­nif­i­cantly worse than those with de novo dis­ exist. Table 2 sum­ma­rizes addi­tional poten­tial poor prog­nos­tic
ease (median OS, 19 vs 46 months). fac­tors in LR patients that fall out­side the IPSS-R. Grade 2 or more
The genet­ ics of tMDS has been inves­ ti­
gated to explain if mar­row fibro­sis may worsen prog­no­sis of patients with MDS.11
their “risk het­ero­ge­ne­ity” (as displayed by both cases) might RBC trans­fu­sion depen­dency is an inde­pen­dent poor prog­nos­
be par­tially related to inad­ver­tent inclu­sion of coin­ci­den­tal sec­ ti­ca­tor. Recently, Hiwase et al12 exam­ined whether RBC-TD adds
ond dis­ease due to genetic pre­dis­po­si­tion.9 A his­tory of ther­ prog­nos­tic value to the IPSS-R. In a mul­ti­var­i­ate anal­y­sis, their
apy might mean there was clonal selec­tion at some ances­tral cohort dem­on­strates that devel­op­ment of RBC-TD at any time
point, but the biol­ogy is het­ero­ge­neous, and some have more dur­ing the course of MDS is asso­ci­ated with poor OS, inde­pen­
indo­lent dis­ease, as in patient 2. Previous can­cer ther­apy with dent of IPSS-R.12 The LeukaemiaNet MDS reg­is­try recently dem­on­
radi­at­ ion, plat­i­num, and topoisomerase II inhib­i­tors, as in patient strated that a rel­at­ ive PLT drop of more than 25% results in a 22%
1, pref­er­en­tially selects for muta­tions in DNA dam­age response OS at 5 years.13 When the PLT drop is com­bined with RBC-TD at
genes (TP53, PPM1D, CHEK2) in a recent large series.10 These lat­ 6 months from diag­no­sis, the OS is a stag­ger­ingly low 9%.13 This
ter patients will likely all­behave in an HR fash­ion, regard­less of is a straight­for­ward and non­in­va­sive way to pre­dict evo­lu­tion
IPSS-R, and should be treated more aggres­sively with seri­ally to HR dis­ease for LR MDS. The addi­tion of the inter­me­di­ate-risk
reassessment and alter­ations to ther­a­peu­tics. The man­age­ment group to the IPSS-R has also been a clin­i­cal chal­lenge as it is
of patients with tMDS remains chal­leng­ing, and care­ful sur­veil­ most often con­sid­ered still LR yet is highly het­ero­ge­neous. Ben­
lance and open­ness to change ther­apy as course dic­tates—to ton et al14 performed an anal­y­sis of 298 inter­me­di­ate-risk patients
lower inten­sity or higher inten­sity—are crit­i­cal. to assess HR fea­tures. Age older than 66 years, periph­eral blood
blasts of 2% or more, and RBC trans­fu­sion were sig­nif­i­cantly
asso­ci­ated with infe­rior sur­vival.14 Patient 3 high­lights sev­eral of
these issues.

CLINICAL CASE 3 Adverse molec­ul­ar fea­tures may “upstage” an LR patient


A pre­vi­
ously healthy 39-year-old man sought treat­ment for Most cases of MDS have 1 or sev­eral somatic gene muta­tions.
fatigue and cytopenias. He had no spleno­ meg­aly. His total SF3B1MUT is gen­er­ally con­sid­ered a favor­able prog­nos­tic marker.15

430  |  Hematology 2021  |  ASH Education Program


Despite this, SF3B1MUT MDS is het­ero­ge­neous in its path­o­logic
fea­tures, treat­ment responses, and clin­i­cal out­comes,16 as in
patient 3. Understanding this het­ero­ge­ne­ity is cru­cial for dis­
ease prog­nos­ti­ca­tion and man­age­ment, and it is rel­e­vant to a
recent pro­posal by the MDS International Working Group that
SF3B1MUT MDS should be clas­si­fied as a dis­tinct noso­logic enti­
ty.15 Although his­tor­i­cally, SF3B1MUT gen­er­ally rep­re­sents indo­lent
dis­ease in LR MDS, comutation with RUNX1 and EZH2 has been
asso­ci­ated with a worse prog­no­sis.15
The inde­ pen­ dent poor prog­ nos­
tic value of many somatic
muta­tions, includ­ing ASXL1, SRSF2, RUNX1, and TP53, has been
established ret­ro­spec­tively.17 The inte­gra­tion of these unfa­vor­
able muta­tions in spe­cific scor­ing sys­tems for LR MDS has been

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/428/1851953/428dezern.pdf by guest on 13 December 2021


reported. In addi­tion, an increased num­ber of muta­tions pres­ent
in a sin­gle patient also sug­gests a less favor­able out­come.18
Of par­tic­u­lar impor­tance is the TP53 muta­tion. Specifically,
this is pres­ent in 20% of cases of LR MDS with iso­lated dele­tion
5q minus syn­drome (del5q; but rare in other LR MDS) and asso­ci­
ated with an HR of AML and poorer sur­vival.19 However, a recent
study ana­ lyzed more than 3000 patients with MDS for TP53
muta­tions and alle­lic imbal­ances.19 One-third of TP53-mutated
patients had monoallelic muta­tions, whereas two-thirds had mul­
ti­ple mutations con­sis­tent with biallelic targeting. Established
asso­ci­a­tions with com­plex kar­yo­type, few co-occur­ring muta­
tions, high-risk pre­sen­ta­tion, and poor out­comes were spe­cific to
multihit patients only. Interestingly, monoallelic patients did not
dif­fer from TP53 wild-type patients in out­comes and response to Figure 1. Therapeutic paradigm in LR MDS. MDS-RS, myelodys­
ther­apy in this series.19 Nonetheless, most still use the pres­ence plastic syndrome with ring sideroblasts.
of TP53 muta­tion (even monoallelic) to upstage LR dis­ease given
its long asso­ci­a­tion with adverse out­comes, as in patient 1.
dif­fer­en­ti­a­tion to inherited pre­dis­po­si­tion beyond poten­tial bio­
logic under­stand­ing of increased risk fea­tures to LR dis­ease.20

Outcomes in LR dis­ease with cur­rently avail­­able ther­a­pies


CLINICAL CASE 3 (Con­t in­u ed) Figure 1 depicts many of the stan­dard treat­ments24-27 often em­
Four months after diag­no­sis, the patient remained well, but blood ployed in LR dis­ease, regard­less of HR fea­tures, as well as fre­quent
counts were lower and treat­ ment warranted for cytopenias, need for reassessment if response to treat­ment is not pos­i­tive. Al­
despite still inter­me­di­ate risk by IPSS-R on repeat mar­row. Con­ though hypomethylating agents (HMAs), such as azacitidine and
cern for his MDS diag­no­sis at age less than 40 years prompted decitabine, and lenalidomide (LEN) are not approved in many
deeper genetic assess­ment. Ultimately, a germline pre­dis­po­si­ countries for LR dis­ease, the focus here is on drugs for con­sid­
tion gene asso­ci­ated with mye­loid malig­nan­cies was iden­ti­fied er­ation in our patients and the avail­­able data in the lit­er­a­ture.
in the patient (and also in his brother and father) with the RTEL1 For throm­bo­cy­to­pe­nic LR patients, eltrombopag is mod­estly
muta­tion. He then under­went unre­lated donor BMT with nor­mal clin­i­cally effec­tive in rais­ing PLT counts and reduc­ing bleed­ing
counts and full donor chi­me­rism with­out com­pli­ca­tions at 1 year. events.28 It is not cur­rently approved for MDS and not yet known
if it will be safe. The assess­ment of long-term safety and effi­cacy
Inherited pre­dis­po­si­tion to MDS may alter of eltrombopag and its effect on sur­vival (phase 2 part of study) is
risk assess­ment still ongo­ing28 and likely will be held to a high stan­dard to ensure
Inherited var­i­ants are rec­og­nized increas­ingly as predisposing no increased clonal evo­lu­tion. Romiplostim has also been stud­ied
patients to MDS, and germline sus­cep­ti­bil­ity to mye­loid malig­nan­ in MDS and reduces PLT trans­fu­sions and bleed­ing events with
cies is included in the lat­est WHO clas­si­fi­ca­tion of hema­to­poi­etic good OS with­out increas­ing rates of AML.29 Clinical tri­als of novel
malig­nan­cies. Moreover, clin­i­cal guide­lines now call for assess­ ther­a­pies in LR dis­ease, such as imetelstat,30 may yet be a ­ ble to
ment of germline pre­dis­po­si­tion at MDS diag­no­sis.20,21 A recent mod­ify dis­ease risk and change the nat­u­ral his­tory.
series showed that the fre­quency of germline var­i­ants in adults Prebet et  al. have looked at out­comes posttreatment with
aged 18 to 40 years diag­nosed with MDS is high (con­sis­tent with LEN in LR MDS with and with­out del5q.31,32 In patients with del5q,
other reports), and the asso­ci­ated germline syn­drome is often not OS fol­low­ing LEN fail­ure was a mere 23 months,31 whereas in
appar­ent from the patient’s med­i­cal or fam­ily his­tory.22 Patient 3, non-del5q, it was 43 months.32 LEN resis­tance in del5q is also
with his youn­ger age, illus­trates this need for deeper genet­ics asso­ci­ated with a high risk of AML and a 5-year prob­a­bil­ity of
assess­ment. These patients may behave in a vari­ety of ways with sur­vival of only 25%.31 For non-del5q, sub­se­quent ther­apy with
HR man­i­fes­ta­tions; coexisting somatic muta­tions are not uncom­ HMAs was asso­ci­ated with a prolonged sur­vival com­pared with
mon in these patients.23 There are also many ben­e­fits of real-time best sup­port­ive care (median OS, 51 vs 36 months, P = .01).32 This
Prakash Singh Shekhawat
Lower risk but high risk | 431
sug­gests an early switch to HMAs in LEN-resis­tant patients is Transplant Research. The authors reported 1514 patients with
rea­son­able. The time to resis­tance or fail­ure of ther­apy varies in MDS who under­went trans­plan­ta­tion, of whom 116 were con­sid­
the lit­er­at­ure, but a trial prob­a­bly should be no lon­ger than 16 ered LR. These patients were con­sid­ered favor­able, with an age
weeks for non-del5q patients as more than 90% of respond­ers at trans­plant from less than 1 year to less than 40 years, no tMDS,
were seen in the phase 3 MDS-005 trial after those 4 cycles.25 no TP53 or Ras path­way gene muta­tion, no throm­bo­cy­to­pe­nia,
In the same study, the authors explored gene muta­tions and and mar­row blasts less than 15%.40 Although not a clas­si­cal def­i­ni­
response to LEN.33 The ana­ly­ses revealed that patients with 4 or tion of LR patients, this cohort of chil­dren and young adults had a
more muta­tions were more likely to have a high serum eryth­ro­ favor­able 3-year OS of 82%.40 The sup­ple­men­tary data review OS
poi­et­in (EPO) level (>500 mU/mL); in addi­tion, high serum EPO curves by IPSS spe­cif­i­cally and sug­gest that low-risk and inter­me­
level was asso­ci­ated with the pres­ence of muta­tions in any of di­ate 1 patients with­out TP53 muta­tions have an OS at 3 years of
5 genes asso­ci­ated with poorer out­comes in the Bejar et al34 approx­i­ma­tely 50%.
anal­y­sis (ASXL1, ETV6, EZH2, RUNX1, and TP53). Taken together To aid in deci­sion mak­ing, some series from the EBMT and
with the observed asso­ci­a­tion between ASXL1 muta­tions and Center for International Blood and Marrow Transplant Research

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/428/1851953/428dezern.pdf by guest on 13 December 2021


low response rate to LEN, this pro­vi­des fur­ther insight into pre­ have devel­oped scor­ing sys­tems or deci­sion mod­els that adjust
vi­ous data that high base­line EPO level pre­dicts for non­re­sponse to indi­vid­ual patient risk. Although not spe­cific to LR patients, the
to LEN in these LR patients.25,33 Finally, even in LR patients, non­ molec­u­lar and other clin­i­cal fea­tures asso­ci­ated with less desir­
re­sponse to HMAs was asso­ci­ated with a median sur­vival of 17 able out­comes can been seen. Both of these series had an OS in
months in a US MDS con­sor­tium study,35 which cor­re­lates with LR patients of about 70% at 3 years or 4 years.41,44 The Gruppo
increas­ing num­ber of muta­tions.36 Thus, response to all­lines of Italiano Trapianto Midollo Osseo e Terapia Cellulare reported a
ther­apy in LR MDS should also be con­sid­ered for tran­si­tions of BMT Mar­kov anal­y­sis on 1728 patients with MDS cat­e­go­rized by
ther­apy. Clinical tri­als should also always be con­sid­ered. Increas­ the IPSS-R. Similar to older ana­ly­ses, a sur­vival advan­tage was
ingly, tri­als for patients with MDS include a clause for eli­gi­bil­ity seen when delaying trans­plant in very low- and low-risk patients,
that allows for upstaging, such as “patients with inter­me­di­ate whereas post­pone­ment was del­e­te­ri­ous in inter­me­di­ate-risk
risk by R-IPSS with high-risk molec­ul­ar fea­tures includ­ing TP53, patients.45 This is in keep­ing with the cur­rent approach to only
ASXL1, EZH2, and/or RUNX1 muta­tions are also eli­gi­ble.” I believe con­sider BMT in LR patients with dis­ease evo­lu­tion or lack of
this is a rea­son­able prac­tice to apply broadly. response to pri­mary treat­ments. Patients with increased RBC
needs, lack of response to HMAs, bone mar­ row fibro­ sis, HR
Consideration of allo­ge­neic trans­plan­ta­tion (BMT) in LR molec­u­lar fea­tures, and tMDS are asso­ci­ated with a poor prog­
dis­ease no­sis, and early con­sid­er­ation for BMT is often suggested.35,46,47
Historically, BMT has been reserved for patients with HR MDS, Genomic fea­tures are rel­e­vant for predicting sur­vival after BMT,
given its poten­tial cura­tive ben­e­fit is off­set by the mor­bid­ity and supporting the ratio­nale to include this infor­ma­tion for trans­plan­
mor­tal­ity that can result from the pro­ce­dure.37,38 Nonetheless, ta­tion deci­sion mak­ing in MDS.48,49 Many of these fea­tures would
efforts have been made to weigh the toxicities of this “higher have been appli­ca­ble in both patients 1 and 3.
risk, higher reward” pro­ce­dure in LR MDS, and many LR patients Additional data includ­ing pro­spec­tive stud­ies, poten­tially
are offered BMT. The National Comprehensive Network Guide­ with a ran­ dom­ ized design com­ pared with the cur­ rent stan­
lines and other con­sen­sus guide­lines rec­om­mend BMT for pa­ dard of delaying BMT, and includ­ing a wider donor pool with
tients with MDS early in their dis­ease if they have inter­me­di­ate 2 haploidentical donors are needed for an improved under­stand­
or high-risk cat­e­gory per the IPSS and later dur­ing their dis­ease ing and estab­lish­ment of the BMT approach for LR patients. An
course (prior to AML pro­gres­sion) if they have LR dis­ease. ongo­ ing pro­ spec­tive nonrandomized trial in France is enroll­
Although no pro­ spec­ tive series exist cur­ rently, there are ing patients with LR dis­ ease and at least 1 high-risk fea­ ture
reported out­ comes of BMT in LR patients included in many (NCT02757989), and results will be use­ful to the field. Nonethe­
ret­ro­spec­tive series.39-43 Only 1 ded­i­cated ret­ro­spec­tive study less, BMT remains the only poten­tially cura­tive option for MDS,
reviewed out­ comes in LR patients, and the results were not and no options avail­­able at pres­ent seem to be a ­ ble to replace
encour­ag­ing.42 In a study of patients who under­went trans­plan­ta­ this path to poten­tial cure; we must use this for patients when
tion between 2000 and 2011 by the European Society for Blood the risk/ben­e­fit pro­file is ratio­nal.
and Marrow Transplantation (EBMT), 246 IPSS low-risk (21%) or
inter­me­di­ate 1 (79%) patients had a 3-year OS and PFS of 57% Conclusions
and 54%, respec­tively. There was a high nonrelapse mor­tal­ity LR MDS is a het­ero­ge­neous group of dis­or­ders. Currently avail­­
at 30%. The best out­come was seen in patients who received able prog­nos­tic scor­ing sys­tems aid in risk strat­i­fi­ca­tion but do
periph­eral blood stem cells and had a matched related donor, not cap­ture all­impor­tant var­i­ables, and serial reassessment of
although alter­na­tive donors were rel­a­tively rare. The con­di­tion­ the patient to fully clas­sify indi­vid­ual risk is impor­tant. The goal
ing reg­i­men inten­sity and patient age did not appear to influ­ence is a pro­ac­tive approach in all­LR patients to extend quan­tity of
out­come in this het­er­og­e­nous series.42 In a series from Asia, LR life with qual­ity through use of all­avail­­able treat­ments avail­­able.
patients had a sig­nif­i­cantly bet­ter out­come than HR patients.43
Prognosis was also sig­nif­i­cantly influ­enced by the genetic risk. Conflict-of-inter­est dis­clo­sure
LR patients who dis­play HR fea­tures such as a com­plex kar­yo­ Amy E. DeZern has received hon­ o­
raria from Bristol Meyers
type or TP53 muta­tions do poorly, whereas LR 5q minus patients Squibb, Novartis, Taiho, and Abbvie as a con­sul­tant.
had an OS of about 40%.43 Additional expla­na­tions of incor­po­
ra­tion of bio­logic data into BMT out­come ana­ly­ses are seen in a Off-label drug use
large series from the Center for International Blood and Marrow Amy E. DeZern: imetelstat or eltrombopag for MDS.

432  |  Hematology 2021  |  ASH Education Program


Correspondence 22. Feurstein S, Churpek JE, Walsh T, et al. Germline var­i­ants drive myelodys­
Amy E. DeZern, Division of Hematologic Malignancies, Sidney plastic syn­drome in young adults. Leukemia. 2021;35(8):2439-2444.
23. Schratz KE, Haley L, Danoff SK, et al. Cancer spec­trum and out­comes in the
Kimmel Cancer Center at Johns Hopkins, 1650 Orleans St, CRBI Men­de­lian short telo­mere syn­dromes. Blood. 2020;135(22):1946-1956.
Room 3M87, Baltimore, MD 21287-0013; e-mail: adezern1@jhmi​ 24. Fenaux P, Platzbecker U, Mufti GJ, et al. Luspatercept in patients with low­
­.edu. er-risk myelodysplastic syn­dromes. N Engl J Med. 2020;382(2):140-151.
25. Santini V, Almeida A, Giagounidis A, et  al. Randomized phase III study
References of lenalidomide ver­ sus pla­ cebo in RBC trans­ fu­sion-depen­dent patients
1. Greenberg PL, Tuechler H, Schanz J, et al. Revised inter­na­tional prog­nos­tic with lower-risk non-del(5q) myelodysplastic syn­ dromes and inel­ i­
gi­
scor­ing sys­tem for myelodysplastic syn­dromes. Blood. 2012;120(12):2454- ble for or refrac­tory to eryth­ro­poi­e­sis-stim­u­lat­ing agents. J Clin Oncol.
2465. 2016;34(25):2988-2996.
2. Bejar R et al. Somatic mutations in MDS patients are associated with clin­ 26. Jabbour E, Short NJ, Montalban-Bravo G, et al. Randomized phase 2 study
ical features and predict prognosis independent of the IPSS-R: Analysis of of low-dose decitabine vs low-dose azacitidine in lower-risk MDS and
combined datasets from the International Working Group for Prognosis in MDS/MPN. Blood. 2017;130(13):1514-1522.
MDS-Molecular Committee/Blood/American Society of Hematology (ash­ 27. Stahl M, DeVeaux M, de Witte T, et al. The use of immu­no­sup­pres­sive ther­
publications.org). Blood. 2015;126(23):907. apy in MDS: clin­i­cal out­comes and their pre­dic­tors in a large inter­na­tional

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/428/1851953/428dezern.pdf by guest on 13 December 2021


3. Kuendgen A, Nomdedeu M, Tuechler H, et  al. Therapy-related myelo­ patient cohort. Blood Adv. 2018;2(14):1765-1772.
dysplastic syn­dromes deserve spe­cific diag­nos­tic sub-clas­si­fi­ca­tion and 28. Oliva EN, Alati C, Santini V, et al. Eltrombopag ver­sus pla­cebo for low-risk
risk-strat­i­fi­ca­tion-an approach to clas­si­fi­ca­tion of patients with t-MDS. Leu- myelodysplastic syn­dromes with throm­bo­cy­to­pe­nia (EQoL-MDS): phase 1
kemia. 2021;35(3):835-849. results of a sin­gle-blind, randomised, con­trolled, phase 2 supe­ri­or­ity trial.
4. Bersanelli M, Travaglino E, Meggendorfer M, et al. Classification and per­ Lancet Haematol. 2017;4(3):e127-e136.
son­al­ized prog­nos­tic assess­ment on the basis of clin­i­cal and geno­mic fea­ 29. Kantarjian HM, Fenaux P, Sekeres MA, et  al. Long-term fol­low-up for up
tures in myelodysplastic syn­dromes. J Clin Oncol. 2021;39(11):1223-1233. to 5 years on the risk of leukaemic pro­ gres­sion in throm­ bo­cy­to­
pe­nic
5. Duncavage EJ, Schroeder MC, O’Laughlin M, et al. Genome sequenc­ing as patients with lower-risk myelodysplastic syn­dromes treated with romi­
an alter­na­tive to cyto­ge­netic anal­y­sis in mye­loid can­cers. N Engl J Med. plostim or pla­cebo in a randomised dou­ble-blind trial. Lancet Haematol.
2021;384(10):924-935. 2018;5(3):e117-e126.
6. Swerdlow S. WHO Classification of Tumours of Haematopoietic and Lym- 30. Steensma DP, Fenaux P, Van Eygen K, et al. Imetelstat achieves mean­ing­ful
phoid Tissues. Lyon International Agency for Research on Cancer; 2017. and dura­ble trans­fu­sion inde­pen­dence in high trans­fu­sion-bur­den patients
7. Nazha A, Seastone DP, Keng M, et al. The Revised International Prognostic with lower-risk myelodysplastic syn­dromes in a phase II study. J Clin Oncol.
Scoring System (IPSS-R) is not pre­dic­tive of sur­vival in patients with sec­ 2021;39(1):48-56.
ond­ary myelodysplastic syn­dromes. Leuk Lymphoma. 2015;56(12):3437- 31. Prebet T, Cluzeau T, Park S, et al. Outcome of patients treated for myelo­
3439. dysplastic syn­dromes with 5q dele­tion after fail­ure of lenalidomide ther­
8. Zeidan AM, Al Ali N, Barnard J, et al. Comparison of clin­i­cal out­comes and apy. Oncotarget. 2017;8(47):81926-81935.
prog­nos­tic util­ity of risk strat­i­fi­ca­tion tools in patients with ther­apy-related 32. Prebet T, Toma A, Cluzeau T, et al. Outcome of patients treated for myel­
vs de novo myelodysplastic syn­dromes: a report on behalf of the MDS Clin­ odysplastic syn­dromes with­out dele­tion 5q after fail­ure of lenalidomide
ical Research Consortium. Leukemia. 2017;31(6):1391-1397. ther­apy. Oncotarget. 2017;8(23):37866-37874.
9. Kuzmanovic T, Patel BJ, Sanikommu SR, et al. Genomics of ther­apy-related 33. Santini V, Fenaux P, Giagounidis A, et  al. Impact of somatic muta­tions on
mye­loid neo­plasms. Haematologica. 2020;105(3):e98-e101. response to lenalidomide in lower-risk non-del(5q) myelodysplastic syn­
10. Bolton KL, Ptashkin RN, Gao T, et  al. Cancer ther­apy shapes the fit­ness dromes patients. Leukemia. 2021;35(3):897-900.
land­scape of clonal hema­to­poi­e­sis. Nat Genet. 2020;52(11):1219-1226. 34. Bejar R, Stevenson K, Abdel-Wahab O, et al. Clinical effect of point muta­
11. Ramos F, Robledo C, Izquierdo-García FM, et al; Span­ish Group for Myelo­ tions in myelodysplastic syndromes. N Engl J Med. 2011;364:2496-506.
dysplastic Syndromes (GESMD). Bone mar­row fibro­sis in myelodysplastic 35. Jabbour EJ, Garcia-Manero G, Strati P, et al. Outcome of patients with low-
syn­dromes: a pro­spec­tive eval­u­a­tion includ­ing muta­tional anal­y­sis. Onco- risk and inter­ me­di­ate-1-risk myelodysplastic syn­ drome after hypometh­
target. 2016;7(21):30492-30503. ylating agent fail­ ure: a report on behalf of the MDS Clinical Research
12. Hiwase DK, Singhal D, Strupp C, et al. Dynamic assess­ment of RBC-trans­fu­sion Consortium. Cancer. 2015;121(6):876-882.
depen­dency improves the prog­nos­tic value of the revised-IPSS in MDS 36. Montalban-Bravo G, Takahashi K, Patel K, et  al. Impact of the num­ber of
patients. Am J Hematol. 2017;92(6):508-514. muta­tions in sur­vival and response out­comes to hypomethylating agents
13. Itzykson R, Crouch S, Travaglino E, et al; Euro­pean MDS Registry mem­bers. in patients with myelodysplastic syn­dromes or myelodysplastic/mye­lo­
Early plate­let count kinet­ics has prog­nos­tic value in lower-risk myelodys­ pro­lif­er­a­tive neo­plasms. Oncotarget. 2018;9(11):9714-9727.
plastic syn­dromes. Blood Adv. 2018;2(16):2079-2089. 37. Cutler CS, Lee SJ, Greenberg P, et al. A deci­sion anal­y­sis of allo­ge­neic bone
14. Benton CB, Khan M, Sallman D, et al. Prognosis of patients with inter­me­di­ mar­row trans­plan­ta­tion for the myelodysplastic syn­dromes: delayed trans­
ate risk IPSS-R myelodysplastic syn­drome indi­cates var­i­able out­comes and plan­ta­tion for low-risk myelodysplasia is asso­ci­ated with improved out­
need for mod­els beyond IPSS-R. Am J Hematol. 2018;93(10):1245-1253. come. Blood. 2004;104(2):579-585.
15. Malcovati L, Stevenson K, Papaemmanuil E, et al. SF3B1-mutant MDS as a 38. Koreth J, Pidala J, Perez WS, et al. Role of reduced-inten­sity con­di­tion­ing
dis­tinct dis­ease sub­type: a pro­posal from the International Working Group allo­ge­neic hema­to­poi­etic stem-cell trans­plan­ta­tion in older patients with
for the Prognosis of MDS. Blood. 2020;136(2):157-170. de novo myelodysplastic syn­dromes: an inter­na­tional col­lab­o­ra­tive deci­
16. Dalton WB, Helmenstine E, Pieterse L, et al. The K666N muta­tion in SF3B1 sion anal­y­sis. J Clin Oncol. 2013;31(21):2662-2670.
is asso­ci­ated with increased pro­gres­sion of MDS and dis­tinct RNA splic­ing. 39. Polona Novak P, Zabelina T, Wolschke C, Ayuk F, Christopeit M, Kröger N.
Blood Adv. 2020;4(7):1192-1196. Allogeneic stem cell trans­plan­ta­tion for patients with lower-risk myelodys­
17. Bejar R, Stevenson KE, Caughey BA, et al. Validation of a prog­nos­tic model plastic syn­drome. Biol Blood Marrow Transplant. 2020;26(11):2047-2052.
and the impact of muta­tions in patients with lower-risk myelodysplastic 40. Lindsley RC, Saber W, Mar BG, et al. Prognostic muta­tions in myelodysplas­
syn­dromes. J Clin Oncol. 2012;30(27):3376-3382. tic syn­drome after stem-cell trans­plan­ta­tion. N Engl J Med. 2017;376(6):536-
18. DeZern AE. Treatments targeting MDS genet­ics: a fool’s errand? Hematol- 547.
ogy Am Soc Hematol Educ Program. 2018;2018(1):277-285. 41. Gagelmann N, Eikema D-J, Stelljes M, et  al. Optimized EBMT trans­plant-
19. Bernard E, Nannya Y, Hasserjian RP, et al. Implications of TP53 alle­lic state spe­cific risk score in myelodysplastic syn­dromes after allo­ge­neic stem-cell
for genome sta­bil­ity, clin­i­cal pre­sen­ta­tion and out­comes in myelodysplas­ trans­plan­ta­tion. Haematologica. 2019;104(5):929-936.
tic syn­dromes. Nat Med. 2020;26(10):1549-1556. 42. Robin M, Porcher R, Zinke-Cerwenka W, et al. Allogeneic haematopoietic
20. Schratz KE, DeZern AE. Genetic pre­dis­po­si­tion to myelodysplastic syn­ stem cell trans­plant in patients with lower risk myelodysplastic syn­drome:
drome in clin­i­cal prac­tice. Hematol Oncol Clin North Am. 2020;34(2):333- a ret­ro­spec­tive anal­y­sis on behalf of the Chronic Malignancy Working Party
356. of the EBMT. Bone Marrow Transplant. 2017;52(7):1081.
21. Greenberg PL, Stone RM, Al-Kali A, et al. Myelodysplastic syn­dromes, ver­ 43. Yoshizato T, Nannya Y, Atsuta Y, et al. Genetic abnor­mal­i­ties in myelodys­
sion 2.2017, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr plasia and sec­ond­ary acute mye­loid leu­ke­mia: impact on out­come of stem
Canc Netw. 2017;15(1):60-87. cell trans­plan­ta­tion. Blood. 2017;129(17):2347-2358.
Prakash Singh Shekhawat
Lower risk but high risk | 433
44. Shaffer BC, Ahn KW, Hu Z-H, et al. Scoring sys­tem prog­nos­tic of out­come 48. Della Porta MG, Alessandrino EP, Bacigalupo A, et al; Gruppo Italiano Trapi­
in patients under­go­ing allo­ge­neic hema­to­poi­etic cell trans­plan­ta­tion for anto di Midollo Osseo. Predictive fac­tors for the out­come of allo­ge­neic
myelodysplastic syn­drome. J Clin Oncol. 2016;34(16):1864-1871. trans­plan­ta­tion in patients with MDS strat­i­fied according to the revised
45. Della Porta MG, Jackson CH, Alessandrino EP, et  al. Decision anal­y­sis of IPSS-R. Blood. 2014;123(15):2333-2342.
allo­ge­neic hema­to­poi­etic stem cell trans­plan­ta­tion for patients with myel­ 49. Della Porta MG, Gallì A, Bacigalupo A, et al. Clinical effects of driver somatic
odysplastic syn­ drome strat­ i­fied according to the revised International muta­tions on the out­comes of patients with myelodysplastic syn­dromes
Prognostic Scoring System. Leukemia. 2017;31(11):2449-2457. treated with allo­ge­neic hema­to­poi­etic stem-cell trans­plan­ta­tion. J Clin
46. de Witte T, Bowen D, Robin M, et al. Allogeneic hema­to­poi­etic stem cell Oncol. 2016;34(30):3627-3637.
trans­plan­ta­tion for MDS and CMML: rec­om­men­da­tions from an inter­na­
tional expert panel. Blood. 2017;129(13):1753-1762.
47. Prébet T, Gore SD, Esterni B, et al. Outcome of high-risk myelodysplastic
syn­drome after azacitidine treat­ment fail­ure. J Clin Oncol. 2011;29(24):3322- © 2021 by The Amer­i­can Society of Hematology
3327. DOI 10.1182/hema­tol­ogy.2021000277

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/428/1851953/428dezern.pdf by guest on 13 December 2021

434  |  Hematology 2021  |  ASH Education Program


MDS: BEYOND A ONE - SIZE - FITS -ALL APPROACH

EVIDENCE-BASED MINIREVIEW

Molecular precision and clinical uncertainty:

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/435/1852132/435richardson.pdf by guest on 13 December 2021


should molecular profiling be routinely used
to guide risk stratification in MDS?
Daniel R. Richardson1 and Amy E. DeZern2
Division of Hematology, Department of Medicine, Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC;
1

Department of Oncology, Sidney Kimmel Cancer Center at Johns Hopkins, Baltimore, MD


2

This is a focused clinical vignette and review of the literature in MDS to discuss the application of molecular sequencing
for risk stratification in MDS. The authors utilize an exemplar patient case and explain the advantages and disadvantages,
based on available data, of routine use of this testing for MDS patients.

LEARNING OBJECTIVES
• Review the prognostic significance of mutational profiles in MDS
• Identify appropriate approaches to use molecular profiling for risk stratification in MDS

development of next­generation sequencing (NGS) assays


CLINICAL CASE has allowed for the identification of more than 50 different
A 67­year­old woman sought treatment for progressive recurrent driver gene mutations that are associated with
fatigue initially attributed to caring for her 18­month­old MDS.1,2 Most patients with MDS will have at least 1 identi­
grandson. She was found to have pancytopenia (hemo­ fied somatic mutation, whereas a typical patient will har­
globin of 10.2 g/dL, mean cell volume of 104 fL, platelet bor a median of 2 or 3 driver mutations.3 NGS assays are
count of 77 × 109/L, and white blood cell count of 2.1 × 109/L increasingly used in the diagnostic evaluation of patients
with absolute neutrophil count of 0.7 × 109/L). Bone mar­ with cytopenias.4 Numerous studies have also evaluated the
row biopsy specimen demonstrated trilineage dysplasia prognostic significance of such mutations. Best practices
with 1% blasts. Cytogenetics were 46,XX [20]. Interna­ for integrating molecular testing into existing risk prediction
tional Prognostic Scoring System – Revised (IPSS­R) risk modeling have yet to be established, and substantial clinical
category was low. Mutational testing demonstrated muta­ variability exists among practicing hematologists.5 Here we
tions in ASXL1 (variant allele fraction [VAF] 34%), SRSF2 review the current evidence for use of molecular profiling to
(VAF 42%), TET2 (VAF 18%), and SETBP1 (VAF 12%). Does guide risk stratification in clinical practice.
this molecular profile modify risk stratification or manage­
ment decisions in this patient? Molecular precision
Several notable studies using NGS and high­throughput
sequencing have demonstrated the independent prog­
Introduction nostic significance of recurrent molecular mutations in
Myelodysplastic syndromes (MDS) are a group of related patients with MDS. Consistently identified driver mutant
hematologic neoplasms that cause dysplasia and ineffective genes include mutations involved in RNA splicing (SF3B1,
hematopoiesis and have a propensity to progress to acute SRSF2, U2AF1, and ZRSR2), DNA methylation (TET2, DN-
myeloid leukemia (AML). The diagnosis of MDS requires mor­ MT3A, IDH1, and IDH2), chromatin modification (ASXL1 and
phologic dysplasia in 1 or more cell lines and/or character­ EZH2), transcription regulation (RUNX1), DNA repair (TP53),
istic cytogenetic abnormalities. Over the past decade, the and signal transduction (CBL, NRAS, and KRAS).6
Prakash Singh Shekhawat
Routine molecular profiling in MDS for risk | 435
Papaemmanuil et al1 sequenced 111 genes across 738 patients IPSS-R, dem­on­strat­ing the fea­si­bil­ity and impor­tance of incor­po­
with MDS, chronic myelomonocytic leukemia, and MDS/mye­lo­ rat­ing molec­u­lar data into prog­nos­tic mod­els. By apply­ing com­
pro­lif­er­a­tive neo­plasms. Mutations in SF3B1 (24%), TET2 (22%), and pu­ta­tional approaches to large inter­na­tional data­bases, Bersanelli
SFSF2 (14%) were most com­mon, followed by ASXL1, DNMT3A, et al16 explored the com­plex­ity of mul­ti­ple coexisting muta­tions
RUNX1, U2AF1, TP53, and EZH2. Mutations in TP53, U2AF1, RUNX1, and iden­ti­fied 8 prognostically dis­tinct “geno­mic sub­types” of
SRSF2, IDH2, CUX1, ASXL1, and BCOR were neg­a­tively asso­ci­ated MDS such as MDS with TP53 muta­tions and/or com­plex kar­yo­
with over­all sur­vival (OS), whereas muta­tions in SF3B1 were asso­ type, MDS with AML-like muta­tional pat­terns, and SRSF2-related
ci­ated with improved OS. Leukemia-free sur­vival was neg­a­tively MDS. Furthermore, by inte­grat­ing 63 clin­i­cal and geno­mic var­i­
asso­ci­ated with increased num­ber of driver muta­tions and cyto­ ables, the authors were a ­ ble to develop a model to gen­er­ate an
ge­netic abnor­mal­i­ties. esti­mated per­son­al­ized prob­a­bil­ity of sur­vival, which also sig­nif­i ­
Bejar et al7 in a study using NGS and mass spec­trom­e­try– cantly improved upon IPSS-R prog­nos­ti­ca­tion. Such approaches
based genotyping of 439 patients with MDS dem­on­strated that likely bet­ter rep­re­sent the dis­ease biol­ogy in MDS, but fur­ther
muta­tions in 10 genes were asso­ci­ated with OS (TET2, ASXL1, work needs to be done to dem­on­strate the util­ity of such com­

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/435/1852132/435richardson.pdf by guest on 13 December 2021


RUNX1, TP53, EZH2, NRAS, JAK2, ETV6, CBL, and IDH2). Mutations plex risk strat­i­fi­ca­tion sys­tems for clin­i­cal deci­sion mak­ing.
in 6 genes (TP53, EZH2, ETV6, RUNX1, CBL, and ASXL1) neg­a­tively
affected OS after adjusting for IPSS risk group and age. The pres­ Clinical uncer­tainty
ence of a TP53 muta­tion had the larg­est inde­pen­dent asso­ci­a­tion Despite the remark­ able tech­ no­log­

cal prog­ ress that has en­
with sur­vival among muta­tions (haz­ard ratio, 2.48). abled rapid molec­u­lar pro­fil­ing of patients with MDS and pre­
Haferlach et al3 used high-through­put deep sequenc­ing of cur­sor states, sub­stan­tial clin­i­cal uncer­tainty remains about
104 genes to ana­lyze the geno­mic and clin­i­cal data of more how to apply molec­ul­ar data to indi­vid­ual patients. Numerous
than 900 patients with MDS. Twenty-five genes were iden­ti­fied prog­nos­tic mod­els have already been val­i­dated for use in MDS
to be asso­ci­ated with sur­vival out­comes on uni­var­i­ate anal­y­sis. and are in wide­ spread use, includ­ ing the IPSS, MDS WHO
Only muta­tions in SF3B1 were asso­ci­ated with an improved clin­ Classification-based Prognostic Scoring System (WPSS), Lower-Risk
i­cal out­come. Other recent ana­ly­ses con­firm the impor­tance of Prognostic Scoring System (LR-PSS), Global MD Anderson Prog­
muta­tions iden­ti­fied in these stud­ies, high­light­ing the prog­nos­ nostic Scoring System (MDAPSS), French Prognostic Scoring
tic value of spe­cific muta­tions inde­pen­dent of IPSS-R.5,8 System (FPSS), and IPSS-R, among oth­ers. Multiple novel mod­els
Variant allele fre­quency is the per­cent­age of var­i­ant reads have been pro­posed that seek to com­bine existing prog­nos­tic
in the total num­ber of reads (var­i­ant plus ref­er­ence reads) and scor­ing sys­tems with muta­tional data from emerg­ing stud­ies.1,3,16
is used to approx­ im
­ ate the pro­ por­tion of DNA mol­ e­
cules in Clinical use of these advanced mod­els incor­po­rat­ing genetic
the orig­i­nal spec­i­men that carry the spe­cific var­i­ant. Tradition­ data is cur­rently lim­ited due to the com­plex­ity of the mod­els
ally, muta­tions with a VAF of 5% or less are not thought to be and the lack of fur­ther val­i­da­tion stud­ies. A stan­dard scor­ing sys­
clin­i­cally sig­nif­i­cant. More data are emerg­ing that prog­nos­tic tem incor­po­rat­ing molec­u­lar data and clin­i­co­path­o­logic data
out­comes asso­ci­ated with indi­vid­ual muta­tions may be depen­ remains to be established.
dent on the VAF of muta­tions, although this effect may be lim­ The lack of an accepted prog­nos­tic scor­ing sys­tem incor­
ited to cer­tain muta­tions (such as TP53, TET2, and SF3B1).9-12 In po­rat­ing molec­u­lar data makes prog­nos­ti­ca­tion for indi­vid­ual
TP53-mutated MDS, for exam­ple, increas­ing VAF was asso­ci­ated patients par­tic­ul­arly chal­leng­ing. Hematologists already face the
with worse prog­no­sis, with an increase in haz­ard ratio of 1.02% chal­lenge of deter­min­ing which existing risk pre­dic­tion model
per 1% VAF increase.13 Patients with a low TP53-muta­tional bur­ best applies to each patient; the addi­tion of molec­u­lar pro­fil­
den (VAF <10%) and with­out a com­plex kar­yo­type may have out­ ing adds yet another layer of com­ plex­ity and clin­i­
cal uncer­
comes bet­ter than expected. Those patients with a high VAF are tainty. Multiple ques­ tions remain: Are the adverse effects of
less likely to respond to hypomethylating agents.13 muta­tions on out­comes uni­form across stan­dard­ized risk mod­
Molecular pro­ fil­
ing is also essen­ tial in esti­
mat­
ing risk for els? Are they uni­form across risk categories? Is there a ceil­ing
patients with clonal hema­to­poi­e­sis of inde­ter­mi­nate poten­ effect to adverse prog­nos­tic fac­tors? For patients with mul­ti­ple
tial (CHIP) and clonal cytopenia of unde­ter­mined sig­nif­i­cance muta­ tions iden­ ti­fied, how should risk be esti­ mated? Multiple
(CCUS), pre­cur­sor states to MDS. For exam­ple, in patients with stud­ies have dem­on­strated the adverse effect of hav­ing mul­ti­
CCUS, muta­ tional sta­ tus is pre­dic­
tive of risk of pro­gres­
sion: ple coexisting muta­tions.1,3,7 Should this “comutation” effect be
muta­tions in U2AF1, ZRSR2, SRSF2, JAK2, or RUNX1 are asso­ci­ thought of in par­al­lel or in sequence to the effect of indi­vid­ual
ated with a high risk of pro­gres­sion to MDS (as high as 80%- muta­tions? These ques­tions, along with many oth­ers, remain to
90% at 5 years), whereas iso­lated muta­tions in TET2, ASXL1, and be addressed to improve risk strat­i­fi­ca­tion for patients with MDS.
DNMT3A are asso­ci­ated with a lower risk of pro­gres­sion (~50%
at 5 years).14 Similar to find­ings in MDS, emerg­ing data have dem­ Implications for clin­i­cal prac­tice
on­strated that dif­fer­ent muta­tional pat­terns and VAF val­ues are Although the use of molec­u­lar pro­fil­ing adds com­plex­ity to prog­
likely impor­tant in predicting over­all risk in CHIP and CCUS.15 nos­ti­ca­tion and obtaining NGS is often hin­dered by the lack of
Although iden­ti­fic ­ a­tion of somatic muta­tions by NGS is an insur­ance cov­er­age, we sug­gest com­plet­ing molec­u­lar pro­fil­ing
impor­tant step for­ward in risk strat­i­fi­ca­tion for patients with MDS, on all­patients suspected or con­firmed to have MDS. The rea­son
indi­vid­ual muta­tions are only a part of a more com­plex land­scape for this is straight­for­ward: in some patients, molec­u­lar pro­fil­ing
of geno­mic abnor­mal­it­ies that drive pathol­ogy and deter­mine will sub­stan­tially change man­age­ment. For the patient men­tioned
out­comes. Using the same cohort of 944 patients men­tioned above, although IPSS-R risk is low, sev­eral fea­tures of the molec­u­lar
above, Haferlach et al3 pro­posed novel prog­nos­tic mod­els incor­ pro­file are impor­tant. The ASXL1 muta­tion, assum­ing it is a canon­
po­rat­ing genetic data that strat­ify patients into 4 prognostically i­cal muta­tion resulting in a read­ing frame­shift, has been shown
dis­tinct sub­groups. These mod­els con­sis­tently outperformed to be asso­ci­ated with worse out­comes inde­pen­dent of IPSS-R.7

436  |  Hematology 2021  |  ASH Education Program


Furthermore, the fact that 4 onco­genic muta­tions were iden­ti­fied enroll­ment in a clin­i­cal trial for patients with a com­plex kar­yo­
also por­tends worse out­comes. The median leu­ke­mia-free sur­vival type and a TP53 muta­tion due to recently emerg­ing encour­ag­ing
for patients with 4 iden­ti­fied muta­tions was only 18 months in the results of small mol­e­cules that induce apo­pto­sis in TP53-mutated
study by Papaemmanuil et al.1 So, although by IPSS-R, this patient cells such as APR-246. Response rates of more than 70% with a 50%
is low risk (esti­mated OS of 5.3 years and leu­ke­mia-free sur­vival of com­plete remis­sion rate were shown in 29 patients with MDS
10.8 years), the molec­u­lar pro­file should prompt more fre­quent receiv­ing APR-246 on the phase 1b/2 trial.28
clin­i­cal mon­i­tor­ing and make con­sid­er­ation of early ther­apy rea­
son­able, includ­ing dis­cus­sions of an allo­ge­neic stem cell trans­ Targetable muta­tions
plant can­di­dacy. Several addi­tional sce­nar­ios require men­tion­ing. Although there are cur­rently no ther­a­pies approved by the US Food
and Drug Administration that tar­get indi­vid­ual muta­tions in MDS,
Upstaging lower-risk dis­ease the iden­ti­fi­ca­tion of spe­cific muta­tions may make a patient eli­gi­ble
Patients with lower-risk MDS often expe­ri­ence long-term sur­ for AML-style treat­ments in which targeted agents are approved or
vival with­out the need for aggres­sive ther­apy such as allo­ge­ for a clin­i­cal trial. Current ther­a­peu­tic clin­i­cal tri­als include patients

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/435/1852132/435richardson.pdf by guest on 13 December 2021


neic hema­to­poi­etic cell trans­plant. The iden­ti­fi­ca­tion of patients with iden­ti­fied muta­tions in TP53 (ClinicalTrials​­.gov Identifier:
who are higher risk for dis­ease com­pli­ca­tions is impor­tant to NCT04638309), IDH1 (numer­ous stud­ies includ­ing NCT03173248,
guide ther­a­peu­tic deci­sions. In a large anal­y­sis of patients pre­ NCT03503409, NCT03839771), IDH2 (NCT04522895, NCT03839771,
dicted to have lower-risk MDS by MDS-LR, the iden­ti­fi­ca­tion of NCT03383575), RUNX1 (NCT04874194), TET2 (NCT03397173), FLT3
a muta­tion in EZH2 predicted worse-than-expected out­comes (NCT04493138, NCT04027309), CBL (NCT04493138), and mul­ti­ple
after adjusting for mul­ti­ple rel­e­vant covariates.17 Similarly, the muta­tions (NCT02670525).
find­ing of a muta­tion with an inde­pen­dent poor prog­nos­tic sig­
nif­i­cance, includ­ing ASXL1, SRSF2, ETV6, RUNX1, and TP53, in
patients should prompt con­sid­er­ation of ear­lier ini­ti­a­tion of dis­ Existing chal­lenges
ease-mod­i­fy­ing ther­apy and/or closer clin­i­cal mon­i­tor­ing as in Obtaining qual­ity molec­u­lar pro­fil­ing on some patients remains
the case above. Furthermore, the iden­ti­fi­ca­tion of a muta­tion in a chal­lenge. Many insur­ance com­pa­nies deny cov­er­age for NGS
NPM1 (~4% of cases) has been asso­ci­ated with an aggres­sive dis­ assays or require prior autho­ri­za­tion that may be dif­fi­cult to ob­
ease course and rel­a­tively rapid pro­gres­sion to AML often with­ tain. We encour­age clear cod­ing as MDS to help with bill­ing cov­
in 12 months of diag­no­sis.18 Treating patients with NPM1 muta­ er­age. A let­ter of med­i­cal neces­sity cit­ing the National Compre­
tions sim­i­lar to patients with AML with inten­sive che­mo­ther­apy hensive Cancer Network guide­lines may also be of use.
and allo­ge­neic stem cell trans­plant has been asso­ci­ated with Some ref­er­ence lab­o­ra­to­ries are not rou­tinely reporting VAF,
improved out­comes.19 Similarly, FLT3 muta­tions, which are less which sub­stan­tially lim­its the inter­pre­ta­tion of data, and/or are
fre­quent in MDS com­pared with AML (~2% vs 35%), are asso­ci­ not clearly delin­eat­ing whether indi­vid­ual muta­tions are path­o­
ated with a com­plex kar­yo­type and a more rapid pro­gres­sion to genic. A con­sen­sus panel by the Association for Molecular Pathol­
AML, although the data are mixed.20-22 The iden­ti­fi­ca­tion of a new ogy with liai­son rep­re­sen­ta­tion from the Amer­i­can College of
FLT3-ITD muta­tion at the time of pro­gres­sion from MDS to AML is Medical Genetics and Genomics, Amer­ i­
can Society of Clinical
asso­ci­ated with dis­mal out­comes (1 month median OS).23 Oncology, and College of Amer­i­can Pathologists rec­om­mends
a 4-tiered reporting approach: tier I, var­i­ants with strong clin­i­cal
SF3B1-mutated MDS sig­nif­i­cance; tier II, var­i­ants with poten­tial clin­i­cal sig­nif­i­cance; tier
SF3B1-muated MDS is often con­ sid­
ered a dis­
tinct sub­
type of III, var­i­ants of unknown clin­i­cal sig­nif­i­cance; and tier IV, var­i­ants
MDS with an indo­lent course.24 SF3B1 muta­tions have a pos­i­ deemed benign or likely benign.29 Importantly, according to the
tive prog­nos­tic value on OS and risk of pro­gres­sion. This favor­ Amer­i­can College of Medical Genetics and Genomics guide­lines, a
able effect on prog­no­sis is inde­pen­dent of IPSS-R for very low var­i­ant of uncer­tain sig­nif­i­cance should not influ­ence clin­i­cal deci­
and low IPSS-R categories but is not retained for high- or very sion mak­ing. NGS may also iden­tify a germline var­i­ant (eg, biallelic
high-risk groups.24 Comutations medi­ate this effect. Mutations in muta­tion in CEBPA, RUNX1, or other genes often evidenced by a
epi­ge­netic reg­u­la­tors, includ­ing TET2, DNMT3A, and ASXL1, do VAF of around 50%), which may have imme­di­ate prac­ti­cal impli­
not appear to affect sur­vival of patients with an SF3B1 muta­tion, ca­tions for donor selec­tion if the patient is a trans­plant can­di­date,
whereas RUNX1 and EZH2 muta­tions have a sig­nif­i­cant asso­ci­a­ and influ­ence fam­ily mem­bers.30 Referral to a genetic coun­selor is
tion with increased risk of dis­ease pro­gres­sion and decreased rea­son­able when germline muta­tions are a con­sid­er­ation.
OS inde­pen­dent of IPSS-R.24,25
Conclusion
The pres­ence or absence of a TP53 muta­tion In sum­mary, we rec­om­mend the use of NGS to pro­vide cli­ni­cians
The adverse prog­nos­tic effect of TP53 muta­tions in MDS has with addi­tional infor­ma­tion for risk strat­i­fi­ca­tion for patients with
long been rec­og­nized. In patients with an iso­lated dele­tion in MDS. Conclusive rec­om­men­da­tions of how to inte­grate cur­rent
5q, 20% of patients har­bor a TP53 muta­tion that con­fers a higher risk prog­nos­ti­ca­tion sys­tems and molec­u­lar pro­files can­not be
risk to AML pro­gres­sion, worse OS, and early pro­gres­sion in lena­ made based on the var­i­abil­ity in data qual­ity and het­er­og­e­nous ef­
lidomide-treated patients.26 TP53 muta­tion sta­tus also appears fects of muta­tions on patients. Treatment deci­sions must be made
crit­i­cal to medi­ate the adverse effect of a com­plex kar­yo­type. on a case-by-case basis with ther­apy aligned with patient pref­er­
Patients with a com­plex kar­yo­type with­out a TP53 muta­tion ences. Increasingly, we are see­ing the dem­on­stra­tion of the asso­
have sig­nif­i­cantly bet­ter sur­vival than would be suggested from ci­a­tion between molec­u­lar pro­files and clin­i­cal fea­tures that can
prog­nos­ti­ca­tion based on karyotyping alone.27 In both cases, the pre­dict prog­no­sis inde­pen­dent of clin­i­cal scor­ing sys­tems. The
pres­ence or absence of a TP53 muta­tion could sub­stan­tially al­ devel­op­ment of val­i­dated prog­nos­tic mod­els, induc­ing molec­u­lar
ter clin­i­cal man­age­ment. Specifically, Prakash
we wouldSingh Shekhawat
highly con­
sider data, will bring great value to the field. Hopefully, we are not too

Routine molec­u­lar pro­fil­ing in MDS for risk  |  437


far away from this being a real­ity. The International Working Group 8. Tefferi A, Gangat N, Mudireddy M, et al. Mayo alli­ance prog­nos­tic model
for the Prognosis of MDS and the MDS Foundation are progressing for myelodysplastic syn­dromes: inte­gra­tion of genetic and clin­i­cal infor­ma­
tion. Mayo Clin Proc. 2018;93(10):1363-1374.
in the devel­op­ment of an inte­grated clin­i­cal-molec­u­lar data­base 9. Sallman DA, Komrokji R, Vaupel C, et al. Impact of TP53 muta­tion var­i­ant
that should sup­port the devel­op­ment of “IPSSR-Molecular.” allele fre­ quency on phe­ no­type and out­ comes in myelodysplastic syn­
dromes. Leukemia. 2016;30(3):666-673.
10. Hirsch CM, Nazha A, Kneen K, et al. Consequences of mutant TET2 on clon­
Recommendations ality and subclonal hier­ar­chy. Leukemia. 2018;32(8):1751-1761.
The rec­om­men­da­tions are labeled as either “strong” or “con­di­ 11. Pellagatti A, Roy S, Di Genua C, et al. Targeted resequencing anal­y­sis of
tional” according to the GRADE approach. 31 genes com­monly mutated in mye­loid dis­or­ders in serial sam­ples from
myelodysplastic syn­drome patients show­ing dis­ease pro­gres­sion. Leuke-
(1) For all­patients suspected of hav­ing or con­firmed to have mia. 2016;30(1):247-250.
MDS, the authors rec­om­mend molec­u­lar pro­fil­ing with NGS to 12. Bernard E, Nannya Y, Hasserjian RP, et al. Implications of TP53 alle­lic state
iden­tify rel­e­vant muta­tions (strong rec­om­men­da­tion based for genome sta­bil­ity, clin­i­cal pre­sen­ta­tion and out­comes in myelodysplastic
syn­dromes. Nat Med. 2020;26(10):1549-1556.
on mod­er­ate cer­tainty in the evi­dence of effects).
13. Montalban-Bravo G, Kanagal-Shamanna R, Benton CB, et al. Genomic con­

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/435/1852132/435richardson.pdf by guest on 13 December 2021


(2) For patientpdfs found to have canon­i­cal muta­tions in genes text and TP53 allele fre­quency define clin­i­cal out­comes in TP53-mutated
known to con­trib­ute to sur­vival out­comes (eg, SF3B1, ASXL1, myelodysplastic syn­dromes. Blood Adv. 2020;4(3):482-495.
SRSF2, ETV6, RUNX1, and TP53), the authors sug­gest cli­ni­ 14. Malcovati L, Gallì A, Travaglino E, et  al. Clinical sig­nif­i­cance of somatic
muta­tion in unex­plained blood cytopenia. Blood. 2017;129(25):3371-3378.
cians incor­po­rate the poten­tial addi­tional risk into existing
15. Galli A, Todisco G, Catamo E, et al. Relationship between clone met­rics and
prog­nos­tic scor­ing sys­tems (con­di­tional rec­om­men­da­tion clin­i­cal out­come in clonal cytopenia [published online 13 July 2021]. Blood.
based on low cer­tainty in the evi­dence of effects). 16. Bersanelli M, Travaglino E, Meggendorfer M, et al. Classification and per­
(3) For patients who are eli­gi­ble based on molec­u­lar pro­files, the au­ son­al­ized prog­nos­tic assess­ment on the basis of clin­ic ­ al and geno­mic fea­
thors sug­gest cli­ni­cians should offer enroll­ment in a clin­i­cal trial to tures in myelodysplastic syn­dromes. J Clin Oncol. 2021;39(11):1223-1233.
17. Bejar R, Stevenson KE, Caughey BA, et al. Validation of a prog­nos­tic model
patients (con­di­tional rec­om­men­da­tion based on low cer­tainty in
and the impact of muta­tions in patients with lower-risk myelodysplastic
the evi­dence of effects). syn­dromes. J Clin Oncol. 2012;30(27):3376-3382.
(4) For all­patients with MDS, the authors rec­om­mend assess­ 18. Forghieri F, Paolini A, Morselli M, et al. NPM1 muta­tions may reveal acute mye­
ing patient pref­er­ences to align treat­ment rec­om­men­da­ loid leu­ke­mia in cases oth­er­wise mor­pho­log­i­cally diag­nosed as myelodys­
plastic syn­dromes or myelodysplastic/mye­lo­pro­lif­er­a­tive neo­plasms. Leuk
tions with patients’ will­ing­ness to tol­er­ate risks for ben­e­fits
Lymphoma. 2015;56(11):3222-3226.
(strong rec­om­men­da­tion based on mod­er­ate cer­tainty in 19. Montalban-Bravo G, Kanagal-Shamanna R, Sasaki K, et al. NPM1 muta­tions
the evi­dence of effects). define a spe­cific sub­group of MDS and MDS/MPN patients with favor­able
out­comes with inten­sive che­mo­ther­apy. Blood Adv. 2019;3(6):922-933.
20. Bains A, Luthra R, Medeiros LJ, Zuo Z. FLT3 and NPM1 muta­tions in myelo­
Conflict-of-inter­est dis­clo­sure dysplastic syn­dromes: fre­quency and poten­tial value for predicting pro­
Dan­iel R. Richardson: no conflicts to disclose. gres­sion to acute mye­loid leu­ke­mia. Am J Clin Pathol. 2011;135(1):62-69.
21. Daver N, Strati P, Jabbour E, et  al. FLT3 muta­ tions in myelodysplas­
Amy E. DeZern has received con­sul­tancy fees from BMS, Abbvie, tic syn­ drome and chronic myelomonocytic leu­ ke­mia. Am J Hematol.
Novartis, and Taiho. 2013;88(1):56-59.
22. Xu F, Han R, Zhang J, et al. The role of FLT3-ITD muta­tion on de novo MDS in
Chi­nese pop­u­la­tion. Clin Lymphoma Myeloma Leuk. 2019;19(2):e107-e115.
Off-label drug use 23. Badar T, Patel KP, Thompson PA, et al. Detectable FLT3-ITD or RAS muta­tion
Dan­iel R. Richardson: nothing to disclose. at the time of trans­for­ma­tion from MDS to AML pre­dicts for very poor out­
Amy E. DeZern: nothing to disclose. comes. Leuk Res. 2015;39(12):1367-1374.
24. Malcovati L, Stevenson K, Papaemmanuil E, et al. SF3B1-mutant MDS as a
dis­tinct dis­ease sub­type: a pro­posal from the International Working Group
Correspondence for the Prognosis of MDS. Blood. 2020;136(2):157-170.
Amy E. DeZern, Department of Oncology, Sidney Kimmel Cancer 25. Malcovati L, Karimi M, Papaemmanuil E, et al. SF3B1 muta­tion identifies a
Center at Johns Hopkins, 1650 Orleans Street, CRBI Room 3M87 dis­tinct sub­set of myelodysplastic syn­drome with ring sideroblasts. Blood.
2015;126(2):233-241.
Baltimore, MD 21287-0013; e-mail: adezern1@jhmi​­.edu. 26. Jädersten M, Saft L, Smith A, et al. TP53 muta­tions in low-risk myelodys­
plastic syn­dromes with del(5q) pre­dict dis­ease pro­gres­sion. J Clin Oncol.
References 2011;29(15):1971-1979.
1. Papaemmanuil E, Gerstung M, Malcovati L, et al; Chronic Myeloid Disorders 27. Haase D, Stevenson KE, Neuberg D, et al; International Working Group for
Working Group of the International Cancer Genome Consortium. Clinical MDS Molecular Prognostic Committee. TP53 muta­tion sta­tus divi­des myel­
and bio­log­i­cal impli­ca­tions of driver muta­tions in myelodysplastic syn­ odysplastic syn­dromes with com­plex kar­yo­types into dis­tinct prog­nos­tic
dromes. Blood. 2013;122(22):3616-3627, quiz 3699. sub­groups. Leukemia. 2019;33(7):1747-1758.
2. Ogawa S. Genetics of MDS. Blood. 2019;133(10):1049-1059. 28. Sallman DA, DeZern AE, Garcia-Manero G, et al. Eprenetapopt (APR-246)
3. Haferlach T, Nagata Y, Grossmann V, et al. Landscape of genetic lesions in and azacitidine in TP53-mutant myelodysplastic syn­dromes. J Clin Oncol.
944 patients with myelodysplastic syn­dromes. Leukemia. 2014;28(2):241- 2021;39(14):1584-1594.
247. 29. Li MM, Datto M, Duncavage EJ, et al. Standards and guide­lines for the inter­
4. Bejar R. Myelodysplastic syn­dromes diag­no­sis: what is the role of molec­u­ pre­ta­tion and reporting of sequence var­i­ants in can­cer: a joint con­sen­sus
lar test­ing? Curr Hematol Malig Rep. 2015;10(3):282-291. rec­om­men­da­tion of the Association for Molecular Pathology, Amer­i­can
5. Nazha A, Sekeres MA, Gore SD, Zeidan AM. Molecular test­ing in myelodys­ Society of Clinical Oncology, and College of Amer­i­can Pathologists. J Mol
plastic syn­dromes for the prac­tic­ing oncol­o­gist: will the prog­ress ful­fill the Diagn. 2017;19(1):4-23.
prom­ise? Oncologist. 2015;20(9):1069-1076. 30. Kennedy AL, Shimamura A. Genetic pre­dis­po­si­tion to MDS: clin­i­cal fea­tures
6. Cazzola M, Della Porta MG, Malcovati L. The genetic basis of myelodyspla­ and clonal evo­lu­tion. Blood. 2019;133(10):1071-1085.
sia and its clin­i­cal rel­e­vance. Blood. 2013;122(25):4021-4034.
7. Bejar R, Stevenson K, Abdel-Wahab O, et al. Clinical effect of point muta­
tions in myelodysplastic syn­ dromes. N Engl J Med. 2011;364(26):2496- © 2021 by The Amer­i­can Society of Hematology
2506. DOI 10.1182/hema­tol­ogy.2021000320

438  |  Hematology 2021  |  ASH Education Program


MDS: BEYOND A ONE - SIZE - FITS -ALL APPROACH

Oral hypomethylating agents: beyond


convenience in MDS

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/439/1852126/439griffiths.pdf by guest on 13 December 2021


Elizabeth A. Griffiths
Roswell Park Comprehensive Cancer Center, Buffalo, NY

Oral hypomethylating agents (HMAs) represent a substantial potential boon for patients with myelodysplastic syndrome
(MDS) who have previously required between 5 and 7 visits per month to an infusion clinic to receive therapy. For patients
who respond to treatment, ongoing monthly maintenance visits represent a considerable burden to quality of life, and
for those who are early in therapy, these sequential visits may tax transportation and financial resources that would be
optimally distributed over the treatment cycle to facilitate transfusion support. The availability of oral HMAs may support
the optimal application of these agents by contributing to adherence and lessening the burden of therapy, potentially
encouraging patients to stay on longer-term treatment. Distinct pharmacokinetic profiles for the recently approved oral
HMAs (oral azacitidine and decitabine-cedazuridine) result in differential toxicity profiles and have prompted their clinical
trial development in lower- and higher-risk MDS, respectively.

LEARNING OBJECTIVES
• Describe the challenges of effective therapy with HMAs for patients with MDS and the barriers to this therapy
• Understand the PK profiles for HMAs given parenterally and contrast these with the PK profiles following oral
administration of unmodified oral azacitidine and decitabine/cedazuridine
• Recognize how the PK for oral HMAs may differ from an established parenteral regimen and anticipate the chal-
lenges associated with transitioning from one regimen to another

with parenteral azacitidine 75 mg/m2 given subcutane-


CLINICAL CASE 1 ously (SQ ) 7 days of a 28-day schedule. The initial treat-
A 78-year-old woman presented to our clinic after routine ment was characterized by the development of red blood
blood work demonstrated thrombocytopenia and neutro- cell (RBC) and platelet transfusion dependence in cycles
penia. The patient was otherwise in reasonable health for 1 and 2. She presented prior to cycle 3 of therapy, and the
her age with comorbid medically managed hypertension. CBC showed a WBC count of 1.0 × 109/µL with absolute
She was widowed but lived independently, with minimal neutrophil count 0.2 × 109/µL, an Hg of 8 g/µL, and plate-
support from her local adult children. A complete blood lets, 105 k/µL. She came to the clinic accompanied by
count (CBC) at the initial evaluation demonstrated a white her children. She stated that she was no longer willing to
blood cell (WBC) count of 1.4 × 109/µL, with an abso- continue therapy. She reported intolerable loss of quality
lute neutrophil count of 0.5 × 109/µL, hemoglobin (Hg) of life and independence due to the daily infusion clinic
of 11 g/µL, and platelet count of 25 k/µL. She was dem- visits in the first week of each cycle and the burden of
onstrated to be nutrient replete for B12, folate, iron, and transfusion dependence. She was adamant that she would
copper. A bone marrow (BM) aspiration and biopsy were take no further chemotherapy despite an early response
performed, showing refractory anemia with excess blast-2 to treatment that suggested she was likely to derive a sur-
(15% blasts); cytogenetics were normal. Next-generation vival benefit from continued treatment (early hematologic
sequencing (NGS) revealed mutations in IDH2 and SRSF2. improvement [HI] of the platelet lineage). She requested
A diagnosis of higher-risk myelodysplastic syndrome enrollment in home hospice care and passed away 1 month
(MDS) was rendered (International Prognostic Scoring after therapy discontinuation.Today, the availability of oral
System [IPSS]-Revised [R], 5.5 points [high risk]; IPSS 2.0 agents might have convinced this patient to continue
points [intermediate-2]), and the patient started therapy treatment, optimizing her survival and improving her
Prakash Singh Shekhawat
Oral hypomethylating agents for MDS | 439
Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/439/1852126/439griffiths.pdf by guest on 13 December 2021
Figure 1. Chemical structure of cytidine, azacitidine analogues, and the breakdown products of CDA.

qual­ity of life. As it stood, this patient accrued all­the tox­ic­ity bind it irre­vers­ibly, cre­at­ing an adduct that results in both DNA
from the use of this agent, includ­ing ini­tial loss of qual­ity of life, dam­age and the deple­tion of DNMT enzymes.2 With repeated
with­out real­iz­ing the lon­ger-term poten­tial ben­e­fits of trans­fu­ cycles of DNA rep­li­ca­tion, loss of the meth­yl­at­ion enzymes re-
sion inde­pen­dence (TI) and sur­vival, which she might have been sults in the pas­sive demethylation of DNA at mul­ti­ple loci. At low
expected to achieve given her early evi­dence of response. doses that do not cause over­whelm­ing cyto­tox­ic­ity, these drugs
induce tran­sient gene-spe­cific and global hypomethylation that
are hypoth­e­sized to medi­ate their response. Historically, activ­ity
Introduction has been pos­tu­lated to result from the reexpression of epi­ge­
Hypomethylating agents (HMAs) have been the main­ stay of net­i­cally silenced tumor sup­pres­sor genes, but more recently,
treat­ment for patients with higher-risk MDS since the ini­tial reg­u­ reexpression of endog­e­nous ret­ro­vi­ral ele­ments with enhanced
la­tory approval of monthly treat­ments with par­en­teral azacitidine immune rec­og­ni­tion of MDS pro­gen­i­tors has been hypoth­e­sized
and decitabine in the early 2000s.1 The shared active metab­ol­ite as another puta­tive mech­a­nism.3 Despite ongo­ing con­tro­versy
of both agents (~15% of the admin­is­tered azacitidine dose, con- over the defin­i­tive expla­na­tion for ther­a­peu­tic effi­cacy, opti­mal
verted by the action of ribo­nu­cle­o­tide reduc­tase) is decitabine patient out­comes from HMA ther­apy seem to require long-term
tri­phos­phate, an ana­logue of the nucle­o­side cyti­dine (Figure 1). treat­ment on a reg­u­lar and unin­ter­rupted sched­ule.4-7
Although 85% of par­en­ter­ally admin­is­tered azacitidine is incor­
po­rated into RNA, interrupting pro­tein syn­the­sis and induc­ing Dose, sched­ule, and treat­ment con­tin­u­a­tion are crit­i­cal
apo­pto­sis, the clin­ic
­ al impact of this incor­po­ra­tion on its activ­ to opti­mal HMA response
ity remains an area of active inves­ti­ga­tion.1 When decitabine tri­ When given on a monthly basis for repeated cycles, these agents
phos­phate is incor­po­rated into DNA whose par­ent strand bears improve cytopenias, lower BM blasts, improve qual­ity of life, de-
a methyl group in the con­text of a cyto­sine-phospho-gua­nine crease trans­for­ma­tion to acute mye­loid leu­ke­mia (AML), and im-
sequence, DNA methyltransferase enzymes (par­tic­u­larly DNMT-1) prove sur­vival for select patients.8-10 Despite years of study, no

440  |  Hematology 2021  |  ASH Education Program


effec­tive pre­treat­ment pre­dic­tors of response are clin­i­cally appli­ quate dose inten­sity dur­ing early cycles prob­a­bly com­pro­mises
ca­ble.11 About half of HMA-treated patients will respond, most response.30 Such treat­ment deci­sions are likely to have an adverse
with improve­ments in cytopenias (HI of the affected lin­e­age) impact on qual­ity of life and com­pro­mise sur­vival for those MDS
and a minor­ity (per­haps 15%) with com­plete response (CR), but patients, who will accrue all­the tox­ic­ity from HMA ther­apy and
responses can take sev­eral months to develop (National Com- none of the ben­e­fits (HI, CR, or sur­vival) of sustained ther­apy.24
prehensive Cancer Center guide­lines rec­om­mend a min­i­mum of The impor­tance of ade­quate sup­port and ther­apy per­sis­tence for
6 cycles of treat­ment before response assess­ment) and are gen­ patients with higher-risk MDS can­not, there­fore, be overstated.
er­ally of lim­ited dura­bil­ity.12,13 Critically, hema­to­log­i­cal improve­ Oral agents, which might limit the need for treat­ment-related
ment (HI) in any lin­e­age is asso­ci­ated with sur­vival ben­e­fit for infu­sion clinic vis­its and allow patients a sched­ule based solely on
patients who con­tinue ther­apy; with sustained treat­ment some trans­fu­sion needs, have the poten­tial to decrease rates of early
indi­vid­u­als will have improved blood counts across more than dis­con­tin­u­a­tion due to treat­ment bur­den and thereby result in
one lin­e­age, even converting to CR.7,13,14 Unfortunately, when pa- improved qual­ity of life, effi­cacy, and sur­vival for patients with
tients discontinue ther­apy, rapid pro­gres­sion and relapse with MDS. Furthermore, although most responses to HMAs have been

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/439/1852126/439griffiths.pdf by guest on 13 December 2021


poor sur­vival are likely.15-21 Good responses and improved sur­vival shown to occur within 4 to 6 cycles of treat­ment, con­tin­u­a­tion
are often accom­pa­nied by recov­ery of the plate­let count in the of ther­apy beyond the first HI results in fur­ther improve­ment
first 1 to 3 cycles of treat­ment, an early bio­marker for those who in response cat­e­gory for about half of those on treat­ment.7,18
are likely benefit­ing from treat­ment.22,23 Indeed, the best documented response seems to man­i­fest at
As for the patient in our first case, early cycles of HMA ther­apy least 2 to 3 cycles after the first documented HI response, with
are often char­ac­ter­ized by the wors­en­ing of cytopenias and the opti­mal responses in 1 study manifesting as late as cycle 12 for a
devel­op­ment of trans­fu­sion depen­dence. This has the poten­tial major­ity of patients.13,18
to cre­ate a sub­stan­tial bur­den for patients and fam­i­lies, espe­cially Given the ongo­ ing bur­
den of treat­ ment, con­ tin­
ued ther­
since each monthly cycle has his­tor­i­cally required 5 to 7 con­sec­ apy on sched­ule must be empha­sized to patients and should
u­tive daily doses of intra­ve­nous (IV; decitabine, azacitidine) or SQ not be underrecognized. Early on, trans­fu­sion depen­dence may
(azacitidine) treat­ment at a des­ig­nated infu­sion cen­ter. Given the increase patient will­ing­ness to come to the office for treat­ment,
early wors­en­ing of cytopenias, opti­mal sup­port­ive care requires but in the long term, these vis­its can become oner­ous. Ongo-
weekly or even twice-weekly vis­its dur­ing the sub­se­quent 3 weeks ing stud­ies are test­ing whether patients on an established par­
to assess trans­fu­sion needs as well as the pro­phy­lac­tic use of anti­ en­teral HMA reg­i­men can be switched to a lower-inten­sity oral
mi­cro­bi­als.24 The devel­op­ment of RBC and plate­let trans­fu­sion reg­i­men to max­i­mize adher­ence with­out com­pro­mis­ing effi­cacy
depen­dence and neutropenia early in the course of treat­ment can (eg, NCT04806906). Such changes to our ther­a­peu­tic approach
trig­ger cli­ni­cians to inap­pro­pri­ately reduce doses, delay, or even may also improve the real-world out­comes asso­ci­ated with HMA
discontinue ther­apy; deci­sions known to com­pro­mise effi­cacy.25,26 treat­ment when com­pared with those seen in clin­i­cal tri­als. If
Such deci­sions may explain the dif­fer­ence between the sur­vival pre­ma­ture dis­con­tin­u­a­tion in responding patients is driven by
ben­e­fit documented in con­trolled pop­u­la­tions of patients treated doc­tors and patients unwill­ing to con­tinue ther­apy, at least in
with HMAs in the con­text of clin­i­cal tri­als com­pared to out­comes part due to the bur­den of infu­sion clinic appoint­ments, I believe
in unse­lected pop­u­la­tions of patients, where improved sur­vival oral ther­a­pies may help us max­i­mize HMA ben­e­fit for patients.
rates have been less con­sis­tent.10,21,27,28
A set of recent ret­ro­spec­tive ana­ly­ses using the sur­veil­lance,
epi­de­mi­­ol­ogy, and end results Medi­care-linked data­base in a large
cohort of 644 patients with higher-risk MDS eval­u­ated between
2011 and 2015 sought to inves­ti­gate the way in which patients CLINICAL CASE 2
treated in a real-world set­ting rou­tinely receive ther­apy. This study A 69-year-old man with a med­ i­
cal his­
tory nota­ble for obe­
sity,
dem­on­strated that almost 30% of patients discontinued HMAs dia­be­tes, and bilat­eral knee oste­o­ar­thri­tis was referred to our
before com­ plet­ing 4 cycles—a major­ ity after com­ plet­ing only prac­tice after a pre­op­er­a­tive CBC performed for a planned knee
1 cycle of treat­ment.29 A more detailed anal­y­sis of this same cohort replace­ment sur­gery dem­on­strated throm­bo­cy­to­pe­nia. The CBC
reveals that an addi­tional 20% of patients had early dose delays of showed a nor­mal WBC count, plate­lets of 40 000/µL, and ane­mia
>90 days between cycles, which likely obvi­ates response.30 Over- with an Hb of 10 g; the mean cor­pus­cu­lar vol­ume was 106 fL. The
all, among the approx­i­ma­tely 50% of patients with higher-risk remain­der of the CBC and a com­pre­hen­sive pro­file were nor­mal,
MDS, those who did not receive their HMA on sched­ule for at least and nutri­tional workup dem­on­strated a suf­fi­ciency of vita­min B12,
4 cycles had sub­stan­tially higher rates of health care uti­li­za­tion folate, and iron; fer­ri­tin was 600 (min­i­mally ele­vated), and the
char­ac­ter­ized by increased emer­gency room vis­its, higher rates serum erythropoietin level was 580 IU (ele­vated). The periph­eral
of hos­pi­tal­i­za­tion, and more admis­sions to skilled nurs­ing facil­i­ties blood smear was nota­ble for pelgeroid neu­tro­phils, throm­bo­cy­
and hos­pice. Suboptimally treated patients in this cohort were to­pe­nia, and RBC anisocytosis. A BM aspi­rate and biopsy were
more likely to be older and unpartnered. hypercellular and ery­throid pre­dom­i­nant with 6% blasts and trilin-
Real-world stud­ies fur­ther dem­on­strate the rel­a­tively lim­ited eage dyspoeisis. Cytogenetics were 47 XY, + 8 in 21 meta­phases.
ini­ti­a­tion of HMA treat­ment for high-risk patients, with only 12% NGS was nota­ble for muta­tions in TET2 and ASXL1. A diag­no­sis
to 30% of pre­sum­ably eli­gi­ble patients receiv­ing ther­apy at all­, of inter­me­di­ate-risk MDS (IPSS score, inter­me­di­ate-1; IPSS-R,
depending upon the report.20,27,30 These data high­light the fact 5 points [high risk]) was ren­dered. After dis­cus­sion of prog­no­
that despite the avail­abil­ity of HMAs as pri­mary ther­apy for MDS sis and options, he elected to enroll in a phase 1b phar­ma­co­
for more than 15 years, many patients do not receive any ther­apy ki­
netic (PK) study of oral ­ decitabine-cedazuridine fixed-dose
at all­, and among those who do, early dis­con­tin­u­a­tion or inad­e­ tab­let 35 mg/100 mg. Initial cycles were char­ac­ter­ized by grade
Prakash Singh Shekhawat
Oral hypomethylating agents for MDS  |  441
Table 1. PK char­ac­ter­is­tics of the HMAs

Bioavailability of sin­gle oral Cmax in ng/mL (%


Agent dose (% of par­en­teral) T1/2 Tmax (range) coef­fi­cient of var­i­a­tion)
Azacitidine IV34,40 100% 4 h 0.5 h Similar to SQ
Azacitidine SQ34,40 89% 4 h 0.5 h (0.2-1.1) 750 (54%)
CC-486 40,52
11% 0.5 h 1 h (0.47-2) 145 (64%)
Decitabine IV35 100% 0.5 h 1 h 147 (49%)
Decitabine PO 41
3.9%-14.1% 0.36-0.93 h 0.5 h —
C-DEC43 60% (55-65) D1; 106% (98-114) D5 1.5 h 1 h (0.3-3.0) 145 (55%)

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/439/1852126/439griffiths.pdf by guest on 13 December 2021


4 throm­bo­cy­to­pe­nia and neutropenia and plate­let trans­fu­sion 35/100 mg), for patients with higher-risk MDS (inter­me­di­ate/high
depen­dence. After cycle 5 the patient had recov­ery of the plate­let risk by IPSS).43 In con­trast with another recently approved agent,
count. A BM biopsy after cycle 4 dem­on­strated a blast per­cent­age unmod­i­fied oral azacitidine (CC-486), which will be discussed
of 4% with ongo­ing dys­pla­sia. Referral for trans­plant was made, sub­se­quently, C-DEC has been shown in a ran­dom­ized phase 3
and a fully matched sib­ling donor was iden­ti­fied. The patient cur­ study to reli­ably reca­pit­u­late the blood drug lev­els over time
rently remains on pro­to­col cycle 12 of oral decitabine-cedazuridine, (cumu­la­tive area under the con­cen­tra­tion-time curve; AUC∞; Fig-
given 4 days of a 28-day cycle (the dose was reduced after cycle ure 2) resulting from daily treat­ment for 5 days with IV decit-
8 for neutropenia last­ing more than 2 weeks and BM show­ing abine, and the 2 can there­fore be treated inter­change­ably for
ongo­ing ther­a­peu­tic response). He has declined to pro­ceed with the pur­poses of clin­i­cal man­age­ment.44,45
trans­plant due to con­cerns about trans­plant-related mor­tal­ity. He As expected, based upon the ubiq­ui­tous expres­sion of CDA
enjoys an excel­lent qual­ity of life and remains trans­fu­sion inde­ in the liver and GI tract, there is sub­stan­tial first-pass metab­o­
pen­dent (TI) and nonneutropenic with a plate­ let count above lism of oral decitabine. Single-agent oral decitabine was shown
100 k/µL. These counts allow him to use non­ste­roi­dal agents for in a phase 1 study to result in low bio­avail­abil­ity with sub­stan­
man­age­ment of his knee pain with­out bleed­ing risk. tial var­i­a­tion across indi­vid­u­als when com­pared with IV decit-
abine.41 To over­come the var­i­able PKs resulting from dif­fer­en­tial
CDA lev­els within and across indi­vid­u­als, a novel inhib­i­tor of CDA
PKs of par­en­teral vs oral HMAs (CDAi) given the des­ig­na­tion E7727 (sub­se­quently cedazuridine)
Parenteral daily HMA treat­ment with azacitidine and decitabine was devel­oped and exten­sively tested in ani­mals.46 This agent
results in blood drug lev­els that peak between 15 and 30 min­ dem­on­strated excel­lent bio­avail­abil­ity and a wide safety mar­gin
utes after admin­ is­tra­
tion and an elim­ i­na­tion half-life between in non­hu­man pri­ma­tes.46,47 In a series of early-phase stud­ies, the
1 to 2 hours for azacitidine and 35 to 40 min­utes for decitabine com­bi­na­tion of cedazuridine with oral decitabine (devel­oped
(Table 1).31-35 The short half-life of par­en­ter­ally admin­is­tered HMAs ini­tially as ASTX 727 and sub­se­quently des­ig­nated decitabine-
is medi­ated largely by the ubiq­ui­tous expres­sion of the enzyme cedazuridine) was tested to develop a com­bi­na­tion pill designed
cyti­dine deam­i­nase (CDA), which is highly expressed in a vari­ety to reca­pit­u­late the PK and phar­ma­co­dy­namic (PD) admin­is­tra­tion
of human tis­sues, includ­ing the liver, the BM, and the gas­tro­in­ of IV decitabine dosed at 20 mg/m2 for 5 days.47 Serial blood col­
tes­ti­nal (GI) tract. CDA rap­idly degrades these agents into the lec­tion was performed, and changes in global meth­yl­at­ ion using
inac­tive metab­o­lites of uri­dine (Figure 1).32,36 the repet­i­tive DNA sequence long inter­spersed nuclear ele­ment
The early devel­op­ment of oral HMAs was char­ac­ter­ized by 1 (a sur­ro­gate for global DNA meth­yl­a­tion) was used as a tool for
the rec­og­ni­tion that when given alone oral admin­is­tra­tion results com­par­ing phar­ma­co­dy­namic (PD) effi­cacy.48 Given the sub­stan­
in mark­ edly lower peak plasma con­ cen­ tra­
tions with a wide tial intra- and inter­in­di­vid­ual var­i­abil­ity in CDA lev­els, the devel­
range of bio­avail­abil­ity when com­pared with IV or SQ dos­ing op­ment pro­gram for this agent used each patient as their own
strat­e­gies.33,37-41 Table 1 pro­vi­des a com­par­i­son of half-life, sin­gle- con­trol and com­pared the PK/PD for IV decitabine against the
dose bio­avail­abil­ity (based upon area under the con­cen­tra­tion PK/PD for the oral com­bi­na­tion.47-49 Early stud­ies used PK/PD
time curve), time to max­im ­ al blood drug level (Tmax), and max­ read­outs to titrate the oral doses of E7727 and decitabine in
i­mal blood con­cen­tra­tion achieved after the admin­is­tered dose order to cre­ate a near iden­tity with IV decitabine admin­is­tra­tion
(Cmax) for each drug depending upon the route of admin­is­tra­ (Figure 2a). Ultimately, the 2 were com­bined into a fixed-dose
tion. Differences in bio­avail­abil­ity are medi­ated by wide inter- com­bi­na­tion tab­let containing 35 mg of decitabine and 100 mg
and intraindividual ranges of CDA expres­sion, with higher lev­els of cedazuridine.43-45
in men, in those with spe­cific sin­gle-nucle­o­tide poly­mor­phisms, The phase 3 study of C-DEC, upon which approval was based,
and in the con­text of inflam­ma­tory driv­ers.32,36,42 ran­dom­ized and treated 133 patients in a cross­over design to
receive either sequence A: C-DEC orally for 5 out of 28 days
Oral decitabine/cedazuridine in cycle 1 followed by IV decitabine 20 mg/m2 × 5 out of 28 days in
Based upon the dem­on­stra­tion of phar­ma­co­log­i­cal equiv­a­ cycle 2 and sub­se­quently C-DEC for cycles 3 onward or sequence
lence to IV decitabine, in July 2020 the US reg­u­la­tory author­i­ B: IV decitabine 20 mg/m2   ×   5 out of 28 days in cycle 1 followed
ties approved the first oral HMA, decitaine-cedazuridine (C-DEC; by C-DEC orally daily for 5/28 days in cycle 2 and sub­se­quently

442  |  Hematology 2021  |  ASH Education Program


Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/439/1852126/439griffiths.pdf by guest on 13 December 2021
Figure 2. Concentration time curves for (a) IV decitabine vs oral C-DEC and (b) SQ azacitidine vs CC-486.40,46 Figure 2a was re-
produced from Future Oncol. 2021;17(16):2077-2087 and was modified only by renumbering for the purpose of this article. Figure
2b was reproduced from Leukemia 2016;30(4):889-896 and was also modified only by renumbering. Both works are licensed under
the Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License. http:​/​/creativecommons​.org​/licenses​
/by​-nc​-nd​/4​.0​/

C-DEC for cycles 3 onward; cycles were to be repeated every tee. Enrolled patients were of median age 71 years (range, 44-88),
28 days (Figure 3). The pri­mary end point for this phase 3 study was 65% were male, 12% had chronic myelomonocytic leu­ke­mia, the
PK for cumu­la­tive AUC equiv­a­lence between IV and oral dos­ing of remain­der had MDS, and 42% had >5% mar­row blasts. Transfu-
decitabine. Eligible patients had MDS by French-Amer­i­can-­­Brit­ish sion depen­dence for either RBCs or plate­lets was pres­ent in 43%.
clas­si­fi­ca­tion, includ­ing chronic myelomonocytic leu­ke­mia, or After a median fol­low-up of 12.6 months, com­plete remis­sion was
MDS deemed inter­me­di­ate 1, 2, or high risk by the IPSS. The clin­ reported in 28 (21%) and HI in 30 (23%) patients for an over­all
i­cal response to ther­apy was assessed according to ­International response rate of 43%. An addi­tional 23 patients dem­on­strated
Working Group 2006 cri­te­ria by an inde­pen­dent review com­mit­ mar­row CR with­out HI, although blast clear­ance with­out blood

Figure 3. Summary comparison of the 2 completed phase 3 studies of C-DEC and CC-486 in MDS. Data abstracted from ­Garcia-Manero
et al40,49,55 and Savona et al.45 AZA, azacitidine; DAC, decitabine.
Prakash Singh Shekhawat
Oral hypomethylating agents for MDS  |  443
count improve­ ment is an end point of unclear clin­ ic
­al sig­
nif­i­ As shown pre­vi­ously for sin­gle-agent decitabine, the exten­
cance. Adverse events (AEs) asso­ci­ated with C-DEC were con­ sive expres­sion of CDA results in low bio­avail­abil­ity of unmod­i­
sis­tent with the famil­iar toxicities from early cycles of par­en­teral fied oral azacitidine. The pilot study of CC-486 ini­tially dosed it
HMA ther­apy, nota­bly cytopenias (≥grade 3: neutropenia, 52%; daily for 7 days in an attempt to reca­pit­u­late the stan­dard par­
throm­bo­cy­to­pe­nia, 50%; ane­mia, 40%; leu­ko­pe­nia, 21%) and en­teral dos­ing sched­ule. In this study the max­i­mum tol­er­ated
infec­tions (febrile neutropenia, 26%; pneu­mo­nia, 12%; sep­sis, 7%); dose was iden­ti­fied to be 480 mg, with a mean oral bio­avail­
in con­trast to CC-486, GI toxicities were not prominent. abil­ity between 6% and 20%. Relatively poor oral bio­avail­abil­ity
While phase 3 data have been reported only in abstract form, could not be over­come due to dose-lim­it­ing grades 3 and 4 GI
they largely reca­pit­u­late the recently published results from a tox­ic­ity (diar­rhea [12.2%], nau­sea [7.3%], vomiting [7.3%]) at the
phase 2 mul­ti­cen­ter ran­dom­ized cross­over study of sim­i­lar design highest doses; other grade 3/4 AEs included febrile neutropenia
that enrolled and treated 80 patients. In this phase 2 study, (19.5%) and fatigue (9.8%).53 With this reg­i­men, hypomethylation
responses (CR + HI) were observed in 44% of patients, with a was low­est at day 15 and recov­ered to base­line by the end of
major­ity of first responses seen by cycle 3 and best responses by the cycle.53,54 While some responses (mostly HI) were observed

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/439/1852126/439griffiths.pdf by guest on 13 December 2021


cycles 5 or beyond. Among base­line trans­fu­sion-depen­dent (TD) in this early study, decreases in meth­yl­at­ion across a range of
patients, about 50% became TI. Overall responses and tox­ic­ity tested loci (the PD bio­marker for activ­ity) were less robust with
from C-DEC appear com­men­su­rate with those from par­en­ter­ally CC-486 com­pared with the par­en­teral azacitidine for­mu­la­tion.
admin­is­tered HMAs, and this is not sur­pris­ing given the iden­ti­cal Given the prevailing hypoth­e­sis that the opti­mal hypometh-
blood drug lev­els (AUC) achieved with this drug. Many patients, ylating activ­ity of HMAs requires incor­po­ra­tion dur­ing S phase as
like our patient in Case 2, will have increased trans­fu­sion needs well as the rel­a­tively slow pro­lif­er­a­tive rate of MDS pro­gen­i­tors,
dur­ing early cycles of ther­apy. The same aggres­ sive sup­port- sub­se­quent drug devel­op­ment focused on extending the dos­ing
­ive care, includ­ing pro­phy­lac­tic anti­mi­cro­bi­als (a mold-active sched­ule to 14 or 21 days to allow opti­mal drug incor­po­ra­tion.38
anti­fun­ gal, an oral agent that cov­ ers gram neg­ a­
tives, and an Lower peak blood drug lev­els, with the sustained low-level expo­
anti-herpes simplex virus agent) for those with prolonged neu- sure asso­ci­ated with this dos­ing strat­egy, would be expected to
tropenia and reg­u­lar eval­u­a­tion of blood work for deter­mi­na­tion improve response rates. Indeed, exten­sion of the dos­ing sched­ule
of trans­fu­sion needs (man­dated weekly dur­ing cycles 1-3 by the in this man­ner in a PK/PD study dem­on­strated effec­tive induc­
clin­i­cal trial and gen­er­ally performed at least 1-2 times per week tion of hypomethylation using doses of 200 mg twice daily for 14
in my own prac­tice as a stan­dard of care), par­tic­u­larly dur­ing days or 300 mg daily for 14 or 21 days that were low­est at day 22
early ther­apy cycles, is crit­i­cal to opti­mize responses from this of treat­ment; hypomethylation of selected loci persisted at day
agent, as for all­HMAs.4,24,25 28.33 This dose sched­ule was tol­er­a­ble, and although a major­ity of
Oral C-DEC is cur­rently the only oral HMA approved for MDS. patients expe­ri­enced GI (84%) and hema­to­log­ic ­ al (81%) toxicities,
This agent was approved only for higher-risk MDS dis­ease cate- these were not dose lim­it­ing.38 The oral admin­is­tra­tion of CC-486
gories, but alter­nate dos­ing strat­e­gies with this agent are cur­ results in rapid absorp­tion from the GI tract, with food con­sump­
rently under inves­ti­ga­tion to deter­mine the poten­tial ben­e­fit tion and gas­tric pH hav­ing lim­ited impacts on blood drug lev­els.37
for those with lower-risk dis­ ease (NCT03502668). Preliminary Subset ana­ly­ses of early CC-486 stud­ies have suggested that low-
reports of effi­cacy in higher-risk MDS from the phase 2 and 3 er-risk patients, par­tic­u­larly those with throm­bo­cy­to­pe­nia, might
tri­als dem­on­strate clin­i­cal activ­ity and response that are at least derive par­tic­u­lar ben­e­fit from such a low-dose oral reg­i­men.39
com­pa­ra­ble with (and pos­si­bly supe­rior to) the response to IV Recently, a phase 3 study of CC-486 in TD lower-risk MDS
decitabine. Although cur­rent guide­lines favor SQ azacitidine as patients (by IPSS cri­te­ria) reported results on 216 patients ran­
the first-choice HMA for patients with MDS given the sur­vival dom­ized 1:1 between pla­cebo and CC-486 dosed at 300 mg daily
ben­e­fit reported for this agent, decitabine and C-DEC are both for 21 days of a 28-day sched­ule (Figure 3).55 Eligibility required
con­sid­ered accept­able alter­na­tives.10,50 Despite this caveat, sub­ an Eastern Cooperative Oncology Group per­for­mance sta­tus of
se­quent reports from clin­ic ­ al tri­als of azacitidine and decitabine at least 2, throm­bo­cy­to­pe­nia (plate­let count ≤75   ×   109/L), and
sug­gest that sur­vival dif­fer­ences in the ini­tial clin­i­cal tri­als may RBC trans­fu­sion depen­dence of at least 2 units per month for at
stem from issues of trial design rather than true dif­fer­ences in least 2 months (International Working Group 2006 cri­te­ria).12 The
effi­cacy, and a major­ity of prac­tic­ing MDS experts would con­ pri­mary end point for this study was RBC TI; the use of exog­e­
sider these agents largely inter­change­able.25 nous growth fac­tors was not per­mit­ted.
Enrolled and treated patients in this study were of median
Unmodified oral azacitidine age 74 years (range, 30-89), with IPSS-defined inter­me­di­ate-1 risk
In Sep­tem­ber 2020 unmod­i­fied oral azacitidine (sub­se­quently dis­ease (by IPSS-R, 28% had high or very high-risk MDS). They
des­ig­nated CC-486) at a dose of 300 mg daily for 14 out of 28 were heavily RBC TD (requir­ing about 3 units/month) and base­
days (not 21 days, as tested in the MDS study) per month was line throm­bo­cy­to­pe­nic (median 25   ×   109/L; ~30% plate­let TD; 40%
approved by the US Food and Drug Administration as main­te­ with base­line plate­let count <20   ×   109/L). The study met its pri­mary
nance ther­apy for inter­me­di­ate- and poor-cyto­ge­netic-risk AML end point of RBC TI in 30% of the CC-486 patients vs 11% of pla­
patients in com­plete remis­sion fol­low­ing an inten­sive induc­tion cebo-treated patients; HI of the plate­let count was also higher
reg­i­men; this agent is not cur­rently approved for patients with for CC-486-treated patients—24% vs 4.6%. Responses took on
MDS.51,52 Although this agent shares its name with par­en­ter­ally aver­age 2.4 months, and decreased trans­fu­sion needs for both
admin­is­tered azacitidine, the dis­tinct PK pro­files resulting from RBCs and plate­lets were sustained with CC-486 treat­ment (for a
oral admin­is­tra­tion of this agent ren­der it a dif­fer­ent drug (Figure median of 10 months for RBCs and 12 months for plate­lets); bilin-
2b). Substitution of CC-486 for IV or SQ azacitidine is not accept­ eage improve­ments (in both Hb and plate­let counts) were seen
able for patients with MDS. in about a quar­ter of the CC-486-treated patients. With a median

444  |  Hematology 2021  |  ASH Education Program


fol­low-up of about 13 months, an under­pow­ered over­all sur­vival remained TI and nonneutropenic. Due to con­cern about the risk
esti­mate was the same in both arms, although the rate of AML pro­ from COVID-19, he requested tran­si­tion to an oral reg­i­men, hav­
gres­sion was lower in the CC-486 arm. Toxicities were as expected ing learned from a recent patient edu­ca­tion event about the
with an HMA-based ther­apy and were worst in the first cycles of approval of C-DEC. His phy­ si­
cian agreed to the switch, and
treat­ment. Most patients reported low-grade GI tox­ic­ity asso­ci­ 3 weeks into cycle 1, the patient presented with short­ness of
ated with tak­ing CC-486. Grades 3 and 4 cytopenias (neutropenia, breath and fatigue. A CBC dem­on­strated an Hb of 5.7 g, an ANC
47% vs 12%; throm­bo­cy­to­pe­nia, 29% vs 16%) and infec­tions (febrile of 0.6   ×   109/µL, and plate­lets of 150   ×   109/µL. The patient and his
neutropenia, 28% vs 10%) were more com­mon in the CC-486- phy­si­cian became concerned about the pos­si­bil­ity of dis­ease
treated patients vs those who got pla­cebo. A higher rate of early pro­gres­sion, and an urgent BM biopsy was performed.
death was reported in the CC-486 arm (16 vs 6 patients), pre­dom­
i­nantly asso­ci­ated with bac­te­rial infec­tions as well as neutropenia.
The phase 3 data reviewed above sug­gest sub­stan­tial activ­ Switching from par­en­teral to oral ther­apy
ity in TD lower-risk MDS patients, likely supporting an expan­sion Recent short­ages of par­en­teral azacitidine and con­cerns about

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/439/1852126/439griffiths.pdf by guest on 13 December 2021


of the labeled indi­ca­tion in the near future.55 High rates of early the risk of COVID-19 infec­tion posed to patients with can­cer by
infec­tious death in this lower-risk pop­u­la­tion should at least war­ expo­sure to infu­sion clin­ics have resulted in rapid sub­sti­tu­tion of
rant the rou­tine use of pro­phy­lac­tic anti-infec­tives in order to C-DEC for par­en­teral HMAs.59,60 It is impor­tant to rec­og­nize that
mit­i­gate risk and may give the Food and Drug Administration C-DEC results in iden­ti­cal PKs with IV decitabine. However, and as
pause in the deci­sion to approve this agent.24 Since CC-486 has empha­sized in the Introduction, the PKs of decitabine and azaciti-
a mark­edly dis­tinct PK/PD pro­file from par­en­teral azacitidine, dine are not iden­ti­cal. For patients who have been maintained on
the two agents are not inter­change­able (Figure 2b); addi­tional dose-reduced azacitidine, the sub­sti­tu­tion of oral C-DEC rep­re­
stud­ies are needed to deter­mine whether CC-486 is appro­pri­ sents a sub­stan­tial dose increase in terms of both peak lev­els and
ate for indi­vid­u­als with higher-risk MDS. Presently, CC-486 use in total AUC expo­sure to HMA (Table 1). C-DEC is avail­­able only as a
patients with MDS is lim­ited to the con­text of clin­i­cal tri­als. fixed-dose tab­let; thus, dose reduc­tions require either a decrease
in the num­ber of days of ther­apy or dos­ing on an interrupted
Other oral HMAs in devel­op­ment sched­ule. The pack­age insert for C-DEC sug­gests dose reduc­tions
Other oral HMAs are in clin­i­cal devel­op­ment for patients with from 5 to 4 days, sub­se­quently to 3 days, and ulti­mately dos­ing
MDS. Among these are NTX-301 (NCT04167917), a novel orally bio­ every other day on days 1, 3, and 5. Due to the action of cedazuri-
avail­able DNMT1 inhib­i­tor, and ASTX-030, a com­bi­na­tion of azacit- dine, which accu­mu­lates dur­ing the 5-day dos­ing period, the AUC
idine with oral cedazuridine (ASTX-030; NCT04256317). The lat­ter for decitabine when C-DEC is dosed daily is sub­stan­tially higher
is being devel­oped using a com­bined phase 1 to 3 clin­i­cal trial than when it is dosed every other day. Therefore, dose reduc­tions
based upon the PK equiv­a­lence model pioneered for C-DEC (Fig- to a sched­ule of every other day result in sub­stan­tially lower decit-
ure 3). It is likely that in the near future we will have approval of at abine expo­sures. When transitioning patients from an established
least 3 oral HMAs for patients with MDS. Moreover, sev­eral stud­ies par­en­teral HMA reg­i­men, par­tic­u­larly when dose reduc­tions have
are either planned or under­way to com­bine oral HMAs with other been made, it is impor­tant to rec­og­nize these dif­fer­ences in drug
prom­is­ing MDS drugs, includ­ing pevonidostat, venetoclax, and sa- deliv­ery and to either plan for up-front dose reduc­tion to mit­i­gate
batolimab, among oth­ers (NCT04655755, NCT04878432). Histori- cytopenias or to reinstitute appro­pri­ate mon­i­tor­ing for tox­ic­ity
cally, com­bi­na­to­rial stud­ies have failed to dem­on­strate improve­ dur­ing the ini­tial treat­ment period. The patient described in case
ments over the sin­gle agent.2,56 The hypoth­e­ses for com­bi­na­tion 3 under­went a BM biopsy to exclude dis­ease pro­gres­sion, which
fail­ures include pos­tu­lated inad­e­quate dose inten­sity for the HMA dem­ on­
strated only hypocellularity. After a 2-week delay from
and antag­o­nism between agents due to con­ve­nience-dic­tated his sched­uled cycle of treat­ment, blood counts fully recov­ered.
overlapping sched­ules, among other rea­sons.2,56 The sub­sti­tu­tion C-DEC ther­apy was resumed, dose reduced to 3 days per cycle
of an oral HMA in such com­bi­na­tions might facil­i­tate improved this time, and blood counts were checked weekly dur­ing the next
dose/sched­ule com­bi­na­tions that could the­o­ret­i­cally enhance 2 cycles of ther­apy. No fur­ther trans­fu­sions were required, and the
effi­cacy and poten­tially decrease treat­ment mod­i­fi­ca­tions.2,56-58 patient remained nonneutropenic. A fail­ure to rec­og­nize the dif­
fer­ence between SQ azacitidine and oral C-DEC in this case could
have resulted in sig­nif­i­cant risk to this patient. Failure to plan for,
or to pre­emp­tively adjust dos­ing in antic­i­pa­tion of these dif­fer­
ences, might prompt both patient and phy­si­cian reluc­tance to
CLINICAL CASE 3 con­tinue with oral ther­apy and thereby con­trib­ute to a loss of dis­
A 78-year-old gen­tle­man with a his­tory of high-risk MDS (base­ ease response or frank relapse.
line hypercellular dys­plas­tic mar­row with blasts of 15%, nor­mal
kar­yo­type in 20 meta­phases, NGS with 2 dis­tinct TET2 muta­ Conclusions
tions, base­line trilineage cytopenias) has been maintained on SQ The advent of orally bio­avail­able HMAs rep­re­sents a sub­stan­
azacitidine 50 mg/m2 daily for 7 days every 28 days. The patient tial poten­tial ben­e­fit for patients with MDS. While both oral
achieved com­plete remis­sion after 6 cycles of ther­apy dosed decitabine (C-DEC) and oral azacitidine (CC-486) have received
at 75 mg/m2 daily for 7 days every 28 days and has been non- reg­u­
la­
tory approval, these two agents are dis­ tinct, and only
TD since cycle 5. Due to the devel­op­ment of neutropenia after C-DEC is cur­ rently approved for patients with MDS. C-DEC
cycle 9, a BM biopsy was repeated that showed ongo­ing CR fixed-dose tab­lets given for 5 days pro­vide iden­ti­cal PK expo­
with a hypocellular mar­row. The azacitidine dose was reduced sure to IV decitabine for 5 days, while CC-486 is dosed daily for
to 50 mg/m2 for 7 days of a 28-day cycle, and the patient 14 or 21 days and results in lower peak blood-drug lev­els than SQ
Prakash Singh Shekhawat
Oral hypomethylating agents for MDS  |  445
or IV azacitidine but a more prolonged expo­sure win­dow. The dif­ 4. Santini V, Prebet T, Fenaux P, et  al. Minimizing risk of hypomethylating
fer­ences in PK and PD pro­files for these two agents ren­der them agent fail­ure in patients with higher-risk MDS and prac­ti­cal man­age­ment
rec­om­men­da­tions. Leuk Res. 2014;38(12):1381-1391.
quite dis­tinct in terms of both poten­tial tox­ic­ity and mech­a­nisms 5. Voso MT, Breccia M, Lunghi M, et  al. Rapid loss of response after with­
of action. As a treating cli­ni­cian, I sub­sti­tute C-DEC directly 1:1 for drawal of treat­ment with azacitidine: a case series in patients with higher-
IV decitabine at a dose of 20 mg/m2 for 5 days. By con­trast, CC- risk myelodysplastic syn­dromes or chronic myelomonocytic leu­ke­mia. Eur
486 can­not be con­sid­ered equiv­a­lent to IV or SQ azacitidine, and J Haematol. 2013;90(4):345-348.
6. Tendas A, Lissia MF, Piccioni D, et  al. Obstacles to adher­ence to azaciti-
in the con­text of MDS, its effi­cacy must be tested pro­spec­tively.
dine admin­is­tra­tion sched­ule in out­pa­tient myelodysplastic syn­drome and
Orally bio­avail­able HMAs have the poten­tial to prove more related dis­or­ders. Support Care Cancer. 2015;23(2):303-305.
con­ve­nient for MDS patients and fam­i­lies, spar­ing them repeated 7. Silverman LR, Fenaux P, Mufti GJ, et  al. Continued azacitidine ther­ apy
vis­its to infu­sion clin­ics for the sole pur­pose of drug deliv­ery, but beyond time of first response improves qual­ity of response in patients with
these agents result in sub­stan­tial cytopenias that require rou­tine higher-risk myelodysplastic syn­dromes. Cancer. 2011;117(12):2697-2702.
8. Steensma DP, Baer MR, Slack JL, et  al. Multicenter study of decitabine
assess­ment of trans­fu­sion needs and con­sid­er­ation for pro­phy­ admin­is­tered daily for 5 days every 4 weeks to adults with myelodysplastic
lac­tic anti­mi­cro­bi­als. Most patients treated with these agents in syn­dromes: the Alternative Dosing for Outpatient Treatment (ADOPT) trial.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/439/1852126/439griffiths.pdf by guest on 13 December 2021


clin­i­cal trial devel­oped neutropenia, which was espe­cially prom- J Clin Oncol. 2009;27(23):3842-3848.
inent dur­ing early cycles of ther­apy, and neutropenic fever was 9. Kantarjian H, Issa JP, Rosenfeld CS, et al. Decitabine improves patient out­
comes in myelodysplastic syn­dromes: results of a phase III ran­dom­ized
among the most fre­quently reported AEs. As with par­en­teral
study. Cancer. 2006;106(8):1794-1803.
HMA ther­apy, opti­mal response to oral HMAs will require sus- 10. Fenaux P, Mufti GJ, Hellstrom-Lindberg E, et al; International Vidaza High-
tained on-sched­ule treat­ment cycles and opti­mized sup­port­ive Risk MDS Survival Study Group. Efficacy of azacitidine com­ pared with
care. When we con­sider switching to an oral reg­i­men in an effort that of con­ven­tional care reg­i­mens in the treat­ment of higher-risk myel-
to opti­mize qual­ity of life for a patient, par­tic­ul­ar care should odysplastic syn­dromes: a randomised, open-label, phase III study. Lancet
Oncol. 2009;10(3):223-232.
be taken to rec­og­nize the dif­fer­ent PK pro­files for the new oral 11. Santini V, Ossenkoppele GJ. Hypomethylating agents in the treat­ment of
reg­i­men and to antic­ip ­ ate the poten­tial toxicities resulting from acute mye­loid leu­ke­mia: a guide to opti­mal use. Crit Rev Oncol Hematol.
this change. 2019;140(August):1-7.
12. Cheson BD, Greenberg PL, Bennett JM, et al. Clinical appli­ca­tion and pro­
Acknowledgments  posal for mod­i­fi­ca­tion of the International Working Group (IWG) response
cri­te­ria in myelodysplasia. Blood. 2006;108(2):419-425.
Elizabeth A. Griffith received research sup­port from the Rappa-
13. Silverman LR, Fenaux P, Mufti GJ, et al. The effects of con­tin­ued azacitidine
port Family Foundation and the Charles D and Mary A Bauer Foun- treat­ment cycles on response in higher risk patients with myelodysplastic
dation. This work was supported by Roswell Park Comprehensive syn­dromes: an update. Ecancermedicalscience. 2008;2(12 August):118.
Cancer Center and National Cancer Institute grant P30CA016056. 14. Gore SD, Fenaux P, Santini V, et al. A mul­ti­var­i­ate anal­y­sis of the rela­tion­
ship between response and sur­vival among patients with higher-risk myel-
The author would also like to acknowl­ edge David Eifrig,
odysplastic syn­ dromes treated within azacitidine or con­ ven­tional care
Michael Nemeth, Valeria Santini, and Sophia Balderman for edi­ reg­i­mens in the ran­dom­ized AZA-001 trial. Haematologica. 2013;98(7):
to­rial review of the arti­cle. 1067-1072.
15. Prébet T, Gore SD, Esterni B, et al. Outcome of high-risk myelodysplastic
Conflict-of-inter­est dis­clo­sure syn­drome after azacitidine treat­ment fail­ure. J Clin Oncol. 2011;29(24):3322-
3327.
Elizabeth A. Griffiths: research funding: Apellis Pharmaceu-
16. Jabbour E, Garcia-Manero G, Batty N, et  al. Outcome of patients with
ticals, Alexion Pharmaceuticals, Astex Pharmaceuticals, Cel- myelodysplastic syn­ drome after fail­ ure of decitabine ther­ apy. Cancer.
gene/Bristol Myers-Squibb, Celldex Therapeutics, Genentech; 2010;116(16):3830-3834.
hon­o­raria: Abbvie, Astex Pharmaceuticals, Bos­ton Biomedical, 17. Jabbour EJ, Garcia-Manero G, Strati P, et al. Outcome of patients with low-
Celgene/Bristol Myers-Squibb, Novartis Oncology, Takeda On- risk and inter­ me­ di­
ate-1-risk myelodysplastic syn­ drome after hypometh-
ylating agent fail­ ure: a report on behalf of the MDS Clinical Research
cology, Taiho Oncology. Consortium. Cancer. 2015;121(6):876-882.
18. Cabrero M, Jabbour E, Ravandi F, et al. Discontinuation of hypomethylating
Off-label drug use agent ther­apy in patients with myelodysplastic syn­dromes or acute mye­
Elizabeth A. Griffiths: This review discusses the use of CC-486/ log­e­nous leu­ke­mia in com­plete remis­sion or par­tial response: ret­ro­spec­tive
anal­y­sis of sur­vival after long-term fol­low-up. Leuk Res. 2015;39(5):520-
unmodified oral azacitidine for MDS, an indication that is not FDA
524.
approved. 19. Dinmohamed AG, van Norden Y, van de Loosdrecht AA, Jongen-Lavrencic
M. Effectiveness of azacitidine in higher-risk myelodysplastic syn­dromes.
Correspondence Leukemia. 2016;30(8):1795-1796.
Elizabeth A. Griffiths, Roswell Park Comprehensive Cancer 20. Bernal T, Martínez-Camblor P, Sánchez-García J, et al; Span­ish Group on
Myelodysplastic Syndromes; PETHEMA Foundation; Span­ ish Society of
Center, Elm and Carlton St, Buffalo, NY 14263; e-mail: elizabeth​
Hematology. Effectiveness of azacitidine in unse­ lected high-risk myel-
­.griffiths@roswellpark​­.org. odysplastic syn­ dromes: results from the Span­ ish reg­is­
try. Leukemia.
2015;29(9):1875-1881.
References 21. Zeidan AM, Stahl M, Hu X, et al. Long-term sur­vival of older patients with
1. Stomper J, Rotondo JC, Greve G, Lübbert M. Hypomethylating agents MDS treated with HMA ther­apy with­out sub­se­quent stem cell trans­plan­ta­
(HMA) for the treat­ment of acute mye­loid leu­ke­mia and myelodysplastic tion. Blood. 2018;131(7):818-821.
syn­dromes: mech­a­nisms of resis­tance and novel HMA-based ther­a­pies. 22. Jung HA, Maeng CH, Kim M, Kim S, Jung CW, Jang JH. Platelet response
Leukemia. 2021;35(7):1873-1889. dur­ing the sec­ond cycle of decitabine treat­ment pre­dicts response and
2. Fandy TE, Herman JG, Kerns P, et  al. Early epi­ge­netic changes and DNA sur­vival for myelodysplastic syn­drome patients. Oncotarget. 2015;6(18):
dam­age do not pre­dict clin­i­cal response in an overlapping sched­ule of ­16653-16662.
5-azacytidine and entinostat in patients with mye­loid malig­nan­cies. Blood. 23. Itzykson R, Thépot S, Quesnel B, et al; Groupe Francophone des Myelodys-
2009;114(13):2764-2773. plasies. Prognostic fac­tors for response and over­all sur­vival in 282 patients
3. Jones PA, Ohtani H, Chakravarthy A, De Carvalho DD. Epigenetic ther­apy in with higher-risk myelodysplastic syn­ dromes treated with azacitidine.
immune-oncol­ogy. Nat Rev Cancer. 2019;19(3):151-161. Blood. 2011;117(2):403-411.

446  |  Hematology 2021  |  ASH Education Program


24. Santini V, Fenaux P, Mufti GJ, et al. Management and sup­port­ive care mea­ 44. Garcia-Manero G, McCloskey J, Griffiths EA, et al. Pharmacokinetic expo­
sures for adverse events in patients with myelodysplastic syn­ dromes sure equiv­a­lence and pre­lim­i­nary effi­cacy and safety from a ran­dom­ized
treated with azacitidine. Eur J Haematol. 2010;85(2):130-138. cross over phase 3 study (ASCERTAIN study) of an oral hypomethylating
25. Zeidan AM, Hu X, Zhu W, et al. Does pro­vider expe­ri­ence treating patients agent ASTX727 (cedazuridine/decitabine) com­ pared to IV decitabine.
(pts) with myelodysplastic syn­dromes (MDS) explain dura­tion of hypometh- Blood. 2019;134(suppl 1):846.
ylating agent (HMA) ther­apy and over­all sur­vival (OS)? A large pop­u­la­tion- 45. Savona MR, McCloskey JK, Griffiths EA, et  al. Clinical effi­cacy and safety
based anal­y­sis. Blood. 2018;132(suppl 1):370. of oral decitabine/cedazuridine in 133 patients with myelodysplastic syn­
26. Stein EM, Bonifacio G, Latremouille-Viau D, et  al. Treatment pat­ terns dromes (MDS) and chronic myelomonocytic leu­ ke­ mia (CMML). Blood.
and out­comes in patients with myelodysplastic syn­dromes treated with 2020;136(suppl 1):37-38.
hypomethylating agents: a SEER-Medi­ care anal­

sis. Leuk Lymphoma. 46. Thota S, Oganesian A, Azab M, Griffiths EA. Role of cedazuridine/decit-
2021;62(6):1411-1421. abine in the man­age­ment of myelodysplastic syn­drome and chronic myel-
27. Grinblatt DL, Sekeres MA, Komrokji RS, Swern AS, Sullivan KA, Narang M. omonocytic leu­ke­mia. Future Oncol. 2021;17(16):2077-2087.
Patients with myelodysplastic syn­dromes treated with azacitidine in clin­i­ 47. Savona MR, Odenike O, Amrein PC, et  al. Results of first in human (FIH)
cal prac­tice: the AVIDA reg­is­try. Leuk Lymphoma. 2015;56(4):887-895. phase 1 phar­ma­co­ki­netic (PK) guided dose-esca­la­tion study of ASTX727,
28. Davidoff AJ, Hu X, Bewersdorf JP, et al. Hypomethylating agent (HMA) ther­ a com­bi­na­tion of the oral cyti­dine deam­i­nase inhib­i­tor (CDAi) E7727 with
apy use and sur­vival in older adults with refrac­tory ane­mia with excess oral decitabine in sub­jects with myelodysplastic syn­dromes (MDS). Blood.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/439/1852126/439griffiths.pdf by guest on 13 December 2021


blasts (RAEB) in the United States (USA): a large pro­pen­sity score-matched 2015;126(23):1683.
pop­u­la­tion-based study. Leuk Lymphoma. 2020;61(5):1178-1187. 48. Savona MR, Odenike O, Amrein PC, et al. An oral fixed-dose com­bi­na­tion
29. Zeidan AM, Joshi N, Kale H, et al. Predictors of hypomethylating agent dis­ of decitabine and cedazuridine in myelodysplastic syn­dromes: a multi-
con­tin­u­a­tion among patients with higher-risk myelodysplastic syn­dromes. centre, open-label, dose-esca­ la­
tion, phase 1 study. Lancet Haematol.
J Clin Oncol. 2021;39(suppl 15):7043. 2019;6(4):e194-e203.
30. Corman S, Joshi N, Wert T, Kale H, Hill K, Zeidan AM. Under-use of 49. Garcia-Manero G, Griffiths EA, Steensma DP, et al. Oral cedazuridine/decit-
hypomethylating agents in patients with higher-risk myelodysplastic syn­ abine for MDS and CMML: a phase 2 phar­ma­co­ki­netic/phar­ma­co­dy­namic
drome in the United States: a large pop­u­la­tion-based anal­y­sis. Clin Lym- ran­dom­ized cross­over study. Blood. 2020;136(6):674-683.
phoma Myeloma Leuk. 2021;21(2):e206-e211. 50. National Comprehensive Cancer Network. Myelodysplastic syn­ dromes:
31. Marcucci G, Silverman L, Eller M, Lintz L, Beach CL. Bioavailability of azaciti- Clinical prac­tice guide­lines in oncol­ogy. J Nat Compr Canc Netw. 2003;1(4):
dine sub­cu­ta­ne­ous ver­sus intra­ve­nous in patients with the myelodysplastic 456-471.
syn­dromes. J Clin Pharmacol. 2005;45(5):597-602. 51. Wei AH, Döhner H, Pocock C, et al; QUAZAR AML-001 Trial Investigators.
32. Mahfouz RZ, Jankowska A, Ebrahem Q , et  al. Increased CDA expres­sion/ Oral azacitidine main­te­nance ther­apy for acute mye­loid leu­ke­mia in first
activ­ity in males con­trib­utes to decreased cyti­dine ana­log half-life and likely remis­sion. N Engl J Med. 2020;383(26):2526-2537.
con­trib­utes to worse out­comes with 5-azacytidine or decitabine ther­apy. 52. Onureg (azacitidine) tab­lets. Package insert, https://www.accessdata.fda​
Clin Cancer Res. 2013;19(4):938-948. .gov/drugsatfda_docs/label/2020/​214120s​000lbl.pdf accessed 10/1/21.
33. Laille E, Shi T, Garcia-Manero G, et al. Pharmacokinetics and phar­ma­co­ 53. Garcia-Manero G, Stoltz ML, Ward MR, Kantarjian H, Sharma S. A pilot phar­
dy­nam­ics with extended dos­ing of CC-486 in patients with hema­to­logic ma­co­ki­netic study of oral azacitidine. Leukemia. 2008;22(9):1680-1684.
malig­nan­cies. PLoS One. 2015;10(8):e0135520. 54. Garcia-Manero G, Gore SD, Cogle C, et al. Phase I study of oral azacitidine
34. Vidaza (azacitidine) for injec­tion. Package insert, https://www.accessdata in myelodysplastic syn­ dromes, chronic myelomonocytic leu­ ke­
mia, and
.fda.gov/drugsatfda_docs/label/2008/​050794s​011lbl.pdf accessed 10/1/21. acute mye­loid leu­ke­mia. J Clin Oncol. 2011;29(18):2521-2527.
35. Dacogen® (decitabine) for injec­tion. Package insert, https://www.accessdata​ 55. Garcia-Manero G, Santini V, Almeida A, et  al. Phase III, ran­dom­ized, pla­
.fda.gov/drugsatfda_docs/label/2018/​021790s021lbl­.pdf accessed 10/­1/21. cebo-con­trolled trial of CC-486 (oral azacitidine) in patients with lower-risk
36. Watanabe S, Uchida T. Expression of cyti­dine deam­i­nase in human solid myelodysplastic syn­dromes. J Clin Oncol. 2021;39(13):1426-1436.
tumors and its reg­u­la­tion by 1 alpha, 25-dihydroxyvitamin D3. Biochim Bio- 56. Sekeres MA, Othus M, List AF, et al. Randomized phase II study of azacitidine
phys Acta. 1996;1312(2):99-104. alone or in com­bi­na­tion with lenalidomide or with vorinostat in higher-risk
37. Laille E, Savona MR, Scott BL, Boyd TE, Dong Q , Skikne B. Pharmacokinetics myelodysplastic syn­dromes and chronic myelomonocytic leu­ke­mia: North
of dif­fer­ent for­mu­la­tions of oral azacitidine (CC-486) and the effect of food Amer­i­can Intergroup Study SWOG S1117. J Clin Oncol. 2017;35(24):2745-
and mod­i­fied gas­tric pH on phar­ma­co­ki­net­ics in sub­jects with hema­to­logic 2753.
malig­nan­cies. J Clin Pharmacol. 2014;54(6):630-639. 57. Cameron EE, Bachman KE, Myöhänen S, Herman JG, Baylin SB. Synergy of
38. Savona MR, Kolibaba K, Conkling P, et  al. Extended dos­ing with CC-486 demethylation and his­tone deacetylase inhi­bi­tion in the re-expres­sion of
(oral azacitidine) in patients with mye­loid malig­nan­cies. Am J Hematol. genes silenced in can­cer. Nat Genet. 1999;21(1):103-107.
2018;93(10):1199-1206. 58. Gore SD, Baylin S, Sugar E, et al. Combined DNA methyltransferase and his­
39. Garcia-Manero G, Scott BL, Cogle CR, et al. CC-486 (oral azacitidine) in patients tone deacetylase inhi­bi­tion in the treat­ment of mye­loid neo­plasms. Cancer
with myelodysplastic syn­dromes with pre­treat­ment throm­bo­cy­to­pe­nia. Leuk Res. 2006;66(12):6361-6369.
Res. 2018;72(Sep­tem­ber):79-85. 59. Kuderer NM, Choueiri TK, Shah DP, et al; COVID-19 and Cancer Consortium.
40. Garcia-Manero G, Gore SD, Kambhampati S, et  al. Efficacy and safety of Clinical impact of COVID-19 on patients with can­cer (CCC19): a cohort
extended dos­ing sched­ules of CC-486 (oral azacitidine) in patients with study. Lancet. 2020;395(10241):1907-1918.
lower-risk myelodysplastic syn­dromes. Leukemia. 2016;30(4):889-896. 60. Zeidan AM, Boddu PC, Patnaik MM, et  al. Special con­sid­er­ations in the
41. Mistry B, Jones MM, Kubiak P, et al. A phase 1 study to assess the abso­lute man­age­ment of adult patients with acute leu­kae­mias and mye­loid neo­
bio­avail­abil­ity and safety of an oral solu­tion of decitabine in sub­jects with plasms in the COVID-19 era: rec­om­men­da­tions from a panel of inter­na­-
myelodysplastic syn­dromes (MDS). Blood. 2011;118(21):3801. tional experts. Lancet Haematol. 2020;7(8):e601-e612.
42. Frances A, Cordelier P. The emerg­ing role of cyti­dine deam­i­nase in human
dis­eases: a new oppor­tu­nity for ther­apy? Mol Ther. 2020;28(2):357-366.
43. Inqovi® (decitabine and cedazuridine) tab­lets. Package insert, https://www
.accessdata.fda.gov/drugsatfda_docs/label/2020/212576s000lbl.pdf © 2021 by The Amer­i­can Society of Hematology
accessed 10/1/2021. DOI 10.1182/hema­tol­ogy.2021000278

Prakash Singh Shekhawat


Oral hypomethylating agents for MDS  |  447
MPN: NEW DIRECTIONS

EVIDENCE-BASED MINIREVIEW

Are DOACs an alternative to vitamin K

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/448/1851806/448schieppati.pdf by guest on 13 December 2021


antagonists for treatment of venous
thromboembolism in patients with MPN?
Francesca Schieppati1 and Anna Falanga1,2
1
Department of Transfusion Medicine and Hematology, Hospital Papa Giovanni XXIII, Bergamo, Italy; and 2Department of Medicine and Surgery,
University of Milan Bicocca, Milan, Italy

LEARNING OBJECTIVES
• Understand the current strategies of VTE treatment and prevention of recurrences in patients with MPN
• Identify the pros and cons of traditional VTE treatment with VKAs vs newer approaches with DOACs in light of the
available evidence

ent with a thrombotic event before or at MPN diagnosis.3


CLINICAL CASE Although arterial thrombotic events (ATEs) are twice as
A 55-year-old man with a history of JAK2-mutated essen- common as venous events, MPN patients have a 4-fold
tial thrombocythemia (ET) presented to the emergency increased risk of ATEs and a 10-fold increased risk of venous
room for the sudden onset of severe pain and swelling thromboembolism (VTE) shortly after diagnosis compared
in his right leg during the night. He denied any recent to the general population, and the thrombotic rate remains
trauma, surgery, or infection. He was in good general con- significantly elevated throughout the follow-up.2
dition except for a modest shortness of breath. An elec- MPN-related VTE manifests most often as deep vein
trocardiogram showed a sinus tachycardia; the oxygen thrombosis (DVT) of the legs and/or pulmonary embo-
saturation was 95% in room air. At physical examination, a lism (PE; accounting for 40%-90% of all cases in different
palpable cord at the right thigh was appreciable, associ- reports), although thromboses at unusual sites, ie, involv-
ated with marked edema and erythema of the calf. A com- ing the splanchnic and cerebral districts, are remarkably
pression ultrasonography revealed an absence of color frequent in these patients.4
flow compatible with complete deep vein thrombosis of In PV and ET, treatment is aimed at preventing throm-
the femoral and popliteal veins of the right leg. A chest botic complications, and risk-stratification for treatment
computed tomography scan also showed bilateral seg- decisions is based on thrombotic risk factors, including an
mental pulmonary embolism. His past medical history was age over 60, a history of thrombosis, and, only in ET, the
otherwise mute except for being a heterozygous carrier presence of the JAK2V617F mutation.5,6 In this regard, the
of the factor V Leiden variant and having hemorrhoids in substantial involvement of JAK2V617F and clonal hemato-
the past. He was on low-dose aspirin as his sole medica- poiesis in thrombosis development in MPN has emerged.7
tion. What is the best treatment of venous thromboembo- Current strategies of thromboprophylaxis include the
lism in this ET patient? use of low-dose aspirin (75-100 mg) once daily in both high-
and low-risk PV patients (aged <60 years and no thrombo-
sis history) and in low-/intermediate-risk ET patients (JAK2
Introduction mutated OR aged >60 years and no thrombosis history),5
Classical BCR/ABL-negative myeloproliferative neoplasms based on 2 randomized controlled trials in PV and on 1 ret-
(MPN) include polycythemia vera (PV), essential thrombo- rospective analysis in ET, respectively.8-10 Low-dose aspi-
cythemia (ET), and primary myelofibrosis (PMF). Patients rin showed a considerable but non-statistically significant
with MPN are at high risk of thrombotic manifestations, beneficial effect on mortality from thrombotic events and
which considerably affect morbidity and mortality, espe- did not prevent major cardiovascular and venous throm-
cially in younger patients.1,2 Up to 30% of patients pres- botic events, taken individually.8,11 Thromboprophylaxis also

448 | Hematology 2021 | ASH Education Program


Table 1. Principal stud­ies includ­ing MPN patients on VKA treat­ment for usual site VTE

Median
N included Overall throm­bo­sis fol­low-up
Reference Study pop­u­la­tion in the study N on VKAs recur­rence (A/V) VTE recur­rence Major bleed­ing (years)
De Stefano PV/ET with at least 494 90 33.6% (7.6% pt-y)* 13.1% (3% pt-y)* 5.4% (0.9% pt-y)* 5.3
et al14 1 epi­sode of throm­bo­sis 7.7% (0.9% pt-y)†
(ATE and VTE) (2.8 pt-y)‡
Hernandez- PV/ET receiv­ing VKA for a 150 150 28% (6.0% pt-y)† 24% (2.7% ON vs 11.3% (over­all 1.7% pt-y, 7.7
Boluda first VTE or ATE epi­sode 9.0% OFF, p)† 1.8% ON vs 1.5%
et al16 OFF)†
De Stefano PV/ET/PMF on sys­temic 206 155 21.8% (6.5% pt-y)* 17.4% (5.2% pt-y)* 6.4% (2.4% ON vs 0.7 3
et al15 anticoagulation for a 12.2% (4.7% pt-y)† 9.6% (4.2% pt-y ON OFF)†
first VTE epi­sode vs 9.6% pt-y OFF)†

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/448/1851806/448schieppati.pdf by guest on 13 December 2021


Wille et al17 PV/ET/PMF with a first 78 40 — 20.5% (6.0% pt-y)* 26.9% 2
VTE epi­sode
*Entire cohort.
†Patients on VKA only.
‡Patients on VKA and aspi­rin.
A/V, arterial/venous; N, num­ber of patients; ON, on VKA; OFF, off VKA; Pt-y, patient-years.

includes phle­bot­omy in all­PV patients and myelosuppression 29%, respec­tively).15 However, it is impor­tant to con­sider that the
with cytoreductive ther­apy in high-risk PV and ET patients.5,12 cumu­la­tive inci­dence of VTE recur­rence in MPN patients receiv­
ing ade­quate VKA treat­ment still remains greater than that of the
Treatment of acute VTE and sec­ond­ary pre­ven­tion of VTE gen­eral pop­u­la­tion (7.8% vs 1.8%-3.5% at 1 year, respec­tively).15
recur­rence In the stud­ies shown in Table 1, the bleed­ing inci­dence dur­
Despite pro­phy­laxis, the reported inci­dence of VTE is 0.6% to ing VKA treat­ment ranged between 0.9% and 2.8% patient-years
1.0% in patient-years across the dif­fer­ent MPN sub­types and is and sig­nif­i­cantly increased only when admin­is­tered in com­bi­na­
con­sid­er­ably higher than the annual inci­dence of 0.1% to 0.2% tion with aspi­rin (com­pared to patients off VKAs). Nevertheless,
observed in the gen­eral pop­u­la­tion.4 Detected risk fac­tors for bleed­ing com­pli­ca­tions with VKAs look higher in MPN com­pared
a first VTE epi­sode in MPN patients include an age >60, a pre­ to non-MPN patients (up to 2.8% vs 1.2%-2.2% in patient-years,
vi­ous his­tory of VTE, a his­tory of major bleed­ing, leu­ko­cy­to­sis, respec­tively),4 with dis­ease-related fac­tors con­trib­ut­ing to the
inherited thrombophilia (in youn­ger patients), and JAK2V617F (in over­all higher hem­or­rhagic risk in MPN patients com­pared to the
ET and PMF).4 gen­eral pop­u­la­tion.3 This is a very rel­e­vant aspect when choos­
In light of a lack of pro­spec­tive stud­ies addressing the effi­ ing the type and dura­tion of anti­co­ag­u­lant ther­apy. Improving
cacy and safety of the newer direct oral anti­co­ag­u­lants (DOACs) the effi­cacy and safety of anticoagulation in MPN patients with
in MPN patients, based on expert opin­ion the ini­tial treat­ment VTE still rep­re­sents an open issue.
for acute VTE in MPN patients should start with low-molec­u­lar- In the last years, more ther­ a­
peu­ tic options for VTE have
weight hep­a­rin (LMWH) or fondaparinux followed by vita­min K become avail­­able with the advent of the DOACs, includ­ing the
antag­o­nists (VKAs), targeting an inter­na­tional nor­mal­ized ratio fac­tor IIa inhib­i­tor dabigatran and the fac­tor Xa (FXa) inhib­i­tors
(INR) of 2.5 for at least 3 to 6 months.4,13 rivaroxaban, apixaban, edoxaban, and oth­ers. In the non-MPN
The effi­cacy and safety of VKAs in the MPN set­ting has been set­ting, DOACs have become the first treat­ment choice for DVT
eval­u­ated in 4 ret­ro­spec­tive stud­ies, includ­ing a very recent one and PE.19 Moreover, FXa inhib­i­tors have been tested spe­cif­i­cally
(Table 1).14-17 The annual inci­dence rate of VTE recur­rence ranged in the can­cer pop­u­la­tion by means of RCTs, show­ing a good effi­
between 3% and 6% in patient-years.14-17 VKA treat­ment was cacy and safety pro­file com­pared to the stan­dard ther­apy with
asso­ci­ated with a sig­nif­i­cant reduc­tion in VTE recur­rences in all LMWH.20-22 Thus, expert guide­lines have recently included these
4 stud­ies and ATEs in 1 study.16 drugs in the recommended treat­ment options for can­cer-asso­
With regard to the over­ all dura­ tion of anticoagulation in ci­ated VTE.23,24 No pro­spec­tive con­trolled stud­ies on the use
these patients, while there is con­sen­sus on con­tinu­ing life­long of DOACs have been conducted with MPN patients so far. In
treat­ment in patients with splanch­nic vein throm­bo­sis,18 the opti­ any case, some obser­va­tional ret­ro­spec­tive stud­ies have been
mal treat­ment dura­tion for VTE at the usual sites is uncer­tain. published in recent years eval­u­at­ing the effi­cacy and safety of
Two stud­ies com­par­ing VKA indef­i­nite treat­ment vs dis­con­tin­ DOACs in MPN (Table 2).
u­a­tion after 6 months showed a sig­nif­i­cantly greater inci­dence Three small stud­ies described an over­all throm­botic recur­
of recur­rence in the group that discontinued VKAs (2.7%-4.2% rence of 0% to 4% involv­ing only arte­rial dis­tricts.25-27 Major
in patient-years vs 9%-9.6%).15,16 Indeed, VKA sus­pen­sion resulted bleed­ing was reported in 0% to 12% of patients, with 3 cases of
in a 2- to 3-fold increased risk of recur­rence.15,16 In addi­tion, MPN clin­i­cally rel­e­vant non­ma­jor bleed­ing in asso­ci­a­tion with aspi­
patients showed a higher rate of 5-year recur­rence after anti­ rin.26 A recent large ret­ro­spec­tive study includ­ing 442 patients
co­ag­u­lant with­drawal com­pared to non-MPN patients (42% vs treated with DOACs for atrial fibril­la­tion (AF) or for VTE pro­vided
Prakash Singh Shekhawat
DOACs vs VKAs in MPN-asso­ci­ated VTE treat­ment  |  449
Table 2. Studies includ­ing at least 20 MPN patients on DOAC treat­ment for usual site VTE

Median
Study N on N on N on N on N on Overall throm­botic fol­low-up
Reference pop­u­la­tion DOAC rivar apix edox dabig recur­rence VTE recur­rence Major bleed­ing (years)
Ianotto et al25
PV/ET receiv­ing 25* 16 9 — — 4% (1 stroke) 0 12% 2.1
DOAC for AF
or VTE
Curto-Garcia PV/ET/PMF/ 32 17 14 1 0 3% (1 mes­en­teric 0 0% 2.1
et al26 MDS-MPN ische­mia)
receiv­ing
DOAC for VTE
Serrao et al27 PV/ET/PMF 71† 26 21 14 10 0% — 0% 1
receiv­ing

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/448/1851806/448schieppati.pdf by guest on 13 December 2021


DOAC for AF
or VTE
Barbui et al28 PV/ET/PMF 442‡ 187 157 48 50 4.9% (2.1% pt-y) (AF) 1.5% (0.6% pt-y) (AF) 6.9% (3.0 pt-y) (AF) 1.7
receiv­ing 9.2% (4.5% pt-y) (VTE) 7.1% (3.4% pt-y) (VTE) 5.0% (2.3% pt-y) (VTE)
DOAC for AF
or VTE
*13 patients receiv­ing DOAC for AF, 4 for AF and stroke, and 8 for VTE.
†35 patients receiv­ing DOAC for AF; 36 for VTE.
‡203 patients receiv­ing DOAC for AF; 239 for VTE.
Apix, apixaban; dabig, dabigatran; edox, edoxaban; rivar, rivaroxaban.

more exten­sive infor­ma­tion on the rates of thrombohemor- sim­i­lar for the 2 anti­co­ag­u­lants.29,30 Interestingly, a sig­nif­i­cantly
rhagic com­pli­ca­tions in this set­ting.28 Specifically, the inci­ higher VTE recur­rence rate was observed after the dis­con­tin­u­
dence of a first VTE event in patients receiv­ing a DOAC for AF a­tion of either drug.30
was 0.6% in patient-years, while the inci­dence of recur­rent In this set­ting it is impor­tant to recall that cytoreduction is
VTE in patients receiv­ing a DOAC for a prior VTE was 3.4% in recommended in PV/ET who have a his­tory of throm­bo­sis or
patient-years, which was no dif­fer­ent from the recur­rence inci­ are expe­ri­enc­ing a first VTE epi­sode in the fol­low-up.5 However,
dence observed in the VKA stud­ies.14-17 Moreover, annual rates of recent stud­ ies show that despite the dem­ on­
strated effi­ cacy
major bleed­ing ranged from 2.3% to 3% in patient-years,28 also of hydroxy­urea (HU) at cytoreduction to pre­vent pri­mary and
sim­i­lar to VKAs. Therefore, on the basis of lim­ited avail­­able evi­ recur­rent arte­rial events,14,31 its action in the pre­ven­tion of first or
dence, DOACs and VKAs seem to have a com­pa­ra­ble risk/ben­ recur­rent venous throm­bo­sis is more lim­ited.32,33 Other cytore-
e­fit pro­file in the treat­ment and sec­ond­ary pre­ven­tion of VTE in ductive or dis­ease-mod­i­fy­ing agents (ie, ruxolitinib, anagrelide,
MPN patients. Finally, 2 recent small stud­ies tried to ret­ro­spec­ inter­ feron alpha, and ropeginterferon) have proved valu­ able
tively com­pare the out­comes of MPN patients treated with alter­ na­tives to HU for dis­ ease con­ trol, although for most of
VKAs or DOACs29,30 (Table 3). In the study by Huenerbein et al, these drugs there is no con­trolled evi­dence show­ing their supe­
despite a higher relapse rate seen in the VKA group com­pared ri­or­ity over HU at pre­vent­ing VTE. Since the inci­dence of ATEs
to the DOAC group, throm­bo­sis-free sur­vival was no dif­fer­ent and VTE remains high in MPN patients despite cytoreduction,
between the 2 groups.29 In the study by Fedorov et al, the rates fur­ther ther­a­peu­tic pro­pos­als are needed, pos­si­bly addressing
of throm­bo­sis were also com­pa­ra­ble between the 2 anti­co­ag­ addi­tional mech­a­nisms besides myelosuppression and targeting
u­lant reg­i­mens. In both stud­ies the rate of major bleed­ing was other thrombogenic path­ways.7

Table 3. Retrospective stud­ies com­par­ing throm­bo­sis recur­rence and major bleed­ing in MPN patients receiv­ing VKA or DOAC

Overall throm­botic
Median
recur­rence VTE recur­rence Major bleed­ing
N on N on fol­low-up
Reference Study pop­u­la­tion VKA DOAC VKA DOAC VKA DOAC VKA DOAC (years)
Huenerbein PV/ET/PMF/ 45 26 48.8% 15.3% 24.4% 11.5% 8.88% 7.6% 3.2
et al29 MPN-U on sys­temic anticoagulation for
VTE or ATE
Fedorov PV/ET/PMF/ 31 22 19.4% 22.7% — — 6.4% 4.5% 1.2
et al30 MPN-U on sys­temic anticoagulation for
VTE or ATE
MPN-U, mye­lo­pro­lif­er­a­tive neo­plasm-unclas­si­fi­able: PV, plycythemia vera.

450  |  Hematology 2021  |  ASH Education Program


When it comes to choos­ing an anti­co­ag­u­lant agent, the pros our treat­ment strat­e­gies in case of changes in the patient’s dis­
and cons of treat­ment with VKAs or DOACs for MPN-asso­ci­ated ease pat­tern or the avail­abil­ity of new evi­dence on anticoagu-
VTE can be sum­ma­rized as fol­lows: lation modal­i­ties in MPN.
VKA pros:
•  Larger stud­ies eval­u­at­ing effi­cacy and safety
•  Longer fol­low-up Conflict-of-inter­est dis­clo­sure
•  Reduced VTE recur­rence Francesca Schieppati: no com­ pet­ ing finan­ cial inter­
ests to
•  Reduced ATE recur­rence declare.
•  Long-term pro­tec­tion from VTE recur­rence Anna Falanga: no com­pet­ing finan­cial inter­ests to declare.

VKA cons: Off-label drug use


•  Laboratory mon­i­tor­ing of INR Francesca Schieppati: nothing to disclose.
•  Increased bur­den in case of life­long treat­ment Anna Falanga: nothing to disclose.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/448/1851806/448schieppati.pdf by guest on 13 December 2021


•  Higher bleed­ing risk in MPN than in non-MPN patients
•  Residual risk of recur­rence even with an INR in the ther­a­peu­tic Correspondence
range Francesca Schieppati, Department of Transfusion Medicine and
Hematology, Hospital Papa Giovanni XXIII, Piazza OMS n 1, Berga-
DOACs pros: mo 24127, Italy; e-mail: fschieppati@gmail​­.com.
•  Easier to admin­is­ter
•  Routine lab­o­ra­tory mon­i­tor­ing is unnec­es­sary References
•  Good effi­ cacy and safety pro­ file in stud­ies with can­
cer 1. Falanga A, Marchetti M, Schieppati F. Prevention and man­ age­
ment of
throm­bo­sis in BCR/ABL-neg­a­tive mye­lo­pro­lif­er­a­tive neo­plasms. Hamo-
patients staseologie. 2021;41(1):48-57.
•  More appeal­ing than VKAs for indef­i­nite treat­ment 2. Hultcrantz M, Björkholm M, Dickman PW, et al. Risk for arte­rial and venous
throm­bo­sis in patients with mye­lo­pro­lif­er­a­tive neo­plasms: a pop­u­la­tion-
DOACs cons: based cohort study. Ann Intern Med. 2018;168(5):317-325.
3. Rungjirajittranon T, Owattanapanich W, Ungprasert P, Siritanaratkul N,
•  Limited stud­ies eval­u­at­ing effi­cacy and safety
Ruchutrakool T. A sys­tem­atic review and meta-anal­y­sis of the prev­a­lence of
•  Shorter fol­low-up throm­bo­sis and bleed­ing at diag­no­sis of Philadelphia-neg­a­tive mye­lo­pro­
•  Higher bleed­ing risk in MPN than in non-MPN patients lif­er­a­tive neo­plasms. BMC Cancer. 2019;19(1):184.
•  Residual risk of recur­rence even dur­ing treat­ment 4. De Stefano V, Finazzi G, Barbui T. Antithrombotic ther­apy for venous throm­
bo­em­bo­lism in mye­lo­pro­lif­er­a­tive neo­plasms. Blood Cancer J. 2018;8(7):65.
In con­clu­sion, the cur­rent evi­dence, although lim­ited, shows 5. Tefferi A, Barbui T. Polycythemia vera and essen­tial thrombocythemia: 2021
sim­i­lar pat­terns in terms of the effi­cacy and safety of VKAs and update on diag­no­sis, risk-strat­i­fi­ca­tion and man­age­ment. Am J Hematol.
2020;95(12):1599-1613.
DOACs for the treat­ment and pre­ven­tion of recur­rent VTE in
6. Barbui T, Finazzi G, Carobbio A, et  al. Development and val­i­da­tion of an
MPN. While we wait for a ran­dom­ized com­par­i­son between International Prognostic Score of throm­bo­sis in World Health Organization-
the 2 reg­i­mens, treat­ment deci­sions should be guided accord- essen­tial thrombocythemia (IPSET-throm­bo­sis). Blood. 2012;120(26):5128-
ing to indi­vid­ual fac­tors (ie, renal func­tion, bleed­ing risk pro­file, 5133, quiz 5252.
con­com­i­tant med­i­ca­tions) as well as patient pref­er­ences. The 7. Moliterno AR, Ginzburg YZ, Hoffman R. Clinical insights into the ori­gins
of throm­bo­sis in mye­lo­pro­lif­er­a­tive neo­plasms. Blood. 2021;137(9):1145-
opti­mal dura­tion of anticoagulation is uncer­tain, but sub­stan­ 1153.
tial evi­dence indi­cates that recur­rence risk is par­tic­u­larly high in 8. Landolfi R, Marchioli R, Kutti J, et al; Euro­pean Collaboration on Low-Dose
these patients even years after the index event. To help decide Aspirin in Polycythemia Vera Investigators. Efficacy and safety of low-dose
on the dura­tion, it is wise to per­form a care­ful assess­ment of the aspi­rin in poly­cy­the­mia vera. N Engl J Med. 2004;350(2):114-124.
9. Italiano Studio Policitemia GRUPPO. Low-dose aspi­rin in polycythaemia
patient’s risk fac­tors for recur­rence (such as unpro­voked VTE,
vera: a pilot study. Br J Haematol. 1997;97(2):453-456.
prox­i­mal DVT, pul­mo­nary embo­li­za­tion, thrombophilia, etc) and 10. Alvarez-Larrán A, Cervantes F, Pereira A, et  al. Observation ver­sus anti-
to plan a peri­od­i­cal reassessment of risk fac­tors for throm­bo­sis platelet ther­apy as pri­mary pro­phy­laxis for throm­bo­sis in low-risk essen­tial
and bleed­ing dur­ing the fol­low-up. thrombocythemia. Blood. 2010;116(8):1205-1210, quiz 1387.
11. Squizzato A, Romualdi E, Passamonti F, Middeldorp S. Antiplatelet drugs for
polycythaemia vera and essen­tial thrombocythaemia. Cochrane Database
Syst Rev. 2013;(4):CD006503.
12. Marchioli R, Finazzi G, Specchia G, et al; CYTO-PV Collaborative Group.
Cardiovascular events and inten­sity of treat­ment in poly­cy­the­mia vera. N
CLINICAL CASE (Con­t in­u ed)
Engl J Med. 2013;368(1):22-33.
We discussed pros and cons with our patient, who was rel­a­ 13. Barbui T, De Stefano V, Falanga A, et al. Addressing and pro­pos­ing solu­tions
tively young and in good form. We agreed to start HU and an for unmet clin­i­cal needs in the man­age­ment of mye­lo­pro­lif­er­a­tive neo­
plasm-asso­ci­ated throm­bo­sis: a con­sen­sus-based posi­tion paper. Blood
FXa inhib­i­tor for VTE treat­ment. This deci­sion was based on
Cancer J. 2019;9(8):61.
the patient’s pref­er­ence for a less oner­ous reg­i­men and on our 14. De Stefano V, Za T, Rossi E, et al; GIMEMA CMD-Working Party. Recurrent
expe­ri­ence with DOACs in can­cer patients. We aimed to pur­ throm­bo­sis in patients with poly­cy­the­mia vera and essen­tial thrombo-
sue a long-term treat­ment, con­sid­er­ing his unpro­voked prox­i­ cythemia: inci­dence, risk fac­tors, and effect of treat­ments. Haematolog-
mal DVT and PE and the pres­ence of addi­tional risk fac­tors such ica. 2008;93(3):372-380.
15. De Stefano V, Ruggeri M, Cervantes F, et al. High rate of recur­rent venous
as JAK2 muta­tion and inherited thrombophilia. We suspended throm­bo­em­bo­lism in patients with mye­lo­pro­lif­er­a­tive neo­plasms and effect
aspi­rin to reduce his bleed­ing risk. Nevertheless, we peri­od­i­ of pro­phy­laxis with vita­min K antag­o­nists. Leukemia. 2016;30(10):2032-
cally assess his throm­bo­sis and bleed­ing risk, will­ing to adjust 2038.
Prakash Singh Shekhawat
DOACs vs VKAs in MPN-asso­ci­ated VTE treat­ment  |  451
16. Hernández-Boluda JC, Arellano-Rodrigo E, Cervantes F, et al; Grupo venous throm­bo­em­bo­lism in a large ter­tiary cen­tre in the UK. Br J Haema-
Español de Enfermedades Mieloproliferativas Filadelfia Negativas. Oral tol. 2020;189(3):e79-e81.
anticoagulation to pre­vent throm­bo­sis recur­rence in poly­cy­the­mia vera 27. Serrao A, Breccia M, Napolitano M, et al. A mul­ti­cen­ter real-life study on
and essen­tial thrombocythemia. Ann Hematol. 2015;94(6):911-918. anti­co­ag­u­lant treat­ment with direct oral anti­co­ag­u­lants in patients with Ph-
17. Wille K, Sadjadian P, Becker T, et al. High risk of recur­rent venous throm­bo­ neg­a­tive mye­lo­pro­lif­er­a­tive neo­plasms. Am J Hematol. 2020;95(12):E329-
em­bo­lism in BCR-ABL-neg­a­tive mye­lo­pro­lif­er­a­tive neo­plasms after ter­mi­ E332.
na­tion of anticoagulation. Ann Hematol. 2019;98(1):93-100. 28. Barbui T, De Stefano V, Carobbio A, et  al. Direct oral anti­co­ag­u­lants for
18. Finazzi G, De Stefano V, Barbui T. Splanchnic vein throm­ bo­
sis in mye­ mye­lo­pro­lif­er­a­tive neo­plasms: results from an inter­na­tional study on 442
lo­pro­lif­er­a­tive neo­plasms: treat­ment algo­rithm 2018. Blood Cancer J. patients. Blood. 2020;136(suppl 1):42-43.
2018;8(7):64. 29. Huenerbein K, Sadjadian P, Becker T, et  al. Direct oral anti­ co­ag­u­
lants
19. Kearon C, Akl EA, Ornelas J, et al. Antithrombotic ther­apy for VTE dis­ease: (DOAC) for pre­ven­tion of recur­rent arte­rial or venous throm­bo­em­bolic
CHEST guide­line and expert panel report. Chest. 2016;149(2):315-352. events (ATE/VTE) in mye­lo­pro­lif­er­a­tive neo­plasms. Ann Hematol. 2021;
20. Raskob GE, van Es N, Verhamme P, et al; Hokusai VTE Cancer Investigators. 100(8):2015-2022.
Edoxaban for the treat­ment of can­cer-asso­ci­ated venous throm­bo­em­bo­ 30. Fedorov K, Goel S, Kushnir M, Billett HH. Direct oral anti­co­ag­u­lants for pre­
lism. N Engl J Med. 2018;378(7):615-624. ven­tion of recur­rent throm­bo­sis in mye­lo­pro­lif­er­a­tive neo­plasms. Blood.
21. Young AM, Marshall A, Thirlwall J, et  al. Comparison of an oral fac­tor Xa 2019;134(suppl 1):4193.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/448/1851806/448schieppati.pdf by guest on 13 December 2021


inhib­i­tor with low molec­u­lar weight hep­a­rin in patients with can­cer with 31. Barbui T, Vannucchi AM, Finazzi G, et  al. A reappraisal of the ben­e­fit-risk
venous throm­bo­em­bo­lism: results of a ran­dom­ized trial (SELECT-D). J Clin pro­file of hydroxy­urea in poly­cy­the­mia vera: a pro­pen­sity-matched study.
Oncol. 2018;36(20):2017-2023. Am J Hematol. 2017;92(11):1131-1136.
22. Agnelli G, Becattini C, Meyer G, et al; Caravaggio Investigators. Apixaban 32. De Stefano V, Rossi E, Carobbio A, et al. Hydroxyurea pre­vents arte­rial and
for the treat­ment of venous throm­bo­em­bo­lism asso­ci­ated with can­cer. N late venous throm­botic recur­rences in patients with mye­lo­pro­lif­er­a­tive
Engl J Med. 2020;382(17):1599-1607. neo­plasms but fails in the splanch­nic venous dis­trict: pooled anal­y­sis of
23. Lyman GH, Carrier M, Ay C, et al. Amer­i­can Society of Hematology 2021 1500 cases. Blood Cancer J. 2018;8(11):112.
guide­lines for man­age­ment of venous throm­bo­em­bo­lism: pre­ven­tion and 33. Barbui T, De Stefano V, Ghirardi A, Masciulli A, Finazzi G, Vannucchi AM. Dif-
treat­ment in patients with can­cer. Blood Adv. 2021;5(4):927-974. ferent effect of hydroxy­urea and phle­bot­omy on pre­ven­tion of arte­rial and
24. Key NS, Khorana AA, Kuderer NM, et al. Venous throm­bo­em­bo­lism pro­phy­ venous throm­bo­sis in poly­cy­the­mia vera. Blood Cancer J. 2018;8(12):124.
laxis and treat­ment in patients with can­cer: ASCO clin­i­cal prac­tice guide­
line update. J Clin Oncol. 2020;38(5):496-520.
25. Ianotto JC, Couturier MA, Galinat H, et al. Administration of direct oral anti­
co­ag­u­lants in patients with mye­lo­pro­lif­er­a­tive neo­plasms. Int J Hematol.
2017;106(4):517-521.
26. Curto-Garcia N, Doyle AJ, Breen KA, et  al. Outcomes of patients receiv­ © 2021 by The Amer­i­can Society of Hematology
ing direct oral anti­co­ag­u­lants for mye­lo­pro­lif­er­a­tive neo­plasm-asso­ci­ated DOI 10.1182/hema­tol­ogy.2021000318

452  |  Hematology 2021  |  ASH Education Program


MPN: NEW DIRECTIONS

Novel therapies vs hematopoietic cell


transplantation in myelofibrosis: who, when, how?

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/453/1851800/453england.pdf by guest on 13 December 2021


James England and Vikas Gupta
Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada

Myelofibrosis is one of the classical Philadelphia chromosome–negative myeloproliferative neoplasms characterized by


progressive marrow failure and chronic inflammation. Discovery of the JAK2 mutation paved the way for development
of small molecular inhibitors and further facilitated the research in understanding of molecular biology of the disease.
Development of novel medications and synergistic combinations with standard JAK inhibitor (JAKi) therapy may have the
potential to improve depth and duration of disease control and symptomatic benefit, whereas advancements in alloge-
neic hematopoietic stem cell transplantation (HCT) have improved tolerability and donor availability, allowing for more
patients to pursue this potentially curative therapy. The increase in options for medical therapy and changing risk profile
of HCT is leading to increased complexity in counseling patients on choice of management strategy. In this case-based
review, we summarize our approach to symptom-directed medical therapy, including the use of novel drugs and combi-
nation therapies currently under study in advanced clinical trials. We outline our recommendations for optimal timing of
HCT, including risk-adapted selection for early HCT as opposed to delayed HCT after upfront JAKi therapy, as well as the
use of pretransplant JAKi and alternative donor sources.

LEARNING OBJECTIVES
• Outline a risk-adapted approach for selection of upfront HCT vs nontransplant therapies for MF in chronic phase
• Overview the progress of symptom-directed JAKi therapy toward disease-modifying novel therapies for MF

Introduction ent but is limited by significant mortality and morbidity.


Myelofibrosis (MF) encompasses a group of clonal stem Accurate estimation of risk to inform patient-centered
cell disorders that are either primary myelofibrosis or arise decision making is a priority concern in managing MF. The
from a preceding polycythemia vera (post-PV MF) or essen- approval of JAK inhibitor (JAKi) therapy with ruxolitinib
tial thrombocythemia (ET; post-ET MF). Although somatic and, more recently, fedratinib provides an alternative
mutations and disease biology may differ between primary effective approach to manage constitutional and spleno-
and secondary MF, the management approach remains megaly-related symptoms in patients with MF who are not
similar. Somatic mutations in myeloproliferative neoplasm candidates or do not wish to pursue HCT. JAKi therapy is
(MPN) driver (JAK2, CALR, MPL) and other myeloid malig- not curative and has limited duration of clinical benefit
nancy genes perpetuate increased cytokine expression, as half of patients experience treatment failure after 2 to
inflammation, and fibrosis within the niche environment 3 years.2,3 A variety of novel agents are in advanced clini-
of the bone marrow (BM).1 The clinical manifestations of cal trials for MF, and these emerging therapies may lead
MF include cytokine-mediated constitutional symptoms to disease modification or improved depth and duration
(night sweats, weight loss, fever), cytopenias from progres- of symptom control.4 Conversely, progress in support-
sive marrow failure, extramedullary hematopoiesis, and ive care, conditioning, graft-versus-host disease (GVHD)
mechanical symptoms from splenomegaly. MF is a heterog- prophylaxis, and increasing safety of alternative donor
enous disease with variable rates of progression to death sources may allow for more patients to benefit from HCT.
and blastic transformation depending on underlying clini- In this case-based review, we outline our approach and
cal and genetic risk factors. framework for individualized decision making for patients
Allogeneic hematopoietic stem cell transplantation with MF on selection of HCT vs nontransplant strategies,
(HCT) is the only therapy with potential for cure at pres- including emerging therapies through clinical trials.
Prakash Singh Shekhawat
Therapeutic landscape for transplant eligible MF | 453
Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/453/1851800/453england.pdf by guest on 13 December 2021
Figure 1.  Timing options for consideration of HCT in MF.

2/high-risk dis­ease and inter­me­di­ate-1 risk with addi­tional risk


CLINICAL CASES fac­tors.5,6 Transplant stud­ies have reported bet­ter out­comes in
Case 1: A 60-year old white woman had symp­toms of slowly patients treated ear­lier in the dis­ease course.7 However, the same
pro­gres­sive ane­mia, abdom­i­nal dis­com­fort, and early sati­ety. group of patients also has bet­ter and more dura­ble response
Physical exam­in ­ a­tion showed a spleen pal­pa­ble 12 cm below the with JAKi ther­apy.8,9 To our knowl­edge, there is no pro­spec­tive
cos­tal mar­gin (BCM). Hemoglobin was 95 g/L, white blood cell study to com­pare the out­comes of HCT vs nontransplant ther­a­
count was 7.0 × 109/L, plate­lets were 150 × 109/L, and periph­eral pies in MF, and lim­ited ret­ro­spec­tive stud­ies were either done
blood (PB) blasts were 1.2%. Bone mar­row biopsy (BMBx) spec­i­ in the pre-JAKi era or had a very small pro­por­tion of patients
men was con­sis­tent with diag­no­sis of overt pri­mary mye­lo­fi­bro­ treated with JAKi.7,10 Lack of com­par­a­tive data in this area has led
sis with World Health Organization grade 3 fibro­sis and blasts to sig­nif­i­cant var­i­a­tions in prac­tice on appli­ca­tion of HCT in MF.
less than 5%. Cytogenetics showed nor­mal kar­yo­type. Targeted When discussing with a patient regard­ing the trans­plant tim­
next-gen­er­a­tion sequenc­ing dem­on­strated type 1 CALR muta­ ing, var­i­ous time points for trans­plant can be con­sid­ered (Figure 1):
tion as the sole abnor­mal­ity (variant allele frequency (VAF) 35%).
a.  Upfront trans­plan­ta­tion: early HCT usu­ally within a few months
Case 2: A 63-year-old man of east Indian ori­gin with a long-
of diag­no­sis
stand­ing diag­no­sis of PV lost the need for phle­bot­omy. He was
b.  Delayed trans­plan­ta­tion: HCT is delayed as long as one can,
asymp­tom­atic, and phys­i­cal exam­i­na­tion showed pal­pa­ble
trans­plant offered at early signs of fail­ure of nontransplant
spleen 7 cm BCM. Complete blood count showed the fol­low­ing:
ther­apy
hemo­glo­bin, 130 g/L; white blood cell count, 15.3 × 109/L; plate­
c.  Leukemic pro­gres­sion: HCT at time of trans­for­ma­tion to acute
lets, 47 × 109/L; and PB blasts, 2%. BMBx spec­i­men was con­sis­
phase (AP) (PB or BM blasts 10%-19%) or blast phase (BP) (PB
tent with post-PV MF with World Health Organization grade 2
or BM blasts ≥20%)
fibro­sis and blasts less than 5%. Cytogenetics dem­on­strated a
dele­tion of 11q in 15 of 20 meta­phases. The fol­low­ing path­o­genic There is no one-size-fits-all­approach in patients with MF;
muta­tions were noted in next-gen­er­a­tion sequenc­ing stud­ies: how­ever, we strongly rec­om­mend against waiting for dis­ease to
JAK2 V617F (VAF 80%), ASXL1 (VAF 27%), and IDH1 (VAF 36%). prog­ress to AP/BP as out­comes after the HCT are poor and many
patients never make it to the trans­plant stage.11-13
Our goal in the begin­ning is to under­stand the nat­u­ral his­
Optimal tim­ing of HCT in MF? tory of the dis­ ease based on best avail­­ able infor­ma­ tion on
Optimal tim­ing of HCT is not well defined, but cur­rent expert clin­i­cal and genetic risk fac­tors. This infor­ma­tion helps in under­
opin­ions rec­om­mend HCT to patients with MF who have Dynam- stand­ing the expected sur­vival and the like­li­hood of response
ic International Prognostic Scoring System (DIPSS) inter­me­di­ate- and the dura­bil­ity of nontransplant ther­apy. Patients with an

454  |  Hematology 2021  |  ASH Education Program


Table 1.  Identifying high-risk patients with sur­vival less than 5 years in mye­lo­fi­bro­sis

Risk strat­i­fi­ca­tion model Risk group Median OS, y Pros Cons


DIPSS14 Intermediate-2 4.0 Easy appli­ca­bil­ity based on Does not include any infor­ma­tion
clin­i­cal/lab­o­ra­tory var­i­ables on genetic var­i­ables that may
High 1.5
have sig­nif­i­cant impact on the
nat­u­ral his­tory of the dis­ease
DIPSS +15 Intermediate-2 2.9 Includes cytopenias and ­ No muta­tional data
cyto­ge­netic data
High 1.3
MIPSS70*16 High 3.1 Includes impact of MPN driver No cyto­ge­netic data
genes and HMR† genes
MIPSS70 + 2.0*17 High 4.1 Cytogenetic data
Driver/HMR‡ genes

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/453/1851800/453england.pdf by guest on 13 December 2021


Very high 1.8
MPN per­son­al­ized risk§18 TP53/–17p/–5/–5q 2.4 Combination of Study heavily weighted toward
clin­ical/genetic/ PV/ET patients
cyto­ge­netic data Copy num­ber var­i­a­tion not
included in most clin­i­cal NGS
reports
*http:​­/​­/www​­.mipss70score​­.it​­/.
†HMR (ASXL1, IDH1/2, EZH2, SRSF2).
‡Includes ASXL1, IDH1/2, EZH2, SRSF2; adds U2AF1 Q157.
§
https:​­/​­/www​­.sanger​­.ac​­.uk​­/science​­/tools​­/progmod​­/progmod​­/.
NGS, next-gen­er­a­tion sequenc­ing.

expected sur­vival of less than 5 years and/or less like­li­hood CALR, and MPL) sub­se­quently showed a more indo­lent course
of dura­ble response from nontransplant ther­apy are defined asso­ci­ated with the type 1 CALR muta­tion, whereas other mye­
as hav­ing high-risk MF (Table 1). We rec­om­mend the option of loid malig­nancy high molec­u­lar risk (HMR) muta­tions, includ­
upfront HCT in these patients as part of shared deci­sion mak­ing ing ASXL1, EZH2, IDH1/2, and SRSF2, were linked with poorer
incor­po­rat­ing patient fit­ness, val­ues, and pref­er­ences, not­ing prog­no­sis.26-28 Mutational data alone have been used as the
that many patients will choose to delay or forgo trans­plant fol­ basis for the Genetically Inspired Prognostic Scoring System,
low­ing coun­sel­ing.19,20 whereas inte­ gra­
tion of all­the above fac­ tors resulted in the
Mutation-Enhanced International Prognostic Scoring System
Evolution of risk assess­ment in MF (MIPSS70), which was fur­ ther refined to include cyto­ ge­ netic
Significant prog­ ress has been made in the past decade on risk in the MIPSS70 plus model.16,24 Mutations in the U2AF1 Q157
under­stand­ing the nat­u­ral his­tory of MF (Figure 2). Early prog­ hotspot were included as an addi­tional HMR in the MIPSS70 plus
nos­tic mod­els incor­po­rated only basic lab­o­ra­tory var­i­ables.21,22 v2.0 (http:​­/​­/www​­.mipss70score​­.it).17
Using clin­i­cal and lab­o­ra­tory param­e­ters, the International Using a large cohort of 2035 patients with MPN (1321 with
Working Group for Myelofibrosis Research and Treatment ET, 356 with PV, and 309 with pri­mary or sec­ond­ary MF and
devel­oped the International Prognostic Scoring System valid at 49 with other MPNs), Grinfeld et al18 iden­ti­fied 8 geno­mic sub­
the time of diag­no­sis of MF and fur­ther val­i­dated in the DIPSS groups with dis­tinct clin­i­cal and prog­nos­tic fea­tures. This was
appli­ca­ble at any time dur­ing the dis­ease course.14,23 Mayo Clinic used to develop a per­son­al­ized MPN risk cal­cu­la­tor that incor­po­
inves­ti­ga­tors refined this score by adding trans­fu­sion requir­ing rates clin­i­cal, cyto­ge­netic, and muta­tion data to pre­dict sur­vival
ane­mia, throm­bo­cy­to­pe­nia, and cyto­ge­net­ics lead­ing to the and leu­ke­mic trans­for­ma­tion (https:​­/​­/cancer​­.sanger​­.ac​­.uk​­/mpn​
DIPSS plus score.15 The impact of MPN driver muta­tions (JAK2, ­-multistage). A key obser­va­tion in this study was the poor sur­vival

Figure 2.  Timeline of prognostic risk models for MF and their components. Reproduced with permission from Davidson and Gupta.25
Lille,21 Cervantes,22 IPSS,23 DIPSS,14 DIPSS-plus,15 MIPSS70/MIPSS70 plus,16 GIPSS,24 MIPSS70 + v.2.0,17 and Personalized.18
Prakash Singh Shekhawat
Therapeutic land­scape for trans­plant eli­gi­ble MF | 455
Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/453/1851800/453england.pdf by guest on 13 December 2021
Figure 3.  Management algorithm for transplant-eligible patients with MF in the chronic phase.

in patients with mutated TP53, a rare yet influ­en­tial muta­tion in sig­nif­i­cant dis­ease pro­gres­sion, and symp­toms were ame­na­ble
chronic phase MF but one whose prog­nos­tic sig­nif­i­cance had to JAKi ther­apy; there­fore, we would delay the HCT for such a
not been pre­vi­ously described. patient until time of JAKi fail­ure.The patient in case 2 had high-risk
Mutation anal­y­sis in mye­loid neo­plasms is a rap­idly progress- genetic abnor­mal­i­ties, includ­ing an adverse cyto­ge­netic abnor­
ing field, and inter­ac­tions of var­i­ous subclonal pop­u­la­tions and mal­ity and 2 HMR muta­tions. Although the patient had no symp­
their down­stream effects on clin­i­cal out­comes are just begin­ning toms and was clin­i­cally well, there was high risk of dis­ease pro­
to be under­stood. The def­i­ni­tion of HMR muta­tions will likely gres­sion or leu­ke­mic trans­for­ma­tion. Therefore, we would refer
evolve fur­ther with inclu­sion of other can­di­date gene muta­tions, such a patient for con­sid­er­ation of upfront HCT.
such as the RAS path­way (NRAS, KRAS, and CBL) and NFE2, Our algo­rith­mic approach to use of HCT in trans­plant-eli­gi­ble
which have been shown to con­fer poorer prog­no­sis in MF.29,30 patients is sum­ma­rized in Figure 3.
Survival fol­ low­ing HCT has been shown to cor­ re­
late with
DIPSS cat­ eg
­ ory, but this does not include trans­ plant-spe­
cific
var­i­ables that may influ­ence clin­i­cal out­comes.7 The clin­i­cal- Nontransplant ther­a­pies in MF
molec­u­lar MF trans­plant scor­ing sys­tem, incor­po­rat­ing clin­i­cal Splenomegaly and con­sti­tu­tional symp­toms
data, donor type, and muta­ tion sta­tus for ASXL1/CALR/MPL, Although rec­om­men­da­tion for HCT is based on risk assess­ment,
pre­dicts over­all sur­vival (OS) and nonrelapse mor­tal­ity (NRM) in cur­rent med­i­cal ther­apy is directed at man­age­ment of symp­
pri­mary and sec­ond­ary MF.31 toms. Ruxolitinib has dem­ on­
strated improve­ments in spleno­
We strongly rec­om­mend that a phy­si­cian should use the max­ meg­aly, MF-related symp­tom bur­den, and health-related qual­ity
i­mal avail­­able infor­ma­tion on clin­i­cal, lab­o­ra­tory, and genetic of life.32,33 Initial tri­als did not include patients with lower IPSS
param­ et­ers depending on the access to the tests. If muta­ risk scores, and sub­se­quent stud­ies have shown symp­tom ben­
tional test­ing is avail­­able, we rec­om­mend using the MIPSS70 or e­fit from ruxolitinib in a lower-risk pop­u­la­tion.8 Rates of spleen
MIPSS70 plus v2.0 and oth­er­wise use the DIPSS. response cor­re­late with ruxolitinib dos­ing, and cytopenias are
the main dose-lim­it­ing tox­ic­ity. Other JAKi have been stud­ied
in phase 3 tri­als with fedratinib approved by the US Food and
Drug Administration for use in MF.34 Momelotinib and pacritinib
CLINICAL CASES (Con­t in­u ed) are other JAKi going through addi­tional phase 3 tri­als (Table 2)
In case 1, although the patient had a DIPSS score 3 (ie, intermediate- and may have dif­fer­en­ti­at­ing value for patients with ane­mia and
2 risk), the tempo of the dis­ease was indo­lent likely due to the severe throm­bo­cy­to­pe­nia, respec­tively. Factors that pre­dict a
type 1 CALR muta­tion with­out addi­tional somatic muta­tions or shorter dura­tion of response to front-line JAKi ther­apy include
cyto­ge­netic abnor­mal­i­ties reflected in a low MIPSS70 + v2.0 risk. higher DIPSS score, trans­fu­sion depen­dence, num­ber of muta­
Although symp­tom­atic, the patient was at rel­a­tively lower risk of tions, and ASXL1/EZH2 muta­tions spe­cif­i­cally.9,35

456  |  Hematology 2021  |  ASH Education Program


Table 2.  Ongoing phase 3 tri­als in mye­lo­fi­bro­sis

Class of investigational Population/key inclu­sion


Agent agent Trial n Phase 2 clin­i­cal ben­e­fit Primary out­come Key Secondary out­comes Comparator cri­te­ria
JAKi-naive patients
Pelabresib + BET inhib­i­tor MANIFEST-2 310 Spleen reduc­tion SVR ≥35% at 24 TSS at 24 weeks Placebo +
ruxolitinib Less ane­mia weeks ruxolitinib
BM fibro­sis reduc­tion
Navitoclax + BCL2 inhib­i­tor TRANSFORM-1 230 Spleen reduc­tion SVR ≥35% at 24 TSS at 24 weeks Placebo +
ruxolitinib weeks OS ruxolitinib
Reduction in BM fibro­sis
Parsaclisib + PI3Kδ inhib­i­tor LIMBER-313 440 Spleen reduc­tion SVR ≥35% at 24 TSS Placebo +
ruxolitinib weeks OS ruxolitinib
Luspatercept + ActRII ligand trap INDEPENDENCE 309 Transfusion inde­pen­dence/​ RBC trans­fu­sion Anemia improve­ment Placebo + JAKi Transfusion depen­dent
JAKi ane­mia improve­ment inde­pen­dence at Duration of ben­e­fit
24 weeks
Pacritinib JAKi PACIFICA 348 Spleen reduc­tion SVR ≥35% at 24 TSS Randomized 2:1 Platelets <50 000/µL
Better tol­er­a­bil­ity in weeks OS Physician selected Includes patients with prior
throm­bo­cy­to­pe­nic BAT JAKi expo­sure
patients
Jaktinib JAKi — 105 SVR ≥35% at 24 Transfusion depen­dence Hydroxyurea
weeks
Following first-line JAKi expo­sure or fail­ure
Momelotinib JAKi MOMENTUM 180 Anemia improve­ment TSS at 24 weeks Transfusion inde­pen­dence Randomized 2:1 Patients with ane­mia <100 g/L
Spleen response rate Danazol

Prakash Singh Shekhawat


Fedratinib JAKi FREEDOM-2 192 Spleen reduc­tion SVR ≥35% at 24 TSS Randomized 2:1
weeks OS BAT
Navitoclax + BCL2 inhib­i­tor TRANSFORM-2 330 Spleen reduc­tion SVR ≥35% at 24 TSS at 24 weeks BAT
ruxolitinib weeks OS
Reduction in BM fibro­sis
Parsaclisib + PI3Kδ inhib­i­tor LIMBER-304 212 Spleen reduc­tion SVR ≥35% 24 weeks TSS Placebo +
ruxolitinib OS ruxolitinib
Imetelstat Telomerase inhib­i­tor IMpactMF 320 Symptom score reduc­tion OS TSS at 24 weeks BAT
OS PFS
SVR ≥35% at 24 weeks
Reduction in BM fibro­sis
BAT, best avail­­able ther­apy; BCL2, B-cell lymphoma 2; BET, bromodomain and extraterminal; PFS, pro­gres­sion-free sur­vival; SVR, spleen vol­ume response; TSS, total symp­tom score.
Source: https:​­/​­/www​­.clinicaltrials​­.gov accessed on April 30, 2021.

Therapeutic land­scape for trans­plant eli­gi­ble MF | 457


Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/453/1851800/453england.pdf by guest on 13 December 2021
Deepening and prolonging the spleen and symp­ tom After 6 years, her spleen began to increase in size again, up
response is a pri­mary goal of many novel com­bi­na­tion strat­e­ to 15 cm BCM, with recur­rence of abdom­in ­ al pain and early sati­
gies adding addi­tive/syn­er­gis­tic agents to ruxolitinib in symp­ ety. The patient was started on fedratinib and had sig­nif­i­cant
tom­atic treat­ment-naive patients (Table 2). In addi­tion to the improve­ment in spleen size. Subsequently, this patient under­
con­sti­tu­tive acti­va­tion of JAK-STAT path­ways, there is evi­dence went 10/10 matched unre­lated donor (MUD) trans­plant using
that cyto­kine bur­den, dis­ease pro­gres­sion, and JAKi resis­tance reduced-inten­sity con­di­tion­ing and remains well after HCT.
can be affected through aber­rant acti­va­tion of other path­ways
that medi­ate cell pro­lif­er­a­tion, sur­vival, and cyto­kine sig­nal­
ing.1,4 Promising agents used in com­bi­na­tion with ruxolitinib
JAKi fail­ure
cur­ rently in phase 3 eval­ u­

tion are sum­ ma­
rized in Table 2.
Prior stud­ies eval­u­at­ing out­comes fol­low­ing dis­con­tin­u­a­tion of
Pelabresib is a bromodomain and extra-ter­mi­nal domain inhib­
ruxolitinib report a median sur­vival of 11 to 16 months (Table 3),
i­tor that reduces nuclear fac­tor–κB–medi­ated inflam­ma­tion.
with very short sur­vival in those with AP/BP trans­for­ma­tion.39-42
Navitoclax is a BCL-2/BCL-XL inhib­ i­
tor that reduces prosur-
Although infor­ma­tive, we note that in real-world prac­tice, many

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/453/1851800/453england.pdf by guest on 13 December 2021


vival sig­nal­ing and pro­motes apo­pto­sis in the malig­nant clone.
patients con­tinue on ruxolitinib despite clear evi­dence of dis­
Parsaclisib, a phosphatidylinositol 3-kinase (PI3K) δ inhib­it­or,
ease pro­gres­sion due to the lack of alter­na­tive med­i­cal ther­apy.
reduces abnor­mal PI3K/mTOR/AKT acti­va­tion that pro­motes
With the approval of fedratinib and avail­abil­ity of novel com­bi­na­
inflam­ma­tion and JAKi resis­tance. In phase 2 stud­ies, com­bi­na­
tion strat­e­gies as part of clin­i­cal tri­als (Table 2), early rec­og­ni­tion
tions of these agents with ruxolitinib have exhibited good rates
of JAKi ther­apy fail­ure and its pat­tern is of par­am ­ ount impor­tance
of spleen reduc­tion and symp­tom score improve­ment, with
for selecting appro­pri­ate options for these patients. We use the
pelabresib and navitoclax containing reg­i­mens dem­on­strat­ing
cri­te­ria pro­posed by the Cana­dian MPN Group and clas­sify the
evi­dence of reduced BM fibro­sis in some patients.4 Pelabresib,
JAKi fail­ure by categories such as loss of spleen/symp­tom re-
navitoclax, and parsaclisib com­ bi­
na­tions with ruxolitinib are
sponse, severe cytopenias, or pro­gres­sion to AP/BP.43
being com­pared with ruxolitinib monotherapy in treat­ment-
Due to poor prog­no­sis of JAKi fail­ure, we rec­om­mend HCT in
naive patients in the phase 3 MANIFEST-2, TRANSFORM-1, and
any patient with first-line JAKi fail­ure (Figure 3), an approach sup-
LIMBER-313 tri­als, respec­tively.
ported by lim­ited ret­ro­spec­tive data dem­on­strat­ing improved
sur­vival with HCT com­pared with sup­port­ive care.40
Cytopenias
Management of cytopenias is an unmet need in MF, both in
oth­er­wise asymp­ tom­ atic patients and in those whose JAKi Pretransplant JAKi ther­apy
treat­ment is com­pli­cated by ane­mia and throm­bo­cy­to­pe­nia. In patients referred for HCT, mas­ sive spleno­ meg­ aly has been
Supportive care relies on trans­fu­sion of red blood cells and asso­ci­ated with delayed count recov­ery and pos­si­bly worse OS.44
plate­lets, whereas phar­ma­co­logic ther­apy is lim­ited to danazol Limited evi­ dence is avail­­
able to define an appro­ pri­
ate tar­get
and eryth­ro­poi­e­sis-stim­u­lat­ing agents. Single-agent pelabresib spleen size, although sple­nec­tomy is con­sid­ered in patients with
is being inves­ti­gated for improv­ing ane­mia as monotherapy for mas­sive spleno­meg­aly that is refrac­tory to med­i­cal ther­apy.44,45
patients who do not have an indi­ca­tion for JAKi ther­apy.4 En- Use of peritransplant JAKi has sig­nif­i­cant inter­est given the
hancement of late-stage eryth­ro­cyte devel­op­ment through the poten­tial for both dis­ease con­trol and impact on GVHD (Table
inhi­bi­tion of transforming growth fac­tor β/SMAD sig­nal­ing with 4). Preliminary results from the JAK-ALLO trial raised safety
activin recep­tor ligand traps, such as luspatercept, has dem­on­ con­cerns with tumor lysis syn­drome and car­dio­genic shock,
strated effi­cacy in improv­ing ane­mia and reduc­ing trans­fu­sion, poten­tially due to cyto­kine flare fol­low­ing sud­den ces­sa­tion
and it will be eval­ua ­ ted fur­ther in the phase 3 INDEPENDENCE
study (Table 2).
Table 3.  Survival and pre­dic­tive fac­tors fol­low­ing ruxolitinib
Asymptomatic patients fail­ure/dis­con­tin­u­a­tion
There is cur­rently no med­ic ­ al ther­apy that has clear ben­e­fit in
asymp­tom­atic patients; we only con­sider it in the set­ting of a Median OS (95% CI)
fol­low­ing ruxolitinib Factors predicting lower
clin­i­cal trial for an agent with dis­ease-mod­i­fy­ing poten­tial. Im-
Reference fail­ure, mo sur­vival
mune ther­ ap­ ies, includ­
ing inter­feron agents, and antifibrotic
agents such as PRM-151 have shown some prom­ise in early stud­ Newberry 14 (10-18) Platelets <260 at ruxolitinib start
et al. (2017)39 Platelets <100 at dis­con­tin­u­a­tion
ies but require con­fir­ma­tion in con­trolled tri­als.4,36-38 Emergent muta­tions at
dis­con­tin­u­a­tion
Kuykendall 13 Lack of sal­vage ther­apy
et al. (2018)40
CLINICAL CASES (Con­t in­u ed) Mascarenhas 11.1 (8.4-14.5) Age >65 years
In case 1, the patient responded well to upfront ruxolitinib with et al. (2020)41 Charlson Comorbidity Index
res­o­lu­tion of symp­toms and reduc­tion in her spleen size to a na- Palandri et al. 13.2 (8.0-22.7) Blast phase at fail­ure
dir of 6 cm BCM. Such a patient could also be con­sid­ered for an (2020)42 Hb <100 g/L
Peripheral blasts >1%
upfront com­bi­na­tion ther­apy trial.

458  |  Hematology 2021  |  ASH Education Program


Table 4.  Prospective stud­ies of pre-HCT ruxolitinib ther­apy

Number of Number of Splenectomy SAE dur­ing


patients patients who prior to HCT, Ruxolitinib ruxolitinib
Reference recruited under­went HCT n (%) Taper ­dis­con­tin­u­a­tion taper Conditioning OS at 2 y, % NRM at 2 y, % GF, % GVHD
Gupta et al. (2019)47 21 19* 0 (0) 4 days 1 day prior to start None RIC (Flu-Bu) 63 28% 16 aGVHD grade 3-4, 16%
5 MSD of con­di­tion­ing cGVHD 76% at 2 y
14 UD
Salit et al. (2020)48 34 28† 3 (11) 5 mg every After 2-3 days of None Multiple 86 7% 0 aGVHD grade 3-4, 22%
14 MRD 3 days con­di­tion­ing cGVHD, 41%
11 UD ther­apy
3 UCB
Robin et al. (2021)46 64 59‡ 19 (30) 15 days (n = 17) 1 day prior to start TLS 3§ RIC (Flu-Mel) 55 46% 0 aGVHD grade 3-4, 44%
18 MSD of con­di­tion­ing RWS 3 23% MSD cGVHD 37% at 2 y
32 MUD Heart fail­ure 50% MUD
14 UD 9/10 77% UD 9/10
None (n = 42) 1 day prior to start Heart fail­ure
of con­di­tion­ing
*One patient with dis­ease pro­gres­sion to AML, 1 patient with sud­den death dur­ing ruxolitinib phase.
†Three patients with pro­gres­sion to AML dur­ing ruxolitinib phase, 3 patients not meet­ing cri­te­ria for trans­plant.
‡One donor with­drawal. One patient con­tin­ued tak­ing ruxolitinib, 3 patients died with­out HCT; 2 with heart fail­ure fol­low­ing sple­nec­tomy.
§No TLS fol­low­ing pro­to­col amend­ment to abrupt ruxolitinib dis­con­tin­u­a­tion.
aGVHD, acute GVHD; AML, acute meloid leukemia; cGVHD, chronic GVHD; Flu-Bu, fludarabine/busulfan; Flu-Mel, fludarabine-melphalan; GF, graft fail­ure; RIC, reduced-inten­sity con­di­tion­ing;
RWS, ruxolitinib with­drawal syn­drome; SAE, sig­nif­i­cant adverse event; TLS, tumor lysis syn­drome; UCB, umbilical cord blood.

Prakash Singh Shekhawat


Table 5.  HCT using alter­na­tive donors in mye­lo­fi­bro­sis

Reference Donor type n Conditioning GVHD ­pro­phy­laxis OS (95% CI) NRM (95% CI) Graft fail­ure GVHD
55
Raj et al. (2019) Mismatched related donor 56 MAC (70%)/RIC (30%) PTCy in 79% 56% (41%-70%) at 2 y 38% (24%-51%) at 2 y Primary 9% aGVHD* 28%
Secondary 13% cGVHD 45% at 1 y
Murata et al. (2019)56 Unrelated cord blood 13 — — 48% at 1 y 41% (22%-60%) at 1 y — aGVHD* 31%
27% at 4 y 62% (35%-81%) at 4 y cGVHD 15% at 1 y
Kunte et al. (2020)54 Haploidentical 58 NMA (52%)/RIC (45%) PTCy in 100% 69% (55%-80%) at 2 y 21% (11%-34%) at 2 y Primary 9% aGVHD* 44% at 6 m
cGVHD 31% at 2 y
*Acute GVHD encompasses grades II to IV.
MAC, myeloablative con­di­tion­ing; NMA, non-myeloablative; PTCy, posttransplant cyclo­phos­pha­mide.

Therapeutic land­scape for trans­plant eli­gi­ble MF | 459


Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/453/1851800/453england.pdf by guest on 13 December 2021
of ruxolitinib, neces­ si­
tat­
ing a pause on the study.46 How- Conclusion
ever, sub­se­quent tri­als do not sug­gest sub­stan­tial risk to this Advancements in both med­i­cal ther­apy and HCT opti­mi­za­tion
approach.47,48 The JAK-ALLO results dem­ on­strated improved have improved and broad­ened ther­a­peu­tic options for patients
safety, with no tumor lysis syn­drome or ruxolitinib with­drawal with MF. We rec­om­mend a risk-adapted strat­egy for tim­ing of
syn­drome, fol­low­ing pro­to­col amend­ment to an abrupt ruxoli- HCT and symp­tom-directed approach to med­i­cal ther­apy. These
tinib dis­con­tin­u­a­tion prior to con­di­tion­ing.46 The impact of pre- rec­ om­men­ da­tions should be incor­ po­rated as part of shared
transplant JAKi on posttransplant out­comes, includ­ing GHVD, deci­sion mak­ing encompassing patient pref­er­ences and val­ues
is not clear, with ret­ro­spec­tive stud­ies dem­on­strat­ing sim­i­lar to decide on an indi­vid­u­al­ized approach in each patient. Where
out­comes in those with or with­out prior JAKi ther­apy.49 We rec­ pos­si­ble, efforts should be made to enroll these patients in pro­
om­mend using JAKi ther­apy prior to HCT in patients with sig­nif­ spec­tive tri­als or reg­is­tries for bet­ter under­stand­ing of com­par­
i­cant MF-related symp­toms (Figure 3), with a taper­ing sched­ule a­tive out­comes.
lead­ing up to, or a few days after, the start of con­di­tion­ing to
avoid any with­drawal symp­toms.46,47,50 Acknowledgments

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/453/1851800/453england.pdf by guest on 13 December 2021


This pro­ject was supported by a grant from the Elizabeth and
Tony Comper Foundation and the Princess Margaret Cancer
CLINICAL CASES (Con­t in­u ed) Centre Foundation (V.G.).
In case 2, the patient was referred for trans­plant, but no fully
matched sib­ ling donors (MSDs) or unre­ lated donors (UDs) Conflict-of-inter­est dis­clo­sure
could be iden­ti­fied. The patient pre­ferred not to pur­sue hap- James England: No com­pet­ing finan­cial inter­ests to declare.
loidentical trans­ plant ini­
tially. However, there was increase Vikas Gupta: Consultancy and advi­sory board: Novartis, BMS-
in blast counts in PB to 8% within 6 months, and the patient Celgene, Sierra Oncology, Pfizer, Abb Vie, Roche.
opted for haploidentical HCT with his son as a donor at that
time point. Off-label drug use
James England: Off-label use of ruxolitinib in the peri-transplant
period is discussed in the article.
Role of alter­na­tive donor trans­plant in MF Vikas Gupta: Off-label use of ruxolitinib in the peri-transplant pe-
HCT from a matched sib­ling or unre­lated donor is the pre­ferred riod is discussed in the article.
source for HCT in MF, with many early stud­ies dem­on­strat­ing
infe­rior out­comes using a mismatched unre­lated donor.51,52 Re- Correspondence
cent reporting trends from the Center for International Blood Vikas Gupta, Princess Margaret Cancer Centre, University of
and Marrow Transplant Research show slowly increas­ing adap­ Toronto, 700 University Avenue, 6-326, Toronto, Ontario, M5G
tion in the use of haploidentical donors in MF as advance­ments 1Z5 Canada; e-mail: vikas​­.gupta@uhn​­.ca.
in sup­port­ive care and the pre­ven­tion of GVHD, includ­ing the
use of posttransplant cyclo­phos­pha­mide, have allowed for im- References
1. Koschmieder S, Chatain N. Role of inflam­ma­tion in the biol­ogy of mye­lo­pro­
proved safety (Table 5).25 A major lim­i­ta­tion of all­these stud­ies
lif­er­a­tive neo­plasms. Blood Rev. 2020;42(suppl 1):100711.
is small sam­ple size. Bregante et al53 report the increas­ing use 2. Harrison CN, Vannucchi AM, Kiladjian J-J, et  al. Long-term find­ings from
of haploidentical donors from 2011 to 2014 com­pared with 2000 COMFORT-II, a phase 3 study of ruxolitinib vs best avail­­able ther­apy for
to 2010, with sim­i­lar out­comes com­pared with MSDs among the mye­lo­fi­bro­sis. Leukemia. 2016;30(8):1701-1707.
alter­na­tive donor group (77% haploidentical) in the later time 3. Verstovsek S, Mesa RA, Gotlib J, et al; COMFORT-I inves­ti­ga­tors. Long-
term treat­ ment with ruxolitinib for patients with mye­ lo­
fi­
bro­
sis: 5-year
period. Initial results from Kunte et al54 also show encour­ag­ing update from the ran­dom­ized, dou­ble-blind, pla­cebo-con­trolled, phase 3
out­comes in a ret­ro­spec­tive cohort of haploidentical HCT with COMFORT-I trial. J Hematol Oncol. 2017;10(1):55.
posttransplant cyclo­ phos­ pha­
mide. Data fol­ low­ing the use of 4. Bankar A, Gupta V. Investigational non-JAK inhib­i­tors for chronic phase
related donors with 2 or more anti­gen mis­matches have been mye­lo­fi­bro­sis. Expert Opin Investig Drugs. 2020;29(5):461-474.
5. Gupta V, Hari P, Hoffman R. Allogeneic hema­to­poi­etic cell trans­plan­ta­
reported from the Euro­ pean Society for Blood and Marrow
tion for mye­lo­fi­bro­sis in the era of JAK inhib­i­tors. Blood. 2012;120(7):1367-
Transplantation reg­is­try dem­on­strat­ing rea­son­able 2-year OS 1379.
(56%) and GVHD rates but higher rates of graft fail­ure (22%) and 6. Kröger NM, Deeg JH, Olavarria E, et  al. Indication and man­age­ment of
NRM at 2 years (38%).55 Use of unre­lated cord blood also pres­ allo­ge­neic stem cell trans­plan­ta­tion in pri­mary mye­lo­fi­bro­sis: a con­sen­
sus pro­ cess by an EBMT/ELN inter­ na­tional work­ ing group. Leukemia.
ents sim­i­lar con­cerns, with a report from the Japan Society for
2015;29(11):2126-2133.
Hematopoietic Cell Transplantation dem­on­strat­ing lower rates 7. Gowin K, Ballen K, Ahn KW, et al. Survival fol­low­ing allo­ge­neic trans­plant in
of engraft­ment, high NRM, and only 27% 4-year OS.56 Limited patients with mye­lo­fi­bro­sis. Blood Adv. 2020;4(9):1965-1973.
data on haploidentical trans­plants sup­port the poten­tial use in 8. Gupta V, Griesshammer M, Martino B, et  al. Analysis of pre­ dic­tors of
selected patients, although many patients may opt for med­i­ response to ruxolitinib in patients with mye­ lo­
fib
­ ro­
sis in the phase 3b
expanded-access JUMP study. Leuk Lymphoma. 2021;62(4):918-926.
cal ther­apy when coun­seled on the added risks asso­ci­ated with 9. Spiegel JY, McNamara C, Kennedy JA, et al. Impact of geno­mic alter­ations
alter­na­tive donors.15 Ongoing large stud­ies through the Center on out­comes in mye­lo­fi­bro­sis patients under­go­ing JAK1/2 inhib­­i­tor ther­
for International Blood and Marrow Transplant Research com­ apy. Blood Adv. 2017;1(20):1729-1738.
par­ing out­comes of haploidentical trans­plants vs MUDs and mis- 10. Kröger N, Giorgino T, Scott BL, et al. Impact of allo­ge­neic stem cell trans­
plan­ta­tion on sur­vival of patients less than 65 years of age with pri­mary
matched donors should fur­ther clar­ify and sup­port the posi­tion­ mye­lo­fi­bro­sis. Blood. 2015;125(21):3347-3350, quiz 3364.
ing of haploidentical trans­plants in the man­age­ment of MF (Tania 11. McNamara C, Spiegel J, Xu W, et  al. Outcomes fol­ low­ ing fail­
ure of
Jain, Johns Hopkins University, per­sonal com­mu­ni­ca­tion, 2021). JAK1/2 inhib­i­tor ther­apy in patients with mye­lo­fi­bro­sis is depen­dent

460  |  Hematology 2021  |  ASH Education Program


on the pat­tern of fail­ure. Clin Lymphoma Myeloma Leuk. 2019;19(suppl 34. Mullally A, Hood J, Harrison C, Mesa R. Fedratinib in mye­lo­fi­bro­sis. Blood
1):S351–S352. Adv. 2020;4(8):1792-1800.
12. Gupta V, Kennedy JA, Capo-Chichi J-M, et al. Genetic fac­tors rather than 35. Patel KP, Newberry KJ, Luthra R, et al. Correlation of muta­tion pro­file and
blast reduc­ tion deter­ mine out­ comes of allo­ ge­
neic HCT in BCR-ABL-­​ response in patients with mye­lo­fi­bro­sis treated with ruxolitinib. Blood.
neg­a­tive MPN in blast phase. Blood Adv. 2020;4(21):5562-5573. 2015;126(6):790-797.
13. Gupta V, Kim S, Hu Z-H, et  al. Comparison of out­comes of HCT in blast 36. Sørensen AL, Mikkelsen SU, Knudsen TA, et al. Ruxolitinib and inter­feron-α2
phase of BCR-ABL1-MPN with de novo AML and with AML fol­low­ing MDS. com­bi­na­tion ther­apy for patients with poly­cy­the­mia vera or mye­lo­fi­bro­sis:
Blood Adv. 2020;4(19):4748-4757. a phase II study. Haematologica. 2020;105(9):2262-2272.
14. Passamonti F, Cervantes F, Vannucchi AM, et  al. A dynamic prog­nos­tic 37. Kiladjian J-J, Soret-Dulphy J, Resche-Rigon M, et al. Ruxopeg, a multi-cen­
model to pre­dict sur­vival in pri­mary mye­lo­fi­bro­sis: a study by the IWG-MRT ter Bayes­ian phase 1/2 adap­tive ran­dom­ized trial of the com­bi­na­tion of
(International Working Group for Myeloproliferative Neoplasms Research ruxolitinib and pegylated inter­feron alpha 2a in patients with mye­lo­pro­lif­
and Treatment). Blood. 2010;115(9):1703-1708. er­a­tive neo­plasm (MPN)–asso­ci­ated mye­lo­fi­bro­sis. Blood. 2018;132(suppl
15. Gangat N, Caramazza D, Vaidya R, et  al. DIPSS plus: a refined Dynamic 1):581.
International Prognostic Scoring System for pri­ mary mye­ lo­
fi­
bro­sis that 38. Verstovsek S, Talpaz M, Wadleigh M, et  al. S828: a ran­dom­ized, dou­ble
incor­po­rates prog­nos­tic infor­ma­tion from kar­yo­type, plate­let count, and blind phase 2 study of 3 dif­fer­ent doses of PRM-151 in patients with mye­
trans­fu­sion sta­tus. J Clin Oncol. 2011;29(4):392-397. lo­fi­bro­sis who were pre­vi­ously treated with or inel­i­gi­ble for ruxolitinib.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/453/1851800/453england.pdf by guest on 13 December 2021


16. Guglielmelli P, Lasho TL, Rotunno G, et  al. MIPSS70: muta­tion-enhanced HemaSphere. 2019;3(suppl 1):367.
inter­na­tional prog­nos­tic score sys­tem for trans­plan­ta­tion-age patients 39. Newberry KJ, Patel K, Masarova L, et al. Clonal evo­lu­tion and out­comes
with pri­mary mye­lo­fi­bro­sis. J Clin Oncol. 2018;36(4):310-318. in mye­lo­fi­bro­sis after ruxolitinib dis­con­tin­u­a­tion. Blood. 2017;130(9):1125-
17. Tefferi A, Guglielmelli P, Lasho TL, et al. MIPSS70 + ver­sion 2.0: muta­tion and 1131.
kar­yo­type-enhanced inter­na­tional prog­nos­tic scor­ing sys­tem for pri­mary 40. Kuykendall AT, Shah S, Talati C, et al. Between a rux and a hard place: eval­u­
mye­lo­fi­bro­sis. J Clin Oncol. 2018;36(17):1769-1770. at­ing sal­vage treat­ment and out­comes in mye­lo­fi­bro­sis after ruxolitinib dis­
18. Grinfeld J, Nangalia J, Baxter EJ, et al. Classification and per­son­al­ized prog­ con­tin­u­a­tion. Ann Hematol. 2018;97(3):435-441.
no­sis in mye­lo­pro­lif­er­a­tive neo­plasms. N Engl J Med. 2018;379(15):1416– 41. Mascarenhas J, Mehra M, He J, Potluri R, Loefgren C. Patient char­ac­ter­is­
1430. tics and out­comes after ruxolitinib dis­con­tin­u­a­tion in patients with mye­
19. Walker AR. How to approach shared deci­sion mak­ing when deter­min­ing lo­fi­bro­sis. J Med Econ. 2020;23(7):721-727.
con­sol­i­da­tion, main­te­nance ther­apy, and trans­plan­ta­tion in acute mye­loid 42. Palandri F, Breccia M, Bonifacio M, et al. Life after ruxolitinib: rea­sons for dis­
leu­ke­mia. Hematology Am Soc Hematol Educ Program. 2020;2020(1):51– con­tin­u­a­tion, impact of dis­ease phase, and out­comes in 218 patients with
56. mye­lo­fi­bro­sis. Cancer. 2020;126(6):1243-1252.
20. Smith E, Huang J, Viswabandya A, et  al. Association of fac­tors influ­enc­ 43. Gupta V, Cerquozzi S, Foltz L, et  al. Patterns of ruxolitinib ther­apy fail­
ing selec­tion of upfront hema­to­poi­etic cell trans­plan­ta­tion ver­sus non- ure and its man­age­ment in mye­lo­fi­bro­sis: per­spec­tives of the Cana­dian
trans­plant ther­a­pies in mye­lo­fi­bro­sis: trans­plant deci­sions in MF. Transplant Myeloproliferative Neoplasm Group. JCO Oncol Pract. 2020;16(7):351-
Cell Ther. 2021;27(7):600.e1-600.e8. 359.
21. Dupriez B, Morel P, Demory JL, et al. Prognostic fac­tors in agnogenic mye­ 44. Malato A, Rossi E, Tiribelli M, Mendicino F, Pugliese N. Splenectomy in
loid meta­pla­sia: a report on 195 cases with a new scor­ing sys­tem. Blood. mye­lo­fi­bro­sis: indi­ca­tions, effi­cacy, and com­pli­ca­tions. Clin Lymphoma
1996;88(3):1013-1018. Myeloma Leuk. 2020;20(9):588-595.
22. Cervantes F, Barosi G, Demory JL, et  al. Myelofibrosis with mye­loid 45. Polverelli N, Mauff K, Kröger N, et al. Impact of spleen size and sple­nec­
meta­pla­sia in young indi­vid­u­als: dis­ease char­ac­ter­is­tics, prog­nos­tic tomy on out­comes of allo­ge­neic hema­to­poi­etic cell trans­plan­ta­tion
fac­tors and iden­ti­fi­ca­tion of risk groups. Br J Haematol. 1998;102(3): for mye­lo­fi­bro­sis: a ret­ro­spec­tive anal­y­sis by the chronic malig­nan­cies
684-690. work­
­ ing party on behalf of Euro­ pean soci­ ety for blood and mar­ row
23. Cervantes F, Dupriez B, Pereira A, et al. New prog­nos­tic scor­ing sys­tem trans­plan­ta­tion (EBMT). Am J Hematol. 2021;96(1):69-79.
for pri­mary mye­lo­fi­bro­sis based on a study of the International Working 46. Robin M, Porcher R, Orvain C, et al. Ruxolitinib before allo­ge­neic hema­to­
Group for Myelofibrosis Research and Treatment. Blood. 2009;113(13): poi­etic trans­plan­ta­tion in patients with mye­lo­fi­bro­sis on behalf SFGM-TC
2895-2901. and FIM groups. Bone Marrow Transplant. 2021;56(8):1-12.
24. Tefferi A, Guglielmelli P, Nicolosi M, et al. GIPSS: genet­i­cally inspired prog­ 47. Gupta V, Kosiorek HE, Mead A, et  al. Ruxolitinib ther­ apy followed by
nos­tic scor­ing sys­tem for pri­mary mye­lo­fi­bro­sis. Leukemia. 2018;32(7):1631– reduced-inten­sity con­di­tion­ing for hema­to­poi­etic cell trans­plan­ta­tion for
1642. mye­lo­fi­bro­sis: mye­lo­pro­lif­er­a­tive dis­or­ders research Consortium 114 study.
25. Davidson MB, Gupta V. Application of stem cell ther­apy in mye­lo­fi­bro­sis. Biol Blood Marrow Transplant. 2019;25(2):256-264.
Hematol Oncol Clin North Am. 2021;35(2):391-407. 48. Salit RB, Scott BL, Stevens EA, Baker KK, Gooley TA, Deeg HJ. Pre-
26. Guglielmelli P, Rotunno G, Fanelli T, et  al. Validation of the dif­fer­en­tial hema­ to­poi­etic cell trans­ plant ruxolitinib in patients with pri­ mary
prog­nos­tic impact of type 1/type 1-like ver­sus type 2/type 2-like CALR and sec­ond­ary mye­lo­fi­bro­sis. Bone Marrow Transplant. 2020;55(1):
muta­tions in mye­lo­fi­bro­sis. Blood Cancer J. 2015;5(10):e360. 70-76.
27. Vannucchi AM, Lasho TL, Guglielmelli P, et al. Mutations and prog­no­sis in 49. Shahnaz Syed Abd Kadir S, Christopeit M, Wulf G, et al. Impact of ruxoli-
pri­mary mye­lo­fi­bro­sis. Leukemia. 2013;27(9):1861-1869. tinib pre­treat­ment on out­comes after allo­ge­neic stem cell trans­plan­ta­tion
28. Rumi E, Pietra D, Pascutto C, et al; Associazione Italiana per la Ricerca sul in patients with mye­lo­fi­bro­sis. Eur J Haematol. 2018;101(3):305-317.
Cancro Gruppo Italiano Malattie Mieloproliferative Investigators. Clinical 50. Shanavas M, Popat U, Michaelis LC, et al. Outcomes of allo­ge­neic hema­
effect of driver muta­tions of JAK2, CALR, or MPL in pri­mary mye­lo­fi­bro­sis. to­poi­etic cell trans­plan­ta­tion in patients with mye­lo­fi­bro­sis with prior
Blood. 2014;124(7):1062-1069. expo­ sure to Janus kinase ½ inhib­ i­tors. Biol Blood Marrow Transplant.
29. Coltro G, Rotunno G, Mannelli L, et  al. RAS/CBL muta­tions pre­dict resis­ 2016;22(3):432-440.
tance to JAK inhib­i­tors in mye­lo­fi­bro­sis and are asso­ci­ated with poor prog­ 51. Kröger N, Holler E, Kobbe G, et  al. Allogeneic stem cell trans­plan­ta­tion
nos­tic fea­tures. Blood Adv. 2020;4(15):3677-3687. after reduced-inten­sity con­di­tion­ing in patients with mye­lo­fi­bro­sis: a
30. Marcault C, Zhao LP, Maslah N, et  al. Impact of NFE2 muta­tions on AML pro­spec­tive, mul­ti­cen­ter study of the Chronic Leukemia Working Party
trans­for­ma­tion and over­all sur­vival in patients with mye­lo­pro­lif­er­a­tive neo­ of the Euro­ pean Group for Blood and Marrow Transplantation. Blood.
plasms (MPN) [published online 4 May 2021]. Blood. 2009;114(26):5264-5270.
31. Gagelmann N, Ditschkowski M, Bogdanov R, et al. Comprehensive clin­i­cal- 52. Gupta V, Malone AK, Hari PN, et al. Reduced-inten­sity hema­to­poi­etic cell
molec­u­lar trans­plant scor­ing sys­tem for mye­lo­fi­bro­sis under­go­ing stem trans­plan­ta­tion for patients with pri­mary mye­lo­fi­bro­sis: a cohort anal­y­sis
cell trans­plan­ta­tion. Blood. 2019;133(20):2233-2242. from the Center for International Blood and Marrow Transplant Research.
32. Harrison C, Kiladjian J-J, Al-Ali H-K, et  al. JAK inhi­bi­tion with ruxolitinib Biol Blood Marrow Transplant. 2014;20(1):89-97.
ver­sus best avail­­able ther­apy for mye­lo­fi­bro­sis. N Engl J Med. 2012;366(9): 53. Bregante S, Dominietto A, Ghiso A, et  al. Improved out­come of alter­na­
787–798. tive donor trans­plan­ta­tions in patients with mye­lo­fi­bro­sis: from unre­
33. Verstovsek S, Mesa RA, Gotlib J, et al. A dou­ble-blind, pla­cebo-con­trolled lated to haploidentical fam­ ily donors. Biol Blood Marrow Transplant.
trial of ruxolitinib for mye­lo­fi­bro­sis. N Engl J Med. 2012;366(9):799-807. 2016;22(2):324-329.

Prakash Singh Shekhawat


Therapeutic land­scape for trans­plant eli­gi­ble MF | 461
54. Kunte SJ, Rybicki L, Viswabandya A, et  al. Haploidentical allo­ ge­
neic 56. Murata M, Takenaka K, Uchida N, et al. Comparison of out­comes of allo­
hema­to­poi­etic cell trans­plan­ta­tion with post-trans­plant cyclo­phos­pha­mide ge­neic trans­plan­ta­tion for pri­mary mye­lo­fi­bro­sis among hema­to­poi­etic
in patients with mye­lo­fi­bro­sis: a multi-insti­tu­tional expe­ri­ence. Blood. stem cell source groups. Biol Blood Marrow Transplant. 2019;25(8):1536-
2020;136(suppl 1):­33-34. 1543.
55. Raj K, Eikema D-J, McLornan D-P, et al. Family mismatched allo­ge­neic stem
cell trans­plan­ta­tion for mye­lo­fi­bro­sis: report from the Chronic Malignancies
Working Party of Euro­pean Society for Blood and Marrow Transplantation. © 2021 by The Amer­i­can Society of Hematology
Biol Blood Marrow Transplant. 2019;25(3):522-528. DOI 10.1182/hema­tol­ogy.2021000279

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/453/1851800/453england.pdf by guest on 13 December 2021

462  |  Hematology 2021  |  ASH Education Program


MPN: NEW DIRECTIONS

Running interferon interference in treating


PV/ET: meeting unmet needs

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/463/1852047/463traxler.pdf by guest on 13 December 2021


Elizabeth A. Traxler and Elizabeth O. Hexner
Department of Medicine and Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA

Enthusiasm about interferons for the treatment of myeloproliferative neoplasms has recently arisen. How does a non-
targeted therapy selectively target the malignant clone? Many foundational questions about interferon treatment are
unanswered, including who, when, and for how long do we treat. Using an individual case, this review touches on gaps
in risk assessment in polycythemia vera (PV) and essential thrombocythemia (ET) and the history of treatment with inter-
ferons. How is it that this proinflammatory cytokine effectively treats ET and PV, themselves proinflammatory states? We
summarize existing mechanistic and clinical data, the molecular context as a modifier for treatment response, the estab-
lishment of treatment goals, and the challenges that lie ahead.

LEARNING OBJECTIVES
• Define treatment goals in PV/ET
• Summarize molecular responses using IFN in PV/ET

overall prognosis of her disease and wonders about the


CLINICAL CASE safety of future pregnancies, which she desires.
A 32­year­old woman with at least 2 years of blood count
abnormalities is referred to hematology. Two years prior,
early in a complicated pregnancy, she had an abnormal Introduction
complete blood count: white blood cell count, 12.7 thou/µL; Our patient has PV, and her experience is common to many
hemoglobin, 16g/dL; mean corpuscular volume, 82 fL; and when diagnosed: long­standing but nonspecific symptoms
platelets, 415 thou/µL. She was admitted with preeclamp­ and a lag from the time of blood count abnormalities to con­
sia 25 weeks into her pregnancy and was closely observed, firmation of diagnosis. More pernicious is her experience in
treated with low­dose aspirin, and delivered at 32 weeks. pregnancy, fraught with complications that are too com­
Her newborn daughter was born small for gestational age. mon in women with PV, including the overlooked real­time
When first evaluated by hematology 19 months later, she diagnosis. Nevertheless, by standard vascular risk stratifi­
described chronic itching for 7 to 8 years, worse after a cation criteria, our patient is considered to have low­risk
shower and absent during pregnancy but present again disease, a designation of little comfort to many young peo­
and mild and manageable. She had no visual symptoms or ple with myeloproliferative neoplasms (MPN). How can we
headaches and no history of clots. Medications included best address her immediate concerns related to thriving as
an oral contraceptive. A repeat complete blood count a “thirty­something,” the intermediate goal of growing her
showed persistent abnormalities: WBC, 11.3 tho/µL; hemo­ family, and, finally, the long­term and often dominant con­
globin, 17.9g/dL; mean corpuscular volume, 76 fL; platelets, cern related to progression of disease and early mortality?
692 tho/µL. Additional testing, including JAK2V617F muta­ Many are hopeful that interferons (IFNs), especially long­
tion analysis, detected a mutation at an allele frequency acting formulations, might address such patient concerns.
of 32.6%. Serum ferritin was 7ng/mL, and serum erythro­
poietin was 2 mIU/mL (range, 3–19 mIU/mL), confirming a MPN as inflammatory diseases
diagnosis of polycythemia vera (PV). While her symptoms Classical MPN without the Philadelphia­negative chromo­
now feel mild and manageable, she is concerned about the some are clonal diseases classified into 3 subtypes: PV,
Prakash Singh Shekhawat
IFN in PV/ET | 463
essen­tial thrombocythemia (ET), and pri­mary mye­lo­fi­bro­sis (PMF). spec­tive stud­ies of peg-IFN can help elu­ci­date the mech­a­nisms
They are clin­i­cally char­ac­ter­ized by pro­lif­er­a­tive bone mar­row, most rel­e­vant for opti­mal response.
abnor­mal blood counts, spleno­meg­aly, and con­sti­tu­tional symp­
toms, along with a long-term higher risk of sec­ond­ary bone mar­ Goals of ther­apy and risk strat­i­fi­ca­tion
row fibro­ sis and acute leu­ ke­mic trans­for­
ma­ tion. In 2005 the 9 The goals of treating patients with MPN include ame­lio­ra­tion
onco­genic driver muta­tion was iden­ti­fied and found to be pres­ent of symp­toms, reduc­tion in throm­bo­sis risk, con­trol of abnor­mal
in nearly all­patients with PV and about 50% to 60% of patients blood counts, reduc­tion in long-term con­se­quences, and, hope­
with ET or PMF. Activating muta­tions in thrombopoietin recep­ fully, delay of dis­ease pro­gres­sion. In addi­tion to an often dom­i­
tor (MPL) and calreticulin (CALR) account for the large major­ity of nant con­cern for pro­gres­sion to advanced mye­lo­fi­bro­sis and/or
remaining cases. These driver muta­tions func­tion­ally con­verge in a acute leu­ke­mia, addi­tional con­se­quences of MPN ther­a­pies can
com­mon path­way, resulting in aber­rant JAK2-STAT sig­nal­ing. accu­mu­late over time; thus, thought­ful treat­ment rec­om­men­da­
The genetic driv­ers of MPN con­verge, with grow­ing evi­dence tions are cru­cial.
that proinflammatory pro­cesses are cen­tral to MPN patho­bi­­ol­ogy. Therapeutic phle­bot­omy (TP) alone was found to be supe­rior

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/463/1852047/463traxler.pdf by guest on 13 December 2021


MPN are acquired dis­eases of stem cells, and the JAK2 path­way is to alkylating agents in one of the early piv­otal stud­ies of inter­
con­sid­ered an impor­tant inflam­ma­tory driver. Patients with MPN ven­tions for PV, is the back­bone of treat­ment in newly diag­nosed
have increased lev­els of inflam­ma­tory cyto­kines, dysregulation of PV, and per­sists as an impor­tant Hip­po­cratic reminder that sim­
immune-related genes, and enhanced oxi­da­tive stress.1,2 As of the ple can be bet­ter.8 TP is par­tic­u­larly crit­i­cal in the acute set­ting
sum­mer of 2021, JAK inhib­i­tors remain the only approved med­i­ at diag­no­sis when signs or symp­toms of hyper­vis­cos­ity are pres­
ca­tions in the United States for PV and PMF, and cen­tral to their ent. A more recent foun­da­tional study, CYTO-PV, con­firmed that
activ­ity is the abil­ity to mod­u­late cyto­kine pro­files and improve hemat­oc ­ rit con­trol below 45%, by means of TP or cytoreduc­
symp­ toms. Despite off-label use, IFNs predated JAK inhib­i­tors tive med­i­ca­tions, reduced the risk of vas­cu­lar events by approx­
for the treat­ment of MPN. IFNs have been con­sid­ered a stan­dard i­ma­tely 4-fold.9 But TP only con­trols red blood cell count, not
treat­ment for decades, predating even the iden­ti­fi­ca­tion of the plate­lets and white blood cells, so pro­lif­er­at­ive dis­ease is only
driver muta­tions. But we are now left with a cen­tral par­a­dox of par­tially addressed with this sat­is­fy­ingly sim­ple and safe inter­
treating a proinflammatory state with a proinflammatory cyto­kine. ven­tion. In addi­tion, iron defi­ciency—some­times pres­ent even
Optimally, the expe­ri­ence of IFN use and mech­a­nis­tic pre­clin­i­cal prior to phle­bot­omy ini­ti­at­ion in PV patients such as ours—is
and clin­ic­ al data will lead us on a path to res­o­lu­tion of this par­a­ induced or wors­ened by TP, which often drives plate­let counts
dox and, more impor­tantly, improved out­comes for our patients. higher still and in aggre­gate may cause or exac­er­bate micro­vas­
cu­lar or neurocognitive symp­toms. PegIFN, by con­trast, induces
History of IFN use as it relates to the treat­ment trilineage blood cell count low­er­ing, and when responses are
of PV and ET seen, it can offer the pos­si­bil­ity of maintaining blood count con­
Let us now con­sider treating our patient. Recombinant IFN-al­ trol while still correcting iron defi­ciency, thus interrupting the
pha has been a treat­ment option for patients with hema­to­logic vicious TP-iron defi­ciency-thrombocytosis cycle (Figure 1).
malig­nan­cies for decades and was first used to treat MPN over Comparative effi­cacy data to guide the selec­tion of ini­tial
30 years ago. In the pre–tyro­sine kinase era, IFN was ­able to cytoreductive ther­apy in PV and ET are lim­ited, and ther­apy is
induce deep remis­sions in a small sub­set of patients with chronic influ­enced by con­ve­nience, cost, tol­er­a­bil­ity, and safety. PegIFN
mye­loid leu­ke­mia, while con­ven­tional chemotherapies failed.3 In is not cur­rently approved in the US for treat­ment of ET or PV, yet
Philadelphia chro­mo­some-neg­a­tive MPN, an early study reported con­sen­sus guide­lines rec­om­mend its con­sid­er­ation as a first-line
on 12 patients with improve­ment in thrombocytosis with 2 to 10 option for cytoreduction, par­tic­u­larly in youn­ger patients and
weeks of recom­ bi­
nant IFN.4,5 Despite other early tri­ als show­ those who desire preg­nancy.10
ing prom­is­ing responses, the side effects with IFN forced unac­
cept­able dis­con­tin­u­a­tion rates, and con­se­quently, its use was Efficacy of pegIFN
not widely adopted. MPN patients can expe­ri­ence debil­i­tat­ing Hematologic response
con­ sti­
tu­
tional symp­ toms, felt to be cyto­ kine driven, mak­ ing In tri­als using stan­dard for­mu­la­tions of recom­bi­nant IFN, hema­to­
expected but overlapping IFN-related side effects poten­ tially logic responses were observed in about 80% of patients, who also
even less well tol­er­ated in this pop­u­la­tion. The devel­op­ment of reported a sig­nif­i­cant reduc­tion in MPN-asso­ci­ated symp­toms,
pegylated IFNs (pegIFNs), a lon­ger-act­ing for­mu­la­tion that is bet­ includ­ing pruritus. More recent stud­ies with lon­ger-act­ing peg-
ter tol­er­ated and requires less fre­quent dos­ing, renewed inter­ IFN have reported a sim­i­lar effect size. Two inde­pen­dent phase
est in this treat­ment approach. More recent stud­ies, sum­ma­rized 2 stud­ies using peginterferon alfa-2a in PV and ET both dem­on­
here, have shown prom­is­ing responses in hema­to­logic abnor­mal­ strated effec­tive cytoreduction with less dis­con­tin­u­a­tion due to
i­ties, symp­toms, and anticlonal activ­ity with the use of pegIFN. tox­ic­ity. The French “PV-Nord” group (PVN1) conducted a study of
The exact action of IFN in MPN remains an area of intense 37 patients with newly diag­nosed PV and boasted count nor­mal­i­
inves­ti­ga­tion. Supported mech­a­nisms include direct cyto­toxic za­tion in 95% of the cohort at 12 months.11 Testifying to the tol­er­a­
effects on the malig­nant clonal cells in addi­tion to enhanc­ing the bil­ity of this for­mu­la­tion, the vast major­ity of patients remained on
host innate and adap­tive immune response. IFN may increase the treat­ment for at least 1 year. Surprisingly, a great major­ity of
the expres­sion of neoantigens on malig­nant clonal cells, ren­der­ these responses were dura­ble at 6-year fol­low-up, even after stop­
ing them more sus­cep­ti­ble to immunosurveillance,6 and it can ping or switching ther­a­pies.12 In a sep­a­rate study of patients with
also induce emer­gence of the hema­to­poi­etic stem cell from qui­ both PV and ET, response rates were around 80% for both dis­
es­cence to allow for apo­pto­sis (exten­sively reviewed in Kiladjian, ease sub­types.13 Noncomparative stud­ies have defined the effec­
Mesa, and Hoffman et al).7 Ideally, cor­rel­a­tive sci­ence from pro­ tive­ness of pegIFN in hydroxy­urea (HU) resis­tance/intol­er­ance.14

464  |  Hematology 2021  |  ASH Education Program


Polycythemia vera
with hematocrit
above goal

Cytoreductive therapy

Therapeutic
phlebotomy
Progressive
microvascular
symptoms

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/463/1852047/463traxler.pdf by guest on 13 December 2021


Iron deficiency

Reactive
thrombocytosis

Figure 1. Vicious cycle: iron deficiency, polycythemia vera, and thrombocytosis.

While these have both gar­ nered sup­ port for their effi­
cacy in lar response. Defined as an unde­tect­able JAK2V617F allele fre­
nor­mal­iz­ing blood cell counts and reduc­ing phle­bot­omy needs, quency, for the first time these data suggested that exog­e­nous
ran­
dom­ ized tri­als com­par­
ing pegIFN to HU may also pro­ vide IFN could poten­tially be directly targeting the MPN clone.11 The
impor­tant direct com­par­a­tive effi­cacy. MPN-RC-112 is a trial that JAK2V617F allele bur­den was pro­gres­sively reduced in about 70%
ran­dom­izes high-risk PV and ET patients to first-line treat­ment of PV patients and 40% of ET patients. Moreover, the JAK2V617F
with either pegIFN or HU. A final anal­y­sis of the trial has yet to muta­tion became unde­tect­able in 24% of PV patients in the PVN1
be published, but interim trial updates report an over­all response study.15 Slightly lower rates of molec­u­lar remis­sion—15% in PV
rate of 78% with peginterferon alfa-2a, not sig­nif­i­cantly dif­fer­ent and 6% in ET patients—were reported in the US study. This is not
from that seen with HU (Table 1). Symptom ben­e­fit appears to be sur­pris­ing given a more het­ero­ge­neous study pop­u­la­tion with a
most pro­nounced for both agents in patients with a high base­line lon­ger dis­ease dura­tion. In both dis­ease groups, the pres­ence
symp­tom bur­den, though more patient-reported out­come data of addi­tional muta­tions such as TET2, ASXL1, IDH2, and TP53
are needed to make con­clu­sions about symp­tom ben­e­fit for both was asso­ci­ated with poorer molec­u­lar responses.16 Along with
HU and pegIFN. Comparative rates of throm­bo­ses will be a key improve­ments in both symp­toms and cell counts, these molec­
end point for deter­min­ing the best treat­ment across the age span. u­lar responses raise the pos­si­bil­ity and hope that this treat­ment
could selec­tively and dura­bly tar­get the malig­nant clone and
Molecular effects: defin­ing dis­ease mod­i­fi­ca­tion even allow hema­tol­o­gists to con­tem­plate a cura­tive treat­ment.
In addi­tion to impres­sive hema­to­logic responses, stud­ies using This aspi­ra­tion is often cap­tured with the phrase “dis­ease mod­i­
pegIFN have reported patients achiev­ing a com­plete molec­u­ fy­ing” when treating patients with PV and ET.

Table 1. Long-acting interferon trials

Study Phase Comparison Patients Follow-up dura­tion Hematologic CR Molecular response


PVN1 2 peginterferon alfa-2a 37 with PV 31.4 mo 94.6% 18.9% unde­tected
72.4% with >50% reduc­tion
MDACC 2 peginterferon alfa-2a 40 with PV 20 mo 70% (PV) 76% (ET) 14% and 6% unde­tected
39 with ET (PV vs ET)
54% and 38% with >50%
reduc­tion (PV vs ET)
PV-PROUD/ 3 Ropeginterferon vs HU 257 with PV 45.5 mo IFN vs HU 44% vs 51% at 12 mo (IFN
CONTINUATION-PV 71% vs 51% at 36 months vs HU)
66% vs 27% at 36 mo (IFN
vs HU)
Prakash Singh Shekhawat
IFN in PV/ET  |  465
Will I be on this for­ever? Treatment dura­tion with pegIFN enrolled 257 PV patients with early-stage dis­ease, includ­ing
Whether dura­ble dis­ease mod­i­fi­ca­tion or cure is achiev­able with patients newly requir­ing cytoreductive ther­apy or those who had
pegIFN has not yet been answered. How these gen­er­ally encour­ been treated with HU for fewer than 3 years.20 Randomization was
ag­ing results trans­late into clin­i­cal prac­tice can be var­i­able, and strat­i­fied by age, his­tory of throm­bo­em­bo­lism, and prior HU use.
the opti­mal treat­ment dura­tion with pegIFN is unknown. If a com­ The pri­mary out­come was com­posed of hema­to­logic response
plete molec­u­lar remis­sion is achieved, is a main­te­nance phase of and nor­mal­i­za­tion of spleen size at 12 months, achieved in 21%
treat­ment required? Tolerability and safety are crit­i­cal to these of patients in the ropeginterferon alfa-2b group and 28% in the
deci­sions, par­tic­u­larly in a pop­u­la­tion with often near-nor­mal life HU group. Longer treat­ ment and fol­low-up showed that 53%
expec­tancy. Longer-term fol­low-up stud­ies suggested an over­all of patients achieved a com­ plete hema­ to­log­

cal response with
dis­con­tin­u­a­tion rate of 22% for treat­ment-related tox­ic­ity. These improved dis­ease bur­den in the ropeginterferon alfa-2b group
rates decreased over time, though treat­ment-lim­it­ing grade vs 38% of patients receiv­ing HU (P = .044 at 36 months). Throm­
3 or 4 toxicities were still observed after 60 months on ther­apy.17 boembolic events in both arms were sim­i­larly rare. Reductions in
If pro­vider and patient elect to pur­sue a treat­ment-free remis­sion JAK2V617F allele fre­quency appeared to improve over time, pre­

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/463/1852047/463traxler.pdf by guest on 13 December 2021


period in patients achiev­ing a com­plete molec­u­lar response, dom­in ­ antly in the ropeginterferon alfa-2b group, supporting the
what are the param­et­ ers for resum­ing treat­ment? Should blood dis­ease-mod­i­fy­ing claim that appears to be unique to pegIFN as
count mon­i­tor­ing only drive retreatment deci­sions, or is mon­i­ a class.
tor­ing for ear­lier mea­sur­able resid­ual dis­ease by JAK2V617F poly­
mer­ase chain reac­tion a bet­ter stan­dard? If the lat­ter, what is the Dosing: pegIFN and ropeginterferon
thresh­old of detect­abil­ity driv­ing deci­sions? At what fre­quency High doses of IFN are poorly tol­er­ated. This was established
should patients be mon­i­tored? These and many more related in the nonpegylated for­mu­la­tion era and in dose-find­ing stud­
ques­tions remain unanswered. ies of pegIFN using up to 360 µg weekly.13 But deter­mi­na­tion of
the opti­mal starting and main­te­nance dose for both pegIFN and
Vascular risk ropeginterferon is a work in prog­ress. Conventional phar­ma­co­
Many of the major life-lim­it­ing con­se­quences of MPN stem from dy­namic stud­ies are chal­leng­ing when the tar­get(s) and mech­
vas­cu­lar com­pli­ca­tions, with rates of throm­bo­sis reported at a­nism of action of these agents remain some­what obscure. In
5.5 events per 100 patients per year.18 The published lon­gi­tu­ addi­tion, the rel­a­tively long time to response, with slow hema­
di­nal data sug­gest that IFN use may mit­i­gate vas­cu­lar risk, but to­logic responses and sub­se­quent deeper molec­u­lar responses,
com­par­a­tive data are lacking to robustly answer these impor­ begs a phil­os­oph­i­cal dilemma between “hit it hard and hit it
tant ques­tions. Encouragingly, the PVN1 trial did not report any early” vs “slow and steady wins the race” since a lin­ear and pre­
throm­bo­sis events in 6 years of fol­low-up, while the phase 2 US dict­able dose-response and dose-tox­ic­ity rela­tion­ship does not
trial reported a throm­bo­sis inci­dence of 1.22 per 100 patients per appear to exist, and maintaining patients on a tol­er­a­ble dose
year.17 Additional long-term fol­low-up may help define any true for a long period of time may be of par­tic­ul­ar impor­tance. Our
vas­cu­lar ben­efi­ t, but these data sug­gest that pegIFN is asso­ci­ approach is to start at a low dose, 45 µg weekly, of pegIFN alpha-
ated with an over­all low throm­bo­sis risk. 2a (Pegasys, the only cur­rently avail­­able US for­mu­la­tion) for a
Patients with splanch­nic vein throm­bo­sis (SVT) rep­re­sent a min­i­mum of 4 weeks. Close mon­i­tor­ing of mood, liver, and thy­
par­tic­u­larly high-risk group. MPN are a com­mon predisposing roid func­tion dic­tate fur­ther dose mod­i­fi­ca­tions or inter­rup­tions.
risk fac­tor for SVT, involv­ing hepatic, splenic, por­tal, or mes­en­ Some patients stay at 45 µg weekly, and oth­ers increase to 90 µg
teric veins, and it has been rec­og­nized that patients with SVT are weekly; doses as high as 135 or 180 µg weekly are less com­mon
at increased risk of devel­op­ing recur­rent throm­bo­ses, bleed­ing but can also be tol­er­ated by some and may be help­ful in the
com­pli­ca­tions, and arte­rial throm­bo­ses. Furthermore, HU fails absence of a hema­to­logic response at lower doses.
to pre­vent recur­rent SVT.19 The MPD-RC 111 study set to address
whether IFN could poten­tially fill this clin­i­cal need in a pro­spec­ Potential toxicities and mon­i­tor­ing for IFN ther­a­pies
tive trial eval­u­at­ing peg-IFN in 20 patients with a his­tory of SVT.14 Although lon­ger-act­ing IFNs have improved tol­er­a­bil­ity com­
Hematologic response rates were 70% at 12 months, on par pared to stan­dard for­mu­la­tions, patients still com­monly expe­ri­
with pre­vi­ous stud­ies of all­-com­ers, and SVT did not recur in any ence adverse effects. Fatigue and flu-like symp­toms are prev­a­lent.
patients dur­ing 2.2 years of fol­low-up. These tend to respond to acet­amin­op ­ hen or non­ste­roi­dal anti-
inflam­ma­tory med­i­ca­tions and can abate with time. Efforts to
Ropeginterferon alfa-2b min­ im
­ ize intolerabilities include low-dose run-in peri­ ods with
Efforts to fur­ther improve the activ­ity and tol­er­ab­ il­ity of pegIFN dose-esca­la­tion algo­rithms. Toxicity mon­i­tor­ing dur­ing treat­ment
treat­ment resulted in the devel­op­ment of ropeginterferon alfa-2b, includes blood counts as well as period thy­roid and liver func­tion
a monopegylated IFN dosed every 14 days. The Euro­pean Med­ test­ing and depres­sion screen­ing. Dose adjust­ments for cytope­
icines Agency approved ropeginterferon alfa-2b in 2019 for PV nias, liver dys­func­tion, and mood or neurocognitive effects are
with­out symp­tom­atic spleno­meg­aly; a deci­sion from the US Food crit­i­cal to safety and opti­mal response.
and Drug Association was expected in March 2021 but has yet to
be decided at the time of this writ­ing. If approved, ropeginter­ Are there dif­fer­en­tial responses to IFN depending
feron alfa-2b would be the first IFN spe­cif­i­cally approved for MPN on muta­tions?
in the US. Euro­pean Medicines Agency approval was based on a As men­ tioned, a post hoc anal­ y­
sis of an early pegIFN study
reg­is­tra­tion study known as PROUD-PV and its exten­sion study, astutely observed that fail­ing to achieve a com­plete molec­u­lar
CONTINUATION-PV. These stud­ies eval­u­ated ropeginterferon alfa- remis­sion was asso­ci­ated with molec­u­larly more com­plex dis­ease
2b com­pared with HU using a noninferiority design. PROUD-PV beyond the driver muta­tion. The pres­ence of muta­tions in TET2,

466  |  Hematology 2021  |  ASH Education Program


ASXL1, EZH2, DNMT3A, and IDH1/2 decreased the like­li­hood of Elizabeth O. Hexner: off-label drug use includes hydroxyurea, pegin­
achiev­ing a molec­u­lar remis­sion.16 Much more recently, detailed terferon alfa-2a, and Ropeginterferon alfa-2b.
ana­ly­ses of serial sam­ples found that leu­ke­mic pro­gen­i­tor cells
with homo­zy­gous JAK2V617F muta­tions were more sen­si­tive to
IFN com­pared to cells with a sin­gle (het­ero­zy­gous) JAK2V617F Correspondence
muta­tion and CALR-mutated cells. In addi­tion, for the het­ero­zy­ Elizabeth O. Hexner, Division of Hematology Oncology, Perelman
gous JAK2V617F cells, a higher dose of IFN appeared to be impor­ Center for Advanced Medicine, 3400 Civic Center Blvd, Philadel­
tant for effec­tive clear­ance of the clone.21 Thus, response to IFN phia, PA 19104; e-mail: hexnere@pennmedicine​­.upenn​­.edu.
appears to be influ­enced by driver muta­tion type and zygos­ity
and by addi­tional dis­ease-asso­ci­ated muta­tions.
References
1. Bjørn ME, Hasselbalch HC. The role of reac­tive oxy­gen spe­cies in mye­lo­
Pregnancy fi­bro­sis and related neo­plasms. Mediators Inflamm. 2015;2015(Octo­ber):
What will we tell our patient about her desire for addi­tional 648090.
preg­nan­cies? While data on the man­age­ment of PV in the set­

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/463/1852047/463traxler.pdf by guest on 13 December 2021


2. Hasselbalch HC, Thomassen M, Riley CH, et al. Whole blood tran­scrip­tional
ting of preg­nancy are lim­ited, the avail­­able stud­ies indi­cate that pro­fil­ing reveals dereg­u­la­tion of oxi­da­tive and antioxidative defence genes
in mye­lo­fi­bro­sis and related neo­plasms. Potential impli­ca­tions of downreg­
early fetal loss is com­mon, as is intra­uter­ine growth restric­tion.
ulation of Nrf2 for geno­mic insta­bil­ity and dis­ease pro­gres­sion. PLoS ONE.
Preterm deliv­ery is com­mon, and there is an esti­mated fetal 2014;9(11):e112786.
sur­vival of 50%.22 In gen­eral, the con­tin­u­a­tion of aspi­rin is rec­ 3. Talpaz M, Kantarjian HM, McCredie K, Trujillo JM, Keating MJ, Gutterman JU.
ommended for vir­tu­ally all­preg­nant women, and IFNs are con­ Hematologic remis­sion and cyto­ge­netic improve­ment induced by recom­
sid­ered safe and should gen­er­ally be con­tin­ued in patients for bi­nant human inter­feron alpha A in chronic mye­log­e­nous leu­ke­mia. N Engl
J Med. 1986;314(17):1065-1069.
whom cytoreductive treat­ ment was indi­ cated prepregnancy, 4. Silver RT. Recombinant inter­feron-alpha for treat­ment of polycythaemia
either for vas­cu­lar risk reduc­tion or symp­tom­atic con­trol. vera. Lancet. 1988;2(8607):403.
5. Tichelli A, Gratwohl A, Berger C, et al. Treatment of thrombocytosis in mye­
lo­pro­lif­er­a­tive dis­or­ders with inter­feron alpha-2a. Blut. 1989;58(1):15-19.
6. Xiong Z, Yan Y, Liu E, et al. Novel tumor anti­gens elicit anti-tumor humoral
immune reac­tions in a sub­set of patients with poly­cy­the­mia vera. Clin
CLINICAL CASE (Con­t in­u ed) Immunol. 2007;122(3):279-287.
7. Kiladjian JJ, Mesa RA, Hoffman R. The renais­sance of inter­feron ther­apy for
Now 2 years old, our patient’s daugh­ter is thriv­ing and meet­ the treat­ment of mye­loid malig­nan­cies. Blood. 2011;117(18):4706-4715.
ing all­her devel­op­men­tal mile­stones. Our patient started TP, 8. Berk PD, Goldberg JD, Silverstein MN, et al. Increased inci­dence of acute
con­tin­ues on aspi­rin, transitioned to a non-estro­gen-containing leu­ke­mia in poly­cy­the­mia vera asso­ci­ated with chlorambucil ther­apy. N
Engl J Med. 1981;304(8):441-447.
reli­able form of con­tra­cep­tion, and ini­ti­ated pegIFN alpha-2a, 9. Marchioli R, Finazzi G, Specchia G, et al; CYTO-PV Collaborative Group.
ini­tially at a low dose (45 µg weekly) and after 6 weeks, with­out Cardiovascular events and inten­sity of treat­ment in poly­cy­the­mia vera. N
dis­cern­able side effects, increased to 90 µg weekly. Her blood Engl J Med. 2013;368(1):22-33.
counts have improved, and her phle­bot­omy require­ments have 10. Mesa RA, Jamieson C, Bhatia R, et al. NCCN guide­lines insights: mye­lo­pro­lif­
er­a­tive neo­plasms, Version 2.2018. J Natl Compr Canc Netw. 2017;15(10):1193-
decreased. She is fol­low­ing with her obstet­rics group and with
1207.
mater­nal fetal med­i­cine, plan­ning for a sec­ond preg­nancy. She 11. Kiladjian JJ, Cassinat B, Turlure P, et  al. High molec­u­lar response rate of
will con­tinue with both aspi­rin and pegIFN through­out preg­ poly­cy­the­mia vera patients treated with pegylated inter­feron alpha-2a.
nancy if she con­ tin­ues to tol­
er­ate it as well as pro­
phy­lac­tic Blood. 2006;108(6):2037-2040.
doses of low-molec­u­lar-weight hep­a­rin for 6 weeks post­par­tum. 12. Kiladjian JJ. Long-term treat­ment with inter­feron alfa for mye­lo­pro­lif­er­a­tive
neo­plasms. Lancet Haematol. 2017;4(4):e150-e151.
13. Quintás-Cardama A, Kantarjian H, Manshouri T, et al. Pegylated inter­feron
alfa-2a yields high rates of hema­to­logic and molec­u­lar response in patients
Conclusions with advanced essen­tial thrombocythemia and poly­cy­the­mia vera. J Clin
Patients with PV and ET have a gen­er­ally favor­able prog­no­ Oncol. 2009;27(32):5418-5424.
14. Mascarenhas J, Kosiorek H, Prchal J, et al. A pro­spec­tive eval­u­a­tion of pegy-
sis, usu­ally sur­viv­ing decades and liv­ing full lives. But they live
lated inter­feron alfa-2a ther­apy in patients with poly­cy­the­mia vera and
with an incur­able chronic ill­ness that for some has debil­i­tat­ essen­tial thrombocythemia with a prior splanch­nic vein throm­bo­sis. Leu-
ing symp­toms, and harder to cap­ture is the exis­ten­tial uncer­ kemia. 2019;33(12):2974-2978.
tainty patients feel about the pos­si­bil­ity of dis­ease pro­gres­sion. 15. Kiladjian JJ, Cassinat B, Chevret S, et al. Pegylated inter­feron-alfa-2a induces
There is excite­ ment around pegIFNs, nontargeted ther­ a­ pies com­plete hema­to­logic and molec­u­lar responses with low tox­ic­ity in poly­cy­
the­mia vera. Blood. 2008;112(8):3065-3072.
that selec­tively tar­get the malig­nant MPN clone. Many foun­ 16. Quintás-Cardama A, Abdel-Wahab O, Manshouri T, et al. Molecular anal­
da­tional ques­tions—who, when, for how long—require more y­sis of patients with poly­cy­the­mia vera or essen­tial thrombocythemia
pre­cise answers, but the prog­ress toward bet­ter out­comes is receiv­ing pegylated inter­feron α-2a. Blood. 2013;122(6):893-901.
unde­ni­able. 17. Masarova L, Patel KP, Newberry KJ, et  al. Pegylated inter­feron alfa-2a in
patients with essen­tial thrombocythaemia or polycythaemia vera: a post-
hoc, median 83 month fol­low-up of an open-label, phase 2 trial. Lancet
Conflict-of-inter­est dis­clo­sure Haematol. 2017;4(4):e165-e175.
Elizabeth A. Traxler: no com­pet­ing finan­cial inter­ests to declare. 18. Marchioli R, Finazzi G, Landolfi R, et al. Vascular and neo­plas­tic risk in a large
Elizabeth O. Hexner: no com­pet­ing finan­cial inter­ests to declare. cohort of patients with poly­cy­the­mia vera. J Clin Oncol. 2005;23(10):2224-
2232.
19. De Stefano V, Rossi E, Carobbio A, et al. Hydroxyurea pre­vents arte­rial and
Off-label drug use late venous throm­botic recur­rences in patients with mye­lo­pro­lif­er­a­tive
Elizabeth A. Traxler: off-label drug use includes hydroxyurea, pegin­ neo­plasms but fails in the splanch­nic venous dis­trict: pooled anal­y­sis of
terferon alfa-2a, and Ropeginterferon alfa-2b. 1500 cases. Blood Cancer J. 2018;8(11):112.
Prakash Singh Shekhawat
IFN in PV/ET  |  467
20. Gisslinger H, Klade C, Georgiev P, et al; PROUD-PV Study Group. Ropegin­ 22. Gangat N, Joshi M, Shah S, et al. Pregnancy out­comes in mye­lo­pro­lif­er­a­
terferon alfa-2b ver­sus stan­dard ther­apy for polycythaemia vera (PROUD-PV tive neo­plasms: a Mayo Clinic report on 102 preg­nan­cies. Am J Hematol.
and CONTINUATION-PV): a randomised, non-infe­ri­or­ity, phase 3 trial and its 2020;95(5):E114-E117.
exten­sion study. Lancet Haematol. 2020;7(3):e196-e208.
21. Mosca M, Hermange G, Tisserand A, et al. Inferring the dynamic of mutated
hema­to­poi­etic stem and pro­gen­i­tor cells induced by IFNα in mye­lo­pro­lif­ © 2021 by The Amer­i­can Society of Hematology
er­a­tive neo­plasms. Published online 18 August 2021. Blood. DOI 10.1182/hema­tol­ogy.2021000280

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/463/1852047/463traxler.pdf by guest on 13 December 2021

468  |  Hematology 2021  |  ASH Education Program


MULTIDISCIPLINARY HEMATOLOGIC DISORDERS: WHAT IS THE HEMATOLOGIST ’ S ROLE ?

Hereditary hemorrhagic telangiectasia (HHT):


a practical guide to management

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/469/1852038/469hammill.pdf by guest on 13 December 2021


Adrienne M. Hammill,1 Katie Wusik,2 and Raj S. Kasthuri3
Cancer and Blood Diseases Institute, Division of Hematology, Cincinnati Children’s Hospital Medical Center, and Department of Pediatrics,
1

University of Cincinnati College of Medicine, Cincinnati, OH; 2Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH;
and 3Division of Hematology, Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, NC

Hereditary hemorrhagic telangiectasia (HHT), the second most common inherited bleeding disorder, is associated with
the development of malformed blood vessels. Abnormal blood vessels may be small and cutaneous or mucosal (telangi-
ectasia), with frequent complications of bleeding, or large and visceral (arteriovenous malformations [AVMs]), with addi-
tional risks that can lead to significant morbidity and even mortality. HHT can present in many different ways and can be
difficult to recognize, particularly in younger patients in the absence of a known family history of disease or epistaxis, its
most common manifestation. HHT is commonly diagnosed using the established Curaçao clinical criteria, which include
(1) family history, (2) recurrent epistaxis, (3) telangiectasia, and (4) visceral AVMs. Fulfillment of 3 or more criteria provides
a definite diagnosis of HHT, whereas 2 criteria constitute a possible diagnosis of HHT. However, these criteria are insuf-
ficient in children to rule out disease due to the age-dependent development of some of these criteria. Genetic testing,
when positive, can provide definitive diagnosis of HHT in all age groups. Clinical course is often complicated by signif-
icant epistaxis and/or gastrointestinal bleeding, leading to anemia in half of adult patients with HHT. The management
paradigm has recently shifted from surgical approaches to medical treatments aimed at control of chronic bleeding, such
as antifibrinolytic and antiangiogenic agents, combined with aggressive iron replacement with intravenous iron. Guide-
lines for management of HHT, including screening and treatment, were determined by expert consensus and originally
published in 2009 with updates and new guidelines in 2020.

LEARNING OBJECTIVES
• Recognize signs and symptoms of HHT
• Know when to test/screen patients for additional complications of the disease
• Understand the approach to treatment of various manifestations of HHT

ther questioning revealed that her mother and brother also


CLINICAL CASE had frequent epistaxis and migraine headaches and that her
Presentation. A 17-year-old previously healthy girl presented mother had undergone a lung lobectomy for AVMs. At that
to neurology with a complaint of intractable headaches and time, the neurosurgeon informed the family that the patient
a history of possible prior concussions during sports. Mag- likely had hereditary hemorrhagic telangiectasia (HHT).
netic resonance imaging (MRI) without contrast revealed Diagnosis. The patient was referred to our HHT center
some asymmetry of the cerebral vessels, and subsequent 20 months after seeing neurosurgery at the age of 18 years.
MRI with contrast and magnetic resonance arteriography She continued to have significant migraine headaches
revealed an arteriovenous malformation (AVM) involving the despite medication. She had nosebleeds approximately 1/wk,
left perisylvian fissure and insular areas. This was not felt to often gushing, lasting up to 15 minutes (Epistaxis Sever-
be the cause of her headaches, and an electroencephalo- ity Score [ESS] 3.7). Physical examination confirmed tel-
gram ruled out seizure activity. She was referred to neuro- angiectasia on her fingers, lower lip, and tongue. Family
surgery for further management. The neurosurgeon noted history was confirmed, including distant cousins with brain
telangiectasia on her fingertips and elicited a history of and lung AVMs. The patient met Curaçao clinical criteria for
heavy recurrent nosebleeds since the age of 9 years. Fur- definite HHT (+telangiectasia, +epistaxis, +AVM) at this visit

Prakash Singh Shekhawat


The ABCs of HHT | 469
even in the absence of a diag­no­sis in a first-degree rel­a­tive. She ized bleed­ing from the abnor­mal ves­sels, rather than the gen­eral
under­went genetic test­ing to con­firm the diag­no­sis, and a path­o­ risk of bleed­ing and post­sur­gi­cal bleed­ing asso­ci­ated with defi-
genic var­ia
­ nt in the endoglin gene ENG was iden­ti­fied. ciencies of plasma coag­u­la­tion fac­tors or plate­let dys­func­tion.
Screening. Additional screen­ing was performed to eval­ua ­ te
for lung AVMs. A delayed-bub­ble echo was pos­i­tive, and sub­se­ Diagnosis
quent com­puted tomog­ra­phy angi­og­ra­phy of the chest revealed The Curaçao clin­i­cal diag­nos­tic cri­te­ria, first for­mal­ized in 2000,
a left lower lobe AVM with a nidus mea­sur­ing nearly 2 × 1 × 1 cm, are shown in Table 1.12 The first is epi­staxis; to meet this cri­te­rion,
with a feed­ing artery mea­sur­ing 4 mm. nose­bleeds should be recur­rent and spon­ta­ne­ous. These may be
gushing or drip­ping, as there is no require­ment for sever­ity.
When ini­ tial lung AVM screen­ing is neg­a­
tive, it should be
repeated every 5 years in chil­dren1,2 and every 5 to 10 years While there is no widely accepted nose­bleed fre­quency to
in adults. meet this cri­te­rion, we gen­er­ally use more than 4 per year with
no other cause.5 A his­tory of night­time nose­bleeds is par­tic­u­

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/469/1852038/469hammill.pdf by guest on 13 December 2021


Treatment. She under­went embo­li­za­tion of her pul­mo­nary larly sug­ges­tive of HHT.
AVM with dra­matic improve­ment in her migraine head­aches. At
the age of 22 years, she under­went pre­ci­sion radio­ther­apy to mul­ Telangiectases, or small bright red spots, typ­i­cally occur at char­ac­
ti­ple brain AVMs due to con­cern for pro­gres­sive venous ectasia. ter­is­tic sites in HHT, includ­ing lips, oral cav­ity (par­tic­u­larly tongue),
Follow-up. She con­tin­ues to be followed in our HHT Center of fin­gers, and nose (Figure 1). These tel­an­gi­ec­ta­sia can be tiny and
Excellence at reg­u­lar inter­vals to mon­i­tor for new or recanalized appear like red freck­les; they gen­er­ally blanch with pres­sure, with
pul­mo­nary AVMs (bub­ble echo every 3-5 years) and changes in rapid reperfusion upon release. Visceral lesions can also be pres­
epi­staxis (ESS at every visit). She gets annual lab­or­a­tory tests to ent, includ­ing GI tel­an­gi­ec­ta­ses and larger high-flow lesions, includ­
screen for com­pli­ca­tions of liver AVMs (hepatic panel includ­ing ing (in order of fre­quency) liver, lung, brain, and spi­nal AVMs. These
γ-glutamyl trans­fer­ase [GGT] to assess liver func­tion and brain can be asymp­tom­atic and may only be detected with screen­ing.
natri­uretic pep­tide [BNP] for heart func­tion) and ane­mia (com­ A fam­ily his­tory of sim­il­ar symp­toms in a first-degree rel­a­tive (par­
plete blood count [CBC], fer­ ri­tin, total iron bind­
ing capac­ ity ent, sib­ling, or child) is usu­ally pres­ent even if they do not always
[TIBC], and retic­u­lo­cyte count) related to nose­bleeds or poten­ carry a diag­no­sis of HHT. Underrecognition and delayed diag­no­sis
tial gas­tro­in­tes­ti­nal (GI) bleed­ing. She takes anti­bi­ot­ics prior is a con­sid­er­able prob­lem in HHT,13 and there is a need to increase
to den­tal pro­ce­dures, includ­ing reg­u­lar cleaning, for infec­tion aware­ness among health care pro­vid­ers.
pro­phy­laxis, due to her known lung AVMs as per HHT treat­ment
guide­lines. She is also followed in the cere­bro­vas­cu­lar clinic for There is sim­i­larly no stan­dard required tel­an­gi­ec­ta­sia count; in
her cere­bral AVMs. our cen­ters, we use more than 4.5

Annual lab­o­
ra­
tory tests for adults followed in our cen­ ter Clinical cri­te­ria, val­i­dated in adults, may not be suf­fic­ ient to rule out
include CBC, fer­ri­tin, retic­u­lo­cyte count, TIBC, hepatic pro­file, dis­ease in chil­dren youn­ger than 16 years.14 In 1 study by Pahl et al,14
GGT, BNP,2 and vita­min D.3,4 the pos­i­tive pre­dic­tive value of the clin­i­cal cri­te­ria was extremely
high in chil­dren (99% over­all), but the neg­a­tive pre­dic­tive value
Her fam­ily mem­bers also under­went screen­ing for vis­ceral vas­ was only 54% over­all and even lower in chil­dren youn­ger than
cu­lar lesions given their clin­i­cal diag­noses of HHT once our 5 years. This is due to the age-depen­dent devel­op­ment of 2 of the
patient was diag­nosed. Her brother was found to have both
mul­ti­ple brain AVMs, which were treated with radio­ther­apy,
and mul­ti­ple lung AVMs, which were treated with coil embo­li­ Table 1. Curaçao cri­te­ria for the clin­i­cal diag­no­sis of HHT12
za­tion. Her mother was found to have addi­tional lung AVMs that
required embo­ li­
za­
tion. Furthermore, her mother proceeded Telangiectases Multiple, at char­ac­ter­is­tic sites
to develop severe iron defi­ciency ane­mia sec­ond­ary to both   Lips, oral cav­ity, fin­gers, nose
severe epi­staxis and GI bleed­ing and is tak­ing main­te­nance
Epistaxis Recurrent spon­ta­ne­ous nose­bleeds
ther­apy with sys­temic bevacizumab in addi­tion to inter­mit­tent
iron infu­sions to main­tain fer­ri­tin more than 50 ng/mL. Visceral involve­ment GI tel­an­gi­ec­ta­sia
Pulmonary AVM
Hepatic AVM
Introduction
Cerebral VM
Hereditary hem­or­rhagic tel­an­gi­ec­ta­sia is the sec­ond most com­
Spinal AVM
mon inherited bleed­ing dis­or­der, occur­ring in 1/5000 to 1/10,000
peo­ple.6 It is gen­er­ally inherited in an auto­so­mal dom­i­nant fash­ Family his­tory First-degree rel­a­tive with known HHT
ion but can occur de novo, includ­ing in mosaic form in some   Parent, sib­ling, or child
pro­bands.7-11 Bleeding is the most com­mon symp­tom, occur­ring Definite if ≥3 cri­te­ria are pres­ent OR if path­o­genic var­i­ant iden­ti­fied in
from nasal, cuta­ne­ous, or GI tel­an­gi­ec­ta­ses. Unlike bleed­ing dis­ known HHT gene
or­ders such as hemo­philia or von Willebrand dis­ease, bleed­ing in Suspected if 2 cri­te­ria are pres­ent
HHT is sec­ond­ary to devel­op­ment of malformed blood ves­sels,
Unlikely if <2 cri­te­ria are pres­ent
which result in ectasia and increased fra­gil­ity. This leads to local­

470  |  Hematology 2021  |  ASH Education Program


Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/469/1852038/469hammill.pdf by guest on 13 December 2021
Figure 1. Telangiectases in HHT.

cri­te­ria. Nosebleeds typ­i­cally develop in the sec­ond decade of in aortopathy as well.19 Rarely, HHT cases meet­ing clin­i­cal cri­te­ria
life and do not com­monly require med­i­cal atten­tion in child­hood, may be caused by GDF2 (BMP9) or RASA1.20 Overlap has been
although many patients do develop recur­rent and/or severe epi­ observed between HHT and cap­il­lary‑ malformation–AVM syn­
staxis requir­ing med­i­cal and/or sur­gi­cal inter­ven­tions by mid­dle drome caused by EPHB4.21 In prac­tice, most test­ing is now per-
age. Development of tel­an­gi­ec­ta­sia is also age depen­dent, typ­i­ formed as part of multigene pan­els consisting of 5 to 6 genes
cally noted in the sec­ond and third decades. Thus, even when a (ENG, ACVRL1, SMAD4, RASA1, GDF2, and EPHB4).
child has a par­ent with HHT (1 clin­i­cal cri­te­rion), in the absence of Genetic test­ing is recommended to assist in establishing the
imag­ing performed to screen for vis­ceral AVMs, affected chil­dren diag­no­sis in indi­vid­u­als who do not meet Curaçao cri­te­ria or in
may not meet the diag­nos­tic thresh­old for dis­ease. those who are asymp­tom­atic or min­i­mally symp­tom­atic, includ­
Genetic test­ing has become more avail­­able and afford­able ing young chil­dren. Genetic test­ing can also be used to iden­tify
and can be used to make the HHT diag­no­sis.15 HHT-caus­ing muta­ the caus­a­tive muta­tion in a fam­ily with clin­i­cally con­firmed HHT
tions have been iden­ti­fied in sev­eral genes in the transforming or to estab­lish a diag­no­sis in rel­a­tives of a per­son with a known
growth fac­tor β path­way, with the major­ity of path­o­genic var­i­ caus­a­tive muta­tion.15 However, cur­rent test­ing remains unable
ants found in ENG and ACVRL1. In 1 recent study, ENG and ACVRL1 to iden­tify a caus­a­tive genetic change in up to 10% to 15% of
muta­tions were found to com­prise up to 96% of cases of “clas­ patients with a clin­i­cal diag­no­sis.
sic HHT” meet­ing strictly applied Curaçao cri­te­ria.16 SMAD4 gene
muta­tions account for approx­i­ma­tely 10% of ENG- and ACVRL1-
Several lab­ o­
ra­
to­ries now offer free fam­ ily test­
ing within a
neg­a­tive cases of HHT, account­ing for 1% to 2% of cases over­
spec­i­fied time period when a path­o­genic var­i­ant is iden­ti­fied
all. SMAD4 also causes juve­nile polyposis, resulting in GI pol­yps
16,17
in 1 patient.
and a can­cer pre­dis­po­si­tion syn­drome, 18
and has Singh
Prakash been impli­
cated
Shekhawat
The ABCs of HHT  |  471
Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/469/1852038/469hammill.pdf by guest on 13 December 2021
Figure 2. Screening recommendations for HHT, assembled from multiple guidelines.1,15

Guidelines have been established regard­ ing the diag­ no­sis, ber of fac­tors, includ­ing fre­quency, dura­tion, amount of bleed­
screen­ing, and treat­ment of peo­ple liv­ing with HHT. The orig­i­nal ing and pres­ence of ane­mia, and inter­ven­tions needed to con­trol
guide­lines were published in 2011 and cov­ered diag­no­sis of HHT, the bleed­ing.22 This score should be cal­cu­lated at every visit and
epi­staxis, cere­bral vas­cu­lar malformations, pul­mo­nary AVMs, GI before and after any inter­ven­tions to mea­sure their impact (online
bleed­ing, and liver vas­cu­lar malformations.15 The Second Inter- cal­cu­la­tor read­ily avail­­able at https:​­/​­/curehht​­.org​­/resource​­/epi-
national guide­lines were published in 2020, with updated guide­ staxis​­-severity​­-score​­/). ESS can be used to guide treat­ment of epi­
lines for epi­staxis, GI bleed­ing, and liver vas­cu­lar malformations, staxis, as shown in Figure 3. Although his­tor­ic ­ ally approaches were
as well as new guide­lines on ane­mia and anticoagulation, pedi­ pri­mar­ily sur­gi­cal, med­i­cal ther­a­pies includ­ing antifibrinolytics
at­rics, and preg­nancy and deliv­ery.1 (tranexamic acid, epsi­lon aminocaproic acid)23,24 and sys­temic anti-
angiogenics have become the main­stay of treat­ment strat­e­gies in
Clinical man­i­fes­ta­tions and man­age­ment recent years, as discussed in more detail below under “Anemia.”
Initial screen­ing
Once the diag­no­sis of HHT is made or suspected, all­patients GI bleed­ing
should undergo ini­tial screen­ing for poten­tial com­pli­ca­tions. GI bleed­ing is also a com­mon symp­tom in adults with HHT older
Although a long-time area of debate in the care of patients than 50 years, reported in 13% to 30% of patients. Most GI tel­
with HHT, recent pedi­at­ric care guide­lines clar­ify that chil­dren an­gi­ec­ta­sias occur in the stom­ach (46%-75%) and small bowel
require workup and screen­ing as well as adults, because even (56%-91%).25 GI bleed­ing can be highly mor­bid in adults, with
though the out­ward clin­i­cal signs (tel­an­gi­ec­ta­sia) and symp­ chronic GI bleed­ing resulting in severe iron defi­ciency ane­mia
toms (epi­staxis) may so far be absent, pedi­at­ric patients can still requir­ing fre­ quent intra­ ve­nous iron replace­ ment and/or red
have vis­ceral AVMs that can cause mor­bid­ity and even mor­tal­ blood cell trans­ fu­
sions, with decreased QoL. GI bleed­ ing is
ity. Screening pro­ce­dures are sum­ma­rized in Figure 2 from the rarely sig­nif­i­cant in chil­dren except in those car­ry­ing the SMAD4
guide­lines for pul­mo­nary AVMs, liver vas­cu­lar malformations, geno­type and then is typ­i­cally related to bleed­ing from pol­yps.
brain vas­cu­lar malformations, and pedi­at­ric care. GI bleed­ing should be suspected in the pres­ence of iron defi­
ciency with or with­out ane­mia, par­tic­u­larly when this is out of
Epistaxis pro­por­tion to reported epi­staxis. The first step in eval­u­a­tion is
Epistaxis, the most prominent symp­tom in most peo­ple with HHT, gen­er­ally refer­ral to a gas­tro­en­ter­ol­o­gist famil­iar with HHT for
can sig­nif­i­cantly inter­fere with daily activ­i­ties, includ­ing work, upper endos­copy, as stool occult blood tests are not reli­able
school, and qual­ity of life (QoL). The updated guide­lines out­line in the set­ting of ongo­ing epi­staxis. Recommended treat­ment is
a num­ ber of rec­ om­men­ da­
tions for con­
trol of nose­ bleeds in a deter­mined based on the sever­ity of GI bleed­ing and coexisting
step­wise fash­ion.1 The ESS was pre­vi­ously established for use in ane­mia; rec­om­men­da­tions for the workup and treat­ment of GI
HHT to facil­i­tate mea­sure­ment of nose­bleeds based on a num­ bleed­ing can be found in Figure 4.

472  |  Hematology 2021  |  ASH Education Program


Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/469/1852038/469hammill.pdf by guest on 13 December 2021
Figure 3. Epistaxis Severity Score (ESS)22 and management of epistaxis in HHT.1 *Therapy currently under investigation. ENT, ear,
nose, and throat; NSAID, nonsteroidal anti-inflammatory drug.

Anemia stud­ied in a pro­spec­tive man­ner in HHT, it has become the de


Anemia can result from epi­staxis, GI bleed­ing, or the com­bi­na­ facto stan­dard of care for severe/refrac­tory ane­mia in HHT. The
tion of the two and is pres­ent in more than 50% of adults with effi­cacy of the oral antiangiogenic agents pomalidomide (PATH-
HHT.26 Anemia, and even iron defi­ciency with­out ane­mia, should HHT trial; clinicaltrials​­.gov NCT03910244) and pazopanib (clin­i­cal
be man­aged aggres­sively to cor­rect ane­mia and main­tain ade­ trial open­ing soon) are being eval­u­ated pro­spec­tively in clin­i­
quate iron stores. In addi­tion to well-known signs and symp­toms cal tri­als fol­low­ing prom­is­ing data in pilot stud­ies.31,32 Additional
of ane­mia, includ­ing fatigue, short­ness of breath, lightheadedness, agents, includ­ing doxy­cy­cline33 (NCT03397004, NCT04167085)
pal­lor, and car­diac stress, there have been addi­tional asso­ci­a­tions and tacrolimus (NCT04646356), are also being actively eval­u­ated.
reported in patients with HHT. In patients with liver AVMs, iron
defi­ciency results in sub­stan­tial increased risk of aber­rant man­ Visceral AVMs
ga­nese depo­si­tion in the brain, lead­ing to Parkinson-like neu­ro­ Visceral AVMs, although less com­mon than bleed­ing symp­toms,
logic symp­toms.27 Paradoxically, an increased risk for throm­bo­sis can also cause sig­nif­i­cant mor­bid­ity and mor­tal­ity if unde­tected
has been reported in HHT, which appears to cor­re­late with iron and/or untreated. Therefore, it is impor­tant to be aware of poten­
defi­ciency.28 Such throm­botic events are gen­er­ally treated with tial signs and symp­toms of AVMs and fol­low cur­rent screen­ing
anticoagulation, with asso­ci­ated increased bleed­ing risk. While guide­lines. In doing so, indi­vid­u­als with HHT who undergo rec-
anticoagulation is not abso­lutely contraindicated in HHT, it can be ommended screen­ing and treat­ment at an HHT Center of Excel-
quite dif­fi­cult to bal­ance bleed­ing risks and clot­ting risks in these
lence can expect a nor­mal life span.34,35
patients. Complete rec­om­men­da­tions for man­age­ment of ane­mia Brain vas­cu­lar malformations seen in HHT include AVM, arte­
and anticoagulation in HHT can be found in Figure 5. rio­
ve­ nous fis­ tula (AVF), vein of Galen malformations, cere­ bral
cav­ern­ous malformations, devel­op­men­tal venous anom­a­lies, and
Our goal is to try to achieve and main­tain fer­ri­tin above 50 ng/mL cap­il­lary tel­an­gi­ec­ta­sia. Of these, AVF and AVM are con­sid­ered
in the set­ting of ongo­ing recur­rent bleed­ing. highest risk for com­pli­ca­tions. Symptoms of AVM vary by loca­
tion and can include sei­zure, head­ache, or, in the case of rup­ture,
For ane­mia resulting from epi­staxis or GI bleed­ing refrac­tory to hem­or­rhagic stroke; how­ever, most brain AVMs are asymp­tom­
local ther­a­pies and first-line med­i­cal treat­ments, sys­temic anti- atic and found on rou­tine screen­ing. Management of brain vas­
angiogenic ther­a­pies should be con­sid­ered in com­bi­na­tion with cu­lar malformations, once iden­ti­fied, is pur­sued col­lab­o­ra­tively
aggres­ sive iron replace­ ment. To date, the most widely used with neu­ro­ra­di­ol­ogy, neu­rol­ogy, neu­ro­sur­gery, and/or neuroin-
antiangiogenic agent in HHT is bevacizumab, a mono­clo­nal anti– terventional radi­ol­ogy. Treatment options for high-risk lesions
vas­cu­lar endo­the­lial growth fac­tor anti­body given as an intra­ve­ include embo­li­za­tion by a skilled neurointerventional radi­ol­o­gist,
nous infu­sion.29,30 Although sys­temicPrakash
bevacizumab has not been
Singh Shekhawatresec­tion by a neu­ro­sur­geon, or focal radi­a­tion (gamma knife or

The ABCs of HHT  |  473


Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/469/1852038/469hammill.pdf by guest on 13 December 2021
Figure 4. Guidelines for the management of GI bleeding in HHT.1 *Mild GI bleeding defined as meeting hemoglobin goals with oral
iron. **Moderate GI bleeding defined as meeting hemoglobin goals with intravenous oral iron. ***Severe GI bleeding defined as not
meeting hemoglobin goals despite adequate iron replacement or requiring blood transfusions. CRC, colorectal cancer; EGD, esoph-
agogastroduodenoscopy.

pro­ton) admin­is­tered by a radi­a­tion oncol­o­gist; choice of ther­ on the cen­ter) but may also be con­sid­ered for slightly smaller
apy will depend on risk of the lesion, size, and loca­tion, includ­ing AVMs based on symp­toms. After treat­ment, con­tin­ued mon­i­tor­
con­sid­er­ations of acces­si­bil­ity and elo­quence. ing is nec­es­sary to rule out recan­a­li­za­tion or for­ma­tion of new
Lung AVMs can be asymp­tom­atic or may cause a num­ber of AVMs; inter­vals vary on a case-by-case basis. An update to the
signs and symp­toms. These include chronic man­i­fes­ta­tions such pul­mo­nary AVM guide­lines is planned in the near future.
as fatigue, short­ness of breath, hyp­oxia, asthma-like symp­toms Liver AVMs are the most com­mon vis­ceral AVM found in HHT,
poorly respon­sive to β-ago­nists, migraine head­aches, or dra­matic but they are often asymp­tom­atic and have been his­tor­i­cally hard
and life-threat­en­ing pre­sen­ta­tions such as par­a­dox­i­cal embo­li­ to treat. The updated guide­lines rec­om­mend screen­ing, as it may
za­tion lead­ing to stroke, tran­sient ische­mic attack, or cere­bral be pos­si­ble to pick up sequelae of the hepatic AVMs ear­lier this
abscess. AVM rup­ture caus­ing mas­sive hemop­ty­sis and/or spon­ way, includ­ing high-out­put heart fail­ure, pul­mo­nary hyper­ten­
ta­ne­ous hemothorax is a rare pre­sen­ta­tion. Cerebral abscess risk sion, por­tal hyper­ten­sion, and enceph­a­lop­a­thy. When pres­ent,
has been strongly asso­ci­ated with den­tal work, includ­ing rou­tine liver AVMs should be followed by a hepatologist but, depending
cleaning, and anti­bi­otic pro­phy­laxis should be admin­is­tered prior on the man­i­fes­ta­tions, may require car­di­­ol­ogy and/or pulmonol-
to den­tal work to all­patients with known lung AVMs and those ogy or pul­mo­nary hyper­ten­sion exper­tise. Treatment options for
who have not yet under­gone lung AVM screen­ing to pre­vent hepatic AVMs have been lim­ited, as embo­li­za­tion often leads to
this com­pli­ca­tion. Many HHT cen­ters also rec­om­mend anti­bi­otic fur­ther liver com­pli­ca­tions. Current approaches include orthot-
pro­phy­laxis for other “dirty” pro­ce­dures that could poten­tially opic liver trans­plant or sys­temic med­i­cal ther­ap­ ies such as beva-
intro­duce bac­te­ria into the blood­stream, includ­ing colonoscopy cizumab, reviewed in more detail above in the “Anemia” sec­tion.
and other GI/GU (genitourinary) pro­ce­dures, but this is not yet
stan­dard­ized. Treatment of lung AVMs is gen­er­ally car­ried out by Pregnancy and child­birth
interventional radi­ol­o­gists using coil embo­li­za­tion. The deci­sion Genetic coun­ sel­
ing is recommended pre­ con­cep­tion to dis­
to treat lung AVMs is based on size of the AVM nidus and diam­e­ter cuss risks of trans­mis­sion and/or pre­na­tally to explore mul­ti­ple
of the feed­ing ves­sels (typ­i­cally >2-2.5 mm diam­e­ter depending options for genetic test­ing of the child, from pre­im­plan­ta­tion, to

474  |  Hematology 2021  |  ASH Education Program


Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/469/1852038/469hammill.pdf by guest on 13 December 2021
Figure 5. Guidelines for the management of anemia and anticoagulation in HHT.1 HGB, hemoglobin.

Figure 6. Guidelines for pregnancy and delivery in HHT.1 CT, computed tomography; VM, vascular malformation.
Prakash Singh Shekhawat
The ABCs of HHT  |  475
in utero, to post­na­tal options such as cord blood. Guidelines for References
preg­nancy and deliv­ery were also devel­oped, includ­ing screen­ 1. Faughnan ME, Mager JJ, Hetts SW, et al. Second International Guidelines for
the Diagnosis and Management of Hereditary Hemorrhagic Telangiectasia.
ing rec­om­men­da­tions for AVMs, outlined in Figure 6. Women
Ann Intern Med. 2020;173(12):989-1001.
who have not been screened recently or who have known lung 2. Ginon I, Decullier E, Finet G, et al. Hereditary hem­or­rhagic tel­an­gi­ec­ta­sia,
AVMs and/or those with high-risk brain vascular malformations liver vas­cu­lar malformations and car­diac con­se­quences. Eur J Intern Med.
should be followed by a mul­ ti­
dis­
ci­
plin­
ary team, includ­ ing a 2013;24(3):e35-e39.
mater­nal-fetal med­i­cine spe­cial­ist, at a ter­tiary care cen­ter. Lung 3. Weber LM, McDonald J, Whitehead K. Vitamin D lev­els are asso­ci­ated with
epi­staxis sever­ity and bleed­ing dura­tion in hered­i­tary hem­or­rhagic tel­an­
AVMs in need of treat­ment should be embolized in the sec­ond
gi­ec­ta­sia. Biomark Med. 2018;12(4):365-371.
tri­mes­ter if pos­si­ble and patients coun­seled that hemop­ty­sis is 4. Ratjen A, Au J, Carpenter S, John P, Ratjen F. Growth of pul­mo­nary arte­rio­ve­-
an emer­gency, based on increas­ing evi­dence of mater­nal mor­ nous malformations in pedi­at­ric patients with hered­i­tary hem­or­rhagic tel­
bid­ity and even mor­tal­ity dur­ing preg­nancy and child­birth,36 due an­gi­ec­ta­sia. J Pediatr. 2019;208:279-281.
5. McDonald J, Stevenson DA, Whitehead K. Incidence of epi­staxis and tel­an­
at least in part to increased risk of lung AVM rup­ture.37,38
gi­ec­ta­ses in the gen­eral pop­u­la­tion. Angiogenesis. 2017;18:531.
6. Kjeldsen AD, Vase P, Green A. Hereditary haemorrhagic tel­an­gi­ec­ta­sia: a

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/469/1852038/469hammill.pdf by guest on 13 December 2021


Conclusion pop­u­la­tion-based study of prev­a­lence and mor­tal­ity in Dan­ish patients.
Patients with HHT can ini­tially pres­ent to any of sev­eral dif­fer­ent J Intern Med. 1999;245(1):31-39.
med­i­cal spe­cial­ists, depending on their presenting symp­toms, 7. McDonald J, Wooderchak-Donahue WL, Henderson K, Paul E, Morris A,
Bayrak-Toydemir P. Tissue-spe­cific mosa­i­cism in hered­i­tary hem­or­rhagic
and underrecognition of the dis­ease risks diag­nos­tic delays and
tel­an­gi­ec­ta­sia: impli­ca­tions for genetic test­ing in fam­i­lies. Am J Med Genet
increased mor­bid­ity. It is impor­tant that poten­tial pro­vid­ers have A. 2018;176(7):1618-1621.
a high index of sus­pi­cion in the pres­ence of the Curaçao clin­i­ 8. Tørring PM, Kjeldsen AD, Ousager LB, Brusgaard K. ENG muta­tional mosa­
cal cri­te­ria for HHT. These patients can pres­ent to hema­tol­o­gists i­cism in a fam­ily with hered­i­tary hem­or­rhagic tel­an­gi­ec­ta­sia. Mol Genet
in many ways: with ane­mia and iron defi­ciency (due to bleed­ Genomic Med. 2018;6(1):121-125.
9. Best DH, Vaughn C, McDonald J, et  al. Mosaic ACVRL1 and ENG muta­
ing from nasal or GI tel­an­gi­ec­ta­ses), with poly­cy­the­mia (as a nor­
tions in hered­i­tary haemorrhagic tel­an­gi­ec­ta­sia patients. J Med Genet.
mal phys­i­o­logic response in the set­ting of pul­mo­nary AVMs), or 2011;48(5):358-360.
for sys­temic ther­a­pies for other com­pli­ca­tions of HHT (severe 10. Lee NP, Matevski D, Dumitru D, Piovesan B, Rushlow D, Gallie BL. Identifi-
ane­mia requir­ ing sig­
nif­i­
cant iron/blood replace­ ment or pul­ cation of clin­i­cally rel­e­vant mosa­i­cism in type I hered­i­tary haemorrhagic
tel­an­gi­ec­ta­sia. J Med Genet. 2011;48(5):353-357.
mo­nary hyper­ten­sion or high-out­put car­diac fail­ure related to
11. Gedge F, McDonald J, Phansalkar A, et al. Clinical and ana­lyt­i­cal sensitivities
liver AVMs).39 Where HHT Centers of Excellence exist, cur­rently in hered­i­tary hem­or­rhagic tel­an­gi­ec­ta­sia test­ing and a report of de novo
at 30 sites in North America, the cen­ter’s direc­tor or codi­rec­tor muta­tions. J Mol Diagn. 2007;9(2):258-265.
will likely pro­vide a med­ic ­ al home for these often com­pli­cated 12. Shovlin CL, Guttmacher AE, Buscarini E, et al. Diagnostic cri­te­ria for hered­
patients; how­ever, no such cen­ter exists in most of the med­i­cal i­tary hem­or­rhagic tel­an­gi­ec­ta­sia (Rendu-Osler-Weber syn­drome). Am J
Med Genet. 2000;91(1):66-67.
cen­ters across the coun­try. Elsewhere, hema­tol­o­gist-oncol­o­gists
13. Pierucci P, Lenato GM, Suppressa P, et  al. A long diag­ nos­
tic delay in
may find them­selves in the role of pri­mary pro­vider, par­tic­u­larly patients with hered­i­tary haemorrhagic tel­an­gi­ec­ta­sia: a ques­tion­naire-
as more targeted med­i­cal ther­a­pies become avail­­able as, to date, based ret­ro­spec­tive study. Orphanet J Rare Dis. 2012;7:33.
most of these drugs have been used pre­vi­ously in the treat­ment 14. Pahl KS, Choudhury A, Wusik K, et al. Applicability of the Curaçao cri­te­ria
for the diag­no­sis of hered­i­tary hem­or­rhagic tel­an­gi­ec­ta­sia in the pedi­at­ric
of can­cers. Given this and the rel­a­tively high fre­quency of HHT as
pop­u­la­tion. J Pediatr. 2018;197:207-213.
a hered­i­tary bleed­ing dis­or­der, an under­stand­ing of mod­ern HHT 15. Faughnan ME, Palda VA, Garcia-Tsao G, et al; HHT Foundation International—
man­age­ment is cru­cial for the prac­tic­ing hema­tol­o­gist. Guidelines Working Group. International guide­lines for the diag­no­sis and
man­age­ment of hered­i­tary haemorrhagic tel­an­gi­ec­ta­sia. J Med Genet.
Conflict-of-inter­est dis­clo­sure 2011;48(2):73-87.
16. McDonald J, Bayrak-Toydemir P, DeMille D, Wooderchak-Donahue W,
Adrienne M. Hammill: none related to this paper. But AH has clin- Whitehead K. Curaçao diag­nos­tic cri­te­ria for hered­i­tary hem­or­rhagic tel­
ical trials (for other diseases) with novartis, cerecor, Venthera, an­gi­ec­ta­sia is highly pre­dic­tive of a path­o­genic var­i­ant in ENG or ACVRL1
Merck; had study drug provided by Pfizer; serves as a consultant (HHT1 and HHT2). Genet Med. 2020;22(7):1201-1205.
for Novartis. 17. Gallione CJ, Richards JA, Letteboer TG, et  al. SMAD4 muta­tions found in
unse­lected HHT patients. J Med Genet. 2006;43(10):793-797.
Katie Wusik: none reported.
18. Gallione C, Aylsworth AS, Beis J, et al. Overlapping spec­tra of SMAD4 muta­
Raj S. Kasthuri: none reported. tions in juve­nile polyposis (JP) and JP-HHT syn­drome. Am J Med Genet A.
2010;152A(2):333-339.
Off-label drug 19. Heald B, Rigelsky C, Moran R, et al. Prevalence of tho­racic aortopathy in
Adrienne M. Hammill: All drugs listed here are off label. None has patients with juve­nile polyposis syn­drome–hered­i­tary hem­or­rhagic tel­an­
gi­ec­ta­sia due to SMAD4. Am J Med Genet A. 2015;167(8):1758-1762.
FDA approval. They are not attributable to a single author. 20. Hernandez F, Huether R, Carter L, et al. Mutations in RASA1 and GDF2 iden­
Katie Wusik: All drugs listed here are off label. None has FDA ap- ti­fied in patients with clin­i­cal fea­tures of hered­i­tary hem­or­rhagic tel­an­gi­ec­
proval. They are not attributable to a single author. ta­sia. Hum Genome Var. 2015;2:15040.
Raj S. Kasthuri: All drugs listed here are off label. None has FDA 21. Wooderchak-Donahue WL, Akay G, Whitehead K, et al. Phenotype of CM-
AVM2 caused by var­i­ants in EPHB4: how much over­lap with hered­i­tary
approval. They are not attributable to a single author.
hem­or­rhagic tel­an­gi­ec­ta­sia (HHT)? Genet Med. 2019;21(9):2007-2014.
22. Hoag JB, Terry P, Mitchell S, Reh D, Merlo CA. An epi­staxis sever­ity score for
Correspondence hered­i­tary hem­or­rhagic tel­an­gi­ec­ta­sia. Laryngoscope. 2010;120(4):838-843.
Adrienne M. Hammill, Cancer and Blood Diseases Institute, Divi- 23. Gaillard S, Dupuis-Girod S, Boutitie F, et al; ATERO Study Group. Tranexamic
acid for epi­staxis in hered­i­tary hem­or­rhagic tel­an­gi­ec­ta­sia patients: a
sion of Hematology, Cincinnati Children’s Hospital Medical Center,
Euro­pean cross-over con­trolled trial in a rare dis­ease. J Thromb Haemost.
and Department of Pediatrics, University of Cincinnati College of 2014;12(9):1494-1502.
Medicine, 3333 Burnet Ave, Cincinnati, OH 45245; e-mail: adrienne​ 24. Geisthoff UW, Seyfert UT, Kübler M, Bieg B, Plinkert PK, König J. Treatment of
­.hammill@cchmc​­.org. epi­staxis in hered­i­tary hem­or­rhagic tel­an­gi­ec­ta­sia with tranexamic acid—a

476  |  Hematology 2021  |  ASH Education Program


dou­ble-blind pla­cebo-con­trolled cross-over phase IIIB study. Thromb Res. 33. Meek M, Womble P, Jordan A, Kanaan A, Meek J. Oral doxy­cy­cline for the
2014;134(3):565-571. treat­ment of epi­staxis in patients with hered­i­tary hem­or­rhagic tel­an­gi­ec­ta­
25. Canzonieri C, Centenara L, Ornati F, et al. Endoscopic eval­u­a­tion of gas­tro­ sia. Angiogenesis. 2015;18(4):530-530.
in­tes­ti­nal tract in patients with hered­i­tary hem­or­rhagic tel­an­gi­ec­ta­sia and 34. de Gussem EM, Edwards CP, Hosman AE, et  al. Life expec­tancy of par­
cor­re­la­tion with their geno­types. Genet Med. 2014;16(1):3-10. ents with hered­i­tary haemorrhagic tel­an­gi­ec­ta­sia. Orphanet J Rare Dis.
26. Kasthuri RS, Montifar M, Nelson J, et al; Brain Vascular Malformation Consor- 2016;11:46.
tium HHT Investigator Group. Prevalence and pre­dic­tors of ane­mia in hered­ 35. Iyer VN, Brinjikji W, Pannu BS, et al. Effect of cen­ter vol­ume on out­comes
i­tary hem­or­rhagic tel­an­gi­ec­ta­sia. Am J Hematol. 2017;92(10):e591-e593. in hos­pi­tal­ized patients with hered­i­tary hem­or­rhagic tel­an­gi­ec­ta­sia. Mayo
27. Serra MM, Besada CH, Cabana Cal A, et al. Central ner­vous sys­tem man­ga­ Clin Proc. 2016;91(12):1753-1760.
nese induced lesions and clin­i­cal con­se­quences in patients with hered­i­tary 36. Dupuis O, Delagrange L, Dupuis-Girod S. Hereditary haemorrhagic tel­an­
hem­or­rhagic tel­an­gi­ec­ta­sia. Orphanet J Rare Dis. 2017;12(1):92. gi­ec­ta­sia and preg­nancy: a review of the lit­er­a­ture. Orphanet J Rare Dis.
28. Livesey JA, Manning RA, Meek JH, et  al. Low serum iron lev­els are asso­ 2020;15(1):5.
ci­ated with ele­vated plasma lev­els of coag­u­la­tion fac­tor VIII and pul­mo­ 37. de Gussem EM, Lausman AY, Beder AJ, et  al. Outcomes of preg­nancy in
nary emboli/deep venous throm­bo­ses in rep­li­cate cohorts of patients with women with hered­i­tary hem­or­rhagic tel­an­gi­ec­ta­sia. Obstet Gynecol.
hered­i­tary haemorrhagic tel­an­gi­ec­ta­sia. Thorax. 2012;67(4):328-333. 2014;123(3):514-520.
29. Iyer VN, Apala DR, Pannu BS, et al. Intravenous bevacizumab for refrac­tory 38. Shovlin CL, Sodhi V, McCarthy A, Lasjaunias P, Jackson JE, Sheppard MN.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/469/1852038/469hammill.pdf by guest on 13 December 2021


hered­i­tary hem­or­rhagic tel­an­gi­ec­ta­sia-related epi­staxis and gas­tro­in­tes­ti­ Estimates of mater­nal risks of preg­nancy for women with hered­i­tary hae-
nal bleed­ing. Mayo Clin Proc. 2018;93(2):155-166. morrhagic tel­an­gi­ec­ta­sia (Osler-Weber-Rendu syn­drome): suggested
30. Al-Samkari H, Kasthuri RS, Parambil JG, et al. An inter­na­tional, mul­ti­cen­ter approach for obstet­ric ser­vices. BJOG. 2008;115(9):1108-1115.
study of intra­ve­nous bevacizumab for bleed­ing in hered­i­tary hem­or­rhagic 39. Al-Samkari H. Hereditary hem­or­rhagic tel­an­gi­ec­ta­sia: sys­temic ther­a­pies,
tel­an­gi­ec­ta­sia: the InHIBIT-Bleed study. Haematologica. 2020;106(8):2161- guide­lines, and an evolv­ing stan­dard of care. Blood. 2021;137(7):888-895.
2169.
31. McCrae K, Swaidani S, Samour M, Silver B, Parambil J, Thomas S. Pomalido-
mide in HHT: results of a pilot study. Angiogenesis. 2019;22(4):624.
32. Faughnan ME, Gossage JR, Chakinala MM, et al. Pazopanib may reduce bleed­
ing in hered­i­tary hem­or­rhagic tel­an­gi­ec­ta­sia. Angiogenesis. 2019;22(1):145- © 2021 by The Amer­i­can Society of Hematology
155. DOI 10.1182/hema­tol­ogy.2021000281

Prakash Singh Shekhawat


The ABCs of HHT  |  477
MULTIDISCIPLINARY HEMATOLOGIC DISORDERS: WHAT IS THE HEMATOLOGIST ’ S ROLE ?

Chronic thromboembolic pulmonary


hypertension: anticoagulation and beyond

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/478/1852108/478martin.pdf by guest on 13 December 2021


Karlyn A. Martin1 and Michael J. Cuttica2,3
Division of Hematology/Oncology and 2Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University
1

Feinberg School of Medicine, Chicago, IL; and 3Bluhm Cardiovascular Institute at Northwestern Memorial Hospital, Chicago, IL

Chronic thromboembolic pulmonary hypertension (CTEPH) is a rare complication in pulmonary embolism (PE) survivors,
characterized by chronic vascular occlusion and pulmonary hypertension. The identification and diagnosis of CTEPH
requires a stepwise approach, starting with symptom evaluation, functional evaluation, screening imaging, and progress-
ing to interventional hemodynamic assessment. On the backbone of anticoagulation, CTEPH management necessitates
a multidisciplinary approach. Surgical pulmonary thromboendarterectomy (PTE) is the only potentially curative option.
In nonoperable disease or residual disease after PTE, interventional balloon pulmonary angioplasty and/or pulmonary-
vasodilator therapies can be offered, in collaboration with interventional and vascular pulmonary colleagues. As it is a
disease that can cause high morbidity and mortality, CTEPH requires a high index of suspicion to diagnose and treat in
patients following PE.

LEARNING OBJECTIVES
• Understand that CTEPH is a rare complication of acute PE that has high morbidity and mortality, necessitating a
high index of suspicion
• Apply diagnostic algorithms to evaluate patients for CTEPH and understand multidisciplinary treatment strategies,
including anticoagulation, surgical and interventional treatments, and pulmonary hypertension therapy

that is associated with impaired quality of life, dyspnea,


CLINICAL CASE and reduced exercise tolerance.1,2 Post-PE syndrome is de-
A 55-year-old man with a history of unprovoked pulmo- fined as the presence of functional or cardiac impairment
nary embolism (PE) 18 months earlier sought treatment for (without another non-PE explanation), chronic thrombo-
shortness of breath. Although he initially improved, he never embolic disease (CTED), or chronic thromboembolic pul-
returned to baseline functional status. For the past 6 months, monary hypertension (CTEPH), occurring after at least 3
dyspnea on exertion has progressed despite appropriate anti- months of effective anticoagulation for acute PE.3 CTED
coagulation. His oxygen saturation is 92%. Computed tomog- and CTEPH share common features of exertional dyspnea
raphy (CT) angiogram is obtained and reported to show PE and persistent thromboembolic material in the pulmonary
(Figure 1A). Subsequent echocardiogram shows a dilated artery tree. However, CTED lacks pulmonary hypertension
right ventricle (RV) with severely reduced right ventricular (PH) at rest, whereas in CTEPH, resting PH is present, as
function (Figure 1B). He was diagnosed with submassive PE defined by a mean pulmonary artery pressure (mPAP) of
and underwent catheter directed thrombolysis. Following 25mm Hg or more, and a pulmonary capillary wedge pres-
thrombolysis, his hypoxemia worsened, and he was trans- sure of 15mm Hg or less.3 Of note, a change to decrease
ferred to our institution for consideration of embolectomy. mPAP to more than 20mm Hg to define PH has been pro-
posed but is not yet incorporated into diagnostic criteria
of CTEPH.4,5
Although a significant number of patients are diag-
Introduction nosed with CTEPH without a known prior acute PE (estima-
As many as 50% of patients have a chronic functional limita- tes vary from 25% to 67%),6,7 hematologists are more likely
tion up to 1 year after PE, termed the “post-PE syndrome,” to encounter CTEPH following acute PE. CTEPH affects

478 | Hematology 2021 | ASH Education Program


Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/478/1852108/478martin.pdf by guest on 13 December 2021
Figure 1. Clinical Imaging from a representative case. PTE, pulmonary thromboendarterectomy.

approx­ i­
ma­tely 1% to 5% of PE sur­ vi­
vors.8 One meta-anal­y­sis beyond acute RV over­load.14 Symptoms of CTEPH can be insid­
dem­on­strated the pooled inci­dence of CTEPH fol­low­ing acute i­ous, such as exer­cise intol­er­ance and dyspnea on exer­tion, or
PE to be 0.56% (95% CI, 0.1%-1.0%) for “all­-com­ers,” 3.2% (95% more severe, such as leg swell­ing, chest pain, and syn­cope that
CI, 2.0%-4.4%) for “sur­vi­vors,” and 2.8% (95% CI, 1.5%-4.1%) for can occur with right heart (RH) fail­ure. As screen­ing echo­car­dio­
“sur­vi­vors with­out major comorbidities.”8 However, when the gram in all­PE sur­vi­vors has a high false-pos­i­tive rate,15 eval­u­a­tion
expected inci­dence of CTEPH based on the num­ber of inci­dent for CTEPH should be reserved for those with symp­toms.16
acute PE cases per year is com­pared with the num­ber of pul­mo­ Numerous risk fac­tors have been his­tor­i­cally asso­ci­ated with
nary thromboendarterectomy (PTE) sur­ger­ies performed each CTEPH, includ­ing sple­nec­tomy, chronic inflam­ma­tory dis­or­
year, it is highly likely that CTEPH is underdiagnosed and there­ ders, indwell­ing cath­e­ters, ele­vated fac­tor VIII, and unpro­voked
fore undertreated. A study mod­el­ing epi­de­mi­o­logic data from or recur­ rent venous thromboembolism (VTE), among oth­ ers,
Europe, Japan, and the United States, for exam­ple, esti­mated reviewed else­where.17 More recently, in a post hoc patient-level
that only 16% of CTEPH cases would be diag­nosed in 2015, and anal­y­sis of 3 large pro­spec­tive cohorts of more than 700 PE sur­
70% or more of those diag­nosed would be in the set­ting of vi­vors, of whom 2.8% devel­oped CTEPH, pre­dic­tors of CTEPH
advanced heart fail­ure.9 Furthermore, data from both a sur­vey of were unpro­voked PE (odds ratio [OR], 20; 95% CI, 2.7 to >100),
inter­na­tional phy­si­cians and ret­ro­spec­tive claims study showed onset of symp­toms more than 14 days prior to diag­no­sis (OR, 7.9;
that diag­nos­tic tests to eval­u­ate CTEPH are under­used.10,11 As 95% CI, 3.3-19), hypo­thy­roid­ism (OR, 4.3; 95% CI, 1.4-13), and RV
CTEPH has high mor­bid­ity and mor­tal­ity—caus­ing pre­ma­ture dys­func­tion on pre­sen­ta­tion (OR, 4.1; 95% CI, 1.4-12), whereas
death in more than 50% of untreated patients within 5 years dia­be­tes mellitus and throm­bo­lytic ther­apy had infi­nitely low OR
of diag­no­sis—but has poten­tially cura­tive inter­ven­tions, a high for devel­op­ing CTEPH.18
index of sus­pi­cion is crit­i­cal to iden­tify the dis­ease.12
How to iden­tify and diag­nose CTEPH
When to sus­pect CTEPH When CTEPH/CTED is suspected, a step­wise eval­u­a­tion aims
CTEPH and/or CTED should be con­sid­ered in PE sur­vi­vors with to iden­tify pul­mo­nary vas­cu­lar dis­ease related to nonresolving
per­sis­tent dyspnea for more than 3 months after a diag­no­sis of throm­bus (Figure 2). The diag­nos­tic eval­u­a­tion also con­cur­
acute PE, despite ade­quate anticoagulation, or in those who ini­ rently allows for assess­ment of treat­ment options and sur­gi­cal
tially improve but sub­se­quently develop wors­en­ing func­tional can­di­dacy.
lim­i­ta­tions and dyspnea with exer­cise between 3 and 24 months Although no uni­ver­sally agreed-on diag­nos­tic algo­rithm
fol­low­ing acute PE. Onset of CTEPH is rare after 24 months fol­ exists to eval­u­ate for CTEPH in a patient post-PE with dyspnea,
low­ing ini­tial PE diag­no­sis.13 In addi­tion, CTEPH should be sus- we start with an echo­car­dio­gram cou­pled with a basic func­
pected if echo­ car­
dio­gram obtained for suspected acute PE tional test such as the 6-min­ ute walk test (6MWT).19 Several
shows increased RV wall thick­ness or tri­cus­pid valve peak sys­tolic guide­lines also pro­pose diag­nos­tic strat­e­gies.4,14 Notably, how­
gra­di­ent more than 60 mm Hg, both of which sug­gest changes ever, if PH is strongly suspected, then we elim­i­nate the 6MWT.
Prakash Singh Shekhawat
CTEPH: anticoagulation and beyond  |  479
Persistent symptoms despite 3 months of
ancoagulaon following acute PE

Transthoracic echocardiogram Equivocal findings; suspicion


and for CTEPH/CTED remains
6-minute walk test

Persistent or new right heart changes Cardiopulmonary exercise tesng


and/or funconal limitaon on walk test (CPET)

V/Q scan Findings suggesve of

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/478/1852108/478martin.pdf by guest on 13 December 2021


+/- CT angiogram pulmonary vascular limitaon

Refer to CTEPH center

Right heart catheterizaon with pulmonary


angiogram
+/- CT angiogram

Assessment of operability by
muldisciplinary CTEPH team

Technically operable Technically inoperable

PTE Medical therapy BPA

Figure 2. Diagnostic algorithm in evaluation for CTEPH/CTED.

Echocardiogram is used to esti­mate prob­a­bil­ity of PH, with inter­ test­ing in patients with low prob­a­bil­ity of PH on echo­car­di­og­ra­
me­di­ate to high prob­a­bil­ity suggested by esti­mated peak tri­cus­ phy but con­tin­ued sus­pi­cion for CTEPH/CTED based on symp­
pid valve regur­gi­ta­tion veloc­ity more than 2.8m/s, or 2.8 m/s toms.16 The pat­tern of increased dead space ven­ti­la­tion with a
or less associated with at least one of the following: RV end wid­en­ing A-a gra­di­ent and flat­tened stroke vol­ume in response
dia­stolic diam­et­er (EDD) more than 30 mm or RVEDD/left ven- to exer­cise sug­gests a pul­mo­nary vas­cu­lar lim­i­ta­tion and need
tricular EDD more than 0.9, hypokinesia of the RV free wall, or for fur­ther inva­sive test­ing.
exertional dyspnea.3,14 Importantly, echo­car­dio­gram allows for Once a func­tional lim­i­ta­tion and/or appro­pri­ate echo­car­dio­
screen­ing with­out expo­sure to radi­a­tion or con­trast dye.20 The graphic abnor­mal­i­ties raise suf­fi­cient con­cern for CTEPH/CTED,
6MWT is a sim­ple test that can be performed in any clinic. As the next step in eval­u­a­tion is documenting unre­solved vas­cu­lar
a patient walks for 6 min­utes, dis­tance walked, heart rate, and occlu­sion. Although a full dis­cus­sion of the roles of CT angi­og­ra­phy
oxy­gen sat­ur­a­tion by pulse oximetry are mea­sured. The 6MWT (CTA) and ven­ti­la­tion-per­fu­sion (VQ ) scan­ning in the eval­u­a­tion of
pro­vi­des a basic assess­ment of car­dio­pul­mo­nary func­tion and CTEPH is beyond the scope of this arti­cle, both imag­ing modal­i­
fit­ness, along with infor­ma­tion on func­tional, car­dio­vas­cu­lar, ties can add impor­tant infor­ma­tion in the assess­ment and deter­
and gas exchange response to exer­cise. Abnormalities in either min­ing appro­pri­ate inter­ven­tion (Table 1). While the gap between
echo­car­dio­gram or 6MWT in a symp­tom­atic patient more than sen­si­tiv­ity of V/Q and CTA scan is narrowing, VQ scan remains
3 months beyond acute PE should trig­ger fur­ther eval­u­a­tion. the pre­ferred test for screen­ing for CTEPH/CTED,4 as nonocclu-
Occasionally, more advanced exer­cise test­ing is needed to sive changes to the ves­sels that occur in CTEPH (such as mural
eval­u­ate per­sis­tent symp­toms. Formal car­dio­pul­mo­nary exer­cise thrombi, webs, and stric­tures) can be missed on rou­tine CTA reads
test­ing plays a key role in the eval­u­a­tion of dyspnea in a patient but are iden­ti­fia­ ble on VQ because of their effect on per­fu­sion to
post-PE, and guide­lines sug­gest using car­dio­pul­mo­nary exer­cise dis­tal areas of the lung.21 Furthermore, VQ has lower expo­sure

480  |  Hematology 2021  |  ASH Education Program


Table 1. Comparison of diag­nos­tic tests used in the eval­u­a­tion for CTEPH

Diagnostic test Findings in CTEPH Advantages Disadvantages/lim­i­ta­tions


Imaging
 Echocardiogram • Evidence of PH or RH strain: RV •  Non-inva­sive •  Not spe­cific or sen­si­tive to CTEPH
dila­tion, RV sys­tolic dys­func­tion, RA • No expo­sure to radi­a­tion or con­ •  Misses CTED
dila­tion, PASP ele­va­tion, flat­ten­ing of trast dye
inter­ven­tric­u­lar sep­tum
 VQ • Mismatched defects in per­fu­sion and • Highest sen­si­tiv­ity to rule out • Limited access as performed in nuclear
ven­ti­la­tion defects CTEPH med­i­cine
•  Heterogeneity of per­fu­sion •  Less radi­a­tion expo­sure •  Unable to pro­vide alter­na­tive diag­no­sis
•  No con­trast dye expo­sure •  Sensitive but not spe­cific for CTEPH
  CT PA • Pulmonary arteries: PA dila­tion, • Defining vas­cu­lar anat­omy can • CTEPH find­ings can some­times be sub­
webs/bands, eccen­tric fill­ing aid in sur­gi­cal assess­ment tle and require exper­tise and atten­tion

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/478/1852108/478martin.pdf by guest on 13 December 2021


defects, mural thrombi, lumi­nal • Provides data on screen­ing for to diag­nose
narrowing with poststenotic dila­tion, con­com­i­tant lung dis­ease/alter­ •  Exposure to IV con­trast
com­plete occlu­sion, pouch defects nate diag­no­sis that can aid in •  Exposure to radi­a­tion
•  Heart: RV dila­tion, sep­tal flat­ten­ing sur­gi­cal risk assess­ment • Less sen­si­tive than VQ (neg CT does
• Lungs: mosaic atten­u­a­tion, large not exclude CTEPH)
bron­chial artery col­lat­er­als
Invasive test­ing
  Pulmonary angio­gram •  Ring lesions/ring-like ste­no­sis • Provides com­plete hemo­dy­ • Invasive
•  Poststenotic dila­tion namic assess­ment and aids in
•  Total occlu­sion sur­gi­cal plan­ning
•  Vascular webs
  RH cath­e­ter­i­za­tion •  mPAP ≥25 mm Hg • Provides com­plete hemo­dy­ • Invasive
•  Wedge ≤15 mm Hg namic assess­ment and aids in
sur­gi­cal plan­ning
Functional test­ing
 Cardiopulmonary • Increased dead space ven­ti­la­tion • Can detect lim­i­ta­tions in func­tion •  Requires spe­cial­ized test­ing unit
exer­cise test­ing with wid­en­ing A-a gra­di­ent, flat­tened and car­dio­pul­mo­nary response • Requires arte­rial blood gas
stroke vol­ume in response to exer­cise to exer­cise mea­sure­ment
  6-min­ute walk test • Functional lim­i­ta­tions and decreased • Simple to per­form, low cost, •  Not spe­cific to CTEPH
oxy­gen sat­u­ra­tion with exer­cise min­i­mal risk
IV, intra­ve­nous; PASP, pulmonary artery systolic pressure; RA, right atrium.

to radi­a­tion and no con­trast dye expo­sure.21 Notably, how­ever, firmed marked ven­ti­la­tion per­fu­sion mis­matches (Figure 1A and
VQ scans are under­used: in 1 study, 43% of patients under­go­ing 1C). He under­went RHC with PA gram, which con­firmed CTEPH.
workup for PH did not get the recommended VQ scan.22
After asso­ci­at­ing ongo­ing symp­toms, func­tional lim­i­ta­tion, and/
or echo­car­dio­graphic changes to per­fu­sion abnor­mal­i­ties on imag­ Management con­sid­er­ations of CTEPH
ing, patients should be referred to an expe­ri­enced CTEPH cen­ter The back­bone of treat­ment of CTEPH requires life­long anticoag-
for right heart cath­e­ter­i­za­tion (RHC) with pul­mo­nary angio­gram (PA ulation to pre­vent acute VTE recur­rence. There are also 3 other
gram).16 The diag­no­sis of PH of any kind, includ­ing CTEPH, relies on treat­ment arms (detailed below) used to address the chronic
a com­pre­hen­sive hemo­dy­namic assess­ment com­pleted dur­ing an vas­cu­lar occlu­sions and PH to improve hemo­dy­nam­ics and qual­
RHC. These data are crit­i­cal for confirming the cor­rect PH diag­no­ ity of life: (1) sur­gi­cal PTE, (2) interventional bal­loon pul­mo­nary
sis and pro­vid­ing prog­nos­tic infor­ma­tion that informs treat­ment angio­plasty (BPA), and (3) pul­mo­nary vaso­di­la­tor med­i­ca­tions.
deci­sions.23 Coupling a diag­nos­tic RHC with PA gram to bet­ter Treatment strat­egy is selected based on an indi­vid­ual patient
define chronic throm­bo­em­bolic lesions not only allows for diag­ and hemo­dy­namic char­ac­ter­is­tics. Although the focus of PTE
nos­tic and prog­nos­tic infor­ma­tion but are the final data needed to and BPA is typ­i­cally patients with CTEPH, it is impor­tant to note
deter­mine sur­gi­cal can­di­dacy. that patients with CTED may also be offered sur­gi­cal or pro­ce­
dural inter­ven­tions if symp­toms affect qual­ity of life.16 Although
anticoagulation falls within the hema­ tol­
ogy exper­
tise, defin­

tive sur­gi­cal, interventional, and PH med­i­cal ther­apy for CTEPH
neces­si­tates a mul­ti­dis­ci­plin­ary team involv­ing hema­tol­ogy, car­
CLINICAL CASE (Con­t in­u ed) dio­tho­racic sur­gery, radi­ol­ogy, car­di­­ol­ogy, and pulmonology.
On arrival to our insti­tu­tion, the patient had per­sis­tence of his
chronic dyspnea despite thrombolysis and anticoagulation, Anticoagulation
which raised sus­pi­cion for CTEPH. On review of CTA by expe­ Anticoagulation is recommended as stan­dard of care for CTEPH
ri­enced radi­ol­o­gists, find­ings of eccen­tric mural thrombi and (so long as min­ i­
mal bleed­ ing risk). Of note, whether chronic
webs were more con­sis­tent with CTEPH, and VQ scan con­ anticoagulation is indi­cated in CTED is not clear, and no guide­
Prakash Singh Shekhawat
CTEPH: anticoagulation and beyond  |  481
lines offer for­mal rec­om­men­da­tions; in our prac­tice, we con­tinue image guid­ance, a bal­loon is inflated at the site of the obstruc­
anticoagulation in symp­tom­atic CTED to pre­vent VTE recur­rence tive lesion to com­ press the intra­ vas­
cu­
lar fibrotic occlu­ sion
if low bleed­ing risk. Vitamin K antag­o­nists (VKAs) are cur­rently against the wall, in an attempt to restore the vas­cu­lar lumen.
recommended as the anti­co­ag­u­lant of choice in patients with Most patients require mul­ti­ple ses­sions for BPA, with an num­
CTEPH given his­tor­i­cal expe­ri­ence, as data on direct-acting oral ber of ses­sions between 4 and 8, typ­i­cally over sev­eral months.32
anticoagulants (DOACs) are lim­ited.16 A ret­ro­spec­tive study com­ Studies have shown effi­cacy of BPA in reduc­ing pulmonary vas-
pared out­comes fol­low­ing PTE in those treated with war­fa­rin cular resistance (PVR) and increas­ing func­tional activ­ity, with low
(n = 700) and DOACs (n = 200) and found sig­nif­i­cantly higher rates rates of com­pli­ca­tions.33,34 Postprocedural com­pli­ca­tions occur
of VTE recur­rence with DOACs (4.62%/per­son-year) com­pared in approx­i­ma­tely 10% of patients, includ­ing pul­mo­nary vas­cu­lar
with VKA (0.76%/per­ son-year) (P = .008), with no dif­ fer­
ence in injury (eg, wire per­fo­ra­tion), reperfusion injury, and pul­mo­nary
over­all sur­vival and sim­i­lar rates of major bleed­ing (0.67%/per­son- hem­or­rhage. Critically, hema­tol­o­gists should be aware of BPA as
year vs 0.68%/per­son-year for VKA and DOAC, respec­tively).24 a treat­ment option and refer patients with nonoperable CTEPH
Furthermore, VTE recur­rence occurred at a median of 5.8 months to a cen­ter with BPA exper­tise.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/478/1852108/478martin.pdf by guest on 13 December 2021


(interquartile range 5.4 months) after PTE, suggesting choice of
anti­co­ag­u­lant in the first 6 months may be par­tic­u­larly impor­tant, Medical ther­apy of PH
although this requires fur­ther eval­u­a­tion. In the absence of robust Medical ther­apy of PH should be addressed by pul­mo­nary vas­
data, in our prac­tice, we treat with VKA for 6 months fol­low­ing cu­lar spe­cial­ists with exper­tise in PH. Currently, riociguat is the
PTE and then allow for switch to DOAC per patient pref­er­ence. only approved ther­apy for the treat­ment of inop­er­a­ble or resid­
Prospective stud­ies are needed to deter­mine opti­mal anticoagu- ual/recur­rent CTEPH fol­low­ing PTE. A sol­u­ble guanylate cyclase
lation strat­egy fol­low­ing PTE. In addi­tion, pulmonologists and/or stim­u­la­tor, riociguat works via the nitric oxide path­way to pro­
car­dio­tho­racic sur­geons may need hema­tol­ogy exper­tise on mote vaso­di­la­ta­tion of the pul­mo­nary arte­rial bed and has been
opti­mal anti­co­ag­u­lants in dis­tinct clin­i­cal sit­u­a­tions, such as hep­ shown to improve 6-min­ute walk dis­tance and decrease PVR.35
a­rin-induced throm­bo­cy­to­pe­nia and antiphospholipid syn­drome Published case series and clin­i­cal tri­als have explored the role
(APS). Reported rates of APS in CTEPH cohorts range from 3% of other pul­mo­nary vaso­di­la­tors such as syn­thetic pros­ta­cy­
to 7%,25 and although rou­tine thrombophilia test­ing does not af- clins and endothelin recep­tor antag­o­nists for the treat­ment of
fect man­age­ment in patients with CTEPH who require life­long CTEPH, but riociguat cur­rently remains the only approved ther­
anticoagulation, the pres­ence of APS would, given evi­dence of apy.36-38 Although the role of targeted PH ther­a­pies in patients
higher rates of recur­rent throm­bo­sis in patients with APS tak­ing with oper­ a­ble CTEPH remains unclear, lim­ ited avail­­
able data
rivaroxaban com­pared with VKA.26,27 do not sup­port a role for “pre­treat­ment” pend­ing PTE sur­gery.
Further research is needed to deter­mine whether high-risk sub­
Pulmonary thromboendarterectomy sets of patients, such as those with very ele­vated PVR, could
PTE remains the only poten­tially cura­tive option for CTEPH and ben­e­fit.39,40
thus is con­sid­ered the gold stan­dard where appro­pri­ate. All pa-
tients should be referred to an expe­ri­enced cen­ter to deter­mine How to man­age patients with CTEPH after PTE
whether obstruc­tive pul­mo­nary artery lesions are ame­na­ble to Long-term man­age­ment of CTEPH involves anticoagulation and
sur­gi­cal removal (“tech­ni­cally oper­a­ble”) and whether the patient mon­i­tor­ing for recur­rent symp­toms. Currently, all­ patients who
is a sur­gi­cal can­di­date. As PTE is tech­ni­cally chal­leng­ing, patients have a CTEPH diag­no­sis are maintained on indef­i­nite anticoagu-
should be referred to a pro­gram with an expe­ri­enced sur­geon, lation to pre­vent recur­rent VTE. The opti­mal inten­sity is unclear;
con­sid­ered to be more than 30 to 50 PTEs performed annu­ally.16 cur­rently, most are maintained on ther­a­peu­tic doses if bleed­
Between 10% and 50% of patients are deemed nonoperable,6 and ing risk is low. Although PTE is poten­tially cura­tive, up to 35%
these patients should be referred for a sec­ond opin­ion at a cen­ of patients have a mPAP of 25 mm Hg or more post-PTE.41-43 In
ter with sig­nif­i­cant expe­ri­ence. Current in-hos­pi­tal mor­tal­ity rates addi­tion, most stud­ies fol­low patients for only 5 years after PTE,
fol­low­ing PTE are less than 5% (<2% in expe­ri­enced cen­ters),28 and recur­rent PH may occur as late as 10 years after sur­gery, sug-
and sur­vival exceeds more than 90% at 1 year and more than 70% gesting inci­dence of recur­rent PH may be even higher.41 There-
at 10 years.29 Hematologists are fre­quently asked about the role fore, long-term mon­i­tor­ing is essen­tial, with par­tic­u­lar atten­tion
of inferior vena cava (IVC) fil­ters perioperatively. Although IVC fil­ on assessing for symp­toms sug­ges­tive of recur­rent dis­ease. If
ters have fallen out of favor prior to most sur­ger­ies given a lack recur­rent CTEPH occurs, options include BPA and/or med­i­cal PH
of clear mor­tal­ity ben­e­fit and increased com­pli­ca­tion risks, there ther­apy. In rare instances of recur­rent severe dis­ease, repeat PTE
are no ded­i­cated pro­spec­tive or obser­va­tional stud­ies spe­cif­i­ may be performed.
cally in the CTEPH pop­u­la­tion.30 However, given that this patient
pop­u­la­tion is com­mit­ted to life­long anticoagulation, the rou­tine Conclusion
place­ment of IVC fil­ters is not cur­rently recommended.31 CTEPH is a rare but sig­ nif­i­
cant com­pli­ ca­tion in PE sur­ vi­
vors,
with high rates of mor­bid­ity and mor­tal­ity. Therefore, it requires
Balloon pul­mo­nary angio­plasty aware­ness of and atten­tive­ness to symp­toms, which, if pres­
BPA is avail­­able for (1) inop­er­a­ble CTEPH, because of the pres­ ent, should prompt an appro­pri­ate diag­nos­tic eval­u­a­tion. Once
ence of either dis­tal lesions not ame­na­ble to sur­gi­cal inter­ven­ CTEPH is diag­nosed, man­age­ment requires mul­ti­dis­ci­plin­ary
tion (“tech­ni­cally inop­er­a­ble”) or comorbidities that pre­clude care. Although PTE remains the only poten­ tially cura­
tive op-
sur­gery, or (2) per­sis­tent or recur­rent CTEPH fol­low­ing PTE. BPA tion, BPA and med­i­cal ther­apy for PH are options for nonoper-
is performed by cath­e­ter­i­za­tion via fem­o­ral or jug­u­lar access and able CTEPH that can improve symp­toms and car­dio­pul­mo­nary
does not require gen­eral anes­the­sia or cir­cu­la­tory arrest. Using func­tion. Further stud­ies should eval­ u­ ate opti­ mal long-term

482  |  Hematology 2021  |  ASH Education Program


anti­co­ag­u­lant agent and inten­sity for both CTEPH and CTED, the 11. Tapson VF, Platt DM, Xia F, et al. Monitoring for pul­mo­nary hyper­ten­sion fol­
pre­ferred strat­egy (CTEPH vs BPA) for dif­fer­ent patient risk pro­ low­ing pul­mo­nary embolism: the INFORM study. Am J Med. 2016;129(9):978–
985.e2.
files, and role of com­bi­na­tion ther­apy. 12. Riedel M, Stanek V, Widimsky J, Prerovsky I. Longterm fol­low-up of patients
with pul­mo­nary throm­bo­em­bo­lism: late prog­no­sis and evo­lu­tion of hemo­
dy­namic and respi­ra­tory data. Chest. 1982;81(2):151-158.
13. Pengo V, Lensing AW, Prins MH, et al; Thromboembolic Pulmonary Hyper-
CLINICAL CASE (Con­t in­u ed) tension Study Group. Incidence of chronic throm­bo­em­bolic pul­mo­nary
hyper­ten­sion after pul­mo­nary embolism. N Engl J Med. 2004;350(22):2257–
Upon review of the patient’s case in a mul­ti­dis­ci­plin­ary CTEPH 2264.
con­fer­ence, he was deemed a PTE can­di­date based on clot dis­ 14. Konstantinides SV, Meyer G, Becattini C, et al; The Task Force for the diag­
tri­bu­tion, asso­ci­ated per­fu­sion abnor­mal­i­ties and hemo­dy­namic no­sis and man­age­ment of acute pul­mo­nary embolism of the Euro­pean
Society of Cardiology (ESC). 2019 ESC Guidelines for the diag­no­sis and
changes, age, and lack of comorbidities. He under­went suc­cess­ man­age­ment of acute pul­mo­nary embolism devel­oped in col­lab­o­ra­tion
ful PTE with excel­lent hemo­dy­namic response (Figure 1D and 1E). with the Euro­pean Respiratory Society (ERS): the Task Force for the diag­
Two years after PTE, he con­tin­ues on war­fa­rin. He has no symp­ no­sis and man­age­ment of acute pul­mo­nary embolism of the Euro­pean

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/478/1852108/478martin.pdf by guest on 13 December 2021


toms of dyspnea on exer­tion or func­tional lim­i­ta­tions. Society of Cardiology (ESC). Eur Respir J. 2019;54(3):1901647.
15. Klok FA, van Kralingen KW, van Dijk APJ, Heyning FH, Vliegen HW, Huisman
MV. Prospective car­dio­pul­mo­nary screen­ing pro­gram to detect chronic
throm­bo­em­bolic pul­mo­nary hyper­ten­sion in patients after acute pul­mo­
Conflict-of-inter­est dis­clo­sure nary embolism. Haematologica. 2010;95(6):970-975.
Karlyn A. Martin reports funding from Janssen Scientific Affairs for 16. Wilkens H, Konstantinides S, Lang IM, et  al. Chronic throm­ bo­em­ bolic
pul­mo­nary hyper­ten­sion (CTEPH): updated rec­om­men­da­tions from the
an inves­ti­ga­tor-ini­ti­ated study out­side the sub­mit­ted work. Cologne Consensus Conference 2018. Int J Cardiol. 2018;272S:69-78.
Michael J. Cuttica reports grants and con­sul­ting fees from Bayer, 17. Lang IM, Madani M. Update on chronic throm­bo­em­bolic pul­mo­nary hyper­
United Therapeutics, and Actelion. ten­sion. Circulation. 2014;130(6):508-518.
18. Klok FA, Dzikowska-Diduch O, Kostrubiec M, et al. Derivation of a clin­i­cal
pre­dic­tion score for chronic throm­bo­em­bolic pul­mo­nary hyper­ten­sion
Off-label drug use after acute pul­mo­nary embolism. J Thromb Haemost. 2016;14(1):121-128.
Karlyn A. Martin: nothing to disclose. 19. ATS Committee on Proficiency Standards for Clinical Pulmonary Function
Michael J. Cuttica: nothing to disclose. Laboratories. ATS state­ment: guide­lines for the six-min­ute walk test. Am J
Respirat Crit Care Med. 2002;166(1):111-117.
20. Dzikowska-Diduch O, Kostrubiec M, Kurnicka K, et al. The post-pul­mo­nary
Correspondence author syn­drome—results of echo­car­dio­graphic driven fol­low up after acute pul­
Karlyn A. Martin, Division of Hematology/Oncology, Department mo­nary embolism. Thromb Res. 2020;186:30-35.
of Medicine, Northwestern University Feinberg School of Medi- 21. Tunariu N, Gibbs SJR, Win Z, et  al. Ventilation-per­fu­sion scin­tig­ra­phy is
cine, 645 N Michigan Ave, Suite 1020, Chicago, IL 60611; e-mail: more sen­si­tive than multidetector CTPA in detecting chronic throm­bo­em­
bolic pul­mo­nary dis­ease as a treat­able cause of pul­mo­nary hyper­ten­sion. J
karlyn​­.martin@north­west­ern​­.edu. Nucl Med. 2007;48(5):680-684.
22. McLaughlin VV, Langer A, Tan M, et al; Pulmonary Arterial Hypertension-​
References Quality Enhancement Research Initiative (PAH-QuERI) Investigators. Con-
1. Kahn SR, Hirsch AM, Akaberi A, et  al. Functional and exer­cise lim­i­ta­tions temporary trends in the diag­no­sis and man­age­ment of pul­mo­nary arte­rial
after a first epi­sode of pul­mo­nary embolism: results of the ELOPE pro­spec­ hyper­ten­sion: an ini­tia­tive to close the care gap. Chest. 2013;143(2):324–
tive cohort study. Chest. 2017;151(5):1058-1068. 332.
2. Sanchez O, Helley D, Couchon S, et  al. Perfusion defects after pul­mo­ 23. Kovacs G, Dumitrescu D, Barner A, et  al. Definition, clin­i­cal clas­si­fi­ca­tion
nary embolism: risk fac­tors and clin­i­cal sig­nif­i­cance. J Thromb Haemost. and ini­tial diag­no­sis of pul­mo­nary hyper­ten­sion: updated rec­om­men­
2010;8(6):1248-1255. da­tions from the Cologne Consensus Conference 2018. Int J Cardiol.
3. Le Gal G, Carrier M, Castellucci LA, et al; ISTH CDE Task Force. Development 2018;272S:11-19.
and implementation of com­mon data ele­ments for venous throm­bo­em­bo­ 24. Bunclark K, Newnham M, Chiu Y-D, et  al. A mul­ti­cen­ter study of antico-
lism research: on behalf of SSC Subcommittee on offi­cial Communication agulation in oper­a­ble chronic throm­bo­em­bolic pul­mo­nary hyper­ten­sion. J
from the SSC of the ISTH. J Thromb Haemost. 2021;19(1):297-303. Thromb Haemost. 2020;18(1):114-122.
4. Delcroix M, Torbicki A, Gopalan D, et al. ERS state­ment on chronic throm­ 25. Bunclark K, Ansaripour A, Sheares K, Toshner M. Response to let­ter to the
bo­em­bolic pul­mo­nary hyper­ten­sion. Eur Respir J. 2021;57(6):2002828. edi­tor: direct oral anti­ co­
ag­u­
lants in throm­botic antiphospholipid syn­
5. Simonneau G, Montani D, Celermajer DS, et al. Haemodynamic def­i­ni­tions drome asso­ci­ated with chronic throm­bo­em­bolic pul­mo­nary hyper­ten­sion.
and updated clin­i­cal clas­si­fi­ca­tion of pul­mo­nary hyper­ten­sion. Eur Respir J. J Thromb Haemost. 2020;18(3):756-757.
2019;53(1):1801913. 26. Pengo V, Denas G, Zoppellaro G, et al. Rivaroxaban vs war­fa­rin in high-risk
6. Pepke-Zaba J, Delcroix M, Lang I, et al. Chronic throm­bo­em­bolic pul­mo­ patients with antiphospholipid syn­drome. Blood. 2018;132(13):1365-1371.
nary hyper­ten­sion (CTEPH): results from an inter­na­tional pro­spec­tive reg­ 27. Ordi-Ros J, Sáez-Comet L, Pérez-Conesa M, et al. Rivaroxaban ver­sus vita­
is­try. Circulation. 2011;124(18):1973-1981. min K antag­o­nist in antiphospholipid syn­drome: a ran­dom­ized noninferior-
7. Condliffe R, Kiely DG, Gibbs JS, et  al. Prognostic and aetiological fac­ ity trial. Ann Intern Med. 2019;171(10):685-694.
tors in chronic throm­bo­em­bolic pul­mo­nary hyper­ten­sion. Eur Respir J. 28. Madani MM, Auger WR, Pretorius V, et  al. Pulmonary end­ ar­
ter­
ec­tomy:
2009;33(2):332-338. recent changes in a sin­gle insti­tu­tion’s expe­ri­ence of more than 2,700
8. Ende-Verhaar YM, Cannegieter SC, Vonk Noordegraaf A, et al. Incidence of patients. Ann Thorac Surg. 2012;94(1):97-103.
chronic throm­bo­em­bolic pul­mo­nary hyper­ten­sion after acute pul­mo­nary 29. Jenkins D, Madani M, Fadel E, D’Armini AM, Mayer E. Pulmonary end­ar­ter­ec­
embolism: a con­tem­po­rary view of the published lit­er­a­ture. Eur Respir J. tomy in the man­age­ment of chronic throm­bo­em­bolic pul­mo­nary hyper­ten­
2017;49(2):1601792. sion. Eur Respir Rev. 2017;26(143):160111.
9. Gall H, Hoeper MM, Richter MJ, Cacheris W, Hinzmann B, Mayer E. An epi­ 30. Bikdeli B, Chatterjee S, Desai NR, et al. Inferior vena cava fil­ters to pre­vent
de­mi­o­log­i­cal anal­y­sis of the bur­den of chronic throm­bo­em­bolic pul­mo­ pul­mo­nary embolism: sys­tem­atic review and meta-anal­y­sis. J Am Coll Car-
nary hyper­ten­sion in the USA, Europe and Japan. Eur Respir Rev. 2017;​ diol. 2017;70(13):1587-1597.
26(143):160121. 31. Galiè N, Humbert M, Vachiery J-L, et al; ESC Scientific Document Group.
10. Gall H, Preston IR, Hinzmann B, et al. An inter­na­tional phy­si­cian sur­vey of 2015 ESC/ERS Guidelines for the diag­no­sis and treat­ment of pul­mo­nary
chronic throm­bo­em­bolic pul­mo­nary hyper­ten­sion man­age­ment. Pulm hyper­ten­sion: the Joint Task Force for the Diagnosis and Treatment of Pul-
Circ. 2016;​6(4):​472-482. monary Hypertension of the Euro­pean Society of Cardiology (ESC) and
Prakash Singh Shekhawat
CTEPH: anticoagulation and beyond  |  483
the Euro­pean Respiratory Society (ERS): endorsed by: Association for Euro­ for treat­ment of inop­er­a­ble chronic throm­bo­em­bolic pul­mo­nary hyper­ten­
pean Paediatric and Congenital Cardiology (AEPC), International Society sion: BENEFiT (Bosentan Effects in iNopErable Forms of chronIc Thrombo-
for Heart and Lung Transplantation (ISHLT). Eur Heart J. 2016;37(1):67-119. embolic pul­mo­nary hyper­ten­sion), a ran­dom­ized, pla­cebo-con­trolled trial.
32. Fukui S, Ogo T, Morita Y, et al. Right ven­tric­u­lar reverse remodelling after J Am Coll Cardiol. 2008;52(25):2127-2134.
bal­loon pul­mo­nary angio­plasty. Eur Respir J. 2014;43(5):1394-1402. 39. Reesink HJ, Surie S, Kloek JJ, et al. Bosentan as a bridge to pul­mo­nary end­
33. Ogo T, Fukuda T, Tsuji A, et  al. Efficacy and safety of bal­loon pul­mo­nary ar­ter­ec­tomy for chronic throm­bo­em­bolic pul­mo­nary hyper­ten­sion. J Tho-
angio­plasty for chronic throm­bo­em­bolic pul­mo­nary hyper­ten­sion guided rac Cardiovasc Surg. Jan 2010;139(1):85-91.
by cone-beam com­puted tomog­ra­phy and elec­tro­car­dio­gram-gated area 40. Jensen KW, Kerr KM, Fedullo PF, et  al. Pulmonary hyper­ten­sive med­ic ­ al
detec­tor com­puted tomog­ra­phy. Eur J Radiol. 2017;89:270-276. ther­apy in chronic throm­bo­em­bolic pul­mo­nary hyper­ten­sion before pul­
34. Olsson KM, Wiedenroth CB, Kamp J-C, et al. Balloon pul­mo­nary angio­plasty mo­nary thromboendarterectomy. Circulation. 2009;120(13):1248-1254.
for inop­er­a­ble patients with chronic throm­bo­em­bolic pul­mo­nary hyper­ 41. Cannon JE, Su L, Kiely DG, et al. Dynamic risk strat­i­fi­ca­tion of patient long-
ten­sion: the ini­tial Ger­man expe­ri­ence. Eur Respir J. 2017;49(6):1602409. term out­come after pul­mo­nary end­ar­ter­ec­tomy: results from the United
35. Ghofrani H-A, D’Armini A-M, Grimminger F, et al; CHEST-1 Study Group. Rio- Kingdom National Cohort. Circulation. 2016;133(18):1761-1771.
ciguat for the treat­ment of chronic throm­bo­em­bolic pul­mo­nary hyper­ten­ 42. Ogino H, Ando M, Matsuda H, et al. Jap­a­nese sin­gle-cen­ter expe­ri­ence of
sion. N Engl J Med. 2013;369(4):319-329. sur­gery for chronic throm­bo­em­bolic pul­mo­nary hyper­ten­sion. Ann Thorac
36. Sadushi-Kolici R, Jansa P, Kopec G, et al. Subcutaneous treprostinil for the Surg. 2006;82(2):630-636.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/478/1852108/478martin.pdf by guest on 13 December 2021


treat­ment of severe non-oper­a­ble chronic throm­bo­em­bolic pul­mo­nary 43. Condliffe R, Kiely DG, Gibbs JS, et al. Improved out­comes in med­i­cally and
hyper­ten­sion (CTREPH): a dou­ble-blind, phase 3, randomised con­trolled sur­gi­cally treated chronic throm­bo­em­bolic pul­mo­nary hyper­ten­sion. Am J
trial. Lancet Respir Med. 2019;7(3):239-248. Respir Crit Care Med. 2008;177(10):1122-1127.
37. Ghofrani H-A, Simonneau G, D’Armini A-M, et al; MERIT study inves­ti­ga­
tors. Macitentan for the treat­ment of inop­er­a­ble chronic throm­bo­em­bolic
pul­mo­nary hyper­ten­sion (MERIT-1): results from the multicentre, phase 2,
randomised, dou­ble-blind, pla­cebo-con­trolled study. Lancet Respir Med.
2017;5(10):785-794.
38. Jaïs X, D’Armini AM, Jansa P, et al; Bosentan Effects in iNopErable Forms of © 2021 by The Amer­i­can Society of Hematology
chronIc Thromboembolic pul­mo­nary hyper­ten­sion Study Group. Bosentan DOI 10.1182/hema­tol­ogy.2021000282

484  |  Hematology 2021  |  ASH Education Program


MULTIDISCIPLINARY HEMATOLOGIC DISORDERS: WHAT IS THE HEMATOLOGIST ’ S ROLE ?

Rebalanced hemostasis in liver disease:


a misunderstood coagulopathy

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/485/1851749/485roberts.pdf by guest on 13 December 2021


Lara N. Roberts
King’s College Hospital NHS Foundation Trust, Denmark Hill, London, United Kingdom

The combination of frequently abnormal hemostatic markers and catastrophic bleeding as seen with variceal hemorrhage
has contributed to the longstanding misperception that chronic liver disease (CLD) constitutes a bleeding diathesis. Lab-
oratory studies of hemostasis in liver disease consistently challenge this with global coagulation assays incorporating
activation of the protein C pathway demonstrating rebalanced hemostasis. It is now recognized that bleeding in CLD is
predominantly secondary to portal hypertension (rather than a coagulopathy) and additionally that these patients are at
increased risk of venous thrombosis, particularly in the portal venous system. This narrative review describes the current
understanding of hemostasis in liver disease, as well as the periprocedural management of hemostasis and anticoagula-
tion for management of venous thromboembolism in patients with CLD.

LEARNING OBJECTIVES
• Describe the changes leading to rebalanced hemostasis in CLD
• Recognize factors influencing periprocedural bleeding risk in patients with CLD
• Evaluate the need for anticoagulation of PVT in patients with liver disease and select the most suitable agent

liver failure (ACLF).1 Most patients admitted to the hospital


CLINICAL CASE are in a state of AD, often following a precipitating event.
A 46-year-old woman with a new diagnosis of presumed Common complications pertain to liver failure or portal
alcohol-related cirrhosis presents to her local hospital hypertension and comprise jaundice, encephalopathy,
with increasing abdominal swelling, pain, and jaundice. ascites, and variceal bleeding. Although bleeding is com-
She had been admitted 1 year prior with alcohol with- mon in liver disease, it is now recognized that most sponta-
drawal but did not attend planned hepatology outpa- neous bleeding events are predominantly gastrointestinal
tient reviews. She has continued to drink 0.5 L of vodka (GI) and secondary to portal hypertension (refer to Table 1
daily. Initial laboratory investigation reveals the follow- for incidence).2-4
ing: hemoglobin, 90 g/L; platelets, 43 × 109/L; bilirubin, The liver has a key role in the synthesis of both pro- and
199 µmol/L; albumin, 26 g/L; international normalized anticoagulant proteins, along with pro- and antifibrinolyt-
ratio (INR), 2.2; prothrombin time (PT), 33 seconds; and ics and thrombopoietin.9 Portal hypertension in CLD leads
creatinine, 126 µmol/L. The Child-Turcotte-Pugh class is to splenomegaly and thrombocytopenia. Progression of
C and Model for End Stage Liver Disease score is 25. She CLD with synthetic failure is accompanied by prolonga-
commences spironolactone but there is no improvement tion of PT.10 As PT simply measures time to first detection
in ascites or weight, and her renal function further deteri- of clot in plasma following activation with tissue factor and
orates. Therapeutic paracentesis is planned and the med- calcium, it does not reflect in vivo hemostasis in patients
ical team requests authorization of fresh-frozen plasma with CLD in which natural anticoagulants (protein C, pro-
(FFP) (aiming for an INR of <1.5) to proceed. tein S, and antithrombin) are reduced in parallel, and it does
not detect increased factor VIII and von Willebrand factor.
Global coagulation assays, such as thrombin generation
The natural history of chronic liver disease (CLD) is charac- incorporating activation of the protein C pathway, demon-
terized by phases of stable cirrhosis progressing to acute strate that patients with CLD have rebalanced hemostasis
decompensation (AD), from which patients may recover or with some evidence of hypercoagulability. With increasing
progress to multiorgan failure, known as acute-on-chronic disease severity (as in ACLF), wide interindividual variation
Prakash Singh Shekhawat
Rebalanced hemostasis in liver disease | 485
Table 1. Incidence of bleed­ing and por­tal vein throm­bo­sis in CLD reported in pro­spec­tive stud­ies

Child-Pugh
Study set­ting Study design Participants, n class A, n (%) Incidence
Bleeding
  Italy, 2012-20143 Prospective, mul­ti­cen­ter 280 53 Overall: 5.5%/year
Major: 3.6%/year
Minor: 1.9%/year
Portal hyper­ten­sive: ~3.3%/year
Nonvariceal GI: 1.9%/year
Intracranial: 0.17%/year
  United States, Decem­ber Prospective, sin­gle-cen­ter 83 0 Overall: 40%
2009 to Jan­u­ary 20104 hos­pi­tal­ized cohort Variceal: 17%
Nonvariceal GI: 12%

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/485/1851749/485roberts.pdf by guest on 13 December 2021


Portal vein throm­bo­sis
  Italy, 2012-20145 Prospective, mul­ti­cen­ter 753 397 (53) 6.1/100 patient years
  No PVT at enroll­ment 692 4.1
  Prior PVT at enroll­ment 61 18.9
  France/Belgium, 2000-2006 6
Prospective, mul­ti­cen­ter 1243 863 (69) 1 y: 4.6%
Cumulative 5 y: 10.7%
  France, 2014-20177 Prospective, sin­gle cen­ter 108 75 (77) 1 y: 10.8%
  Portugal, 2014-20198 Prospective, sin­gle cen­ter 241 184 (76) 1 y: 3.7%
Cumulative 3 y: 7.6%
Modified from Roberts and Bernal (2020)2 with per­mis­sion from Thieme.

in throm­bin gen­er­a­tion poten­tial is seen with a more pre­car­i­ous in the pro­ce­dural set­ting.18,19 In Spain, a national sur­vey (of pre­
hemostatic bal­ance.10-12 Similarly, throm­bo­cy­to­pe­nia and plate­let dom­i­nantly hepatologists, 69%) from 2017 reported the major­
dys­func­tion are coun­tered by increased von Willebrand fac­tor, ity of respon­dents would attempt to cor­rect PT prior to major
with increased high molec­u­lar weight multimers due to reduced pro­ce­dures, with 17% also attempting to cor­rect prior to low-
ADAMTS13.13,14 The changes in hemo­sta­sis asso­ci­ated with CLD risk pro­ce­dures.19 Almost all­respon­dents reported attempting
are sum­ma­rized in Table 2. to cor­rect throm­bo­cy­to­pe­nia prior to major sur­gery, with 35%
also attempting to cor­rect prior to low-risk pro­ce­dures, pre­dom­
Periprocedural bleed­ing risk i­nantly using a thresh­old for plate­lets of less than 50 × 109/L. How-
It has been repeat­edly dem­on­strated over the past 40 years that ever, attempts to cor­rect throm­bo­cy­to­pe­nia and coagulopathy
nei­ther prolonged PT/INR nor throm­bo­cy­to­pe­nia inde­pen­dently with trans­fu­sion are asso­ci­ated with addi­tional risks. Administra-
pre­dict periprocedural bleed­ing.9,15-17 These param­e­ters con­tinue tion of a “ther­a­peu­tic” vol­ume of FFP pre­dis­poses to both trans­
to be mea­sured with attempts at cor­rec­tion com­mon in this set­ fu­sion-asso­ci­ated cir­cu­la­tory over­load and trans­fu­sion-related
ting, despite poten­tial for harm asso­ci­ated with trans­fu­sion.18,19 acute lung injury.25 Furthermore, due to sub­se­quent increases in
Procedural bleed­ ing risk in patients with CLD is not well por­tal venous pres­sure,26 FFP admin­is­tra­tion may par­a­dox­i­cally
defined; var­i­able prac­tice in attempting to cor­rect hemo­static increase the risk of bleed­ing (eg, dur­ing endo­scopic variceal
mark­ers and a lack of con­sis­tent def­i­ni­tion for major bleed­ing band liga­tion or transjugular portosystemic shunt place­ment).
are key con­trib­u­tory fac­tors.20 It is pro­posed that pro­ce­dures In addi­tion, lab­o­ra­tory stud­ies dem­on­strate that patients with
with an inci­dence of major bleed­ing of more than 2% or bleed­ CLD have preexisting nor­mal to hyper­co­ag­u­la­ble pro­files, with
ing with poten­tial to cause organ dam­age/death be con­sid­ered FFP trans­fu­sion hav­ing a min­i­mal impact on improv­ing PT but
high risk.20 Ascites is a fre­quent com­pli­ca­tion of CLD, devel­op­ing increas­ing hyper­co­ag­u­la­bil­ity.27,28 Platelet trans­fu­sion results in
in approaching two-thirds of patients within 10 years of cir­rho­sis var­i­able plate­let count incre­ment,28,29 and ran­dom­ized con­trolled
diag­no­sis.21 Therapeutic paracentesis is recommended as first- tri­als of plate­let trans­fu­sion in other pop­u­la­tions (eg, intra­ce­re­
line treat­ment for large-vol­ume asci­tes, and this sce­nario is com­ bral bleed­ing) are asso­ci­ated with worse out­comes.30 Throm-
monly encoun­tered. The risk of bleed­ing fol­low­ing paracentesis bopoietin recep­tor ago­nists have been dem­on­strated to bet­ter
is low, esti­mated at 0.2%.22 Acute kid­ney injury is a rec­og­nized cor­rect plate­let count prior to elec­tive pro­ce­dures.31,32 However,
inde­pen­dent risk fac­ tor for postparacentesis hematoperito- the sem­i­nal stud­ies included both low- and high-risk pro­ce­dures
neum.23 and lacked clin­i­cally impor­tant pri­mary out­comes. Meta-anal­y­sis
sug­gests their use may reduce periprocedural bleed­ing.33
Prophylactic hemo­static inter­ven­tions Given the risks asso­ci­a­tion with plasma trans­fu­sion and lack
Despite evi­
dence that major sur­ gery such as liver trans­plan­ of evi­dence to dem­on­strate ben­e­fit, cor­rec­tion of prolonged PT
ta­
tion can be safely performed with­ out correcting abnor­ mal prior to paracentesis has been advised against since 2003 by
hemo­static mark­ers,24 attempts at cor­rec­tion remain com­mon inter­na­tional soci­e­ties. Recommendations for high-risk bleed­ing

486  |  Hematology 2021  |  ASH Education Program


Table 2. Summary of hemostatic changes in CLD and lab­o­ra­tory evi­dence supporting rebalanced hemo­sta­sis

Laboratory evi­dence to sup­port


Characteristic Factors asso­ci­ated with bleed­ing Factors asso­ci­ated with throm­bo­sis rebalanced hemo­sta­sis
Platelets • Thrombocytopenia •  ↑ vWF • Weak
•  Platelet dys­func­tion • ↓ADAMTS13 • Difficult to rep­li­cate plate­let-
• Anemia •  ↑Platelet acti­va­tion endo­the­lial inter­ac­tion in lab­o­ra­tory
•  Endothelial acti­va­tion set­ting
Coagulation •  ↓FII, V, VII, IX, X, XI • ↓Protein C, pro­tein S, anti­throm­bin • Thrombin gen­er­a­tion nor­mal to
•  ↓Fibrinogen (in AD, ACLF) • ↑FVIII increased when mea­sured with pro­
• ↓FXIII •  ↓Fibrin clot per­me­abil­ity tein C path­way acti­va­tion
•  ↓Rates of fibrin poly­mer­i­za­tion • Weak evi­dence for rebalancing of
fibrin clot strength/sta­bil­ity
Fibrinolysis •  ↑tPA (not rebalanced by ↑PAI-1) • ↓Plasminogen • Uncertain; both

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/485/1851749/485roberts.pdf by guest on 13 December 2021


• ↓α2-Antiplasmin hyper/hypofibrinolysis seen in
• ↓TAFI ACLF, poten­tially reflecting the del­i­
cate nature of rebalance
Adapted from Northup et al20 with per­mis­sion from Wiley.
ADAMTS13, a disintegrin and metalloproteinase thrombospondin type 1 motif; F, fac­tor; PAI-1, plas­min­o­gen acti­va­tor inhib­i­tor 1; TAFI, throm­bin
activatable fibri­no­ly­sis inhib­i­tor; tPA, tis­sue plas­min­o­gen acti­va­tor; vWF, von Willebrand fac­tor.

pro­ce­dures are sum­ma­rized in Table 3.20,34-36 Vitamin K defi­ciency cir­rho­sis was asso­ci­ated with an increased odds ratio for venous
may be rel­e­vant in those with poor diet and/or mal­ab­sorp­tion, throm­bo­em­bo­lism (VTE) of 1.7 (95% CI, 1.3-2.2).39 The inci­dence
and a sin­gle dose of 10 mg more than 12 hours prior to inter­ven­ of PVT in recent pro­spec­tive stud­ies is sum­ma­rized in Table 1.
tions in such patients is advo­cated by some.35,36 The Ital­ian obser­va­tional cohort of 753 patients with cir­rho­sis
reported an inci­dence rate of 6 per 100 patient years.5 Of note,
half of the events were asymp­tom­atic and detected on rou­tine
screen­ing for hepa­to­cel­lu­lar car­ci­noma. Those with prior PVT at
study entry had a sig­nif­i­cantly higher rate of PVT com­pared with
CLINICAL CASE (Con­t in­u ed) those with­out (18.9 vs 4.0 per 100 patient years).5 The only inde­
This patient received a sin­gle dose of vita­min K on admis­sion pen­dent risk fac­tors for PVT iden­ti­fied in this study were p ­ re­vi­ous
with no improve­ment in the PT. After reassuring the med­i­cal PVT and degree of throm­bo­cy­to­pe­nia (with lower counts asso­
team that the pro­ce­dural bleed­ing risk of paracentesis is low ci­ated with increased risk). A fur­ther pro­spec­tive obser­va­tional
and that PT/INR and plate­let count are not a mea­sure of bleed­ cohort of 241 patients with cir­rho­sis reported a cumu­la­tive inci­
ing risk, ther­ a­
peu­tic paracentesis is performed under ultra­ dence of 3.7% and 7.6% at 1 and 3 years, respec­tively.8 Throm-
sound guid­ance with­out com­pli­ca­tion. bocytopenia and pre­vi­ous decom­pen­sa­tion were iden­ti­fied as
She recov­ers from this acute epi­sode of decom­pen­sa­tion and inde­pen­dent pre­dic­tors of PVT. Of note, in both cohorts, most
remains absti­nent from alco­hol fol­low­ing hos­pi­tal dis­charge. On patients were Child-Pugh class A (see Table 1).
rou­tine screen­ing ultra­sound for hepa­to­cel­lu­lar car­ci­noma 2 This sce­nario illus­trates again that con­ven­tional lab­o­ra­tory
years later, she is diag­nosed with occlu­sive por­tal vein throm­bo­ tests do not reflect the under­ly­ing hemo­static milieu in patients
sis (PVT) extending to the supe­rior mes­en­teric venous junc­tion. with liver dis­ease. The par­a­dox­i­cal increase in PVT risk asso­ci­
The spleen is enlarged (20 cm), and the patient has the fol­low­ ated with throm­bo­cy­to­pe­nia likely reflects increas­ing sever­ity of
ing lab­o­ra­tory val­ues: hemo­glo­bin, 110  g/L; plate­lets, 57  ×  109/L; both liver dis­ease and por­tal hyper­ten­sion with reduced por­tal
bil­i­ru­bin, 16 µmol/L; albu­min, 40 g/L; PT, 18 sec­onds; INR, 1.3; venous flow.8,40 Only a sin­gle, small ret­ro­spec­tive study (n = 53)
sodium, 138 µmol/L; and cre­at­i­nine, 116 µmol/L. She is Child- has exam­ined the influ­ence of hyper­co­ag­u­la­bil­ity mea­sured
Turcotte-Pugh class A, and Model for End Stage Liver Disease with throm­ bin gen­ er­a­
tion on PVT risk.41 Patients with base­
score is 13. An upper GI endos­copy performed dur­ing the pre­ line “thrombomodulin resis­tance” (defined as an endog­e­nous
vi­ous admis­sion revealed mild por­tal hyper­ten­sive gastropathy throm­bin poten­tial ratio >95th per­cen­tile of nor­mal con­trols) had
with no esoph­a­geal varices. The med­i­cal team seek advice on an 8-fold increase in risk of PVT over 4 years. Local hyper­co­ag­
the role for anticoagulation in the con­text of throm­bo­cy­to­pe­nia. u­la­bil­ity medi­ated by bac­te­rial trans­lo­ca­tion, inflam­ma­tion, and
endotoxemia are pro­posed as addi­tional con­trib­u­tors but have
not been con­firmed in pro­spec­tive stud­ies.42
Venous throm­bo­em­bo­lism in CLD
PVT is the most com­mon throm­botic event affect­ing patients Anticoagulation in CLD
with CLD, with its prev­a­lence reported in up to 26% in those The need for anticoagulation in this sce­nario of inci­den­tal PVT in
listed for liver trans­plan­ta­tion and increas­ing in par­al­lel with dis­ the absence of intes­ti­nal ische­mia or planned liver trans­plan­ta­
ease sever­ity.38 CLD is also asso­ci­ated with an increased risk of tion (in which anas­to­mo­sis may be com­pro­mised) is uncer­tain.
deep vein throm­bo­sis (DVT) and pul­mo­nary embolism. A meta- A recent meta-anal­y­sis of 33 stud­ies com­pris­ing 1696 patients
anal­y­sis of pre­dom­i­nantly ret­ro­spec­tive cohort stud­ies reported with PVT reported spon­ta­ne­ous com­plete recan­a­li­za­tion in 18%
Prakash Singh Shekhawat
Rebalanced hemo­sta­sis in liver dis­ease  |  487
Table 3. Thresholds for coag­ul­a­tion param­e­ters prior to high-risk pro­ce­dures in patients with CLD

Characteristic AASLD 202120 ACG 202037 AGA 201935 SIR 201936


PT/INR Do not cor­rect Do not cor­rect Do not cor­rect INR >2.5*
Platelet count Do not cor­rect >50 × 10 /L
9
>50 × 10 /L
9
>30 × 109/L
Fibrinogen Do not cor­rect No spe­cific rec­om­men­da­tion >1.2 g/L >1 g/L
VHA Do not use rou­tinely May be use­ful No spe­cific rec­om­men­da­tion No spe­cific rec­om­men­da­tion
*Give vita­min K if INR >2.5; do not use FFP/prothrombin complex concentrate.
Table adapted from Northup et al20 with per­mis­sion from Wiley.
AASLD, Amer­i­can Association for the Study of Liver Disease; ACG, Amer­i­can College of Gastroenterology; AGA, Amer­i­can Gastroenterology
Association; SIR, Society of Interventional Radiology; VHA, vis­co­elas­tic hemo­static assay.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/485/1851749/485roberts.pdf by guest on 13 December 2021


of patients (n  = 
33/180) not receiv­ ing anticoagulation. How- Thrombocytopenia is a rec­og­nized risk fac­tor for bleed­ing
ever, anticoagulation was asso­ci­ated with a sig­nif­i­cant 2-fold with anticoagulation; a small obser­va­tional cohort reported a
improve­ment in over­all and com­plete recan­a­li­za­tion rates com­ plate­let count of less than 50 × 109/L as a pre­dic­tor for bleed­
pared with no anticoagulation. The risk of throm­bus pro­gres­sion ing.46 In these patients, I con­sider ini­ti­at­ing LMWH at pro­phy­
was also sig­nif­i­cantly reduced (risk ratio [RR], 0.26; 95% CI, 0.14- lac­tic to inter­me­di­ate doses and titrate the dose as tol­er­ated. I
0.49). Six stud­ies reported over­all sur­vival, with anticoagulation favor inter­me­di­ate doses for symp­tom­atic and/or exten­sive PVT.
asso­ci­ated with improve­ment (RR, 1.11; 95% CI, 1.03-1.21).43 There For lim­ited extent/asymp­tom­atic events in which a deci­sion for
are clearly a num­ber of lim­it­a­tions to this anal­y­sis. In our case, anticoagulation is made, I favor pro­phy­lac­tic doses at ini­ti­a­tion.
given the throm­bus is occlu­sive and extends to the mes­en­teric Severe throm­bo­cy­to­pe­nia (<30 × 109/L) is uncom­ mon, and in
junc­tion, anticoagulation is likely to be ben­e­fi­cial in pre­vent­ing such cases, I would look for and treat con­trib­ut­ing fac­tors but
fur­ther exten­sion to the mes­en­teric vas­cu­la­ture and achiev­ing still con­sider pro­phy­lac­tic LMWH on a case-by-case basis. If the
recan­a­li­za­tion, with poten­tial reduc­tion in por­tal hyper­ten­sion plate­let count improves, ther­ap ­ eu­tic doses can then be used. As
(given the lack of portosystemic col­lat­eral cir­cu­la­tion). Guide- this patient has a plate­let count more than 50 × 109/L, full-dose
lines rec­om­mend obtaining cross-sec­tional imag­ing (com­puted anticoagulation should be used.
tomog­ra­phy/mag­netic res­o­nance imag­ing) to con­firm the diag­ For venous throm­bo­em­bo­lism in patients with­out CLD, direct
no­sis and extent of throm­bo­sis, as well as to exclude malig­nant oral anti­co­ag­u­lants (DOACs) are now the agents of choice due
obstruc­tion prior to treat­ment ini­ti­a­tion.20 There is a clear role to their pre­dict­able phar­ma­co­ki­net­ics, elim­i­nat­ing the need for
for anticoagulation in patients with symp­tom­atic PVT to pre­vent rou­tine drug mon­i­tor­ing and favor­able safety pro­file.47 Charac-
pro­gres­sion and intes­ti­nal ische­mia. teristics of avail­­able anti­co­ag­u­lants are sum­ma­rized in Table 4.
The prin­ci­pal con­cern in anticoagulating patients with liver Of note, patients with CLD were excluded from the ran­dom­ized
dis­ease is the con­com­i­tant increased risk of bleed­ing. In the con­trolled tri­als establishing their effi­cacy in VTE, and there is
afore­men­tioned meta-anal­y­sis, the pooled over­all rate of bleed­ lit­tle evi­dence for use of DOACs in treat­ment of PVT in cir­rho­sis.
ing asso­ci­ated with anticoagulation from 19 stud­ies was 2.8%, The use of DOACs is not recommended in patients with increased
with fatal bleed­ing 0.7% (from 25 stud­ies).43 The pooled rate of sever­ity of CLD (see Table 4). A recent sys­tem­atic review iden­ti­
esoph­a­geal variceal bleed­ing from 17 stud­ies was 2%. The use fied 5 ret­ro­spec­tive cohort stud­ies com­pris­ing 239 patients with
of anticoagulation did not increase the risk of bleed­ ing (RR, CLD treated for DVT, splanch­nic vein throm­bo­sis, or atrial fibril­la­
0.78; 95% CI, 0.47-1.3) from 4 stud­ ies reporting out­ comes in tion.48 The anal­y­sis sug­gests com­pa­ra­ble effi­cacy and safety but
those both on and off anticoagulation, and inter­est­ingly, anti- is lim­ited by the small sam­ple size and ret­ro­spec­tive nature of
coagulation was asso­ci­ated with a reduced risk of both upper the stud­ies. A small ran­dom­ized con­trolled trial (n = 80) in Egypt
GI and variceal bleed­ing (RR, 0.26; 95% CI, 0.11-0.65).43 These com­pared rivaroxaban 10 mg daily with war­fa­rin for treat­ment
data sug­gest anticoagulation does not increase the risk of vari- of acute PVT in patients with com­pen­sated hep­a­ti­tis C cir­rho­
ceal hem­or­rhage (and may in fact reduce the risk, poten­tially by sis (pre­dom­i­nantly after sple­nec­tomy) for a var­i­able treat­ment
reduc­ing por­tal hyper­ten­sion). It is recommended that upper GI dura­tion based on throm­bus extent and recan­a­li­za­tion.49 Patients
endos­copy be performed to iden­tify and enable treat­ment of were mon­i­tored fortnightly with ultra­sound imag­ing with a pri­
high-risk varices.20 In prac­tice, anticoagulation can be ini­ti­ated mary out­come of com­plete recan­a­li­za­tion. The pri­mary out­come
while awaiting fur­ther endo­scopic eval­u­a­tion. Endoscopic vari- was achieved in sig­nif­i­cantly more patients receiv­ing rivarox-
ceal banding is a low bleed­ing risk pro­ce­dure,20 with pro­ce­dural aban (85% vs 45%), with recan­a­li­za­tion occur­ring at a mean of 2.5
bleed­ing uncom­mon and the major­ity of bleed­ing sec­ond­ary to months. There was no major bleed­ing reported in the rivaroxaban
band ulcer­at­ion, presenting some days later.15,44 A sin­gle obser­ arm and no recur­rent events at 12 months. A fur­ther small study
va­tional study found no increased bleed­ing risk asso­ci­ated with com­par­ing out­comes in those treated with edoxaban (n = 20) to
use of low molec­u­lar weight hep­a­rin (LMWH) (169 pro­ce­dures war­fa­rin (n = 30) fol­low­ing 2 weeks of danaparoid also reported
in 80 patients) at the time of variceal band liga­tion.45 However, supe­rior clot regres­sion with edoxaban.50 Although these find­
proceduralists may tem­po­rar­ily inter­rupt anticoagulation fol­low­ ings are prom­is­ing, it is impor­tant to note that nei­ther war­fa­rin
ing variceal banding when the per­ceived bleed­ing risk due to arm pro­vided stan­dard of care in that there was no bridg­ing until
sec­ond­ary band ulcer­a­tion is thought to out­weigh the risk of ther­a­peu­tic INR was reached, with a sub­ther­a­peu­tic INR tar­get
throm­bo­sis exten­sion. (1.5-2.0) in 1 study. This is impor­tant as ­recan­a­li­za­tion rates are

488  |  Hematology 2021  |  ASH Education Program


Table 4. Available anti­co­ag­u­lants and con­sid­er­ations for use in man­age­ment of VTE in CLD

Characteristic VKA LMWH Dabigatran Apixaban Edoxaban Rivaroxaban


Dosing Variable od, based on Weight based, once or Twice daily Twice daily Once daily Twice daily for 3 weeks,
INR twice daily then once daily
Initiation Overlap with LMWH until As above ≥5 days LMWH, then 10 mg twice daily for 5 days LMWH, then switch 15 mg twice daily for 3
INR >2. Use alter­nate switch 7 days weeks
agent if base­line INR
increased
Standard dose Variable as above As above 150 mg bd 5 mg twice daily from 60 mg od 20 mg once daily from
day 8 day 22
Dose reduc­tion NA CrCl 15-30 mL, reduce 110 mg bd for age ≥80 Consider dose reduc­tion 30 mg od for CrCl <50 mL/min Consider dose reduc­tion
dose years or con­com­i­tant to 2.5 mg bd from or weight <60 kg to 10 mg from
verap­a­mil and con­sider 6 months 6  months
for other high-risk
groups
Drug mon­i­tor­ing Y N N N N N
Hepatobiliary/intes­ti­nal Predominant Minimal 20% 73% 50% 34%
elim­i­na­tion

Prakash Singh Shekhawat


CLD
CPC A Y Y Y Y Caution, avoid if ALT/AST Y
<2 × ULN and bil­i­ru­bin
CPC B Y Y Limited expe­ri­ence, no Y in absence of N
<1.5 × ULN
change in expo­sure coagulopathy
in n = 12
CPC C Y Y N N N N
CKD
CrCl, 15-30 mL/min Y ↓Dose Do not use Limited expe­ri­ence Limited expe­ri­ence Limited expe­ri­ence
<15 mL/min Y Avoid Do not use Do not use Do not use Do not use
Modified from Northup et al20 with per­mis­sion from Wiley.
ALT, ala­nine ami­no­trans­fer­ase; AST, aspar­tate ami­no­trans­fer­ase; bd, twice daily; CKD, chronic kid­ney dis­ease; CPC, Child-Pugh clas­si­fi­ca­tion; CrCl, cre­at­i­nine clear­ance; N, no; NA, not applicable;
od, once daily; ULN, upper limit of nor­mal; Y, yes.

Rebalanced hemo­sta­sis in liver dis­ease  |  489


Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/485/1851749/485roberts.pdf by guest on 13 December 2021
improved with early anticoagulation.46 Further ade­quately pow- most fre­quent bleed­ing man­i­fes­ta­tion. There is lit­tle evi­dence
ered stud­ies with the use of LMWH bridg­ing until ther­a­peu­tic to sup­port pro­phy­lac­tic cor­rec­tion of hemo­static mark­ers in the
INR is achieved in the com­par­a­tor arm are required. In prac­tice, periprocedural set­ting, and use of pre­emp­tive trans­fu­sion sup­
LMWH is often ini­ti­ated until com­ple­tion of upper GI endos­copy, port should be avoided. Anticoagulation is likely to improve out­
fol­low­ing which a vita­min K antag­o­nist (VKA) can be ini­ti­ated. comes fol­low­ing occlu­sive por­tal vein throm­bo­sis, even in the
DOACs may be a rea­son­able alter­na­tive in cases with dif­fi­culty pres­ence of hemo­static abnor­mal­i­ties. However, the opti­mal
com­ply­ing to var­i­able dos­ing of VKA or the mon­i­tor­ing require­ dura­tion of anticoagulation and role of DOACs remains uncer­tain.
ments (eg, lim­ited venous access and/or unable to use a point-
of-care INR device). For patients with sig­nif­i­cantly prolonged PT Conflict-of-inter­est dis­clo­sure
at base­line, I favor extended LMWH pro­vided con­tin­ued par­ Lara N. Roberts: no com­pet­ing finan­cial inter­ests to declare.
en­teral admin­is­tra­tion is accept­able to the patient, given the
opti­mal INR tar­get is unknown in those with sig­nif­i­cantly raised Off-label drug use
base­line INR. Lara N. Roberts: nothing to disclose.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/485/1851749/485roberts.pdf by guest on 13 December 2021


For PVT, in which a deci­sion to ini­ti­ate anticoagulation is made,
the min­i­mum recommended dura­tion of anti­co­ag­u­lant ther­apy is Correspondence
6 months.20,37 Guidelines sug­gest reimaging to eval­ua ­ te recan­a­ Lara N. Roberts, King’s Thrombosis Centre, Department of Haema-
li­za­tion to guide the ongo­ing need for anticoagulation.20 There tological Medicine, King’s College Hospital NHS Foundation Trust,
are, how­ever, no high-qual­ity data to inform whether incom­plete Denmark Hill, London SE5 9RS, UK; e-mail: lara​­.roberts@nhs​­.net.
recan­al­i­za­tion is asso­ci­ated with a higher risk of recur­rence or
other adverse out­comes related to por­tal hyper­ten­sion. Small References
obser­va­tional stud­ies sug­gest the risk of early recur­rent PVT is 1. D’Amico G, Garcia-Tsao G, Pagliaro L. Natural his­tory and prog­nos­tic indi­
high (up to 38%) fol­low­ing dis­con­tin­u­a­tion of anticoagulation ca­tors of sur­vival in cir­rho­sis: a sys­tem­atic review of 118 stud­ies. J Hepatol.
2006;44(1):217-231.
in patients with advanced CLD.42,46 In addi­tion, pre­vi­ous PVT is 2. Roberts LN, Bernal W. Incidence of bleed­ing and throm­bo­sis in patients
a strong pre­dic­tor for recur­rence in pro­spec­tive obser­va­tional with liver dis­ease. Semin Thromb Hemost. 2020;46(6):656-664.
cohorts.5 However, given the lack of ran­dom­ized con­trolled tri­ 3. Basili S, Raparelli V, Napoleone L, et al; PRO-LIVER Collaborators. Platelet
als to defin­i­tively inform the risk of both PVT recur­rence and count does not pre­dict bleed­ing in cir­rhotic patients: results from the PRO-
LIVER study. Am J Gastroenterol. 2018;113(3):368-375.
bleed­ing, the fea­si­bil­ity of a mul­ti­cen­ter pro­spec­tive study to
4. Shah NL, Northup PG, Caldwell SH. A clin­i­cal sur­vey of bleed­ing, throm­
deter­ mine the opti­ mal dura­ tion of anticoagulation should be bo­sis, and blood prod­uct use in decompensated cir­rho­sis patients. Ann
eval­u­ated. Currently, those listed for liver trans­plan­ta­tion fre­ Hepatol. 2012;11(5):686-690.
quently remain on extended anticoagulation to reduce the risk 5. Violi F, Corazza GR, Caldwell SH, et al; PRO-LIVER Collaborative Group.
of recur­rent PVT, par­tic­ul­arly as the pres­ence of occlu­sive PVT Incidence and recur­rence of por­tal vein throm­bo­sis in cir­rhotic patients.
Thromb Haemost. 2019;119(3):496-499.
at trans­plan­ta­tion affects both tech­ni­cal suc­cess and posttrans- 6. Nery F, Chevret S, Condat B, et al; Groupe d’Etude et de Traitement du
plant sur­vival.51 For patients with DVT/pul­mo­nary embolism, the Carcinome Hépatocellulaire. Causes and con­ se­
quences of por­ tal vein
min­im ­ um dura­tion of anticoagulation is 3 months. All patients throm­bo­sis in 1,243 patients with cir­rho­sis: results of a lon­gi­tu­di­nal study.
should be reviewed prior to com­plet­ing 3 months of treat­ment Hepatology. 2015;61(2):660-667.
7. Nery F, Correia S, Macedo C, et al. Nonselective beta-block­ers and the risk
to con­sider the need for extended anticoagulation in the pres­
of por­tal vein throm­bo­sis in patients with cir­rho­sis: results of a pro­spec­tive
ence of per­sis­tent risk fac­tors (eg, inflam­ma­tory bowel dis­ease) lon­gi­tu­di­nal study. Aliment Pharmacol Ther. 2019;49(5):582-588.
and/or for events not asso­ci­ated with a major pro­vok­ing fac­tor 8. Noronha Ferreira C, Marinho RT, Cortez-Pinto H, et al. Incidence, pre­dic­tive
(eg, major sur­gery/hos­pi­tal­i­za­tion).52 fac­tors and clin­i­cal sig­nif­i­cance of devel­op­ment of por­tal vein throm­bo­sis
in cir­rho­sis: a pro­spec­tive study. Liver Int. 2019;39(8):1459-1467.
9. Lisman T, Hernandez-Gea V, Magnusson M, et al. The concept of rebal-
anced hemostasis in patients with liver disease: Communication from the
ISTH SSC working group on hemostatic management of patients with liver
CLINICAL CASE (Con­t in­u ed) disease. Thromb Haemost. 2021;19(4):1116-22.
Therapeutic anticoagulation with LMWH was ini­ti­ated while 10. Fisher C, Patel VC, Stoy SH, et al. Balanced haemostasis with both hypo- and
hyper-coag­u­la­ble fea­tures in crit­i­cally ill patients with acute-on-chronic-
fur­
ther endos­copy was planned (unchanged from pre­ vi­
ous,
liver fail­ure. J Crit Care. 2018;43:54-60.
with no esoph­a­geal varices seen with mild por­tal hyper­ten­sive 11. Gatt A, Riddell A, Calvaruso V, Tuddenham EG, Makris M, Burroughs AK.
gastropathy). Following this, war­fa­rin was ini­ti­ated with a tar­ Enhanced throm­bin gen­er­a­tion in patients with cir­rho­sis-induced coagu-
get INR of 2.5. LMWH was con­tin­ued until an INR of more than lopathy. J Thromb Haemost. 2010;8(9):1994-2000.
2 was achieved. She remained on war­fa­rin for 12 months, and 12. Tripodi A, Primignani M, Chantarangkul V, et  al. Thrombin gen­er­a­tion in
patients with cir­rho­sis: the role of plate­lets. Hepatology. 2006;44(2):440-
reimaging con­firmed par­tial recan­a­li­za­tion of the por­tal vein. 445.
Apart from minor trau­matic bruis­ing, there were no sig­nif­i­cant 13. Lisman T, Bongers TN, Adelmeijer J, et al. Elevated lev­els of von Willebrand
bleed­ing com­pli­ca­tions dur­ing treat­ment. fac­tor in cir­rho­sis sup­port plate­let adhe­sion despite reduced func­tional
capac­ity. Hepatology. 2006;44(1):53-61.
14. Reuken PA, Kussmann A, Kiehntopf M, et al. Imbalance of von Willebrand
fac­tor and its cleav­ing pro­te­ase ADAMTS13 dur­ing sys­temic inflam­ma­tion
Conclusion superimposed on advanced cir­rho­sis. Liver Int. 2015;35(1):37-45.
In sum­ mary, despite fre­ quently deranged hemo­ static mark­ 15. Ewe K. Bleeding after liver biopsy does not cor­re­late with indi­ces of
periph­eral coag­u­la­tion. Dig Dis Sci. 1981;26(5):388-393.
ers in patients with CLD, there is a dual pro­pen­sity to bleed­ing 16. Vieira da Rocha EC, D’Amico EA, Caldwell SH, et al. A pro­spec­tive study of
and throm­bo­sis (with a pre­pon­der­ance of por­tal vein throm­bo­ con­ven­tional and expanded coag­u­la­tion indi­ces in predicting ulcer bleed­ing
sis). Variceal bleed­ing sec­ond­ary to por­tal hyper­ten­sion is the after variceal band liga­tion. Clin Gastroenterol Hepatol. 2009;7(9):988-993.

490  |  Hematology 2021  |  ASH Education Program


17. Napolitano G, Iacobellis A, Merla A, et  al. Bleeding after inva­ sive pro­ and bleed­ing risk in patients under­go­ing per­cu­ta­ne­ous image-guided
ce­dures is rare and unpredicted by plate­let counts in cir­rhotic patients inter­ven­tions—part II: rec­om­men­da­tions. J Vasc Interv Radiol. 2019;30
with throm­bo­cy­to­pe­nia. Eur J Intern Med. 2017;38:79-82. ­(8):1168-1184.
18. Desborough MJ, Hockley B, Sekhar M, et al; National Audit Collaborative. 37. Simonetto DA, Singal AK, Garcia-Tsao G, Caldwell SH, Ahn J, Kamath PS.
Patterns of blood com­po­nent use in cir­rho­sis: a nation­wide study. Liver Int. ACG clin­i­cal guide­line: dis­or­ders of the hepatic and mes­en­teric cir­cu­la­tion.
2016;36(4):522-529. Am J Gastroenterol. 2020;115(1):18-40.
19. Fortea JI, Puente Á, Ezcurra I, et al. Management of haemostatic alter­ations 38. Ponziani FR, Zocco MA, Senzolo M, Pompili M, Gasbarrini A, Avolio AW.
and asso­ci­ated dis­or­ders in cir­rho­sis in Spain: a national sur­vey. Dig Liver Portal vein throm­bo­sis and liver trans­plan­ta­tion: impli­ca­tions for waiting
Dis. 2019;51(1):95-103. list period, sur­gi­cal approach, early and late fol­low-up. Transplant Rev
20. Northup PG, Garcia–Pagan JC, Garcia–Tsao G, et  al. Vascular liver dis­or­ (Orlando). 2014;28(2):92-101.
ders, por­tal vein throm­bo­sis, and pro­ce­dural bleed­ing in patients with 39. Ambrosino P, Tarantino L, Di Minno G, et al. The risk of venous throm­bo­em­
liver dis­ease: 2020 Practice Guidance by the Amer­i­can Association for the bo­lism in patients with cir­rho­sis: a sys­tem­atic review and meta-anal­y­sis.
Study of Liver Diseases. Hepatology. 2021;73(1):366-413. Thromb Haemost. 2017;117(1):139-148.
21. Ginés P, Quintero E, Arroyo V, et al. Compensated cir­rho­sis: nat­u­ral his­tory 40. Abdel-Razik A, Mousa N, Elhelaly R, Tawfik A. De-novo por­tal vein throm­
and prog­nos­tic fac­tors. Hepatology. 1987;7(1):122-128. bo­ sis in liver cir­rho­sis: risk fac­tors and cor­ re­la­
tion with the model for
22. Pache I, Bilodeau M. Severe haemorrhage fol­low­ing abdom­i­nal paracen- end-stage liver dis­ ease scor­ ing sys­ tem. Eur J Gastroenterol Hepatol.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/485/1851749/485roberts.pdf by guest on 13 December 2021


tesis for asci­tes in patients with liver dis­ease. Aliment Pharmacol Ther. 2015;27(5):585-592.
2005;21(5):525-529. 41. La Mura V, Tripodi A, Tosetti G, et al. Resistance to thrombomodulin is asso­
23. Hung A, Garcia-Tsao G. Acute kid­ney injury, but not sep­sis, is asso­ci­ated ci­ated with de novo por­tal vein throm­bo­sis and low sur­vival in patients
with higher pro­ce­dure-related bleed­ing in patients with decompensated with cir­rho­sis. Liver Int. 2016;36(9):1322-1330.
cir­rho­sis. Liver Int. 2018;38(8):1437-1441. 42. Senzolo M, Garcia-Tsao G, García-Pagán JC. Current knowl­edge and man­
24. Massicotte L, Denault AY, Beaulieu D, et al. Transfusion rate for 500 con­sec­ age­ment of por­tal vein throm­bo­sis in cir­rho­sis. J Hepatol. 2021;75(2):442-
u­tive liver trans­plan­ta­tions: expe­ri­ence of one liver trans­plan­ta­tion cen­ter. 453.
Transplantation. 2012;93(12):1276-1281. 43. Wang L, Guo X, Xu X, et  al. Anticoagulation favors throm­bus recan­a­li­za­
25. Green L, Bolton-Maggs P, Beattie C, et al. Brit­ish Society of Haematology tion and sur­vival in patients with liver cir­rho­sis and por­tal vein throm­bo­sis:
guide­lines on the spec­trum of fresh fro­zen plasma and cryoprecipitate results of a meta-anal­y­sis. Adv Ther. 2021;38(1):495-520.
prod­ucts: their han­dling and use in var­i­ous patient groups in the absence 44. Giannini EG, Giambruno E, Brunacci M, et al. Low fibrin­o­gen lev­els are asso­
of major bleed­ing. Br J Haematol. 2018;181(1):54-67. ci­ated with bleed­ing after varices liga­tion in throm­bo­cy­to­pe­nic cir­rhotic
26. Zimmon DS, Kessler RE. The por­tal pres­sure-blood vol­ume rela­tion­ship in patients. Ann Hepatol. 2018;17(5):830-835.
cir­rho­sis. Gut. 1974;15(2):99-101. 45. Bianchini M, Cavani G, Bonaccorso A, et al. Low molec­u­lar weight hep­a­rin
27. Rassi AB, d’Amico EA, Tripodi A, et al. Fresh fro­zen plasma trans­fu­sion in does not increase bleed­ing and mor­tal­ity post-endo­scopic variceal band
patients with cir­rho­sis and coagulopathy: effect on con­ven­tional coag­u­la­ liga­tion in cir­rhotic patients. Liver Int. 2018;38(7):1253-1262.
tion tests and thrombomodulin-mod­i­fied throm­bin gen­er­a­tion. J Hepatol. 46. Delgado MG, Seijo S, Yepes I, et al. Efficacy and safety of anticoagulation
2020;72(1):85-94. on patients with cir­rho­sis and por­tal vein throm­bo­sis. Clin Gastroenterol
28. von Meijenfeldt FA, van den Boom BP, Adelmeijer J, Roberts LN, Lisman T, Hepatol. 2012;10(7):776-783.
Bernal W. Prophylactic fresh fro­zen plasma and plate­let trans­fu­sion have 47. Ortel TL, Neumann I, Ageno W, et  al. Amer­i­can Society of Hematology
a prothrombotic effect in patients with liver dis­ease. J Thromb Haemost. 2020 guide­lines for man­age­ment of venous throm­bo­em­bo­lism: treat­
2021;19(3):664-676. ment of deep vein throm­ bo­sis and pul­ mo­ nary embolism. Blood Adv.
29. Tripodi A, Primignani M, Chantarangkul V, et al. Global hemo­sta­sis tests in 2020;4(19):4693-4738.
patients with cir­rho­sis before and after pro­phy­lac­tic plate­let trans­fu­sion. 48. Hoolwerf EW, Kraaijpoel N, Büller HR, van Es N. Direct oral anti­co­ag­u­
Liver Int. 2013;33(3):362-367. lants in patients with liver cir­rho­sis: a sys­tem­atic review. Thromb Res.
30. Baharoglu MI, Cordonnier C, Al-Shahi Salman R, et al; PATCH Investigators. 2018;170:102-108.
Platelet trans­fu­sion ver­sus stan­dard care after acute stroke due to spon­ta­ 49. Hanafy AS, Abd-Elsalam S, Dawoud MM. Randomized con­trolled trial of
ne­ous cere­bral haemorrhage asso­ci­ated with antiplatelet ther­apy (PATCH): rivaroxaban ver­sus war­fa­rin in the man­age­ment of acute non-neo­plas­tic
a randomised, open-label, phase 3 trial. Lancet. 2016;387(10038):2605- por­tal vein throm­bo­sis. Vascul Pharmacol. 2019;113:86-91.
2613. 50. Nagaoki Y, Aikata H, Daijyo K, et  al. Efficacy and safety of edoxaban for
31. Peck-Radosavljevic M, Simon K, Iacobellis A, et  al. Lusutrombopag for treat­ ment of por­ tal vein throm­ bo­sis fol­
low­ing danaparoid sodium in
the treat­ment of throm­bo­cy­to­pe­nia in patients with chronic liver dis­ease patients with liver cir­rho­sis. Hepatol Res. 2018;48(1):51-58.
under­go­ing inva­sive pro­ce­dures (L-PLUS 2). Hepatology. 2019;70(4):1336- 51. Rodríguez-Castro KI, Porte RJ, Nadal E, Germani G, Burra P, Senzolo M.
1348. Management of nonneoplastic por­tal vein throm­bo­sis in the set­ting of liver
32. Terrault N, Chen Y-C, Izumi N, et  al. Avatrombopag before pro­ce­dures trans­plan­ta­tion: a sys­tem­atic review. Transplantation. 2012;94(11):1145-
reduces need for plate­let trans­fu­sion in patients with chronic liver dis­ease 1153.
and throm­bo­cy­to­pe­nia. Gastroenterology. 2018;155(3):705-718. 52. Kearon C, Ageno W, Cannegieter SC, et al; Subcommittees on Control of
33. Lindquist I, Olson SR, Li A, et al. The effi­cacy and safety of thrombopoeitin Anticoagulation, and Predictive and Diagnostic Variables in Thrombotic
recep­tor ago­nists in patients with chronic liver dis­ease under­go­ing elec­ Disease. Categorization of patients as hav­ing pro­voked or unpro­voked
tive pro­ce­dures: a sys­tem­atic review and meta-anal­y­sis [published online venous throm­bo­em­bo­lism: guid­ance from the SSC of ISTH. J Thromb Hae-
18 Jan­u­ary 2021]. Platelets. most. 2016;14(7):1480-1483.
34. Moore KP, Wong F, Gines P, et  al. The man­age­ment of asci­tes in cir­rho­
sis: report on the con­sen­sus con­fer­ence of the International Ascites Club.
Hepatology. 2003;38(1):258-266.
35. O’Leary JG, Greenberg CS, Patton HM, Caldwell SH. AGA clin­i­cal prac­tice
update: coag­u­la­tion in cir­rho­sis. Gastroenterology. 2019;157(1):34-43.
36. Patel IJ, Rahim S, Davidson JC, et al. Society of interventional radi­ol­ogy © 2021 by The Amer­i­can Society of Hematology
con­sen­sus guide­lines for the periprocedural man­age­ment of throm­botic DOI 10.1182/hema­tol­ogy.2021000283

Prakash Singh Shekhawat


Rebalanced hemo­sta­sis in liver dis­ease  |  491
NEUTROPENIA: DOING MORE WITH LESS

Diagnosis and therapeutic decision-making


for the neutropenic patient

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/492/1851746/492connelly.pdf by guest on 13 December 2021


James A. Connelly1 and Kelly Walkovich2
Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN; and 2Department of Pediatrics, University of Michigan, Ann Arbor, MI
1

Determining the cause of a low neutrophil count in a pediatric or adult patient is essential for the hematologist’s clini-
cal decision-making. Fundamental to this diagnostic process is establishing the presence or lack of a mature neutrophil
storage pool, as absence places the patient at higher risk for infection and the need for supportive care measures. Many
diagnostic tests, eg, a peripheral blood smear and bone marrow biopsy, remain important tools, but greater understand-
ing of the diversity of neutropenic disorders has added new emphasis on evaluating for immune disorders and genetic
testing. In this article, a structure is provided to assess patients based on the mechanism of neutropenia and to prioritize
testing based on patient age and hypothesized pathophysiology. Common medical quandaries including fever manage-
ment, need for growth factor support, risk of malignant transformation, and curative options in congenital neutropenia
are reviewed to guide medical decision-making in neutropenic patients.

LEARNING OBJECTIVES
• Recognize the different pathophysiologic mechanisms of neutropenia
• Evaluate the cause of neutropenia
• Assess the infectious and malignancy risk in neutropenia patients
• Provide directed therapy and supportive care to neutropenia patients

Introduction and no complaints concerning for an underlying rheuma-


Neutropenia, defined as an absolute neutrophil count (ANC) tologic condition or malignancy. He takes no medications
<1500/µL after the first year of life, is a common abnormal- and denies recreational drug use. His mother denies any
ity confronting the hematologist. Establishing the correct family history of malignancy, bone marrow failure (BMF)
diagnosis and differentiating benign vs severe pathologic syndromes, or immunodeficiency from the maternal side
mechanisms of neutropenia are critical to inform medical of the family. His height is in the 10th percentile; weight is
decision-making regarding infectious risk, the utility of sup- in the 10th percentile. The physical exam is unremarkable,
portive care measures, the possibility of malignant trans- including a detailed oral, skeletal, and skin assessment.
formation, and curative approaches. This article provides A review of his peripheral smear is consistent with mild neu-
a diagnostic algorithm for evaluation of the pediatric and tropenia; neutrophils are normal in morphologic appear-
adult neutropenic patient followed by evidence-based ance. Hemoglobin level, platelet counts, mean corpuscular
approaches, where available, or expert consensus recom- volume, and the remainder of differential are unremarkable.
mendations on the management of medical complications. A repeat complete blood count (CBC) obtained within
Additionally, the complexity inherent to diagnosing and 1 month of the referral redemonstrates an ANC of 1100/µL.
managing neutropenia is illustrated via a patient case. The patient remains in excellent health and as such is coun-
seled that his ANC result is most likely secondary to the
absence of the Duffy antigen, a phenotype most often seen
in individuals of African, Caribbean, Middle Eastern, and/or
CLINICAL CASE West Indian descent.1 Return instructions are provided.
A 16-year-old generally healthy African American adoles- Author commentary: This adolescent is healthy, with an
cent boy is referred for an ANC of 1200/µL incidentally iden- incidental finding of neutropenia during a well-child exami-
tified on labs obtained as part of a sports-related physical. nation. There are no other CBC abnormalities, and the ANC
He has no history of recent, recurrent, or unusual infections is stable on repeat examination. There are no concerning

492 | Hematology 2021 | ASH Education Program


find­ings on his­tory or phys­i­cal exam to sus­pect a con­gen­i­tal fi­ciently, as seen in inap­pro­pri­ate seques­tra­tion of neu­tro­phils
disorder or under­ly­ing malig­nancy that would impair ­neu­tro­phil within the mar­gin­ated splenic pool in hypersplenism.3 Immune-
pro­duc­tion dur­ ing this ini­
tial visit. Individuals with Duffy-null medi­ated destruc­tion is com­mon after infec­tions or drug insults,
(Fy[a−b−]) sta­tus have a lower neu­tro­phil count but are at no can be driven by T- and B-cell auto­im­mu­nity (eg, pri­mary auto­
increased risk of infec­tions and should not be labeled as neu- im­mune neutropenia) as well as innate immune cell i­nflam­ma­tory
tropenic. Providing edu­ca­tion for concerning signs and symp­ cyto­ kine pro­duc­tion (eg, chronic idi­ o­
pathic neutropenia), or
toms of an evolv­ing mar­row pro­cess with return instruc­tions is may man­ i­
fest sec­ond­
ary to broader immune syn­ dromes (eg,
an appro­pri­ate action. sec­ond­ary auto­im­mune neutropenia).4 Increased con­sump­tion is
typ­i­cally driven by height­ened demand dur­ing infec­tions.

Neutropenia clas­si­fi­ca­tion by path­o­phys­i­­ol­ogy Differential diag­no­sis and eval­u­a­tion of neutropenia


Classically, the degree of neutropenia is subdivided into mild Most patients presenting for eval­u­a­tion of neutropenia will have
(ANC of 1000-1499/µL), mod­ er­
ate (ANC of 500-999/µL), or benign causes. The most likely eti­­ol­o­gies are pri­mar­ily driven by

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/492/1851746/492connelly.pdf by guest on 13 December 2021


severe (ANC <500/µL), with the term “agran­u­lo­cy­to­sis” reserved age (Table 1), high­light­ing the impor­tance of an age-focused his­
for patients with an ANC <200/µL and an absence of neu­tro­phil tory. For neo­na­tes, it is impor­tant to assess the mater­nal health
pre­cur­sors on bone mar­row exam­i­na­tion. This clas­si­fi­ca­tion sys­ with regard to any mater­nal hyper­ten­sion/pre­eclamp­sia asso­
tem is applied to noninfants only because neo­na­tes have an ele­ ci­
ated with restricted pla­ cen­ tal blood flow and sub­ se­quent
vated ANC dur­ing the first 2 weeks of life followed by a lower reduced neo­na­tal bone mar­row pro­duc­tion as well as known
ANC limit of 1000/µL until 1 year of age. Although this orga­ni­za­ auto­im­mu­nity,5 eg, sys­temic lupus erythematosus or prior preg­
tion is instruc­tive for indi­vid­u­als with a decreased capac­ity for nan­cies com­pli­cated by known antineutrophil immu­no­glob­u­lin
neu­tro­phil pro­duc­tion, in which the risk for infec­tion increases G (IgG) antibodies that may cross the pla­centa and result in allo-
as the ANC falls below 1000/µL and the dura­tion of neutropenia immune or isoimmune neutropenia. Significant com­pli­ca­tions at
length­ens, it is not as valu­able for predicting infec­tious risk for deliv­ery, eg, the prolonged rup­ture of mem­branes with chorio-
other mech­a­nisms of neutropenia. It also fails to rec­og­nize the amnionitis or neo­na­tal sep­sis, can exhaust neu­tro­phil supplies,
eth­nic dif­fer­ences in the ANC sec­ond­ary to Duffy-null sta­tus that resulting in prolonged neutropenia. Additionally, pre­ ma­ture
fre­quently result in an ANC <1500/µL.1 infants are more likely to have neutropenia as the pro­duc­tion of
An alter­ na­ tive approach is to clas­ sify neutropenia by the neu­tro­phils occurs pri­mar­ily dur­ing the third tri­mes­ter.6
path­o­phys­i­­ol­ogy driv­ing the decreased periph­eral blood ANC.2 For infants and chil­dren with benign eti­­ol­o­gies, includ­ing pri­
The advan­tage of this clas­si­fi­ca­tion sys­tem is that it pro­vi­des a mary auto­im­mune neutropenia due to the devel­op­ment of anti-
frame­work to inter­pret the periph­eral blood ANC in the con­text neutrophil autoantibodies, symp­toms are usu­ally lacking, and
of diag­nos­tic inves­ti­ga­tions such as a bone mar­row ­exam­i­na­tion the dis­cov­ery of neutropenia is inci­den­tal. Additional ques­tions
and pro­vi­des insights into man­age­ment. Central to this approach to screen for more concerning or revers­ible eti­­ol­o­gies of neu-
is appre­ci­at­ing whether the neutropenic patient has a decreased tropenia include detailing the pres­ence or absence of recur­rent
or nor­mal bone mar­row reserve because cir­cu­lat­ing neu­tro­phils fevers, oral ulcers, gin­gi­vi­tis, den­tal infec­tions, recur­rent or deep-
account for only 3% to 5% of the total body neu­tro­phil count and seated pyo­genic infec­tions, coli­tis, and nutri­tional deficiencies
there­fore may not reflect the abil­ity to mobi­lize mye­loid cells in sec­ond­ary to restric­tive diets (eg, veg­an­ism or extended total
response to exter­nal threats. par­en­teral nutri­tion use) or mal­ab­sorp­tion. Care should be taken
Patients may have hin­dered pro­lif­er­a­tion and/or ­mat­u­ra­tion of to assess for recent infec­tion as tran­sient postinfectious neutro-
their neu­tro­phils from intrin­sic defects or extrin­sic fac­tors result- penia is com­mon. The med­i­ca­tion his­tory should be reviewed for
ing in decreased mar­row reserves. Intrinsic defects in myelopoi- com­monly offending agents used in chil­dren, eg, anti­ep­i­lep­tics
esis stem from germline path­o­genic var­i­ants, eg, ELANE-related and anti­bi­ot­ics, as with­drawal, close mon­i­tor­ing, or tran­si­tion to
neutropenia, or somatic genetic alter­ations, eg, acquired myel- an alter­nate agent may be indi­cated.
odysplastic syn­drome (MDS), that have a neg­a­tive impact on the Further fea­tures ele­vat­ing con­cern for an under­ly­ing inborn
effi­ciency or devel­op­ment of mye­loid pro­gen­i­tors into func­tional error of immu­nity,7 such as a per­sonal or fam­ily his­tory of pan­cre­
poly­mor­pho­nu­clear neu­tro­phils. These patients are at high risk for atic insuf­fi­ciency, malig­nancy (espe­cially mye­loid and lym­phoid),
infec­tion with­out sup­port­ive and/or defin­i­tive care and may have early edentulism or abnor­mal teeth, early graying, liver or lung
a risk of pro­gres­sion to MDS and acute mye­loid leu­ke­mia (AML). fibro­sis, myco­bac­te­rial infec­tions, warts, lymphedema, non­ma­
In con­trast, patients with extrin­sic lim­i­ta­tions on myelopoi- lig­nant lymphoproliferation, vas­cu­li­tis/stroke, auto­im­mu­nity, or
esis have exter­nal pres­sures that inhibit an oth­er­wise nor­mal child­ hood death from infec­ tion should be solicited because
bone mar­row from ade­quately pro­duc­ing neu­tro­phils either many con­gen­i­tal neutropenias or syn­dromes inclu­sive of neutro-
through injury to or phys­i­cal obscu­ra­tion of the mar­row niche, penia pres­ent in the first years of life but may have an incom­plete
eg, ­neu­ro­blas­toma or fibro­sis or nutri­tional deficiencies. These spec­trum of symp­toms.
patients are also at increased risk of infec­tion; how­ever, many For ado­les­cents and young adults, a his­tory sim­i­lar to infants
of the exter­nal pres­sures are treat­able with prompt rec­og­ni­tion. and chil­dren should be obtained with addi­tional screen­ing ques­
Patients with a nor­mal mar­row reserve can have neutropenia tions related to rheumatologic dis­or­ders since the inci­dence of
through 3 dif­fer­ent mech­a­nisms: inef­fec­tive traf­fick­ing, immune- sec­ond­ary auto­im­mune neutropenia increases with age. Addi-
medi­ated destruc­tion, or increased con­sump­tion. For patients tional his­tory ques­tions aimed at assessing risk for eat­ing dis­
with inef­fec­tive traf­fick­ing, neu­tro­phils mature appro­pri­ately but or­ders, eg, anorexia nervosa, or rec­re­a­tional drug use should
are either unable to egress from the mar­row, eg, due to defects be com­pleted. Neutropenia sec­ond­ary to the use of cocaine
in the CXCL12/CXCR4 axis, as in warts, hypogammaglobulinemia, adul­ ter­
ated with levamisole is documented,8 as are cases of
infec­tions, myelokathexis (WHIM) syn­ Prakash
drome, or Singh Shekhawat
are stored inef­ aplastic ane­mia sec­ond­ary to ecstasy (3,4-methylenedioxymeth-

Diagnosis and deci­sion-mak­ing for neutropenia  |  493


Table 1. Etiologies of neutropenia by mech­a­nism and age

Neonates Infants/chil­dren Adolescents and young adults Adults


Decreased bone mar­row reserve
Intrinsic defects in - Inborn errors of - Inborn errors of - Inborn errors of myelopoiesis - Acquired MDS
myelopoiesis myelopoiesis (eg, ELANE- myelopoiesis (eg, SDS, (eg, GATA2 haploinsufficiency, - Inborn errors of
related neutropenia) ELANE-related neutropenia, BMF syn­dromes) myelopoiesis (eg, GATA2
GATA2 haploinsufficiency) haploinsufficiency)
Extrinsic lim­i­ta­tions on - Maternal hyper­ten­sion - Viral infec­tion - Viral infec­tion - Medications
myelopoiesis - Prematurity - Nutritional deficiencies - Medications - Viral infec­tion
- Viral infec­tion - Medications - Idiopathic aplastic ane­mia - Nutritional deficiencies (eg,
- Infiltration of the mar­row - Infiltration of the mar­row - Nutritional deficiencies (eg, vita­min B12 defi­ciency, cop­
space (eg, neu­ro­blas­toma, space (eg, acute leu­ke­mia) anorexia nervosa) per defi­ciency)
osteopetrosis) - Drug abuse - Idiopathic aplastic ane­mia

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/492/1851746/492connelly.pdf by guest on 13 December 2021


- Alcoholism - Infiltration of the mar­row
space (eg, T-LGL, fibro­sis,
mye­loma)
- Thymoma with pure WBC
aplasia
- Drug abuse
- Alcoholism
Normal bone mar­row reserve
Ineffective traf­fick­ing - Inborn errors of immu­nity - Inborn errors of immu­nity - Inborn errors of immu­nity (eg, - Hypersplenism
(eg, WHIM syn­drome) (eg, WHIM syn­drome) WHIM syn­drome)
- Hypersplenism
Immune-medi­ated - Postinfectious anti­body- - Postinfectious anti­body- - Postinfectious anti­body- - Drug-related anti­body-
destruc­tion medi­ated neutropenia medi­ated neutropenia medi­ated neutropenia medi­ated neutropenia
- Isoimmune neutropenia - Autoimmune neutropenia - Drug-related anti­body- - Postinfectious anti­body-
- Alloimmune neutropenia of infancy medi­ated neutropenia medi­ated neutropenia
- Drug-related anti­body- - Primary auto­im­mune - Autoimmune neutropenia
medi­ated neutropenia neutropenia asso­ci­ated with auto­im­mune
- Autoimmune neutropenia dis­or­ders
asso­ci­ated with inborn errors - Primary auto­im­mune
of immu­nity neutropenia
- Cytokine-induced apo­pto­
sis (eg, chronic idi­o­pathic
neutropenia)
Increased con­sump­tion - Neonatal sep­sis - Bacterial infec­tion - Bacterial infec­tion - Bacterial infec­tion
WBC, white blood cell.

amphetamine) and inhal­ing glue.9,10 Alcoholism can also result Diagnostically, all­patients ben­efi ­ t from a CBC and dif­fer­en­tial,
in neutropenia from impaired granulopoiesis.11 Many of the BMF a mea­sure­ment of liver and renal func­tion, and a direct review
syn­dromes pres­ent with clin­i­cally sig­nif­i­cant cytopenias in the of the periph­eral blood smear to ensure a lack of pseudoneutro-
sec­ond and third decades of life, so like youn­ger chil­dren, a full penia due to in vitro leu­ko­cyte aggre­ga­tion (due to ethylenedi-
his­tory should be obtained to screen for under­ly­ing eti­­ol­o­gies aminetetraacetic acid or cold agglu­ti­nins). The remain­der of the
that may have been pre­vi­ously qui­es­cent. diag­nos­tic eval­u­a­tion should be tai­lored to both the age and pre-
For adults, acquired eti­­ol­o­gies, par­tic­u­larly drug expo­sures, senting clin­ic
­ al fea­tures of the patient (Table 2). An impor­tant fac­
remain the most likely eti­­ol­ogy, and thus a care­ful review of tor in deter­min­ing risk of infec­tion in patients with a low periph­eral
the patient’s med­i­ca­tion expo­sures is crit­i­cal. Nutritional defi- ANC is the pres­ence or absence of a bone mar­row reserve pool of
ciencies are also com­ mon, eg, cop­ per defi­ciency sec­ond­
ary mature mye­loid cells. Although a bone mar­row biopsy can pro­vide
to gas­tric bypass.12 Relative to pedi­at­ric patients, both auto­ this infor­ma­tion, it usu­ally is not indi­cated in the ini­tial eval­u­a­tion if
im­mu­nity and malig­nancy are more likely in adults, so focused the his­tory and phys­i­cal are nonconcerning for bone mar­row dys­
ques­tions regard­ing arthral­gias, arthri­tis, rashes, fatigue, unex­ func­tion. Alternative meth­ods to assess for a bone mar­row reserve
plained fevers, bone pain, weight loss, and night sweats should include documenting a rise in the ANC either dur­ing an infec­tious
be included in the his­tory. epi­sode, although fail­ure to increase the ANC may be a false-neg­
The phys­i­cal exam eval­ua ­ ­tion for all­patients should focus a­tive in the set­ting of a viral infec­tion, or through a granulocyte
on the oral cav­ity to assess for aphthous ulcers, gin­gi­vi­tis, and col­ony-stim­u­lat­ing fac­tor (G-CSF) stim­u­la­tion test in which an ANC
den­tal abnor­mal­i­ties sig­nal­ing clin­i­cally sig­nif­i­cant neutropenia is mea­sured before and 4 to 6 hours after a sin­gle dose.
as well as on the assess­ment of height, skel­e­tal abnor­mal­i­ties, For neo­na­tes, screen­ing for mater­nal IgG antineutrophil anti-
skin changes (eg, albi­nism, malar rash, vit­il­igo, cuta­ne­ous vas­ bodies may assist in secur­ing a diag­no­sis of alloimmune or
cu­li­tis, fol­lic­u­li­tis, warts, eczema), nail anom­a­lies, lymph­ade­nop­ isoimmune neutropenia. For infants and chil­dren, antineutro-
a­thy, and hepatosplenomegaly, which are poten­tial clues to an phil antibodies can be assessed to lend sup­port to a suspected
under­ly­ing eti­­ol­ogy. diag­no­sis of auto­im­mune neutropenia of infancy; how­ever, the

494  |  Hematology 2021  |  ASH Education Program


Table 2. Laboratory eval­u­a­tion of neutropenia by age

Neonates Infants/chil­dren Adolescents and young adults Adults


Recommended for all­patients
CBC with dif­fer­en­tial CBC with dif­fer­en­tial CBC with dif­fer­en­tial CBC with dif­fer­en­tial
Liver and renal func­tion Liver and renal func­tion Liver and renal func­tion Liver and renal func­tion
Peripheral blood smear Peripheral blood smear Peripheral blood smear Peripheral blood smear
HIV anti­body screen­ing
T-LGL flow cytom­e­try
To be con­sid­ered based on patient-spe­cific his­tory
Maternal antineutrophil antibodies Antineutrophil antibodies Vitamin B12, folate, cop­per Vitamin B12, folate, cop­per
Vitamin B12, folate, HIV Urine drug screen
cop­per Urine drug screen ANA, rheu­ma­toid fac­tor
Amylase iso­en­zymes Amylase iso­en­zymes ESR, CRP

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/492/1851746/492connelly.pdf by guest on 13 December 2021


Fecal elas­tase Fecal elas­tase Amylase iso­en­zymes
Telomere length Telomere length Fecal elas­tase
Chromosomal break­age Chromosomal break­age Telomere length
ADA2 enzyme activity ADA2 enzyme activity Chromosomal break­age
IgG/A/M/E ANA, ESR, CRP ADA2 enzyme activity
T/B/NK sub­sets IgG/A/M/E IgG/A/M/E
T/B/NK sub­sets T/B/NK sub­sets
T/B/NK, T cell/B cell/nat­u­ral killer cell.

absence of such antibodies does not pre­clude the d ­ iag­no­sis. rec­og­ni­tion of neutropenia with inborn errors of hema­to­poi­e­sis
If an anti­body is not detected, though, the neutropenia is and immu­nity and lim­i­ta­tions in the clin­i­cal clas­si­fi­ca­tion of dis­
often referred to as chronic idi­o­pathic neutropenia of child­ ease,18 genetic sequenc­ing is increas­ingly being uti­lized to secure
hood.13 Additionally, pos­i­tive antineutrophil antibodies do not a molec­u­lar diag­no­sis.19 Confirmation of a genetic diag­no­sis is
guar­an­tee the absence of other eti­­ol­o­gies, includ­ing severe con­ valu­able because it facil­i­tates bio­log­i­cally ratio­nal and per­son­
gen­i­tal neutropenia (SCN).14 Secondary to con­cerns regard­ing the ally tai­lored prog­nos­ti­ca­tion, sup­port­ive care, and ther­apy. For
poor sen­si­tiv­ity and spec­i­fic­ity of avail­­able assays, the mea­sure­ patients with neutropenia, it can offer spe­cific insights into the
ment of antineutrophil antibodies is not uni­formly prac­ticed by util­ity of G-CSF and the risk of trans­for­ma­tion to malig­nan­cies
hema­tol­o­gists. Test per­for­mance can be improved by includ­ing and inform deci­sions related to donor selec­tion and con­di­tion­
both the granulocyte immu­no­flu­o­res­cence test and the granulo- ing reg­i­mens for hema­to­poi­etic stem cell trans­plant (HSCT). In
cyte agglu­ti­na­tion test and by repeat­ing the assay.15 some cases, par­tic­u­larly in patients with neutropenia as part of
Depending on the solicited his­ tory, screen­ ing for pan­cre­ an inborn error of immu­nity, the genetic diag­no­sis may iden­tify
atic insuf­fi­ciency with isoamylase, fecal elas­tase, and a pan­cre­ an exploit­able molec­u­lar path­way that is amenable to targeted
atic ultra­sound and/or skel­e­tal imag­ing may be use­ful because ther­apy. For fur­ther infor­ma­tion on the inter­sec­tion between neu-
many patients with Shwachman-Diamond syn­drome (SDS) have tropenia and inborn errors of immu­nity, the read­ers are directed
clin­i­cally sub­tle pan­cre­atic or skel­e­tal defects.16 Other screen­ing to the accom­pa­ny­ing arti­cle “Understanding Neutropenia Sec-
tests for BMF, eg, telo­mere length (short telo­mere syn­dromes), ondary to Intrinsic or Iatrogenic Immune Dysregulation” by Dr.
chro­ mo­somal break­ age (Fanconi ane­ mia), or ADA2 enzyme Walkovich. Patients with suspected benign causes of neutrope-
activ­ity (a defi­ciency of ADA2), may be appro­pri­ate. Similarly, nia should have repeat CBCs every few months to mon­i­tor for
screen­ing for under­ly­ing immune defi­ciency with quan­ti­ta­tive res­o­lu­tion of neutropenia. If the neutropenia does not resolve
Igs and enu­mer­a­tion of lym­pho­cyte sub­sets and func­tion or for in an expected time frame based on the suspected mech­a­nism,
auto­im­mune dis­ease with an anti­nu­clear anti­body (ANA) may be reassessment is warranted. This may include repeat genetic test­
of value. Evaluation with folate, vita­min B12, and cop­per is rec- ing if the ini­tial test­ing was conducted more than 2 years ear­lier
ommended in all­older adult patients with suspected nutri­tional to account for newly dis­cov­ered inborn errors of hema­to­poi­e­sis
deficiencies. and immu­nity.
For adults, ANA, rheu­ma­toid fac­tor, eryth­ro­cyte sed­i­men­ta­
tion rate (ESR), and C-reac­tive pro­tein (CRP) tests are valu­able to
screen for auto­im­mune dis­ease. Screening for HIV and for T-cell
large gran­u­lar lym­pho­cyte leu­ke­mia (T-LGL) is recommended CLINICAL CASE (Con­tin­ued)
even in patients with­out lym­pho­cy­to­sis or a his­tory of auto­im­ Four years later at age 20, the patient returns prior to a planned
mune dis­ease.17 Depending on the his­tory, addi­tional tests such sur­gi­cal pro­ce­dure to remove his wis­dom teeth and is con-
as a serum pro­tein elec­tro­pho­re­sis or imag­ing may be warranted. cerned his neutropenia may place him at risk of infec­tion. He is
Regardless of age, if a sat­is­fac­tory eti­­ol­ogy of the neutropenia clin­i­cally doing well, but a CBC is nota­ble for an ANC of 1000/µL
is not deter­mined or other key phys­i­cal anom­a­lies or his­tory ele­ and a plate­let count of 90 000/µL. Given the new throm­bo­cy­
ments prompt con­cern for an under­ly­ing dis­or­der, then a bone to­pe­nia, a bone mar­row biopsy is performed that dem­on­strates
mar­row aspi­rate and biopsy to assess myelopoiesis and evi­dence hypocellularity (50%) and mild mye­loid and mega­kar­yo­cyte dys­
of clonal evo­lu­tion is warranted. Additionally, given the increased pla­sia. Karyotyping reveals a small del(20q) clone but no abnor­
Prakash Singh Shekhawat
Diagnosis and deci­sion-mak­ing for neutropenia  |  495
mal blast pop­u­la­tion by flow cytom­e­try. The com­pre­hen­sive lead to harm­ful con­se­quences, includ­ing aller­gic reac­tions, an
mar­row report men­tions the pos­si­bil­ity of SDS, but there is no increase in bac­te­rial resis­tance to anti­bi­ot­ics, hos­pi­tal-acquired
his­tory of diar­rhea, and an inherited neutropenia next-gen­er­a­ infec­tions, large costs to the health care sys­tem, and a decreased
tion sequenc­ing (NGS) panel is neg­at­ive for path­o­genic var­i­ qual­ity of life for patients with chronic neutropenia. This care­
ants. Given the lack of mor­pho­log­ic ­ al evi­dence of MDS or AML, ful bal­ance must be weighed for each neutropenic patient who
it is recommended that the patient be followed closely with pres­ents with fever, and the approach will dif­fer if the patient
monthly CBCs and a repeat bone mar­row in 3 months. is a known neutropenic patient or pres­ents with newly iden­ti­
Author com­men­tary: Unlike his first visit, there are sev­ fied neutropenia in the con­text of fever. Additional fac­tors such
eral fea­tures dur­ing this eval­u­a­tion concerning for a con­gen­i­ as the pos­si­bil­ity of neu­tro­phil dys­func­tion, the dura­tion of the
tal neutropenia dis­or­der. His mar­row aspi­ra­tion dem­on­strated neutropenia, the sta­tus of vac­ci­na­tions, the pres­ence of a cen­
hypocellularity and cel­lu­lar dys­cra­sias, and his cyto­ge­net­ics tral venous cath­et­ er, and any con­cur­rent immune defects related
iden­ ti­
fied a com­ mon and likely benign clonal hema­ to­
poi­
e­ to the under­ly­ing dis­or­der or immu­no­sup­pres­sive med­i­ca­tions
sis seen in SDS. The phy­si­cian rec­og­nized the asso­ci­at­ion of must also be con­sid­ered in deter­min­ing the risk of bac­ter­emia in

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/492/1851746/492connelly.pdf by guest on 13 December 2021


del(20q) with SDS but assumed that a lack of ste­at­or­rhea and a the neutropenic patient.
neg­a­tive NGS panel was suf­fic ­ ient to rule out SDS. However, the
clas­sic pre­sen­ta­tion of diar­rhea and neutropenia is only seen Fever man­
age­
ment in the newly diag­
nosed neutropenic
in approx­i­ma­tely 50% of SDS patients.15 It is also impor­tant to ­patient
review the genes pres­ent in NGS pan­els to ascer­tain whether The iden­ti­fi­ca­tion of iso­lated neutropenia in a pre­vi­ously healthy
they incor­po­rate suspected genes of inter­est. SBDS may not patient presenting with fever is chal­leng­ing, as the abil­ity to
be included on some NGS pan­els sec­ond­ary to the pres­ence mount neu­tro­phil immu­nity to lower the risk of bac­ter­emia is
of a pseudogene (SBDSP1), requir­ing high-qual­ity map­ping and likely unknown. Several stud­ies have exam­ined the risk of severe
infor­mat­ics or other approaches, such as Sanger sequenc­ing, bac­te­rial infec­tion (SBI) in pre­vi­ously healthy chil­dren who pres­
for dif­fer­en­ti­a­tion. Additional genes (EFL1, DNAJC21, SRP54) ent with fever and neutropenia, and the major­ity have dem­on­
have recently been dis­cov­ered to be asso­ci­ated with SDS and strated these patients to be at low risk, with a sim­i­lar prev­a­lence
may not yet have been added to some pan­els. A high sus­pi­cion of bac­te­rial blood and uri­nary tract infec­tions (UTIs) as nonneu-
of SDS should prompt other screens to detect pan­cre­atic exo­ tropenic patients presenting with fever.20 The larg­ est study,
crine insuf­fi­ciency, includ­ing serum pan­cre­atic isoamylase level conducted at the Bos­ ton Children’s Emergency Department,
and fecal elas­tase tests, and an abdom­in ­ al ultra­sound to eval­ US, eval­ua ­ ted the out­comes of 1888 pedi­at­ric patients who pre-
u­ate for pan­cre­atic atro­phy/fatty replace­ment. The phy­si­cian sented for eval­u­a­tion of unsus­pected and iso­lated neutropenia.21
should have ordered these tests to help deter­mine a clin­i­cal For febrile infants <3 months of age, the SBI rate was 6.2%, which
diag­no­sis of SDS. The patient’s mar­row find­ings are com­mon was sim­i­lar to the SBI rate of 7% reported in a prior study of non-
for SDS, but as the diag­no­sis is still unknown in this patient, neutropenic infants.22 SBI was very rare in chil­dren over 3 months
close fol­low-up as pre­scribed by the phy­si­cian is appro­pri­ate. of age (2.0%; 1 bac­ter­emia, 15 UTIs, 0 men­in­gi­tis), with the sin­
gle case of bac­ter­emia presenting with obvi­ous clin­i­cal signs of
sep­tic shock in a 9-month-old with Streptococcus pneumoniae.
Therapeutic deci­sion-mak­ing for the neutropenic patient A study in Barcelona, Spain, eval­u­ated 190 well-appearing febrile
Following iden­ti­fi­ca­tion of the neutropenic patient and eval­ua ­­ and unsus­ pected neutropenia patients and dem­ on­
strated an
tion for an under­ly­ing cause, chal­lenges arise regard­ing treat­ SBI in 4 patients (2.1%; 2 UTIs, 2 pneu­mo­nias), but 101 patients
ment deci­sions, mon­i­tor­ing for infec­tion and/or MDS and AML, (53.2%) were admit­ted, and 48 (47.5%) received broad-spec­trum
and, for some dis­or­ders, con­sid­er­ation of cura­tive options, eg, anti­bi­ot­ics.23 A case con­trol study com­pared pedi­at­ric patients
HSCT. Unfortunately, for many neutropenic dis­or­ders and com­ >3 months of age who presented with fever and unsus­pected
pli­ca­tions the evi­dence to sup­port med­i­cal deci­sion-mak­ing is neutropenia with an age-matched con­trol group with­out neutro-
lacking or reli­ant on expe­ri­ence gained in onco­logic neutrope- penia. SBIs were iden­ti­fied at a higher rate in the con­trol group
nic patients. The devel­op­ment of neutropenia reg­is­tries such as (19 vs 6 patients), which may be explained by the asso­ci­a­tion of
the Severe Chronic Neutropenia International Registry (SCNIR) viral infec­tions with fever and neutropenia (Assaf Harofeh Medi-
and the Shwachman-Diamond Syndrome Registry has pro­vided cal Center, Israel).24 Two addi­tional stud­ies dem­on­strated sim­i­lar
impactful data to close this infor­ma­tion gap, but much guid­ance low SBI rates in only 1 of 52 (1.9%) patients aged 1.5 to 36 months
on clin­i­cal deci­sion-mak­ing still depends on expert and con­sen­ (Children’s Hospital of Wisconsin, US) and in 0 of 51 well-appear-
sus opin­ion. Knowing this lim­it­a­tion in the field, the fol­low­ing ing chil­dren but in 5 of 17 (30%) who appeared ill (Dana-Dwek
dis­cus­sion on treating the neutropenic patient is based on evi­ Children Hospital, Israel).25,26 Higher rates of bac­te­rial infec­tions
dence where avail­­able but also observes stan­dard treat­ment have been reported in stud­ies focused on hos­pi­tal­ized neutro-
prac­ tices in which expert con­ sen­
sus has been achieved and penic patients (12.7%-14.9%, with 1 study dem­on­strat­ing 0%),27
areas of ongo­ing man­age­ment debate among hema­tol­o­gists. which may have resulted from the admis­sion of sicker patients
and the infec­tious envi­ron­ment of the pop­u­la­tion, as bru­cel­lo­
Management of fever sis and rick­ett­sia, bac­te­rial infec­tions known to be asso­ci­ated
Historically, fevers have been treated aggres­sively, even in well- with neutropenia, were reported eti­­ol­o­gies in 3 out of 4 of these
looking patients, over con­cerns that signs of inflam­ma­tion may ­inpa­tient-only stud­ies.28-31
be dimin­ished with­out infil­trat­ing neu­tro­phils and that the risk of The con­clu­sion from these stud­ies is that the risk of bac­te­rial
not treating an occult bac­ter­emia will lead to poor out­comes. infec­tion in the out­pa­tient set­ting for the well-appearing pedi­
However, fre­quent hos­pi­tal admis­sions and anti­bi­otic use may at­ric patient with iso­lated and unsus­pected neutropenia is low

496  |  Hematology 2021  |  ASH Education Program


and sim­i­lar to patients with nonneutropenic fever. Screening for emer­gent eval­u­a­tion for any fever is nec­es­sary sec­ond­ary to the
bac­te­rial infec­tion, includ­ing a blood cul­ture, a uri­nal­y­sis with risk of bac­te­rial sep­sis. However, patients who have con­sis­tent
urine cul­ture, and, for symp­tom­atic patients, a chest x-ray, is ANCs above 1000/µL are at low risk of infec­tion,13 and man­age­
suggested, but aggres­sive man­age­ment includ­ing admis­sion ment of fever may be based on the clin­i­cal appear­ance of the
and empiric anti­bi­ot­ics may not be nec­es­sary for all­patients. patient, with unwell patients eval­u­ated in the emer­gency room
Patients who are unwell, have addi­tional cytopenias that raise and well patients eval­u­ated in out­pa­tient clin­ics.
con­cern for malig­nancy or BMF, have other con­sti­tu­tional fea­
tures, have a his­tory of infec­tions, or have a fam­ily his­tory of Indications for G-CSF
hema­ to­logic malig­ nan­cies or cytopenias should enact more The use of G-CSF is mainly employed in patients with defec­tive
aggres­ sive mea­ sures includ­ ing admis­
sion for empiric anti­ bi­ myelopoiesis and has proven to be a life­sav­ing inter­ven­tion in
ot­ics that include Pseudomonas spp. and gram-neg­a­tive cov­ patients with SCN. The SCNIR rec­om­mends a starting dose of
er­
age. Similarly, adults who pres­ ent with severe neutropenia 5  µg/kg/d for patients with SCN with esca­la­tion to 10  µg/kg/d
(ANC <500/µL) are more likely to have seri­ous dis­or­ders such as and then an increase by incre­ments of 10  µg/kg/d at 14-day inter­

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/492/1851746/492connelly.pdf by guest on 13 December 2021


under­ly­ing malig­nancy and should be admit­ted for eval­u­a­tion for vals while the ANC remains below 1000/µL.38 The main­te­nance
SBIs.17 Medications asso­ci­ated with the sup­pres­sion of myelopoi- dose of G-CSF for SCN is var­i­able depending on the genetic dis­
esis that are not of vital impor­tance should also be discontinued or­der as well as the spe­cific path­o­genic var­i­ant within the gene,
because drug-induced agran­u­lo­cy­to­sis can lead to fatal­ity with with a median dose of 7.3  µg/kg/d needed to achieve an ANC
con­tin­ued expo­sure.32 between 1000 and 1500/µL.39 Higher ANCs are not nec­es­sary in
most patients, and maintaining the smallest effec­tive dose avoids
Fever man­age­ment in the established neutropenic patient the con­se­quences of higher dos­ing, includ­ing bone pain, spleno­
The approach to the patient with known neutropenia is often less meg­aly, and the­o­ret­i­cal higher selec­tive pres­sure for hema­to­poi­
chal­leng­ing, as the eti­­ol­ogy of the neutropenia and the his­tory etic clones more respon­sive to G-CSF.40 For cyclic neutropenia,
of infec­tious com­pli­ca­tions may guide man­age­ment. Disorders the dose is much lower, typ­i­cally 1 to 3  µg/kg/d given every 1 to
such as pri­mary auto­im­mune neutropenia and chronic idi­o­pathic 3 days, which short­ens the dura­tion of the neutropenic period
neutropenia typ­ i­
cally have ade­ quate bone mar­ row stor­ age and effec­tively elim­i­na­tes sep­sis and death from sep­sis based on
pools of neu­tro­phils to respond to infec­tion and there­fore pres­ out­come data from the SCNIR.13 Granulocyte-mac­ro­phage-CSF
ent a low risk for SBIs. In a study of 43 pedi­at­ric patients with treat­ment is inef­fec­tive in most forms of con­gen­i­tal neutropenia,
pri­mary auto­im­mune or chronic idi­o­pathic neutropenia, there as, unlike G-CSF, it is unable to induce nic­o­tin­amide phosphori-
were 133 emer­gency room encoun­ters for fever with no patients bosyl trans­fer­ase-depen­dent granulopoiesis.41
iden­ti­fied as hav­ing a true bac­te­rial infec­tion (Texas Children’s G-CSF use in acquired causes of neutropenia is typ­i­cally guided
Cancer and Hematology Centers, US).33 In the Ital­ian Neutrope- by the patient his­tory and is likely safe, as no defin­i­tive asso­ci­a­tion
nia Registry, over the course of 9.5 years they iden­ti­fied only 1 with G-CSF use and can­cer devel­op­ment has been established in
blood­stream infec­tion in 38 patients with auto­im­mune neutro- this patient pop­u­la­tion.35 G-CSF treat­ment for pri­mary auto­im­mune
penia and 2 blood­stream infec­tions in 23 idi­o­pathic neutropenia and chronic idi­o­pathic neutropenia should not be deter­mined
patients.34 For the pedi­at­ric patient with auto­im­mune or chronic based on the detec­tion of antineutrophil antibodies but rather is
idi­o­pathic neutropenia, if a rise in the neu­tro­phil count can be recommended for patients with severe neutropenia with recur­
dem­on­strated dur­ing times of infec­tion and there is no his­tory rent infec­tions or sto­ma­ti­tis.42 As mye­loid pro­duc­tion is rel­a­tively
of severe infec­tions, eval­u­a­tion by the child’s pri­mary care phy­si­ pre­served in these con­di­tions, small doses of 0.5 to 3  µg/kg/d
cian should be suf­fi­cient, with emer­gency room vis­its lim­ited to of G-CSF given every 1 to 3 days can be used to increase the
when the patient appears unwell.35 ANC to a tar­get main­te­nance of 1000 to 1500/uL.43 In the French
Adults with pri­mary auto­im­mune and chronic idi­o­pathic neu- Severe Chronic Neutropenia Registry, the use of G-CSF pro­vided
tropenia usu­ally do not develop severe infec­tions, although they a clin­i­cal ben­e­fit in adult patients with pri­mary auto­im­mune or
may suf­fer from recur­rent sto­ma­ti­tis and respi­ra­tory infec­tions.35 chronic idi­o­pathic neutropenia in 23 of 24 and 20 of 26 rou­tinely
This was dem­on­strated in a French Severe Chronic Neutropenia sched­uled and spo­radic G-CSF cohorts, respec­tively.36 Intermit-
Registry report in which adult patients with pri­mary auto­im­mune tent dos­ing of G-CSF may be required in patients who pres­ent
and chronic idi­o­pathic neutropenia had low rates of SBIs at 3.85 with severe infec­tion or in the set­ting of sur­gi­cal pro­ce­dures at
per 100 patient-years.36 Secondary auto­im­mune neutropenia in high risk for infec­tion or poor wound healing.44 G-CSF is almost
both chil­dren and adults is often asso­ci­ated with addi­tional cyto- always given in patients with suspected drug-induced agran­u­
penias, immune defects, or immu­no­sup­pres­sive med­i­ca­tions lo­cy­to­sis, although evi­dence supporting this rec­om­men­da­tion is
and car­ries a higher risk of infec­tion. The Ital­ian Neutropenia lacking,35 and may also be used in patients with only mod­er­ate
Registry reported SBIs in 10 of 26 (40%) patients with sec­ond­ drug-induced sup­pres­sion of myelopoiesis in whom con­tin­u­a­tion
ary auto­im­mune neutropenia, which is sig­nif­i­cantly higher than of the offending agent is nec­es­sary.
in the 11.8% of patients with pri­mary auto­im­mune neutropenia,
over the course of 15 years.37 Extra cau­tion should there­fore be Role of immune sup­pres­sion in auto­im­mune neutropenia
taken in patients with sec­ond­ary auto­im­mune neutropenia who Immune sup­pres­sion to treat pri­mary auto­im­mune and chronic
pres­ent with fever, includ­ing con­sid­er­ation of fun­gal and other idi­o­pathic neutropenia is not recommended for chil­dren due to
atyp­i­cal infec­tions related to addi­tional T-cell immu­no­de­fi­ciency. poor response and low infec­tious risk and has lim­ited suc­cess in
Disorders of impaired myelopoiesis, includ­ing SCN, will lead adults.44 In the French Severe Congenital Neutropenia Registry,
to an impaired abil­ity to mount neu­tro­phil immu­nity in the set­ response to com­monly used immu­no­sup­pres­sant agents (gluco-
ting of infec­tion. For patients with a poor response to G-CSF, an corticosteroids, meth­o­trex­ate, cyclo­spor­ine) was observed in half
Prakash Singh Shekhawat
Diagnosis and deci­sion-mak­ing for neutropenia  |  497
Table 3. Congenital neutropenia dis­or­ders at risk for MDS/AML

Disease, gene, and inher­i­tance Additional infor­ma­tion includ­ing


pat­tern Risk of MDS/AML Common somatic alter­ations nonhematologic fea­tures
ELANE SCN 15%-20%51; increased inci­dence CSF3R, RUNX1, ASXL1, SUZ12, Acquired CSF3R muta­tions
ELANE, AD of MDS/AML in patients on high EP300, mono­somy 7, tri­somy com­mon (approx­i­ma­tely 50%) in
doses of G-CSF with poor 21.75 patients with­out MDS/AML.75
neu­tro­phil response.54
Cyclic neutropenia AML rarely reported.76 CSF3R, mono­somy 7, tri­somy 21 in Acquired CSF3R muta­tions rarely
ELANE, AD the 1 patient with AML.76 iden­ti­fied in patients with­out
MDS/AML.76
HAX1-SCN MDS/AML reported, but inci­ Somatic alter­ations not published. Acquired CSF3R muta­tions
HAX1, AR dence of leu­ke­mia not pre­cisely com­mon (approx­i­ma­tely 45%) in
reported.77 patients with­out MDS/AML.75

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/492/1851746/492connelly.pdf by guest on 13 December 2021


G6PC3 defi­ciency AML rarely reported.78 T(8;21)(q22;q22) iden­ti­fied by kar­ Inflammatory bowel dis­ease,
G6PC3, AR yo­type in one patient.78 con­gen­i­tal heart and uro­gen­i­tal
defects, and endo­crine dis­or­
ders. Acquired CSF3R muta­tions
iden­ti­fied in patients with­out
MDS/AML.75
X-linked neutropenia MDS/AML rarely reported.79 Somatic alter­ations not published. Acquired CSF3R muta­tions
WAS GOF, XL iden­ti­fied in patients with­out
MDS/AML.75
Glycogen stor­age dis­ease 4% with MDS/AML in the SCNIR.80 Monosomy 7. Short telo­meres also Hypoglycemia, hepa­to­meg­aly, and
type 1b reported.81 entero­co­li­tis.
G6PT1, AR
Poikiloderma with neutropenia MDS/AML reported, but inci­ Somatic alter­ations not published. Poikiloderma, nail dys­tro­phy, and
USB1, AR dence of leu­ke­mia not pre­cisely pal­mar/plan­tar hyper­ker­a­to­sis.
reported.82 Myelodysplastic
changes in most patients.
SDS 20%-30%83,84; patients also at risk Biallelic TP53, RUNX1, SETBP1, Pancreatic exo­crine insuf­fi­ciency,
SBDS, AR of BMF.16 BRAF, NRAS, ETNK1, alter­ations skel­e­tal abnor­mal­i­ties. Somatic
in chro­mo­somes 3, 5, 7, and alter­ations not asso­ci­ated with
20.85 MDS/AML include i(7)(q10),
del(20q), and EIF6 muta­tions.
GATA2 haploinsufficiency 80%86; occur­rence typ­ic
­ ally in later Monosomy 7, tri­somy 8, ASXL1, Warts, lymphedema, panniculitis,
GATA2, AD ado­les­cence and early adult­ der(1;7)(q10;p10).87 deaf­ness, infec­tion, and pul­mo­
hood and may be presenting nary alve­o­lar proteinosis.
fea­ture.
SAMD9/SAMD9L GOF dis­or­der Risk of MDS and AML unknown. Monosomy 7, del(7q), RUNX1, SAMD9: adre­nal hypo­pla­sia,
SAMD9 or SAMD9L, AD Patients also at risk of BMF that SETBP1, ETV6.89 growth restric­tion, gen­i­tal
may spon­ta­ne­ously resolve.88 abnor­mal­i­ties, enter­op­a­thy,
infec­tions.
SAMD9L: neu­ro­logic symp­toms,
infec­tions.
AD, auto­so­mal dom­in
­ ant; AR, auto­so­mal reces­sive, GOF, gain of func­tion; XL, X-linked.

of the adult patients with pri­mary auto­im­mune or chronic idi­o­ increase the neu­tro­phil count but is asso­ci­ated with the exac­er­
pathic neutropenia, but the response was mostly par­tial and was ba­tion of spleno­meg­aly and the acute flare of joint symp­toms
lost upon taper­ing or discontinuing the treat­ment.36 In the SCNIR, while cyclo­phos­pha­mide, meth­o­trex­ate, and cyclo­spor­ine can
almost all­of the 48 adult patients with auto­im­mune or chronic idi­o­ be effec­tive.46
pathic neutropenia on immune mod­u­la­tion (glucocorticosteroids,
gamma glob­u­lin, meth­o­trex­ate, cyclo­spor­ine) discontinued this Monitoring con­gen­i­tal neutropenia patients at risk
ther­apy with­out resump­tion once placed on G-CSF.42 Additional for MDS/AML
reports have also been published dem­on­strat­ing the low response Many con­gen­i­tal neutropenia dis­or­ders impose a risk of trans­
rate and high relapse rate with glucocorticosteroids, gamma glob­ for­ma­tion to a mye­loid malig­nancy (see Table 3). The mech­a­
u­lin, cyclo­spor­ine, and rituximab.45 However, treat­ment with these nism of leu­ke­mo­gen­e­sis in these dis­or­ders is still actively under
agents may be con­sid­ered if patients fail to respond to G-CSF. inves­ti­ga­tion and is unique to each dis­or­der. As knowl­edge of
In con­trast, although rarely requir­ing treat­ment, ­sec­ond­ary leu­ke­mo­gen­e­sis in these dis­or­ders increases, the abil­ity to pre­
auto­im­mune neutropenia often responds to the immune sup­pres­ dict and detect early malig­nant cells will hope­fully be a nat­ur­al
sion used to treat other auto­im­mune symp­toms.17 LGL-asso­ci­ated con­se­quence. Historic and cur­rent rec­om­men­da­tions are to per­
neutropenia is often severe and requires treat­ment. G-CSF can form fre­quent (every 3-4 months) CBCs and annual bone mar­row

498  |  Hematology 2021  |  ASH Education Program


­ spi­ra­tion and biop­sies to mon­i­tor for cytopenias and mor­pho­
a has spe­cial con­sid­er­ations in con­gen­i­tal neutropenia. The mea­
logic changes concerning for MDS/AML devel­op­ment in con­gen­i­ sure­ment of hematopoietic progenitor cell (HPC) chi­me­rism is
tal neutropenias sec­ond­ary to muta­tions in ELANE, HAX1, G6PC3, dif­fi­cult in the clin­i­cal set­ting, and typ­i­cally, the mea­sure­ment of
SBDS, WAS (X-linked neutropenia), and GATA2.43 However, data mature mye­loid cells in the periph­eral blood is used as a sur­ro­
are needed to inform sur­veil­lance prac­tices and their effect on gate for HPC engraft­ment. However, mye­loid mark­ers, such as
out­comes for patients with con­gen­i­tal neutropenia. CD33, that are pres­ent on neu­tro­phils will not be an accu­rate
proxy for HPC chi­me­rism because recip­i­ent HPCs will have a
sig­nif­i­cant dis­ad­van­tage com­pared to donor HPCs in con­trib­ut­
ing to the periph­eral blood neu­tro­phil pool. Therefore, markers
CLINICAL CASE (Con­t in­u ed) weakly expressed or absent on neutrophils, such as CD14 (mono-
The patient undergoes CBCs every 3 to 4 months and 2 sur­veil­ cytes), CD56 (natural killer cells), CD19 (B cells), CD34 (HPCs),
lance bone mar­row exam­i­na­tions over the fol­low­ing year. His and CD3 (T cells), should be included in measuring peripheral
CBCs con­tinue to show mild neutropenia and throm­bo­cy­to­pe­ blood chimerism, particulalry in disorders such as GATA2 haplo-

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/492/1851746/492connelly.pdf by guest on 13 December 2021


nia, and his bone mar­row results are con­sis­tent with find­ings insufficiency, which affect multiple cell lineages.
from his first exam­i­na­tion. At the age of 22, he undergoes another The impact of par­ tial chi­
me­rism and long-term out­ comes
bone mar­row biopsy, which dem­on­strates an i(7)(q10) clone on for patients with con­gen­i­tal neutropenia is poorly under­stood,
cyto­ge­net­ics and a TP53 path­o­genic var­i­ant in a somatic NGS in large part because myeloablative reg­i­mens have dom­i­nated
panel. There con­tin­ues to be no mor­pho­logic or flow cytomet- early trans­plant approaches, and lit­er­a­ture reports have not rou­
ric evi­dence of increas­ing dys­pla­sia or an abnor­mal blast count. tinely focused on this trans­plant result or relied on neu­tro­phil-
Genetic test­ing is revisited, and Sanger sequenc­ing of the SBDS spe­cific mark­ers for chi­me­rism reporting. Mixed chi­me­rism has
locus identifies 2 path­o­genic muta­tions inde­pen­dently inherited been shown to cure the neutropenia in con­gen­i­tal neutropenia
from each par­ent. The pres­ence of a chro­mo­some 7 abnor­mal­ patients.47 Although the per­cent­age required to elim­i­nate the
ity and a new TP53 clonal hema­to­poi­e­sis raises the con­cern for risk of bac­te­rial infec­tion and G-CSF inde­pen­dence is uncer­tain,
a malig­nancy, and the patient is referred for HSCT con­sul­ta­tion. from anec­dotal reports and expe­ri­ence it is likely 20% to 30%
Author com­men­tary: The phy­si­cian was ­able to diag­nose HPC donor chi­me­rism.47
SDS in this patient with genetic test­ing, but this case illus­trates The more dif­fi­cult ques­tion is whether the per­sis­tence of recip­
the com­plex­ity of clin­i­cal sequenc­ing in that NGS tech­nol­ogy i­ent hema­to­poi­e­sis fol­low­ing expo­sure to che­mo­ther­apy will con­
has sev­eral short­com­ings, includ­ing the inabil­ity to dif­fer­en­ti­ate tinue to pres­ent a risk for mye­loid malig­nan­cies post trans­plant.
genes from pseudogenes with­out excel­lent map­ping and ana­lyt­ Our cur­rent under­stand­ing of leu­ke­mo­gen­e­sis in each dis­or­der pro­
ics and to iden­tify deep intronic var­i­ants. In most clin­i­cal sce­nar­ vi­des the­o­ret­i­cal hypoth­e­ses, but prolonged fol­low-up in patients
ios, chro­mo­some 7 aber­ra­tions raise the con­cern for MDS/AML. with mixed chi­me­rism will be required to answer this ques­tion
But sim­i­lar to del(20q), i(7)(q10) is a somatic change that does not defin­i­tively. In patients with ELANE SCN, a mar­row micro­en­vi­ron­
have an impact on tumor sur­veil­lance machin­ery. TP53 muta­tions ment with high lev­els of G-CSF pro­vi­des a com­pet­i­tive set­ting to
increase cel­lu­lar fit­ness in SDS, but at the expense of decreased select out clones with enhanced response to G-CSF. However, in
tumor sur­veil­lance, and likely increase the risk of MDS/AML within the posttransplant set­ting with abla­tion of the neutropenia defect
the clone. However, TP53 clonal hema­to­poi­e­sis is com­mon in by donor HPCs, the selec­tive envi­ron­ment driven by high lev­els of
SDS with­out malig­nancy, and iden­ti­fi­ca­tion by itself is not an indi­ G-CSF is elim­i­nated. If this pos­tu­late is cor­rect, the risk of malig­
ca­tion for HSCT. After dis­cus­sions with experts in SDS, a deci­sion nant trans­for­ma­tion should also be min­i­mized, if not abolished,
is made for con­tin­ued close mon­i­tor­ing that includes CBCs every with donor hema­to­poi­e­sis. In patients with SDS, the leu­ke­mo­gen­
3 to 4 months and annual bone mar­row mon­i­tor­ing only. e­sis hypoth­e­sis is less reli­ant on exter­nal pres­sures for hema­to­
poi­e­sis but depen­dent on indi­vid­ual cell fit­ness through reliev­ing
ribo­somal stress through TP53 muta­tions. Whether the reduc­tion
Curative approach with HSCT for con­gen­i­tal neutropenia of rep­li­ca­tive and inflam­ma­tory stress through donor hema­to­poi­
Absolute and pos­si­ble indi­ca­tions to pro­ceed to trans­plant e­sis is suf­fi­cient to pre­vent leu­ke­mo­gen­e­sis in recip­i­ent stem cells
depend on the under­ly­ing genetic con­di­tion, but com­mon under ribo­somal stress needs fur­ther study. As leu­ke­mia may not
causes to pro­ceed to HSCT include poor response to G-CSF, be a com­mon event in posttransplant patients, research eval­u­at­
recur­rent and severe infec­tions, BMF, and trans­for­ma­tion to a ing clonal dynam­ics with a par­tic­u­lar focus on CSF3R in SCN and
mye­loid malig­nancy. The approach to HSCT is not uni­form within TP53 in SDS will be incred­i­bly valu­able to under­stand­ing hema­to­
or between genetic dis­or­ders, but each trans­plant method shares poi­e­sis dynam­ics in the mixed-chi­mera patient.
unique obsta­cles to suc­cess­in neutropenic patients. The risk Graft rejec­tion for both malig­nant and non­ma­lig­nant indi­ca­
of bac­te­rial and fun­gal infec­tion is higher early in trans­plant due tions in con­gen­i­tal neutropenia is rel­a­tively high com­pared to
to pre­ced­ing neutropenia that rap­idly declines with the dis­con­ other com­pa­ra­ble con­di­tions. Several hypoth­e­ses have been
tin­u­a­tion of G-CSF. Evaluation for occult infec­tion pretransplant gen­er­ated to explain this phe­nom­e­non, includ­ing ­com­pe­tent
with sinus, chest, abdo­men, and pel­vis imag­ing is employed, lym­pho­cyte immu­nity pre­ced­ing con­di­tion­ing ther­apy, an inflam­
with more inva­sive mea­sures such as bron­chos­copy or biopsy ma­tory micro­en­vi­ron­ment induced from recur­rent infec­tion,
indi­cated for areas concerning for infec­tion. Bacterial and mold alter­ations in the bone mar­row niche sec­ond­ary to the under­ly­
pro­phy­laxis at the start of con­di­tion­ing ther­apy is ini­ti­ated to ing defect or high expo­sure to G-CSF,48 and prior expo­sure to for­
mit­i­gate the very early neutropenia in these patients. eign blood prod­ucts in patients with BMF. Serotherapy is almost
Chimerism anal­y­sis to dif­fer­en­ti­ate between resid­ual and stan­ dard today for non­ ma­lig­
nant trans­ plant to help address
engrafted donor hema­to­poi­e­sis is impor­tant for all­HSCTs but con­cerns for increased alloreactivity, but ques­tions regard­ing
Prakash Singh Shekhawat
Diagnosis and deci­sion-mak­ing for neutropenia  |  499
the mar­row niche are more dif­fic ­ ult and will require con­tin­ued entire cohort, but relapse was only seen in 2 of 14 (14%) patients.
research in this area, par­tic­u­larly if pre- and posttransplant mar­ An Amer­i­can Society of Hematology abstract from the Euro­pean
row aspi­ra­tion sam­ples are avail­­able for study. branch of the SCNIR reported out­comes on 51 patients after the
Congenital neutropenia patients who prog­ress to malig­nancy 2001 implementation of guide­lines to avoid induc­tion che­mo­ther­
pres­ent the most chal­leng­ing and most con­tro­ver­sial sce­nario for apy and dem­on­strated an OS of 77.8% and 83.3% in SCN patients
cli­ni­cians. First, the diag­no­sis of trans­for­ma­tion is not straight­for­ treated with and with­out malig­nancy.57 Despite the fact that not
ward because dif­fer­en­ti­at­ing hema­to­poi­etic dys­cra­sias pres­ent all­patients with SCN and sec­ ond­ ary malig­
nancy pro­ceed to
in many inborn errors of hema­to­poi­e­sis from dys­plas­tic clones HSCT in remis­sion, the reported relapse rates are much lower in
can be dif­fi­cult even for expe­ri­enced hematopathologists. This com­par­i­son to de novo MDS/AML (10%-15% vs 25%-40% for MDS
was illus­trated in a study of SDS patients with MDS/AML who and 40%-50% for AML).55,56,58-60 The rea­son for reduced relapsed
under­went a cen­tral path­o­logic review that showed a 56% dis­ rates may be sec­ond­ary to the growth restraint on malig­nant
crep­ancy from the orig­i­nal pathol­ogy report.49 Clonal hema­to­ cells afforded by the germline defect, a fea­ture reflected in some
poi­e­sis is also com­mon in con­gen­i­tal neutropenia, and some of case reports of SCN patients with “indo­lent” AML.61 Although

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/492/1851746/492connelly.pdf by guest on 13 December 2021


these changes are not believed to be induc­ers of leu­ke­mo­genic reported OS between myeloablation (MA) and reduced inten­
changes. Knowledge of path­o­logic clonal hema­to­poi­e­sis is key sity con­di­tion­ing (RIC) is sim­i­lar in SCN patients, myeloablative
in interpreting these find­ings. (MA) con­di­tion­ing is recommended for patients with under­ly­ing
Second, cytoreductive che­mo­ther­apy to reduce blast counts, malig­nancy for elim­i­na­tion of the malig­nant clone.
as is typ­i­cally done for de novo AML, can be incred­i­bly toxic to
con­gen­i­tal neutropenia patients who have dif­fi­culty recov­er­ing HSCT out­comes for SDS
neu­tro­phils for infec­tion erad­i­ca­tion and tis­sue repair or who HSCT for SDS is conducted in patients with BMF or hema­to­logic
have under­ly­ing organ dys­func­tion from the under­ly­ing genetic malig­nancy. Unlike some patients with SCN, the con­sen­sus is to
defect or recur­rent infec­tions.50 As such, recent approaches often not per­form HSCT pre­emp­tively in this dis­or­der. Early expe­ri­
elim­ i­
nate cytoreductive ther­ apy com­ pletely or have adopted ence in HSCT dem­on­strated high nonrelapse trans­plant mor­tal­
reg­i­mens used in med­i­cally frag­ile patients to bridge the patient ity, as shown in a recent long-term out­come study reporting that
while awaiting iden­ti­fi­ca­tion of a suit­able bone mar­row donor. 21% of patients died from tox­ic­ity.62 This real­i­za­tion prompted
Driving blast pro­duc­tion with G-CSF has been dem­on­strated in many cli­ni­cians to use RIC for all­SDS patients with BMF.63,64 In a
SCN, and judi­cious use fol­low­ing che­mo­ther­apy is warranted. The study by the Center for International Blood and Marrow Trans-
admis­sion of all­ patients fol­low­ing che­mo­ther­apy to mon­i­tor for plant Research, early deaths were seen in 4 of 13 (31%) SDS BMF
infec­tion and insti­tu­tion of G-CSF for evi­dence of severe bac­te­rial patients receiv­ing a MA con­di­tion­ing reg­i­men com­pared to
or fun­gal dis­ease is a com­mon approach to bal­ance these risks. 4 of 26 (15%) early deaths in patients receiv­ing RIC.65 This report
G-CSF is not recommended post HSCT for patients with SCN and showed a 5-year OS of 72% in patients with BMF and graft rejec­
malig­nancy with­out severe infec­tion, but whether G-CSF use fol­ tion in 3 of 39 patients (8%). The larg­est study to date published
low­ing trans­plant increases the risk of relapse is unknown. by the Severe Aplastic Anemia Working Party of the Euro­pean
Society for Blood and Marrow Transplantation on 61 patients
HSCT out­comes for SCN (1988-2016) with BMF dem­on­strated a 5-year OS of 70.7% with a
HSCT for SCN is conducted for G-CSF refrac­to­ri­ness or the devel­ graft fail­ure rate of 13%.62 OS with RIC and MA was iden­ti­cal, but
op­ment of MDS/AML. Patients with ELANE muta­tions at high risk the seg­re­ga­tion of con­di­tion­ing ther­apy by indi­ca­tion (BMF vs
of pro­gres­sion to AML, includ­ing p.G214R, p.C151Y, and p.C223ter, leu­ke­mia) was not pro­vided, and the authors con­cluded that MA
may be con­sid­ered for pre­emp­tive HSCT, but data to inform reg­i­mens should be reserved for malig­nant trans­for­ma­tion only.
the risks and ben­e­fits of this prac­tice are cur­rently lacking.51,52 Transplant sur­vival for SDS patients who develop malig­nancy
Some groups also con­sider high-dose G-CSF (>15 µg/kg/d) as an are abys­mal across lit­er­a­ture reports. A mul­ti­cen­ter study focused
indi­ca­tion to offer trans­plant because patients on higher G-CSF on malig­nant trans­plant out­comes dem­on­strated a 3-year OS
doses with decreased ANC response have shown an increased of 51% and 11% for patients with MDS and AML, respec­tively.49
fre­quency of leu­ke­mia devel­op­ment.53,54 This approach has not This result is in con­ trast to recent SCN out­ comes for malig­
been uni­ver­sally adopted sec­ond­ary to the desire to avoid HSCT nancy, indi­cat­ing a higher degree of resis­tance to trans­plant,
expo­sure to the major­ity of patients on high-dose G-CSF who per­haps in part related to increased che­mo­ther­apy resis­tance
will not develop malig­nancy. from mutated TP53, as seen in other can­cers.66 The impor­tance
Most lit­er­a­ture on HSCT for SCN is based on case reports of sur­veil­lance was suggested by an improved 3-year OS of 62%
and small case series that have dem­on­strated good out­comes vs 28% in patients who had reg­u­lar bone mar­row eval­u­at­ion,
in patients with­out malig­nancy (over­all sur­vival [OS], approx­i­ma­ but reg­u­lar CBCs had poor sen­si­tiv­ity in iden­ti­fy­ing mar­row dis­
tely 90%; event-free sur­vival [EFS], approx­im ­ a­tely 75%) despite ease.49 Although early detec­tion may pro­vide an incre­men­tal
high rates of graft fail­ure (approx­i­ma­tely 20%).55 Historical out­ improve­ment in sur­vival, novel approaches to avoid exces­sive
comes with malig­nancy have been less suc­cess­ful, with an OS of tox­ic­ity from con­di­tion­ing ther­apy while pre­vent­ing relapse are
approx­i­ma­tely 45% and an EFS of approx­i­ma­tely 40%, but many clearly needed for SDS patients who develop malig­nancy.
cases were transplanted prior to the adop­tion of mod­ern prac­
tices to avoid inten­sive cytoreductive ther­apy prior to trans­plant. Gene ther­apy and other poten­tial treat­ment modal­i­ties
A larger report of 136 SCN patients from the Euro­pean Society for con­gen­i­tal neutropenia
for Blood and Marrow Transplantation dem­on­strated a 3-year OS As our under­stand­ing of the mech­a­nism of neutropenia and leu­
and EFS of 87% and 77% for patients with­out malig­nancy and ke­mia for­ma­tion evolves, new ther­a­peu­tic modal­i­ties have been
79% and 64% with malig­nancy.56 Graft rejec­tion was 10% for the tested in pre­clin­i­cal mod­els with a pri­mary focus on increas­ing

500  |  Hematology 2021  |  ASH Education Program


neu­tro­phil mat­u­ra­tion and pro­duc­tion. Investigative ther­a­pies References
include nic­o­tin­amide to increase the expres­sion of G-CSF and 1. Reich D, Nalls MA, Kao WH, et al. Reduced neu­tro­phil count in peo­ple of
G-CSF recep­tors in SCN,67,68 neu­tro­phil elas­tase inhib­i­tors to Afri­can descent is due to a reg­u­la­tory var­i­ant in the Duffy anti­gen recep­tor
for chemokines gene. PLoS Genet. 2009;5(1):e1000360.
pre­vent the unfolded pro­tein response from mutated ELANE,69,70 2. Frater JL. How I inves­ti­gate neutropenia. Int J Lab Hematol. 2020;42(suppl
and gene editing to repair mutated ELANE or reduce trans­la­tion 1):121-132.
of the ELANE gene.71-73 Nicotinamide has been used in a small 3. Schaffner A, Fehr J. Granulocyte demargination by epi­neph­rine in eval­u­a­
clin­i­cal trial with pos­i­tive results, but other modal­i­ties have tion of hypersplenic states. Scand J Haematol. 1981;27(4):225-230.
4. Papadaki HA, Eliopoulos AG, Kosteas T, et  al. Impaired gran­u­lo­cy­to­poi­
only reported results in the pre­clin­i­cal set­ting. The use of small-
e­sis in patients with chronic idi­ o­pathic neutropenia is asso­ ci­ated with
mol­e­cule inhib­i­tors to pro­mote non­sense sup­pres­sion in increased apo­pto­sis of bone mar­row mye­loid pro­gen­i­tor cells. Blood.
patients with ter­mi­na­tion SBDS muta­tions (c.183-184TA > TC) 2003;101(7):2591-2600.
has also dem­ on­strated prom­ ise in reduc­ ing apo­pto­sis and 5. Marins LR, Anizelli LB, Romanowski MD, Sarquis AL. How does pre­eclamp­
increas­ing neu­tro­phil mat­u­ra­tion in bone mar­row pro­gen­i­ sia affect neo­na­tes? Highlights in the dis­ease’s immu­nity. J Matern Fetal
Neonatal Med. 2019;32(7):1205-1212.
tor cells.74 Inhibition of EIF6, a pro­tein removed by SBDS to 6. Melvan JN, Bagby GJ, Welsh DA, Nelson S, Zhang P. Neonatal sep­sis and

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/492/1851746/492connelly.pdf by guest on 13 December 2021


advance ribo­some for­ma­tion, is also an attrac­tive ther­a­peu­tic neu­tro­phil insufficiencies. Int Rev Immunol. 2010;29(3):315-348.
tar­get for SDS treat­ment but still requires pre­clin­i­cal inves­ti­ 7. Bousfiha A, Jeddane L, Picard C, et  al. Human inborn errors of immu­
ga­tion. nity: 2019 update of the IUIS phe­no­typ­i­cal clas­si­fi­ca­tion. J Clin Immunol.
2020;40(1):66-81.
8. Larocque A, Hoffman RS. Levamisole in cocaine: unex­pected news from an
Summary old acquain­tance. Clin Toxicol (Phila). 2012;50(4):231-241.
The diag­no­sis and man­age­ment of the neutropenia patient are 9. Marsh JC, Abboudi ZH, Gib­son FM, et al. Aplastic anae­mia fol­low­ing expo­
sig­nif­i­cantly aided by clas­si­fy­ing dis­or­ders into the pres­ence or sure to 3,4-methylenedioxymethamphetamine (“ecstasy”). Br J Haematol.
absence of ade­quate bone mar­row reserves. Patients with inef­ 1994;88(2):281-285.
10. Powars D. Aplastic ane­ mia sec­ ond­ary to glue sniffing. N Engl J Med.
fi­cient myelopoiesis have a lower thresh­old to ini­ti­ate empiric 1965;273(13):700-702.
treat­ment for fever and may require mon­i­tor­ing for trans­for­ 11. Shi X, DeLucia AL, Bao J, Zhang P. Alcohol abuse and dis­or­der of granulo-
ma­tion into a mye­loid malig­nancy. Intact myelopoiesis patients poiesis. Pharmacol Ther. 2019;198(June):206-219.
with ade­quate mar­row neu­tro­phil reserves are at much lower 12. Yarandi SS, Griffith DP, Sharma R, Mohan A, Zhao VM, Ziegler TR. Optic
neu­rop­a­thy, mye­lop­a­thy, ane­mia, and neutropenia caused by acquired
risk of these com­pli­ca­tions despite a low periph­eral blood ANC.
cop­ per defi­ ciency after gas­ tric bypass sur­ gery. J Clin Gastroenterol.
G-CSF is com­monly used for poor neu­tro­phil pro­duc­tion and 2014;48(10):862-865.
may pro­vide a ben­e­fit in some sce­nar­ios of competent neu­ 13. Dale DC. How I man­ age chil­ dren with neutropenia. Br J Haematol.
tro­phil pro­duc­tion. HSCT offers an oppor­tu­nity for cure in con­ 2017;178(3):351-363.
gen­i­tal neutropenias, but ques­tions regard­ing the indi­ca­tions, 14. Shaver AK, Walkovich K, Connelly JA. Recurrence of neo­na­tal lupus post-
cord blood trans­ plant for severe con­ gen­i­
tal neutropenia. Pediatrics.
the impor­ tance of posttransplant chi­ me­rism, and the tech­ 2015;136(2):e535-e538.
niques to reduce tox­ic­ity and malig­nant relapse in SDS limit 15. Farruggia P, Fioredda F, Puccio G, et al. Autoimmune neutropenia of infancy:
the opti­mi­za­tion of patient selec­tion, con­di­tion­ing approaches, data from the Ital­ ian Neutropenia Registry. Am J Hematol. 2015;90(12):
and out­comes. Gene editing may pro­vide an alter­na­tive option E221-E222.
16. Myers KC, Bolyard AA, Otto B, et al. Variable clin­i­cal pre­sen­ta­tion of Shwa-
to HSCT to cure the neutropenia, but the anal­o­gous ques­tion
chman-Diamond syn­drome: update from the North Amer­i­can Shwachman-
to HSCT chi­me­rism of resid­ual uncor­rected hema­to­poi­etic Diamond Syndrome Registry. J Pediatr. 2014;164(4):866-870.
stem and pro­gen­i­tor cells resulting in con­tin­ued can­cer risk is 17. Gib­son C, Berliner N. How we eval­u­ate and treat neutropenia in adults.
unknown. Continued col­lab­o­ra­tive research in clin­i­cal reg­is­tries Blood. 2014;124(8):1251-1258, quiz 1378.
and basic sci­ence inves­ti­ga­tion are nec­es­sary to answer these 18. Michniacki TF, Connelly JA, Sturza J, et al. Neutropenia is an underrecog-
nized find­ing in pedi­at­ric pri­mary immu­no­de­fi­ciency dis­eases: an anal­y­sis
ques­tions and pro­vide addi­tional evi­dence for impor­tant clin­i­ of the United States Immunodeficiency Network Registry. J Pediatr Hema-
cal deci­sions for neutropenic patients. tol Oncol. 2020;42(7):e601-e605.
19. Furutani E, Newburger PE, Shimamura A. Neutropenia in the age of genetic
test­ing: advances and chal­lenges. Am J Hematol. 2019;94(3):384-393.
Conflict-of-inter­est dis­clo­sure
20. Hao R, Saleh M, Liang T, et  al. The prev­a­lence of seri­ous bac­te­rial infec­
James A. Connelly: advi­sory board mem­ber: X4 Pharmaceuticals, tions in neutropenic immu­no­com­pe­tent febrile chil­dren. Am J Emerg Med.
Horizon Therapeutics. 2021;45(July):1-6.
Kelly Walkovich: con­ sul­
tancy: Swed­ ish Orphan Biovitrum AB; 21. Melendez E, Harper MB. Risk of seri­ous bac­te­rial infec­tion in iso­lated and
advi­sory board mem­ber: Horizon Therapeutics, Pharming; local unsus­pected neutropenia. Acad Emerg Med. 2010;17(2):163-167.
22. Bachur RG, Harper MB. Predictive model for seri­ ous bac­te­
rial infec­tions
prin­ci­pal inves­ti­ga­tor: clin­i­cal trial on mavorixafor spon­sored by among infants youn­ger than 3 months of age. Pediatrics. 2001;108(2):311-316.
X4 Pharmaceuticals. 23. Pascual C, Trenchs V, Hernández-Bou S, Català A, Valls AF, Luaces C. Out-
comes and infec­tious eti­­ol­o­gies of febrile neutropenia in non-immu­no­com­
pro­mised chil­dren who pres­ent in an emer­gency depart­ment. Eur J Clin
Off-label drug use Microbiol Infect Dis. 2016;35(10):1667-1672.
James A. Connelly: nothing to disclose. 24. Barg AA, Kozer E, Mordish Y, Lazarovitch T, Kventsel I, Goldman M. The risk
Kelly Walkovich: nothing to disclose. of seri­ous bac­te­rial infec­tion in neutropenic immu­no­com­pe­tent febrile
chil­dren. J Pediatr Hematol Oncol. 2015;37(6):e347-e351.
25. Bonadio WA, Stremski E, Shallow K. Clinical char­ac­ter­is­tics of chil­dren with
Correspondence fever and tran­sient neutropenia who expe­ri­ence seri­ous bac­te­rial infec­
tions. Pediatr Emerg Care. 1989;5(3):163-165.
James A. Connelly, Department of Pediatrics, Vanderbilt Universi-
26. Wittmann O, Rimon A, Scolnik D, Glatstein M. Outcomes of immu­no­com­
ty Medical Center, 389 PRB, 2220 Pierce Ave, Nashville, TN 37232; pe­ tent chil­dren presenting with fever and neutropenia. J Emerg Med.
e-mail: james​­.a​­.connelly@vumc​­.org. 2018;54(3):315-319.

Prakash Singh Shekhawat


Diagnosis and deci­sion-mak­ing for neutropenia  |  501
27. Husain EH, Mullah-Ali A, Al-Sharidah S, Azab AF, Adekile A. Infectious eti­­ol­o­ 50. Choi SW, Boxer LA, Pulsipher MA, et al. Stem cell trans­plan­ta­tion in patients
gies of tran­sient neutropenia in pre­vi­ously healthy chil­dren. Pediatr Infect with severe con­gen­i­tal neutropenia with evi­dence of leu­ke­mic trans­for­ma­
Dis J. 2012;31(6):575-577. tion. Bone Marrow Transplant. 2005;35(5):473-477.
28. Tschernin D, Fruchtman Y, Sergienko R, et al. The eti­o­logic, micro­bi­o­logic, 51. Makaryan V, Zeidler C, Bolyard AA, et  al. The diver­sity of muta­tions and
clin­i­cal and out­come char­ac­ter­is­tics of immu­no­com­pe­tent young chil­dren clin­i­cal out­comes for ELANE-asso­ci­ated neutropenia. Curr Opin Hematol.
<2 years of age hos­pi­tal­ized with acute neutropenia. Pediatr Neonatol. 2015;22(1):3-11.
2021;62(1):26-35. 52. Dale DC, Makaryan V, Kelly ML, et al. Termination and frame­shift muta­tions
29. Leibovitz E, Kapelushnik J, Alsanaa S, et  al. Comparison of the eti­o­logic, in ELANE are asso­ci­ated with adverse out­comes in patients with severe
micro­bi­o­logic, clin­i­cal and out­come char­ac­ter­is­tics of febrile vs. non-fe- chronic neutropenia. Blood. 2016;128(22):1326.
brile neutropenia in hos­pi­tal­ized immu­no­com­pe­tent chil­dren. Eur J Clin 53. Rotulo GA, Beaupain B, Rialland F, et al. HSCT may lower leu­ke­mia risk in
Microbiol Infect Dis. 2020;39(12):2415-2426. ELANE neutropenia: a before-after study from the French Severe Congeni-
30. Alexandropoulou O, Kossiva L, Haliotis F, et  al. Transient neutropenia in tal Neutropenia Registry. Bone Marrow Transplant. 2020;55(8):1614-1622.
chil­dren with febrile ill­ness and asso­ci­ated infec­tious agents: 2 years’ 54. Rosenberg PS, Zeidler C, Bolyard AA, et al. Stable long-term risk of leu­kae­
fol­low-up. Eur J Pediatr. 2013;172(6):811-819. mia in patients with severe con­gen­i­tal neutropenia maintained on G-CSF
31. Karavanaki K, Polychronopoulou S, Giannaki M, et al. Transient and chronic ther­apy. Br J Haematol. 2010;150(2):196-199.
neutropenias detected in chil­dren with dif­fer­ent viral and bac­te­rial infec­ 55. Connelly JA, Choi SW, Levine JE. Hematopoietic stem cell trans­plan­ta­tion

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/492/1851746/492connelly.pdf by guest on 13 December 2021


tions. Acta Paediatr. 2006;95(5):565-572. for severe con­gen­i­tal neutropenia. Curr Opin Hematol. 2012;19(1):44-51.
32. Palmblad J, Dufour C, Papadaki HA. How we diag­ nose neutropenia in 56. Fioredda F, Iacobelli S, van Biezen A, et al; Severe Aplastic Anemia the
the adult and elderly patient. Haematologica. 2014;99(7):1130-1133. Inborn Error; Pediatric Disease Working Parties of the Euro­pean Society
33. Kirk SE, Grimes AB, Shelke S, Despotovic JM, Powers JM. The cost of a for Blood and Bone Marrow Transplantation and Stem Cell Transplant for
“benign” con­di­tion: healthcare uti­li­za­tion and infec­tious out­comes in Immunodeficiencies in Europe. Stem cell trans­plan­ta­tion in severe con­gen­
young chil­dren with pri­mary auto­im­mune neutropenia. Pediatr Blood Can- i­tal neutropenia: an anal­y­sis from the Euro­pean Society for Blood and Mar-
cer. 2020;67(4):e28146. row Transplantation. Blood. 2015;126(16):1885-1892, quiz 1970.
34. Fioredda F, Calvillo M, Burlando O, et al. Infectious com­pli­ca­tions in chil­ 57. Zeidler C, Nickel A, Skyora KW, Welte K. Improved out­come of stem cell
dren with severe con­gen­i­tal, auto­im­mune or idi­o­pathic neutropenia: a ret­ trans­plan­ta­tion for severe chronic neutropenia with or with­out sec­ond­ary
ro­spec­tive study from the Ital­ian Neutropenia Registry. Pediatr Infect Dis J. leu­ke­mia: a long-term anal­y­sis of Euro­pean data for more than 25 years by
2013;32(4):410-412. the SCNIR. Blood. 2013;122(21):3347.
35. Newburger PE. Autoimmune and other acquired neutropenias. Hematol- 58. Choi EJ, Lee JH, Lee JH, et al. Treatment and clin­i­cal out­comes of patients
ogy Am Soc Hematol Educ Program. 2016;2016(1):38-42. relaps­ing after allo­ge­neic hema­to­poi­etic cell trans­plan­ta­tion for myelodys-
36. Sicre de Fontbrune F, Moignet A, Beaupain B, et al; French Severe Chronic plastic syn­drome. Blood Res. 2018;53(4):288-293.
Neutropenia Registry. Severe chronic pri­mary neutropenia in adults: report 59. Bitan M, He W, Zhang MJ, et al. Transplantation for chil­dren with acute mye­
on a series of 108 patients. Blood. 2015;126(14):1643-1650. loid leu­ke­mia: a com­par­i­son of out­comes with reduced inten­sity and mye-
37. Farruggia P, Puccio G, Fioredda F, et al. Autoimmune neutropenia of child­ loablative reg­i­mens. Blood. 2014;123(10):1615-1620.
hood sec­ ond­ary to other auto­ im­ mune dis­or­
ders: data from the Ital­ ian 60. Rautenberg C, Germing U, Haas R, Kobbe G, Schroeder T. Relapse of acute
Neutropenia Registry. Am J Hematol. 2017;92(9):E546-E549. mye­loid leu­ke­mia after allo­ge­neic stem cell trans­plan­ta­tion: pre­ven­tion,
38. Welte K, Zeidler C, Dale DC. Severe con­gen­i­tal neutropenia. Semin Hema- detec­tion, and treat­ment. Int J Mol Sci. 2019;20(1):228.
tol. 2006;43(3):189-195. 61. Connelly JA, Mody RJ, Wu YM, et al. Identification of novel MECOM gene
39. Fioredda F, Calvillo M, Bonanomi S, et al; Neutropenia Committee of the fusion and per­son­al­ized ther­a­peu­tic tar­gets through inte­gra­tive clin­i­cal
Marrow Failure Syndrome Group of the AIEOP (Associazione Italiana Ema- sequenc­ing in sec­ond­ary acute mye­loid leu­ke­mia in a patient with severe
to-Oncologia Pediatrica). Congenital and acquired neutropenias con­sen­ con­gen­i­tal neutropenia: a case report and lit­er­a­ture review. Cold Spring
sus guide­lines on ther­apy and fol­low-up in child­hood from the Neutropenia Harb Mol Case Stud. 2018;4(2):a002204.
Committee of the Marrow Failure Syndrome Group of the AIEOP (Associazi- 62. Cesaro S, Pillon M, Sauer M, et  al. Long-term out­come after allo­ge­neic
one Italiana Emato-Oncologia Pediatrica). Am J Hematol. 2012;87(2):238- hema­to­poi­etic stem cell trans­plan­ta­tion for Shwachman-Diamond syn­
243. drome: a ret­ ro­
spec­tive anal­ y­
sis and a review of the lit­ er­

ture by the
40. Link DC. Mechanisms of leu­ke­mic trans­for­ma­tion in con­gen­i­tal neutropenia. Severe Aplastic Anemia Working Party of the Euro­pean Society for Blood
Curr Opin Hematol. 2019;26(1):34-40. and Marrow Transplantation (SAAWP-EBMT). Bone Marrow Transplant.
41. Koch C, Samareh B, Morishima T, et al. GM-CSF treat­ment is not effec­tive 2020;55(9):1796-1809.
in con­gen­i­tal neutropenia patients due to its inabil­ity to acti­vate NAMPT 63. Burroughs LM, Shimamura A, Talano JA, et al. Allogeneic hema­to­poi­etic cell
sig­nal­ing. Ann Hematol. 2017;96(3):345-353. trans­plan­ta­tion using treosulfan-based con­di­tion­ing for treat­ment of mar­
42. Dale DC, Bolyard AA. An update on the diag­no­sis and treat­ment of chronic row fail­ure dis­or­ders. Biol Blood Marrow Transplant. 2017;23(10):1669-1677.
idi­o­pathic neutropenia. Curr Opin Hematol. 2017;24(1):46-53. 64. Bhatla D, Davies SM, Shenoy S, et  al. Reduced-inten­sity con­di­tion­ing is
43. Dale DC. How I diag­ nose and treat neutropenia. Curr Opin Hematol. effec­tive and safe for trans­plan­ta­tion of patients with Shwachman-Dia-
2016;23(1):1-4. mond syn­drome. Bone Marrow Transplant. 2008;42(3):159-165.
44. Farruggia P, Dufour C. Diagnosis and man­ age­ment of pri­ mary auto­im­ 65. Myers K, Hebert K, Antin J, et  al. Hematopoietic stem cell trans­plan­ta­
mune neutropenia in chil­dren: insights for cli­ni­cians. Ther Adv Hematol. tion for Shwachman-Diamond syn­drome. Biol Blood Marrow Transplant.
2015;6(1):15-24. 2020;26(8):1446-1451.
45. Dungarwalla M, Marsh JC, Tooze JA, et al. Lack of clin­i­cal effi­cacy of ritux- 66. Hientz K, Mohr A, Bhakta-Guha D, Efferth T. The role of p53 in can­cer drug
imab in the treat­ment of auto­im­mune neutropenia and pure red cell aplasia: resis­tance and targeted che­mo­ther­apy. Oncotarget. 2017;8(5):8921-8946.
impli­ca­tions for their path­o­phys­i­­ol­ogy. Ann Hematol. 2007;86(3):191-197. 67. Skokowa J, Lan D, Thakur BK, et  al. NAMPT is essen­tial for the G-CSF-
46. Lamy T, Loughran TP Jr. How I treat LGL leu­ke­mia. Blood. 2011;117(10):2764- induced mye­loid dif­fer­en­ti­a­tion via a NAD(+)-sirtuin-1-depen­dent path­way.
2774. Nat Med. 2009;15(2):151-158.
47. Zeidler C, Welte K, Barak Y, et al. Stem cell trans­plan­ta­tion in patients with 68. Deordieva E, Shvets O, Voronin K, et al. Nicotinamide (vita­min B3) treat­
severe con­gen­i­tal neutropenia with­out evi­dence of leu­ke­mic trans­for­ma­ ment improves response to G-CSF in severe con­ gen­ i­
tal neutropenia
tion. Blood. 2000;95(4):1195-1198. patients. Br J Haematol. 2021;192(4):788-792.
48. Bardelli D, Dander E, Bugarin C, et  al. Mesenchymal stro­mal cells from 69. Nanua S, Murakami M, Xia J, et  al. Activation of the unfolded pro­ tein
Shwachman-Diamond syn­ drome patients fail to rec­ re­
ate a bone mar­ response is asso­ci­ated with impaired granulopoiesis in trans­genic mice
row niche in vivo and exhibit impaired angio­ gen­ e­sis. Br J Haematol. expressing mutant Elane. Blood. 2011;117(13):3539-3547.
2018;182(1):114-124. 70. Makaryan V, Kelley ML, Fletcher B, Bolyard AA, Aprikyan AA, Dale DC. Elas-
49. Myers KC, Furutani E, Weller E, et  al. Clinical fea­tures and out­comes of tase inhib­i­tors as poten­tial ther­a­pies for ELANE-asso­ci­ated neutropenia.
patients with Shwachman-Diamond syn­drome and myelodysplastic syn­ J Leukoc Biol. 2017;102(4):1143-1151.
drome or acute mye­loid leu­kae­mia: a multicentre, ret­ro­spec­tive, cohort 71. Ritter MU, Secker B, Klimiankou M, et al. Efficient cor­rec­tion of ELANE muta­
study. Lancet Haematol. 2020;7(3):e238-e246. tions in pri­mary HSPCs of severe con­gen­i­tal neutropenia patients using

502  |  Hematology 2021  |  ASH Education Program


CRISPR/Cas9 and rAVV6 HDR repair tem­ plates. Blood. 2019;134(suppl 82. Negri G, Crescenzi B, Colombo EA, et al. Expanding the role of the splic­ing
1):1036. USB1 gene from poikiloderma with neutropenia to acquired mye­loid neo­
72. Nasri M, Ritter M, Mir P, et  al. CRISPR/Cas9-medi­ated ELANE knock­out plasms. Br J Haematol. 2015;171(4):557-565.
enables neu­tro­philic mat­u­ra­tion of pri­mary hema­to­poi­etic stem and pro­ 83. Donadieu J, Fenneteau O, Beaupain B, et al; Associated Investigators of
gen­i­tor cells and induced plu­rip­o­tent stem cells of severe con­gen­i­tal neu- the French Severe Chronic Neutropenia Registry*. Classification of and
tropenia patients. Haematologica. 2020;105(3):598-609. risk fac­tors for hema­ to­
logic com­ pli­
ca­
tions in a French national cohort
73. Rao S, Yao Y, Soares de Brito J, et al. Dissecting ELANE neutropenia path­ of 102 patients with Shwachman-Diamond syn­ drome. Haematologica.
o­ge­nic­ity by human HSC gene editing. Cell Stem Cell. 2021;28(5):833-845. 2012;97(9):1312-1319.
e5845e5. 84. Cada M, Segbefia CI, Klaassen R, et  al. The impact of cat­e­gory, cytopa-
74. Bezzerri V, Bardelli D, Morini J, et  al. Ataluren-driven res­ to­
ra­
tion of thology and cyto­ ge­net­ics on devel­ op­
ment and pro­ gres­
sion of clonal
­Shwachman-Bodian-Diamond syn­drome pro­tein func­tion in Shwachman- and malig­nant mye­loid trans­for­ma­tion in inherited bone mar­row fail­ure
Diamond syn­drome bone mar­row cells. Am J Hematol. 2018;93(4):527-536. ­syn­dromes. Haematologica. 2015;100(5):633-642.
75. Klimiankou M, Uenalan M, Kandabarau S, et al. Ultra-sen­si­tive CSF3R deep 85. Kennedy AL, Myers KC, Bowman J, et al. Distinct genetic path­ways define
sequenc­ing in patients with severe con­gen­i­tal neutropenia. Front Immu- pre-malig­nant ver­sus com­pen­sa­tory clonal hema­to­poi­e­sis in Shwachman-
nol. 2019;10(Feb­ru­ary):116. Diamond syn­drome. Nat Commun. 2021;12(1):1334.
76. Klimiankou M, Mellor-Heineke S, Klimenkova O, et al. Two cases of cyclic 86. Spinner MA, Sanchez LA, Hsu AP, et al. GATA2 defi­ciency: a pro­tean dis­or­

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/492/1851746/492connelly.pdf by guest on 13 December 2021


neutropenia with acquired CSF3R muta­ tions, with 1 devel­ op­ing AML. der of hema­to­poi­e­sis, lym­phat­ics, and immu­nity. Blood. 2014;123(6):809-
Blood. 2016;127(21):2638-2641. 821.
77. Carlsson G, Fasth A, Berglöf E, et al. Incidence of severe con­gen­i­tal neu- 87. Wlodarski MW, Hirabayashi S, Pastor V, et al; Euro­pean Working Groups
tropenia in Sweden and risk of evo­lu­tion to myelodysplastic syn­drome/ of Myelodysplastic Syndromes. Prevalence, clin­i­cal char­ac­ter­is­tics, and
leu­kae­mia. Br J Haematol. 2012;158(3):363-369. prog­no­sis of GATA2-related myelodysplastic syn­dromes in chil­dren and
78. Desplantes C, Fremond ML, Beaupain B, et al. Clinical spec­trum and long- ado­les­cents. Blood. 2016;127(11):1387-1397, quiz 1518.
term fol­low-up of 14 cases with G6PC3 muta­tions from the French Severe 88. Bluteau O, Sebert M, Leblanc T, et al. A land­scape of germ line muta­tions in
Congenital Neutropenia Registry. Orphanet J Rare Dis. 2014;9(10 Decem­ a cohort of inherited bone mar­row fail­ure patients. Blood. 2018;131(7):717-
ber):183. 732.
79. Ancliff PJ, Blundell MP, Cory GO, et al. Two novel acti­vat­ing muta­tions in the 89. Wong JC, Bryant V, Lamprecht T, et al. Germline SAMD9 and SAMD9L muta­
Wiskott-Aldrich syn­drome pro­tein result in con­gen­i­tal neutropenia. Blood. tions are asso­ci­ated with exten­sive genetic evo­lu­tion and diverse hema­to­
2006;108(7):2182-2189. logic out­comes. JCI Insight. 2018;3(14).
80. Dale DC, Bolyard AA, Marrero T, et  al. Neutropenia in gly­ co­gen stor­
age dis­ease Ib: out­comes for patients treated with granulocyte col­ony-
stim­u­lat­ing fac­tor. Curr Opin Hematol. 2019;26(1):16-21.
81. Li AM, Thyagu S, Maze D, et al. Prolonged granulocyte col­ony stim­u­lat­ing
fac­tor use in gly­co­gen stor­age dis­ease type 1b asso­ci­ated with acute mye­
loid leu­ke­mia and with short­ened telo­mere length. Pediatr Hematol Oncol. © 2021 by The Amer­i­can Society of Hematology
2018;35(1):45-51. DOI 10.1182/hema­tol­ogy.2021000284

Prakash Singh Shekhawat


Diagnosis and deci­sion-mak­ing for neutropenia  |  503
NEUTROPENIA: DOING MORE WITH LESS

Understanding neutropenia secondary to intrinsic


or iatrogenic immune dysregulation

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/504/1851486/504walkovich.pdf by guest on 13 December 2021


Kelly Walkovich1 and James A. Connelly2
Department of Pediatrics, University of Michigan, Ann Arbor, MI; and 2Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN
1

As a key member of the innate and adaptive immune response, neutrophils provide insights into the hematopoietic and
inflammatory manifestations of inborn errors of immunity (IEI) and the consequences of immunotherapy. The facile recog-
nition of IEI presenting with neutropenia provides an avenue for hematologists to facilitate early diagnosis and expedite
biologically rationale care. Moreover, enhancing the understanding of the molecular mechanisms driving neutropenia
in IEI—decreased bone marrow reserves, diminished egress from the bone marrow, and decreased survival—offers an
opportunity to further dissect the pathophysiology driving neutropenia secondary to iatrogenic immune dysregulation,
eg, immune checkpoint inhibitors and chimeric antigen receptor T-cell therapy.

LEARNING OBJECTIVES
• Identify neutropenia as a potential first sign of underlying inborn errors of immunity and utilize a targeted history
and physical exam to assess the need for additional evaluation
• Describe peripheral blood smear and bone marrow findings associated with IEI
• Discuss the benefits of early IEI diagnosis in relation to patient­specific, biologically rationale therapeutic
decision­making
• Explain the pathophysiology of iatrogenic neutropenia secondary to immunotherapy
• Illustrate potential correlations between neutropenia secondary to immunotherapy and known immune disorders

Introduction IEI­mediated neutropenia provide insights into the mecha­


While neutrophils have long been recognized as critical medi­ nism and management of neutropenia secondary to the
ators of first­line host defense and inflammation­induced immune­related therapies used by oncologists that lever­
injury, emerging evidence suggests a more nuanced and age the same molecular pathways to enhance immunologic
extensive role for neutrophils in both the innate and adap­ antitumor activity.
tive immune response.1 In parallel, neutropenia, defined as an
absolute neutrophil count (ANC) <1500 cells per microliter for Distinguishing patients with neutropenia related
patients over 1 year of age, is increasingly appreciated as a to IEI from other etiologies
clinical feature of inborn errors of immunity (IEI) and immune Differentiating neutropenia associated with IEI from more
dysregulation.2 Patients with IEI also frequently present with common postinfectious, drug­related, nutritional, or primary
other hematologic clinical features, eg, cytopenias, bone autoimmune etiologies can be challenging, particularly as
marrow failure, splenomegaly, and malignancy.3,4 the number of IEI are rapidly expanding and may present
Neutropenia in IEI and other immune dysregulation with variable or incomplete clinical features.4 The history
states can be a consequence of infection or a drug effect; should be inclusive of screening for phagocyte disorders
however, for several disorders neutropenia is the result of an and humoral and combined immunodeficiency as well as
intrinsic defect of myelopoiesis or an outcome of an improper specific questions to probe for bone marrow failure or
immune response. Understanding the pathophysiology driv­ immune dysregulation syndromes (see Table 1). Careful
ing neutropenia in IEI and immune dysregulation provides attention to the exam, particularly the oral cavity (eg, oral
an opportunity to intervene in a biologically rational man­ ulcers, gingivitis, abnormal dentition, periodontal disease),
ner. Hematologists adept at distinguishing neutropenia sec­ integumentary (eg, eczema, hemangiomas, hypopigmenta­
ondary to IEI or immune dysregulation from other etiologies tion, warts, prominent superficial veins, vasculitis, etc), and
can facilitate early diagnosis. Additionally, investigations into hematologic/lymphatic systems, can provide clues to an

504 | Hematology 2021 | ASH Education Program


Table 1. Targeted screen­ing by his­tory for IEI in patients presenting with neutropenia

Targeted screen­ing by his­tory for IEI Additional stud­ies to con­sider


Phagocyte dis­or­ders
Recurrent fevers Serial CBCPDs
Persistent or recur­rent oral ulcers DHR flow
Bleeding and/or reced­ing gums Genetic sequenc­ing
Gingivitis Echocardiogram
Dental anom­a­lies and/or den­tal infec­tions Renal ultra­sound
Frequent or deep-seated skin infec­tions Skeletal sur­vey
Perirectal infec­tions or inflam­ma­tory bowel dis­ease Genetic sequenc­ing
Developmental delay
Cardiac, renal, or skel­e­tal anom­a­lies
Personal or fam­ily his­tory of MDS/AML

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/504/1851486/504walkovich.pdf by guest on 13 December 2021


Bone mar­row fail­ure syn­dromes
Personal or fam­ily his­tory of bone mar­row fail­ure, MDS, AML, sar­co­mas, fat mal­ab­sorp­tion, Pancreatic amy­lase
warts, lymphedema, pul­mo­nary or hepatic fibro­sis, early graying of hair, nail Fecal elas­tase
abnor­mal­i­ties, stroke, vas­cu­li­tis Pancreatic ultra­sound
Telomere length
Chromosomal break­age
ADA2 level
Quantitative immu­no­glob­u­lins
T/B/NK-cell enu­mer­a­tion
Genetic sequenc­ing
Humoral defi­ciency
Four or more new ear infec­tions in 1 year Quantitative immu­no­glob­u­lins
Two or more sinus infec­tions or pneu­mo­nias requir­ing anti­bi­ot­ics within the past 1 year T/B/NK-cell enu­mer­a­tion
Requirement for prolonged course or mul­ti­ple courses of oral anti­bi­ot­ics to clear “sim­ple” Vaccine anti­body responses
infec­tions BTK flow cytom­e­try
Need for intra­ve­nous anti­bi­ot­ics and/or hos­pi­tal­i­za­tions for infec­tion treat­ment Genetic sequenc­ing
Combined immu­no­de­fi­ciency
Abnormal TREC new­born screen Quantitative immu­no­glob­u­lins
Failure to thrive T/B/NK-cell enu­mer­a­tion
Developmental delay Vaccine anti­body responses
Persistent oral thrush or fun­gal skin infec­tions Lymphocyte mito­gen pro­lif­er­a­tion
Two or more deep-seated infec­tions Lymphocyte anti­gen pro­lif­er­a­tion
Atypical or oppor­tu­nis­tic infec­tions TRECs
CD40L flow cytom­e­try
STAT1 func­tional screen­ing
Toll-like recep­tor-medi­ated sig­nal­ing screen­ing
Genetic sequenc­ing
Immune dysregulation
Personal or fam­ily his­tory of inflam­ma­tory bowel dis­ease, par­tic­u­larly with onset prior to Ferritin
age 6, auto­im­mune cytopenias, pro­gres­sive neu­ro­log­i­cal dys­func­tion, hemophagocytic TCR alpha-beta CD3+CD4−CD8− T cells
lymphohistiocytosis Genetic sequenc­ing
Evidence of non­ma­lig­nant lymphoproliferation with chronic lymph­ade­nop­a­thy or
spleno­meg­aly
Differences in hair or skin pig­men­ta­tion, par­tic­u­larly in rela­tion to imme­di­ate fam­ily
mem­bers
AML, acute mye­loid leu­ke­mia; CBCPD, com­plete blood count with plate­lets and dif­fer­en­tial; DHR, dihydrorhodamine; MDS, myelodysplastic
syn­drome; TCR, T-cell recep­tor; TREC, T-cell recep­tor exci­sion cir­cle.

under­ly­ing IEI. Notably, the hema­tol­o­gist can also raise con­cern With recent advance­ments in sequenc­ing meth­ods and acces­
for an under­ly­ing IEI by review of the periph­eral blood smear and, si­bil­ity, genetic test­ing is increas­ingly pro­moted as a key tool
if com­pleted, bone mar­row biopsy for signs sug­ges­tive of dis­ in the defin­i­tive diag­no­sis of IEI.5-7 A molec­u­lar diag­no­sis is valu­
rupted neu­tro­phil devel­op­ment/func­tion or mar­row stress/infil­ able for unequivocally establishing the IEI diag­no­sis, per­mit­ting
tra­tion (see Table 2). Functional lab­o­ra­tory eval­u­a­tion tai­lored appro­ pri­
ate genetic coun­ sel­ing to iden­ tify other at-risk fam­
to the patient pre­sen­ta­tion can pro­vide sup­port­ive evi­dence of ily mem­bers and future preg­nan­cies, facil­i­tat­ing geno­type/
an IEI. The Clinical Immunology Society main­tains a DLI Labo- phe­no­type cor­re­la­tions to inform patient-spe­cific clin­i­cal expec­
ratory Directory that is pub­licly acces­si­ble and can assist with ta­tions, and iden­ ti­
fy­
ing patients most likely to ben­ e­fit from
the iden­ti­fi­ca­tion of labs cur­rently pro­vid­ing Clinical Laboratory gene-spe­cific ther­a­pies.
Improvement Amendments-cer­ti­fied immune test­ing (Accessed
30 Sep­tem­ber 2021. https:​­/​­/clinimmsoc​­.org​­/CIS​­/Resources​­/DLI​ Pathophysiology of neutropenia in IEI
­-Lab​­-Directory​­.htm). A defin­i­tive diag­no­sis gen­er­ally requires the While the gen­eral mech­a­nisms for neutropenia in IEI have been
iden­ti­fi­ca­tion of path­o­genic var­i­ants via genetic sequenc­ing. pro­posed, ie, dimin­ished sur­vival, flawed devel­op­ment, phys­i­cal
Prakash Singh Shekhawat
Neutropenia and immune dysregulation  |  505
Table 2. Peripheral blood smear and bone mar­row find­ings in IEI asso­ci­ated with neutropenia

Hematopathologic find­ing IEI to con­sider in dif­fer­en­tial Additional com­ments


Absent or decreased neu­tro­phil gran­ules GFI1-related neutropenia In spe­cific gran­ule defi­ciency, neu­tro­phils with bilobed nuclei are
JAGN1-related neutropenia com­monly observed.
Specific gran­ule defi­ciency
Atypical mega­kar­yo­cytes GATA2 haploinsufficiency Megakaryocytes may be small or mono­nu­clear or may have sep­a­
rated nuclear lobes. Often asso­ci­ated with mar­row fibro­sis.21
Giant cyto­plas­mic gran­ules Chediak-Higashi syn­drome Giant azurophilic gran­ules within the lyso­somes are con­sid­ered
patho­gno­monic for the dis­or­der and are eas­ily vis­i­ble in neu­tro­
phils, eosin­o­phils, and other granulocytes.51
Additional find­ings in the mar­row include exten­sive vacuolization,
aci­do­philic inclu­sion (esp in promyelocytes), spe­cific gran­ules
eas­ily rec­og­nized as early as the mye­lo­cyte stage.9

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/504/1851486/504walkovich.pdf by guest on 13 December 2021


Myelofibrosis DADA215 DADA2 is also asso­ci­ated with lym­phoid aggre­gates in the mar­row.
GATA2 haploinsufficiency21
VPS45-related neutropenia52
Myelokathexis WHIM syn­drome23 Neutrophils are also often hypersegmented with thin, long isthmi,
G6PC3-related neutropenia53 pyknotic nuclei, and cyto­plas­mic vac­u­oles in WHIM syn­drome.
Neutrophil nuclei her­ni­a­tion WDR1 defi­ciency54 In addi­tion to her­ni­a­tion of the nuclear lobes, neu­tro­phils also have
agranular regions within the cyto­sol.
PHA NBAS defi­ciency Pseudo-PHA is asso­ci­ated with var­i­ous med­i­ca­tions (eg, immu­no­
sup­pres­sive agents, anti­mi­cro­bi­als, growth fac­tors). The pseudo-
PHA is gen­er­ally revers­ible with med­i­ca­tion ces­sa­tion.54 PHA also
seen in asso­ci­a­tion with MDS.
Promyelocyte arrest Severe, per­ma­nent arrest:55 Copper defi­ciency can also result in mat­u­ra­tion arrest; revers­ible
ELANE-related neutropenia (severe with cop­per admin­is­tra­tion.
con­gen­i­tal phe­no­type) Promyelocytic AML can mimic the promyelocytic arrest asso­ci­ated
HAX-1-related neutropenia with IEI but is eas­ily dif­fer­en­ti­ated by addi­tional flow and/or
CSF3R-related neutropenia cyto­ge­netic stud­ies.
G6PC3-related neutropenia
WAS-related neutropenia
JAGN1-related neutropenia56
Mild, inter­mit­tent arrest:
GFI1-related neutropenia
Neutropenia asso­ci­ated with
poikiloderma
AML, acute mye­loid leu­ke­mia; MDS, myelodysplastic syn­drome; PHA, Pelger-Huet anom­aly.

replace­ment, or fail­ure to egress from the mar­row,8 defin­i­tive smear clas­si­cally dem­on­strates giant azurophilic gran­ules within
mech­an ­ isms for neutropenia in the major­ity of IEI out­side of granulocyte lyso­somes. The treat­ment of neutropenia is pri­mar­ily
the con­gen­i­tal dis­or­ders of iso­lated neutropenia (eg, ELANE- sup­port­ive with anti­mi­cro­bi­als and granulocyte col­ony-stim­u­lat­ing
related neutropenia) have not been established. As such, this fac­tor (G-CSF), although hema­to­poi­etic stem cell trans­plant (HSCT)
arti­cle focuses on IEI with well-established asso­ci­a­tions with can resolve the hema­to­poi­etic aspects of the dis­or­der.
neutropenia accom­pa­nied by the addi­tional immune def­i­cits
most likely to be encoun­tered by hema­tol­o­gists (Table 3). Common var­i­able immu­no­de­fi­ciency
Common var­i­able immu­no­de­fi­ciency (CVID) is a het­ero­ge­neous
Chediak-Higashi syn­drome dis­or­der char­ac­ter­ized by impaired B-cell dif­fer­en­ti­a­tion and
Chediak-Higashi syn­drome is an auto­so­mal reces­sive dis­or­der defec­tive immu­no­glob­u­lin (Ig) pro­duc­tion. Autoimmune cytope­
sec­ond­ary to path­o­genic var­i­ants in CHS1/LYST. Patients clas­si­ nias are a com­mon clin­i­cal fea­ture of CVID, with immune throm­
cally pres­ent with oculocutaneous albi­nism, recur­rent pyo­genic bo­cy­to­pe­nia and auto­im­mune hemo­lytic ane­mia more prev­a­lent
infec­tions, bleed­ing diath­e­sis, and pro­gres­sive neu­ro­log­i­cal than auto­im­mune neutropenia.10 Cytopenias, includ­ing iso­lated
dys­func­tion and are at high risk of devel­op­ing hemophagocytic neutropenia, fre­quently pre­cede the diag­no­sis of CVID and her­
lymphohistiocytosis (HLH) sec­ond­ary to defec­tive lyso­somal traf­ ald a worse out­come.11,12 Autoimmune clear­ance sec­ond­ary to
fick­ing. Neutropenia is com­monly observed and is attrib­uted to self-reac­tive IgG to hema­to­poi­etic anti­gens is the pri­mary sus­
abnor­mal bone mar­row reserves and defec­tive granulocyte mobi­ pected eti­­ ol­
ogy of neutropenia, although hypersplenism and
li­za­tion from the mar­row space compounded by intramedullary drug-related (eg, sec­ond­ary to rituximab) and infec­tion-related
granulocyte destruc­tion.9 While not explic­itly stud­ied, it is likely that neutropenia is documented. Patients may be treated with cor­
infec­tion-related mar­row sup­pres­sion and/or accel­er­ated immune ti­co­ste­roids and/or G-CSF. Splenectomy and rituximab are not
clear­ance also con­trib­utes to the neutropenia. The periph­eral use­ful,11 but rapamycin is emerg­ing as an effec­tive ther­apy.13

506  |  Hematology 2021  |  ASH Education Program


Table 3. Characteristics of neutropenia in IEI com­monly encoun­tered by hema­tol­o­gists

Proposed mech­a­nism Neutropenia-directed


IEI Frequency of neutropenia of neutropenia ther­apy Comments
Chediak-Higashi syn­drome Common 57
-Accelerated granulocyte G-CSF -Phagocyte intra­cel­lu­lar
turn­over sec­ond­ Antimicrobials kill­ing and NK cyto­tox­ic­ity
ary to intramedullary HSCT58 is impaired.
granulocyte destruc­
tion compounded by
hypersplenism.9
-Abnormal bone mar­row
reserves with defec­tive
granulocyte mobi­li­za­
tion from the mar­row
space.9

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/504/1851486/504walkovich.pdf by guest on 13 December 2021


CVID 1%-8.4%10,12 -Autoimmune clear­ance as Corticosteroids -Neutropenia does not improve
supported by the iden­ti­ G-CSF with IgG replace­ment
fi­ca­tion of antineutrophil Rapamycin -Neutropenia is asso­ci­ated
antibodies and evi­dence with increased infec­
of hyper­plas­tic yet inef­ tions, increased poly­clonal
fi­cient ger­mi­nal cen­ter lymphoproliferation, and auto­
responses.11,59 im­mu­nity.12
-Hypersplenism -Increased mor­tal­ity rate asso­ci­
-Postinfectious ated with neutropenia in CVID
-Drug-related patients.11,12

DADA2 7%-15%14,60 -Decreased ADA2 pro­tein Corticosteroids -ADA2 is highly expressed in


func­tion in severely Rituximab mye­loid cells and pro­duced
del­e­te­ri­ous var­i­ants is Anti-TNF agents by acti­vated mac­ro­phages,
pro­posed to lead to HSCT61 mono­cytes, and den­dritic cells
decreased mar­row when stim­u­lated by an inflam­
pro­duc­tion.17 ma­tory response.14
-Immune-medi­ated
destruc­tion is also
hypoth­e­sized.
Hyper IgM syn­drome 41%18 -Decreased CD16 expres­ G-CSF -Neutropenia may be chronic
sion and dysregulated or inter­mit­tent.
transcriptome resulting in -Bone mar­row eval­u­a­tion may
impaired dif­fer­en­ti­a­tion.62 dem­on­strate mat­u­ra­tion
-Disrupted cyto­kine or arrest.62
growth fac­tor sup­port in
the bone mar­row.63
GATA2 haploinsufficiency 47%21 -Reduction of the prim­i­tive G-CSF -Mild chronic neutropenia may
HSC pool20 HSCT be the first man­i­fes­ta­tion of
dis­ease with other clin­i­cal fea­
tures, eg, monocytopenia or
MDS/AML presenting later.
-Maturation of neu­tro­phils in the
bone mar­row is gen­er­ally pre­
served.20
WHIM syn­drome Near uni­ver­sal -Diminished egress from G-CSF -Bone mar­row is hypercellular
the bone mar­row CXCR4 with full mat­u­ra­tion and clas­sic
sec­ond­ary to gain of antag­o­nists pyknotic nuclei.
func­tion muta­tions in
CXCR423
XLA 10%-26%64-66 -Decreased bone mar­row G-CSF -BTK expressed in mye­loid and
pre­cur­sors25 IVIG B-cell dif­fer­en­ti­a­tion (lim­ited
-Decreased mat­u­ra­tion to hema­to­poi­etic cells).
of mye­loid pre­cur­sors -Neutropenia may be a
sec­ond­ary to changes in presenting sign of XLA.
BTK-related sig­nal trans­ -Neutropenia gen­er­ally resolves
duc­tion25 with ini­ti­a­tion of IVIG, allowing
-Decreased cyto­ G-CSF with­drawal.
kine/chemokine pro­duc­ -Neutropenia gen­er­ally
tion from mono­cytes, documented only in con­junc­
esp decreased IL-1825 tion with an active infec­tion.
AML, acute mye­loid leu­ke­mia; MDS, myelodysplastic syn­drome.
Prakash Singh Shekhawat
Neutropenia and immune dysregulation  |  507
Deficiency of aden­o­sine deam­i­nase 2 dom­i­nant path­o­genic var­i­ants in CXCR4 that result in the abnor­
A defi­ ciency of aden­ o­sine deam­i­
nase 2 (DADA2) results from mal reten­tion of neu­tro­phils in the bone mar­row and sub­se­quent
homo­zy­gous or com­pound het­ero­zy­gous path­o­genic var­i­ants periph­eral neutropenia. Besides neutropenia, patients with WHIM
that result in a loss of func­tion of ADA2. Originally described as syn­drome often have quan­ti­ta­tive defects in mono­cytes, B cells,
an eti­­ol­ogy for mono­genic vas­cu­li­tis and early-onset stroke, the and CD4+ T cells. Patients are sus­ cep­
ti­
ble to sinopulmonary
phe­no­type is now rec­og­nized to be highly var­i­able and inclu­ infec­tions due to hypogammaglobulinemia and HPV-related dis­
sive of hema­to­logic and immune def­i­cits.14 Hematologic pre­sen­ ease, includ­ing warts and malig­nancy.23 The major­ity of WHIM
ta­tions include cytopenias—eg, pure red blood cell aplasia and patients will spon­ta­ne­ously release neu­tro­phils from the bone
bone mar­row fail­ure—spleno­meg­aly, and lymph­ade­nop­a­thy,15 mar­row dur­ing times of “stress,” eg, infec­tion, and do not require
with cytopenias often the first sign of dis­ease. Age of pre­sen­ta­ pro­phy­lac­tic G-CSF. However, for patients who suf­fer recur­rent
tion is also var­ia­ ble, with 24% of cases diag­nosed in infancy and infec­tions and/or fail to dem­on­strate an ade­quate release of
77% before the age of 10, but adult-onset symp­toms are increas­ neu­tro­phils from the bone mar­row dur­ing infec­tious chal­lenges,
ingly rec­og­nized.14,16 The mech­a­nism of neutropenia in DADA2 is G-CSF is recommended.24 CXCR4 antag­o­nists are dem­on­strat­ing

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/504/1851486/504walkovich.pdf by guest on 13 December 2021


unclear; how­ever, both immune-medi­ated destruc­tion and bone prom­ise as ther­ap­ eu­tic options to improve both neu­tro­phil and
mar­row fail­ure are suspected to con­trib­ute. Genotype-phe­no­type lym­pho­cyte counts.
cor­re­la­tions are emerg­ing, with more del­e­te­ri­ous var­i­ants result­
ing in hema­to­logic dis­ease with var­i­able response to anti-tumor
necro­sis fac­tor (TNF) agents.17 Corticosteroids, rituximab, and HSCT X-linked agam­ma­glob­u­lin­emia
have been uti­lized for the treat­ment of cytopenias. Bruton’s X-linked agam­ma­glob­u­lin­emia (XLA) is sec­ond­ary to
path­o­genic var­i­ants in the sig­nal trans­duc­tion gene BTK that
Hyper IgM syn­drome result in the near absence of CD19+ B cells and severe hypogam­
Hyper IgM syn­drome is a genetic het­ero­ge­neous group of dis­ maglobulinemia. Neutropenia is com­ mon and is most often
or­ders typ­i­fied by the inabil­ity to pro­duce class-switched immu­ iden­ti­
fied dur­ing peri­ ods of mar­ row “stress,” eg, dur­ing an
no­glob­u­lin. X-linked path­o­genic var­i­ants in CD40L are the most active infec­tion. The exact eti­­ol­ogy of neutropenia in XLA is
com­mon eti­­ol­ogy of hyper IgM syn­drome. Patients often pres­ uncer­tain; how­ever, given that Bruton’s tyro­sine kinase (BTK)
ent in early infancy or child­hood with recur­rent sinopulmonary is expressed in mye­loid and B-cell dif­fer­en­ti­a­tion, pro­posed
infec­tions and/or oppor­tu­nis­tic infec­tions such as Pneumocystis mech­a­nisms include decreased mat­u­ra­tion of mye­loid pre­cur­
jirovecii, his­to­plas­mo­sis, or cryp­to­spo­rid­ium. Neutropenia can sors sec­ond­ary to changes in BTK-related sig­nal trans­duc­tion
be chronic or inter­mit­tent in CD40L defi­ciency and is asso­ci­ated compounded by changes in mono­cyte IL-18 pro­duc­tion.25 Neu­
with an increased risk of oral and rec­tal ulcers, gin­gi­vi­tis, and tropenia can be the first clin­i­cal sign of XLA and is respon­sive
proctitis.18 Neutropenia can be the ini­tial clin­ic ­ al sign of hyper to G-CSF. Intravenous immu­no­glob­u­lin (IVIG) is the main­stay of
IgM syn­ drome and can mimic severe con­ gen­ it­al neutropenia ther­apy for patients with XLA, often allowing for the with­drawal
sec­ond­ ary to the pres­ ence of mat­ ur­a­
tion arrest in the bone of G-CSF.
mar­row. The exact mech­a­nism of neutropenia remains obscure,
although immune clear­ance, defec­tive mye­loid dif­fer­en­ti­a­tion,
and/or flawed bone mar­row egress have been hypoth­e­sized.19
Patients are gen­er­ally respon­sive to G-CSF. Of note, lym­pho­
mas and gas­tro­in­tes­ti­nal-related malig­nan­cies are com­mon in CLINICAL CASE 1
patients with hyper IgM syn­drome. An 8-month-old boy is admit­ted with severe neutropenia (ANC,
300/µL) and thrombocytosis (plate­let count, 650,000/µL) in
GATA2 haploinsufficiency the set­ting of fever and rash. The phys­i­cal exam is nota­ble for
GATA2 haploinsufficiency occurs sec­ond­ary to auto­so­mal dom­i­ exten­sive impe­tigo with­out lymph­ade­nop­a­thy and the absence
nant path­o­genic var­i­ants in GATA2, an early hema­to­poi­etic tran­ of ton­sil­lar tis­sue. A com­pre­hen­sive met­a­bolic panel reveals
scrip­ tion fac­tor active in mye­ loid devel­op­ ment. Neutropenia low total pro­tein with nor­mal albu­min. Flow cytometry for
occurs sec­ond­ary to neg­a­tive impacts on the hema­to­poi­etic B-cell enu­mer­a­tion dem­on­strates a near absence of CD19+ B
stem cell (HSC) pool.20 Patients with GATA2 haploinsufficiency cells. G-CSF and IVIG are ini­ti­ated. Genetic test­ing con­firms
have var­i­able clin­i­cal pre­sen­ta­tions inclu­sive of neutropenia; a path­o­genic var­i­ant in BTK con­sis­tent with XLA. Following
monocytopenia; B-cell, CD4+ T-cell and NK-cell lymphopenia; sev­eral IVIG infu­sions, his ANC nor­mal­izes, and the G-CSF is
myelodysplastic syn­drome; acute mye­loid leu­ke­mia; nontu­ with­drawn.
berculous myco­bac­te­rial infec­tions; viral infec­tions (espe­cially Author com­men­tary: Humoral deficiencies are often masked
var­i­cella, Epstein-Barr virus [EBV], and human pap­il­loma virus in early infancy due to the pla­cen­tal trans­fer of mater­nal IgG.
[HPV]); and/or lymphedema.21 Chronic neutropenia may be the A lack of local­ized lymph­ade­nop­a­thy despite an exten­sive skin
ini­tial sign of GATA2 haploinsufficiency, par­tic­u­larly in pedi­at­ric infec­tion, an absence of ton­sil­lar tis­sue, and a low total pro­tein
patients.22 G-CSF and con­ sid­
er­ation of HSCT are ther­ ap
­ eu­tic (sur­mised to be due to low immu­no­glob­u­lin, as albu­min is nor­
options to man­age neutropenia. mal) raise con­cern for a B-cell defect. Understanding that the
neutropenia in X-linked agam­ma­glob­u­lin­emia gen­er­ally occurs
Warts, hypogammaglobulinemia, infec­tions, myelokathexis dur­ing peri­ods of mar­row stress and is respon­sive to G-CSF is
syn­drome key to early man­age­ment. Additionally, aware­ness that the neu­
The clin­i­
cal tet­
rad of warts, hypogammaglobulinemia, infec­ tropenia in XLA resolves with IVIG ini­ti­at­ ion lends con­fi­dence to
tion, and myelokathexis (WHIM) syn­drome is due to auto­so­mal with­draw­ing the G-CSF post infec­tion res­o­lu­tion.

508  |  Hematology 2021  |  ASH Education Program


Patients with prior B-cell dis­or­ders includ­ing a his­tory of auto­
CLINICAL CASE 2 im­mu­nity31 or chronic lym­pho­cytic leu­ke­mia may be at increased
A 16-year-old-girl pres­ents with iso­lated mild neutropenia (ANC, risk of irAEs.28 There is uncer­tainty if PD-1/PD-L1 vs CTLA-4
1100/µL) inci­den­tally iden­ti­fied on a screen­ing sports phys­i­cal. She block­ade results in higher hema­to­logic irAEs, although the devel­
has no fam­ily his­tory of neutropenia, and her phys­i­cal exam is unre­ op­ment of HLH may be more com­mon with anti-CTLA-4.27,29,33 The
mark­able. Serial test­ing con­firms chronic neutropenia. Her bone devel­op­ment of autoreactive T cells and B cells, the increased
mar­row is mod­er­ately hypocellular, prompting next-gen­er­a­tion secre­tion of cyto­kine with the sub­se­quent infil­tra­tion of CD8+
genetic screen­ing, but no path­o­genic var­i­ants are iden­ti­fied. Sub­ T cells, and the immune-medi­ated dys­func­tion of HSC mat­u­ra­
sequently, she is admit­ted twice for dehy­dra­tion and cytopenias tion or pro­lif­er­a­tion have all­ been pos­tu­lated as gen­eral mech­a­
related to a sig­nif­i­cant bout of mono­nu­cle­o­sis. Post dis­charge, her nisms of neutropenia.26 It is prob­a­ble that dif­fer­ent mech­a­nisms
abso­lute lym­pho­cyte and abso­lute mono­cyte counts remain low. exist, based on bone mar­row biopsy find­ings dem­on­strat­ing that
Following dis­cus­sion with a genet­i­cist, addi­tional test­ing is sent to a minor­ity of patients have gran­u­lo­cytic hypo­pla­sia with lym­pho­
eval­u­ate for intronic var­i­ants in GATA2, and a path­o­genic var­i­ant is cytic infil­trate (a cen­tral or hypo­plas­tic type), while the major­ity

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/504/1851486/504walkovich.pdf by guest on 13 December 2021


iden­ti­fied con­sis­tent with GATA2 haploinsufficiency. of patients have nor­mal or hyper­plas­tic bone mar­row with or
Author com­men­tary: This case high­lights that many IEI have with­out the iden­ti­fi­ca­tion of a neu­tro­phil anti­body (periph­eral or
sub­tle pre­sen­ta­tions and clin­i­cal symp­toms that evolve over hyper­plas­tic type).31
time, neces­ si­
tat­ing the need for the hema­ tol­
o­gist to remain Patients on ICIs presenting with neutropenia should undergo
vig­i­lant and to con­sider gaps in test­ing, eg, the lim­i­ta­tions of bone mar­row eval­u­a­tion to dif­fer­en­ti­ate cen­tral vs periph­eral
genetic-sequenc­ ing tech­ niques such as with intronic regions neutropenia and to rule out under­ly­ing malig­nancy. A review of
or pseudogenes, in sus­pi­cious cases. The unusual require­ment other med­i­ca­tions and screen­ing for infec­tious causes of neutro­
for mul­ti­ple admis­sions for a pri­mary EBV infec­tion should raise penia are also man­da­tory. Examination of the periph­eral blood
con­cern for immune dys­func­tion. In GATA2 haploinsufficiency, and bone mar­ row for large gran­ u­
lar lym­ pho­ cytes should be
severe EBV infec­tions are fre­quently encoun­tered as den­dritic under­taken and immunophenotyped for large gran­u­lar lym­pho­
cells, which are reduced or absent in GATA2 haploinsufficiency, cyte counts greater than 0.5/µL.27 The pres­ence of high fever
are required for EBV anti­gen pre­sen­ta­tion to T cells. Early diag­ should alert to the pos­si­bil­ity of HLH and pan­cy­to­pe­nia to aplas­
no­sis of GATA2 haploinsufficiency in this patient greatly affects tic ane­mia, 2 diag­noses with poor treat­ment responses and out­
her sup­port­ive care plan, with rec­om­men­da­tions for G-CSF and comes.27,33 Therapy for neutropenia is not spe­cif­i­cally addressed
other pre­ven­ta­tive mea­sures, eg, HPV vac­ci­na­tion and malig­ by the Amer­i­can Society of Clinical Oncology or the Euro­pean
nancy sur­veil­lance. Additionally, early diag­no­sis affords her the Society for Medical Oncology,34,35 but gen­eral lit­er­a­ture rec­
oppor­tu­nity to con­sider cura­tive ther­apy with HSCT. om­ men­ da­tions are to closely mon­ i­
tor patients with grade 1
neutropenia.31 For grade 2 tox­ic­ity, the ICI should be with­held
and low-dose cor­ti­co­ste­roids ini­ti­ated if no recov­ery occurs in
Pathophysiology of neutropenia sec­ond­ary 1 week. For grade 3 to 4 tox­ic­ity, ICIs should be discontinued and
to immu­no­ther­apy high-dose cor­ti­co­ste­roids admin­is­tered with taper­ing over 4 to
The role of the immune sys­tem in tumor sur­veil­lance has long been 6 weeks for responding patients.26,27 G-CSF should be pre­scribed
appre­ci­ated, but only recently have sci­en­tists and cli­ni­cians har­ to has­ten recov­ery for all­grade 2 to 4 toxicities. IVIG is likely
nessed the power of T cells to treat can­cer. Two mod­ern immune safe and may be more effec­tive for the periph­eral destruc­tion of
ther­a­pies include the use of genet­i­cally engineered T cells pro­ neu­tro­phils,31 but there is no con­sen­sus in the lit­er­a­ture regard­
grammed to attack malig­nant cells, eg, chi­me­ric anti­gen recep­tor ing whether it should be used as up-front ther­apy or as sal­vage
(CAR) T cells, and block­ade of sig­nals that drive T-cell exhaus­tion for cor­ti­co­ste­roid non­re­spond­ers. For patients with hypo­plas­tic/
in the tumor micro­en­vi­ron­ment. The over­all tox­ic­ity from immu­no­ cen­tral neutropenia, cyclo­spor­ine may be trialed if there is a
ther­apy is less com­pared to con­ven­tional che­mo­ther­apy, but the poor response to cor­ti­co­ste­roids,31 and antithymocyte glob­u­lin
devel­op­ment of immune-related adverse events (irAEs), includ­ is an option for aplastic ane­mia.36 Most patients (approx­i­ma­tely
ing immune-related neutropenia, can result in severe tox­ic­ity. The 75%-85%) have improve­ment or nor­mal­i­za­tion of the neu­tro­phil
under­stand­ing of the immu­no­logic mech­a­nisms under­pin­ning count, with res­o­lu­tion more likely in patients with a hyper­plas­
immune-related neutropenia is incom­plete, but our knowl­edge of tic/periph­eral type,31,32 but reinstitution of the ICI fre­ quently
neutropenia driven by IEI may pro­vide help­ful clues to uncover the results in neutropenia recur­rence.28,31
path­o­phys­i­­ol­ogy and sug­gest poten­tial treat­ments (see Table 4).
CAR T cells
Immune check­point inhib­i­tors CAR T cells are genet­i­cally engineered to express a mod­i­fied
Immune check­point inhib­i­tors (ICIs) include agents that inhibit T-cell recep­tor with costimulatory domains that tar­get com­mon
CTLA-4, programmed cell death 1 pro­tein (PD-1), or programmed can­cer anti­gens, such as CD19 and B-cell mat­u­ra­tion anti­gen.
cell death 1 ligand 1 (PD-L1) sig­nal­ing and are inte­gral to the CAR T cells have dem­on­strated activ­ity against a range of B-cell
treat­ment of many can­cers.26 The reported inci­dence of all­-grade malig­nan­cies, but effi­cacy has been lim­ited by cyto­kine release
hema­to­logic irAEs is 0.4% to 8.3% with high-grade (grade 3-5) at syn­drome (CRS) and neuroinflammation. High lev­els of IL-1, IL-6,
0% to 1.7%.27-29 High-grade neutropenia in iso­la­tion or in con­cert inter­feron gamma, monocyte chemo-attractant protein-1, and
with addi­tional cytopenias is reported at a fre­quency of 0.2% to granulocyte-mac­ro­phage (GM) CSF largely pro­duced by mono­
0.94%.28,30 Neutropenia is almost always severe with ANCs near cytes and mac­ro­phages drive this inflam­ma­tory tox­ic­ity.37,38
zero and can result in severe infec­tion and death. 27,28
The median Cytopenias are the most com­mon grade 3 or greater irAE, with
time to onset is 8.5 to 10.5 weeks after ICI ini­ti­a­tion, but neutro­ neutropenia being the most com­mon defi­ciency.39 CAR stud­ies
penia can occur at any point dur­ing Prakash
ICI treat­ment. Singh
27,31,32 Shekhawat
have dem­on­strated an inci­dence of grade 3 to 4 neutropenia in

Neutropenia and immune dysregulation  |  509


Table 4. Neutropenia sec­ond­ary to ICIs and CAR T-cells with cor­re­la­tion to known immune dis­or­ders

Treatment of Correlate to known Treatment of


Treatment irAE Proposed mech­a­nism cytopenias in irAE immune dis­or­ders cytopenias in IEI
ICI Hypoplastic T-cell infil­tra­tion of the bone Corticosteroids Immune-medi­ated Cyclosporine
(PD-1/PD-L1 and neutropenia/aplastic mar­row with sub­se­quent Cyclosporine aplastic ane­mia67 ATG
CTLA-4 block­ ane­mia destruc­tion of hema­to­poi­ ATG
ade) etic pre­cur­sors26 G-CSF
Immune-medi­ated dys­func­ Corticosteroids HSC dys­func­tion in ADA2 TNF inhi­bi­tion15
tion of HSC mat­u­ra­tion and G-CSF defi­ciency69
pro­lif­er­a­tion68
Hyperplastic/periph­ Peripheral destruc­tion of Corticosteroids Fas/Fas ligand defi­ Corticosteroids
eral neutropenia neu­tro­phils medi­ated by IVIG ciency and devel­op­ Rapamycin
autoantibodies31 G-CSF ment of auto­im­mune Mycophenolate

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/504/1851486/504walkovich.pdf by guest on 13 December 2021


cytopenias70 Mofetil
HLH T-cell and mac­ro­phage Corticosteroids HLH and MAS dis­or­ders Corticosteroids
acti­va­tion, per­haps from Etoposide with nor­mal phago­ Etoposide
immune cell tumor inva­sion cytic and cyto­toxic IFNG anti­body
and malig­nant cell apo­ func­tion; malig­nancy- IL-1 anti­body
pto­sis71 asso­ci­ated HLH72 IL-6R anti­body
Jak inhib­i­tors
ICI Multiple irAE, includ­ing Regulatory T-cell inhi­bi­tion Corticosteroids CTLA-4 haploinsufficiency Corticosteroids
CTLA-4 block­ade neutropenia and/or deple­tion73,74 (to sup­press LRBA defi­ciency CTLA-4-Ig
expanded effec­ IPEX syn­drome (FOXP3 Rapamycin
tor T cells) defi­ciency) Calcineurin
G-CSF CD25 defi­ciency inhib­i­tors
CAR-T Early neutropenia Lymphodepleting ther­apy IL-6R anti­body HLH and MAS syn­dromes Corticosteroids
and CRS75 Corticosteroids with nor­mal phago­ Etoposide
cytic and cyto­toxic IFNG anti­body
func­tion IL-1 anti­body
IL-6R anti­body
Jak inhib­i­tors
Late neutropenia Disrupted CXCL12 bone G-CSF WHIM syn­drome (CXCR4- G-CSF
mar­row gra­di­ent inhibiting GOF) CXCR4 inhi­bi­tion
neu­tro­phil egress46 Late-onset neutropenia
sec­ond­ary to rituximab48
ATG, antithymocyte glob­u­lin; GOF, gain-of-func­tion; IFNG, inter­feron gamma; Ig, immu­no­glob­u­lin; IPEX, immunodysregulation, polyendocrinopathy,
and enter­op­a­thy, X-linked; MAS, mac­ro­phage acti­va­tion syn­drome.

60% to 85% of patients and “late neutropenia” occur­ring 28 days infu­sion.45 Both hypocellularity and nor­mal cel­lu­lar­ity have been
or later in 3% to 53%.40-44 reported, per­haps indi­cat­ing dif­fer­ing mech­a­nisms of neutro­
Early neutropenia has been attrib­uted to immunodepleting penia.45,46 G-CSF may has­ten neu­tro­phil recov­ery but should be
ther­apy with fludarabine and cyclo­phos­pha­mide preinfusion and avoided early (within 3 weeks) of infu­sion given the poten­tial for
hema­to­poi­etic sup­pres­sion from CRS.45 However, late neutrope­ height­ened CRS. Data to sup­port this con­cern include higher lev­
nia, which can per­sist well after che­mo­ther­apy and res­o­lu­tion of els of murine G-CSF in mice cor­re­lat­ing with CRS sever­ity,49 ele­
CRS, is more enig­matic. Possible expla­na­tions for late neutrope­ vated lev­els of G-CSF and GM-CSF in patients with neu­ro­tox­ic­ity,50
nia include poor bone mar­row reserves and exces­sive inflam­ma­ and 1 small study dem­on­strat­ing that G-CSF use prox­i­mal to CAR
tory dam­age given the asso­ci­a­tion with prior HCT/mul­ti­ple lines T-cell infu­sion was asso­ci­ated with higher CRS sever­ity.37 Eltrom­
of ther­apy and higher-grade CRS,45-47 respec­tively, in patients bopag has been used with suc­cess in patients with aplasia, and
with prolonged hema­to­logic tox­ic­ity. Interestingly, a sub­set of autol­o­gous stem cell infu­sion should be con­sid­ered for very late
patients with late neutropenia show an ini­tial recov­ery of neu­ neutropenia (greater than 3 months) if cryopreserved cells are
tro­phils only to be followed by a sec­ond trough with­out any avail­­able.45
inter­ven­ing ther­apy.45,46 One group eval­ua ­ ted serum CXCL12 lev­
els and reported a cor­re­la­tion in patients with late neutropenia,
hypoth­e­siz­ing that the con­sump­tion of CXCL12 from an expand­
ing pre­cur­sor B-cell pop­u­la­tion may dis­rupt the nor­mal CXCL12
bone mar­row gra­di­ent required for nor­mal neu­tro­phil egress, CLINICAL CASE 3
resulting in a recur­rence of periph­eral neutropenia in some CD19 A 65-year-old woman with met­as­ tatic mel­a­noma is treated with
CAR T-cell recip­i­ents.46 A sim­i­lar mech­a­nism has been pro­posed sin­gle-agent ipilimumab (anti-CTLA-4). Two weeks fol­low­ing
for late-onset neutropenia fol­low­ing rituximab ther­apy.48 her third cycle of ther­apy, she pres­ents with fever and an ANC
Guidance for the eval­ ua
­t­ion and treat­ ment of neutropenia of 80/µL. A bone mar­row biopsy dem­on­strates increased mye­
fol­low­ing CAR T cells is cur­rently lacking, but many sug­gest a loid cells with­out a lym­pho­cytic infil­trate or mat­u­ra­tion block.
bone mar­row biopsy for neutropenia persisting 1 month fol­low­ing Additional cycles of ipilimumab are tem­ po­
rar­
ily discontinued,

510  |  Hematology 2021  |  ASH Education Program


and she is admin­is­tered cor­ti­co­ste­roids, G-CSF, and IVIG, with a of immune dysregulation. While commonly utilized in these rare
ris­ing ANC within 4 days of treat­ment ini­ti­a­tion. Her ste­roids are diseases, these conditions are off-label uses from the formal FDA
tapered over 4 weeks with a con­tin­ued nor­mal ANC. Her oncol­o­ label.
gist tri­als another cycle of ipilimumab, but her neutropenia recurs. James A. Connelly: therapy with rituximab, rapamycin, CXCR4 in­
Author com­men­tary: Neutropenia as an irAE is a rare but seri­ hibition, cyclosporine, ATG, MMG, JAK inhibitors, IL-1 antibodies,
ous com­pli­ca­tion of ICIs. The loss of CTLA-4 through iat­ro­genic IL-6R antibodies, and CTLA-4-Ig are briefly discussed as options
block­ade or genetic haploinsufficiency results in the loss of reg­ for treating immunodysregulation from IEI or iatrogenic etiolo­
u­la­tory T-cell sup­pres­sive activ­ity and immune dysregulation. gies of immune dysregulation. While commonly utilized in these
Subsequent neutropenia can develop through sev­eral pro­posed rare diseases, these conditions are off-label uses from the formal
mech­a­nisms, includ­ing T-cell medi­ated attack on the bone mar­ FDA label.
row, auto­an­ti­body for­ma­tion, hypercytokinemia, and immune-
medi­ ated HSC dys­ func­tion. In this case, mye­ loid hyper­ pla­
sia Correspondence
with­out CD8+ lym­pho­cy­to­sis in the bone mar­row would favor Kelly Walkovich, Department of Pediatrics, University of Mich­

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/504/1851486/504walkovich.pdf by guest on 13 December 2021


periph­eral destruc­tion by antineutrophil antibodies, a mech­a­nism igan, 1540 E Medical Center Dr, Ann Arbor, MI 48109; e-mail:
more likely to respond over­all and to IVIG treat­ment. Although kwalkovi@med​­.umich​­.edu.
most patients with immune-related neutropenia recover with
treat­ment, recur­rence with reinitiation of the ICI occurs in the References
major­ity of patients and should only be trialed if deemed essen­ 1. Silvestre-Roig C, Hidalgo A, Soehnlein O. Neutrophil het­ero­ge­ne­ity: impli­
tial for oncol­ogy care and with care­ful com­plete blood counts ca­tions for homeo­sta­sis and path­o­gen­e­sis. Blood. 2016;127(18):2173-2181.
with plate­lets and dif­fer­en­tial mon­i­tor­ing. 2. Michniacki TF, Connelly JA, Sturza J, et al. Neutropenia is an underrecog­
nized find­ing in pedi­at­ric pri­mary immu­no­de­fi­ciency dis­eases: an anal­y­sis
of the United States immu­no­de­fi­ciency net­work reg­is­try. J Pediatr Hema-
tol Oncol. 2020;42(7):e601-e605.
Conclusion 3. Seidel MG. Autoimmune and other cytopenias in pri­ mary immunodefi­
As an inte­grated com­po­nent of the immune sys­tem, the neu­tro­ ciencies: pathomechanisms, novel dif­fer­en­tial diag­noses, and treat­ment.
phil func­tions as a strong com­pan­ion with other immune cells to Blood. 2014;124(15):2337-2344.
pro­tect the body under nor­mal phys­i­­ol­ogy. However, with dis­ 4. Derpoorter C, Bordon V, Laureys G, Haerynck F, Lammens T. Genes at the
cross­road of pri­mary immunodeficiencies and can­cer. Front Immunol.2018;
rup­ tion of tolerogenic path­ ways through genetic alter­ ation or
9(Novem­ber):2544.
iat­ro­genic inhib­i­tors, the neu­tro­phil can quickly turn from friend 5. Yska HAF, Elsink K, Kuijpers TW, Frederix GWJ, van Gijn ME, van Montfrans
to foe to unre­strained immune cells. Early and cor­rect iden­ti­fi­ca­ JM. Diagnostic yield of next gen­er­a­tion sequenc­ing in genet­i­cally undi­ag­
tion of immune dysregulation presenting with or com­pli­cated by nosed patients with pri­mary immunodeficiencies: a sys­tem­atic review. J
neutropenia is crit­i­cal to improv­ing patient out­comes related to Clin Immunol. 2019;39(6):577-591.
6. Heimall JR, Hagin D, Hajjar J, et al. Use of genetic test­ing for pri­mary immu­
selecting vastly expanding targeted ther­a­peu­tic options, min­i­miz­
no­de­fi­ciency patients. J Clin Immunol. 2018;38(3):320-329.
ing infec­tions through the early ini­ti­a­tion of anti­mi­cro­bial pro­phy­ 7. Bonilla FA, Khan DA, Ballas ZK, et al; Joint Task Force on Practice Parame­
laxis and vac­ci­na­tion strat­e­gies, mon­i­tor­ing for malig­nan­cies, and ters, representing the Amer­i­can Academy of Allergy, Asthma and Immunol­
lim­it­ing end organ dam­age. Establishing an IEI diag­no­sis is also ogy, the Amer­i­can College of Allergy, Asthma and Immunology, and the
nec­es­sary for future preg­nancy coun­sel­ing and HSCT donor eval­u­ Joint Council of Allergy, Asthma and Immunology. Practice param­e­ter for
a­tions. While many of the mech­a­nisms of neutropenia in IEI remain the diag­no­sis and man­age­ment of pri­mary immu­no­de­fi­ciency. J Allergy
Clin Immunol. 2015;136(5):1186-1205.e1.
incom­pletely under­stood, the com­mon themes of disrupted neu­
8. Sullivan KE. Neutropenia as a sign of immu­ no­de­
fi­
ciency. J Allergy Clin
tro­phil pro­duc­tion resulting in decreased mar­row reserves, ham­ Immunol. 2019;143(1):96-100.
pered mobi­li­za­tion from the mar­row, and exces­sive destruc­tion 9. Blume RS, Bennett JM, Yan­kee RA, Wolff SM. Defective granulocyte reg­u­
are appar­ent. Leveraging fur­ther mech­a­nis­tic eval­u­a­tion into la­tion in the Chediak-Higashi syn­drome. N Engl J Med. 1968;279(19):1009-
IEI-related neutropenia may also illu­mi­nate the path­o­phys­i­­ol­ogy 1015.
10. Feuille EJ, Anooshiravani N, Sullivan KE, Fuleihan RL, Cunningham-Rundles
of neutropenia as a con­se­quence of malig­nancy immu­no­ther­apy.
C. Autoimmune cytopenias and asso­ci­ated con­di­tions in CVID: a report
Through such research, the neu­tro­phil can hope­fully be protected from the USIDNET reg­is­try. J Clin Immunol. 2018;38(1):28-34.
from the con­se­quences of induced immune acti­va­tion with­out 11. Guffroy A, Mourot-Cottet R, Gérard L, et al; DEFI Study Group. Neutropenia
sac­ri­fic­ing the T-cell onslaught on malig­nant cells. in patients with com­mon var­i­able immu­no­de­fi­ciency: a rare event asso­ci­
ated with severe out­come. J Clin Immunol. 2017;37(7):715-726.
12. Ghorbani M, Fekrvand S, Shahkarami S, et al. The eval­u­a­tion of neutropenia
Conflict-of-inter­est dis­clo­sure in com­mon var­i­able immune defi­ciency patients. Expert Rev Clin Immunol.
Kelly Walkovich: con­ sul­tancy: Swed­ ish Orphan Biovitrum AB; 2019;15(11):1225-1233.
advi­sory board mem­ ber: Horizon Therapeutics, AstraZeneca, 13. Bride KL, Vincent T, Smith-Whitley K, et  al. Sirolimus is effec­ tive in
Pharming; local prin­ci­pal inves­ti­ga­tor: mavorixafor clin­i­cal trial relapsed/refrac­ tory auto­ im­
mune cytopenias: results of a pro­ spec­tive
multi-insti­tu­tional trial. Blood. 2016;127(1):17-28.
spon­sored by X4 Pharmaceuticals.
14. Meyts I, Aksentijevich I. Deficiency of aden­o­sine deam­i­nase 2 (DADA2):
James A. Connelly: advi­sory board mem­ber: X4 Pharmaceuticals, updates on the phe­no­type, genet­ics, path­o­gen­e­sis, and treat­ment. J Clin
Horizon Therapeutics. Immunol. 2018;38(5):569-578.
15. Michniacki TF, Hannibal M, Ross CW, et  al. Hematologic man­i­fes­ta­tions
Off-label drug use of defi­ciency of aden­o­sine deam­i­nase 2 (DADA2) and response to tumor
necro­sis fac­tor inhi­bi­tion in DADA2-asso­ci­ated bone mar­row fail­ure. J Clin
Kelly Walkovich: therapy with rituximab, rapamycin, CXCR4 inhi­
Immunol. 2018;38(2):166-173.
bition, cyclosporine, ATG, MMG, JAK inhibitors, IL-1 antibodies, 16. Sharma A, Naidu G, Sharma V, et al. Deficiency of aden­o­sine deam­i­nase 2 in
IL-6R antibodies, and CTLA-4-Ig are briefly discussed as options adults and chil­dren: expe­ri­ence from India. Arthritis Rheumatol. 2021;73(2):
Prakash
for treating immunodysregulation from Singhetiologies
IEI or iatrogenic Shekhawat 276-285.

Neutropenia and immune dysregulation  |  511


17. Lee PY, Kellner ES, Huang Y, et al. Genotype and func­tional cor­re­lates of 42. Raje N, Berdeja J, Lin Y, et  al. Anti-BCMA CAR T-cell ther­apy bb2121 in
dis­ease phe­no­type in defi­ciency of aden­o­sine deam­i­nase 2 (DADA2). J relapsed or refrac­tory mul­ti­ple mye­loma. N Engl J Med. 2019;380(18):1726-
Allergy Clin Immunol. 2020;145(6):1664-1672.e101672e10. 1737.
18. Leven EA, Maffucci P, Ochs HD, et al. Hyper IgM syn­drome: a report from 43. Schuster SJ, Bishop MR, Tam CS, et al; JULIET Investigators. Tisagenlecleu­
the USIDNET reg­is­try. J Clin Immunol. 2016;36(5):490-501. cel in adult relapsed or refrac­tory dif­fuse large B-cell lym­phoma. N Engl J
19. Rezaei N, Moazzami K, Aghamohammadi A, Klein C. Neutropenia and pri­ Med. 2019;380(1):45-56.
mary immu­no­de­fi­ciency dis­eases. Int Rev Immunol. 2009;28(5):335-366. 44. Maude SL, Laetsch TW, Buechner J, et al. Tisagenlecleucel in chil­dren and
20. Rodrigues NP, Janzen V, Forkert R, et al. Haploinsufficiency of GATA-2 per­ young adults with B-cell lym­pho­blas­tic leu­ke­mia. N Engl J Med. 2018;378
turbs adult hema­to­poi­etic stem-cell homeo­sta­sis. Blood. 2005;106(2): ­(5):439-448.
477-484. 45. Nagle SJ, Murphree C, Raess PW, et  al. Prolonged hema­to­logic tox­ic­ity
21. Spinner MA, Sanchez LA, Hsu AP, et al. GATA2 defi­ciency: a pro­tean dis­ fol­low­ing treat­ment with chi­me­ric anti­gen recep­tor T cells in patients with
or­der of hema­to­poi­e­sis, lym­phat­ics, and immu­nity. Blood. 2014;123(6): hema­to­logic malig­nan­cies. Am J Hematol. 2021;96(4):455-461.
809-821. 46. Fried S, Avigdor A, Bielorai B, et al. Early and late hema­to­logic tox­ic­ity fol­
22. Pasquet M, Bellanné-Chantelot C, Tavitian S, et  al. High fre­ quency of low­ing CD19 CAR-T cells. Bone Marrow Transplant. 2019;54(10):1643-1650.
GATA2 muta­tions in patients with mild chronic neutropenia evolv­ing to 47. Chakraborty R, Hill BT, Majeed A, Majhail NS. Late effects after chi­me­ric
MonoMac syn­drome, myelodysplasia, and acute mye­loid leu­ke­mia. Blood. anti­gen recep­tor T cell ther­apy for lym­phoid malig­nan­cies. Transplant Cell

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/504/1851486/504walkovich.pdf by guest on 13 December 2021


2013;121(5):822-829. Ther. 2021;27(3):222-229.
23. Heusinkveld LE, Majumdar S, Gao JL, McDermott DH, Mur­phy PM. WHIM 48. Dunleavy K, Hakim F, Kim HK, et  al. B-cell recov­ery fol­low­ing rituximab-
syn­drome: from path­o­gen­e­sis towards per­son­al­ized med­i­cine and cure. J based ther­apy is asso­ci­ated with per­tur­ba­tions in stro­mal derived fac­tor-1 and
Clin Immunol. 2019;39(6):532-556. granulocyte homeo­sta­sis. Blood. 2005;106(3):795-802.
24. Badolato R, Donadieu J; WHIM Research Group. How I treat warts, 49. Giavridis T, van der Stegen SJC, Eyquem J, Hamieh M, Piersigilli A, Sadelain
hypogammaglobulinemia, infec­tions, and myelokathexis syn­drome. Blood. M. CAR T cell-induced cyto­kine release syn­drome is medi­ated by mac­ro­
2017;130(23):2491-2498. phages and abated by IL-1 block­ade. Nat Med. 2018;24(6):731-738.
25. Jacobs ZD, Guajardo JR, Anderson KM. XLA-asso­ci­ated neutropenia treat­ 50. Santomasso BD, Park JH, Salloum D, et al. Clinical and bio­log­i­cal cor­re­lates
ment: a case report and review of the lit­er­a­ture. J Pediatr Hematol Oncol. of neu­ro­tox­ic­ity asso­ci­ated with CAR T-cell ther­apy in patients with B-cell
2008;30(8):631-634. acute lym­pho­blas­tic leu­ke­mia. Cancer Discov. 2018;8(8):958-971.
26. Omar NE, El-Fass KA, Abushouk AI, et  al. Diagnosis and man­age­ment of 51. Kjeldsen L, Calafat J, Borregaard N. Giant gran­ules of neu­tro­phils in
hema­to­log­i­cal adverse events induced by immune check­point inhib­i­tors: a Chediak-Higashi syn­drome are derived from azurophil gran­ules but not
sys­tem­atic review. Front Immunol. 2020;11(21 Octo­ber):1354. from spe­cific and gelatinase gran­ules. J Leukoc Biol. 1998;64(1):72-77.
27. Michot JM, Lazarovici J, Tieu A, et  al. Haematological immune-related 52. Vilboux T, Lev A, Malicdan MC, et al. A con­gen­i­tal neu­tro­phil defect syn­
adverse events with immune check­point inhib­i­tors, how to man­age? Eur J drome asso­ci­ated with muta­tions in VPS45. N Engl J Med. 2013;369(1):
Cancer. 2019;122(Novem­ber):72-90. 54-65.
28. Delanoy N, Michot JM, Comont T, et  al. Haematological immune-related 53. McDermott DH, De Ravin SS, Jun HS, et al. Severe con­gen­i­tal neutropenia
adverse events induced by anti-PD-1 or anti-PD-L1 immu­ no­ther­
apy: a resulting from G6PC3 defi­ciency with increased neu­tro­phil CXCR4 expres­
descrip­tive obser­va­tional study. Lancet Haematol. 2019;6(1):e48-e57. sion and myelokathexis. Blood. 2010;116(15):2793-2802.
29. Kramer R, Zaremba A, Moreira A, et  al. Hematological immune related 54. Kuhns DB, Fink DL, Choi U, et al. Cytoskeletal abnor­mal­i­ties and neu­tro­phil
adverse events after treat­ment with immune check­point inhib­i­tors. Eur J dys­func­tion in WDR1 defi­ciency. Blood. 2016;128(17):2135-2143.
Cancer. 2021;147(April):170-181. 55. Donadieu J, Fenneteau O, Beaupain B, Mahlaoui N, Chantelot CB. Congen­
30. Petrelli F, Ardito R, Borgonovo K, et  al. Haematological toxicities with ital neutropenia: diag­ no­sis, molec­
ul­ar bases and patient man­ age­ ment.
immu­no­ther­apy in patients with can­cer: a sys­tem­atic review and meta- Orphanet J Rare Dis. 2011;6(May):26.
anal­y­sis. Eur J Cancer. 2018;103(Novem­ber):7-16. 56. Boztug K, Järvinen PM, Salzer E, et  al. JAGN1 defi­ ciency causes aber­
31. Finkel I, Sternschuss M, Wollner M, et al; Israel Lung Cancer Group. Immune- rant mye­loid cell homeo­sta­sis and con­gen­i­tal neutropenia. Nat Genet.
related neutropenia fol­low­ing treat­ment with immune check­point inhib­i­tors. 2014;46(9):1021-1027.
J Immunother. 2020;43(2):67-74. 57. Introne W, Boissy RE, Gahl WA. Clinical, molec­ul­ar, and cell bio­log­i­cal
32. Boegeholz J, Brueggen CS, Pauli C, et al. Challenges in diag­no­sis and man­ aspects of Chediak-Higashi syn­drome. Mol Genet Metab. 1999;68(2):
age­ment of neutropenia upon expo­sure to immune-check­point inhib­i­tors: 283-303.
meta-anal­y­sis of a rare immune-related adverse side effect. BMC Cancer. 58. Lozano ML, Rivera J, Sánchez-Guiu I, Vicente V. Towards the targeted man­age­
2020;20(1):300. ment of Chediak-Higashi syn­drome. Orphanet J Rare Dis. 2014;9(August):132.
33. Davis EJ, Salem JE, Young A, et al. Hematologic com­pli­ca­tions of immune 59. Romberg N, Le Coz C, Glauzy S, et  al. Patients with com­mon var­i­able
check­point inhib­i­tors. Oncologist. 2019;24(5):584-588. immu­no­de­fi­ciency with auto­im­mune cytopenias exhibit hyper­plas­tic yet
34. Brahmer JR, Lacchetti C, Thompson JA. Management of immune-related inef­fi­cient ger­mi­nal cen­ter responses. J Allergy Clin Immunol. 2019;143(1):
adverse events in patients treated with immune check­point inhib­i­tor 258-265.
ther­apy: Amer­i­can Society of Clinical Oncology Clinical Practice Guide­ 60. Pinto B, Deo P, Sharma S, Syal A, Sharma A. Expanding spec­trum of DADA2:
line Summary. J Oncol Pract. 2018;14(4):247-249. a review of phe­no­types, genet­ics, path­o­gen­e­sis and treat­ment. Clin Rheu-
35. Haanen JBAG, Carbonnel F, Robert C, et al. Management of toxicities from matol. 2021;40(10):3883-3896.
immu­no­ther­apy: ESMO Clinical Practice Guidelines for diag­no­sis, treat­ 61. Hashem H, Kumar AR, Müller I, et al; Deficiency of Adenosine Deaminase
ment and fol­low-up. Ann Oncol. 2017;28(suppl 1):iv119-iv142. Type 2 Foundation. Hematopoietic stem cell trans­plan­ta­tion res­cues the
36. Zhuang J, Du J, Guo X, et al. Clinical diag­no­sis and treat­ment rec­om­men­ hema­to­log­i­cal, immu­no­log­i­cal, and vas­cu­lar phe­no­type in DADA2. Blood.
da­tions for immune check­point inhib­i­tor-related hema­to­log­i­cal adverse 2017;130(24):2682-2688.
events. Thorac Cancer. 2020;11(3):799-804. 62. Cabral-Marques O, França TT, Al-Sbiei A, et  al. CD40 ligand defi­ciency
37. Gaut D, Tang K, Sim MS, Duong T, Young P, Sasine J. Filgrastim asso­ci­a­tions causes func­tional defects of periph­eral neu­tro­phils that are improved by
with CAR T-cell ther­apy. Int J Cancer. 2021;148(5):1192-1196. exog­e­nous IFN-γ. J Allergy Clin Immunol. 2018;142(5):1571-1588.e91588e9.
38. Sterner RM, Sakemura R, Cox MJ, et al. GM-CSF inhi­bi­tion reduces cyto­kine 63. Solanilla A, Déchanet J, El Andaloussi A, et  al. CD40-ligand stim­ u­
lates
release syn­drome and neuroinflammation but enhances CAR-T cell func­tion myelopoiesis by reg­u­lat­ing flt3-ligand and thrombopoietin pro­duc­tion in
in xeno­grafts. Blood. 2019;133(7):697-709. bone mar­row stro­mal cells. Blood. 2000;95(12):3758-3764.
39. Yáñez L, Sánchez-Escamilla M, Perales MA. CAR T cell tox­ic­ity: cur­rent man­ 64. Ochs HD, Smith CI. X-linked agam­ma­glob­u­lin­emia. A clin­i­cal and molec­u­lar
age­ment and future direc­tions. Hemasphere. 2019;3(2):e186. anal­y­sis. Medicine (Baltimore). 1996;75(6):287-299.
40. Neelapu SS, Locke FL, Bartlett NL, et  al. Axicabtagene ciloleucel CAR 65. Farrar JE, Rohrer J, Conley ME. Neutropenia in X-linked agam­ma­glob­u­lin­
T-cell ther­apy in refrac­tory large B-cell lym­phoma. N Engl J Med. 2017;377 emia. Clin Immunol Immunopathol. 1996;81(3):271-276.
­(26):2531-2544. 66. Winkelstein JA, Marino MC, Lederman HM, et  al. X-linked agam­ma­glob­u­
41. Abramson JS, Palomba ML, Gordon LI, et  al. Lisocabtagene maraleucel lin­emia: report on a United States reg­is­try of 201 patients. Medicine (Balti-
for patients with relapsed or refrac­tory large B-cell lym­pho­mas (TRAN­ more). 2006;85(4):193-202.
SCEND NHL 001): a multicentre seam­less design study. Lancet. 2020;396 67. Wang L, Liu H. Pathogenesis of aplastic ane­mia. Hematology. 2019;24(1):
­(10254):839-852. 559-566.

512  |  Hematology 2021  |  ASH Education Program


68. Inadomi K, Kumagai H, Arita S, et  al. Bi-cytopenia pos­si­bly induced by 73. Sharma A, Subudhi SK, Blando J, et al. Anti-CTLA-4 immu­no­ther­apy does
anti-PD-1 anti­body for pri­
mary malig­ nant mel­
a­noma of the esoph­ a­
gus: not deplete FOXP3. Clin Cancer Res. 2019;25(4):1233-1238.
a case report. Medicine (Baltimore). 2016;95(29):e4283. 74. Simpson TR, Li F, Montalvo-Ortiz W, et  al. Fc-depen­ dent deple­ tion of
69. Lee PY. Vasculopathy, immu­no­de­fi­ciency, and bone mar­row fail­ure: the tumor-infil­trat­ing reg­u­la­tory T cells co-defi­nes the effi­cacy of anti-CTLA-4
intrigu­ing syn­
drome caused by defi­ ciency of aden­ o­
sine deam­ i­
nase 2. ther­apy against mel­a­noma. J Exp Med. 2013;210(9):1695-1710.
Front Pediatr. 2018;6(18 Octo­ber):282. 75. Neelapu SS, Tummala S, Kebriaei P, et al. Chimeric anti­gen recep­tor T-cell
70. Rieux-Laucat F, Magérus-Chatinet A, Neven B. The auto­im­mune lymphop­ ther­apy—assess­ment and man­age­ment of toxicities. Nat Rev Clin Oncol.
roliferative syn­drome with defec­tive FAS or FAS-ligand func­tions. J Clin 2018;15(1):47-62.
Immunol. 2018;38(5):558-568.
71. Hantel A, Gabster B, Cheng JX, Golomb H, Gajewski TF. Severe hemophago­
cytic lymphohistiocytosis in a mel­a­noma patient treated with ipilimumab +
nivolumab. J Immunother Cancer. 2018;6(1):73.
72. Daver N, McClain K, Allen CE, et  al. A con­sen­sus review on malig­nancy-
asso­ci­ated hemophagocytic lymphohistiocytosis in adults. Cancer. 2017; © 2021 by The Amer­i­can Society of Hematology
123(17):3229-3240. DOI 10.1182/hema­tol­ogy.2021000285

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/504/1851486/504walkovich.pdf by guest on 13 December 2021

Prakash Singh Shekhawat


Neutropenia and immune dysregulation  |  513
NEUTROPENIA: DOING MORE WITH LESS

Impaired myelopoiesis in congenital neutropenia:


insights into clonal and malignant hematopoiesis

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/514/1851470/514warren.pdf by guest on 13 December 2021


Julia T. Warren1 and Daniel C. Link2
Division of Hematology-Oncology, Department of Pediatrics, and 2Division of Oncology, Department of Internal Medicine, Washington University
1

School of Medicine, St. Louis, MO

A common feature of both congenital and acquired forms of bone marrow failure is an increased risk of developing acute
myeloid leukemia (AML) or myelodysplastic syndrome (MDS). Indeed, the development of MDS or AML is now the major
cause of mortality in patients with congenital neutropenia. Thus, there is a pressing clinical need to develop better strat-
egies to prevent, diagnose early, and treat MDS/AML in patients with congenital neutropenia and other bone marrow
failure syndromes. Here, we discuss recent data characterizing clonal hematopoiesis and progression to myeloid malig-
nancy in congenital neutropenia, focusing on severe congenital neutropenia (SCN) and Shwachman-Diamond syndrome.
We summarize recent studies showing excellent outcomes after allogenic hematopoietic stem cell transplantation for
many (but not all) patients with congenital neutropenia, including patients with SCN with active myeloid malignancy
who underwent transplantation. Finally, we discuss how these new data inform the current clinical management of
patients with congenital neutropenia.

LEARNING OBJECTIVES
• Understand the indications for and interpretation of somatic gene pane sequencing and karyotyping in congenital
neutropenia
• Integrate recent improvements in hematopoietic stem cell transplantation outcome in congenital neutropenia
into clinical management
• Appreciate the contribution of stressors in the development of clonal hematopoiesis and myeloid malignancy in
congenital neutropenia

Introduction
There has been a recent increase in the use of somatic gene CLINICAL CASE 1
panel sequencing in patients with congenital neutropenia This patient presented in early childhood with recurrent
to identify patients at risk of leukemic transformation. We infections and an absolute neutrophil count (ANC) of <200
present three cases of congenital neutropenia in which cells/mm3. Bone marrow biopsy specimen showed mye-
results of molecular testing raise important clinical man- loid maturation arrest, and genetic analysis revealed a het-
agement questions. To answer these questions, we review erozygous germline pathogenic variant in ELANE (p.S126L).
recent advances on the following topics: (1) the frequency Serial surveillance bone marrow biopsy specimens dem-
and molecular features of myeloid malignancy in congenital onstrated no overt clonal abnormalities as detected by
neutropenia, (2) clonal hematopoiesis in congenital neutro- karyotype and fluorescence in situ hybridization. However,
penia and mechanisms of clonal evolution to myeloid malig- somatic sequencing of a surveillance bone marrow sample
nancy, and (3) recent studies of hematopoietic stem cell obtained at age 45 years showed a high variant allele fre-
transplantation (HSCT) outcomes in congenital neutropenia. quency (41%) truncating pathogenic variant in CSF3R. Lower
Finally, we explore how these new data inform the clinical frequency clones in DNMT3A (1.7%) and ASXL1 (2.6%) also
management of congenital neutropenia, including the case were detected.
presentations.

514 | Hematology 2021 | ASH Education Program


of SCN is asso­ci­ated with dis­tinct molec­u­lar fea­tures com­pared
CLINICAL CASE 2 with de novo MDS/AML.10 Most nota­bly, somatic trun­cat­ing var­
This patient presented at age 2 years with neutropenia, fail­ure i­ants in CSF3R and mis­sense var­i­ants in RUNX1 are observed in
to thrive with chronic diar­rhea, mild devel­op­men­tal delay, and ~80% and 63% of cases, respec­tively.11 In addi­tion, sec­ond­ary
pel­vic dysostosis. The ANC was 320 cells/mm3, hemo­glo­bin MDS/AML in patients with SCN is fre­ quently asso­ ci­
ated with
was 11.7 g/dL, and plate­lets were 132 000 cells/mm3. Initial bone mono­somy 7 and tri­somy 21.12-14
mar­row biopsy performed at age 2 years dem­on­strated hypo- SDS is an inherited bone mar­row fail­ure syn­drome char­ac­ter­
cellularity with a decrease in mature mye­loid cells and nor­mal ized by a con­stel­la­tion of extra-hema­to­poi­etic man­i­fes­ta­tions
cyto­ge­net­ics. Sequencing revealed biallelic germline path­o­genic with high var­i­abil­ity (exo­crine pan­cre­atic insuf­fi­ciency, hepa-
var­i­ants in SBDS (c. 183_184TA _CT and c. 258 + 2T>C), confirming topathy, bony abnor­mal­i­ties, poor growth, and other endo­crine
the diag­ no­sis of Shwachman-Diamond syn­ drome (SDS). Over man­i­fes­ta­tions).15,16 Neutropenia is pres­ent in approx­i­ma­tely 90%
time, the patient devel­oped mod­er­ate ane­mia with mac­ro­cy­to­sis of cases of SDS, with ane­mia, throm­bo­cy­to­pe­nia, and pan­cy­to­
and con­tin­ued with inter­mit­tent mild to mod­er­ate neutropenia. pe­nia observed less fre­quently. SDS is caused, in most cases,

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/514/1851470/514warren.pdf by guest on 13 December 2021


Repeat bone mar­row exam­i­na­tion revealed mild dysmorphology by biallelic germline var­i­ants in SBDS.17 The esti­ma­tes of risk for
pre­dom­i­nantly in the mye­loid lin­e­ages with­out dys­pla­sia and hema­to­poi­etic malig­nancy vary from 12% to 20% depending in
mega­kar­yo­cyte hypo­pla­sia. Serial somatic gene panel sequenc­ part on the age of the cohort.8,18,19 MDS/AML that arises in the set­
ing over a 5-year time span iden­ti­fied mul­ti­ple het­ero­zy­gous low- ting of SDS is char­ac­ter­ized by biallelic TP53 var­i­ants and com­
abun­dance TP53 path­o­genic mis­sense var­i­ants. plex chro­mo­somal abnor­mal­i­ties.8,20,21

Clonal hema­to­poi­e­sis in con­gen­i­tal neutropenia


During hema­to­poi­e­sis, the acqui­si­tion of somatic var­i­ants over
CLINICAL CASE 3 time leads to the devel­op­ment of a genet­i­cally het­ero­ge­neous
blood cell pop­u­la­tion. Some of these var­i­ants con­fer a fit­ness
The patient presented in infancy with omphalitis and was noted
advan­tage, lead­ing to the expan­sion of hema­to­poi­etic stem/
to have severe neutropenia with an ANC of <200 cells/mm3.
pro­gen­i­tor cells (HSPCs) car­ry­ing cer­tain var­i­ants over time in
Before ini­ti­a­tion of granulocyte col­ony-stim­u­lat­ing fac­tor
a pro­cess known as clonal hema­to­poi­e­sis.22,23 Variants in genes
(G-CSF), the patient expe­ ri­
enced recur­ rent sinopulmonary
impli­cated in mye­ loid malig­ nancy are the most fre­ quent in
infec­tions and cel­lu­li­tis. Germline sequenc­ing revealed a path­
age-related clonal hema­to­poi­e­sis (ARCH), includ­ing in the epi­ge­
o­genic ELANE var­i­ant (p.V65F). Somatic gene panel sequenc­ing
netic mod­i­fi­ers DNTM3A, TET2, and ASXL1. Of note, in oth­er­wise
obtained at age 7 years did not dem­on­strate any severe con­
healthy per­sons, the pres­ence of clonal hema­to­poi­e­sis con­fers an
gen­i­tal neutropenia (SCN)–asso­ci­ated or other clonal hema­to­
increased risk of devel­op­ing hema­to­poi­etic malig­nancy, depend-
poi­e­sis var­i­ants, and serial bone mar­row exam­i­na­tions did not
ing on the gene involved, clone size, and whether mul­ti­ple var­i­
reveal any dys­pla­sia or chro­mo­somal abnor­mal­i­ties. Although
ants are pres­ent.24
infec­tious com­pli­ca­tions ini­tially improved while on G-CSF ther­
Recent stud­ ies dem­ on­strate that the fre­ quency of clonal
apy, the patient even­ tu­
ally expe­ ri­
enced severe neutropenia
hema­to­poi­e­sis in patients with con­gen­i­tal neutropenia is mark­
again with con­com­i­tant recur­rent cel­lu­li­tis despite esca­lat­ing
edly increased. We showed that in both SCN and SDS, the inci­
doses of G-CSF (>10 µg/kg/dose).
dence of clonal hema­to­poi­e­sis was mark­edly increased com­pared
with age-matched healthy donors.25 However, dis­ tinct genes
were respon­si­ble for this increase. In SCN, the increase in clonal
Myeloid malig­nancy in con­gen­i­tal neutropenia hema­to­poi­e­sis is entirely due to trun­cat­ing var­i­ants in CSF3R;
SCN is a rare syn­drome char­ac­ter­ized by chronic neutropenia no increase in clonal hema­to­poi­e­sis due to other genes com­
pres­ent from birth and recur­ring bac­te­rial infec­tions.1 Pathogenic monly mutated in clonal hema­to­poi­e­sis (eg, DNMT3A, TET2, and
var­i­ants of ELANE are the most com­mon cause of SCN, account­ ASXL1) was observed (Figure 1). In con­trast, in SDS, the increase
ing for approx­i­ma­tely 50% of cases, with var­i­ants in CLPB, HAX1, in clonal hema­to­poi­e­sis is due to a marked increase in TP53 var­i­
and G6PC3 account­ing for an addi­tional 10% to 20% of cases ants, with nearly 50% of patients car­ry­ing 1 or more TP53 var­i­ants.
depending on geo­graphic loca­tion (eg, HAX1 var­i­ants have not Of note, TP53 var­i­ants were not seen in patients with SCN or in
been observed in North America).2-5 Treatment with G-CSF is age-matched healthy donors. Conversely, CSF3R var­i­ants were
the stan­dard of care for SCN; it increases the level of cir­cu­lat­ not seen in patients with SDS or age-matched healthy donors.
ing neu­tro­phils in most patients and reduces infec­tion-related What accounts for the strik­ing dif­fer­ence in clonal hema­to­
mor­tal­ity.6 The devel­op­ment of mye­loid malig­nancy is now the poi­e­sis between per­sons with SCN and SDS? Current evi­dence
major cause of mor­tal­ity in SCN. Indeed, after 15 years of G-CSF points to dif­fer­ences in cel­lu­lar stress­ors in healthy per­sons and
ther­apy, the cumu­la­tive inci­dence of myelodysplastic syn­drome per­sons with SCN or SDS. The CSF3R var­i­ants in patients with
(MDS) or acute mye­loid leu­ke­mia (AML) in patients with SCN was SCN typ­i­cally pro­duce a trun­cated G-CSF recep­tor, which trans­
esti­mated at 22%.7 In this study, trans­for­ma­tion to MDS/AML was mits a sustained, dysregulated sig­nal in response to G-CSF.26-28
asso­ci­ated with a higher G-CSF dose (>8 µg/kg/d) and with a Studies in mice show that expres­ sion of a trun­ cated G-CSF
reduced neu­tro­phil response to G-CSF. Similar data from the recep­ tor con­ fers a com­ pet­i­
tive advan­ tage to HSPCs that is
French Neutropenia Registry reported an 11% cumu­la­tive inci­ depen­dent on chronic G-CSF treat­ment.29 Thus, the high level
dence of MDS/AML by age 20 years.8 Of note, almost all­genetic of G-CSF pres­ent in patients with SCN (either through endog­
sub­types of SCN have been asso­ci­ated with an increased risk e­nous pro­duc­tion or phar­ma­co­logic admin­is­tra­tion) may drive
for trans­for­ma­tion to MDS/AML.9 MDS/AML aris­ing in the set­ting the expan­sion of HSPCs car­ry­ing trun­ca­tion CSF3R var­i­ants
Prakash Singh Shekhawat
Clonal evo­lu­tion in con­gen­i­tal neutropenia  |  515
Figure 1. Graphical representation of the observed clonal hematopoiesis rates by somatic genetic alteration within healthy age-
matched controls, patients with SCN, or patients with SDS.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/514/1851470/514warren.pdf by guest on 13 December 2021


(Figure 2). Interestingly, a recent study exam­ined clonal hema­ fer­ence is due to the sever­ity of neutropenia or sec­ond­ary to
to­poi­e­
sis in 185 patients with chronic idi­ o­
pathic neutropenia cell-intrin­sic changes related to germline var­i­ants in SCN will
(also termed idi­o­pathic neutropenia of unde­ter­mined sig­nif­i­ require fur­ther study.
cance–neutropenia).30 Clonal hema­to­poi­e­sis was iden­ti­fied in Germline var­i­ants in SBDS that are pres­ent in most cases of
21 of 185 (11.4%) patients, which is sim­i­lar to that expected for age- SDS result in impaired ribo­some bio­gen­e­sis, which in turn induces
matched con­trols. The most fre­quent somatic var­i­ants observed p53 expres­sion and growth arrest.31-33 Pathogenic somatic var­
were in DNMT3A and TET2, as observed in ARCH. In sharp con­ i­ants in TP53 in HSPCs are predicted to atten­u­ate this growth
trast to SCN, somatic CSF3R var­ i­
ants were not detected in arrest, resulting in their selec­tive expan­sion in patients with SDS.
patients with chronic idi­o­pathic neutropenia. Whether this dif­ An ele­gant study by Kennedy et al20 pro­vi­des fur­ther sup­port for

Figure 2. Modes of evolution from clonal hematopoiesis to malignant transformation. In the presence of high exogenous or endoge-
nous G-CSF, subclones with truncating CSF3R variants have a competitive advantage and expand over time. With continued pressure
from excessive G-CSF signaling, cooperating RUNX1 variants (*) can contribute to malignant transformation. In SDS, ribosome biogen-
esis stress selects for subclones that have adapted through acquisition of EIF6 or TP53 somatic variants. Chronic ribosome biogenesis
stress can lead to either continued adaptive clonal hematopoiesis or, in TP53 subclones clones that have acquired a second TP53
variant on the other allele, malignant transformation.

516  |  Hematology 2021  |  ASH Education Program


the key role of impaired ribo­some bio­gen­e­sis in the devel­op­ Table 1. Overall sur­vival fol­low­ing HSCT with or with­out
ment of clonal hema­to­poi­e­sis in patients with SDS. Specifically, in malig­nancy in mod­ern-era tri­als
addi­tion to TP53 var­i­ants, patients with SDS fre­quently develop
inactivating var­i­ants of EIF6. These var­i­ants par­tially res­cue the Post-HSCT over­all sur­vival Without malig­nancy With malig­nancy
ribo­some bio­gen­e­sis defect in SDS, thereby reduc­ing p53 acti­ SCN
va­tion. Together, these obser­va­tions illus­trate the impor­tance  *EBMT39 87% (n = 73) 79% (n = 14)
of cel­lu­lar stress­ors in the devel­op­ment of clonal hema­to­poi­e­sis
  *SCNIR (Euro­pean)40 78% (n = 27) 83% (n = 24)
(and likely mye­loid malig­nancy). In SCN, ele­vated lev­els of G-CSF
drive the expan­sion of HSPCs car­ry­ing trun­ca­tion CSF3R var­i­ants, SDS
  ^CIBMTR42 72% (n = 39) 15% (n = 13)
and in SDS, impaired ribo­some bio­gen­e­sis selects for HSPCs car­
ry­ing TP53 var­i­ants or var­i­ants (ie, EIF6) that reduce p53 acti­va­   ^SAAWP-EBMT41 71% (n = 61) 29% (n = 13)
tion (Figure 2).  *SDSR43 — 46% (n = 15)
*Three-year overall survival; ^five-year overall survival.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/514/1851470/514warren.pdf by guest on 13 December 2021


Clonal evo­lu­tion to mye­loid malig­nancy in con­gen­i­tal CIBMTR, Center for International Blood and Marrow Transplant Research;
neutropenia EBMT, Euro­pean Society for Bone Marrow Transplantation; SAAWP-EMBT,
The pres­ence of clonal hema­to­poi­e­sis in patients with con­gen­i­ Severe Aplastic Anemia Working Party of the EBMT; SCNIR, Severe Chronic
tal neutropenia does not inev­i­ta­bly lead to mye­loid malig­nancy. Neutropenia International Registry; SDSR, Shwachman-Diamond Syndrome
Registry of North America.
Although the pres­ence of trun­cat­ing CSF3R var­i­ants is asso­ci­ated
with the devel­op­ment of mye­loid malig­nancy in patients with
SCN, it is not suf­fi­cient. Indeed, lon­gi­tu­di­nal sequenc­ing stud­ies tain but sug­gests that SCN leu­ke­mic stem cells have reduced
show that CSF3R-mutant clonal hema­to­poi­e­sis can per­sist for fit­ness and/or increased sus­cep­ti­bil­ity to allo­genic immune cell
many years with­out pro­gres­sion, even in cases with large CSF3R- clear­ance.
mutant clones.34 There is evi­dence that somatic loss-of-func­tion HSCT for patients with SDS is gen­er­ally performed for bone
var­i­ants of RUNX1 may be an early pro­gres­sion event, with acqui­ mar­row fail­ure or mye­loid malig­nancy. HSCT out­comes are infe­
si­tion of chro­mo­somal abnor­mal­i­ties (mono­somy 7 and tri­somy 21) rior to that that seen with patients with SCN and remains espe­
late events.11 Consistent with this con­clu­sion, Dannenmann et al35 cially dis­mal for those patients who have already devel­oped a
used SCN patient-derived induced plu­ rip­o­
tent stem cells that mye­loid malig­nancy41-43 (Table 1). The rea­ sons for the infe­
rior
expressed mutant CSF3R with our with­out mutant RUNX1 to show out­comes are likely mul­ti­fac­to­rial, includ­ing impaired bone mar­
that these var­i­ants coop­er­ate to block gran­u­lo­cytic dif­fer­en­ti­a­ row stro­mal cell func­tion, nonhematopoietic organ tox­ic­ity, and,
tion, pro­duc­ing an AML-like phe­no­type. most impor­tant, the intrin­sic resis­tance of TP53 mutant mye­loid
Variants in TP53 are pres­ent in nearly 50% of patients with SDS malig­nan­cies to ther­apy. Together, these data sug­gest that an
and may per­sist for many years with­out pro­gres­sion.20,25 Thus, effec­tive method to iden­tify patients with SDS at high risk of
hav­ing a TP53 var­i­ant within the hema­to­poi­etic com­part­ment is pro­gres­sion to mye­loid malig­nancy for early HSCT is warranted.
clearly not suf­fi­cient to drive leu­ke­mo­gen­e­sis. We sug­gest that However, this must be bal­anced by the like­li­hood of increased
con­tin­ued ribo­some bio­gen­e­sis stress in SDS HSPCs car­ry­ing a trans­plant-related mor­bid­ity and mor­tal­ity in patients with SDS
het­ero­zy­gous path­o­genic TP53 var­i­ant selects for clones that who have under­ly­ing extra-hema­to­poi­etic organ dys­func­tion.
inac­ti­vate the sec­ond TP53 allele (Figure 2). In con­trast, var­i­ants
in EIF6, by reduc­ing ribo­some bio­gen­e­sis stress, may be pro­tec­ Timing of HSCT in SCN
tive. Consistent with this model, biallelic TP53 var­i­ants, but not There are a num­ber of clin­i­cal and molec­u­lar data to con­sider
EIF6 var­i­ants, are very com­mon in the malig­nant clone in patients regard­ing tim­ing of trans­plant for patients with SCN, includ­ing
with SDS who develop mye­loid malig­nancy.20 (1) response to G-CSF, (2) results of sur­veil­lance bone mar­row
stud­ies to iden­tify dys­pla­sia or chro­mo­somal abnor­mal­i­ties, and
HSCT in con­gen­i­tal neutropenia (3) somatic var­i­ant detec­tion. Prior stud­ies have dem­on­strated
Because the devel­op­ment of mye­loid malig­nan­cies is the major that G-CSF dose and neu­tro­phil response are pre­dic­tors of pro­
cause of death in con­gen­i­tal neutropenia, a key ques­tion in the gres­sion to mye­loid malig­nancy.7,19 Based on these data, the
clin­i­cal man­age­ment is when to offer HSCT. French Severe Neutropenia Registry recommended that trans­
Current indi­ca­tions for HSCT in patients with SCN include plant be offered to all­patients with SCN requir­ing high-dose
G-CSF refrac­to­ri­ness or devel­op­ment of MDS/AML, with some G-CSF (>15 µg/kg/d). Since insti­tut­ing this pol­icy in 2005, none
inves­­ti­ga­tors also con­sid­er­ing high-dose G-CSF require­ment of the patients in this cohort have devel­oped a mye­loid malig­
(>15 µg/kg/d)36 or cer­tain high-risk ELANE path­o­genic var­i­ants nancy.36 Given the excel­lent HSCT out­comes for patients with
(p.G214R, p.C151Y, and p.C223ter).37,38 Studies of out­ comes in SCN who have mye­loid malig­nancy, a wait-and-see approach for
patients with SCN after HSCT are con­founded by the dif­fer­ent such patients also is rea­son­able. Of note, an inad­e­quate neu­
eras of trans­plant, con­di­tion­ing reg­i­mens, and donor sources. tro­phil response (ANC <1000 cells/mm3) to G-CSF, despite high
Nonetheless, recent reports39,40 dem­on­strate that out­comes are G-CSF dose, is a strong indi­ca­tion for HSCT.
good for patients who undergo trans­plan­ta­tion with or with­out It is com­mon prac­tice to per­form annual bone mar­row biopsy
mye­loid malig­nancy (Table 1). Also high­lighted in these stud­ies is eval­u­a­tions on patients with SCN to iden­tify chro­mo­somal abnor­
the remark­ably low relapse rate after HSCT in patients with SCN mal­i­ties and/or dys­pla­sia as early signs of malig­nant trans­for­ma­
who have mye­loid malig­nancy, espe­cially con­sid­er­ing that many tion.44-46 Historically, this prac­tice was based on expert opin­ion
patients with SCN have active mye­loid malig­nancy at the time rather than strong clin­i­cal or exper­i­men­tal evi­dence. However,
of trans­plant. The rea­son for the very low relapse rate is uncer­ bone mar­row biopsy is an inva­sive and expen­sive pro­ce­dure, and
Prakash Singh Shekhawat
Clonal evo­lu­tion in con­gen­i­tal neutropenia  |  517
it also car­ries asso­ci­ated risks of seda­tion in youn­ger patients. show that a subclone car­ry­ing biallelic TP53 var­i­ants was detect­
In our opin­ion, in light of recent data show­ing good out­comes able 4 years prior to malig­nant trans­for­ma­tion, expanded over
for patients with SCN who have mye­loid malig­nancy, the value time, and was pres­ent in the AML clone. However, sin­gle-cell
of annual sur­veil­lance bone mar­rows is debat­able. That said, a sequenc­ing is expen­sive, labor inten­sive, and not yet clin­i­cally
bone mar­row biopsy for patients with SCN with wors­en­ing blood avail­­able, lim­it­ing its cur­rent use to research appli­ca­tions.
counts or decreased respon­sive­ness to G-CSF is warranted.
There has been a recent increased use of gene panel sequenc­
ing to iden­tify somatic var­ia ­ nts in patients with SCN. An advan­ CLINICAL CASES (Con­tin­ued)
tage of this approach is the abil­ity to ana­lyze blood sam­ples, Case 1
obvi­at­ing the needed for an inva­sive bone mar­row biopsy. On This patient with ELANE-mutated SCN was noted to have a high-
the other hand, the test is expen­sive and insur­ance cov­er­age fre­quency CSF3R path­o­genic var­i­ant and low-fre­quency DNTM3A
var­i­able. Interpreting the sequenc­ing results also is chal­leng­ing. and ASXL1 var­i­ants at age 45 years. As we have discussed, the
Truncating CSF3R var­i­ants in SCN are com­mon and may per­sist pre­dic­tive value of CSF3R var­i­ants for leu­ke­mic pro­gres­sion is

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/514/1851470/514warren.pdf by guest on 13 December 2021


for years or even dis­ap­pear with leu­ke­mic pro­gres­sion.34 Thus, low. Likewise, the sig­nif­i­cance of the low-fre­quency DNTM3A or
the value of trun­cat­ing CSF3R var­i­ants to iden­tify patients at risk ASXL1 var­i­ants in this older patient is unclear. We favor con­tin­ued
for pro­gres­sion is lim­ited. The pre­dic­tive value of RUNX1 var­i­ants obser­va­tion of this patient, reserv­ing HSCT for the devel­op­ment
appears more prom­ is­
ing. As noted pre­ vi­
ously, the fre­
quency of MDS/AML. We would not rec­om­mend repeat somatic gene
of RUNX1 var­i­ants occur­ring in the set­ting of SCN-asso­ci­ated panel sequenc­ing out­side of a research study. Ultimately, this
MDS/AML is high (68%),11 and these var­ia ­ nts are often detect­ patient devel­oped AML at age 56 years, which was suc­cess­fully
able in the blood or bone mar­row months prior to pro­gres­sion. treated by bone mar­row trans­plan­ta­tion.
However, in con­trast to CSF3R var­i­ants, var­i­ants in RUNX1 appear
to be rare in patients with SCN with­out mye­loid malig­nancy. In Case 2
our study of clonal hema­to­poi­e­sis in 40 patients with SCN with­ This patient with SDS presented with mul­ti­ple low-abun­dance
out mye­loid malig­nancy, none had detect­able RUNX1 var­i­ants.25 TP53 var­i­ants by somatic gene panel sequenc­ing but ini­tially
Thus, RUNX1 var­i­ants may be pre­dic­tive of leu­ke­mic pro­gres­sion had no dys­pla­sia or chro­mo­somal abnor­mal­ity. The pre­dic­tive
in patients with SCN, although pro­spec­tive data supporting this value for leu­ke­mic pro­gres­sion of TP53 detected by bulk somatic
con­clu­sion are needed. Altogether, con­sid­er­ing the good out­ DNA sequenc­ing is likely low. It is also unknown whether, sim­
comes for patients with SCN who undergo trans­plan­ta­tion with i­lar to patients with ARCH, the pres­ence of mul­ti­ple var­i­ants
malig­nancy, cur­rent evi­dence does not sup­port the rou­tine use may con­fer a higher risk of trans­for­ma­tion. We would favor con­
of sur­ veil­lance somatic gene panel sequenc­ ing (out­
side of a tin­ued obser­ va­
tion in this patient with annual bone mar­ row
research study) to strat­ify patients for HSCT. eval­u­a­tion for dys­pla­sia and chro­mo­somal abnor­mal­i­ties. The
value of repeat bulk somatic gene panel sequenc­ing to mon­i­
Timing of HSCT in SDS tor TP53-mutant clone size is uncer­tain and not recommended
One approach for early detec­ tion of clonal pro­ gres­
sion in out­side of a research study. This patient devel­oped AML at age
patients with SDS is annual bone mar­row sur­veil­lance. Abnormal 22 years, and the malig­nant clone dem­on­strated biallelic TP53
find­ings on cyto­ge­netic anal­y­sis are pres­ent in approx­i­ma­tely var­i­ants detect­able 4 years before trans­for­ma­tion. Although a
50% of patients with SDS, the most com­mon being iso­chro­mo­ pro­spec­tive study is required, this case high­lights the poten­tial
some 7 and del(20q).47 Isochromosome 7 results in an extra copy for sin­gle-cell sequenc­ing to detect early biallelic TP53 events
of the SBDS gene, which, although typ­i­cally car­ry­ing a splice and serve as a bio­marker for con­sid­er­ing early HSCT.
site var­i­ant, is capa­ble of pro­duc­ing a small amount of full-length
SBDS pro­tein.48 del(20q) deletes 1 copy of EIF6, which, as dis- Case 3
cussed ear­lier, atten­u­ates the defec­tive ribo­some bio­gen­e­sis in This 12-year-old patient with ELANE-mutant SCN was noted to
SDS.49 Perhaps not sur­pris­ingly, iso­chro­mo­some 7 and del(20q) have decreased G-CSF respon­ sive­
ness and recur­ rent cel­lu­
li­
are asso­ci­ated with a lower risk of pro­gres­sion to mye­loid malig­ tis. Although results of cyto­ge­netic and somatic gene panel
nancy. In con­trast, mono­somy 7 or del(7q) is asso­ci­ated with sequenc­ing were neg­a­tive, we would favor early HSCT in this
rapid pro­gres­sion to mye­loid malig­nancy and is an indi­ca­tion for patient. Ultimately, the patient under­went matched unre­lated
trans­plant.50 donor hema­to­poi­etic stem cell trans­plant at age 13 years and
The role for somatic gene panel sequenc­ing in SDS is uncer­ over 10 years later is alive and well with­out trans­plant-related
tain. Clonal hema­to­poi­e­sis due to TP53 var­i­ants is pres­ent in com­pli­ca­tions.
approx­ i­
ma­ tely 50% of patients with SDS and increases with
age.25 The pres­ence of TP53-mutant clonal hema­to­poi­e­sis is not
asso­ci­ated with more severe cytopenias, and its pre­dic­tive value Summary and future direc­tions
for leu­ke­mic pro­gres­sion is likely to be low. Indeed, TP53 var­i­ Recent advances in our under­stand­ing of dis­ease path­o­gen­e­sis
ants can per­sist for sev­eral years with a sta­ble clone size with­out and clonal evo­lu­tion have affected the clin­i­cal man­age­ment of
pro­gres­sion to mye­loid malig­nancy. Kennedy et al20 showed that patients with con­gen­i­tal neutropenia. Cellular stress­ors play a
most mye­loid malig­nan­cies aris­ing in patients with SDS carry key role in clonal evo­lu­tion and depend, in part, on the under­ly­
biallelic TP53 var­i­ants, rais­ing the pos­si­bil­ity that detec­tion of ing genetic eti­­ol­ogy of con­gen­i­tal neutropenia. Thus, iden­ti­fy­ing
hema­to­poi­etic clones car­ry­ing biallelic TP53 var­i­ants may be the genetic cause of neutropenia is desir­able, and we rec­om­
more pre­dic­tive of leu­ke­mic pro­gres­sion. Indeed, the authors mend panel-based test­ing rather than sin­gle-gene test­ing as part
used sin­gle-cell sequenc­ing in an infor­ma­tive patient with SDS to of the diag­nos­tic eval­u­a­tion. An effec­tive sur­veil­lance strat­egy

518  |  Hematology 2021  |  ASH Education Program


to iden­tify patients at risk for trans­for­ma­tion to mye­loid malig­ 12. Freedman MH, Bonilla MA, Fier C, et  al. Myelodysplasia syn­drome and
nancy is needed, par­tic­u­larly for patients with SDS. Surveillance acute mye­loid leu­ke­mia in patients with con­gen­i­tal neutropenia receiv­ing
G-CSF ther­apy. Blood. 2000;96(2):429-436.
strat­e­gies for patients with SCN will need to account for recent 13. Kalra R, Dale D, Freedman M, et al. Monosomy 7 and acti­vat­ing RAS muta­
improve­ments in HSCT out­comes even in the pres­ence of active tions accom­pany malig­nant trans­for­ma­tion in patients with con­gen­i­tal
mye­loid malig­nancy. neutropenia. Blood. 1995;86(12):4579-4586.
Many open ques­tions in the field need to be addressed and 14. Link DC, Kunter G, Kasai Y, et al. Distinct pat­terns of muta­tions occur­ring in
de novo AML ver­sus AML aris­ing in the set­ting of severe con­gen­i­tal neutro-
may affect clin­i­cal man­age­ment of patients with con­gen­i­tal neu-
penia. Blood. 2007;110(5):1648-1655.
tropenia. Prospective tri­als are needed to deter­mine whether 15. Burroughs L, Woolfrey A, Shimamura A. Shwachman-Diamond syn­drome: a
RUNX1 or biallelic TP53 var­i­ants are pre­dic­tive of leu­ke­mic trans­ review of the clin­i­cal pre­sen­ta­tion, molec­u­lar path­o­gen­e­sis, diag­no­sis, and
for­ma­tion in SCN or SDS, respec­tively. The fre­quency and clin­i­cal treat­ment. Hematol Oncol Clin North Am. 2009;23(2):233-248.
16. Myers KC, Bolyard AA, Otto B, et  al. Variable clin­ i­
cal pre­ sen­ta­
tion of
impli­ca­tions of clonal hema­to­poi­e­sis due to chro­mo­somal abnor­
Shwachman-Diamond syn­ drome: update from the North Amer­ i­
can
mal­i­ties in patients with CN are unknown. Prospective studies to Shwachman-Diamond Syndrome Registry. J Pediatr. 2014;164(4):866-870.
detect somatic single nucleotide, small insertions/deletions, or 17. Shimamura A. Shwachman-Diamond syn­ drome. Semin Hematol. 2006;

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/514/1851470/514warren.pdf by guest on 13 December 2021


copy number alterations in patients with CN are needed. Finally, 43(3):178-188.
it will be impor­tant to define risks of mye­loid malig­nancy and 18. Hashmi SK, Allen C, Klaassen R, et al. Comparative anal­y­sis of Shwachman-
Diamond syn­drome to other inherited bone mar­row fail­ure syn­dromes and
pat­terns of clonal evo­lu­tion in the expanding genetic sub­types geno­type-phe­no­type cor­re­la­tion. Clin Genet. 2011;79(5):448-458.
of con­gen­i­tal neutropenia. 19. Rosenberg PS, Alter BP, Bolyard AA, et al; Severe Chronic Neutropenia
International Registry. The inci­dence of leu­ke­mia and mor­tal­ity from sep­sis
in patients with severe con­gen­i­tal neutropenia receiv­ing long-term G-CSF
Conflict-of-inter­est dis­clo­sure
ther­apy. Blood. 2006;107(12):4628-4635.
Julia T. Warren: no com­pet­ing finan­cial inter­ests to declare. 20. Kennedy AL, Myers KC, Bowman J, et al. Distinct genetic path­ways define
Dan­iel C. Link: no com­pet­ing finan­cial inter­ests to declare. pre-malig­nant ver­sus com­pen­sa­tory clonal hema­to­poi­e­sis in Shwachman-
Diamond syn­drome. Nat Commun. 2021;12(1):1334.
21. Lindsley RC, Saber W, Mar BG, et al. Prognostic muta­tions in myelodysplas-
Off-label drug use
tic syn­drome after stem-cell trans­plan­ta­tion. N Engl J Med. 2017;376(6):536-
Julia T. Warren: no off-label drug use. 547.
Dan­iel C. Link: no off-label drug use. 22. Genovese G, Kähler AK, Handsaker RE, et  al. Clonal hema­to­poi­e­sis and
blood-can­ cer risk inferred from blood DNA sequence. N Engl J Med.
2014;371(26):2477-2487.
Correspondence
23. Jaiswal S, Fontanillas P, Flannick J, et al. Age-related clonal hema­to­poi­e­sis
Dan­iel C. Link, Division of Oncology, Department of Inter- asso­ci­ated with adverse out­comes. N Engl J Med. 2014;371(26):2488-2498.
nal Medicine, Washington University School of Medicine, 660 24. Warren JT, Link DC. Clonal hema­to­poi­e­sis and risk for hema­to­logic malig­
S. Euclid Ave, Campus Box 8007, St. Louis, MO 63110; e-mail: nancy. Blood. 2020;136(14):1599-1605.
danielclink@wustl​­.edu. 25. Xia J, Miller CA, Baty J, et al. Somatic muta­tions and clonal hema­to­poi­e­sis
in con­gen­i­tal neutropenia. Blood. 2018;131(4):408-416.
26. Hermans MH, Antonissen C, Ward AC, Mayen AE, Ploemacher RE, Touw IP.
References Sustained recep­tor acti­va­tion and hyperproliferation in response to gran-
1. Skokowa J, Dale DC, Touw IP, Zeidler C, Welte K. Severe con­gen­i­tal neutro- ulocyte col­ony-stim­u­lat­ing fac­tor (G-CSF) in mice with a severe con­gen­
penias. Nat Rev Dis Primers. 2017;3:17032. i­tal neutropenia/acute mye­loid leu­ke­mia-derived muta­tion in the G- CSF
2. Boztug K, Appaswamy G, Ashikov A, et al. A syn­drome with con­gen­i­tal neu- recep­tor gene. J Exp Med. 1999;189(4):683-692.
tropenia and muta­tions in G6PC3. N Engl J Med. 2009;360(1):32-43. 27. Hermans MH, Ward AC, Antonissen C, Karis A, Löwenberg B, Touw IP. Per-
3. Horwitz M, Benson KF, Person RE, Aprikyan AG, Dale DC. Mutations in ELA2, turbed granulopoiesis in mice with a targeted muta­tion in the granulocyte
encoding neu­tro­phil elas­tase, define a 21-day bio­log­i­cal clock in cyclic col­ony-stim­u­lat­ing fac­tor recep­tor gene asso­ci­ated with severe chronic
haematopoiesis. Nat Genet. 1999;23(4):433-436. neutropenia. Blood. 1998;92(1):32-39.
4. Klein C, Grudzien M, Appaswamy G, et  al. HAX1 defi­ciency causes auto­ 28. McLemore ML, Poursine-Laurent J, Link DC. Increased granulocyte col­ony-
so­mal reces­sive severe con­gen­i­tal neutropenia (Kostmann dis­ease). Nat stim­u­lat­ing fac­tor respon­sive­ness but nor­mal rest­ing granulopoiesis in
Genet. 2007;39(1):86-92. mice car­ry­ing a targeted granulocyte col­ony-stim­u­lat­ing fac­tor recep­tor
5. Xia J, Bolyard AA, Rodger E, et al. Prevalence of muta­tions in ELANE, GFI1, muta­tion derived from a patient with severe con­gen­i­tal neutropenia. J Clin
HAX1, SBDS, WAS and G6PC3 in patients with severe con­gen­i­tal neutrope- Invest. 1998;102(3):483-492.
nia. Br J Haematol. 2009;147(4):535-542. 29. Liu F, Kunter G, Krem MM, et al. Csf3r muta­tions in mice con­fer a strong
6. Dale DC, Bonilla MA, Davis MW, et al. A ran­dom­ized con­trolled phase III trial clonal HSC advan­tage via acti­va­tion of Stat5. J Clin Invest. 2008;118(3):946-
of recom­bi­nant human granulocyte col­ony-stim­u­lat­ing fac­tor (filgrastim) 955.
for treat­ment of severe chronic neutropenia. Blood. 1993;81(10):2496-2502. 30. Tsaknakis G, Gallì A, Papadakis S, et al. Incidence and prog­no­sis of clonal
7. Rosenberg PS, Zeidler C, Bolyard AA, et al. Stable long-term risk of leu­kae­ hema­to­poi­e­sis in patients with chronic idi­o­pathic neutropenia. Blood.
mia in patients with severe con­gen­i­tal neutropenia maintained on G-CSF 2021;138(14):1249-1257.
ther­apy. Br J Haematol. 2010;150(2):196-199. 31. Menne TF, Goyenechea B, Sánchez-Puig N, et al. The Shwachman-Bodian-
8. Donadieu J, Leblanc T, Bader Meunier B, et  al.; French Severe Chronic Diamond syn­drome pro­tein medi­ates trans­la­tional acti­va­tion of ribo­somes
Neutropenia Study Group. Analysis of risk fac­tors for myelodysplasias, leu­ in yeast. Nat Genet. 2007;39(4):486-495.
ke­mias and death from infec­tion among patients with con­gen­i­tal neutro- 32. Wong CC, Traynor D, Basse N, Kay RR, Warren AJ. Defective ribo­some
penia. Experience of the French Severe Chronic Neutropenia Study Group. assem­bly in Shwachman-Diamond syn­drome. Blood. 2011;118(16):4305-4312.
Haematologica. 2005;90(1):45-53. 33. Zambetti NA, Bindels EM, Van Strien PM, et al. Deficiency of the ribo­some
9. Furutani E, Newburger PE, Shimamura A. Neutropenia in the age of genetic bio­gen­e­sis gene Sbds in hema­to­poi­etic stem and pro­gen­i­tor cells causes
test­ing: advances and chal­lenges. Am J Hematol. 2019;94(3):384-393. neutropenia in mice by atten­u­at­ing lin­e­age pro­gres­sion in mye­lo­cytes.
10. Ley TJ, Miller C, Ding L, et al; Cancer Genome Atlas Research Network. Haematologica. 2015;100(10):1285-1293.
Genomic and epigenomic land­scapes of adult de novo acute mye­loid leu­ 34. Touw IP. Game of clones: the geno­mic evo­lu­tion of severe con­gen­i­tal neu-
ke­mia. N Engl J Med. 2013;368(22):2059-2074. tropenia. Hematol Am Soc Hematol Educ Program. 2015;2015:1-7.
11. Skokowa J, Steinemann D, Katsman-Kuipers JE, et  al. Cooperativity of 35. Dannenmann B, Klimiankou M, Oswald B, et al. iPSC mod­el­ing of stage-spe­
RUNX1 and CSF3R muta­tions in severe con­gen­i­tal neutropenia: a unique cific leu­ke­mo­gen­e­sis reveals BAALC as a key onco­gene in severe con­gen­i­
path­way in mye­loid leu­ke­mo­gen­e­sis. Blood. 2014;123(14):2229-2237. tal neutropenia. Cell Stem Cell. 2021;28(5):906.e6-922.e6.
Prakash Singh Shekhawat
Clonal evo­lu­tion in con­gen­i­tal neutropenia  |  519
36. Rotulo GA, Beaupain B, Rialland F, et al. HSCT may lower leu­ke­mia risk in 44. Fioredda F, Calvillo M, Bonanomi S, et al; Neutropenia Committee of the
ELANE neutropenia: a before-after study from the French Severe Congeni- Marrow Failure Syndrome Group of the AIEOP (Associazione Italiana Ema-
tal Neutropenia Registry. Bone Marrow Transplant. 2020;55(8):1614-1622. to-Oncologia Pediatrica). Congenital and acquired neutropenias con­sen­
37. Dale DC, Makaryan V, Kelley ML, et al. Termination and frame­shift muta­tions sus guide­lines on ther­apy and fol­low-up in child­hood from the Neutropenia
in ELANE are asso­ci­ated with adverse out­comes in patients with severe Committee of the Marrow Failure Syndrome Group of the AIEOP (Associazi-
chronic neutropenia. Blood. 2016;128(22):1326. one Italiana Emato-Oncologia Pediatrica). Am J Hematol. 2012;87(2):238-
38. Makaryan V, Zeidler C, Bolyard AA, et  al. The diver­sity of muta­tions and 243.
clin­i­cal out­comes for ELANE-asso­ci­ated neutropenia. Curr Opin Hematol. 45. Newburger PE, Dale DC. Evaluation and man­age­ment of patients with iso­
2015;22(1):3-11. lated neutropenia. Semin Hematol. 2013;50(3):198-206.
39. Fioredda F, Iacobelli S, van Biezen A, et al; Severe Aplastic Anemia the 46. Porter CC, Druley TE, Erez A, et  al. Recommendations for sur­ veil­
lance
Inborn Error, and the Pediatric Disease Working Parties of the Euro­pean for chil­dren with leu­ke­mia-predisposing con­di­tions. Clin Cancer Res.
Society for Blood and Bone Marrow Transplantation (EBMT) and Stem Cell 2017;23(11):e14-e22.
Transplant for Immunodeficiencies in Europe (SCETIDE). Stem cell trans­ 47. Maserati E, Pressato B, Valli R, et  al. The route to devel­op­ment of myel-
plan­ta­tion in severe con­gen­i­tal neutropenia: an anal­y­sis from the Euro­pean odysplastic syn­drome/acute mye­loid leu­kae­mia in Shwachman-Diamond
Society for Blood and Marrow Transplantation. Blood. 2015;126(16):1885- syn­drome: the role of age­ing, kar­yo­type insta­bil­ity, and acquired chro­mo­
1892; quiz 1970. some anom­a­lies. Br J Haematol. 2009;145(2):190-197.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/514/1851470/514warren.pdf by guest on 13 December 2021


40. Zeidler C, Nickel A, Sykora K-W, Welte K. Improved out­come of stem cell 48. Minelli A, Maserati E, Nicolis E, et al. The iso­chro­mo­some i(7)(q10) car­ry­ing
trans­plan­ta­tion for severe chronic neutropenia with or with­out sec­ond­ary c.258 + 2t>c muta­tion of the SBDS gene does not pro­mote devel­op­ment of
leu­ke­mia: a long-term anal­y­sis of Euro­pean data for more than 25 years by mye­loid malig­nan­cies in patients with Shwachman syn­drome. Leukemia.
the SCNIR. Blood. 2013;122(21):3347. 2009;23(4):708-711.
41. Cesaro S, Pillon M, Sauer M, et  al. Long-term out­come after allo­ge­neic 49. Pressato B, Valli R, Marletta C, et al. Deletion of chro­mo­some 20 in bone
hema­to­poi­etic stem cell trans­plan­ta­tion for Shwachman-Diamond syn­ mar­row of patients with Shwachman-Diamond syn­drome, loss of the EIF6
drome: a ret­ ro­
spec­tive anal­y­sis and a review of the lit­ er­
at­ure by the gene and benign prog­no­sis. Br J Haematol. 2012;157(4):503-505.
Severe Aplastic Anemia Working Party of the Euro­pean Society for Blood 50. Donadieu J, Fenneteau O, Beaupain B, et al; Associated inves­ti­ga­tors of
and Marrow Transplantation (SAAWP-EBMT). Bone Marrow Transplant. the French Severe Chronic Neutropenia Registry. Classification of and
2020;55(9):1796-1809. risk fac­tors for hema­ to­
logic com­ pli­
ca­
tions in a French national cohort
42. Myers K, Hebert K, Antin J, et  al. Hematopoietic stem cell trans­plan­ta­ of 102 patients with Shwachman-Diamond syn­ drome. Haematologica.
tion for Shwachman-Diamond syn­drome. Biol Blood Marrow Transplant. 2012;97(9):1312-1319.
2020;26(8):1446-1451.
43. Myers KC, Furutani E, Weller E, et  al. Clinical fea­tures and out­comes of
patients with Shwachman-Diamond syn­drome and myelodysplastic syn­
drome or acute mye­loid leu­kae­mia: a multicentre, ret­ro­spec­tive, cohort © 2021 by The Amer­i­can Society of Hematology
study. Lancet Haematol. 2020;7(3):e238-e246. DOI 10.1182/hema­tol­ogy.2021000286

520  |  Hematology 2021  |  ASH Education Program


PERIOPERATIVE CONSULTATIVE HEMATOLOGY: CAN YOU CLEAR MY PATIENT FOR SURGERY ?

Perioperative consultative hematology:


can you clear my patient for a procedure?

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/521/1851845/521burnett.pdf by guest on 13 December 2021


Allison Elaine Burnett,1 Bishoy Ragheb,2 and Scott Kaatz3
University of New Mexico, Health Sciences Center, College of Pharmacy, Albuquerque, NM; 2Tennessee Valley Health Systems (TVHS) Veterans
1

Affairs, Nashville, TN; and 3Henry Ford Hospital, Detroit, MI

Periprocedural management of antithrombotics is a common but challenging clinical scenario that renders patients
vulnerable to potential adverse events such as bleeding and thrombosis. Over the past decade, periprocedural anti-
thrombotic approaches have changed considerably with the advent of direct oral anticoagulants (DOACs), as well as
a paradigm shift away from bridging in many warfarin patients. Successfully navigating this high-risk period relies on a
number of individualized patient assessments conducted within a framework of standardized, systematic approaches.
It also requires a thorough understanding of antithrombotic pharmacokinetics, multidisciplinary coordination of care,
and comprehensive patient education and empowerment. In this article, we provide clinicians with a practical, stepwise
approach to periprocedural management of antithrombotic agents through case-based examples of relevant clinical
scenarios.

LEARNING OBJECTIVES
• Explain significant differences between perioperative management of DOAC and warfarin, including the rationale
for these differences
• Determine if temporary interruption of anticoagulant therapy is needed through evaluation of factors, including
bleeding and thrombotic potential of the procedure and the patient
• Identify warfarin patients who do and do not warrant consideration for perioperative bridging
• Develop safe, effective perioperative anticoagulation plans for DOAC and warfarin patients based on existing evi-
dence and expert consensus

Introduction vulnerability to medication discrepancies, therapeutic


Although exact numbers are not known, it is estimated that overlap, failure to resume anticoagulation, communica-
6 to 8 million people in the United States are prescribed tion errors, and missed coordination of follow-up.8,9
oral anticoagulation (OAC) with either warfarin or a direct • Anecdotal experience suggests providers lack familiar-
oral anticoagulant (DOAC).1,2 Approximately 10% to 15% of ity with the pharmacokinetics of the DOACs, which differ
these patients will require temporary interruption of anti- dramatically from those of warfarin. It is not uncom-
coagulation for surgery or an invasive procedure, which mon in clinical practice to discover DOAC patients with
equates to 600 000 to 1 200 000 interruptions annually.3,4 periprocedural plans with inappropriately prolonged
The periprocedural period is a time of significant risk for hold times and/or overlapping therapy with low molec-
anticoagulation patients for a multitude of reasons, includ- ular weight heparin (LMWH) as providers try to manage
ing but not limited to the following: these agents in a similar manner as they would warfarin.
• A number of complex individualized assessments must The increased risk for adverse events in the periproce-
to be made to estimate bleeding and thrombotic risks dural period has prompted a number of quality improve-
of both the procedure and the patient, which are largely ment and patient safety initiatives and shifts in practice.
based on expert consensus.5,6
• It is estimated that surgical patients will experience up to • In 2019, the Joint Commission revised its National
15 transitions of care.7 Each transition is associated with Patient Safety Goal (NPSG) 03.05.01 pertaining to anti-

Prakash Singh Shekhawat


Perioperative management of antithrombotics | 521
co­ag­u­lants to include a new ele­ment of per­for­mance (EP3) Table 1. Minor sur­ger­ies or pro­ce­dures that may not require
call­ing for hos­pi­tals to use approved pro­to­cols and evi­dence- inter­rup­tion of OAC
based prac­tice guide­lines for periprocedural man­age­ment of
all­ patients tak­ing oral anti­co­ag­u­lants.10 •  Minor den­tal (eg, 1-2 tooth extrac­tion, cleaning)
•  The Centers for Medi­care & Med­ic­aid Services offers merit- •  Minor der­ma­to­logic or cuta­ne­ous
based incen­tive pay­ments to phy­si­cians who pro­vide doc­
• Cataract
u­men­ta­tion of periprocedural anticoagulation man­age­ment
plans, includ­ing tim­ing of inter­rup­tion, man­age­ment of con­ •  Cardiac implant­able devices (pace­mak­ers, defi­bril­la­tors)
com­i­tant antithrombotics, bridg­ing (if indi­cated), lab­o­ra­tory •  Cardiac abla­tions, cardioversion, elec­tro­phys­i­­ol­ogy stud­ies
mea­sure­ments, tim­ing of resump­tion, and dis­cus­sion of plan
•  Endovascular pro­ce­dures (eg, angio­plasty)
with the patient.11
•  An increas­ing num­ber of health sys­tems are using clin­i­cal phar­ •  Endoscopy with­out resec­tion or biopsy
ma­ cists and anticoagulation stew­ ard­
ship pro­
grams to opti­ •  Intramuscular vac­ci­na­tion

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/521/1851845/521burnett.pdf by guest on 13 December 2021


mize devel­op­ment and appli­ca­tion of guide­line-recommended •  Percutaneous cor­o­nary inter­ven­tions (radial approach)
periprocedural plans.12,13
It is impor­tant to acknowl­edge that perioperative con­sul­ clude throm­bo­cy­to­pe­nia, renal dys­func­tion with ure­mia, sig­nif­i­
ta­
tion may be accom­ pa­nied by dif­fer­
ences of opin­ ion and cant hepatic impair­ment with base­line coagulopathy, his­tory of
approaches between involved dis­ci­plines that requires thor­ bleed­ing, and con­com­i­tant med­i­ca­tions such as antiplatelets or
ough dis­cus­sion and con­sen­sus build­ing. In addi­tion, a patient non­ste­roi­dal anti-inflam­ma­tory drugs. Although many of these
who is cleared for sur­gery should not be deemed “risk-free” are not mod­ i­
fi­
able, being aware of their pres­ ence may help
from adverse events. Multidisciplinary col­ lab­or­a­
tion before, antic­i­pate and address com­pli­ca­tions.
dur­ing, and after sur­gery to assess for and mit­ig­ ate risk as much
as pos­si­ble is essen­tial for opti­mized patient care. Minimal bleed risk pro­ce­dures
Studies have shown that many low bleed­ing risk minor sur­ger­
Atrial fibril­la­tion ies or pro­ce­dures (which con­sti­tute up to 20% of cases) can
be safely done with­out war­fa­rin inter­rup­tion.19 In addi­tion, ran­
dom­ized con­trolled tri­als and meta-ana­ly­ses have shown that
CLINICAL CASE war­fa­rin inter­rup­tion with or with­out LMWH bridg­ing leads to
A 65-year-old female patient is to undergo an elec­tive right hip more adverse events than no war­fa­rin inter­rup­tion.20-23 There is
replace­ment. Her med­i­cal his­tory includes a bioprosthetic aor­ less cer­tainty with con­tinu­ing DOACs around minor pro­ce­dures.
tic valve, nonvalvular atrial fibril­la­tion, hyper­ten­sion, and dia­be­ However, a grow­ing body of evi­dence sug­gests unin­ter­rupted
tes. She is referred for periprocedural DOAC rec­om­men­da­tions. DOACs may be rea­son­able24-26 and pos­si­bly safer than unin­ter­
Laboratory val­ues, includ­ing renal and liver func­tion, are nor­mal. rupted war­fa­rin in many pro­ce­dures (Table 1).27,28

Our patient is under­go­ing a hip replace­ment that we would


Step 1: Does anticoagulation need to be interrupted? con­sider a high bleed risk pro­ce­dure, and DOAC should be
Bleeding risk of the pro­ce­dure interrupted.
The need for OAC inter­ rup­tion is deter­ mined pri­ mar­ily by the
bleed­ing risk of the pro­ce­dure. Unfortunately, a high-qual­ity, evi­
dence-based schema to cat­e­go­rize pro­ce­dural bleed­ing risk has Step 2: If OAC inter­rup­tion is needed, does the patient
not been well established and has led to dif­fer­ences across guide­ require bridg­ing?
lines and var­i­a­tions in prac­tice.14-16 Recently, the International Soci- DOACs
ety on Thrombosis and Haemostasis issued guid­ance on this with There are sig­nif­i­cant phar­ma­co­ki­netic dif­fer­ences between DOACs
the intent of pro­mot­ing more stan­dard­ized approaches.17 In our and war­fa­rin, and thus their perioperative man­age­ment requires
prac­tice, we also often refer to the very com­pre­hen­sive pro­ce­ dif­fer­ent approaches.26,29 In patients with nor­mal renal func­tion,
dural bleed­ing risk appen­dix published with the 2017 ACC Expert DOACs have a much shorter half-life than war­fa­rin (approx­i­ma­
Consensus Decision Pathway for Periprocedural Management of tely 12 hours vs approx­i­ma­tely 40 hours, respec­tively) and much
Anticoagulation in Patients With Nonvalvular Atrial Fibrillation.18 In faster off­set. Thus, with­hold­ing DOACs for a prolonged period
Table 1, we have pro­vided a list (nonexhaustive) of pro­ce­dures with of time (eg, 5 days as is rou­tinely done with war­fa­rin) poten­tially
min­i­mal bleed­ing risk that likely do not require inter­rup­tion that places the patient at risk for a throm­botic event. Also, given
can greatly sim­plify man­age­ment and mit­i­gate risk for harm. For the rapid off­set of DOACs, it is not nec­es­sary to inter­rupt them
other pro­ce­dures, we sug­gest cli­ni­cians use existing pro­ce­dure preprocedurally and replace with LMWH. In the postprocedure
cat­e­go­ri­za­tion tools as a frame­work for dis­cus­sion with sur­geons set­ting, it is imper­a­tive to rec­og­nize the faster onset of DOAC
and other interventionalists when devel­op­ing a perioperative plan. anti­co­ag­u­lant action (approx­i­ma­tely 3 hours) com­pared with
war­ fa­rin (approx­ i­
ma­tely 4-5 days) and the need for care­ fully
Bleeding risk of the patient timed resump­tion to mit­i­gate bleed­ing risk. Very impor­tantly,
The intrin­sic bleed­ing risk of the patient should also be con­ DOACs should never be overlapped with par­en­teral anti­co­ag­u­
sid­
ered dur­ ing periprocedural plan­ ning. Characteristics that lants, such as LMWH, as this is not nec­es­sary due to their rapid
we rou­tinely con­sider when assessing patient bleed­ing risk in- onset and stud­ies show­ing sig­nif­i­cantly increased bleed­ing.3,4,25

522  |  Hematology 2021  |  ASH Education Program


The Perioperative Anticoagulation Use for Surgery Evaluation neuraxial anes­the­sia and lon­ger for dabigatran if the cre­at­i­nine
(PAUSE) cohort study30 used a sim­ ple DOAC inter­ rup­
tion and clear­ance is low.34
resump­tion pro­to­col with­out any bridg­ing in patients with atrial
fibril­la­tion under­go­ing sur­gi­cal pro­ce­dures. This was based in part
on suc­cess­ful use of a sim­i­lar approach in an ear­lier pro­spec­tive • We would hold the DOAC for 2 days before sur­gery and restart
mul­ti­cen­ter trial eval­u­at­ing perioperative man­age­ment of dabiga- the DOAC on post­op­er­a­tive day 2 or 3.
tran.31 Timing of inter­rup­tion and resump­tion was based on DOAC •  In the postprocedural interim until ther­a­peu­tic anticoagulation
phar­ma­co­ki­netic prop­er­ties, pro­ce­dural bleed risk, and renal func­ is resumed, we would ensure ade­quate pro­phy­laxis (eg, enox-
tion. The inves­ti­ga­tors hoped to show a major bleed­ing rate of less aparin 40 mg subcutaneously [SQ ] once daily) to pre­vent post­
than 2.0% and a stroke or tran­sient ische­mic attack (TIA) rate of op­er­a­tive VTE.
less than 1.5% with 95% cer­tainty. Of note, this study used num­
ber of days and not hours for inter­rup­tion and resump­tion tim­ing,
as shown in Table 2. Because dabigatran is pri­mar­ily elim­i­nated CLINICAL CASE (Continued)

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/521/1851845/521burnett.pdf by guest on 13 December 2021


through the kid­neys (80%), cre­at­i­nine clear­ance was esti­mated The patient does well, and a year later, she is going to have her
(using the Cockcroft-Gault equa­tion and actual body weight), and other hip replaced. Unfortunately, her insur­ance has changed, and
hold times were dou­bled in dabigatran patients with an esti­mated she can no lon­ger afford a DOAC and is tak­ing war­fa­rin. Her med­
clear­ance of less than 50 mL/min. Results showed with 95% con­fi­ i­cal his­tory is unchanged, and her lab­o­ra­tory tests remain nor­mal.
dence that bleed­ing rates were not greater than 2.00%, 1.73%, and
2.65% with apixaban, dabigatran, and rivaroxaban, respec­tively,
and arte­ rial throm­bo­em­ bo­lism was not greater than 1.5% with Warfarin
any DOAC. In addi­tion, it was shown that more than 90% of DOAC Due to the long off­set of war­fa­rin (approx­i­ma­tely 5 days), cli­ni­
patients col­lec­tively had lit­tle to no resid­ual anti­co­ag­u­lant effect cians must deter­mine tim­ing of inter­rup­tion to pro­vide the de-
from more than 30 ng/mL (which has been suggested as an accept­ sired resid­ual anti­co­ag­u­lant effect, as well as any role for bridg­ing
able pre­op­er­a­tive plasma con­cen­tra­tion32), pre­clud­ing the need with a par­en­teral anti­co­ag­u­lant. For clar­ity, when we use the term
for rou­tine pre­op­er­a­tive lab­o­ra­tory assess­ment when the PAUSE bridg­ing, we mean ther­a­peu­tic-inten­sity anticoagulation, such as
pro­to­col is followed. The Scientific and Standardization Commit- enoxaparin 1 mg/kg twice daily. The deci­sion to bridge or not is
tee of the International Society on Thrombosis and Haemostasis based on the patient’s indi­ca­tion for anticoagulation, under­ly­ing
sug­gests that if a pre­op­er­a­tive quan­ti­ta­tive DOAC level is 30 ng/mL throm­botic risk, and the throm­botic risk of the pro­ce­dure. Al-
or less, it is rea­son­able to pro­ceed with an inva­sive pro­ce­dure.33 To though bridg­ing of war­fa­rin was once rou­tinely employed, ret­ro­
our knowl­edge, there are no data to sug­gest enhanced util­ity of spec­tive obser­va­tional evi­dence published over the past decade
mea­sur­ing DOAC con­cen­tra­tions before elec­tive pro­ce­dures com­ has con­sis­tently shown bridg­ing to be asso­ci­ated with net harm
pared with a sim­ple stan­dard­ized, phar­ma­co­ki­netic-based pro­to­ and sug­gests it should be avoided in most cases.6,35-37
col as was used in the PAUSE trial. Urgent or emer­gent pro­ce­dures The ran­ dom­ ized con­ trolled Bridging Anticoagulation in
pose a chal­lenge, and there may be a role for DOAC mea­sure­ment Patients Who Require Temporary Interruption of Warfarin Ther-
in select sit­u­a­tions. However, it is crit­i­cal to bear in mind that these apy for an Elective Invasive Procedure or Surgery (BRIDGE) trial
quan­ti­ta­tive DOAC assays are not avail­­able in many hos­pi­tals, and sought to answer whether fore­go­ing LMWH bridg­ing in atrial
if they are, turn­around times may pre­clude any util­ity. fibril­la­tion patients requir­ing war­fa­rin inter­rup­tion for a pro­ce­dure
An unanswered ques­tion is what to do in patients with a his­ would result in less major bleed­ing with­out an increase in arte­
tory of venous throm­bo­em­bo­lism (VTE), and we extrap­o­late rial throm­bo­em­bo­lism. Per stan­dard­ized pro­to­col, war­fa­rin was
the PAUSE study tim­ing of inter­rup­tion and resump­tion to these held for 5 days preprocedurally, and patients were ran­dom­ized to
patients with con­fi­dence in the major bleed­ing rates but uncer­ pla­cebo or LMWH (dalteparin) with 30-day pri­mary out­comes of
tainty in VTE recur­rence. major bleed­ing (supe­ri­or­ity) and arte­rial throm­bo­em­bolic event
An addi­tional point is the PAUSE study dif­fers slightly from the (noninferiority). There was no dif­fer­ence in arte­rial throm­bo­em­
rec­om­men­da­tions by the Amer­i­can Society of Regional Anes- bo­lism 0.4% vs 0.3%, and major bleed­ing was reduced with no
thesia, which rec­om­mend hold­ing DOACs for 72 hours prior to bridg­ing (1.3%) vs LMWH bridg­ing (3.2%) (P < .005).37 This trial

Table 2. Periprocedural DOAC inter­rup­tion pro­to­col from PAUSE study

Characteristic Apixaban Rivaroxaban Dabigatran CrCl ≥50 Dabigatran CrCl <0 Edoxaban
Preprocedural inter­rup­tion Days Days Days Days Not stud­ied
  Low bleed­ing risk pro­ce­dure 1 1 1 2
  High bleed­ing risk pro­ce­dure 2 2 2 4
Postprocedural resump­tion
  Low bleed­ing risk pro­ce­dure 1 1 1 1
  High bleed­ing risk pro­ce­dure 2-3 2-3 2-3 2-3
For pro­ce­dural bleed­ing risk strat­i­fi­ca­tion, we sug­gest using soci­ety-based guid­ance in the appen­dix to the ACC con­sen­sus deci­sion path­way18 or
cat­e­go­ri­za­tion from PAUSE study.30
CrCl, cre­at­i­nine clear­ance.
Prakash Singh Shekhawat
Perioperative man­age­ment of antithrombotics  |  523
prompted a par­a­digm shift away from bridg­ing in most patients and she now has a bileaflet mechan­i­cal aor­tic valve. Her other med­
with atrial fibril­la­tion. i­cal his­tory is unchanged, and lab­o­ra­tory val­ues remain nor­mal. She
In the BRIDGE trial, war­fa­rin was resumed the night of the pro­ pres­ents for perioperative war­fa­rin man­age­ment again.
ce­dure at the patient’s usual home dose. The mean time to rees-
tablish a ther­a­peu­tic inter­na­tional nor­mal­ized ratio (INR) was 8
The Amer­i­can College of Chest Physicians (ACCP) 2012 guide­
days. To min­i­mize this period of sub­ther­a­peu­tic anticoagulation,
lines and the Amer­i­can College of Cardiology (ACC)/Amer­i­can
it is rea­son­able to con­sider a boosted dose of war­fa­rin for 1 to 2
Heart Association 2020 guide­lines are con­cor­dant in their clas­
days after the pro­ce­dure in the absence of high bleed­ing risk.38
si­fi­ca­tion of mechan­i­cal heart valves as (1) high risk if any mitral,
caged-ball, or tilting disk valves or recent (within 6 months)
Thromboembolic risk strat­i­fi­ca­tion
stroke or TIA; (2) mod­er­ate risk with bileaflet aor­tic valve and
We use a throm­bo­em­bolic risk strat­i­fi­ca­tion approach based
any other risk fac­tor, which includes atrial fibril­la­tion, prior stroke
on avail­­able evi­dence and expert con­sen­sus rec­om­men­da­tions
or TIA, hyper­ten­sion, dia­be­tes, con­ges­tive heart fail­ure, or older
from mul­ti­ple orga­ni­za­tions to guide deci­sions on bridg­ing war­
than 75 years; and (3) low risk with bileaflet aor­tic valves with no

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/521/1851845/521burnett.pdf by guest on 13 December 2021


fa­rin patients who require tem­po­rary inter­rup­tion for a pro­ce­
other risk fac­tors (Table 3).19,39
dure (Table 3).18,19,39,40
The only ran­dom­ized trial for LMWH bridg­ing for war­fa­rin inter­
rup­tion in patients with mechan­i­cal heart valves that we are aware
• We would not use bridg­ing anticoagulation in our atrial fibril­la­tion of is the recently published Postoperative low molecular weight
patient because her CHADS2-VA2Sc (Congestive heart failure = 1, heparin bridging treatment for patients at high risk of arterial
Hypertension = 1, age >/= 75 years = 2, Diabetes = 1, Stroke/TIA = 2, thromboembolism (PERIOP 2) trial.41 Patients with atrial fibril­la­tion
Vascular disease = 1, Age 65-74 = 1, Sex category = 1) score is 4. or non-high-risk mechan­i­cal valves who required war­fa­rin inter­rup­
•  We would resume her war­fa­rin the night of the pro­ce­dure. tion for a pro­ce­dure (n = 1471) were ran­dom­ized to LMWH bridg­ing
• While the patient is still in the hos­pi­tal, we would ensure ade­ or pla­cebo and followed for 90 days. LMWH bridg­ing consisted
quate pro­phy­laxis with low-dose anticoagulation (eg, enoxa- of ther­a­peu­tic dose dalteparin 200 IU/kg sub­cu­ta­ne­ously on days
parin 40 mg SQ once daily) to pre­vent post­op­er­a­tive VTE until −3 and −2 before pro­ce­dure and half the ther­a­peu­tic dose of
the INR is 2 or more. 100 IU/kg sub­cu­ta­ne­ously the day before the pro­ce­dure. Post-
procedurally (same day or next day), patients in the inter­ven­tion
arm at high bleed risk were given fixed-dose pro­phy­lac­tic dalte-
Mechanical heart valve parin 5000 IU, and those at low bleed risk were given dalteparin
200 IU/kg sub­cu­ta­ne­ously until the INR was more than 1.9.
In the sub­group of 304 patients with non-high-risk mechan­
i­cal valves, no bridg­ing vs bridg­ing major throm­bo­em­bo­
CLINICAL CASE (Continued) lism event rates were 0% vs 0.7% (P = .49), and major bleed­ing
The patient did well after her sec­ond hip sur­gery and has been occurred in 2% vs 0.7% (P = .62) respec­tively. The inves­ti­ga­tors
doing so much walk­ing that her left knee now needs replace­ment. con­cluded there is no ben­e­fit from postprocedure LMWH bridg­
Unfortunately, her bioprosthetic aor­tic valve gave out in the interim, ing in patients with non-high-risk mechan­i­cal valves.

Table 3. Thromboembolic risk strat­i­fi­ca­tion and bridg­ing con­sid­er­ations

Indication Mechanical heart valve Atrial fibril­la­tion VTE


Guideline(s) ACCP 2012, ACC/AHA 2020 ACCP 2012, ACC 2017 ACCP 2012, ASH 2018
Thrombotic risk Criteria Recommendation Criteria* Recommendation Criteria Recommendation
High •  All mitral valve Suggest bridg­ing/ CHADS2 CHA2DS-VA2Sc Suggest bridg­ing • Within 3 CHEST 2012:
• Caged-ball and rea­son­able to >4 >7 months Suggest
tilting disc bridge • Severe bridg­ing
• Stroke or TIA in thrombophilia ASH 2018: Not
past 6 months addressed
Moderate Bileaflet aor­tic valve Individualized CHADS2 CHA2DS-VA2Sc Individualized • Past 3-12 CHEST 2012:
+ risk fac­tors deci­sion based 3-4 5 or 6 deci­sion based months Individualized
•  Atrial fibril­la­tion on patient and on patient and •  Recurrent VTE deci­sion based
•  Prior stroke or TIA pro­ce­dural pro­ce­dural •  Active can­cer on patient and
• Hypertension nuances nuances • Nonsevere pro­ce­dural
• Diabetes thrombophilia nuances
•  Congestive heart ASH 2018: Do not
fail­ure bridge
•  Age >75 years
Low Bileaflet aor­tic Do not bridge CHADS2 CHA2DS-VA2Sc Do not bridge More than 12 CHEST 2012: Do
valve with­out risk ≤2 <4 months ago not bridge
fac­tors ASH 2018: Do not
bridge
*ACCP 2012 based on CHADS2; 2017 ACC expert con­sen­sus deci­sion path­way for periprocedural man­age­ment of anticoagulation based on CHA2DS-VA2Sc.
ACC, American College of Cardiology; ACCP, American College of Chest Physicians; AHA, Amer­i­can Heart Association.

524  |  Hematology 2021  |  ASH Education Program


Table 4. Special sit­u­a­tions that may influ­ence perioperative antithrombotic man­age­ment

Situation or issue Comments Suggested actions


Urgent/emer­gent • If there is not ade­quate time to allow nat­u­ral nor­mal­i­za­tion • Shared deci­sion mak­ing with the patient, mul­ti­dis­ci­
pro­ce­dures of the patient’s coag­u­la­tion sta­tus before sur­gery, use of plin­ary dis­cus­sion, and con­sul­ta­tion with a throm­bo­sis
rever­sal agents, prohemostatic agents, or spe­cific anti­ spe­cial­ist
dotes may be indi­cated and should be done judi­ciously • Clinicians are referred to existing guid­ance on rever­sal
and thought­fully. of anticoagulation.40,45,46
• Rapidly returning a patient to their native, prothrombotic
state along with any intrin­sic risk of throm­bo­sis posed by
the rever­sal agents or anti­dotes them­selves may increase
the risk for adverse events.
Patients on con­com­i­tant • This is an oppor­tune time to eval­u­ate the over­all clin­i­cal • Shared deci­sion mak­ing with the patient, mul­ti­dis­ci­
antiplatelet ther­a­pies neces­sity of con­com­i­tant antiplatelet ther­apy. If not indi­ plin­ary dis­cus­sion, and con­sul­ta­tion with pre­scriber of
cated, cli­ni­cians should dis­cuss per­ma­nent dis­con­tin­u­a­tion antiplatelet ther­apy (eg, car­di­­ol­o­gist, neu­rol­o­gist) and

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/521/1851845/521burnett.pdf by guest on 13 December 2021


with the patient and pre­scriber. throm­bo­sis spe­cial­ist
• Whether to tem­po­rar­ily inter­rupt con­com­i­tant antiplatelet • Clinicians are referred to existing guid­ance on
ther­a­pies is a com­plex deci­sion that is based on sev­eral perioperative antiplatelet man­age­ment.47,48
fac­tors, includ­ing indi­ca­tion, recency of events, bleed­ing,
and throm­botic risks of the pro­ce­dure and patient.
• Perioperative antiplatelet strat­e­gies should be indi­vid­u­ally
tai­lored based on mul­ti­dis­ci­plin­ary input.
History of hep­a­rin-induced • Patients with a his­tory of HIT should not receive any hep­ • Use an alter­na­tive, nonheparin anti­co­ag­u­lant such as
throm­bo­cy­to­pe­nia (HIT) a­rin or LMWH prod­ucts, includ­ing small doses such as bivalirudin, fondaparinux, or a DOAC as appro­pri­ate
flushes or VTE pro­phy­laxis. based on patient’s clin­i­cal sta­tus and clin­i­cal sit­u­a­tion.
• Clinicians are referred to existing guid­ance on HIT.49
Inferior vena cava (IVC) • The esti­mated inci­dence of VTE recur­rence in the first • If the patient is antic­i­pated to be off anticoagulation for
fil­ters month after an acute event off of anti­co­ag­u­lant ther­apy is <48 hours, aggres­sive phar­ma­co­logic pro­phy­laxis with
esti­mated to be 40%.50 expe­di­ent esca­la­tion to ther­a­peu­tic dos­ing is pre­ferred.
• If pos­si­ble, delay non­ur­gent/emer­gent pro­ce­dures to • If a retriev­able fil­ter is con­sid­ered, a plan for timely
allow at least 3 months of anticoagulation ther­apy fol­low­ removal should be clearly delin­eated prior to place­ment.
ing an acute VTE. • Clinicians are referred to existing guid­ance on IVC fil­ters.51
• If the pro­ce­dure can­not be delayed and the VTE occurred
in the pre­vi­ous 30 days, a retriev­able IVC fil­ter may be
con­sid­ered.
Severe renal impair­ment • Warfarin patients with severe renal impair­ment or on • These patients may need to have their war­fa­rin held at a
hemo­di­al­y­sis who have a clear indi­ca­tion for bridg­ing can­ prespecified time in the out­pa­tient set­ting and then be
not be man­aged with LMWH. admit­ted for bridg­ing ther­apy with intra­ve­nous hep­a­rin.

In a sys­temic review of 5 stud­ies of unfractioned hep­a­rin or rup­tion in atrial fibril­la­tion, there is no strong evi­dence to
LMWH bridg­ing in patients with mechan­i­cal heart valves, major guide us.
bleed­ing rates ranged from 4.39% to 10.07%. Bleeding def­i­ni­ •  After much dis­cus­sion, it is ulti­mately decided she does not
tions var­ied, pre­clud­ing the pooling of results. Although obser­ require bridg­ing ther­apy based on her mod­er­ate throm­bo­
va­tional data, this anal­y­sis sug­gests that bleed­ing risk asso­ci­ated em­bolic risk from her bileaflet mechan­i­cal aor­tic valve.
with bridg­ing in valve patients is not neg­li­gi­ble and under­scores
the need for lim­it­ing to patients at increased throm­botic risk.42
Some obser­va­tional data sug­gest that pro­phy­lac­tic dose
anticoagulation may be a via­ ble perioperative approach for VTE
select mechan­i­cal valve patients requir­ing tem­po­rary inter­rup­
tion in war­fa­rin, as well as those with newly implanted valves as
a bridge to a ther­a­peu­tic INR of 2 or more.43,44 This may mit­i­gate CLINICAL CASE (Continued)
bleed­ing risk but also pro­vi­des impor­tant post­op­er­a­tive VTE
pro­phy­laxis. However, it can­not be recommended as a rou­tine •  A year later, the patient is now going to have her other (right)
approach for all­patients until bet­ter data are avail­­able. knee replaced (fourth major ortho­pe­dic sur­gery), and this
con­sul­ta­tion is more com­plex.
•  After her last knee sur­gery, the deci­sion to not use bridg­
•  We would check her INR about 7 to 10 days prior to sur­gery ing with ther­a­peu­tic dose LMWH was interpreted as to not
and, if in the ther­a­peu­tic range, would hold war­fa­rin 5 days use any LMWH, and the patient never received any venous
pre­op­er­a­tively to allow nadir of anti­co­ag­u­lant effect at the thromboembolism pro­phy­laxis post­op­er­a­tively while war­fa­
time of the pro­ce­dure. rin rose to the appro­pri­ate INR tar­get.
•  We would have care­ ful shared deci­ sion mak­ ing with the •  She devel­oped a deep vein thrombosis, which was treated
patient and her ortho­ pe­dic sur­
geon regard­ ing the poten­ in the usual man­ ner, and she remains on war­ fa­
rin for her
tial ben­e­fits and harms of bridg­ing with LMWH because her mechan­i­cal heart valve and atrial fibril­la­tion.
throm­bo­em­bolic risk is mod­er­ate, and unlike war­fa­rin inter­
Prakash Singh Shekhawat
Perioperative man­age­ment of antithrombotics  |  525
Table 5. Additional clin­i­cal resources for periprocedural tools an anti­co­ag­ul­ant that does not require oral intake or absorp­tion
and guid­ance would be pre­ferred.

Resource Location Special sit­u­a­tions


Some addi­tional clin­i­cal sit­u­a­tions war­rant dis­cus­sion but are be-
Anticoagulation Forum Centers of https:​­/​­/acforum​­-excellence​­.org
Excellence yond the scope this arti­cle. We have sum­ma­rized these in Tables
4 and 5.45-51
Managing Anticoagulation in the http:​­/​­/mappp​­.ipro​­.org
Perioperative Period (MAPP)
Summary
Michigan Anticoagulation Quality http:​­/​­/www​­.maqi2​­.org Each year, a large num­ber of patients tak­ing OACs undergo an
Improvement Initiative (MAQI2)
inva­sive pro­ce­dure, with many requir­ing tem­po­rary inter­rup­tion
Thrombosis Canada https://thrombosiscanada​­.ca of ther­apy. This is a high-risk period for patients that requires
thought­ ful and method­ i­
cal approaches using the best avail­­
able evi­dence and expert con­sen­sus to bal­ance bleed­ing and

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/521/1851845/521burnett.pdf by guest on 13 December 2021


throm­botic risks. It is impor­tant for cli­ni­cians to be famil­iar with
The ACCP classifies VTE as (1) high, VTE within 3 months or pro­ce­dures where OAC does not require inter­rup­tion, as this
severe thrombophilia; (2) mod­er­ate, VTE within 3 to 12 months, will greatly sim­plify man­age­ment and likely min­i­mize adverse
nonsevere thrombophilia (eg, het­ero­zy­gous fac­tor V Leiden or pro­ events. For sit­u­at­ ions when inter­rup­tion is indi­cated, it is imper­
throm­bin gene muta­tion), recur­rent VTE, or active can­cer within 6 a­tive for cli­ni­cians to be famil­iar with key dif­fer­ences in phar­ma­
months; and (3) low, VTE more than 12 months ago and no other co­ki­netic prop­er­ties between DOACs and war­fa­rin, as these lead
risk fac­tors. They sug­gest bridg­ing for high risk, no bridg­ing for low to sig­nif­i­cantly dif­fer­ent perioperative man­age­ment approaches.
risk, and indi­vid­u­al­ized deci­sion mak­ing for mod­er­ate risk.19 The A step­wise sys­tem-level pro­cess, mul­ti­dis­ci­plin­ary col­lab­o­ra­tion,
Amer­i­can Society of Hematology (ASH) 2018 guide­lines on opti­mal shared deci­sion mak­ing with patients, and clear com­mu­ni­ca­tion
man­age­ment of anti­co­ag­u­lant ther­apy, which use the same VTE and doc­um ­ en­ta­tion of the plan are all­key ele­ments of antithrom-
risk strat­i­fi­ca­tion as the ACCP, dif­fer slightly, in that they sug­gest no bosis stew­ard­ship nec­es­sary for suc­cess­ful nav­i­ga­tion and im-
bridg­ing in patients with VTE at mod­er­ate risk (Table 3).40 plementation of perioperative plans.
There are no ran­dom­ized tri­als for bridg­ing in patients with
VTE who require periprocedural war­fa­rin inter­rup­tion that we Conflict-of-inter­est dis­clo­sure
are aware of. A sys­tem­atic review of 28 obser­va­tional stud­ies Allison Elaine Burnett: no rel­e­vant con­flicts of inter­est.
com­par­ing bridg­ing vs no bridg­ing in 6915 pro­ce­dures showed Bishoy Ragheb: no rel­e­vant con­flicts of inter­est.
no dif­fer­ence in VTE rates (0.7% vs 0.5%), an increase in major Scott Kaatz: research funding: Janssen, BMS, Osmosis Research,
bleed­ing (1.8% vs 0.4%), and an increase in all­bleed­ing (3.9% vs National Institutes of Health; consultancy: Janssen, BMS, Alexion/
0.4%), respec­tively. The 95% con­fi­dence inter­vals overlapped for Portola, Novartis, CSL Behring, Gilead.
VTE and major bleed­ing but did not for all­bleed­ing.35
•  There is no clear answer for this patient. She is at low risk from Off-label drug use
a purely VTE stand­point based on ACCP and ASH guide­lines. Allison Elaine Burnett: There are currently no anticoagulants ap-
•  However, we would likely bridge just on prac­ti­cal­ity of her proved specifically for bridging therapy in warfarin patients, thus
col­lec­tive risk fac­tors. any discussion on this is off-label.
•  We would ini­ti­ate perioperative bridg­ing with ther­a­peu­tic Bishoy Raghe: There are currently no anticoagulants approved
dose LMWH and hold for 24 hours prior to the pro­ce­dure. specifically for bridging therapy in warfarin patients, thus any
•  We would resume ther­a­peu­tic dose LMWH 2 to 3 days post­op­ discussion on this is off-label.
er­a­tively because knee sur­gery is a high bleed­ing risk sur­gery. Scott Kaatz: There are currently no anticoagulants approved
•  This time, how­ever, pro­phy­lac­tic dose LMWH is to be given on specifically for bridging therapy in warfarin patients, thus any
post­op­er­a­tive days 1 and 2 for VTE pre­ven­tion until ther­a­peu­tic discussion on this is off-label.
dose LMWH is started for the mod­er­ate-risk mechan­ic ­ al valve
(and will also “cover” the low-risk atrial fibril­la­tion and prior VTE). Correspondence
Allison Elaine Burnett; University of New Mexico, Health Sciences
Center, College of Pharmacy, 2211 Lomas Blvd NE, Albuquerque,
NM 87106; e-mail: aburnett@salud​­.unm​­.edu.
Postprocedure VTE pro­phy­laxis
As high­lighted in each of the case vignettes above, most if not References
all­ sur­gi­cal patients war­rant post­op­er­a­tive VTE pro­phy­laxis in the 1. Colacci M, Tseng EK, Sacks CA, Fralick M. Oral anti­co­ag­u­lant uti­li­za­tion in
interim until they can be safely increased to ther­a­peu­tic anti- the United States and United Kingdom. J Gen Intern Med. 2020;35(8):2505-
coagulation at the appro­pri­ate time (ie, 24 hours for low bleed 2507.
2. Bleeding Risk Survey. Blood clots. Accessed 24 April 2021. https:​­/​­/www​
risk pro­ce­dures, 48-72 hours for high bleed risk pro­ce­dures). This
­.stoptheclot​­.org​­/news​­/bleeding​­-risk​­-survey​­/
is termed a “step-up” approach, and the most com­monly used 3. Sherwood MW, Douketis JD, Patel MR, et al; ROCKET AF Investigators.
pro­phy­lac­tic agents are LMWH and hep­a­rin. Although DOACs Outcomes of tem­po­rary inter­rup­tion of rivaroxaban com­pared with war­
could be con­sid­ered a means of VTE pro­phy­laxis in the step- fa­rin in patients with nonvalvular atrial fibril­la­tion: results from the rivarox-
up period, patients may not imme­di­ately be ­able to tol­er­ate aban once daily, oral, direct fac­tor Xa inhi­bi­tion com­pared with vita­min K
antag­o­nism for pre­ven­tion of stroke and embolism trial in atrial fibril­la­tion
oral med­i­ca­tions. In addi­tion, some patients may have altered (ROCKET AF). Circulation. 2014;129(18):1850-1859.
absorp­tion, includ­ing those with post­op­er­a­tive ileus or patients 4. Healey JS, Eikelboom J, Douketis J, et al; RE-LY Investigators. Periprocedural
who have under­gone bariatric sur­gery. For these patients, use of bleed­ing and throm­bo­em­bolic events with dabigatran com­pared with

526  |  Hematology 2021  |  ASH Education Program


war­fa­rin: results from the ran­dom­ized eval­u­a­tion of long-term anticoagula- 26. Steffel J, Collins R, Antz M, et  al. 2021 Euro­pean Heart Rhythm Associa-
tion ther­apy (RE-LY) ran­dom­ized trial. Circulation. 2012;126(3):343-348. tion prac­ti­cal guide on the use of non-vita­min K antag­o­nist oral anti­co­ag­
5. Shaw JR, Kaplovitch E, Douketis J. Periprocedural man­age­ment of oral anti- u­lants in patients with atrial fibril­la­tion [published online 25 April 2021]. EP
coagulation. Med Clin North Am. 2020;104(4):709-726. Europace.
6. Rose AJ, Allen AL, Minichello T. A call to reduce the use of bridg­ing antico- 27. Romero J, Cerrud-Rodriguez RC, Alviz I, et al. Significant ben­e­fit of unin­
agulation. Circ Cardiovasc Qual Outcomes. 2016;9(1):64-67. ter­rupted DOACs ver­sus VKA dur­ing cath­e­ter abla­tion of atrial fibril­la­tion.
7. Bougeard A-M, Watkins B. Transitions of care in the perioperative peri- JACC Clin Electrophysiol. 2019;5(12):1396-1405.
od—a review. Clin Med (Lond). 2019;19(6):446-449. 28. Sticherling C, Marin F, Birnie D, et  al. Antithrombotic man­ age­ ment in
8. Michota F. Transitions of care in anticoagulated patients. J Multidiscip patients under­go­ing elec­tro­phys­i­o­log­i­cal pro­ce­dures: a Euro­pean Heart
Healthc. 2013;6:215-228. Rhythm Association (EHRA) posi­ tion doc­ u­
ment endorsed by the ESC
9. Steib A, Mertes P-M, Marret E, Albaladejo P, Fusciardi J. Compliance with Working Group Thrombosis, Heart Rhythm Society (HRS), and Asia Pacific
guide­lines for the perioperative man­age­ment of vita­min K antag­o­nists. Heart Rhythm Society (APHRS). Europace. 2015;17(8):1197-1214.
Thromb Res. 2014;133(6):1056-1060. 29. Burnett AE, Mahan CE, Vazquez SR, Oertel LB, Garcia DA, Ansell J. Guidance
10. Dager WE, Ansell J, Barnes GD, et al. “Reduce the like­li­hood of patient harm for the prac­ti­cal man­age­ment of the direct oral anti­co­ag­u­lants (DOACs) in
asso­ci­ated with the use of anti­co­ag­u­lant ther­apy”: Commentary from the VTE treat­ment. J Thromb Thrombolysis. 2016;41(1):206-232.
Anticoagulation Forum on the Updated Joint Commission NPSG.03.05.01 30. Douketis JD, Spyropoulos AC, Duncan J, et al. Perioperative man­age­ment

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/521/1851845/521burnett.pdf by guest on 13 December 2021


ele­ments of per­for­mance. Jt Comm J Qual Patient Saf. 2020;46(3):173-180. of patients with atrial fibril­la­tion receiv­ing a direct oral anti­co­ag­u­lant. JAMA
11. Centers for Medi­ care & Med­ ic­
aid Services. Quality pay­ ment pro­gram. Intern Med. 2019;179(11):1469-1478.
Explore mea­sures & activ­i­ties. 2021 improve­ment activ­i­ties. Anticoagulant 31. Schulman S, Carrier M, Lee AY, et al; Periop Dabigatran Study Group.
man­age­ment improve­ments. Accessed 25 April 2021. https:​­/​­/qpp​­.cms​­.gov​ Perioperative man­age­ment of dabigatran: a pro­spec­tive cohort study. Cir-
­/mips​­/explore​­-measures​­?tab=improvementActivities&py=2020 culation. 2015;132(3):167-173.
12. Kataruka A, Renner E, Barnes GD. Evaluating the role of clin­i­cal phar­ma­ 32. Godier A, Dincq A-S, Martin A-C, et al. Predictors of pre-pro­ce­dural con­cen­
cists in pre-pro­ce­dural anticoagulation man­age­ment. Hosp Pract (1995). tra­tions of direct oral anti­co­ag­u­lants: a pro­spec­tive multicentre study. Eur
2018;46(1):16-21. Heart J. 2017;38(31):2431-2439.
13. Marquez J, Togami JC, Dant CR, Herrera A, Marshik P, Burnett AE. Peri-pro­ 33. Levy JH, Ageno W, Chan NC, et al; Subcommittee on Control of Antico-
ce­dural antithrombotic man­age­ment: time to burn the bridge? J Thromb agulation. When and how to use anti­dotes for the rever­sal of direct oral
Thrombolysis. 2018;45(3):337-344. anti­co­ag­u­lants: guid­ance from the SSC of the ISTH. J Thromb Haemost.
14. Collins R, Scrimgeour A, Yusuf S, Peto R. Reduction in fatal pul­mo­nary 2016;14(3):623-627.
embolism and venous throm­bo­sis by perioperative admin­is­tra­tion of sub­ 34. Horlocker TT, Vandermeuelen E, Kopp SL, Gogarten W, Leffert LR, Benzon
cu­ta­ne­ous hep­a­rin. Overview of results of ran­dom­ized tri­als in gen­eral, HT. Regional anes­the­sia in the patient receiv­ing antithrombotic or throm­
ortho­pe­dic, and uro­logic sur­gery. N Engl J Med. 1988;318(18):1162-1173. bo­lytic ther­apy: Amer­i­can Society of Regional Anesthesia and Pain Med-
15. Baron TH, Kamath PS, McBane RD. Management of antithrombotic ther­apy icine Evidence-Based Guidelines (Fourth Edition). Reg Anesth Pain Med.
in patients under­go­ing inva­sive pro­ce­dures. N Engl J Med. 2013;368(22):​ 2018;43(3):263-309.
2113-2124. 35. Baumgartner C, de Kouchkovsky I, Whitaker E, Fang MC. Periprocedural
16. Spyropoulos AC, Douketis JD. How I treat anticoagulated patients under­ bridg­ing in patients with venous throm­bo­em­bo­lism: a sys­tem­atic review.
go­ing an elec­tive pro­ce­dure or sur­gery. Blood. 2012;120(15):2954-2962. Am J Med. 2019;132(6):722-732.e7732e7.
17. Spyropoulos AC, Brohi K, Caprini J, et al; SSC Subcommittee on Perioper- 36. Siegal D, Yudin J, Kaatz S, Douketis JD, Lim W, Spyropoulos AC. Periproce-
ative and Critical Care Thrombosis and Haemostasis of the International dural hep­ a­rin bridg­ing in patients receiv­ ing vita­min K antag­ o­nists: sys­
Society on Thrombosis and Haemostasis. Scientific and stan­dard­i­za­tion tem­atic review and meta-anal­y­sis of bleed­ing and throm­bo­em­bolic rates.
com­mit­tee com­mu­ni­ca­tion: guid­ance doc­u­ment on the periprocedural Circulation. 2012;126(13):1630-1639.
man­age­ment of patients on chronic oral anti­co­ag­u­lant ther­apy: rec­om­men­ 37. Douketis JD, Spyropoulos AC, Kaatz S, et al; BRIDGE Investigators. Periop-
da­tions for stan­dard­ized reporting of pro­ce­dural/sur­gi­cal bleed risk and erative bridg­ing anticoagulation in patients with atrial fibril­la­tion. N Engl J
patient-spe­cific throm­bo­em­bolic risk. J Thromb Haemost. 2019;17(11):1966– Med. 2015;373(9):823-833.
1972. 38. Schulman S, Hwang H-G, Eikelboom J-W, Kearon C, Pai M, Delaney J. Load-
18. Doherty JU, Gluckman TJ, Hucker WJ, et al. 2017 ACC expert con­sen­sus deci­ ing dose vs. main­te­nance dose of war­fa­rin for reinitiation after inva­sive pro­
sion path­way for periprocedural man­age­ment of anticoagulation in patients ce­dures: a ran­dom­ized trial. J Thromb Haemost. 2014;12(8):1254-1259.
with nonvalvular atrial fibril­la­tion. J Am Coll Cardiol. 2017;69(7):871–898. 39. Otto Catherine M., Nishimura Rick A., Bonow Robert O., et  al. 2020
19. Douketis JD, Spyropoulos AC, Spencer FA, et  al. Perioperative man­age­ ACC/AHA guide­line for the man­age­ment of patients with val­vu­lar heart
ment of antithrombotic ther­apy: antithrombotic ther­apy and pre­ven­tion of dis­ease: a report of the Amer­i­can College of Cardiology/Amer­i­can Heart
throm­bo­sis, 9th ed: Amer­i­can College of Chest Physicians Evidence-Based Association Joint Committee on Clinical Practice Guidelines. Circulation.
Clinical Practice Guidelines. Chest. 2012;141(2, suppl):e326S-e350S. 2021;​143(5):​e72-e227.
20. Birnie DH, Healey JS, Wells GA, et al; BRUISE CONTROL Investigators. Pace- 40. Witt DM, Nieuwlaat R, Clark NP, et  al. Amer­i­can Society of Hematology
maker or defi­bril­la­tor sur­gery with­out inter­rup­tion of anticoagulation. N 2018 guide­lines for man­age­ment of venous throm­bo­em­bo­lism: opti­mal
Engl J Med. 2013;368(22):2084-2093. man­age­ment of anticoagulation ther­apy. Blood Adv. 2018;2(22):3257-3291.
21. Di Biase L, Burkhardt JD, Santangeli P, et  al. Periprocedural stroke and 41. Kovacs MJ, Wells PS, Anderson DR, et al; PERIOP2 Investigators. Postop-
bleed­ing com­pli­ca­tions in patients under­go­ing cath­e­ter abla­tion of atrial erative low molec­u­lar weight hep­a­rin bridg­ing treat­ment for patients at
fibril­la­tion with dif­fer­ent anticoagulation man­age­ment: results from the high risk of arte­rial throm­bo­em­bo­lism (PERIOP2): dou­ble blind randomised
role of coumadin in pre­vent­ing throm­bo­em­bo­lism in atrial fibril­la­tion (AF) con­trolled trial. BMJ. 2021;373:n1205.
patients under­go­ing cath­e­ter abla­tion (COMPARE) ran­dom­ized trial. Circu- 42. Torres M. Effects of heparinoid bridg­ing in patients with mechan­i­cal heart
lation. 2014;129(25):2638-2644. valves [published online 16 Novem­ber 2020]. J Am Assoc Nurse Pract.
22. Kowalewski M, Suwalski P, Raffa GM, et al. Meta-anal­y­sis of unin­ter­rupted as 43. Mathew JG, Spyropoulos AC, Yusuf A, et  al. Efficacy and safety of early
com­pared to interrupted oral anticoagulation with or with­out bridg­ing in par­en­teral anticoagulation as a bridge to war­fa­rin after mechan­i­cal valve
patients under­go­ing cor­o­nary angi­og­ra­phy with or with­out per­cu­ta­ne­ous replace­ment. Thromb Haemost. 2014;112(6):1120-1128.
cor­o­nary inter­ven­tion. Int J Cardiol. 2016;223:186-194. 44. Hjellström L, Labaf A. Prophylactic doses of low-molec­u­lar weight hep­a­
23. Shahi V, Brinjikji W, Murad MH, Asirvatham SJ, Kallmes DF. Safety of unin­ rin as periprocedural bridg­ing ther­apy in mechan­i­cal heart valve patients.
ter­rupted war­fa­rin ther­apy in patients under­go­ing car­dio­vas­cu­lar endo- Thromb Res. 2018;163:180-184.
vascular pro­ce­dures: a sys­tem­atic review and meta-anal­y­sis. Radiology. 45. Tomaselli GF, Mahaffey KW, Cuker A, et  al. 2020 ACC expert con­sen­sus
2016;278(2):383-394. deci­sion path­way on man­age­ment of bleed­ing in patients on oral anti­co­
24. Birnie DH, Healey JS, Wells GA, et al. Continued vs. interrupted direct oral ag­u­lants. J Am Coll Cardiol. 2020;76(5):594-622.
anti­co­ag­u­lants at the time of device sur­gery, in patients with mod­er­ate 46. Cuker A, Burnett A, Triller D, et al. Reversal of direct oral anti­co­ag­u­lants:
to high risk of arte­rial thrombo-embolic events (BRUISE CONTROL-2). Eur guid­ance from the anticoagulation forum. Am J Hematol. 2019;94(6):697–
Heart J. 2018;39(44):3973-3979. 709.
25. Beyer-Westendorf J, Gelbricht V, Förster K, et al. Peri-interventional man­ 47. Filipescu DC, Stefan MG, Valeanu L, Popescu WM. Perioperative man­age­
Prakash Singh Shekhawat
age­ment of novel oral anti­co­ag­u­lants in daily care: results from the pro­ ment of antiplatelet ther­apy in non­car­diac sur­gery. Curr Opin Anaesthesiol.
spec­tive Dresden NOAC reg­is­try. Eur Heart J. 2014;35(28):1888-1896. 2020;33(3):454-462.

Perioperative man­age­ment of antithrombotics  |  527


48. Roberta R, Giuseppe T, Giuseppe M, et al. A mul­ti­dis­ci­plin­ary approach on 51. Kelkar AH, Rajasekhar A. Inferior vena cava fil­ters: a frame­work for evi­
the perioperative antithrombotic man­age­ment of patients with cor­o­nary dence-based use. Hematology Am Soc Hematol Educ Program. 2020;​
stents under­go­ing sur­gery. JACC: Cardiovasc Interv. 2018;11(5):417-434. 2020(1):619-628.
49. Cuker A, Arepally GM, Chong BH, et al. Amer­i­can Society of Hematology
2018 guide­lines for man­age­ment of venous throm­bo­em­bo­lism: hep­a­rin-
induced throm­bo­cy­to­pe­nia. Blood Adv. 2018;2(22):3360-3392.
50. Kearon C, Hirsh J. Management of anticoagulation before and after elec­tive
sur­gery. N Engl J Med. 1997;336(21):1506-1511. DOI 10.1182/hema­tol­ogy.2021000287

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/521/1851845/521burnett.pdf by guest on 13 December 2021

528  |  Hematology 2021  |  ASH Education Program


PERIOPERATIVE CONSULTATIVE HEMATOLOGY: CAN YOU CLEAR MY PATIENT FOR SURGERY ?

How to manage bleeding disorders in aging


patients needing surgery

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/529/1851841/529abou-ismail.pdf by guest on 13 December 2021


Mouhamed Yazan Abou-Ismail1 and Nathan T. Connell2,3
Division of Hematology and Hematologic Malignancies, Department of Internal Medicine, University of Utah, Salt Lake City, UT; 2Hematology
1

Division, Department of Medicine, Brigham and Women’s Hospital, Boston, MA; and 3Harvard Medical School, Boston, MA

With improvements in medical care, the life expectancy of patients with bleeding disorders is approaching that of the gen-
eral population. A growing population of older adult patients with bleeding disorders is at risk of age-related comorbidities
and in need of various elective and emergent age-related procedures. The increased risk of thrombosis and volume over-
load in older adults complicates perioperative hemostatic management. Furthermore, antithrombotic treatment such as
antiplatelet or anticoagulant therapy, which is frequently required for various cardiovascular interventions, requires a metic-
ulous individualized approach. Evidence-based guidelines for the management of aging patients with bleeding disorders
are lacking, largely due to the underrepresentation of older adult patients in clinical trials as well as the rarity of many such
bleeding disorders. We discuss the current guidelines and recommendations in the perioperative hemostatic management
of older adult patients with hemophilia and von Willebrand disease as well as other rare bleeding disorders. The optimal
management of these patients is often complex and requires a thorough multidisciplinary and individualized approach
involving hematologists, surgeons, anesthesiologists, and the specialists treating the underlying disorder.

LEARNING OBJECTIVES
• Recognize the challenges unique to the aging patient, such as age-related risks for thrombosis and volume overload
• Review the guidelines for the peri-operative hemostatic management of various bleeding disorders
• Apply an optimal approach in complex management scenarios, utilizing an individualized treatment plan and
multi-disciplinary approach

Introduction teins, have been suggested as mechanisms for the increased


With improvements in medical care, the life expectancy thrombotic risk seen in older adults (Figure 1).4-9 Chronic
of patients with bleeding disorders, such as hemophilia, cardiovascular comorbidities, such as hypertension, diabe-
is approaching that of the general population.1 As a result, tes, and hypercholesterolemia, further exacerbate this risk.
there is a growing population of older adult patients with Despite having bleeding tendencies, older adult patients with
bleeding disorders in need of various elective and emer- bleeding disorders are prone to these age-related changes
gent age-related procedures.1-3 These commonly include that contribute to thrombosis and are not considered pro-
orthopedic procedures such as joint arthroplasties and spi- tected from these complications. While the incidence of
nal surgery as well as cardiac interventions such as percuta- cardiovascular disease in hemophilia patients, for example,
neous coronary intervention (PCI), coronary artery bypass has been reported to be less than that of the general popu-
grafting (CABG), and heart valve replacement, all of which lation (15% vs 25.8%),10 events such as myocardial infarction
are increasingly prevalent with age. In addition to the sig- and stroke still occur and should be prevented.2,3,10-13 Partic-
nificant bleeding and thrombotic risks that many of these ular attention should be given to older adults with bleeding
procedures carry, several challenges may further compli- disorders undergoing surgery in order to prevent periop-
cate perioperative management in older adult patients in erative thrombotic complications as well as bleeding. This
comparison to children and young adults. necessitates the judicious use of hemostatic agents, such as
One such major challenge is the increased risk of throm- coagulation factors or hemostatic bypass agents, that may
bosis that occurs with age. Several age-related physiological subject patients to a higher risk of thrombosis.14 In addition,
changes, such as vessel wall remodeling, endothelial dys- another considerable age-related risk is that of volume over-
function, venous stasis, and an increase in procoagulant pro- load, which may occur as a complication of receiving the
Prakash Singh Shekhawat
Bleeding disorders and surgery in aging patients | 529
Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/529/1851841/529abou-ismail.pdf by guest on 13 December 2021
Figure 1. Proposed age-related mechanisms contributing to increased risk of thrombosis with age, based on Virchow’s triad. FVII,
factor VII; FVIII, factor VIII; Fg, fibrinogen; PAI-1, plasminogen activator inhibitor 1; VWF, von Willebrand factor.

large vol­umes of fresh fro­zen plasma (FFP) and plate­let trans­fu­sions Intraoperative and post­op­er­a­tive hemo­sta­sis
required in man­ag­ing cer­tain bleed­ing dis­or­ders. In the above sce­nario, metic­u­lous hemo­static sup­port is cru­cial
Evidence-based guide­ lines for the man­ age­
ment of aging for the suc­cess of the sur­gi­cal pro­ce­dure. The over­all hemo­static
patients with bleed­ ing dis­ or­
ders are lacking, largely due to man­age­ment and avail­­able agents vary sig­nif­i­cantly among
the under­rep­re­sen­ta­tion of older adult patients in clin­i­cal tri­als dif­fer­ent bleed­ing dis­or­ders, and we dis­cuss each sep­a­rately
as well as the rar­ity of many bleed­ing dis­or­ders. The opti­mal below. However, regard­less of the type of bleed­ing dis­or­der, the
perioperative man­age­ment of these patients is often com­plex man­age­ment con­cepts are sim­i­lar: to pro­vide effec­tive and suf­
and requires a thor­ough mul­ti­dis­ci­plin­ary approach that involves fi­cient hemo­static sup­port to pre­vent sur­gi­cal-related bleed­ing,
hema­tol­o­gists, sur­geons, anes­the­si­­ol­o­gists, and the spe­cial­ists to sup­port wound healing, and to avoid side effects and com­
treating the under­ly­ing dis­or­der.15 pli­ca­tions related to the ther­apy used. The World Federation of
Hemophilia (WFH) 2020 guide­ lines rec­
om­ mend that patients
with hemo­philia requir­ing sur­gery should always be man­aged
at or in con­sul­ta­tion with rec­og­nized HTCs with appro­pri­ate
CLINICAL CASE 1 hema­to­logic sup­port, reli­able lab­o­ra­tory mon­i­tor­ing, and avail­­
An 88-year-old man with severe hemo­philia A (HA; base­line fac­tor able and suf­fi­cient clot­ting fac­tor con­cen­trates.16 This con­cept
VIII [FVIII] activ­ity level <1%, with­out inhib­i­tors) has severe, debil­ applies to patients with any bleed­ing dis­or­der. The HTC should
i­tat­ing left hip arthrop­a­thy. He is oth­er­wise healthy and does not be supported by a mul­ti­dis­ci­plin­ary team that includes nurses,
have any other med­i­cal comorbidities. He self-admin­is­ters pro­ social work­ers, and phys­i­cal ther­a­pists famil­iar with the needs of
phy­lac­tic fac­tor replace­ment 3 times a week and receives his care hemo­philia patients under­go­ing sur­gery.17 As in the case above,
within a hemo­philia treat­ment cen­ter (HTC). He is eval­u­ated by pro­ce­dures should be performed elec­tively when pos­si­ble, early
an ortho­pe­dic sur­geon and deemed a fit can­di­date for elec­tive in the week, and early in the day by expe­ri­enced sur­geons and
left total hip arthroplasty. The hema­tol­o­gist is consulted to assist anes­the­si­­ol­o­gists.15 Preoperative in vivo recov­ ery stud­ies are
with perioperative man­age­ment to pre­vent bleed­ing com­pli­ca­ used to eval­u­ate the phar­ma­co­ki­netic prop­er­ties of coag­u­la­tion
tions. The patient is treated with 50 U/kg of anti­he­mo­philic fac­tor fac­tors, which may dif­fer based on the prod­uct and the patient.
(Advate) before sur­gery and achieves a peak FVIII level of 108% and In hemo­philia patients this allows for a more pre­cise esti­ma­tion
good post­op­er­at­ive hemo­sta­sis. He is then maintained on daily of peak FVIII and fac­tor IX (FIX) lev­els and can help tai­lor the dos­
fac­tor replace­ment for the rest of the week and begins phys­ic ­ al ing and fre­quency of fac­tor replace­ment according to the need
reha­bil­i­ta­tion. He is not placed on pro­phy­lac­tic anticoagulation. of the indi­vid­ual patient, espe­cially before major sur­gery. The
pres­ence of inhib­i­tors should also be assessed prior to sur­gery.

530  |  Hematology 2021  |  ASH Education Program


Table 1. Suggested fac­tor replace­ment sched­ule for older adult per­sons with hemo­philia A or B with­out inhib­i­tors based on WFH
guide­lines

Hemophilia A Hemophilia B
Indication Target peak lev­els (%) Duration Target peak lev­els (%) Duration
Minor sur­gery 50-80 Day of sur­gery 50-80 Day of sur­gery
30-80 Days 1-5 30-80 Days 1-5
Major sur­gery 80-100 Day of sur­gery 60-80 Day of sur­gery
60-80 Days 1-3 40-60 Days 1-3
40-60 Days 4-6 30-50 Days 4-6
30-50 Days 7-14 20-40 Days 7-14

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/529/1851841/529abou-ismail.pdf by guest on 13 December 2021


Adapted with per­mis­sion from Srivastava et al.16

Hemophilia A dence of these events remains low and both rFVIIa and aPCC are
The hemo­static agents avail­­able for HA include desmopressin and gen­er­ally con­sid­ered safe in older adults,27,30-32 patients should
FVIII replace­ment. Desmopressin may be con­sid­ered in patients be very cau­tiously mon­i­tored for throm­bo­sis. The lack of reli­able
with mild HA under­go­ing minor sur­gery in whom a response to ther­a­peu­tic mon­i­tor­ing param­e­ters for bypass agents fur­ther
desmopressin has been dem­on­strated, par­tic­u­larly when the cost com­pli­cates their opti­mal dos­ing strat­egy, and metic­u­lous clin­
or devel­op­ment of inhib­i­tors due to expo­sure to FVIII prod­ucts is i­cal eval­u­a­tion of bleed­ing and throm­bo­sis is cru­cial. Data from
of con­cern. Issues with the use of desmopressin include free water the Euro­ pean Acquired Hemophilia Registry sug­ gests a sim­ i­
reten­tion, hyponatremia, fluid shifts, and exac­er­ba­tion of car­dio­ lar safety and effi­cacy pro­file for both rFVIIa and aPCC27; how­
vas­cu­lar dis­ease. For those rea­sons it should be used with con­sid­ ever, par­al­lel use of dif­fer­ent bypass agents should be avoided
er­able cau­tion in older adult patients prone to vol­ume over­load because it may con­fer a higher risk for throm­bo­sis.33 Suggested
and car­dio­vas­cu­lar mor­bid­ity and is contraindicated in patients dos­ing of bypass agents is sum­ma­rized in Table 2.
with active car­dio­vas­cu­lar dis­ease or sei­zure dis­or­der.16,18 A sim­i­lar approach applies to acquired HA, an auto­im­mune
FVIII replace­ment ther­apy is the treat­ment of choice for all­ dis­or­der caused by antibodies against FVIII that is more com­mon
other patients with­out high-titer inhib­i­tors, such as the patient in older adults.34 In addi­tion to bypass agents, these patients
in our case above. Suggested plasma FVIII tar­ get lev­ els are also have the option of recom­bi­nant por­cine FVIII as a hemo­
pro­vided in Table 1, adapted from WFH guide­lines.16 However, static agent.34,35 While the recommended ini­tial dose of recom­
high-qual­ity data guid­ing this prac­tice are lacking.19 Addition- bi­nant por­cine FVIII is 200 U/kg, it has been suggested that an
ally, patients under­go­ing car­diac pro­ce­dures indi­cated for ini­tial dose of 100 U/kg may be suf­fi­cient for most patients and
antithrombotic treat­ment may require an extended dura­tion of can be con­sid­ered for those at risk of throm­bo­sis.
fac­tor replace­ment to allow for safe admin­is­tra­tion of the for­mer.
This is discussed sep­a­rately below. Perioperative thromboprophylaxis
The WFH rec­om­mends against the rou­tine use of phar­ma­co­logic
Hemophilia B thromboprophylaxis in patients with HA or HB under­go­ing major
While the over­all clin­i­cal man­i­fes­ta­tions and man­age­ment non­car­diac sur­gery.16 For ortho­pe­dic pro­ce­dures, such as the case
approaches in hemo­philia B (HB) are sim­i­lar to those in HA, a above, the Amer­i­can Academy of Orthopaedic Surgeons and the
few dif­fer­ences should be noted. In con­trast to HA, the bleed­ Amer­i­can College of Chest Physicians also sug­gest for­go­ing the
ing phe­no­type in HB may be milder,19-25 and the recommended rou­tine use of antithrombotic agents in patients with bleed­ing
tar­get lev­els in the WFH 2020 guide­lines are lower for major sur­ dis­or­ders, as does the WFH, with the excep­tion of high plasma
gery (Table 1).16 The dos­ing fre­quency should also con­sider the lev­els of coag­u­la­tion fac­tors.36 As pre­vi­ously men­tioned, how­ever,
lon­ger half-life of FIX. Patients with HB do not have desmopressin older adult patients with bleed­ing dis­or­ders are not nec­es­sar­ily
as an avail­­able option, and FIX replace­ment is the treat­ment of protected from throm­botic out­comes, includ­ing venous or arte­
choice for major pro­ce­dures. While the inci­dence of inhib­i­tors rial throm­bo­sis, and this risk may increase with age due to age-re-
is much rarer in HB than in HA,26 their pres­ence should also be lated prothrombotic phys­i­o­log­i­cal changes (Figure 1). In older
assessed prior to sur­gery. patients, an indi­vid­u­al­ized risk/ben­e­fit assess­ment is nec­es­sary
when addi­tional throm­botic risk fac­tors are pres­ent, espe­cially
Patients with inhib­i­tors in patients with corrected fac­tor lev­els after ortho­pe­dic sur­gery.
Hemophilia patients with high-titer inhib­ i­
tors to FVIII, FIX, or
fac­tor XI (FXI) require the use of bypass agents, mainly recom­
bi­nant fac­tor VIIa (rFVIIa) or acti­vated pro­throm­bin com­plex con­
cen­trate (aPCC), which pose an addi­tional risk for throm­bo­sis, CLINICAL CASE 2
espe­cially in older adults. Several stud­ies note that the risk of A 79-year-old woman with hyper­ ten­
sion, dia­
be­ tes, and type
throm­bo­sis due to these agents increases with age, and arte­ 2A von Willebrand dis­ ease (VWD; VWF [von Willebrand fac­
rial throm­bo­sis is a par­tic­u­lar con­cern.27-29 While the over­all inci­ tor]:Ag = 35, VWF:RCo = 15, FVIII:C = 110) is sched­uled for elec­tive
Prakash Singh Shekhawat
Bleeding dis­or­ders and sur­gery in aging patients  |  531
Table 2. Suggested dos­ing reg­i­mens of bypass agents in hemo­philia patients with inhib­i­tors

Agent Dosing for minor sur­gery Dosing for major sur­gery Comments
rFVIIa 90 µg/kg/dose imme­di­ately before 90 µg/kg/dose imme­di­ately before • Preferred agent for patients with HB
sur­gery and every 2 hours for 2 sur­gery and every 2 hours for 2 days. and high-titer inhib­i­tors per WFH
days and then every 2-6 hours until Then every 2-3 hours for 5 days, then guide­lines since aPCC con­tains FIX
healed. every 4 hours until days 7-10, then and may cause or worsen an aller­gic
every 6 hours until days 14-21 reac­tion.
• For patients with FIX defi­ciency, low
dose (15-30 µg/kg) in com­bi­na­tion
with TXA has been shown to be
effec­tive for major sur­gery in lim­ited
stud­ies and can be con­sid­ered for
those at risk of throm­bo­sis.33

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/529/1851841/529abou-ismail.pdf by guest on 13 December 2021


Activated pro­throm­bin 50-100 U/kg imme­di­ately before 50-100 U/kg imme­di­ately before sur­ • Maximum: 100 units/kg/dose; 200
con­cen­trate com­plex sur­gery and then every 6-12 hours gery, then every 6-12 hours units/kg/d).
• Less pre­ferred for HB patients with
high-titer inhib­i­tors per WFH guide­
lines.

cho­le­cys­tec­tomy. She is admit­ted and receives 60 U/kg of VWF increases in VWF.37-42 The 2021 guide­ lines sug­
gest reconsider-
con­cen­ trate prior to sur­ gery, achiev­ing VWF:RCo  = 104, and ing the diag­no­sis in these patients, as opposed to remov­ing it.43
FVIII:C = 225. Her sur­gi­cal course is uncom­pli­cated, but the next Whether the nor­mal­i­za­tion of VWF lev­els in older patients results
morn­ing she com­plains of substernal chest pain. She is found to in an ame­lio­ra­tion of bleed­ing symp­toms has not been established.
have non-ST ele­va­tion myo­car­dial infarc­tion. With her VWF activ­ While that may be pos­si­ble in some patients, it has been sug-
ity lev­els still nor­mal, she undergoes PCI and is found to have gested that supranormal VWF lev­els may be required in oth­ers.44
severe tri­ ple ves­sel dis­ease. She is recommended for elec­ tive Those with nor­mal­ized VWF lev­els in whom bleed­ing symp­toms
CABG. The patient’s man­age­ment is ­discussed in a mul­ti­dis­ci­plin­ have resolved may be harmed by the unnec­ es­sary admin­
is­tra­
ary approach involv­ing car­di­­ol­ogy, car­dio­tho­racic sur­gery, and tion of VWF con­cen­trates, espe­cially if they have car­dio­vas­cu­lar
hema­tol­ogy. risk fac­tors.44 There are cur­rently insuf­fic
­ ient data to guide opti­mal
man­age­ment in these cases. An indi­vid­u­al­ized risk/ben­e­fit assess­
ment is cru­cial in patients with nor­mal­ized VWF lev­els, with care­ful
Von Willebrand dis­ease clin­i­cal eval­u­a­tion of bleed­ing and throm­botic risk fac­tors.
The 2021 guide­lines by the Amer­i­can Society of Hematology, Acquired von Willebrand syn­drome may result from the shear­
International Society of Thrombosis and Hemostasis, National ing of  VWF (eg, across a ste­notic car­diac valve or through mechan­
Hemophilia Foundation, and WFH rec­om­mend a goal VWF activ­ i­cal cir­cu­la­tory sup­port devices) or less com­monly may arise as an
ity and FVIII level of >50% for at least 3 days for major sur­ger­ies auto­im­mune dis­or­der with antibodies directed against VWF, often
using VWF/FVIII con­cen­trates.18 This is often extended to a dura­ seen in patients with lymphoproliferative dis­or­ders and plasma
tion of 7 to 14 days based on the indi­vid­ual type of sur­gery. In cell dys­cra­sias. When due to an auto­an­ti­body, acquired von Wil-
older adults, spe­cial pre­cau­tion must be taken to avoid overdos- lebrand syn­drome can be effec­tively man­aged with intra­ve­nous
ing of fac­tor replace­ment, as prolonged supraphysiologic FVIII immune glob­ u­lin in addi­
tion to fac­tor replace­ ment for major
lev­els may lead to throm­botic com­pli­ca­tions. Available options sur­gery, as well as the use of continuous-infusion factor replace-
include plasma-derived for­ mu­la­
tions of VWF/FVIII as well as ment.45,46,47 In older adult patients with a risk of throm­bo­sis or renal
recom­bi­nant VWF when avail­­able. Older adult patients may have dys­func­tion, approaches such as ther­a­peu­tic plasma exchange,
nor­mal or ele­vated base­line FVIII lev­els, and this should be taken rFVIIa, and TXA have been uti­lized, although data are lacking.45
into con­sid­er­ation when choos­ing the appro­pri­ate VWF replace­
ment prod­ uct. This includes plasma-derived prod­ ucts with a Cardiovascular inter­ven­tions
higher VWF to FVIII ratio, such as Humate-P, or dose adjust­ment Percutaneous cor­o­nary inter­ven­tion
of con­com­i­tant FVIII replace­ment given with recom­bi­nant VWF. PCI is a com­mon pro­ce­dure among older adult patients, and its
Patients with type 1 VWD respon­sive to desmopressin with­out chal­lenges include the need for intraoperative heparinization
active car­dio­vas­cu­lar dis­ease may be con­sid­ered for desmopressin as well as post­op­er­a­tive antiplatelet ther­apy. In patients with
for minor sur­gery, with a goal VWF level >50 per the 2021 guide­ bleed­ing dis­or­ders, a best effort should be made to min­i­mize
lines.18 The same risks and pre­cau­tions for desmopressin described the dura­tion of dual antiplatelet ther­apy (DAPT). Earlier rec­om­
under HA above apply for VWD. Close mon­i­tor­ing for throm­botic men­da­tions favored the use of bare metal stents over drug-elut­
com­pli­ca­tions is nec­es­sary for older adult patients, espe­cially if ing stents to limit the dura­tion of DAPT to 1 month.48,49 However,
con­com­i­tant tranexamic acid (TXA) is given. Patients with mild more recent data sug­gest that 1 month of DAPT may be con­
type 1 VWD under­go­ing minor muco­sal pro­ce­dures may be con­ sid­ered with newer-gen­er­a­tion drug-elut­ing stents with biore-
sid­ered for TXA alone in some cases,18 which would min­i­mize the sorbable poly­mers,50-52 which was the approach reported in a
risks asso­ci­ated with fac­tor replace­ment or desmopressin. recent case series of hemo­philia patients under­go­ing PCI.53 The
Several stud­ies on patients with type 1 VWD have dem­on­strated indi­vid­ual cor­o­nary anat­omy, risks of restenosis, and bleed­ing
that VWF lev­els may nor­mal­ize with age due to phys­i­o­log­i­cal pro­pen­sity should all­ be fac­tored into a mul­ti­dis­ci­plin­ary deci­

532  |  Hematology 2021  |  ASH Education Program


sion-mak­ing pro­cess. Due to the bleed­ing risk of DAPT, patients acquired coagulopathy. In high-risk patients, off-pump var­i­a­tions
with bleed­ing dis­or­ders may require pro­phy­lac­tic hemo­static of such pro­ce­dures should be con­sid­ered, or less inva­sive alter­
ther­apy through­out the dura­tion of DAPT, even if they had not na­tives such as multivessel PCI in lieu of CABG. As with PCI, an
been receiv­ ing it before. For hemo­ philia patients, the WFH effort should be made to min­i­mize the dura­tion of DAPT when
guide­lines rec­om­mend trough lev­els of 15% to 30% of the defi­ pos­si­ble by weighing the risks and ben­e­fits of shorter dura­tions.
cient fac­tor dur­ing the dura­tion of DAPT and lev­els of 1% to 5% In patients requir­ing valve replace­ment, bioprosthetic valves
dur­
ing sin­ gle antiplatelet agent ther­ apy.16 Similarly, the 2021 should be used instead of mechan­i­cal valves that require indef­i­
VWD guide­lines rec­om­mend pro­phy­laxis with VWF con­cen­trate nite anticoagulation. An approach uti­liz­ing low-molec­u­lar-weight
dur­ing DAPT in patients with VWD and a severe bleed­ing phe­no­ hep­a­rin for 10 days post­op­er­a­tively followed by 3 months of
type.18 In the patient above under­go­ing PCI, VWF:RCo lev­els >30 vita­min K antag­o­nist, with con­com­i­tant coag­u­la­tion fac­tor cor­
may be con­sid­ered through­out the dura­tion of DAPT. rec­tion through­out that dura­tion, has been suggested for hemo­
philia patients.54,55 A sim­i­lar approach can be implemented in
Cardiac sur­gery patients with other bleed­ing dis­or­ders, with cor­rec­tion of the

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/529/1851841/529abou-ismail.pdf by guest on 13 December 2021


Data on car­dio­vas­cu­lar pro­ce­dures such as CABG, valve replace­ hemo­static defect applied through­out the dura­tion of anticoag-
ment, or device implan­ta­tion in patients with bleed­ing dis­or­ders ulation. An addi­tional con­sid­er­ation in HB patients is the height­
are lim­ited. Management is more com­plex than other pro­ce­dures ened sever­ity of the dis­ease that can occur as a result of vita­min
due to the fre­quent need for post­op­er­a­tive antithrombotic ther­ K antag­o­nist ther­apy lead­ing to decreased FIX lev­els, as well as
apy or the need for car­dio­pul­mo­nary bypass in cer­tain cases. the inter­fer­ence with inter­na­tional nor­mal­ized ratio mon­i­tor­ing
Procedures that may require car­dio­pul­mo­nary bypass, such as caused by FIX replace­ment.2
CABG, con­sti­tute sev­eral high risks for patients with bleed­ing dis­ The lit­er­a­ture sug­gests that despite the major hemo­static
or­ders, includ­ing the need for sys­temic anticoagulation as well as chal­lenges of var­i­ous car­diac inter­ven­tions, care­ful, mul­ti­dis­
the pos­si­ble occur­rence of con­sump­tive coagulopathies.54 Con- ci­
plin­ary plan­ ning can help achieve opti­ mal out­ comes with
tinuous replace­ment of the defi­cient fac­tor may bet­ter main­tain min­i­mal mor­bid­ity and mor­tal­ity in patients with bleed­ing dis­
steady lev­els com­pared to IV boluses, and addi­tional hemo­static or­ders.53,54,56-60 However, these data may be sub­ject to poten­tial
prod­ucts such as FFP or plate­lets may be nec­es­sary in cases of pub­li­ca­tion bias, and older adult patients are under­rep­re­sented.

Table 3. Hemostatic man­age­ment of rare bleed­ing dis­or­ders and pre­cau­tions in older adult patients

Recommended hemo­static Recommended hemo­static Suggested tar­get trough


Bleeding dis­or­der treat­ment for minor sur­gery treat­ment for major sur­gery level of defi­cient fac­tor
Rare fac­tor deficiencies
 II FFP (15-20 mL/kg) FFP (15-20 mL/kg) >30%
PCC* (20-30 U/kg)
 V FFP (15-20 mL/kg) FFP (15-20 mL/kg) >20%
  V + VIII FFP (15-20 mL/kg) FFP (15-20 mL/kg) Factor V: >20% FVIII:
as in Table 1
 VII rFVIIa (15-30 µg/kg rFVIIa (15-30 µg/kg every 4-6 hours) >20%
every 4-6 hours) PCC* (20-30 mL/kg)
 X Factor X con­cen­trate (10-20 U/kg) Factor X con­cen­trate (10-20 U/kg) >20%
FFP 15-20 mL/kg FFP (15-20 mL/kg)
PCC* (20-30 U/kg)
 XIII Plasma-derived fac­tor XIII con­cen­trate Plasma-derived fac­tor XIII con­cen­trate 5%
(40 U/kg) (40 U/kg)
Recombinant fac­tor XIII con­cen­trate Recombinant fac­tor XIII con­cen­trate
(35 U/kg) (35 U/kg)
Cryoprecipitate (1 bag per 10 kg) Cryoprecipitate (1 bag per 10 kg)
Other rare bleed­ing dis­or­ders
  Platelet func­tion defects Intravenous or oral TXA (500-1000 mg Platelet trans­fu­sions (4-6 units)
every 6-12 hours) Bypass agents†
Desmopressin
 Hypo/dysfibrinogenemia† Cryoprecipitate (1 bag per 10 kg) Cryoprecipitate (1 bag per 10 kg) >100 mg/dL
Fibrinogen con­cen­trate (20-30 mg/kg) Fibrinogen con­cen­trate (20-30 mg/kg)
FFP (15-20 mL/kg)
  Disorders of fibri­no­ly­sis Intravenous or oral TXA (500-1000 mg Intravenous TXA (10-15 mg/kg)
every 6-12 hours) every 6-12 hours
*After PCC treat­ment, fac­tors II, VI, IX, and X should not exceed 150%.
†Dysfibrinogenemia often pres­ents with a throm­botic phe­no­type. Hypofibrinogenemia and afibrinogenemia usu­ally pres­ent with bleed­ing, although
both venous and arte­rial throm­bo­sis can occur. Replacement rec­om­men­da­tions apply to patients with a bleed­ing phe­no­type, and con­com­i­tant
antithrombotic pro­phy­laxis may be nec­es­sary in some, par­tic­u­larly older adults.
Adapted with per­mis­sion from Mannucci et al.67
Prakash Singh Shekhawat
Bleeding dis­or­ders and sur­gery in aging patients  |  533
Management of such patients under­ go­
ing car­
diac inter­ ven­ Nathan T. Connell: The use of bypass agents mentioned in this
tions requires a metic­u­lous, indi­vid­u­al­ized, and mul­ti­dis­ci­plin­ary article, namely aPCC and rFVIIa, is considered off-label in various
approach, with care­ful mon­i­tor­ing of hemo­static param­e­ters. rare hematologic conditions due to paucity of data.

Correspondence
Nathan T. Connell, Hematology Division, Brigham and Women’s
CLINICAL CASE 3 Hospital, SR322, 75 Francis St, Bos­ton, MA 02115; e-mail: ntcon-
An 86-year-old man is eval­ua ­ ted by a urol­o­gist for con­sid­er­ation nell@bwh​­.harvard​­.edu.
of transurethral resec­tion of the pros­tate for a new diag­no­sis of
early-stage pros­tate can­cer. He has a his­tory of con­gen­i­tal FXI defi­ References
ciency, and his most recent tests show an FXI level of 24%. He has 1. Hassan S, Monahan RC, Mauser-Bunschoten EP, et al. Mortality, life expec­
a his­tory of hyper­ten­sion, chronic kid­ney dis­ease, aor­tic ste­no­sis, tancy, and causes of death of per­sons with hemo­philia in the Netherlands
2001-2018. J Thromb Haemost. 2021;19(3):645-653.
and advanced heart fail­ure with reduced ejec­tion frac­tion. Because

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/529/1851841/529abou-ismail.pdf by guest on 13 December 2021


2. Mannucci PM, Schutgens RE, Santagostino E, Mauser-Bunschoten EP. How
of his prior his­tory of cir­cu­la­tory over­load with FFP, the option of I treat age-related morbidities in elderly per­sons with hemo­philia. Blood.
TXA in com­bi­na­tion with sin­gle low-dose rFVIIa is discussed. 2009;114(26):5256-5263.
3. Franchini M, Tagliaferri A, Mannucci PM. The man­age­ment of hemo­philia in
elderly patients. Clin Interv Aging. 2007;2(3):361-368.
4. Engbers MJ, Van Hylckama Vlieg A, Rosendaal FR. Venous throm­bo­sis in
FXI defi­ciency
the elderly: inci­dence, risk fac­tors and risk groups. J Thromb Haemost.
Compared to HA and HB, FXI defi­ciency has its own unique chal­ 2010;8(10):2105-2112.
lenges in man­age­ment. The poor cor­re­la­tion between FXI lev­els 5. Nurmohamed MT, Büller HR, ten Cate JW. Physiological changes due to
and bleed­ing phe­no­type lim­its the abil­ity to pre­dict bleed­ing out­ age: impli­ca­tions for the pre­ven­tion and treat­ment of throm­bo­sis in older
comes.61 In gen­eral, any FXI level <20% is con­sid­ered severe.61 The patients. Drugs Aging. 1994;5(1):20-33.
6. Yamamoto K, Takeshita K, Kojima T, Takamatsu J, Saito H. Aging and
lack of FXI con­cen­trates in the United States neces­si­tates the use plas­min­o­gen acti­va­tor inhib­i­tor-1 (PAI-1) reg­u­la­tion: impli­ca­tion in the
of FFP infu­sions (usu­ally 10-20 mL/kg) to replace FXI periopera- path­o­gen­e­sis of throm­botic dis­or­ders in the elderly. Cardiovasc Res.
tively or the use of bypass agents or antifibrinolytic agents.59,61-63 2005;66(2):276-285.
In older adult patients, large vol­umes of FFP are a major risk fac­ 7. Kiechl S, Willeit J; Bruneck Study Group. The nat­u­ral course of ath­ero­scle­ro­
sis, II: vas­cu­lar remodeling. Arterioscler Thromb Vasc Biol. 1999;19(6):1491-
tor for cir­cu­la­tory over­load, espe­cially in those with valve dis­ease
1498.
or con­ges­tive heart fail­ure. In such patients, major sur­gery would 8. Balleisen L, Bailey J, Epping PH, Schulte H, van de Loo J. Epidemiological
require thor­ough fac­tor replace­ment and large vol­umes of plasma, study on fac­tor VII, fac­tor VIII and fibrin­o­gen in an indus­trial pop­u­la­tion, I:
which may pose a substantial risk. Ther­a­peu­tic plasma exchange base­line data on the rela­tion to age, gen­der, body-weight, smok­ing, alco­
hol, pill-using, and men­o­pause. Thromb Haemost. 1985;54(2):475-479.
has been uti­lized as a method to over­come this risk and should be
9. Sagripanti A, Carpi A. Natural anti­co­ag­u­lants, aging, and throm­bo­em­bo­
con­sid­ered as a poten­tially safer method of fac­tor cor­rec­tion.64,65 lism. Exp Gerontol. 1998;33(7-8):891-896.
The opti­mal tar­get FXI level is unknown; how­ever, lev­els of >45% 10. Sood SL, Cheng D, Ragni M, et al. A cross-sec­tional anal­y­sis of car­dio­vas­
have been recommended for major sur­gery, with con­sid­er­ation cu­lar dis­ease in the hemo­philia pop­u­la­tion. Blood Adv. 2018;2(11):1325-
of antifibrinolytic agents in areas of high fibri­no­lytic activ­ity, such 1333.
11. Girolami A, Ruzzon E, Fabris F, Varvarikis C, Sartori R, Girolami B. Myocardial
as the blad­der, endo­me­trium, and oro­phar­ynx.61 For patients who
infarc­tion and other arte­rial occlu­sions in hemo­philia a patients: a car­di­o­
can­not receive blood prod­ucts or are at risk of cir­cu­la­tory over­ log­i­cal eval­u­a­tion of all­ 42 cases reported in the lit­er­a­ture. Acta Haematol.
load, as in the patient above, the use of a sin­gle low-dose rFVIIa 2006;116(2):120-125.
(Table 2) in com­bi­na­tion with TXA has been shown to be effec­tive 12. Aledort LM. Comparative throm­ botic event inci­ dence after infu­ sion of
recom­bi­nant fac­tor VIIa ver­sus fac­tor VIII inhib­i­tor bypass activ­ity. J Thromb
in lim­ited stud­ies.66
Haemost. 2004;2(10):1700-1708.
13. Srámek A, Kriek M, Rosendaal FR. Decreased mor­tal­ity of ischaemic heart
Other rare bleed­ing dis­or­ders dis­ease among car­ ri­
ers of haemophilia. Lancet. 2003;362(9381):351-
Very lim­ited data exist to guide the opti­mal perioperative man­ 354.
age­ment of older adults with rare bleed­ing dis­or­ders.66 The avail­ 14. O’Donnell JS, Lavin M. Perioperative man­age­ment of patients with von Wille-
brand dis­ease. Hematology Am Soc Hematol Educ Program. 2019;2019(1):
abil­ity of reli­able lab­o­ra­tory test­ing and appro­pri­ate hemo­static
604-609.
prod­ucts is nec­es­sary in the man­age­ment of these patients as 15. Mensah PK, Gooding R. Surgery in patients with inherited bleed­ing dis­or­
guided by a hemo­sta­sis expert. Table 3 sum­ma­rizes some of the ders. Anaesthesia. 2015;70(suppl 1):112-120, e39-40.
con­cerns that may arise in using spe­cific hemo­static agents in 16. Srivastava A, Santagostino E, Dougall A, et al; WFH Guidelines for the Man-
each of these dis­or­ders in older adult patients, and nec­es­sary agement of Hemophilia pan­el­ists and coau­thors. WFH guide­lines for the
man­age­ment of hemo­philia, 3rd edi­tion. Haemophilia. 2020;26 (suppl 6):1-
pre­cau­tions should be implemented. 158.
17. Rodríguez-Merchán EC. The role of ortho­pae­dic sur­gery in haemophilia:
Conflict-of-inter­est dis­clo­sure cur­rent ratio­nale, indi­ca­tions and results. EFORT Open Rev. 2019;4(5):165-
Mouhamed Yazan Abou-Ismail: no com­pet­ing finan­cial inter­ests 173.
18. Connell NT, Flood VH, Brignardello-Petersen R, et al. ASH ISTH NHF WFH
to declare.
2021 guide­lines on the man­age­ment of von Willebrand dis­ease. Blood Adv.
Nathan T. Connell: no com­pet­ing finan­cial inter­ests to declare. 2021;5(1):301-325.
19. Coppola A, Windyga J, Tufano A, Yeung C, Di Minno MN. Treatment for
Off-label drug use pre­vent­ing bleed­ing in peo­ple with haemophilia or other con­gen­i­tal
Mouhamed Yazan Abou-Ismail: The use of bypass agents men- bleed­ing dis­or­ders under­go­ing sur­gery. Cochrane Database Syst Rev.
2015;(2):Cd009961.
tioned in this article, namely aPCC and rFVIIa, is considered 20. Ludlam CA, Lee RJ, Prescott RJ, et al. Haemophilia care in cen­tral Scotland
off-label in various rare hematologic conditions due to paucity 1980-94, I: demo­graphic char­ac­ter­is­tics, hos­pi­tal admis­sions and causes
of data. of death. Haemophilia. 2000;6(5):494-503.

534  |  Hematology 2021  |  ASH Education Program


21. Soucie JM, Cianfrini C, Janco RL, et  al. Joint range-of-motion lim­i­ta­tions 45. Abou-Ismail MY, Rodgers GM, Bray PF, Lim MY. Acquired von Willebrand
among young males with hemo­philia: prev­a­lence and risk fac­tors. Blood. syn­drome in mono­clo­nal gammopathy—a scop­ing review on hemo­static
2004;103(7):2467-2473. man­age­ment. Res Pract Thromb Haemost. 2021;5(2):356-365.
22. Schulman S, Eelde A, Holmström M, Ståhlberg G, Odeberg J, Blombäck 46. Tiede A. Diagnosis and treat­ment of acquired von Willebrand syn­drome.
M. Validation of a com­pos­ite score for clin­i­cal sever­ity of hemo­philia. J Thromb Res. 2012;130(suppl 2):S2-S6.
Thromb Haemost. 2008;6(7):1113-1121. 47. Meizlish ML, et al. A neutrophil activation signature predicts critical illness
23. Santagostino E, Mancuso ME, Tripodi A, et al. Severe hemo­philia with mild and mortality in COVID-19. Blood Adv. 2021;5(14):2813-2816.
bleed­ing phe­no­type: molec­u­lar char­ac­ter­iza­tion and global coag­u­la­tion 48. Jabbar AY, Baydoun H, Janbain M, Ferdinand KC. Current con­cepts in the
pro­file. J Thromb Haemost. 2010;8(4):737-743. man­age­ment of sta­ble ische­mic heart dis­ease and acute cor­o­nary syn­
24. Nagel K, Walker I, Decker K, Chan AK, Pai MK. Comparing bleed fre­quency drome in patients with hemo­philia. Ann Transl Med. 2018;6(15):299.
and fac­tor con­cen­trate use between haemophilia A and B patients. Hae- 49. Staritz P, de Moerloose P, Schutgens R, Dolan G; ADVANCE Working Group.
mophilia. 2011;17(6):872-874. Applicability of the Euro­pean Society of Cardiology guide­lines on man­
25. Mahlangu J, Powell JS, Ragni MV, et al; A-LONG Investigators. Phase 3 study age­ment of acute cor­o­nary syn­dromes to peo­ple with haemophilia—an
of recom­bi­nant fac­tor VIII Fc fusion pro­tein in severe hemo­philia A. Blood. assess­ment by the ADVANCE Working Group. Haemophilia. 2013;19(6):833-
2014;123(3):317-325. 840.
26. Ehrenforth S, Kreuz W, Scharrer I, et al. Incidence of devel­op­ment of fac­tor 50. Varenne O, Cook S, Sideris G, et al; SENIOR Investigators. Drug-elut­ing

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/529/1851841/529abou-ismail.pdf by guest on 13 December 2021


VIII and fac­tor IX inhib­i­tors in haemophiliacs. Lancet. 1992;339(8793):594- stents in elderly patients with cor­o­nary artery dis­ease (SENIOR): a ran-
598. domised sin­gle-blind trial. Lancet. 2018;391(10115):41-50.
27. Baudo F, Collins P, Huth-Kühne A, et al; Euro­pean Acquired Haemophilia 51. Urban P, Meredith IT, Abizaid A, et al; LEADERS FREE Investigators.
Registry Contributors. Management of bleed­ing in acquired hemo­philia A: Polymer-free drug-coated cor­o­nary stents in patients at high bleed­ing risk.
results from the Euro­pean Acquired Haemophilia (EACH2) Registry. Blood. N Engl J Med. 2015;373(21):2038-2047.
2012;120(1):39-46. 52. Valgimigli M, Patialiakas A, Thury A, et al; ZEUS Investigators. Zotarolimus-
28. Levi M, Levy JH, Andersen HF, Truloff D. Safety of recom­bi­nant acti­vated elut­ ing ver­ sus bare-metal stents in uncer­ tain drug-elut­
ing stent can­di­
fac­tor VII in ran­dom­ized clin­i­cal tri­als. N Engl J Med. 2010;363(19):1791-1800. dates. J Am Coll Cardiol. 2015;65(8):805-815.
29. Rajpurkar M, Croteau SE, Boggio L, Cooper DL. Thrombotic events with 53. Cohen OC, Bertelli M, Manmathan G, et al. Challenges of antithrombotic
recom­bi­nant acti­vated fac­tor VII (rFVIIa) in approved indi­ca­tions are rare ther­apy in the man­age­ment of car­dio­vas­cu­lar dis­ease in patients with
and asso­ci­ated with older age, car­dio­vas­cu­lar dis­ease, and con­com­i­tant inherited bleed­ing dis­or­ders: a sin­gle-cen­tre expe­ri­ence. Haemophilia.
use of acti­vated pro­throm­bin com­plex con­cen­trates (aPCC). J Blood Med. 2021;27(3):425-433.
2019;2019(10):335-340. 54. Ferraris VA, Boral LI, Cohen AJ, Smyth SS, White GC II. Consensus review of
30. Leebeek FW, Kappers-Klunne MC, Jie KS. Effective and safe use of recom­ the treat­ment of car­dio­vas­cu­lar dis­ease in peo­ple with hemo­philia A and B.
bi­nant fac­tor VIIa (NovoSeven) in elderly mild haemophilia A patients with Cardiol Rev. 2015;23(2):53-68.
high-titre antibodies against fac­tor VIII. Haemophilia. 2004;10(3):250-253. 55. Mannucci PM, Mauser-Bunschoten EP. Cardiovascular dis­ease in haemo-
31. Sumner MJ, Geldziler BD, Pedersen M, Seremetis S. Treatment of acquired philia patients: a con­tem­po­rary issue. Haemophilia. 2010;16(suppl 3):58-66.
haemophilia with recom­bi­nant acti­vated FVII: a crit­i­cal appraisal. Haemo- 56. Lin PS, Yao YT. Perioperative man­age­ment of hemo­philia A patients under­
philia. 2007;13(5):451-461. go­ing car­diac sur­gery: a lit­er­a­ture review of published cases. J Cardiotho-
32. Yin EB, Tan B, Nguyen T, et al. Safety and effec­tive­ness of fac­tor VIII inhib­ rac Vasc Anesth. 2021;35(5):1341-1350.
i­tor bypassing activ­ity (FEIBA) and fresh fro­zen plasma in oral anti­co­ag­u­ 57. Fogarty PF, Mancuso ME, Kasthuri R, et al; Global Emerging Hemophilia
lant-asso­ci­ated intra­cra­nial hem­or­rhage: a ret­ro­spec­tive anal­y­sis. Neurocrit Panel. Presentation and man­age­ment of acute cor­o­nary syn­dromes among
Care. 2017;27(1):51-59. adult per­sons with haemophilia: results of an inter­na­tional, ret­ro­spec­tive,
33. Ingerslev J, Sørensen B. Parallel use of by-pass­ing agents in haemophilia 10-year sur­vey. Haemophilia. 2015;21(5):589-597.
with inhib­i­tors: a crit­i­cal review. Br J Haematol. 2011;155(2):256-262. 58. Boehnel C, Rickli H, Graf L, Maeder MT. Coronary angi­og­ra­phy with or with­
34. Janbain M, Leissinger CA, Kruse-Jarres R. Acquired hemo­philia A: emerg­ing out per­cu­ta­ne­ous cor­o­nary inter­ven­tion in patients with hemo­philia—sys­
treat­ment options. J Blood Med. 2015;6(8 May):143-150. tem­atic review. Catheter Cardiovasc Interv. 2018;92(1):1-15.
35. Tiede A, Collins P, Knoebl P, et  al. International rec­ om­men­ da­ tions on 59. Shalabi A, Kachel E, Kogan A, et al. Cardiac sur­gery in patients with hemo­
the diag­no­sis and treat­ment of acquired hemo­philia A. Haematologica. philia: is it safe? J Cardiothorac Surg. 2020;15(1):76.
2020;105(7):1791-1801. 60. Lim MY, Abou-Ismail MY. Left atrial append­age occlu­sion for man­age­ment
36. Lieberman JR, Pensak MJ. Prevention of venous throm­bo­em­bolic dis­ease of atrial fibril­la­tion in per­sons with hemo­philia. Thromb Res. 2021;206(Octo­
after total hip and knee arthroplasty. J Bone Joint Surg Am. 2013;95(19):1801- ber):9-13.
1811. 61. Bolton-Maggs PHB. Factor XI defi­ciency—resolv­ing the enigma? Hematol-
37. Sanders YV, Giezenaar MA, Laros-van Gorkom BA, et al; WiN Study Group. ogy. 2009;2009(1):97-105.
von Willebrand dis­ease and aging: an evolv­ing phe­no­type. J Thromb Hae- 62. Gay ND, Azar S, Salomon O, Taylor JA. Management of severe fac­tor XI defi­
most. 2014;12(7):1066-1075. ciency in car­diac sur­gery: a case report and review of the lit­er­a­ture. Hae-
38. Borghi M, Guglielmini G, Mezzasoma AM, et al. Increase of von Willebrand mophilia. 2017;23(6):e512-e514.
fac­tor with aging in type 1 von Willebrand dis­ease: fact or fic­tion? Haema- 63. Royal College of Obstetricians and Gynecologists. Management of inher-
tologica. 2017;102(11):e431-e433. ited bleed­ing dis­or­ders in preg­nancy: green-top guide­line no. 71 (joint
39. Abou-Ismail MY, Ogunbayo GO, Secic M, Kouides PA. Outgrowing the lab­o­ with UKHCDO). BJOG. 2017;124(8):e193-e263.
ra­tory diag­no­sis of type 1 von Willebrand dis­ease: a two decade study. Am 64. Pagano MB, Konkle BA, Wu Y, Josephson N. Preoperative man­age­ment of
J Hematol. 2018;93(2):232-237. fac­tor XI defi­ciency with ther­a­peu­tic plasma exchange: a case report and
40. Flood VH, Christopherson PA, Gill JC, et al. Clinical and lab­o­ra­tory var­i­abil­ lit­er­a­ture review. J Clin Apher. 2016;31(6):579-583.
ity in a cohort of patients diag­nosed with type 1 VWD in the United States. 65. Alsammak MS, Ashrani AA, Winters JL, Pruthi RK. Therapeutic plasma
Blood. 2016;127(20):2481-2488. exchange for perioperative man­age­ment of patients with con­gen­i­tal fac­
41. Atiq F, Meijer K, Eikenboom J, et al; WiN Study Group. Comorbidities asso­ tor XI defi­ciency. J Clin Apher. 2017;32(6):429-436.
ci­ated with higher von Willebrand fac­tor (VWF) lev­els may explain the 66. Salomon O, Budnik I, Avishai E, et al. Single low dose of rFVIIa com­bined
age-related increase of VWF in von Willebrand dis­ease. Br J Haematol. with antifibrinolytic agent is a sim­ple and safe treat­ment for fac­tor XI-
2018;182(1):93-105. defi­cient patients under­go­ing sur­gery. Thromb Haemost. 2019;119(12):1927-
42. Rydz N, Grabell J, Lillicrap D, James PD. Changes in von Willebrand fac­tor 1932.
level and von Willebrand activ­ity with age in type 1 von Willebrand dis­ 67. Mannucci PM, Duga S, Peyvandi F. Recessively inherited coag­u­la­tion dis­or­
ease. Haemophilia. 2015;21(5):636-641. ders. Blood. 2004;104(5):1243-1252.
43. James PD, Connell NT, Ameer B, et al. ASH ISTH NHF WFH 2021 guide­lines
on the diag­no­sis of von Willebrand dis­ease. Blood Adv. 2021;5(1):280-300.
44. Seaman CD, Ragni MV. The effect of age on von Willebrand fac­tor and
bleed­ing symp­toms in von Willebrand dis­ease. Thromb Haemost. © 2021 by The Amer­i­can Society of Hematology
2020;120(8):1159-1165. DOI 10.1182/hema­tol­ogy.2021000288
Prakash Singh Shekhawat
Bleeding dis­or­ders and sur­gery in aging patients  |  535
PERIOPERATIVE CONSULTATIVE HEMATOLOGY: CAN YOU CLEAR MY PATIENT FOR SURGERY ?

Heparin-induced thrombocytopenia
and cardiovascular surgery

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/536/1851441/536pishko.pdf by guest on 13 December 2021


Allyson M. Pishko1 and Adam Cuker1,2
Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA; and 2Department of Pathology and Laboratory
1

Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA

Clinicians generally counsel patients with a history of heparin-induced thrombocytopenia (HIT) to avoid heparin prod-
ucts lifelong. Although there are now many alternative (nonheparin) anticoagulants available, heparin avoidance remains
challenging for cardiac surgery. Heparin is often preferred in the cardiac surgery setting based on the vast experience
with the agent, ease of monitoring, and reversibility. To “clear” a patient with a history of HIT for cardiac surgery, hema-
tologists must first confirm the diagnosis of HIT, which can be challenging due to the ubiquity of heparin exposure and
frequency of thrombocytopenia in patients in the cardiac intensive care unit. Next, the “phase of HIT” (acute HIT, sub-
acute HIT A/B, or remote HIT) should be established based on platelet count, immunoassay for antibodies to platelet
factor 4/heparin complexes, and a functional assay (eg, serotonin release assay). As long as the HIT functional assay
remains positive (acute HIT or subacute HIT A), cardiac surgery should be delayed if possible. If surgery cannot be
delayed, an alternative anticoagulant (preferably bivalirudin) may be used. Alternatively, heparin may be used with either
preoperative/intraoperative plasma exchange or together with a potent antiplatelet agent. The optimal strategy among
these options is not known, and the choice depends on institutional experience and availability of alternative anticoag-
ulants. In the later phases of HIT (subacute HIT B or remote HIT), brief intraoperative exposure to heparin followed by an
alternative anticoagulant as needed in the postoperative setting is recommended.

LEARNING OBJECTIVES
• Recognize the phases of HIT and implications for heparin reexposure for CV surgery
• Understand the indications and potential alternative (nonheparin) anticoagulants for use in CV procedures and
surgeries

Introduction
Heparin­induced thrombocytopenia (HIT) is a highly pro­ CLINICAL CASE
thrombotic state resulting from pathogenic antibodies to A 45­year­old man with ischemic cardiomyopathy and a
platelet factor 4/heparin (PF4/H) complexes.1 Clinicians history of left ventricular thrombosis receiving warfarin is
generally counsel patients who experience this potentially admitted with worsening dyspnea. Warfarin is held and
life­threatening adverse reaction to never receive heparin an unfractionated heparin infusion is started. He develops
again. With the development of many alternative (non­ acute thrombocytopenia on hospital day 7, and a lower
heparin) anticoagulants, avoiding heparin in most circum­ extremity ultrasound reveals a new popliteal vein throm­
stances (eg, venous thromboembolism treatment) is not bosis (Figure 1A). A 4Ts score is calculated to be 7 points
difficult.2 Cardiovascular (CV) surgery is a unique scenario (high probability). The clinical team switches the heparin
in which heparin is highly preferred given the vast experi­ to bivalirudin and sends HIT laboratory testing. The immu­
ence with the drug, the ease of monitoring with a point­of­ noglobulin G–specific PF4/H enzyme­linked immunosor­
care assay (activated clotting time), and a readily available bent assay (ELISA) is 2.2 optical density (OD) units (positive
reversal agent (protamine).3 It is not uncommon for hema­ result ≥0.4 units). A few days later, the serotonin release
tologists to be asked to “clear” a patient with a history of assay (SRA) returns positive.
HIT for CV surgery. Here we present our approach to eval­
uating and managing such patients.

536 | Hematology 2021 | ASH Education Program


A B C D
Phases of HIT

Acute HIT Subacute HIT A Subacute HIT B Remote HIT


HIT lab
tesng

PF4/H ELISA 2.2 OD 1.5 OD 1.0 OD <0.4 OD


SRA POSITIVE POSITIVE NEGATIVE NEGATIVE
Ancoagulant

Warfarin
Heparin Bivalirudin

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/536/1851441/536pishko.pdf by guest on 13 December 2021


250 Bivalirudin for PCI
225
200
Platelet count x

175
Plan for intraoperave
150
109/L

heparin for cardiac


125 Bivalirudin for
transplant then
100 LVAD placement
alternave
75 (nonheparin)
50 ancoagulant
postoperavely
25
0
1 2 3 4 5 6 7 8 9 10 15 16 17 18 19 20 21 22 52 58 75 100
Days from iniaon of heparin

Figure 1. Management of a patient undergoing PCI and cardiac surgery during multiple phases of HIT. (A) Patient develops a fall in
platelet count and lower extremity deep vein thrombosis 7 days after initiation of unfractionated heparin. The 4Ts score is 7. Heparin
is stopped and the patient is started on bivalirudin. HIT laboratory testing reveals a positive PF4/H ELISA and positive SRA. Acute HIT
is diagnosed. (B) At hospital day 15, the platelet count has recovered. The PF4/H ELISA and SRA remain positive, meeting criteria for
subacute HIT A. The patient undergoes left heart catheterization with bivalirudin. At hospital day 20, he remains in subacute HIT A
and requires LVAD placement that cannot be delayed. He receives bivalirudin during LVAD placement. Postprocedurally, he continues
receiving bivalirudin and is bridged to warfarin for discharge to home. (C) The patient is subsequently referred to a hematology clinic
for cardiac transplant evaluation. Repeat anti-PF4/H testing remains positive by ELISA (1.0 OD units) 45 days post-HIT diagnosis, but
the SRA is now negative, satisfying criteria for subacute HIT B. (D) Approximately 3 months after index admission for HIT, both PF4/H
ELISA and SRA are negative. The patient is listed for cardiac transplant with planned brief intraoperative heparin exposure followed
by treatment with an alternative anticoagulant postoperatively. PCI, percutaneous cardiac intervention; LVAD, left ventricular assist
device; PF4/H ELISA, Platelet factor-4/heparin Enzyme linked immunoassay; SRA, serotonin release assay.

Diagnosis of HIT in patients with CV dis­ease or can­celed. In extreme cases, HIT may be the comorbidity that
HIT is a highly feared iat­ro­genic com­pli­ca­tion of CV sur­gery, dur­ deters cli­ni­cians from pro­ceed­ing with life­sav­ing mea­sures such
ing which patients are nearly uni­ver­sally exposed to hep­a­rin.4 as list­ing for car­diac trans­plant. Although the diag­no­sis of HIT
Indeed, when HIT occurs post-CV sur­gery, it is asso­ci­ated with alone should never be an abso­lute con­tra­in­di­ca­tion to nec­es­sary
excess mor­bid­ity and mor­tal­ity. In a pro­pen­sity score–matched urgent car­diac inter­ven­tions, it undoubt­edly com­pli­cates oper­
study of 11 820 CV sur­gery patients, 29.1% of patients who devel­ a­tive man­age­ment. Thus, it is of utmost impor­tance to get the
oped HIT after car­diac sur­gery had a throm­bo­em­bolic event diag­no­sis “right.” The first step of any eval­u­a­tion for “his­tory of
(com­pared with 2.9% who did not develop HIT), and post­op­er­a­ HIT” is to obtain and review the clin­i­cal his­tory and lab­o­ra­tory
tive mor­tal­ity was 21.8% in patients with HIT (vs 5.3% in patients test­ing that led to the diag­no­sis. Heparin aller­gies placed on the
who did not develop HIT). Fortunately, in this study, as in oth­ers, chart for suspected HIT are often not removed after HIT has been
HIT was uncom­mon, occur­ring in 1.1% of CV sur­gery patients.5 ruled out.7 Clarifying the HIT diag­no­sis is vital to deter­min­ing the
Educational ini­tia­tives have increased aware­ness of what can anti­co­ag­u­lant strat­egy for sur­gery.
be a hor­ren­dous com­pli­ca­tion fol­low­ing even the most rou­tine In car­diac inten­sive care unit or CV sur­gery patients, cau­tion
of CV sur­ger­ies. However, they have also con­trib­uted to an epi­ is needed when cal­cu­lat­ing the 4Ts score (Table 1). First, most
demic of overdiagnosis.6 The ram­i­fi­ca­tions of overdiagnosis in patients will have a fall in plate­let count fol­low­ing place­ment of
patients with CV dis­ease are seri­ous; as such, patients fre­quently intra­vas­cu­lar devices, car­dio­pul­mo­nary bypass (CPB), or extra­cor­
require sur­ger­ies or pro­ce­dures neces­si­tat­ing hep­a­rin use. A po­real mem­brane oxy­gen­a­tion that can mimic the degree of fall
patient with a his­tory of HIT may havePrakash
urgent pro­cSingh Shekhawat
e­dures delayed typ­i­cal of HIT (≥30%-50% from base­line).8 With intra-aor­tic bal­

HIT and CV sur­gery  | 537


Table 1. Special con­sid­er­ations in appli­ca­tion of the 4Ts score to car­diac sur­gery patients

Component of 4Ts score Cardiac sur­gery pop­u­la­tion con­sid­er­ations


Timing of plate­let count fall A biphasic plate­let count pat­tern is typ­i­cal of HIT after CPB sur­gery
Early-onset and per­sis­tent throm­bo­cy­to­pe­nia after CPB is rarely HIT
Thrombocytopenia Percent fall in plate­let count is cal­cu­lated from the highest plate­let count that imme­di­ately pre­cedes the
puta­tive HIT-related plate­let count decline, to the nadir value
The nadir plate­let count need not fall below 150 × 109/L, par­tic­u­larly in sur­gi­cal patients who have post­op­
er­a­tive thrombocytosis
Thrombosis or other sequelae Digital ische­mia due to hypo­ten­sion, vaso­pres­sors, non-HIT DIC, and/or under­ly­ing periph­eral arte­rial
dis­ease may mimic HIT-asso­ci­ated small-ves­sel throm­bo­em­bo­lism
Other causes of throm­bo­cy­to­pe­nia Common alter­na­tive causes in car­diac patients include CPB, IABP, ECMO, LVAD, infec­tion, and other med­i­
ca­tions that can cause throm­bo­cy­to­pe­nia (eg, gly­co­pro­tein IIb/IIIa antag­o­nists)

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/536/1851441/536pishko.pdf by guest on 13 December 2021


Adapted from Pishko and Cuker4 with per­mis­sion from Georg Thieme Verlag KG.
CPB, cardiopulmonary bypass; IABP, intra-aortic balloon pump; DIC, dis­sem­i­nated intra­vas­cu­lar coag­u­la­tion; ECMO, extra­cor­po­real mem­brane oxy­
gen­a­tion; LVAD, left ventricular assist device.

loon pump place­ment, the plate­let count falls a mean 40% from diag­no­sis. Unfortunately, the patient’s car­diac sta­tus wors­ens.
base­line shortly after place­ment.9 Left ven­tric­u­lar assist devices An echo­car­dio­gram shows new left ven­tric­u­lar wall motion
(LVADs) also cause plate­let acti­va­tion and shear stress, lead­ing abnor­mal­i­ties. Cardiology rec­om­mends a left heart cath­e­ter­i­za­
to throm­bo­cy­to­pe­nia.10 Following CPB sur­gery, 30% to 50% of tion (Figure 1B).
patients develop throm­bo­cy­to­pe­nia, with a typ­i­cal decrease
of 50% and a pre­dict­able nadir 48 to 72 hours post­op­er­a­tively.8
Thrombocytopenia that occurs soon after cardiac surgery and
persists is rarely indicative of HIT in cardiac surgery patients.11 Phases of HIT: selec­tion of anti­co­ag­ul­ant for car­diac
Rather, a “biphasic fall,” in which the plate­let count recov­ers after pro­ce­dures/sur­gery
the 48- to 72-hour post-CPB fall and then sud­denly drops around HIT gen­er­ally fol­lows a pre­dict­able course, with the plate­let
days 5 to 10 after hep­a­rin expo­sure is more indic­a­tive of HIT.12 Sec­ count returning to base­line in most patients within 7 days fol­low­
ond, “dusky dig­its” are com­monly due to prolonged vaso­pres­sor ing dis­con­tin­u­a­tion of hep­a­rin, followed by neg­a­tive func­tional
use, arte­rial line place­ment, periph­eral arte­rial dis­ease, non-HIT assay at a median of 50 days, and dis­ap­pear­ance of anti-PF4/H
dis­sem­i­nated intra­vas­cu­lar coag­u­la­tion, and/or poor car­diac out­ antibodies at a median of 85 days.17,18 The risk of hep­ar­in reex­
put, not throm­bo­sis.4 There are other pre­dic­tion scores devised posure depends on the “phase of HIT” (Table 2).19,20 The highest
for the CV sur­gery pop­u­la­tion (Lillo–Le Louet score)13 or that incor­ risk is before plate­let count recov­ery and when the func­tional
po­rate fea­tures such as the use of an intra-aor­tic bal­loon pump or assay remains pos­i­tive. Recurrence of HIT with hep­a­rin reexpo­
CPB within the past 96 hours (HIT Expert Probability [HEP] score).14 sure in the “remote HIT” phase after the immu­no­as­say becomes
These scores have not been con­sis­tently proven to have higher neg­a­tive appears to be low.19 However, as most patients avoid
diag­nos­tic accu­racy than the 4Ts score in pro­spec­tive stud­ies.14 hep­a­rin after a HIT diag­no­sis, this risk is not well char­ac­ter­ized.
However, they may be use­ful in con­junc­tion with the 4Ts score,
par­tic­u­larly in high­light­ing “other” causes of throm­bo­cy­to­pe­nia. PCI: acute HIT or his­tory of HIT
Determining an accu­rate pre­test prob­a­bil­ity of HIT is cru­cial There are robust data for alter­na­tive anti­co­ag­u­lants, par­tic­u­
as HIT anti­body test­ing can be mis­lead­ing in the CV sur­gery pop­ larly bivalirudin, for per­cu­ta­ne­ous car­diac inter­ven­tions (PCIs)
u­la­tion. Up to 20% of patients screened before CPB sur­gery had (Table 3). The Amer­i­can College of Cardiology/Amer­i­can Heart
detect­ able anti-PF4/H antibodies (most were the non­ patho­ Association guide­ line gives both hep­ ar­in and bivalirudin a
genic non–immu­ no­ glob­ u­
lin G type). Detection of these anti­ class I indi­ca­tion for use dur­ing pri­mary PCI in patients with
bodies was not asso­ci­ated with an increase in adverse events.15 ST-ele­va­tion myo­car­dial infarc­tion (STEMI).21 A recent unpub­
PF4/H antibodies also fre­ quently develop post­ op­
er­a­tively; a lished meta-anal­y­sis iden­ti­fied 8 ran­dom­ized con­trolled tri­als
mul­ti­cen­ter study of approximately 1000 CV sur­gery patients com­par­ing bivalirudin to hep­ar­in for PCI for STEMI or non-ST
reported a 50% sero­con­ver­sion rate as mea­sured using a poly­ ele­va­tion myo­car­dial infarc­tion. In the pooled data, bivaliru­
specific PF4/H ELISA at 30 days after sur­gery.16 Seroconversion din was asso­ci­ated with a reduc­tion in seri­ous bleed­ing rates
was not asso­ci­ated with increased death or throm­bo­em­bo­lism. com­pared with hep­a­rin and, in the STEMI sub­group, a 30-day
Thus, it is essen­tial to send HIT lab­o­ra­tory test­ing only in patients mor­tal­ity ben­e­fit as well.22,23 One pro­spec­tive study of bivaliru­
with suf­fi­ciently high clin­i­cal sus­pi­cion of HIT. din dur­ing PCI exclu­sively in patients with HIT reported “pro­
ce­dural suc­cess” in 98% of patients; only 1 of 52 patients (1.9%)
had major bleed­ing.24 There are also data supporting the use
of bivalirudin in other min­i­mally inva­sive car­diac pro­ce­dures,
includ­ing transcathether aor­tic valve replace­ment. The effect
CLINICAL CASE (Con­t in­u ed) of Bivalirudin on Aortic Valve Intervention 3 trial ran­dom­ized
The patient con­tin­ues tak­ing bivalirudin for con­firmed HIT. His 802 patients (HIT and non-HIT) to bivalirudin vs hep­a­rin dur­
plate­let count recov­ers to base­line approx­i­ma­tely 1 week after ing transcathether aor­tic valve replace­ment and found sim­i­lar

538  |  Hematology 2021  |  ASH Education Program


Table 2. Phases of acute HIT

Time from dis­con­tin­u­a­tion


Phases Platelet count Functional assay Immunoassay
of hep­a­rin
Suspected HIT Decreased ? ? NA
Acute HIT Decreased + + NA
Subacute HIT A Normal + + Majority recover plate­let
count by 7 days
Subacute HIT B Normal − + Median 50 days for func­tional
assay to become neg­a­tive
Remote HIT Normal − − Median 85 days for PF4/
H antibodies to become
unde­tect­able

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/536/1851441/536pishko.pdf by guest on 13 December 2021


Adapted from Cuker19 with per­mis­sion from Georg Thieme Verlag KG.
NA, not appli­ca­ble.

rates of major bleed­ing and adverse CV out­comes.25 Among HIT guide­lines rec­om­mend delaying CV sur­gery until patients
patients with a his­tory of HIT, there was no sig­nif­i­cant dif­fer­ enter at least the sub­acute HIT B phase, if fea­si­ble. For car­diac
ence in bleed­ing rates in the hep­a­rin and bivalirudin groups sur­gery that can­not be delayed, these guide­lines sug­gest 1 of
(9.0% vs 10.4%).25 3 options2:
Other alter­na­tive anti­co­ag­u­lants stud­ied for PCI include arg­
1. intraoperative anticoagulation with bivalirudin,
atroban and danaparoid (not avail­­ able in the United States).
2. intraoperative hep­a­rin after treat­ment with pre­op­er­a­tive
These agents have less data than bivalirudin but may be con­sid­
and/or intraoperative ther­a­peu­tic plasma exchange (TPE), and
ered when there is a lack of avail­abil­ity or pro­ce­dural expe­ri­ence
3. intraoperative hep­a­rin with a potent antiplatelet agent (eg,
with bivalirudin. In 1 open-label study of 91 patients with HIT who
pros­ta­cy­clin/tirofiban).
received argatroban for PCI, the “pro­ce­dural suc­cess” rate was
94%.26 There are reports of 61 cases of danaparoid use for inva­ There is grow­ing expe­ri­ence with direct thrombin inhibitors
sive vas­cu­lar pro­ce­dures (mostly PCI), but out­comes were not (DTIs), par­tic­u­larly bivalirudin, in CV sur­gery (Table 3). The Evalua­
reported.27 tion of Patients dur­ing Coronary Artery Bypass (EVOLUTION-ON)
In acute HIT/sub­acute HIT A, Amer­i­can Society of Hematol­ study com­pared hep­a­rin with bivalirudin for patients under­go­ing
ogy (ASH) 2018 HIT guide­lines sug­gest using bivalirudin over coronary artery bypass graft (CABG) with CPB (his­tory of HIT was
other alter­na­tive anti­co­ag­u­lants for PCI. If bivalirudin is not avail­­ not required).28 “Procedural suc­cess” was not sig­nif­i­cantly dif­fer­
able, argatroban may also be con­sid­ered.2 In later phases of HIT, ent between study arms, nor was cumu­la­tive median blood loss
sub­acute HIT B/remote B, the panel also sug­gests bivalirudin within the first 24 hours. Notably, 6.1% of patients who received
over hep­a­rin use for PCI because mul­ti­ple stud­ies show sim­i­lar bivalirudin required repeat explor­ atory oper­ a­
tions com­ pared
safety and effi­cacy to hep­a­rin. However, this is sub­ject to the with 1.9% of patients who received hep­a­rin. Limitations of the
avail­abil­ity of the agent and insti­tu­tional expe­ri­ence. study were small sam­ple size and var­i­a­tion in trans­fu­sion thresh­
olds between insti­tu­tions. Similarly, the EVOLUTION-OFF study
showed sim­i­lar rates of major adverse events in patients under­
go­ing elec­tive “off-pump” car­diac sur­gery with bivalirudin com­
pared with hep­a­rin.29 A pro­spec­tive sin­gle-arm study of bivalirudin
CLINICAL CASE (Con­t in­u ed)
only in patients with a his­tory of HIT who required CPB sur­gery
The patient undergoes car­diac cath­e­ter­i­za­tion while receiv­ing enrolled 49 patients (43 with acute HIT and 6 with a his­tory of HIT).
bivalirudin. No intervenable cor­o­nary lesion is iden­ti­fied. The “Procedural suc­cess” was achieved in 42 of 49 patients (85.7%).30
patient remains crit­i­cally ill. The car­diac team rec­om­mends a In a “real-world” ret­ro­spec­tive study, 13 patients received a DTI
LVAD. On hos­pi­tal day 18, HIT lab­o­ra­tory test­ing is repeated. for CPB sur­gery, mostly CABG and/or valve sur­gery.31 Patients
The PF4/H ELISA is 1.5 OD units, and the SRA remains pos­i­tive who received DTI for CPB had no dif­fer­ence in mor­tal­ity, throm­
(Figure 1B). bo­sis, or hem­or­rhage com­pared with those who received hep­a­
rin. The authors cau­tioned that patients treated with a DTI may
have been at intrin­si­cally lower bleed­ing risk or may have under­
gone lower-risk pro­ce­dures than those reexposed to hep­a­rin.31
Cardiac sur­gery: acute HIT/sub­acute HIT A Another strat­egy is to reexpose patients with acute HIT/sub­
Although data sup­port bivalirudin for PCI, hep­a­rin remains the acute HIT A to hep­a­rin intraoperatively fol­low­ing TPE +/− intra­
highly pre­ferred agent for most other CV pro­ce­dures/sur­ger­ ve­nous immu­no­glob­u­lin (IVIG). In a clin­i­cal cohort of 24 patients
ies. Thus, hema­tol­o­gists are asked to deter­mine the safety of under­go­ing TPE for HIT before hep­a­rin use for CPB, 3 non-HIT-
reexposure to hep­a­rin. Figure 2 describes our approach to se­ related deaths and 3 throm­bo­em­bolic events occurred.32 Nota­
lecting an anti­co­ag­u­lant strat­egy for CV sur­gery in patients with bly, a National Inpatient Sample data­ base study iden­ ti­
fied 90
HIT. Due to the pref­er­ence for hep­a­rin in car­diac sur­ger­ies, ASH patients with HIT who received TPE; less than one-fourth under­
Prakash Singh Shekhawat
HIT and CV sur­gery  | 539
Table 3.  Alternative anti­co­ag­u­lants for car­diac inter­ven­tions and CV sur­gery

Recommendation on use by pro­ce­dure*


Drug Mechanism T1/2 Dosing† Monitoring Additional pre­cau­tions
PCI CPB sur­gery Non-CPB sur­gery
Bivalirudin Direct 25 min (nor­mal to Preferred alter­na­ Preferred alter­na­tive Preferred alter­na­ PCI: 0.75 mg/kg PCI: weight-based Prolonged half-life in
throm­bin mildly impaired tive anti­co­ag­u­lant anti­co­ag­u­lant if tive anti­co­ag­ bolus followed by dos­ing, ACT patients with renal
inhib­i­tor renal func­tion) in patients with a acute HIT/sub­ u­lant if acute 1.75 mg/kg/h for CPB†: ACT >2.5 times impair­ment
34 min (mod­er­ate his­tory of HIT acute HIT A and HIT/sub­acute dura­tion of pro­ce­ base­line Avoid sta­sis in the CPB
renal impair­ sur­gery can­not be HIT A and sur­ dure Non-CPB†: ACT cir­cuit
ment) delayed gery can­not be CPB‡: 1 mg/kg bolus >300 s Hypothermia should
57 min (severe renal delayed then 2.5 mg/kg/h be min­i­mized to
impair­ment) +50 mg added to avoid drug accu­mu­
3.5 h (dial­y­sis) prim­ing solu­tion la­tion
for CPB
Non-CPB‡: 0.75 mg/kg
bolus then
1.75 mg/kg/h
Argatroban Direct 39-51 min (nor­mal Bivalirudin pre­ferred Not gen­er­ally Bivalirudin pre­ PCI: begin infu­sion of PCI: ACT 300-450 s Prolonged half-life
throm­bin hepatic func­tion) but may be con­ recommended ferred but may 25 µg/kg/min and CPB: NR in patients with
inhib­i­tor 181 min (hepatic sid­ered based for use in CPB be con­sid­ admin­is­ter bolus of Non-CPB†: ACT hepatic impair­ment

540  |  Hematology 2021  |  ASH Education Program


impair­ment) on insti­tu­tional ered based on 350 µg/kg (over 3- 200-300 s Case reports describe
expe­ri­ence insti­tu­tional 5 min). Then adjust prolonged bleed­
expe­ri­ence infu­sion rate based ing after stop­ping
on ACT drawn 10 argatroban at end
mins following of CPB
bolus as fol­lows: Case reports describe
ACT <300 s: give clot for­ma­tion in
addi­tional cir­cuit with use of
150 µg/kg bolus, argatroban despite
and increase rate ther­a­peu­tic ACT
to 30 µg/kg/min
(recheck ACT in 5-
10 mins)
ACT >450 s: decrease
infu­sion rate to
15 µg/kg/min
(recheck ACT in
5-10 min)
CPB: NR
Non-CPB‡: NR. Infusion
of 2-5 µg/kg/min
with­out bolus
30-60 min prior to
sur­gery to main­tain
ACT 200-300 s has
been described

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/536/1851441/536pishko.pdf by guest on 13 December 2021


Table 3.  Alternative anti­co­ag­u­lants for car­diac inter­ven­tions and CV sur­gery (Continued)

Recommendation on use by pro­ce­dure*


Drug Mechanism T1/2 Dosing† Monitoring Additional pre­cau­tions
PCI CPB sur­gery Non-CPB sur­gery
Danaparoid§ Indirect 25 h (nor­mal renal Not recommended Not recommended Case reports PCI: NR Anti-Xa Inappropriately long
fac­tor Xa func­tion) have been CPB: NR half-life for CPB
inhib­i­tor 29-35 h (renal described, but Non-CPB‡: NR. Bolus sur­gery
impair­ment) not a pre­ferred of 40 U/kg for non-
agent when CPB sur­gery has
other alter­na­ been described.
tive anti­co­ag­u­
lants avail­­able

Prakash Singh Shekhawat


*Recommendation based on opin­ion of authors as well as the ASH 2018 HIT guide­lines (note certainty of evi­dence was “low” for PCI and “very low” for CPB and non-CPB).

Dosing as described in US Food and Drug Administration (FDA) label for approved indi­ca­tions. For off-label use, exam­ple of dos­ing reg­i­men described in selected stud­ies and/or case reports.
Dose adjustments may be needed for hepatic or renal dysfunction depending on the agent. Refer to insti­tu­tional dos­ing algo­rithms when avail­­able.

Off-label use of the med­i­ca­tion. Not FDA approved for this indi­ca­tion.
§
Not avail­­able in the United States.
ACT, activated clotting time; NR, not recommended.

HIT and CV sur­gery  | 541


Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/536/1851441/536pishko.pdf by guest on 13 December 2021
Paent with reported acute HIT or history of HIT in need of
cardiovascular surgery

Review inial HIT presentaon and HIT lab tesng

Current (or prior) presentaon and lab tesng consistent with No Proceed with CV surgery with

Not HIT
diagnosis of HIT? heparin exposure +
postoperave heparin use
Yes
Obtain Platelet Count, Immunoassay (ex. PF4/H ELISA),
and Funconal Assay (ex. Serotonin Release Assay)

Platelets LOW or RECOVERED,


POSITIVE Funconal Assay & Platelets RECOVERED &

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/536/1851441/536pishko.pdf by guest on 13 December 2021


POSITIVE Immunoassay NEGATIVE Funconal Assay &

Subacute HIT B/Remote HIT


POSITIVE OR NEGATIVE Immunoassay
Yes Delay unl
Acute HIT/subacute HIT A

Can surgery be delayed?


funconal assay
NEGATIVE Intraoperave ancoagulaon: Heparin
No
Intraoperave ancoagulaon: one of 3
opons
1. Bivalirudin Postoperave ancoagulaon: alternave
2. TPE before and/or during CPB with (nonheparin) ancoagulant
heparin
3. Heparin + potent anplatelet agent

Postoperave ancoagulaon: alternave


(nonheparin) ancoagulant

Figure 2. Approach to the management of HIT or history of HIT in cardiac surgery patients. For patients with a history of HIT who
require cardiac surgery, the records should first be reviewed to confirm the diagnosis. The platelet count, anti-PF4/H immunoassay,
and functional assay (e.g., SRA) determine the “phase of HIT” and the risks of heparin reexposure.

went TPE for car­diac sur­gery (CPB or non-CPB). Outcomes fol­ HIT A.” After dis­cus­sion with CV sur­gery and anes­the­sia, the
low­ing car­diac sur­gery were not reported, but over­all, TPE with patient undergoes LVAD place­ment with bivalirudin infu­sion.
HIT com­pared with not receiv­ing TPE with HIT was asso­ci­ated Postprocedurally, he con­tin­ues on bivalirudin and is ulti­mately
with a higher like­li­hood of major bleed­ing (mostly gas­tro­in­tes­ti­ discharged on war­fa­rin. The patient pres­ents to the out­pa­tient
nal bleed­ing) and higher length of stay (20.5 vs 10 days).31 Other hema­tol­ogy clinic for car­diac trans­plant eval­u­a­tion. Repeat
cen­ters have suc­cess­fully used IVIG and TPE before emer­gent CV test­ing 45 days post-HIT diag­no­sis shows PF4/H ELISA remains
sur­gery.33,34 pos­i­tive (1.0 OD units), but the SRA is now neg­a­tive (Figure 1C).
A third strat­egy for patients with acute/sub­acute HIT A requir­ Two months later, both PF4/H ELISA and SRA are neg­at­ ive (Fig­
ing CV sur­gery is intraoperative hep­a­rin in com­bi­na­tion with plate­ ure 1D). The patient is listed for car­diac trans­plant with planned
let inhi­bi­tion. Multiple case stud­ies have described this approach, brief intraoperative hep­a­rin expo­sure.
using a vari­ety of potent antiplatelet agents.35,36 A recent report
describes suc­cess­ful hep­a­rin reexposure in a patient with acute
HIT for CPB sur­gery com­bin­ing antiplatelet ther­apy and IVIG.37
Reexposure in sub­acute B/remote HIT
As the 3 afore­men­tioned strat­e­gies have not been directly
After a patient with HIT no lon­ger has a pos­it­ive func­tional
com­pared, the ASH guide­lines do not rec­om­mend 1 strat­egy
assay and/or no lon­ger has PF4/H ELISA antibodies, the risk
over the oth­ers. Rather, the selec­tion of strat­egy should depend
of devel­op­ing HIT with intraoperative hep­a­rin expo­sure ap­
on insti­tu­tional expe­ri­ence and sur­gi­cal pref­er­ence. If the sec­ond
pears to be low.38 Some groups have hypoth­e­sized that the
or third strat­egy is cho­sen, patients’ expo­sure to hep­a­rin should
very high doses of intraoperative intra­ve­nous hep­a­rin do not
be strictly lim­ited to the intraoperative set­ting. Before and after
invoke the same plate­let-acti­vat­ing response as lower con­cen­
sur­gery, they should receive an alter­na­tive anti­co­ag­u­lant.
tra­tions, even if HIT antibodies are pres­ent.39 Warkentin and
Sheppard38 described 17 patients with a his­tory of HIT who
received intraoperative hep­a­rin (but no post­op­er­a­tive hep­a­
rin) for car­diac or vas­cu­lar sur­gery between 8 weeks and 13.5
CLINICAL CASE (Con­t in­u ed) years after HIT diag­no­sis. All patients had a neg­at­ive PF4/H
With a pos­ i­
tive immu­
no­
as­
say and func­ tional assay but nor­ ELISA and SRA pre­op­er­a­tively. Anti-PF4/H anti­body pos­i­tiv­
mal plate­let count, the patient is diag­nosed with “sub­acute ity and SRA pos­i­tiv­ity were com­mon fol­low­ing sur­gery (9/17

542  |  Hematology 2021  |  ASH Education Program


Table 4. Alternative anti­co­ag­u­lants for venous throm­bo­em­bo­lism pro­phy­laxis and con­sid­er­ations in the post­op­er­a­tive set­ting
in patients with a his­tory of HIT

Alternative Dosing for venous throm­bo­em­bo­lism


Considerations in post­op­er­a­tive set­ting
anti­co­ag­u­lants pro­phy­laxis
Fondaparinux 2.5 mg sub­cu­ta­ne­ously once daily Elimination half-life 17-21 h
Not read­ily revers­ible
Use with cau­tion in patients with renal impair­ment (CrCl 30-50 mL/min). Avoid use
if CrCl <30 mL/min
Reduced clear­ance in patients <50 kg
Caution against use in unsta­ble patients who may require urgent sur­ger­ies/
pro­ce­dures
Apixaban* 2.5 mg oral twice daily Elimination half-life 12 h
Reversal agent may not be read­ily avail­­able and is not cur­rently FDA approved for

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/536/1851441/536pishko.pdf by guest on 13 December 2021


use prior to sur­gery
Use with cau­tion in hepatic impair­ment (contraindicated in Child Pugh C cir­rho­sis)
Caution against use in unsta­ble patients who may require urgent sur­ger­ies/
pro­ce­dures
Rivaroxaban* 10 mg oral daily Elimination half-life 5-9 h
Caution with renal impair­ment, contraindicated in acute renal fail­ure
Use with cau­tion in hepatic impair­ment (contraindicated in Child Pugh C cir­rho­sis)
Caution against use in unsta­ble patients who may require urgent sur­ger­ies/
pro­ce­dures
*Off-label use. Drug is not approved by the US Food and Drug Administration (FDA) for venous thromboembolism pro­phy­laxis in hos­pi­tal­ized
nonorthopedic sur­gi­cal patients.

[53%] and 8/17 [47%], respec­tively). Despite the high rate of Acknowledgment
sero­con­ver­sion, recur­rent HIT occurred in only 1 patient, and Allyson M. Pishko is supported by a 2019 HTRS Mentored Re­
this appeared to be a case of “auto­im­mune HIT” char­ac­ter­ized search Award from the Hemostasis and Thrombosis Research
by strong HIT antibodies that also acti­vated plate­lets in the Society (HTRS), which was supported by an educational grant
absence of hep­a­rin. from Sanofi Genzyme.
Given this evi­dence and the vast expe­ri­ence with hep­a­rin in
CV sur­gery, ASH HIT guide­lines rec­om­mend using intraoperative Conflict-of-inter­est dis­clo­sure
hep­a­rin in patients with remote HIT/sub­acute HIT B.2 Again, hep­ Allyson M. Pishko has received research funding from an edu­ca­
a­rin expo­sure should be lim­ited to the intraoperative set­ting and tional grant from Sanofi Genzyme and Novo Nordisk. Adam Cuker
scru­pu­lously avoided before and after sur­gery.19,20 The expected has served as a con­sul­tant for Synergy and has received roy­al­ties
period for HIT recur­rence is 5 to 10 days after intraoperative hep­ from UpToDate, and his insti­tu­tion has received research sup­port
a­rin expo­sure, and thus the plate­let count should be mon­i­tored on his behalf from Alexion, Bayer, Novartis, Novo Nordisk, Pfizer,
dur­ing this time.20 Sanofi, and Spark.

Off-label drug use


Postoperative anticoagulation in patients with a his­tory
Allyson M. Pishko: Off-label use of fondaparinux and direct oral
of HIT
anticoagulants for treatment of heparin-induced thrombocyto­
Postoperatively, regard­less of the phase of HIT, hep­a­rin should
penia are discussed in the manuscript. Off-label use of alterna­
be avoided. Table 4 out­lines alter­na­tive anti­co­ag­u­lants for ve­
tive anticoagulants in cardiac bypass surgery discussed.
nous throm­bo­em­bo­lism pro­phy­laxis and con­sid­er­ations in the
Adam Cuker: Off-label use of fondaparinux and direct oral anti­
post­op­er­a­tive set­ting.
coagulants for treatment of heparin-induced thrombocytopenia
are discussed in the manuscript. Off-label use of alternative anti­
Conclusions coagulants in cardiac bypass surgery discussed.
Clinicians should be aware of the many chal­lenges in diag­nos­ing
HIT in the CV pop­u­la­tion and order HIT lab­o­ra­tory test­ing judi­ Correspondence
ciously. Ideally, CV sur­ gery that requires hep­ a­rin is delayed in Allyson M. Pishko, Department of Medicine, Perelman School of
a patient with acute HIT/sub­acute HIT A until the plate­let count Medicine, University of Pennsylvania, 3400 Spruce St, 3rd Floor Dull­
recov­ers and the func­tional assay becomes neg­a­tive. If delay is es, Philadelphia, PA 19104; e-mail: allyson​­.pishko@pennmedicine​
not pos­si­ble, options include intraoperative bivalirudin, TPE prior ­.upenn​­.edu.
to and/or dur­ing sur­gery with intraoperative hep­a­rin, or intraop­
erative hep­a­rin in com­bi­na­tion with a potent antiplatelet agent. References
Once the patient progresses to sub­acute HIT B and beyond, the 1. Arepally GM. Heparin-induced throm­bo­cy­to­pe­nia. Blood. 2017;129(21):2864-
2872.
ben­e­fits of intraoperative hep­a­rin use out­weigh the risks of brief 2. Cuker A, Arepally GM, Chong BH, et  al. Amer­i­can Society of Hematology
hep­a­rin expo­sure. In all­cases, hep­a­rin should be avoided pre- and 2018 guide­lines for man­age­ment of venous throm­bo­em­bo­lism: hep­a­rin-
post­op­er­a­tively. induced throm­bo­cy­to­pe­nia. Blood Adv. 2018;2(22):3360-3392.
Prakash Singh Shekhawat
HIT and CV sur­gery  | 543
3. Shore-Lesserson L, Baker RA, Ferraris V, et  al. STS/SCA/AmSECT clin­ic ­ al inter­ven­tion in patients with hep­a­rin-induced throm­bo­cy­to­pe­nia (ATBAT)
prac­tice guide­lines: anticoagulation dur­ing car­dio­pul­mo­nary bypass. J study: main results. J Invasive Cardiol. 2003;15(11):611-616.
Extra Corpor Technol. 2018;50(1):5-18. 25. Dangas GD, Lefèvre T, Kupatt C, et al. Bivalirudin ver­sus hep­a­rin anticoag­
4. Pishko AM, Cuker A. Heparin-induced throm­bo­cy­to­pe­nia in car­diac sur­gery ulation in transcatheter aor­tic valve replace­ment the ran­dom­ized BRAVO-3
patients. Semin Thromb Hemost. 2017;43(7):691-698. trial. J Am Coll Cardiol. 2015;66(25):2860-2868.
5. Brown JA, Aranda-Michel E, Kilic A, et al. Outcomes with hep­a­rin-induced 26. Lewis BE, Matthai WH, Cohen M, et al. Argatroban anticoagulation dur­
throm­bo­cy­to­pe­nia after car­diac sur­gery. Ann Thorac Surg. 2021;112(2):487- ing per­cu­ta­ne­ous cor­o­nary inter­ven­tion in patients with hep­a­rin-induced
493. throm­bo­cy­to­pe­nia. Catheter Cardiovasc Intev. 2002;57(2):177-184.
6. Cuker A. Heparin-induced throm­bo­cy­to­pe­nia (HIT) in 2011: an epi­demic of 27. Magnani H, Gallus A. Heparin-induced throm­bo­cy­to­pe­nia (HIT). A report of
overdiagnosis. Thromb Haemos. 2011;106(6):993-994. 1,478 clin­i­cal out­comes of patients treated with danaparoid (Orgaran) from
7. McMahon CM, Tanhehco YC, Cuker A. Inappropriate doc­ u­men­ta­tion of 1982 to mid-2004. Thromb Haemost. 2006;95(6):967-981.
hep­a­rin allergy in the med­i­cal record because of mis­di­ag­no­sis of hep­a­ 28. Dyke CM, Smedira NG, Koster A, et al. A com­par­i­son of bivalirudin to hep­
rin-induced throm­bo­cy­to­pe­nia: fre­quency and con­se­quences. J Thromb a­rin with prot­amine rever­sal in patients under­go­ing car­diac sur­gery with
Haemost. 2017;15(2):370-374. car­dio­pul­mo­nary bypass: the EVOLUTION-ON study. J Thorac Cardiovasc
8. Griffin BR, Bronsert M, Reece TB, et al. Thrombocytopenia after car­dio­pul­ Surg. 2006;131(3):533-539.
mo­nary bypass is asso­ci­ated with increased mor­bid­ity and mor­tal­ity. Ann 29. Smedira NG, Dyke CM, Koster A, et  al. Anticoagulation with bivaliru­

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/536/1851441/536pishko.pdf by guest on 13 December 2021


Thorac Surg. 2020;110(1):50-57. din for off-pump cor­ o­nary artery bypass grafting: the results of the
9. Vonderheide RH, Thadhani R, Kuter DJ. Association of throm­bo­cy­to­pe­nia EVOLUTION-OFF study. J Thorac Cardiovasc Surg. 2006;131(3):686-692.
with the use of intra-aor­tic bal­loon pumps. Am J Med. 1998;105(1):27-32. 30. Koster A, Dyke CM, Aldea G, et  al. Bivalirudin dur­ing car­dio­pul­mo­nary
10. Muslem R, Caliskan K, Leebeek FWG. Acquired coagulopathy in patients bypass in patients with pre­vi­ous or acute hep­a­rin-induced throm­bo­cy­to­
with left ven­tric­u­lar assist devices. J Thromb Haemost. 2018;16(3):429-440. pe­nia and hep­a­rin antibodies: results of the CHOOSE-ON trial. Ann Thorac
11. Selling S, Malowsky B, Strobel U, et al. Early-onset and persisting throm­bo­ Surg. 2007;83(2):572-577.
cy­to­pe­nia in post-car­diac sur­gery patients is rarely due to hep­a­rin-induced 31. Carlson D, Bartholomew J, Gomes M, McCrae K, Chaturvedi S. Outcomes
throm­bo­cy­to­pe­nia, even when anti­body tests are pos­i­tive. J Thromb Hae- of car­dio­vas­cu­lar sur­gery uti­liz­ing hep­a­rin ver­sus direct throm­bin inhib­i­
most. 2010;8(1):30-36. tors in car­dio­pul­mo­nary bypass in patients with pre­vi­ously diag­nosed HIT.
12. Pouplard C, May MA, Regina S, Marchand M, Fusciardi J, Gruel Y. Changes Thromb Haemost. 2019;120(2):300-305.
in plate­let count after car­diac sur­gery can effec­tively pre­dict the devel­ 32. Moreno-Duarte I, Cooter M, Onwuemene OA, Ghadimi K, Welsby IJ.
op­ment of path­o­genic hep­a­rin-depen­dent antibodies. Br J Haematol. Clinical out­comes of car­diac sur­gery patients under­go­ing ther­a­peu­
2005;128(6):837-841. tic plasma exchange for hep­a­rin-induced throm­bo­cy­to­pe­nia. Vox Sang.
13. Louet AL-L, Boutouyrie P, Alhenc-Gelas M, et al. Diagnostic score for hep­ 2021;116(2):217-224.
a­rin-induced throm­bo­cy­to­pe­nia after car­dio­pul­mo­nary bypass. J Thromb 33. Soares Ferreira Júnior A, Boyle SH, Kuchibhatla M, Akinyemiju T, Onwuemene
Haemost. 2004;2(11):1882-1888. OA. Use of ther­a­peu­tic plasma exchange in hep­a­rin-induced throm­bo­cy­to­
14. Pishko AM, Fardin S, Lefler DS, et  al. Prospective com­par­i­son of the HEP pe­nia: a pop­u­la­tion-based study. J Clin Apher. 2021;36(3):398-407.
score and 4Ts score for the diag­no­sis of hep­a­rin-induced throm­bo­cy­to­pe­ 34. Liu VC, Klompas AM, Stulak JM, Yalamuri SM. The tri­ple HIT: periopera­
nia. Blood Adv. 2018;2(22):3155-3162. tive man­age­ment of hep­a­rin-induced throm­bo­cy­to­pe­nia using plasma
15. Selleng S, Selleng K. Heparin-induced throm­bo­cy­to­pe­nia in car­diac sur­ exchange, intra­ve­nous immu­no­glob­u­lin, and prot­amine infu­sion for left
gery and crit­i­cally ill patients. Thromb Haemost. 2016;116(5):843-851. ven­tric­u­lar assist device implan­ta­tion [published online March 30, 2021].
16. Welsby IJ, Krakow EF, Heit JA, et al. The asso­ci­a­tion of anti-plate­let fac­tor J Cardiothor Vasc Anesth.
4/hep­a­rin antibodies with early and delayed throm­bo­em­bo­lism after car­ 35. Palatianos G, Michalis A, Alivizatos P, et  al. Perioperative use of iloprost
diac sur­gery. J Thromb Haemost. 2016;15(1):57-65. in car­diac sur­gery patients diag­nosed with hep­a­rin-induced throm­bo­cy­
17. Warkentin TE, Kelton JG. Temporal aspects of hep­a­rin-induced throm­bo­cy­ to­pe­nia-reac­tive antibodies or with true HIT (HIT-reac­tive antibodies plus
to­pe­nia. N Engl J Med. 2001;344(17):1286-1292. throm­bo­cy­to­pe­nia): an 11-year expe­ri­ence. Am J Hematol. 2015;90(7):608-
18. Mattioli AV, Bonetti L, Zennaro M, Ambrosio G, Mattioli G. Heparin/PF4 617.
antibodies for­ma­tion after hep­a­rin treat­ment: tem­po­ral aspects and long- 36. Koster A, Kukucka M, Bach F, et  al. Anticoagulation dur­ing car­dio­pul­mo­
term fol­low-up. Am Heart J. 2009;157(3):589-595. nary bypass in patients with hep­a­rin-induced throm­bo­cy­to­pe­nia type II
19. Cuker A. Management of the mul­ti­ple phases of hep­a­rin-induced throm­bo­ and renal impair­ment using hep­a­rin and the plate­let gly­co­pro­tein IIb-IIIa
cy­to­pe­nia. Thromb Haemost. 2016;116(05):835-842. antag­o­nist tirofiban. Anesthesiology. 2001;94(2):245-251.
20. Warkentin TE, Anderson JAM. How I treat patients with a his­tory of hep­ar­ in- 37. Koster A, Nazy I, Birschmann IE, Smith JW, Sheppard JI, Warkentin TE. High-
induced throm­bo­cy­to­pe­nia. Blood. 2016;128(3):348-359. dose IVIG plus cangrelor plate­let “anes­the­sia” dur­ing urgent hep­a­rin-CPB
21. O’Gara PT, Kushner FG, Ascheim DD, et  al. 2013 ACCF/AHA guide­ line in a patient with recent SRA-neg­a­tive HIT-throm­bo­sis with persisting plate­
for the man­age­ment of ST-ele­va­tion myo­car­dial infarc­tion. Circulation. let-acti­vat­ing antibodies. Res Pract Thromb Haemost. 2020;4(6):1060-
2013;127(4):e362-e425. 1064.
22. Stone GW. Individual patient data pooled anal­y­sis of ran­dom­ized tri­als of 38. Warkentin TE, Sheppard J-AI. Serological inves­ti­ga­tion of patients with a
bivalirudin ver­sus hep­a­rin in acute myo­car­dial infarc­tion. Presented at: TCT pre­vi­ous his­tory of hep­a­rin-induced throm­bo­cy­to­pe­nia who are reexposed
Connect; Octo­ber 2020; (virtual meeting). to hep­a­rin. Blood. 2014;123(16):2485-2493.
23. Bikdeli B, McAndrew T, Crowley A, et al. Individual patient data pooled anal­ 39. Warkentin TE. Acute intraoperative HIT dur­ing heart sur­gery: why so rare?
y­sis of ran­dom­ized tri­als of bivalirudin ver­sus hep­a­rin in acute myo­car­dial Thromb Res. 2016;146(6, suppl):110-112.
infarc­tion: ratio­nale and meth­od­ol­ogy. Thromb Haemost. 2019;120(2):348-
362.
24. Mahaffey KW, Lewis BE, Wildermann NM, et al. The anti­co­ag­u­lant ther­apy © 2021 by The Amer­i­can Society of Hematology
with bivalirudin to assist in the per­for­mance of per­cu­ta­ne­ous cor­o­nary DOI 10.1182/hema­tol­ogy.2021000289

544  |  Hematology 2021  |  ASH Education Program


PREGNANCY IN SPECIAL POPULATIONS: CHALLENGES AND SOLUTIONS

How to evaluate and treat the spectrum of TMA


syndromes in pregnancy

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/545/1851434/545scully.pdf by guest on 13 December 2021


Marie Scully
Department of Haematology, University College London Hospitals NHS Foundation Trust and Cardiometabolic Programme-NIHR UCLH/UC BRC,
London, UK

Thrombotic microangiopathy (TMA) is the broad definition for thrombocytopenia, microangiopathic hemolytic anemia,
and end-organ damage. Two important categories are thrombotic thrombocytopenic purpura (TTP) and complement-
mediated hemolytic-uremic syndrome (CM-HUS). Pregnancy and the immediate postpartum period are associated with
TMAs specific to pregnancy in rare situations. These include pregnancy-induced hypertension, preeclampsia, and hemo-
lysis, elevated liver enzymes, and low platelets. TTP and CM-HUS may present in pregnancy. However, the diagnosis
may not be immediately obvious as they share characteristics of pregnancy-related TMAs. Within this review, we discuss
investigations, differential diagnosis of TMAs in pregnancy, and management. The importance is a risk of maternal mor-
tality but also poor fetal outcomes in relation to TTP and CM-HUS. Treatment of these disorders at presentation in preg-
nancy is discussed to achieve remission and prolong fetal viability if possible. In subsequent pregnancies, a treatment
pathway is presented that has been associated with successful maternal and fetal outcomes. Critical to this is a multidis-
ciplinary approach involving obstetricians, the fetal medicine unit, and neonatologists.

LEARNING OBJECTIVES
• TTP and CM-HUS are acute life-threatening disorders, require prompt diagnosis and treatment, and may be com-
plicated by PE or HELLP.
• Congenital TTP needs considering in pregnancy. Subsequent pregnancies require multidisciplinary team monitor-
ing and ADAMTS-13 replacement.

Introduction TTP or CM-HUS needs urgent therapy, initially with plasma


Thrombotic microangiopathies (TMAs) presenting in preg- exchange but thereafter specific ongoing treatment, a dis-
nancy or the postpartum period can be difficult to diagnose integrin and metalloproteinase with a thrombospondin type
and have an impact on both maternal and fetal outcomes, 1 motif–member 13 (ADAMTS-13) replacement, immunosup-
and therapies are dependent on the clinical condition. The pression, or complement inhibitor therapy, respectively.5
treatment for pregnancy-related TMAs, such as pregnancy-
induced hypertension (PIH), preeclampsia (PE) or hemolysis,
elevated liver enzymes, and low platelets (HELLP), includes CLINICAL CASE
control of blood pressure and delivery. Very rarely, acute A 21-year-old woman presented at her 28-week checkup
fatty liver of pregnancy (AFLP) may present with some TMA in her first pregnancy. Routine bloods revealed a reduced
features.2 Diagnosis and treatment of TMA in pregnancy may platelet count (21 × 109/L) and raised blood pressure of
be challenging, as it is dependent on the time of presen- 135/110 mm Hg. She was admitted for further investigation.
tation during pregnancy and potential fetal viability at this Cardiotocography (CTG) revealed fetal distress. Remain-
stage. Furthermore, presentation, for example, of PE can ing relevant bloods included alanine aminotransferase
occur in 1% to 20% of women in the postpartum period, (ALT) of 100 IU (normal range [NR], 10–35 IU) and urine
also typical of complement-mediated hemolytic-uremic protein/creatinine ratio of 432 mg/mmol (NR, <15).
syndrome (CM-HUS).3 Alternatively, features suggestive of What is the diagnosis? How was this established, and
PIH, PE, or HELLP may in fact be the result of thrombotic what was the management in this situation?
thrombocytopenic purpura (TTP) or CM-HUS.4 Diagnosis of
Prakash Singh Shekhawat
Pregnancy TMAs | 545
Diagnosis of preg­nancy-related TMA the coag­u­la­tion abnor­mal­i­ties. The con­di­tion appears to be
TMA in preg­ nancy is suggested by plate­ let counts less than the result of mito­chon­drial dys­func­tion in the oxi­da­tion of fatty
100 × 109/L, a raised LDH (lactate dehydrogenase) typ­i­cally twice acids in the liver. Diagnosis can be con­firmed by test­ing for the
the upper limit of nor­mal, and blood film find­ings suggesting enzyme long-chain 3-hydroxyacyl-CoA dehy­dro­ge­nase.10
ane­mia, polychromasia, throm­bo­cy­to­pe­nia, and fragmented red Thrombocytopenia in preg­nancy, defined as below the nor­mal
blood cells. There is often organ involve­ment related to ische­mic lab­o­ra­tory range, typ­i­cally 150 to 400 × 109/L, is not uncom­mon
dam­age affect­ing the kid­neys, pri­mar­ily in CM-HUS, and brain and 70% to 80% of all­cases of throm­ bo­cy­to­
pe­
nia in preg­
and heart in TTP (Table 1). However, multiorgan involve­ment can nancy are due to ges­ta­tional throm­bo­cy­to­pe­nia, which rarely
be seen in any of the TMAs asso­ci­ated with preg­nancy. There is decreases below 75 × 109/L or is asso­ci­ated with immune dis­ease
the addi­tional impact on pla­cen­tal func­tion, which may man­i­fest (eg, immune thrombocytopenic purpura [ITP] in 3%–4%).11 Throm-
as evi­dence of reduced fetal growth, impaired uter­ine Dopp­ler bocytopenia asso­ci­ated with hyper­ten­sive dis­ease accounts for
flow, abnor­mal CTGs, or even in utero fetal death (IUFD).6 20% of all­cases,12 but plate­let lev­els less than 30 × 109/L would
Further lab­o­ra­tory anal­y­sis to assist with the diag­no­sis of be a sig­nal to exclude CM-HUS or TTP. Similarly, with increas­ing
TMAs include raised bil­i­ru­bin, which is uncon­ju­gated; evi­dence of cre­at­i­nine and wors­en­ing AKI with oliguria/anuria, one should

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/545/1851434/545scully.pdf by guest on 13 December 2021


renal impair­ment; urine pro­tein/cre­at­i­nine esti­mate; a coag­u­la­ con­sider a diag­no­sis of CM-HUS, and neu­ro­logic fea­tures should
tion screen; a direct anti­glob­ul­in test; urate lev­els; retic­ul­o­cytes; prompt con­sid­er­ation of TTP. AKI can be dif­fi­cult to diag­nose in
and liver func­tion tests (Table 2). Autoimmune screen—includ­ing preg­nancy, given the reduc­tion in cre­at­i­nine lev­els dur­ing the
ANA and ENA; com­ple­ment lev­els, spe­cif­i­cally C3 and C4; and preg­nant state. However, guid­ance sug­gests AKI in preg­nancy
ADAMTS-13 activ­ity lev­els—may aid dif­fer­en­ti­at­ing the under­ly­ is diag­nosed with a dou­bling in cre­at­i­nine from base­line at the
ing mech­a­nism. begin­ning of preg­nancy or a 26-µmol/L increase and, there­fore, a
In rela­tion to case 1, her retic­u­lo­cyte count was 5.32% (NR, cre­at­i­nine more than 90   µmol/L or a 25% increase from base­line.13
0.45% –2.42%); cre­at­i­nine, 80    µmol/L (NR, 49-92); eGFR, 83   mL/min
(NR, >90); LDH, 558 (214 IU upper limit of normal); hemo­glo­bin, TTP presenting in preg­nancy
100 g/L (NR, 115-155); white blood cell count,  15.8 × 109/L  (NR, 3-10); TTP is asso­ci­ated with severe ADAMTS-13 defi­ciency. ADAMTS-13
and ADAMTS-13 activ­ity, 14 IU/dL (NR,>60). Anti-nuclear antibody is required for cleav­age of von Willebrand fac­tor (VWF) into mul-
and extractable nuclear antigen were neg­a­tive, with C3 and C4 timeric forms. In preg­nancy, there are increases in von Willebrand
within the NR. factor and fac­tor VIII through­out each tri­mes­ter of preg­nancy,
which remain raised in the post­par­tum period. There is a rel­a­tive
Pregnancy-asso­ci­ated TMAs decrease in ADAMTS-13 lev­els but usu­ally within the NR lev­els:
The diag­no­sis of pre­eclamp­sia is defined by a blood pres­sure this is a phys­i­o­logic effect.14,15 Pregnancy is asso­ci­ated with LDH
of 140/90 mm Hg or higher after 20 weeks’ ges­ta­tion and evi­ lev­els within the NR, and increases are related to cell dam­age or
dence of acute kidney injury (serum cre­ at­i­
nine >90   
µmol/L) ische­mia.16 Presentation of TTP in preg­nancy can be con­fused by
or pro­tein­uria.7 Serum urate lev­ els are increased. Pregnancy the pres­ence of hyper­ten­sion and throm­bo­cy­to­pe­nia, but symp­
induced hypertension occurs in patients with blood pres­sure toms may also be like those of patients with TTP out­side of preg­
lev­els not in the pre­eclamp­sia range but above the patients’ nancy, such as head­aches, transient ischemic attack, and stroke.
base­line lev­els.8 HELLP is con­sid­ered in patients with epi­gas­ Pregnancy-asso­ci­ated TTP accounts for approx­i­ma­tely 5% to
tric or right upper quad­ rant pain and ala­ nine ami­no­ trans­
fer­ 10% of all­TTP cases. What is unusual is that there appears to be
ase (ALT) or aspar­tate ami­no­trans­fer­ase (AST) lev­els more than an increased chance of con­gen­i­tal TTP in preg­nancy pre­sen­ta­
40  IU/L. Alkaline phos­pha­tase lev­els are increased phys­i­o­log­i­ tions rather than immune anti­body-medi­ated TTP, which would
cally in preg­nancy (Table 2). Eclampsia is a severe con­di­tion in be more typ­i­cal.17,18 Presentation of immune TTP (iTTP) and con-
which there may be altered men­tal state, severe head­aches, genital TTP (cTTP) is 95% and 5%, respec­tively, but in preg­nancy,
blind­ness, stroke, or sei­zures. AFLP is a rare and life-threat­en­ing cTTP has been reported at an increased rate, seen in 24% to 66%
con­di­tion asso­ci­ated with liver fail­ure, multiorgan involve­ment, of cases. Abnormal liver func­tion tests, as in HELLP, are not nor-
and coagulopathy. It is an impor­tant dif­fer­en­tial given its acute mally asso­ci­ated with pregnancy associated-TTP. TTP can pres­
pre­sen­ta­tion and multiorgan involve­ment but dif­fer­en­ti­ated by ent at any stage of preg­nancy but is more fre­quently diag­nosed

Table 1. Key fea­tures dif­fer­en­ti­at­ing preg­nancy-asso­ci­ated and preg­nancy-related TMAs

Incidence/ Renal Neurologic ADAMT-13


Characteristic preg­nan­cies Thrombocytopenia HBP impair­ment fea­tures activ­ity, IU/dL
Preeclampsia 1/20 +++ +++ +/− ++ >20
HELLP 1/1000 ++++ + +/− +/− >20
TTP 1/200,000 +++ + + +++ <10
CM-HUS 1/25,000 +++ ++ +++ + >20
Note: the degree of involvement is demonstrated by an increasing number of +.
Table adapted from Scully.9
HBP, hypertension.

546  |  Hematology 2021  |  ASH Education Program


Table 2. Investigations in preg­nancy-asso­ci­ated TMA

Pregnancy-related TMAs (eg, PE,


Test TTP CM-HUS HELLP, AFLP)
Hemoglobin Reduced Reduced Reduced
Platelet count Very reduced Reduced Reduced/very reduced
Reticulocytes Increased Increased Normal/increased
Fragmentation on blood film Yes +++ Yes ++ Yes +
Serum cre­at­i­nine Normal/increased Increased Normal/increased
Urine pro­tein/cre­at­i­nine ratio Normal/increased Increased Increased
Coagulation screen Normal Normal Normal (except AFLP; coag­u­la­tion screen
prolonged—decreased fibrin­o­gen)

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/545/1851434/545scully.pdf by guest on 13 December 2021


Bilirubin Increased Increased Mild increase
ALT/AST Normal Normal Increased
Alkaline phos­pha­tase Normal Normal Increased in HELLP and AFLP
Urate Normal Normal Increased
C3/C4 Normal Reduced C3 in some cases Normal
ADAMTS-13 Severely reduced Mildly reduced Mildly reduced
Note: the degree of involvement is demonstrated by an increasing number of +.

in the third tri­mes­ter and post­par­tum period. Undiagnosed TTP,


espe­cially cTTP, is related to sec­ond-tri­mes­ter IUFD. CLINICAL CASE (Con­tin­ued)
There were fea­tures sug­ges­tive of hyper­ten­sion/PE and poten­
CM-HUS presenting in preg­nancy tially HELLP. Indeed, the plan for this woman had been emer­gency
There are nor­ mal phys­ i­
o­logic changes in both the immune cae­sar­ean sec­tion with plate­let trans­fu­sion cover. However, the
and com­ple­ment path­ways dur­ing preg­nancy with the aim of diag­no­sis of TTP was raised. When the patient was trans­ferred, she
protecting the fetus. The increase in com­ple­ment acti­va­tion is received imme­di­ate plasma exchange and the baby was deliv­ered
asso­ci­ated with a com­pa­ra­ble esca­la­tion in con­trol­ling com­ at 28+ weeks’ ges­ta­tion by cae­sar­ean sec­tion under gen­eral anes­
ple­ment pro­teins such as CD46 to pre­vent excess com­ple­ment thetic, with no plate­let trans­fu­sion required. Blood loss related to
acti­va­tion.19 sur­gery was 500 mL. High-dose ste­roid ther­apy was given before
Presentation of CM-HUS was thought to be only in the post­ and after the cae­sar­ean sec­tion. The baby weighed 0.836  kg and
par­tum period. Indeed, 80% of cases are diag­nosed dur­ing this was trans­ferred to the neo­na­tal unit. Blood pres­sure was con­
period, but like TTP, CM-HUS pre­sen­ta­tion can, rarely, occur from trolled with labetalol, and plasma exchange con­tin­ued daily until
the first tri­mes­ter. The detec­tion of com­ple­ment-asso­ci­ated muta­ plate­let nor­mal­i­za­tion, requir­ing 6 in total. ADAMTS-13 activ­ity
tion is com­pa­ra­ble to non­preg­nant CM-HUS cases, and 30% to level at the refer­ring hos­pi­tal was 14  IU/dL and less than 5  IU/dL
50% have no detect­able abnor­mal­ity. Some patients with HELLP preexchange. There was no immu­no­glob­u­lin G (IgG) anti­body to
syn­drome show fea­ tures, on serum sam­ ples, of com­ ple­
ment ADAMTS-13, and sub­se­quent muta­tional anal­y­sis for ADAMTS-13
acti­va­tion,20 and increased pla­cen­tal com­ple­ment acti­va­tion con­firmed a homo­zy­gous mis­sense muta­tion R1060W.24 Diagnosis
asso­ci­ated with com­ple­ment muta­tions has been iden­ti­fied in in the clin­i­cal case was con­gen­i­tal TTP, presenting in preg­nancy.
pre­eclamp­sia.21,22
Vascular endo­the­lial fac­tor pro­tects against aber­rant com­
ple­ment acti­va­tion and is inhibited by sol­u­ble fms-like tyro­sine Treatment of preg­nancy-asso­ci­ated TMA
kinase 1 (sFLT1) in other organs. There is a cor­re­late with sFLT1 In many cases, the pre­cise diag­no­sis may not be imme­di­ately clear,
expres­sion, suggesting a role for com­ple­ment in PE.23 A role for so plasma exchange should be started as soon as a diag­no­sis of
mea­sur­ing sFLT1 and pla­cen­tal growth fac­tor to aid dif­fer­en­ti­a­ TTP or CM-HUS is con­sid­ered (Figure 1). The inves­ti­ga­tions in Table
tion of hyper­ten­sive dis­or­ders in preg­nancy by mea­sur­ing the 1 should be under­taken, with ADAMTS-13 activ­ity lev­els dif­fer­en­ti­at­
ratio of sFLT1/placental growth factor has been suggested. ing CM-HUS and TTP (Figure 1). It may not be pos­si­ble to con­firm
Before the use of com­ple­ment inhib­i­tor ther­apy, there was if TTP is con­gen­i­tal or immune medi­ated; there­fore, ini­tial immu­no­
mater­nal and fetal mor­tal­ity with, nearly two-thirds of women sup­pres­sion with ste­roids may be nec­es­sary. In the cur­rent era, the
requir­ing acute renal replace­ment ther­apy, half of all­cases devel­ nanobody caplacizumab is ini­ti­ated on the diag­no­sis of acute TTP.25
op­ing end-stage renal dis­ease, and an CM-HUS relapse rate of If the diag­ no­sis of TTP is fol­ low­ing deliv­
ery/post­par­
tum, this is
30%. There was also a risk of spon­ta­ne­ous abor­tion and still­birth accept­ able, but it is not licensed in preg­
n ancy, and given the small
that in the past has not been fully appre­ci­ated.22 molec­u­lar size, it should be avoided if the preg­nancy is ongo­ing.
Unlike TTP, CM-HUS in preg­nancy may not be asso­ci­ated with It may be obvi­ous the diag­no­sis is CM-HUS, in which case
throm­bo­cy­to­pe­nia (ie, <100  ×  109/L), with AKI being the over­rid­ ini­ti­a­tion of com­ple­ment inhib­i­tor ther­apy can be con­sid­ered.
ing fea­ture. Eculizumab is a human­ized IgG4κ chi­me­ric anticomplement C5
Prakash Singh Shekhawat
Pregnancy TMAs  |  547
ADAMTS
Delivery -13
acvity
PIH level
Control blood
pressure and
PE monitor cTg
HELLP
for pregnancy-
related TMA =CM-HUS =TTP
and
deterioraon

ADAMTS-13

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/545/1851434/545scully.pdf by guest on 13 December 2021


HUS TTP
Eculizumab replacement/
immunosuppression

AKI Cardiac
Neurologic +/- ADAMTS-13 anbody
Complement mutaon
analysis if negave
renal symptoms analysis/CFH anbody
PEX
ADAMTS-13 mutaon
analysis

Figure 1. Summary of treatment of acute TMA in pregnancy. Highlighted are the conditions that may present as TMA in pregnancy.
Pregnancy-related TMAs generally require delivery. TTP and CM-HUS may present features of pregnancy-related TMAs but require a
different pathway in part dictated by the ADAMTS-13 activity level. TTP is diagnosed when ADAMTS-13 activity is less than 10 IU/dL but
may be up to 20 IU/dL before TTP can be excluded. Repeat ADAMTS-13 activity samples and ADAMTS-13 antibody should be checked.

anti­body, pre­vents ongo­ing acti­va­tion of the mem­brane attack laxis. This is based on evi­dence of pla­cen­tal dys­func­tion his­to­log­
com­plex, but has no effect on the com­ple­ment sys­tem up to C3. i­cally and impaired uter­ine artery Dopp­ler flow dur­ing preg­nancy
Eculizumab has been used in preg­nancy, pri­mar­ily in parox- from affected cases (Figure 2).
ysmal nocturnal haemoglobinuria,26,27 but there are data now of The risk of relapse of CM-HUS in sub­se­quent preg­nan­cies is
its suc­cess­ful use in CM-HUS.28 The global atypical HUS Registry approx­i­ma­tely 25%. The risk appears to be lower in patients for
has con­firmed the ben­e­fit of eculizumab in women with preg­ whom no com­ple­ment muta­tion has been iden­ti­fied.31 Even with
nancy-asso­ci­ated HUS, and it has sig­nif­i­cantly improved renal a com­ple­ment abnor­mal­ity, the risk remains var­i­able. The chance
out­comes.29 of sub­se­quent acute CM-HUS in preg­nancy is greatest in those
Meticulous con­trol of blood pres­sure and sup­port­ive ther­ women with com­ple­ment fac­tor H or I muta­tions. The use of the
apy should be insti­gated, such as renal replacement therapy in com­ple­ment inhib­i­tor, eculizumab, has sig­nif­i­cantly improved
CM-HUS. the out­come for CM-HUS. Currently, ther­apy may be ini­ti­ated
Unlike preg­nancy-related TMAs, for which deliv­ery is asso­ dur­ing preg­nancy if there is a sig­nal to sug­gest exac­er­ba­tion of
ci­ated with nor­mal­i­za­tion of the con­di­tion within 48 hours, in CM-HUS. There are no data on starting treat­ment for high-risk
preg­ nancy-asso­ ci­
ated TMAs (TTP and CM-HUS), achiev­ ing a cases once preg­nancy is con­firmed, but this should be con­sid­
remis­sion is pos­si­ble, which may be impor­tant for women pre- ered in indi­vid­ual cases.31
senting ear­lier in preg­nancy and deliv­ery would be asso­ci­ated In women in whom iTTP was con­firmed, mon­i­tor­ing of ADAMTS-
with fetal com­pro­mise. This deci­sion needs to be under­taken in 13 activ­ity lev­els as well as rou­tine lab­o­ra­tory param­e­ters should
con­junc­tion with obste­tri­cians and neo­na­tol­o­gists. be under­taken, iden­ti­fy­ing the base­line level at the begin­ning
of preg­nancy. Testing of ADAMTS-13 activ­ity is advised in each
tri­mes­ter and in the post­par­tum period. If lev­els decrease, there
What are the risks and treat­ment options in sub­se­quent are sev­eral options, includ­ing ste­roids, aza­thi­op ­ rine, or plasma
preg­nan­cies? exchange, if ADAMTS-13 activ­ity lev­els drop below 10   IU/dL. Rit-
Subsequent preg­nan­cies in patients with both TTP and CM-HUS uximab has been used in preg­nancy, but it is pref­er­ab ­ le to wait
can be supported, assum­ing women and their part­ners are fully until the post­par­tum period.4
aware of the risks to them and the fetus (Figure 2). Management In con­gen­i­tal TTP, ADAMTS-13 replace­ment should begin
in sub­se­quent preg­nan­cies requires a mul­ti­dis­ci­plin­ary approach, once preg­nancy is con­firmed, ini­tially every 2 weeks (approx­i­
with reg­u­lar mon­i­tor­ing of mother and baby dur­ing preg­nancy ma­tely 10 mL/kg). This may need to be increased to weekly from
and into the post­par­tum period. Empirically, women are started the sec­ond tri­mes­ter, targeting nor­mal plate­let count and LDH
on low-dose aspi­rin (e.g., 75/100    mg/d)30 and thromboprophy- and an absence of symp­toms. Further evi­dence for the role of

548  |  Hematology 2021  |  ASH Education Program


iTTP

ADAMTS-13 prepregnancy to ensure in normal range

Iniate LDA or confirmaon of pregnancy

Iniate thromboprophylaxis if confirmed thrombophilia


or previous placental dysfuncon

ADAMTS-13 measurements at least each trimester/postpartum Regular fetal scans/measurement of uterine artery
flow by Doppler

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/545/1851434/545scully.pdf by guest on 13 December 2021


Severely
Abnormal:
Closely
reduced: Risks: TTP Normal: Abnormal:
Normal: Iniaon of Risks: IUGR,
monitor +/- relapse, Connue Ensure no
Delivery PEX complicaon IUFD,
iniaon of down clinical/ preterm
as per frequency s of therapy
steroids/
depending
obstetric ADAMTS- delivery, fetal
obstetric azathioprine eg, PEX,
on roune immunosupp
pathway to 13 TTP prematurity
plan if earlier in
lab ression delivery relapse
pregnancy
parameters

Will need regular laboratory parameters, FBC, LDH and renal funcon

Congenital

Confirmaon of pregnancy and iniate plasma


infusion 10 mL/kg every 2 weeks

Start LDA and thromboprophylaxis


Regular fetal
scans/measurement of uterine
Increase plasma infusion to 10 mL/kg artery flow by Doppler
weekly from second trimester or if
platelets decrease/LDH increase
outside of the normal lab range

Plan delivery 37/38 weeks’ gestaon

Connue regular plasma infusions


for at least 8 weeks postpartum

Will need regular laboratory parameters, FBC, LDH and renal funcon

Figure 2. TTP in subsequent pregnancies, immune mediated and congenital pathways. Proceeding with subsequent pregnancy
requires a discussion of the maternal and fetal risks. Close management throughout pregnancy is required for patients with both
immune-mediated and congenital TTP. Regular fetal imaging ultrasound and uterine artery Doppler measurements would be advised.
IUGR, in utero growth retardation; PEX, plasma exchange.
Prakash Singh Shekhawat
Pregnancy TMAs  |  549
Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/545/1851434/545scully.pdf by guest on 13 December 2021
Figure 3A- From the 1st pregnancy Figure 3B-From the 2nd pregnancy
Figure 3. Placental histology from the clinical case. (A) Distal villous hypoplasia and infarction at 28 weeks’ gestation in untreated
congenital TTP. (B) Normal villi in the subsequent delivery from the same mother after treatment throughout pregnancy. ADAMTS-13
replacement was via plasma infusion.

plasma infu­sion and improved out­comes for both mother and Off-label drug use
fetus has been presented.32 Marie Scully: rituximab for TTP is an off-label use of therapy.
In all 3 of these sub­groups of TMA, deliv­ ery needs to be
planned, aiming for 37 to 38 weeks’ ges­ta­tion. Correspondence
Marie Scully, Department of Haematology, University College
London Hospitals NHS Foundation Trust and Cardiometabolic
Programme-NIHR UCLH/UC BRC, 250 Euston Rd, London NW1
CLINICAL CASE (Con­t in­u ed) 2PG, UK; e-mail: m​­.scully@ucl​­.ac​­.uk.
The patient did have a fur­ther preg­nancy. She had ini­ti­ated a
pro­gram of reg­u­lar plasma infu­sion because of symp­tom­atol­ References
ogy and con­tin­ued with fort­nightly plasma infu­sion until the 1. Scully M, Cataland S, Coppo P, et al; International Working Group for
end of her sec­ond tri­mes­ter. She received weekly plasma infu­ Thrombotic Thrombocytopenic Purpura. Consensus on the stan­dard­i­
sion until 8 weeks post­par­tum. Her sec­ond child was deliv­ered za­tion of ter­mi­nol­ogy in throm­botic throm­bo­cy­to­pe­nic pur­pura and
related throm­botic microangiopathies. J Thromb Haemost. 2017;15(2):
by elec­tive cae­sar­ean sec­tion at 38 weeks’ ges­ta­tion, with no
312-322.
com­pli­ca­tions, and the baby weighed 2.75  kg. A com­par­i­son 2. Nelson DB, Byrne JJ, Cunningham FG. Acute fatty liver of preg­nancy. Clin
of his­tol­ogy from her first preg­nancy and sec­ond preg­nancy is Obstet Gynecol. 2020;63(1):152-164.
pro­vided in Figure 3. 3. Sibai BM. Etiology and man­a ge­m ent of post­p ar­tum hyper­ten­s ion-
pre­eclamp­sia. Am J Obstet Gynecol. 2012;206(6):470-475.
4. Neave L, Scully M. Microangiopathic hemo­ lytic ane­
mia in preg­ nancy.
Transfus Med Rev. 2018;32(4):230-236.
In con­ clu­
sion, TTP and CM-HUS are acute life-threat­ en­
ing 5. Scully M, Goodship T. How I treat throm­botic throm­bo­cy­to­pe­nic pur­pura and
con­di­tions that require diag­nos­tic con­sid­er­ation and imme­di­ate atyp­i­cal haemolytic uraemic syn­drome. Br J Haematol. 2014;164(6):759-766.
ther­apy. Presentation within preg­nancy com­pli­cates the diag­ 6. Thomas MR, Robinson S, Scully MA. How we man­age throm­botic microan-
no­sis fur­ther. Pregnancy has its own TMAs (eg, PE and HELLP). giopathies in preg­nancy. Br J Haematol. 2016;173(6):821-830.
7. O’Gorman N, Wright D, Poon LC, et  al. Multicenter screen­ ing for pre-
Features of these preg­nancy-related TMAs may be part of the eclamp­sia by mater­nal fac­tors and bio­mark­ers at 11-13 weeks’ ges­ta­tion:
TTP/CM-HUS pre­sen­ta­tion. Aside from the mater­nal mor­tal­ity, com­par­i­son with NICE guide­lines and ACOG rec­om­men­da­tions. Ultra-
there are sig­nif­i­cant risks for the fetus, includ­ing in utero growth sound Obstet Gynecol. 2017;49(6):756-760.
retar­da­tion and IUFD, normally evi­dent in the sec­ond tri­mes­ter. 8. Umesawa M, Kobashi G. Epidemiology of hyper­ten­sive dis­or­ders in preg­
nancy: prev­a­lence, risk fac­tors, pre­dic­tors and prog­no­sis. Hypertens Res.
Pathways have been presented to aid remis­sion dur­ing preg­
2017;40(3):213-220.
nancy but also to ensure improved out­ comes in sub­ se­
quent 9. Scully M. Thrombotic throm­bo­cy­to­pe­nic pur­pura and atyp­i­cal hemo­lytic
preg­nan­cies. This includes close obser­va­tion, with reg­u­lar fetal ure­mic syn­drome microangiopathy in preg­nancy. Semin Thromb Hemost.
scans to ensure no impact on pla­cen­tal func­tion. 2016;42(7):774-779.
10. Gutiérrez Junquera C, Balmaseda E, Gil E, et al. Acute fatty liver of preg­
nancy and neo­na­tal long-chain 3-hydroxyacyl-coen­zyme A dehy­dro­ge­nase
Conflict-of-inter­est dis­clo­sure (LCHAD) defi­ciency. Eur J Pediatr. 2009;168(1):103-106.
Marie Scully: speak­
ers fees and advi­ sory boards for Sanofi, 11. Gernsheimer T, James AH, Stasi R. How I treat throm­bo­cy­to­pe­nia in preg­
Takeda, Octapharma, Novartis, Alexion. nancy. Blood. 2013;121(1):38-47.

550  |  Hematology 2021  |  ASH Education Program


12. Eslick R, McLintock C. Managing ITP and throm­bo­cy­to­pe­nia in preg­nancy. and syncytiotrophoblast dam­age in pre­eclamp­sia. Hypertens Pregnancy.
Platelets. 2020;31(3):300-306. 2019;38(3):193-199.
13. Kidney Disease: Improving Global Outcomes (KDIGO) Acute Kidney Injury 24. Alwan F, Vendramin C, Liesner R, et al. Characterization and treat­ment of
Work Group. KDIGO clin­i­cal prac­tice guide­line for acute kid­ney injury. Kid- con­gen­i­tal throm­botic throm­bo­cy­to­pe­nic pur­pura. Blood. 2019;133(15):
ney Int. 2012;2(suppl):1-138. ­1644-1651.
14. Mannucci PM, Canciani MT, Forza I, Lussana F, Lattuada A, Rossi E. Changes 25. Scully M, Cataland SR, Peyvandi F, et al; HERCULES Investigators. Capla-
in health and dis­ease of the metalloprotease that cleaves von Willebrand cizumab treat­ment for acquired throm­botic throm­bo­cy­to­pe­nic pur­pura.
fac­tor. Blood. 2001;98(9):2730-2735. N Engl J Med. 2019;380(4):335-346.
15. Sánchez-Luceros A, Farías CE, Amaral MM, et al. von Willebrand fac­tor-cleav­ing 26. Miyasaka N, Miura O, Kawaguchi T, et al. Pregnancy out­comes of patients
pro­te­ase (ADAMTS13) activ­ity in nor­mal non-preg­nant women, preg­nant and with par­ox­ys­mal noc­tur­nal hemo­glo­bin­uria treated with eculizumab: a
post-deliv­ery women. Thromb Haemost. 2004;92(6):1320-1326. Jap­a­nese expe­ri­ence and updated review. Int J Hematol. 2016;103(6):703-
16. Jaiswar SP, Gupta A, Sachan R, Natu SN, Shaili M. Lactic dehy­dro­ge­nase: a 712.
bio­chem­i­cal marker for pre­eclamp­sia-eclamp­sia. J Obstet Gynaecol India. 27. Kelly RJ, Höchsmann B, Szer J, et al. Eculizumab in preg­nant patients with
2011;61(6):645-648. par­ox­ys­mal noc­tur­nal hemo­glo­bin­uria. N Engl J Med. 2015;373(11):1032-
17. Scully M, Thomas M, Underwood M, et al; col­lab­o­ra­tors of the UK TTP Reg- 1039.
istry. Thrombotic throm­bo­cy­to­pe­nic pur­pura and preg­nancy: pre­sen­ta­tion, 28. Servais A, Devillard N, Frémeaux-Bacchi V, et  al. Atypical haemolytic

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/545/1851434/545scully.pdf by guest on 13 December 2021


man­age­ment, and sub­se­quent preg­nancy out­comes. Blood. 2014;124(2): uraemic syn­drome and preg­nancy: out­come with ongo­ing eculizumab.
211-219. Nephrol Dial Transplant. 2016;31(12):2122-2130.
18. Moatti-Cohen M, Garrec C, Wolf M, et al; French Reference Center for Throm- 29. Fakhouri F, Scully M, Ardissino G, Al-Dakkak I, Miller B, Rondeau E. Pregnancy-
botic Microangiopathies. Unexpected fre­quency of Upshaw-Schulman syn­ trig­gered atyp­i­cal hemo­lytic ure­mic syn­drome (aHUS): a Global aHUS Regis-
drome in preg­nancy-onset throm­botic throm­bo­cy­to­pe­nic pur­pura. Blood. try anal­y­sis [published online 7 April 2021]. J Nephrol. 2021.
2012;119(24):5888-5897. 30. Bujold E, Roberge S, Nicolaides KH. Low-dose aspi­rin for pre­ven­tion of
19. Regal JF, Gilbert JS, Burwick RM. The com­ple­ment sys­tem and adverse preg­ adverse out­comes related to abnor­mal pla­cen­ta­tion. Prenat Diagn. 2014;
nancy outcomes. Mol Immunol. 2015;67(1):56-70. ­34(7):642-648.
20. Vaught AJ, Gavriilaki E, Hueppchen N, et  al. Direct evi­dence of com­ple­ 31. Fakhouri F, Scully M, Provôt F, et al. Management of throm­botic microangi-
ment acti­va­tion in HELLP syn­drome: a link to atyp­i­cal hemo­lytic ure­mic opathy in preg­nancy and post­par­tum: report from an inter­na­tional work­ing
syn­drome. Exp Hematol. 2016;44(5):390-398. group. Blood. 2020;136(19):2103-2117.
21. Salmon JE, Heuser C, Triebwasser M, et al. Mutations in com­ple­ment reg­ 32. Sakai K, Fujimura Y, Nagata Y, et al. Success and lim­i­ta­tions of plasma treat­
u­la­tory pro­teins pre­dis­pose to pre­eclamp­sia: a genetic anal­y­sis of the ment in preg­nant women with con­gen­i­tal throm­botic throm­bo­cy­to­pe­nic
PROMISSE cohort. PLoS Med. 2011;8(3):e1001013. pur­pura. J Thromb Haemost. 2020;18(11):2929-2941.
22. Gaggl M, Aigner C, Csuka D, et al. Maternal and fetal out­comes of preg­
nan­cies in women with atyp­i­cal hemo­lytic ure­mic syn­drome. J Am Soc
Nephrol. 2018;29(3):1020-1029.
23. Yonekura Collier A-R, Zsengeller Z, Pernicone E, Salahuddin S, Khankin E-V, © 2021 by The Amer­i­can Society of Hematology
Karumanchi S-A. Placental sFLT1 is asso­ci­ated with com­ple­ment acti­va­tion DOI 10.1182/hema­tol­ogy.2021000290

Prakash Singh Shekhawat


Pregnancy TMAs  |  551
PREGNANCY IN SPECIAL POPULATIONS: CHALLENGES AND SOLUTIONS

Pregnancy in special populations: challenges


and solutions practical aspects of managing von
Willebrand disease in pregnancy

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/552/1851794/552turan.pdf by guest on 13 December 2021


Ozlem Turan1,2 and Rezan Abdul Kadir1,2
Katharine Dormandy Haemophilia and Thrombosis Unit and Department of Obstetrics and Gynecology, Royal Free Hospital NHS Trust, London, UK;
1

and 2EGA Institute for Women’s Health, University College London, London, UK

Pregnancy and childbirth pose an important hemostatic challenge for women with von Willebrand disease (VWD) and
can be associated with an increased risk of maternal and neonatal bleeding complications. VWD is a genetically
and clinically heterogeneous bleeding disorder caused by a deficiency or an abnormality in the function of von Wille-
brand factor. Understanding inheritance pattern, hemostatic response to pregnancy, and response to treatment is essen-
tial for provision of individualized obstetric care and optimal outcome. A multidisciplinary approach to management with
a close liaison between the obstetric team and the hemophilia treatment center is required for continuity of care from
preconception counseling through to antenatal, peripartum, and postpartum care. Delivery plan must be coordinated
by the multidisciplinary team and include decisions on place and mode of delivery, implementation of safe analgesia/
anesthesia, and peripartum hemostasis. In this clinical case-based review, we aim to deliver evidence-based practical
guidance for challenges encountered during pregnancy and management of childbirth and puerperium.

LEARNING OBJECTIVES
• Evaluate the clinical challenges in the management of pregnancy and childbirth in VWD
• Recognize heterogeneity in the maternal and fetal risks in different types of VWD
• Discuss multidisciplinary management and best practice for optimal maternal and neonatal outcome

diagnosis, perform genetic testing when applicable, and assess


CLINICAL CASE their bleeding risk and response to treatment.
A 20­year­old woman with severe type 2M von Willebrand The inheritance of most cases of type 1, 2A, 2B, and
disease (VWD) is pregnant at 10 weeks’ gestation. This 2M VWD is autosomal dominant, with a 50% chance of an
is her first pregnancy, and she is seen in the multidisci­ affected fetus in each pregnancy. Types 2N and 3 are inher­
plinary clinic for women with bleeding disorders at the ited in an autosomal recessive manner,1 and there is a 25%
hemophilia treatment center (HTC) to discuss and plan risk of having an affected child in each pregnancy when
management of pregnancy and delivery. both parents are carriers of the genetic mutation. Genetic
testing and PND are not routinely performed in VWD but
are considered in families with type 3 VWD.2 PND in late
Preconceptional care and prenatal diagnosis pregnancy (usually at 34 weeks’ gestation) is also consid­
Understanding different types of inherited bleeding disorders ered in women with severe types of VWD if the mutation is
(IBDs), their inheritance pattern, and maternal and fetal/neo­ known to assist management of delivery3,4; for example, if
natal bleeding risks is important for appropriate counseling the baby is affected, delivery can be planned in a tertiary
in relation to prenatal diagnosis (PND) and planning manage­ center, or delivery can be managed without any restric­
ment of pregnancy and childbirth. Ideally, women with IBDs tions and possibly in the local maternity unit if the baby is
should be seen preconceptionally, to review their historic unaffected.

552 | Hematology 2021 | ASH Education Program


Antenatal man­age­ment
CLINICAL CASE (Con­t in­u ed) Women with all­types of IBDs require a mul­ti­dis­ci­plin­ary team
The patient was seen for pre­con­cep­tion coun­sel­ing in the pre­vi­ (MDT) approach for man­age­ment of preg­nancy and deliv­ery, as
ous year. Her diag­no­sis was con­firmed to be type 2M. Her base­ well as a close col­lab­o­ra­tion between the obstet­ric team and
line fac­tor VIII (FVIII) level was 0.58 IU/mL, von Willebrand fac­tor HTC.11,12 Women with VWD do not appear to have an increased
(VWF):antigen (Ag) was 0.28 IU/mL, VWF:Ristocetin cofactor (RCo) risk of mis­car­riage or antepartum hem­or­rhage7,13-15 and do not
was <0.4 IU/mL, and VWF:collagen binding was 0.18 IU/mL, with usu­ally require pro­phy­lac­tic treat­ment in preg­nancy; thus, ante­
nor­mal multimeric anal­y­sis. Genetic anal­y­sis had con­firmed dele­ na­tal care can be pro­vided in their local obstet­ric units in col­
tion on allele 4222_4224delAAG. She was coun­seled by a mul­ti­dis­ lab­o­ra­tion with the HTC, except those with severe type 2 and
ci­plin­ary obstet­ric/hema­tol­ogy team about inher­i­tance, bleed­ing 3 VWD, who require care in a ter­tiary cen­ter.16
risks for her and her baby, and man­age­ment of preg­nancy. There are a num­ber of com­mon early preg­nancy com­pli­ca­
tions asso­ci­ated with high risk of hem­or­rhagic sequalae. Mis­
carriage is the most prev­a­lent one that affects up to 15% of all­
Changes in coag­u­la­tion fac­tors in preg­nancy preg­nan­cies before 12 weeks’ ges­ta­tion.17 In women with VWD,

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/552/1851794/552turan.pdf by guest on 13 December 2021


There are sev­eral phys­i­o­log­i­cal adap­ta­tions from early stages of the preg­nancy-induced rise in fac­tor lev­els is not achieved in
preg­nancy in prep­a­ra­tion for labor and deliv­ery. Hypercoagula­ the first half of preg­nancy, and they remain at an increased risk
bility is one of the phys­i­o­logic changes that involves a pro­gres­ of bleed­ing after a mis­car­riage or inva­sive PND pro­ce­dures. In
sive increase in lev­els of VWF and FVIII through­out preg­nancy, these cir­cum­stances, it is vital to assess their fac­tor lev­els and
with a peak at the time of deliv­ery.5 This preg­nancy-induced con­sider pro­vi­sion of hemo­static sup­port.16
rise in VWF and FVIII is also seen in most preg­nant women with Antenatal mon­i­tor­ing should include checking VWF:Ag and
VWD but not to the same extent that is seen in healthy preg­nant FVIII:procoagulant activity at least dur­ing their first visit and the
con­trols.6 In addi­tion, due to the het­ero­ge­ne­ity of geno­typic third tri­mes­ter of preg­nancy (32-34 weeks), unless first-tri­mes­ter
and phe­no­typic fea­tures of VWD, each type exhib­its a dif­fer­ent lev­els are already within the nor­mal range.10,15,18 VWF:RCo is the
response to preg­nancy. best pre­dic­tor of bleed­ing risk and should be checked at least
Women with type 1 VWD with a prepregnancy base­line VWF once dur­ing the third tri­mes­ter, espe­cially in women with type 2
and FVIII level of >0.3 IU/mL are likely to achieve fac­tor lev­els VWD. Women with type 2B VWD should also have mon­i­tor­ing of
within the nor­mal range by the end of preg­nancy.6 In type 2 VWD, their plate­let count.
WVF and FVIII lev­els increase sig­nif­i­cantly, but lack of high molec­ Women with VWD are not at an increased risk of obstet­
ular weight multimers does not change and the VWF:RCo remains ric com­pli­ca­tions.19 A national data­base from the United States,
reduced.7 Pregnancy may also exac­er­bate preexisting throm­bo­ includ­ing 4067 deliv­er­ies, found no increase in fetal growth restric­
cy­to­pe­nia in type 2B VWD.8 Both VWF and FVIII lev­els go up in tion, pla­cen­tal abrup­tion, or pre­term birth in women with VWD.19
type 2N VWD, but FVIII is usu­ally lower in com­par­i­son to VWF in Therefore, they do not require addi­tional obstet­ric assess­ment or
most cases.9 Women with type 3 VWD typ­i­cally do not show any mon­i­tor­ing.
increase in FVIII and VWF dur­ing preg­nancy10 (Figure 1). Iron defi­ ciency and iron-defi­ ciency ane­ mia, sec­ond­ary to
heavy men­strual bleed­ing, is a com­mon diag­no­sis among women
with VWD. There is an increased iron require­ment, espe­cially at
CLINICAL CASE (Con­t in­u ed) late stages of preg­ nancy. Anemia is asso­ ci­
ated with adverse
fetal and mater­nal out­come, includ­ing risk of pri­mary post­par­
In our patient, changes in her fac­tor lev­els were con­gru­ent with
tum hem­or­rhage (PPH).20-22 In a cohort study of 506 women with
changes described in type 2 VWD; VWF:Ag and FVIII lev­els
VWD, 28% of those who expe­ri­enced PPH were ane­mic ante­na­
increased, whereas VWF:RCo remained unchanged.
tally.23 Therefore, reg­u­lar mon­i­tor­ing and cor­rec­tion of ane­mia
and iron defi­ciency are a very impor­tant part of ante­na­tal care.
VWF:RCo
Women with IBDs should be reviewed by the MDT team in
FVIII
the third tri­ mes­ter of preg­ nancy (32-34 weeks) ahead of the
expected date of deliv­ery for a deliv­ery plan. The plan should
VWF:Ag
include details of place and mode of deliv­ery (MOD), hemo­static
cover for deliv­ery and post­par­tum period, pain relief and anes­
the­sia, and man­age­ment of mother and baby dur­ing labor and
after deliv­ery. The plan is agreed on with the mother and com­
mu­ni­cated to all­involved in her care.
Baseline First Second Third 8 weeks
trimester trimester trimester postpartum
Management of labor and deliv­ery
Women with type 1 VWD who have a VWF:RCo level >0.5 IU/mL
First tri­mes­ter: FVIII: 0.57 IU/mL, VWF:RCo: <0.4 IU/mL, and at 32 to 34 weeks’ ges­ta­tion and a mild bleed­ing phe­no­type
VWF:Ag: 0.29 IU/mL can plan deliv­ery in their local mater­nity unit unless they have
Second tri­mes­ter: FVIII: 0.64 IU/mL, VWF:RCo: <0.4 IU/mL, addi­tional bleed­ing risk such as pre­vi­ous PPH.16 However, for
and VWF:Ag: 0.34 IU/mL women with a severe IBD, includ­ing type 2 and 3 VWD, or those
Third tri­mes­ter: FVIII: 0.97 IU/mL, VWF:RCo: <0.4 IU/mL, and car­ry­ing a baby poten­tially at risk of a severe dis­or­der, it is rec­
VWF:Ag: 0.50 IU/mL ommended that they deliver in a mater­nity unit with an affil­i­
ated HTC, where the nec­es­sary exper­tise in man­age­ment and
Prakash Singh Shekhawat
Management of von Willebrand dis­ease in preg­nancy  |  553
Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/552/1851794/552turan.pdf by guest on 13 December 2021
Figure 1. Modifications of FVIII and VWF levels during normal pregnancy and in women with the more frequent types of VWD.10

resources for lab­o­ra­tory test­ing and clot­ting fac­tor replace­ment risk in the new­borns need to be implemented. These mea­sures
are read­ily avail­­able.16,24 are presented in Table 1, based on risk strat­i­fic
­ a­tion and rec­om­
The aim of plan­ning the MOD is to achieve the least trauma men­da­tions by the Royal College of Obstetricians and Gynaecolo­
to the mother and infant. VWD is usu­ally not an indi­ca­tion for a gists guide­lines on man­age­ment of IBDs in preg­nancy.16
cesar­ean sec­tion, and the deci­sion for MOD should be guided by
obstet­ric indi­ca­tions.18,25 Unlike hemo­philia, there is a lack of data Neuraxial anal­ge­sia/anes­the­sia
on MOD and risk of cra­nial bleed­ing for fetuses at risk of severe Neuraxial anal­ge­sia pro­vi­des effec­tive pain con­trol in labor, but
VWD (types 2 and 3). Labor and deliv­ery are asso­ci­ated with an there is a con­cern of a the­o­ret­ic ­ al increased risk of spi­nal canal
increase in VWF and FVIII in neo­na­tes,26 and thus neo­na­tes with hema­toma in women with bleed­ing dis­or­ders due to their
mild type 1 VWD have a neg­li­gi­ble bleed­ing risk.4 Neonates with coag­u­la­tion defect. In total, 161 550 epi­du­rals were sited for
type 2 and 3 and severe type 1 VWD still have reduced lev­els of obstet­ric anal­ge­sia dur­ing a 2-week national audit period in the
VWF and FVIII at birth and have a the­o­ret­i­cal risk of increased United Kingdom, with a reported inci­dence of 5 cases of spi­nal
bleed­ing.27 Despite this the­o­ret­i­cal risk, bleed­ing risks in neo­na­ canal hema­toma. There were zero cases of spi­nal canal hema­toma
tes with VWD, includ­ing type 3 VWD, are uncom­mon, and life- out of a total of 133 525 obstet­ric spi­nal anes­the­sia sited dur­ing
threat­en­ing intra­cra­nial hem­or­rhage is very rare. Small case series, the same audit period.31 On the other hand, in the event of an
includ­ing 6 from Canada and 2 from the United Kingdom, show oper­a­tive deliv­ery, the alter­na­tive to neuraxial anes­the­sia would
that there were no neo­ na­
tal cases of intra­ cra­
nial hem­ or­
rhage be gen­eral anes­the­sia, which is asso­ci­ated with an 8 times higher
in infants with VWD.27,28 However, cephalohematoma has been risk of uter­ine atony and PPH, a 50 times higher risk of failed intu­
reported.29,30 In a cohort of 113 pedi­at­ric patients (aged 0-16 years) ba­tion, and low neo­na­tal Apgar scores.32 In women with IBDs, the
with VWD, cephalohematoma at birth was reported in 10 of 113 data on the risk of spi­nal canal hema­toma are scarce and lim­ited
(9%) chil­dren29; 50% of them were born by a ventouse deliv­ery. In to case reports and small case series, all­show­ing a safe admin­
the same study, bleed­ing from inva­sive mon­i­tor­ing using a fetal is­tra­tion of neuraxial anal­ge­sia/anes­the­sia when the coag­u­la­tion
scalp elec­trode was reported in 2 cases. In another study, 5 of 7 fac­tors are nor­mal­ized due to phys­i­o­logic changes of preg­nancy
(71%) chil­dren had a cephalohematoma after being born by a ven­ or with appro­pri­ate hemo­static cover. Two of the larg­est series
touse deliv­ery.30 Therefore, pre­cau­tions to reduce hem­or­rhagic included 15 and 33 cases of VWD,33,34 with no hemo­static treat­ment

554  |  Hematology 2021  |  ASH Education Program


Table 1. Risk strat­i­fi­ca­tion of bleed­ing risk for the fetus/neo­nate in women with VWD16

Risk level Possible or con­firmed fetal diag­no­sis Suggested man­age­ment of deliv­ery


High Type 3 VWD • Discuss MOD, tak­ing mater­nal and fetal fac­tors into con­sid­er­ation, but avoid
midcavity for­ceps, ventouse deliv­ery; FBS, FSE, rota­tional for­ceps, and exter­nal
cephalic ver­sion
Medium Type 2 VWD • Avoid midcavity for­ceps, ventouse, rota­tional for­ceps, and exter­nal cephalic ver­sion
• Judicious use of FBS and FSE to facil­i­tate vag­i­nal deliv­ery
Mild Clinically mod­er­ate or severe type • Consider avoid­ance of ventouse and exter­nal cephalic ver­sion
1 VWD in fam­ily • Judicious use of rota­tional for­ceps, FBS, and FSE
Unlikely to be at risk Clinically mild type 1 VWD in fam­ily •  No spe­cial pre­cau­tions
FBS, fetal blood sampling; FSE, fetal scalp electrode.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/552/1851794/552turan.pdf by guest on 13 December 2021


or only tranexamic acid (TXA) for those with mild VWD but fac­tor er­ies.35,37 Lack of con­sis­tency in the def­i­ni­tion used for PPH
con­cen­trate admin­is­tra­tion for mod­er­ate and severe cases. in the VWD lit­er­a­ture was rec­og­nized by a mul­ti­dis­ci­plin­ary
The UK guide­line on the man­age­ment of women with IBDs panel work­ing on the recent VWD guide­line38; the panel has
rec­om­mends avoid­ance of neuraxial block in women with VWD pro­posed a PPH def­i­ni­tion as “blood loss of 1000 mL or more
unless VWF activ­ity is more than 0.5 IU/mL and the hemo­static within 24 hours of birth or any blood loss with the poten­tial to
defect has been corrected.16 A tar­get VWF activ­ity level of 0.50 pro­duce hemo­dy­namic insta­bil­ity” to be used in future VWD
to 1.50 IU/mL is recommended to allow neuraxial block, and VWF research.38 Women with VWD have a greater risk of expe­ri­enc­
activ­ity should be maintained above 0.50 IU/mL while the neurax­ ing PPH,39 and a recent sys­tem­atic review reported an inci­
ial cath­e­ter is in place and for at least 6 hours after removal.35 In dence of pri­mary PPH of 34% among 811 deliv­er­ies in women
cases with a mild risk of bleed­ing (ie, type 1 VWD with corrected with VWD.40 In a large national cohort study includ­ing 4067
VWF activ­ity >0.5 IU/mL), hemo­static sup­port with TXA is suf­fi­ women with VWD,19 the risk of PPH was 6% in women with
cient.16 In women with type 2 and 3 VWD, ade­quate hemo­sta­ VWD com­pared with 4% in those with­out VWD, and women
sis may not be achieved despite fac­tor replace­ment and nor­mal with VWD were 5 times more likely to receive a blood trans­
lab­o­ra­tory VWF activ­ity level; such cases should be indi­vid­u­ally fu­sion after child­birth.19
assessed by an MDT with exper­tise in VWD. Although the eti­­ol­ogy for PPH is often mul­ti­fac­to­rial, uter­
Management of obstet­ric anal­ge­sia and anes­the­sia in women ine atony is the commonest obstet­ric cause.41 Other com­mon
with VWD requires a prior indi­vid­u­al­ized risk assess­ment and obstet­ric causes are retained pla­cen­tal tis­sue and gen­i­tal
plan­ning by the MDT. Risks and ben­e­fits of neuraxial block and tract trauma. In women with IBDs, coag­u­la­tion defect can be
its alter­na­tives should be con­sid­ered to help the mother make an the pri­mary or con­trib­ut­ing fac­tor to PPH. Prevention of PPH
informed deci­sion. Table 2 sum­ma­rizes con­di­tions for the use of should include obstet­ric mea­sures to min­i­mize risk of uter­ine
neuraxial block in women with IBDs. atony and birth trauma as well as hemo­static mea­sures to
cor­rect their coag­u­la­tion defect. Active man­age­ment of the
Postpartum hem­or­rhage third stage of labor, which incor­po­rates pro­phy­lac­tic use of
PPH is defined as blood loss of 500 mL or more within 24 hours uterotonic (oxy­to­cin), early clamping, and con­trolled cord
of child­birth and com­pli­cates on aver­age 3% to 6% of all­deliv­ trac­tion to deliver the pla­centa, reduces the risk of severe pri­mary

Table 2. Conditions for the use of regional block in women with IBDs dur­ing labor and deliv­ery36

Multidisciplinary man­age­ment involv­ing hema­tol­o­gists, anes­the­tists, obste­tri­cians, and the mother


Detailed coun­sel­ing on the ben­e­fits and risks of regional block and its alter­na­tives to help the mother make an informed deci­sion
Careful assess­ment of coag­u­la­tion sta­tus, includ­ing assess­ment of clot­ting fac­tor dur­ing the third tri­mes­ter, and bleed­ing phe­no­type, includ­ing per­
sonal and fam­ily bleed­ing his­tory
Availability of ther­a­peu­tic prod­ucts and lab­o­ra­tory facil­i­ties to ensure ade­quate response to treat­ment
Plan of man­age­ment made ante­na­tally dur­ing the third tri­mes­ter, clearly documented, and read­ily avail­­able to pro­fes­sion­als attend­ing the woman in
labor
Normalization of coag­u­la­tion defect by either a preg­nancy-induced rise in coag­u­la­tion fac­tors or the use of appro­pri­ate pro­phy­lac­tic treat­ment prior
to regional block pro­ce­dures
Meticulous tech­ni­cal skills in the admin­is­tra­tion of regional block by an expe­ri­enced anes­the­tist
Where an epi­du­ral cath­e­ter is placed, ade­quate hemo­sta­sis should be maintained prior to cath­e­ter removal, as the risk of bleed­ing is no less than
with inser­tion
Awareness and sur­veil­lance for symp­toms and signs of poten­tial com­pli­ca­tions

Prakash Singh Shekhawat


Management of von Willebrand dis­ease in preg­nancy  |  555
PPH greater than 1000 mL42 and should be implemented for deliv­ery as pos­si­ble or prior to inser­tion of a neuraxial cath­e­
all­women with IBDs. Additional uterotonic agents such as ter. The peak tar­get VWF activ­ity level is aimed at 1.0 IU/mL,
pros­ta­glan­din E2 (misoprostol) have been recommended for and the level should be maintained above 0.5 IU/mL until
pro­phy­lac­tic use in women with mod­er­ate and severe IBDs.25 hemo­sta­sis is secured.16 Monitoring of pre- and posttreatment
Obstetric man­age­ment should also include avoid­ance of fac­ lev­els of VWF activ­ity and fac­tor VIII lev­els is recommended
tors that increase the risk of PPH, such as prolonged labor, after deliv­ery, if labor is prolonged, or in case of exces­sive
mater­nal birth tract injury dur­ing vag­i­nal deliv­ery, and assur­ bleed­ing.16 There are a num­ber of plasma-derived prod­ucts
ance of ade­quate sur­gi­cal hemo­sta­sis dur­ing repair or dur­ing with dif­fer­ent VWF/FVIII ratios or recom­bi­nant VWF, which is
cesar­ean deliv­ery. devoid of FVIII. The choice of fac­tor con­cen­trate will depend
Hemostatic man­age­ment includes prior assess­ment of PPH on the local avail­abil­ity and FVIII lev­els in the third tri­mes­ter.9
risk and plan­ning an appro­pri­ate level of hemo­static cover. In An increased level of FVIII is asso­ci­ated with a risk of venous
women with VWD, risk assess­ment should be indi­vid­u­al­ized, throm­bo­em­bo­lism (VTE). Products that limit a patient’s expo­
tak­ing into con­sid­er­ation the VWF level dur­ing the third tri­ sure to FVIII allows for more fre­quent dos­ing of VWF if needed,
mes­ter, bleed­ing phe­no­type, pre­vi­ous response to treat­ment, with­out the risk of VTE sec­ond­ary to accu­mu­la­tion of FVIII.49

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/552/1851794/552turan.pdf by guest on 13 December 2021


and obstet­ ric risk fac­tors for PPH (eg, mul­ ti­
ple preg­ nancy, Plasma-derived con­cen­trates with a low FVIII are advis­able for
pla­centa previa). Several stud­ies have shown a high rate of women with a high third-tri­mes­ter FVIII level, such as women
PPH in women with VWD, with a third-tri­mes­ter VWF of <0.5 with type 2 VWD. Recombinant VWF con­cen­trates con­tain no
IU/dL despite pro­phy­lac­tic treat­ment with VWF con­cen­trates FVIII with a lon­ger half-life, now licensed for use in sev­eral
(plasma derived or recom­bi­nant).6,43-45 However, these stud­ies countries, and they can also be used in these women.9,44 In
pro­vide no data on treat­ment regimes, tar­get VWF activ­ity and women with type 2B VWD, plate­let count should be checked
fac­tor-level mon­i­tor­ing dur­ing treat­ment, cause of PPH, and the when in labor and plate­let trans­fu­sion is recommended to
use of other treat­ments such as uterotonics and TXA. main­tain a plate­let count of >50 × 109.18
There is an increased sys­ temic and local fibri­ no­ly­
sis after Women with VWD are also at risk of sec­ond­ary PPH and pro­
child­birth, and TXA inhib­its fibri­no­ly­sis and reduces blood loss longed and heavy lochia loss dur­ing the puer­pe­rium.50 A preg­
and mor­tal­ity with PPH.46 Prophylactic use of TXA is recom­ nancy-induced rise in VWF and FVIII returns to prepregnancy
mended for all­women with VWD: alone for those with a VWF lev­els within a few days after deliv­ery6,51 (Figure 2). For women
level >0.5 IU/mL and in con­junc­tion with desmopressin or VWF who are receiv­ing fac­tor con­cen­trates, it is impor­tant to ensure
con­cen­trate if VWF:RCo is <0.5 IU/mL.16,35 This can be started at VWF and FVIII lev­els are maintained above 0.5 IU/mL for 3 to
the onset of established labor and given 6 hourly and con­tin­ued 5 days after an uncom­pli­cated vag­i­nal deliv­ery and 5 to 7 days
for sev­eral weeks until lochia is light. The intra­ve­nous route may fol­low­ing oper­a­tive deliv­er­ies.52 In women with severe VWD,
be pre­ferred due to slower gas­tric emp­ty­ing and increased risk espe­cially type 3 VWD, VWF con­cen­trate may be required for
of vomiting in labor. a few weeks after deliv­ery.16 In addi­tion, TXA 1 g 6 hourly dur­ing
Desmopressin increases cir­cu­la­tory lev­els of VWF and FVIII the puer­pe­rium should be con­sid­ered in all­women with VWD
due to release of an endo­the­lial store of VWF. It is a valid until the lochia has stopped. All women should be briefed, with a
treat­ment option for VWD, pri­mar­ily type 1 VWD and selected safety net put in place before dis­charge regard­ing increased risk
type 2 cases. It is effec­tive in patients with base­line VWF and of sec­ond­ary PPH, and they are advised to report any exces­sive
FVIII lev­els higher than 0.10 IU/mL, but due to var­ia ­ bil­ity in vag­i­nal bleed­ing.
response, a prior test dose is recommended.10,35 Desmopres­ Pregnancy and the post­par­tum period are asso­ci­ated with an
sin is gen­er­ally contraindicated in type 2B VWD as it can tran­ increased risk of VTE. Women with VWD were no less likely to
siently exac­er­bate throm­bo­cy­to­pe­nia.47 It is not used in type
3 VWD because the clin­i­cal response is very poor or absent.9
A sys­tem­atic review of 216 preg­nan­cies in 30 stud­ies dem­
on­strated a safe and effec­tive use of desmopressin for the
pre­ven­tion and treat­ment of bleed­ing in preg­nancy and child­
birth in most cases.48 The most com­mon indi­ca­tion for its use
was as a pro­phy­lac­tic hemo­static agent for the pre­ven­tion of
PPH in 172 cases with no sig­nif­i­cant bleed­ing in 167 deliv­er­ies.
There were no adverse fetal out­comes. Maternal side effects
included well-tol­er­ated facial flash­ing and head­ache and only
1 case of water intox­i­ca­tion sei­zure.48 Thus, fluid intake should
be mon­i­tored and lim­ited to 1 to 1.5 L in 24 hours, and elec­
tro­lytes should be mon­it­ored if addi­tional fluid is required.
Repeated dos­ing of more than 2 to 3 doses 12 to 24 hours
apart should be avoided. Desmopressin should also be
avoided if fluid restric­tion or mon­i­tor­ing is not fea­si­ble and in
women with pre­eclamp­sia due to its vaso­con­stric­tive effect,
exac­er­bat­ing vas­cu­lar resis­tance and hyper­ten­sion.47
Factor replace­ment is required when desmopressin is inef­
fec­tive or contraindicated. Treatment with fac­tor con­cen­trate
is given when the mother is in active labor and as near to Figure 2. Postnatal changes in VWF.6

556  |  Hematology 2021  |  ASH Education Program


expe­ri­ence a pul­mo­nary embolism after child­birth than women (NSAIDS) use. Misoprostol is also widely used for the prevention and
with­out VWD in a large cohort study.19 Therefore, women should treatment of postpartum haemorrhage.
be care­fully assessed for throm­botic risk post­de­liv­ery against Rezan Abdul Kadir: Misoprostol is currently only approved by the
their risk of bleed­ing, and appro­pri­ate post­na­tal VTE pro­phy­laxis Food and Drug Administration (FDA) in the US for the prevention
should be advised. Low-molec­u­lar-weight hep­a­rin can be used and treatment of gastric ulcers secondary to non-steroidal anti-
for women with ade­quate cor­rec­tion of VWF and FVIII lev­els, but inflammatory drug (NSAID) use. Misoprostol is also widely used
mechan­i­cal meth­ods should be employed when fac­tor lev­els are for the prevention and treatment of postpartum haemorrhage.
less than 0.5 IU/mL.16

Correspondence
Management of neo­na­tes
Rezan Abdul Kadir, Royal Free Hospital NHS Foundation Trust,
VWF level is raised at birth, mak­ing diag­no­sis of mild VWD dif­
Pond Street, Hampstead, London NW3 2QG, UK; e-mail:rezan​
fi­
cult in the neo­ na­tal period and reduc­ ing neo­ na­tal bleed­ ing
­.abdul-kadir@nhs​­.net.
risk. However, fac­tor lev­els in neo­na­tes with severe type 1 and
types 2 and 3 remain low, and thus these neo­na­tes are at risk of

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/552/1851794/552turan.pdf by guest on 13 December 2021


bleed­ing com­pli­ca­tions. It is recommended that a cord blood References
sam­ple is obtained at the time of deliv­ery for assess­ment of FVIII 1. James PD, Goodeve AC. von Willebrand dis­ease. Genet Med. 2011;13(5):365-
and VWF anti­gen and activ­ity in these neo­na­tes. Cranial imag­ing 376.
2. Ludlam CA, Pasi KJ, Bolton-Maggs P, et al; UK Haemophilia Centre Doc­
and short-term pro­phy­laxis with fac­tor con­cen­trate should be
tors’ Organisation. A frame­work for genetic ser­vice pro­vi­sion for haemo­
con­sid­ered in neo­na­tes with type 3 VWD, espe­cially if deliv­ery philia and other inherited bleed­ing dis­or­ders. Haemophilia. 2005;11(2):
was poten­tially trau­matic.16 Neonates with reduced VWF are also 145-163.
at risk of mus­cle bleeds fol­low­ing intra­mus­cu­lar injec­tion, and 3. Cutler J, Chappell LC, Kyle P, Madan B. Third tri­mes­ter amnio­cen­te­sis for
vita­min K should be admin­is­tered orally in neo­na­tes with type diag­no­sis of inherited bleed­ing dis­or­ders prior to deliv­ery. Haemophilia.
2013;19(6):904-907.
2 and 3 VWD. Bleeding post­vac­ci­na­tion has also been reported 4. Leebeek FWG, Duvekot J, Kruip MJHA. How I man­age preg­nancy in car­ri­ers
in these neo­na­tes, and it was the sec­ond most fre­quent pedi­ of hemo­philia and patients with von Willebrand dis­ease. Blood. 2020;136
at­ric bleed­ing symp­tom in 1 study.30 Therefore, all­ immu­ni­za­ ­(19):2143-2150.
tions should be given sub­cu­ta­ne­ously with the smallest gauge 5. Nowak-Göttl U, Limperger V, Kenet G, et al. Developmental hemo­sta­sis: a
lifespan from neo­na­tes and preg­nancy to the young and elderly adult in a
needle.53 Review at the HTC with the pedi­at­ric team should be
Euro­pean white pop­u­la­tion. Blood Cells Mol Dis. 2017;67:2-13.
planned and arranged before dis­charge. 6. James AH, Konkle BA, Kouides P, et al. Postpartum von Willebrand fac­tor
lev­els in women with and with­out von Willebrand dis­ease and impli­ca­tions
for pro­phy­laxis. Haemophilia. 2015;21(1):81-87.
7. Castaman G, Tosetto A, Rodeghiero F. Pregnancy and deliv­ery in women
CLINICAL CASE (Con­t in­u ed) with von Willebrand’s dis­ease and dif­fer­ent von Willebrand fac­tor muta­tions.
Haematologica. 2010;95(6):963-969.
The mother opted for a planned cesar­ean sec­tion for fear of 8. Kruse-Jarres R, Johnsen JM. How I treat type 2B von Willebrand dis­ease.
labor and to avoid any risk for her baby. After appro­pri­ate coun­ Blood. 2018;131(12):1292-1300.
sel­ing, a planned cesar­ean sec­tion at 39 weeks’ ges­ta­tion was 9. Castaman G, James PD. Pregnancy and deliv­ery in women with von Wille­
brand dis­ease. Eur J Haematol. 2019;103(2):73-79.
performed under spi­nal anes­the­sia. As her FVIII was high dur­ 10. Castaman G, Goodeve A, Eikenboom J; Euro­pean Group on von Willebrand
ing the third tri­mes­ter, a plasma-derived con­cen­trate with a Disease. Principles of care for the diag­no­sis and treat­ment of von Wille­
low FVIII level was cho­sen for pro­phy­lac­tic treat­ment dur­ing brand dis­ease. Haematologica. 2013;98(5):667-674.
cesar­ean sec­tion and post­na­tally. First dose was given prior to 11. Kadir RA, Davies J. Hemostatic dis­or­ders in women. J Thromb Haemost.
2013;11(suppl 1):170-179.
inser­tion of a spi­nal cath­e­ter with 1 g intra­ve­nous TXA. Factor
12. van Galen K, Lavin L, Skouw-Rasmussen N, et  al. Euro­pean prin­ci­ples of
con­cen­trate was given 8 hourly for 24 hours, then 12 hourly on care for women and girls with inherited bleed­ing dis­or­ders. 2021. Accessed
day 2, and then daily for 5 days. Tranexamic acid 1 g was given 6/18/2021. https:­/­/eahad­.org­/eahad­-wgbd­-poc­-webinar­-on­-9­-march­/
intra­ve­nously in the first 24 hours and then orally for 4 weeks or 13. Kadir RA, Lee CA, Sabin CA, Pollard D, Economides DL. Pregnancy in
until lochia com­pletely stopped. women with von Willebrand’s dis­ease or fac­tor XI defi­ciency. Br J Obstet
Gynaecol. 1998;105(3):314-321.
She deliv­ered a baby boy weighing 2936 g under spi­nal anes­ 14. Lak M, Peyvandi F, Mannucci PM. Clinical man­i­fes­ta­tions and com­pli­ca­tions
the­sia with no com­pli­ca­tions. Estimated blood loss at deliv­ery of child­birth and replace­ment ther­apy in 385 Ira­nian patients with type
was 450 mL. Cord blood sam­ple indi­cated that her infant was 3 von Willebrand dis­ease. Br J Haematol. 2000;111(4):1236-1239.
affected by VWD. Vitamin K was admin­is­trated orally. Mother and 15. Skeith L, Rydz N, O’Beirne M, Goodyear D, Li H, Poon M-C. Pregnancy loss
in women with von Willebrand dis­ease: a sin­gle-cen­ter pilot study. Blood
baby were discharged home on day 3 post­na­tally with a hemo­
Coagul Fibrinolysis. 2017;28(5):393-397.
philia nurse and a com­mu­nity mid­wife to visit daily for a week. 16. Pavord SR, Rayment R, Madan B, et al. Management of inherited bleed­ing
dis­or­ders in preg­nancy. Green-top Guideline No. 71 (joint with UKHCDO).
BJOG. 2017;124(8):e193-e263.
Conflict-of-inter­est dis­clo­sure 17. Quenby S, Gallos ID, Dhillon-Smith RK, et al. Miscarriage mat­ters: the epi­
de­mi­o­log­i­cal, phys­i­cal, psy­cho­log­i­cal, and eco­nomic costs of early preg­
Ozlem Turan: no com­pet­ing finan­cial inter­ests to declare. nancy loss. Lancet. 2021;397(10285):1658-1667.
Rezan Abdul Kadir: no com­pet­ing finan­cial inter­ests to declare. 18. Millar CM. Von Willebrand dis­ease. In: Kadir RA, James PD, Lee C, eds.
Inherited Bleeding Disorders in Women. 2nd ed. John Wiley; 2019:83-94.
Off-label drug use 19. James AH, Jamison MG. Bleeding events and other com­pli­ca­tions dur­ing
preg­nancy and child­birth in women with von Willebrand dis­ease. J Thromb
Ozlem Turan: Misoprostol is currently only approved by the Food and
Haemost. 2007;5(6):1165-1169.
Drug Administration (FDA) in the US for the prevention and treatment 20. Kavle JA, Stoltzfus RJ, Witter F, Tielsch JM, Khalfan SS, Caulfield LE. Associa­
of gastric ulcers secondary to non-steroidal anti-inflammatory drugs tion between anae­mia dur­ing preg­nancy and blood loss at and after deliv­ery
Prakash Singh Shekhawat
Management of von Willebrand dis­ease in preg­nancy  |  557
among women with vag­i­nal births in Pemba Island, Zanzibar, Tanzania. J 38. Connell NT, James PD, Brignardello-Petersen R, et al. von Willebrand dis­ease:
Health Popul Nutr. 2008;26(2):232-240. pro­pos­ing def­i­ni­tions for future research. Blood Adv. 2021;5(2):565-569.
21. Mirza FG, Abdul-Kadir R, Breymann C, Fraser IS, Taher A. Impact and man­ 39. James AH. More than men­or­rha­gia: a review of the obstet­ric and gynaeco­
age­ment of iron defi­ciency and iron defi­ciency ane­mia in women’s health. logical man­i­fes­ta­tions of bleed­ing dis­or­ders. Haemophilia. 2005;11(4):295-
Expert Rev Hematol. 2018;11(9):727-736. 307.
22. Nair M, Choudhury MK, Choudhury SS, et al. Association between mater­ 40. Punt MC, Waning ML, Mauser-Bunschoten EP, et  al. Maternal and neo­
nal anae­mia and preg­nancy out­comes: a cohort study in Assam, India. BMJ na­tal bleed­ing com­pli­ca­tions in rela­tion to peripartum man­age­ment in
Glob Health. 2016;1(1):e000026. women with von Willebrand dis­ ease: a sys­ tem­
atic review. Blood Rev.
23. Malec LM, Moore CG, Yabes J, Li J, Ragni MV. Postpartum haemorrhage in 2020;39:100633.
women with von Willebrand dis­ease: an obser­va­tional study of the Penn­ 41. Chandraharan E, Krishna A. Diagnosis and man­age­ment of post­par­tum
sylvania Health Care Cost Containment Council (PHC4) data­base. Haemo- haemorrhage. BMJ. 2017;358:j3875.
philia. 2015;21(5):e442-e445. 42. Begley CM, Gyte GM, Devane D, McGuire W, Weeks A, Biesty LM. Active
24. Johnsen JM. Current approaches to preg­nancy and child­birth in women ver­sus expec­tant man­age­ment for women in the third stage of labour.
with von Willebrand dis­ease. Clin Adv Hematol Oncol. 2018;16(4):254-257. Cochrane Database Syst Rev. 2019;2:CD007412.
25. Huq FY, Kadir RA. Management of preg­ nancy, labour and deliv­ ery in 43. Kadir RA, Chi C. Women and von Willebrand dis­ease: con­tro­ver­sies in
women with inherited bleed­ing dis­or­ders. Haemophilia. 2011;17(suppl 1): diag­no­sis and man­age­ment. Semin Thromb Hemost. 2006;32(6):605-615.
20-30. 44. Machin N, Ragni MV. Recombinant vs plasma-derived von Willebrand fac­

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/552/1851794/552turan.pdf by guest on 13 December 2021


26. Kulkarni AA, Osmond M, Bapir M, et al. The effect of labour on the coag­ul­a­ tor to pre­vent post­par­tum hem­or­rhage in von Willebrand dis­ease. Blood
tion sys­tem in the term neo­nate. Haemophilia. 2013;19(4):533-538. Adv. 2020;4(14):3234-3238.
27. Labarque V, Stain AM, Blanchette V, Kahr WH, Carcao MD. Intracranial hae­ 45. Stoof SC, van Steenbergen HW, Zwagemaker A, et al. Primary post­par­tum
morrhage in von Willebrand dis­ease: a report on six cases. Haemophilia. haemorrhage in women with von Willebrand dis­ease or carriership of hae­
2013;19(4):602-606. mophilia despite specialised care: a ret­ro­spec­tive sur­vey. Haemophilia.
28. Chalmers E, Alamelu J, Collins P, et al; Paediatric and Rare Disorders 2015;21(4):505-512.
Working Parties of UKHCDO. Intracranial haemorrhage in chil­dren with 46. Shakur H, Elbourne D, Gülmezoglu M, et al. The WOMAN Trial (World Mater­
inherited bleed­ing dis­or­ders in the UK 2003-2013. J Thromb Haemost. nal Antifibrinolytic Trial): tranexamic acid for the treat­ment of post­par­tum
2018;24(4):641-647. haemorrhage: an inter­ na­tional randomised, dou­ ble blind pla­cebo con­
29. Biss TT, Blanchette VS, Clark DS, et al. Quantitation of bleed­ing symp­toms trolled trial. Trials. 2010;11:40.
in chil­dren with von Willebrand dis­ease: use of a stan­dard­ized pedi­at­ric 47. Laffan MA, Lester E, O’Donnell JS, et al. The diag­no­sis and man­age­ment of
bleed­ing ques­tion­naire. J Thromb Haemost. 2010;8(5):950-956. von Willebrand dis­ease: a United Kingdom Haemophilia Centre Doctors
30. Sanders YV, Fijnvandraat K, Boender J, et al; WiN Study Group. Bleeding Organization guide­line approved by the Brit­ish Committee for Standards
spec­trum in chil­dren with mod­er­ate or severe von Willebrand dis­ease: rel­ in Haematology. Br J Haematol. 2014;167(4):453-465.
e­vance of pedi­at­ric-spe­cific bleed­ing. Am J Hematol. 2015;90(12):1142-1148. 48. Trigg DE, Stergiotou I, Peitsidis P, Kadir RA. A sys­tem­atic review: the use of
31. Cook TM, Counsell D, Wildsmith JA; Royal College of Anaesthetists Third desmopressin for treat­ment and pro­phy­laxis of bleed­ing dis­or­ders in preg­
National Audit Project. Major com­ pli­
ca­
tions of cen­tral neuraxial block: nancy. Haemophilia. 2012;18(1):25-33.
report on the Third National Audit Project of the Royal College of Anaes­ 49. Coppola A, Franchini M, Makris M, Santagostino E, Di Minno G, Mannucci
thetists. Br J Anaesth. 2009;102(2):179-190. PM. Thrombotic adverse events to coag­u­la­tion fac­tor con­cen­trates for
32. Delgado C, Ring L, Mushambi MC. General anaes­the­sia in obstet­rics. BJA treat­ment of patients with haemophilia and von Willebrand dis­ease: a sys­
Educ. 2020;20(6):201-207. tem­atic review of pro­spec­tive stud­ies. Haemophilia. 2012;18(3):e173-e187.
33. Boyd SC, O’Connor AD, Horan MA, et al. Analgesia, anaes­the­sia and obstet­ 50. Chi C, Bapir M, Lee CA, Kadir RA. Puerperal loss (lochia) in women with or
ric out­come in women with inherited bleed­ing dis­or­ders. Eur J Obstet with­out inherited bleed­ing dis­or­ders. Am J Obstet Gynecol. 2010;203(1):56.
Gynecol Reprod Biol. 2019;239:60-63. e1-56.e5.
34. Chi C, Lee CA, England A, Hingorani J, Paintsil J, Kadir RA. Obstetric anal­ge­ 51. Huq FY, Kulkarni A, Agbim EC, Riddell A, Tuddenham E, Kadir RA. Changes
sia and anaes­the­sia in women with inherited bleed­ing dis­or­ders. Thromb in the lev­els of fac­tor VIII and von Willebrand fac­tor in the puer­pe­rium.
Haemost. 2009;101(6):1104-1111. Haemophilia. 2012;18(2):241-245.
35. Connell NT, Flood VH, Brignardello-Petersen R, et al. ASH ISTH NHF WFH 52. National Hemophilia Foundation. MASAC Document 265—MASAC Guide-
2021 guide­lines on the man­age­ment of von Willebrand dis­ease. Blood Adv. lines for Pregnancy and Perinatal Management of Women with Inherited
2021;5(1):301-325. Bleeding Disorders and Carriers of Hemophilia A or B. National Hemophilia
36. Bouthors AS, England A, Kadir RA. Analgesia and anaes­the­sia for preg­nant Foundation; 2021.
women with inherited bleed­ing dis­or­ders. In: Kadir RA, James PD, Lee C, 53. Carcao M, Bouskill V. The new­born. In: Kadir RA, James PD, Lee C, eds.
eds. Inherited Bleeding Disorders in Women. 2nd ed. John Wiley; 2019:191- Inherited Bleeding Disorders in Women. 2nd ed. John Wiley; 2019:205-221.
202.
37. Knight M, Callaghan WM, Berg C, et al. Trends in post­par­tum hem­or­rhage
in high resource countries: a review and rec­om­men­da­tions from the Inter­
national Postpartum Hemorrhage Collaborative Group. BMC Pregnancy © 2021 by The Amer­i­can Society of Hematology
Childbirth. 2009;9:55. DOI 10.1182/hema­tol­ogy.2021000321

558  |  Hematology 2021  |  ASH Education Program


PREGNANCY IN SPECIAL POPULATIONS: CHALLENGES AND SOLUTIONS

Prevention and management of venous


thromboembolism in pregnancy: cutting through
the practice variation

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/559/1851791/559skeith.pdf by guest on 13 December 2021


Leslie Skeith
Division of Hematology and Hematological Malignancies, Department of Medicine, Foothills Medical Centre, University of Calgary, Calgary, Canada

There is clinical practice variation in the area of prevention and management of venous thromboembolism (VTE) in
pregnancy. There are limited data and differing recommendations across major clinical practice guidelines, especially
relating to the role of postpartum low-molecular-weight heparin (LMWH) for patients with mild inherited thrombophilia
and those with pregnancy-related VTE risk factors. This chapter explores the issues of practice variation and related data
for postpartum VTE prevention. Controversial topics of VTE management in pregnancy are also reviewed and include
LMWH dosing and the role of anti-Xa level monitoring, as well as peripartum anticoagulation management around labor
and delivery.

LEARNING OBJECTIVES
• Describe the practice variation and limited data in the area of postpartum VTE prevention
• Describe management of VTE in pregnancy and an approach to anticoagulation management around labor and
delivery

There is an increased risk of VTE (deep vein thrombosis


CLINICAL CASE 1 [DVT] and pulmonary embolism [PE]) during pregnancy
You are seeing a 30-year-old G1P0 woman who is 32 that affects approximately 1.2 per 1000 deliveries, which is
weeks pregnant and is known to be heterozygous for the a 5- to 10-fold increased risk compared with the nonpreg-
factor V Leiden gene mutation. She has no personal his- nant population.1-3 The antepartum period and postpartum
tory of venous thromboembolism (VTE), but her mother period each carry a similar VTE risk (0.6 per 1000 deliveries).1
has a history of an unprovoked VTE. She has no other VTE VTE is a leading cause of direct maternal mortality and can
risk factors (RFs) or medical history. She is referred to you have important long-term consequences, including post-
for counseling relating to the role of postpartum throm- thrombotic syndrome, post-PE syndrome associated with
boprophylaxis. functional limitations, and a reduced quality of life.4-9 One of
the greatest predictors of postthrombotic syndrome after
a pregnancy-associated VTE is having a postpartum DVT.5
The risk of VTE during pregnancy and the postpartum
period is higher in patients with additional VTE RFs, such
CLINICAL CASE 2 as an inherited thrombophilia or pregnancy-related RFs
A 25-year-old G2P2 woman had an urgent unplanned (eg, a woman with a combination of prolonged antepartum
cesarean delivery (CD) overnight because of failure to immobility and elevated body mass index ≥25kg/m2 has an
progress in labor. Her CD was uncomplicated, and she adjusted odds ratio of 40 to develop a postpartum VTE).10,11
had an estimated blood loss of 750 mL. Should the patient Because the VTE incidence in the postpartum period is still
receive postpartum thromboprophylaxis? relatively low (even in these higher-risk groups) and large
randomized controlled trials have been difficult to conduct,

Prakash Singh Shekhawat


VTE prevention and management in pregnancy | 559
clin­i­cal prac­tice guide­line rec­om­men­da­tions regard­ing thrombo- 10 years ago, and so data used to inform deci­sions may be out of
prophylaxis are largely based on obser­va­tional data and expert date.15,17 Experts do not agree on what VTE risk thresh­old to use,
opin­ion. Information about the VTE risk, bleeding risk, other which is high­lighted in the clin­i­cal prac­tice guide­line meth­od­ol­
downsides of low-molecular-weight heparin, related mortality, ogy described.13,26 Although most abso­lute VTE risk thresh­olds in
and patient preferences are taken into account when formulating guide­lines (if reported at all­) are set some­where between 1% and
guideline recommendations.12 3% to con­sider LMWH use, some experts have recommended a
LMWH thromboprophylaxis is indi­ cated dur­ ing preg­nancy lower VTE thresh­old of 0.2% post-CD based on deci­sion mod­el­
for indi­vid­u­als with a prior unpro­voked or hor­mone-asso­ci­ated ing.27 Others argue that set­ting a higher VTE thresh­old or num­
VTE, and LMWH thromboprophylaxis is indi­cated in the 6-week ber needed to treat is required to bal­ance a higher bleed­ing case
post­par­tum period for indi­vid­u­als with any prior VTE (includ­ fatal­ity rate (com­pared with the VTE case fatal­ity rate) or other
ing unpro­voked, hor­mone-asso­ci­ated, or pro­voked VTE).13 In com­pli­ca­tions seen, which is pri­mar­ily extrap­o­lated from non-
patients with­out a per­sonal his­tory of VTE, there is more uncer­ pregnancy data.12,28,29 Understanding what mat­ters to patients is
tainty about the role of thromboprophylaxis. There is gen­eral still poorly under­stood. Bates et al30 interviewed preg­nant patients
guide­line agree­ment (but not com­plete con­sen­sus) on LMWH and iden­ti­fied that patients placed sim­i­lar health state val­ues on

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/559/1851791/559skeith.pdf by guest on 13 December 2021


thromboprophylaxis for an indi­vid­ual with a potent thrombo- recur­rent VTE and obstet­ric bleed­ing, but this also var­ied among
philia. There are more differences across clinical practice guide- indi­vid­u­als. Furthermore, how patient val­ues and pref­er­ences are
lines for the role of LMWH use between suggested LMWH use incor­po­rated into the guide­line rec­om­men­da­tions remains var­i­
and clin­i­cal prac­tice guide­lines for indi­vid­u­als who have a more able and unclear, and fur­ther work is needed in this area.
com­mon inherited thrombophilia (eg, het­ero­zy­gous fac­tor V Even after mak­ing sta­tis­ti­cal assump­tions about the lim­ited
Leiden) or those with preg­nancy-related RFs (eg, pre­eclamp­sia, data and decid­ing on a VTE risk thresh­old to rec­om­mend LMWH,
urgent CD, post­par­tum hem­or­rhage).13-17 we are still learn­ing how this risk is reflected in real-life prac­tice.
LMWH thromboprophylaxis rec­om­men­da­tions dif­fer across For exam­ple, the 2018 ASH guide­line panel recommended a 1%
clin­i­cal prac­tice guide­lines for patients with inherited thrombo- VTE risk thresh­old for post­par­tum LMWH use.13 In a large US data­
philia and are outlined in Table 3 of the 2018 Amer­ic ­ an Society base study of over 1.2 mil­lion cesar­ean deliv­er­ies, patients who
of Hematology (ASH) guide­lines for man­age­ment of VTE in the were iden­ti­fied as “ele­vated risk” by the 2018 ASH guide­lines had
con­text of preg­nancy.13 There has been less empha­sis placed on an actual VTE inci­dence of 20.0 (14.9-25.7) per 1000 cesar­ean
the role and prac­tice var­i­a­tion of LMWH thromboprophylaxis for deliv­er­ies (2% risk) at 6 weeks post­par­tum, whereas other clin­i­cal
preg­nancy-related RFs, includ­ing after CD. A sum­mary of thrombo- prac­tice guide­lines’ “high-risk” categories had lower actual VTE
prophylaxis guide­line state­ments for LMWH use post-CD is listed inci­dences (Table 2).22 In con­trast, in a sin­gle-cen­ter study, the
in Table 1. In a US data­base that cap­tured over 1.2 mil­lion women highest cal­cu­lated VTE risk was approx­im ­ a­tely 0.5% using a risk
post-CD, the pro­por­tion of patients who would have the­o­ret­i­cally score derived by Sultan et al31, which is lower than the VTE risk
received LMWH thromboprophylaxis based on VTE RFs was 0.3%, threshold cutoff suggested by the ASH guidelines.23
16.2%, 73.4%, and 0.2%, according to dif­fer­ing rec­om­men­da­tions There have been preg­nancy and post­par­tum VTE risk scores
by the 2011 Amer­ic ­ an College of Obstetricians and Gynecologists derived and exter­nally val­i­dated from large reg­is­try data­bases,
(ACOG), 2012 Amer­i­can College of Chest Physicians (ACCP), 2015 but these scores have yet to be stud­ied pro­spec­tively or incor­
Royal College of Obstetricians and Gynaecologists (RCOG), and po­rated into guide­lines.31-33 Several lim­i­ta­tions exist in cohort and
the 2018 ASH guide­lines, respec­tively22 (Table 2). Similarly, in a data­base stud­ies, includ­ing dif­fer­ent RF and VTE def­i­ni­tions used
ret­ro­spec­tive review in Geneva, Switzerland, of 344 post­par­tum in admin­is­tra­tive data­bases, miss­ing data for some VTE RFs, low
women who deliv­ered in 1 month at 1 cen­ter, the the­o­ret­i­cal use num­bers of patients with high-risk con­di­tions, and, most impor­
of post­par­tum LMWH thromboprophylaxis after CD included 35% tant, the actual LMWH pro­phy­laxis use is often not accu­rately
(ACOG), 40% (ACCP), 89% (RCOG), and 0% (ASH) (Table 2 for all­ cap­tured.
deliv­er­ies). For both exam­ples, the ASH guide­lines do not focus Although esti­mat­ing what the true ben­e­fit of LMWH is remains
on this spe­cific area of rec­om­men­da­tion.23 unknown, what is equally chal­leng­ing is esti­mat­ing the bleed­
Why do clin­i­cal prac­tice guide­lines dif­fer? The some­what ing risk and pos­si­ble wound com­pli­ca­tions, cost, and bur­den
obvi­ous answer is because lower-qual­ity evi­dence is avail­­able to of LMWH injec­tions. Introducing an anti­co­ag­u­lant post-CD may
inform deci­sions, which includes the lim­ited data on the actual the­ o­ret­

cally affect wound healing of the inci­ sion by caus­ ing
ben­e­fits and risks of thromboprophylaxis in this pop­u­la­tion. In an local­ized bleed­ing, but lit­tle data remain in this area. In a large
updated Cochrane sys­tem­atic review of avail­­able ran­dom­ized tri­ ret­ro­spec­tive cohort study of 24 229 deliv­er­ies, VTE and bleed­ing
als, the lim­ited evi­dence remains “very uncer­tain about the ben­e­ out­comes were assessed before and after a stan­dard­ized throm-
fits and harms of VTE thromboprophylaxis” dur­ing preg­nancy and boprophylaxis hos­pi­tal pro­to­col was implemented. There was
the post­par­tum period.24 More spe­cif­i­cally, there are dif­fer­ences 1.2% and 15.6% of anti­co­ag­u­lant use before and after pro­to­col
in how the lim­ited data are interpreted, which can lead to dif­fer­ implementation, respec­tively. There were no dif­fer­ences in VTE
ent guide­line rec­om­men­da­tions. This includes issues of pub­li­ca­ rates seen before and after pro­to­col implementation, but there
tion date, what sta­tis­ti­cal strat­e­gies are used to com­bine mul­ti­ple was a 2-fold higher rate of super­fi­cial wound hema­to­mas.31 With
VTE RFs together, what VTE risk thresh­old is set to rec­om­mend com­pet­ing risks and pos­si­ble com­pli­ca­tions, under­stand­ing the
LMWH, if a VTE scor­ing sys­tem is used, and the role of expert and patient per­spec­tive on LMWH use, as well as how we best com­
patient opin­ion.12,25 When should older clin­i­cal prac­tice guide­ mu­ni­cate the benefit and risk of LMWH to our patients, is still
lines be con­sid­ered “retired”? Earlier guide­lines, such as those needed.
from the ACCP and the Aus­tra­lian and New Zealand Journal of Two pilot tri­als were conducted that assessed the fea­si­bil­
Obstetrics and Gynaecology (ANZJOG), were published almost ity of ran­dom­iz­ing postpartum women with VTE RFs to LMWH

560  |  Hematology 2021  |  ASH Education Program


Table 1. LMWH pro­phy­laxis rec­om­men­da­tions across major clin­i­cal prac­tice guide­lines after cesar­ean deliv­ery

Characteristic ASH 201813 RCOG 201516 SOGC 201418 ACCP 201215 ANZJOG 201217
Elective For women with no or 1 No No For women under­go­ing No; early mobi­li­za­tion
CD alone clin­i­cal RF (exclud­ing a cesar­ean sec­tion with­ and avoid­ance of
known thrombophilia or out addi­tional throm­ dehy­dra­tion
his­tory of VTE), the ASH bo­sis RFs, we rec­om­
Emergent All women who have had cesar­ean No Following emer­gency
guide­line panel sug­gests mend against the use of
CD alone sec­tions should be con­sid­ered cesar­ean sec­tion
against antepartum throm­bo­sis pro­phy­laxis
for thromboprophylaxis with thromboprophylaxis
post­par­tum pro­phy­laxis other than early mobi­li­
LMWH for 10 days after deliv­ with LMWH or UFH is
(con­di­tional rec­om­men­ za­tion (grade 1B).
ery apart from those hav­ing recommended for at
da­tion, low cer­tainty in
an elec­tive cesar­ean sec­tion least 5 days or lon­ger
evi­dence about effects).
who should be con­sid­ered for until recov­ery of full
thromboprophylaxis with LMWH mobil­ity (group con­sen­
for 10 days after deliv­ery if they sus level 1).
have any addi­tional RFs
(grade C).
Elective Not stated See above; con­sid­ered for Postpartum thromboprophylaxis For women at increased ≥1 major and ≥2 minor
CD + RF thromboprophylaxis with LMWH should be con­sid­ered in the risk of VTE after cesar­ RFs: Postpartum
for 10 days after deliv­ery pres­ence of mul­ti­ple clin­i­cal or ean sec­tion because of thromboprophylaxis
(grade C). preg­nancy-related RFs when the pres­ence of for ≥5 days or until fully
the over­all abso­lute risk is 1 major or at least 2 mobile; 1 major or 2
esti­mated to be greater than minor RFs, minor RFs: Consider
1% drawn from the fol­low­ing we sug­gest phar­ grad­u­ated com­pres­sion
group­ings: ma­co­logic stock­ings.
Any 2 of the fol­low­ing RFs (emer­ thromboprophylaxis
gency cesar­ean sec­tion counts (pro­phy­lac­tic LMWH) or
as 1 RF) (II-2B) mechan­i­cal pro­phy­laxis

Prakash Singh Shekhawat


LMWH until dis­charge up to 2 (elas­tic stock­ings or
weeks if 2 RFs inter­mit­tent pneu­matic
com­pres­sion) in those
with con­tra­in­di­ca­tions
to anti­co­ag­u­lants while
in the hos­pi­tal fol­low­ing
deliv­ery rather than no
pro­phy­laxis (grade 2B).

VTE pre­ven­tion and man­age­ment in preg­nancy  |  561


Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/559/1851791/559skeith.pdf by guest on 13 December 2021
Table 1. (continued)

Characteristic ASH 201813 RCOG 201516 SOGC 201418 ACCP 201215 ANZJOG 201217
Emergent Not stated All women who have had cesar­ean Postpartum thromboprophylaxis Further details in Table 3: Following emer­gency
CD + RF sec­tions should be con­sid­ered should be con­sid­ered in the Requires pres­ence of cesar­ean sec­tion,
for thromboprophylaxis with pres­ence of mul­ti­ple clin­i­cal or at least 1 major RF OR thromboprophylaxis
LMWH for 10 days after deliv­ preg­nancy-related RFs when pres­ence of at least with LMWH or UFH is
ery apart from those hav­ing the over­all abso­lute risk is esti­ 2 minor RFs (planned recommended for at
an elec­tive cesar­ean sec­tion mated to be >1% drawn from cesar­ean sec­tion) OR 1 least 5 days or lon­ger
who should be con­sid­ered for the fol­low­ing group­ings: minor RF in the set­ting until recov­ery of full
thromboprophylaxis with LMWH Any 3 or more of the fol­low­ing of an emer­gency cesar­ mobil­ity (group con­sen­
for 10 days after deliv­ery if they RFs (elec­tive cesar­ean sec­tion ean sec­tion sus level 1).
have any addi­tional RFs counts as 1 RF) (II-2B)
(grade C). LMWH until dis­charge up to 2
weeks if 1 RF
Note: These mostly include preg­nancy-related RFs and do not review inherited thrombophilia guid­ance unless stated below.
ACCP15: Major RFs: Immobility, post­par­tum hem­or­rhage ≥1000 mL with sur­gery, pre­vi­ous VTE, pre­eclamp­sia with fetal growth restric­tion, thrombophilia (AT defi­ciency, fac­tor V Leiden, pro­throm­
bin gene muta­tion), med­i­cal con­di­tions (SLE, heart dis­ease, sickle cell dis­ease), blood trans­fu­sion, post­par­tum infec­tion. Minor RFs: BMI >30 kg/m2, mul­ti­ple preg­nancy, PPH >1 L, smok­ing >10
cig­a­rettes/d, fetal growth restric­tion, thrombophilia (pro­tein C and S defi­ciency), pre­eclamp­sia.
RCOG16: See Table 1 of the 2015 RCOG guide­lines.

562  |  Hematology 2021  |  ASH Education Program


SOGC18: Any 2 of the fol­low­ing RFs: BMI ≥30 kg/m2, smok­ing >10 cig­a­rettes/d, pre­eclamp­sia, intra­uter­ine growth restric­tion, pla­centa previa, emer­gency cesar­ean sec­tion, peripartum or post­
par­tum blood loss of >1 L or blood prod­uct replace­ment, any low-risk thrombophilia (pro­tein C or pro­tein S defi­ciency), het­ero­zy­gous fac­tor V Leiden, or pro­throm­bin gene muta­tion 20210A,
mater­nal car­diac dis­ease, SLE, sickle cell dis­ease, inflam­ma­tory bowel dis­ease, var­i­cose veins, ges­ta­tional dia­be­tes, pre­term deliv­ery, still­birth. Any 3 or more of the fol­low­ing RFs: age >35 years,
par­ity ≥2, any assisted repro­duc­tive tech­nol­ogy, mul­ti­ple preg­nancy, pla­cen­tal abrup­tion, pre­ma­ture rup­ture of mem­branes, elec­tive cesar­ean sec­tion, mater­nal can­cer.
ANZJOG17: Major RFs: Elective cesar­ean sec­tion, BMI ≥30 kg/m2, immo­bi­li­za­tion, med­i­cal comorbidity (eg, inflam­ma­tory bowel dis­ease, SLE, pneu­mo­nia), pre­eclamp­sia, sys­temic infec­tion. Minor
RFs: Age >35 years, prolonged labor (>24 hours), smoker, PPH >1000 mL, exten­sive per­i­neal trauma and prolonged repair, gross var­i­cose veins.
Descriptions used for evi­dence grad­ing and con­sen­sus:
ASH 2018: The panel used the GRADE approach to assess the cer­tainty in the evi­dence and for­mu­late rec­om­men­da­tions.
RCOG 2015: Described in a sep­a­rate meth­od­ol­ogy arti­cle: “using the SIGN meth­od­ol­ogy, the qual­ity of the evi­dence used and the direct­ness of its appli­ca­tion should be incor­po­rated into the
for­mu­la­tion and grad­ing of the rec­om­men­da­tion.”
SOGC 2014: “The qual­ity of evi­dence in this doc­u­ment was rated using the cri­te­ria described in the Report of the Cana­dian Task Force on Preventative Health Care.”
ACCP 2012: “We followed the approach artic­u­lated by Grades of Recommendations, Assessment, Development, and Evaluation for for­mu­la­tion of rec­om­men­da­tions.”
ANZJOG 2012: “To assess the level of con­sen­sus with the rec­om­men­da­tions, all­authors were sent a spread­sheet list­ing all­the rec­om­men­da­tions and were asked to indi­cate whether they agreed
or disagreed with each state­ment. Recommendations were then graded with the fol­low­ing lev­els of con­sen­sus: Group Consensus Level 1—com­plete con­sen­sus: all­ten authors in agree­ment;
Group Consensus Level 2—par­tial con­sen­sus: eight of ten authors in agree­ment; Group Consensus Level 3—no con­sen­sus—two or more authors disagreed with rec­om­men­da­tion.”
AT, anti­throm­bin; BMI, body mass index; GRADE, Grading of Recommendations Assessment, Development and Evaluation; PPH, post­par­tum hem­or­rhage; SLE, sys­temic lupus erythematosus;
SOGC, Society of Obstetricians and Gynaecology of Canada.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/559/1851791/559skeith.pdf by guest on 13 December 2021


Table 2. Differences in clin­i­cal prac­tice guide­lines for the use of post­par­tum thromboprophylaxis and asso­ci­ated VTE risk

Author, year, and Population n (% who Proportion Statements of actual LMWH Incidence of VTE in
loca­tion under­went CD) meet­ing cri­te­ria use women who met
for LMWH guide­line cri­te­ria for
pro­phy­laxis LMWH pro­phy­laxis
Pamerola,19 2016, USA Post-CD; cross-sec­tional 293 ACOG: 1.0% “At the cen­tre where this study NR
chart review at 2 time CD: 100% RCOG: 85% was performed, hep­a­rin is
points in 2013-2014 ACCP: 34.8% admin­is­tered empir­i­cally to
all­women after CD unless
there is a spe­cific con­tra­in­
di­ca­tion.”
Omunakwe,20 2016, UK All deliv­er­ies; 4 weeks in 227 RCOG: 46.6% NR NR
Sep­tem­ber-Octo­ber 2015 CD: 35%

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/559/1851791/559skeith.pdf by guest on 13 December 2021


O’Shaughnessy,21 2019, All deliv­er­ies; cross-sec­tional 21 019 ACOG: 8% NR NR
Ireland study of pro­spec­tively col­ CD: 32% RCOG: 37%
lected data; Jan­u­ary 2015 ACCP: 7%
to Decem­ber 2017 Australia/New
Zealand: 23%
SOGC: 15%
Federspiel et al,22 2021, Post-CD; Nationwide 1 390 603 ACOG: 0.3% Unknown VTE inci­dence per 1000
USA Readmissions Database, CD: 100% RCOG: 73.4% deliv­er­ies:
Octo­ber 2015 to Decem­ ACCP: 16.2% ACOG: 19.5 (14.9-23.9)
ber 2017 ASH: 0.2%* RCOG: 1.9 (1.8-2.0)
ACCP: 4.2 (3.9-4.6)
ASH: 20.0 (14.9-25.7)
Gassmann et al,23 2021, All deliv­er­ies in Jan­u­ary 2019; 344 ACOG: 8.7% 24% Calculated based on
Switzerland ret­ro­spec­tive chart review CD: 23.3% RCOG: 40.1% “Standard of care is to pre­ Sultan risk score:
(34.9%-45.5%) scribe thromboprophylaxis ACOG: 0.20%
ACCP: 9.9% to women with CD. For RCOG: 0.12%
ASH: 0%* women with vag­i­nal deliv­ ACCP: 0.20%;
ery, LMWH is restricted to
those with thrombophilia or
prior VTE, fol­low­ing indi­vid­
ual hemo­sta­sis con­sul­ta­tion.”
*The 2018 ASH guide­lines did not spe­cif­i­cally com­ment on post-CD thromboprophylaxis use (see Table 1).
NR, not reported.

vs pla­cebo/no LMWH for 10 to 21 days. Unfortunately, both pilot PARTUM trial. In the absence of an avail­­able research study, the
tri­als were not fea­si­ble due to low par­tic­i­pant recruit­ment rates 2018 ASH guide­line panel sug­gests against thromboprophylaxis
that aver­aged <1 par­tic­i­pant enrolled per month per cen­ter.32,33 use for this patient, even with the pres­ence of a fam­ily his­tory
In a sur­vey of 306 patients who were eli­gi­ble but did not par­tic­i­ of VTE. Because other clin­i­cal prac­tice guide­lines rec­om­mend
pate, 32% were too overwhelmed or pre­oc­cu­pied in the post­par­ thromboprophylaxis in this sce­nario, fur­ther dis­cus­sion is needed
tum period to con­sider research, and 28.4% declined because with the patient before mak­ing an informed deci­sion, includ­ing
they wanted to avoid LMWH injec­tions.36 Although par­tic­i­pant high­light­ing this prac­tice var­i­a­tion and reviewing the patient’s
num­bers were too small in the pilot PROSPER (PostpaRtum PrO- val­ues and pref­er­ences. Regardless of the choice to use LMWH
phylaxiS for PE Randomized Control Trial Pilot) tri­als to com­ment or not post­par­tum, symp­toms of VTE and when to seek med­i­cal
on the effi­cacy or safety of post­par­tum LMWH use, there was 1 atten­tion should be reviewed. A review of addi­tional preg­nancy-
major bleed­ing event and 2 clin­i­cally rel­e­vant non­ma­jor bleed­ing related RFs should also be com­pleted at the time of deliv­ery.
events among 16 par­tic­i­pants, which high­lights that high-qual­ity
tri­als are still needed to deter­mine the true risk and ben­e­fit of
post­par­tum thromboprophylaxis.36 The pilot PARTUM (Postpar-
tum Aspirin to Reduce Thromboembolism Undue Morbidity) trial CLINICAL CASE 2 (Con­tin­ued)
is ongo­ing, to see the fea­si­bil­ity of low-dose aspi­rin for 6 weeks
to pre­vent VTE in post­par­tum women with VTE RFs, com­pared The abso­lute risk of post­par­tum VTE after urgent CD is less
with pla­cebo (clinicaltrials​­.gov ID NCT04153760). than 1%, and it is still uncer­tain what the true ben­e­fit of LMWH
thromboprophylaxis is in this sit­ u­a­
tion, weighing the side
effects and sys­tem-level cost. Although I would not rec­om­
mend LMWH thromboprophylaxis in this case, oth­ers would
rec­om­mend LMWH use, such as in the 2015 RCOG guide­lines.
CLINICAL CASE 1 (Con­t in­u ed) Instead, I would advo­cate for early mobi­li­za­tion and reassess
Given the lim­
ited data in this area, the patient should be the role of LMWH if addi­tional post­par­tum VTE RFs develop.
approached to par­tic­i­pate in a research study such as the pilot
Prakash Singh Shekhawat
VTE pre­ven­tion and man­age­ment in preg­nancy  |  563
Management of VTE in preg­nancy out­comes. In the larg­est non-ran­dom­ized study of 26 par­tic­i­
pants (11 received anti-Xa level mon­i­tor­ing and 15 did not receive
anti-Xa level mon­i­tor­ing), there was no dif­fer­ence in recur­rent
CLINICAL CASE 3 VTE or bleed­ing reported.53 Based on lim­ited evi­dence, the 2018
You are see­ing a 29-year old G2P1 woman who devel­oped a symp­ ASH guide­line panel sug­gests either a once-daily or twice-daily
tom­atic PE at 20 weeks’ ges­ta­tion based on a high-prob­a­bil­ity LMWH reg­i­men be used and suggested against anti-Xa level
V/Q scan with large mismatched defects in the left lower lobe. mon­i­tor­ing with the poten­tial excep­tion of higher-risk sce­nar­ios,
Her vitals are nor­mal, and she has no exam­i­na­tion find­ings of DVT. such as in those patients with obe­sity or advanced renal dys­
Her preg­nancy has been unre­ mark­able, and she has no other func­tion.13 Table 3 sum­ma­rizes var­i­ous clin­i­cal prac­tice guide­line
med­i­cal his­tory. Her only med­i­ca­tion is a pre­na­tal vita­min. What rec­om­men­da­tions for once- vs twice-daily LMWH, anti-Xa level
anti­co­ag­u­lant reg­i­men do you rec­om­mend for her preg­nancy? mon­i­tor­ing, and peripartum anticoagulation man­age­ment (dis-
cussed fur­ther in case 4).

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/559/1851791/559skeith.pdf by guest on 13 December 2021


Therapeutic-dose LMWH is the recommended treat­ ment
for preg­nant patients with acute VTE because it does not cross CLINICAL CASE 3 (Con­tin­ued)
the pla­centa and has an improved safety pro­file com­pared with
Whatever anti­co­ag­u­lant reg­i­men is cho­sen, it is impor­tant to
unfractionated hep­ a­
rin (UFH). Vitamin K antag­ o­
nists are not
have close fol­low-up and mon­i­tor­ing of symp­toms, espe­cially in
recommended in preg­nancy due to known ter­a­to­ge­nic­ity, and
the first month of treat­ment. The rec­om­men­da­tions presented
direct oral anti­co­ag­u­lants still have lim­ited safety infor­ma­tion
by the 2018 ASH guide­lines are a patient-focused approach
(with an International Society on Thrombosis and Haemostasis
that includes min­i­miz­ing the num­ber of injec­tions and lab­o­ra­
reg­is­try ongo­ing).37,38 Side effects of LMWH use dur­ ing preg­
tory vis­its. Although this aligns with my gen­eral prac­tice and
nancy include a small risk of impor­tant bleed­ing antepartum
approach for the major­ity of patients, I would con­sider esca­
(~0.5%), injec­tion site bruis­ing and reac­tions, and a very rare
lat­ing to a twice-daily LMWH and/or anti-Xa level mon­i­tor­ing
pos­si­bil­ity of hep­a­rin-induced throm­bo­cy­to­pe­nia.39 Unlike UFH,
in the first month of VTE treat­ment in those with higher-risk
pro­phy­lac­tic or inter­me­di­ate-dose LMWH does not reduce bone
fea­tures, such as those with exten­sive VTE bur­den, recur­rent
min­eral den­sity dur­ing preg­nancy, but less data are avail­­able
VTE despite anticoagulation, or patients with a high-risk throm-
for ther­a­peu­tic doses.40,41 Systemic thrombolysis should only be
bophilia such as antiphospholipid syn­drome.
reserved for patients with PE who have life-threat­en­ing hemo­dy­
namic com­pro­mise or car­diac arrest given the exces­sive bleed­
ing risk (among 83 women treated with sys­temic thrombolysis:
mater­ nal sur­ vival, 94%; fetal sur­vival, 88%; antepartum major Peripartum man­age­ment of anticoagulation
bleed­ing, 17.5%; post­par­tum major bleed­ing, 58.3%).42 Beyond
these state­ments, there remains sig­nif­i­cant prac­tice var­i­a­tion on
CLINICAL CASE 4
the details of anticoagulation man­age­ment in preg­nancy.43
A 38-year-old G0 woman had a large PE 2 years ago after start-
As preg­nancy advances, there is an increased vol­ume of dis­
ing a com­bined oral con­tra­cep­tive pill. After receiv­ing 6 months
tri­bu­tion and glo­mer­u­lar fil­tra­tion rate, so there is the poten­
of anticoagulation, she stopped anticoagulation and the con­
tial for increased clear­ance of LMWH.38,44 Because of these
tra­cep­tive pill and had a pro­ges­ter­one-only intra­uter­ine device
preg­nancy-spe­cific changes, there remains con­tro­versy on the
placed. She is now inter­ested in get­ting preg­nant. You meet
use of once- vs twice-daily LMWH dos­ing and the role of anti-
her for a pre­con­cep­tion coun­sel­ing visit and recommended
Xa level mon­i­tor­ing of LMWH. In a 2014 Cana­dian sur­vey of 69
starting pro­phy­lac­tic-dose LMWH dur­ing preg­nancy and the
hema­tol­o­gists, inter­nists, and obste­tri­cians, there was con­sid­
6-week post­par­tum period to pre­vent recur­rent VTE. She has
er­able prac­tice var­i­a­tion in acute VTE man­age­ment. Within the
sev­eral ques­tions about what this means for her deliv­ery and if
first month of a VTE event, par­tic­i­pants used either once-daily
she will be ­able to get an epi­du­ral for pain con­trol.
(36%) or twice-daily (62%) LMWH, and anti-Xa level mon­i­tor­ing
was com­pleted weekly (20%), monthly (26%), weekly or monthly
in spe­cial pop­u­la­tions only (23%), or never (20%).40
A sys­tem­atic review in non­preg­nant patients showed sim­i­lar One of the more chal­ leng­ing areas with lit­ tle high-qual­ity
out­comes of once- vs twice-daily LMWH dos­ing.45 To date, there data is when and how to stop anticoagulation around labor and
has been no clear out­come dif­fer­ence in recur­rent VTE seen in deliv­ery, as well as when to resume post­par­tum anticoagulation.
ret­ro­spec­tive cohorts of preg­nant patients treated with dif­fer­ Systematic reviews report the var­i­able and low-qual­ity ret­ro­
ent reg­i­mens.46-47 Information related to anti-Xa level mon­i­tor­ing spec­tive data avail­­able that phy­si­cians rely on to make deci­sions
is lim­ited to small cohorts of preg­nant patients who received about anti­co­ag­u­lant man­age­ment.37,56,57 One par­tic­u­lar chal­lenge
dif­fer­ent ther­a­peu­tic-dose LMWH reg­i­mens in which phy­si­cians in the lit­er­a­ture has been the dif­fer­ent def­i­ni­tions of peripartum
titrated LMWH doses based on var­i­ous anti-Xa level tar­gets; no bleed­ ing; peripartum bleed­ ing def­i­
ni­tions may be based on
major safety sig­nals were iden­ti­fied across dif­fer­ent strat­e­gies, esti­mated blood loss alone (variably mea­sured across cen­ters),
albeit with lim­ited data from a small num­ber of patients.48,54 One a decrease in hemo­glo­bin, blood trans­fu­sion, hos­pi­tal readmis-
chal­lenge in interpreting anti-Xa lev­els is that a spe­cific “anti- sions, repeat sur­gery, or wound com­pli­ca­tions.57-62 To min­i­mize
Xa level range” in preg­nancy is not known, because there is no this var­i­at­ ion, the International Society on Thrombosis and Hae-
data avail­­able to cor­re­late anti-Xa lev­els with VTE or bleed­ing mostasis Committee of Women’s Health Issues in Thrombosis

564  |  Hematology 2021  |  ASH Education Program


Table 3. Recommendations of VTE man­age­ment in preg­nancy across major clin­i­cal prac­tice guide­lines

Characteristic ASH 201813 RCOG 201552 SOGC 201418 ACCP 201215 ANZJOG 201217
Once- vs twice- For preg­nant women with LMWH should be given in For the treat­ment of acute No rec­om­men­da­tion Treatment of acute VTE in
daily LMWH acute VTE treated with doses titrated against VTE in preg­nancy, we preg­nancy should be with
LMWH, the ASH guide­ the woman’s book­ing or rec­om­mend adher­ing LMWH given once daily or
line panel sug­gests either early preg­nancy weight. to the man­u­fac­turer’s twice daily at ther­a­peu­tic
once-per-day or twice- There is insuf­fi­cient evi­ recommended dos­ing for doses. There is cur­rently
per-day dos­ing reg­i­mens dence to rec­om­mend indi­vid­ual LMWH based insuf­fi­cient evi­dence to
(con­di­tional rec­om­men­da­ whether the dose of on the woman’s cur­rent favor one dose reg­i­men
tion, very low cer­tainty in LMWH should be given weight. (II-1A). over the other (group con­
evi­dence about effects). once daily or in two LMWH can be admin­is­ sen­sus level 1).
divided doses (grade C). tered once or twice a day Women with PE or more
depending on the agent exten­sive DVT (ie,
selected (III-C). iliofemoral throm­bo­sis)
dur­ing preg­nancy should
receive ini­tial treat­ment
with twice-daily LMWH for
at least 8 to 12 weeks, after
which time a reduc­tion to a
once-daily reg­i­men may be
con­sid­ered (group con­sen­
sus level 2).
Anti-Xa level For preg­nant women receiv­ Routine mea­sure­ment of No rec­om­men­da­tion No rec­om­men­da­tion There is insuf­fi­cient evi­dence
mon­i­tor­ing ing ther­a­peu­tic-dose peak anti-Xa activ­ity for In the text: con­sid­er­ation In the text: to rec­om­mend mon­i­tor­ing
LMWH for the treat­ment patients tak­ing LMWH for should be given to ini­tial Given the absence of large of anti-Xa lev­els to guide
of VTE, the ASH guide­line treat­ment of acute VTE in mon­i­tor­ing of anti-Xa activ­ stud­ies using clin­i­cal end dos­ing in women on ther­a­
panel sug­gests against preg­nancy or post­par­ ity, dur­ing the first month points that dem­on­strate an peu­tic dose LMWH (group
rou­tine mon­i­tor­ing of anti- tum is not recommended of treat­ment only, to tar­get opti­mal ther­a­peu­tic anti-Xa con­sen­sus level 1).

Prakash Singh Shekhawat


FXa lev­els to guide dos­ing except in women at a level of 0.6 to 1.0 U/mL LMWH range or that dose
(con­di­tional rec­om­men­ extremes of body weight 4 hours after injec­tion, adjust­ments increase the
da­tion, low cer­tainty in (<50 and 90 kg or more) bear­ing in mind that tar­get safety or effi­cacy of ther­apy,
evi­dence about effects). or with other com­pli­cat­ lev­els will vary with the the lack of accu­racy and
ing fac­tors (eg, with renal LMWH used. However, the reli­abil­ity of the mea­sure­
impair­ment or recur­rent cost of the assay, the lack ment, the lack of cor­re­la­tion
VTE) (grade C). of cor­re­la­tion with clin­i­cal with risk of bleed­ing and
events, and the var­i­abil­ity recur­rence, and the cost of
between assays make the the assay, rou­tine mon­i­
util­ity of mon­i­tor­ing anti- tor­ing with anti-Xa lev­els is
FXa activ­ity in preg­nancy dif­fi­cult to jus­tify.
con­tro­ver­sial.

VTE pre­ven­tion and man­age­ment in preg­nancy  |  565


Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/559/1851791/559skeith.pdf by guest on 13 December 2021
Table 3. (continued)

Characteristic ASH 201813 RCOG 201552 SOGC 201418 ACCP 201215 ANZJOG 201217
Peripartum For preg­nant women receiv­ The woman tak­ing LMWH Women receiv­ing pro­ For preg­nant women receiv­ No rec­om­men­da­tion
anticoagulation ing ther­a­peu­tic-dose for main­te­nance ther­apy phy­lac­tic, inter­me­di­ ing adjusted-dose LMWH
man­age­ment LMWH for the man­age­ment should be advised that ate-dose, or ther­a­peu­tic ther­apy and where deliv­ery
of VTE, the ASH guide­line once she is in established anticoagulation should is planned, we rec­om­mend
panel sug­gests sched­uled labor or thinks that she is have a dis­cus­sion about dis­con­tin­u­a­tion of LMWH
deliv­ery with prior dis­ in labor, she should not options at least 24 hours prior to
con­tin­u­a­tion of anti­co­ag­ inject any fur­ther hep­a­rin. for anal­ge­sia/anes­the­sia induc­tion of labor or cesar­
u­lant ther­apy (con­di­tional When VTE occurs at term, prior to deliv­ery (III-B). ean sec­tion (or expected
rec­om­men­da­tion, very low con­sid­er­ation should Switching from time of neuraxial anes­the­
cer­tainty in evi­dence about be given to the use of thromboprophylactic sia) rather than con­tinu­ing
effects). intra­ve­nous UFH, which is LMWH to a pro­phy­lac­tic LMWH up until the time of
For preg­nant women receiv­ more eas­ily manip­u­lated dose of UFH at term (37 deliv­ery (grade 1B).
ing pro­phy­lac­tic-dose (grade D). weeks) may be con­sid­ered
LMWH, the ASH guide­line Where deliv­ery is planned, to allow for more options
panel sug­gests against either by elec­tive cesar­ with respect to labor anal­
sched­uled deliv­ery with ean sec­tion or induc­tion ge­sia (III-L).
dis­con­tin­u­a­tion of pro­ of labor, LMWH main­te­ Discontinue pro­phy­lac­tic or
phy­lac­tic anticoagulation nance ther­apy should be inter­me­di­ate-dose LMWH

566  |  Hematology 2021  |  ASH Education Program


com­pared with allowing discontinued 24 hours or UFH upon the onset
spon­ta­ne­ous labor (con­ prior to planned deliv­ery of spon­ta­ne­ous labor or
di­tional rec­om­men­da­tion, (grade D). the day before a planned
very low cer­tainty in evi­ induc­tion of labor or cesar­
dence about effects). ean sec­tion (II-3B).
Time of neuraxial Regional anes­thetic or anal­ For women tak­ing LMWH, For preg­nant women receiv­ Table 3: LMWH—pro­phy­lac­tic
anes­the­sia after ge­sic tech­niques should neuraxial anes­the­sia can ing adjusted-dose LMWH dose: ensure a min­i­mum of
last dose of not be under­taken until be admin­is­tered as a: ther­apy and where deliv­ery 12 hours after LMWH dose
LMWH at least (a) Prophylactic dose: a min­i­ is planned, we rec­om­mend before the per­for­mance of
24 hours after the last mum of 10 to 12 hours after dis­con­tin­u­a­tion of LMWH a neuraxial block or removal
dose of ther­a­peu­tic the last dose (III-B) at least 24 hours prior to of a neuraxial cath­e­ter.
LMWH (grade D). (b) Therapeutic dose: after induc­tion of labor or cesar­ LMWH—ther­a­peu­tic dose:
24 hours since the last ean sec­tion (or expected pref­er­a­ble to avoid ther­a­
dose (III-B) time of neuraxial anes­the­ peu­tic dos­ing with cath­e­ter
Neuraxial anes­the­sia must be sia) rather than con­tinu­ing in situ; wait at least 24 hours
avoided in a woman who LMWH up until the time of after the last dose of LMWH
is fully anticoagulated or in deliv­ery (grade 1B). before performing neuraxial
whom there is evi­dence of block­ade or remov­ing a
altered coag­u­la­tion (II-3A). neuraxial cath­e­ter.
See Table 1 for descrip­tions used for evi­dence grad­ing and con­sen­sus.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/559/1851791/559skeith.pdf by guest on 13 December 2021


and Haemostasis devel­oped and published stan­dard­ized preg­ Two mul­ti­cen­ter inter­na­tional pro­spec­tive cohort stud­ies are
nancy and peripartum bleed­ ing out­ come def­i­ni­tions, which ongo­ing (PREP and GO and the PANDA stud­ies) and will use stan­
focus on the inter­ven­tions needed to treat impor­tant blood loss dard­ized VTE and bleed­ing def­i­ni­tions to pro­vide more infor­ma­
rather than on esti­mated blood loss alone.63 tion on the risk esti­ma­tes of VTE, bleed­ing, patient-focused and
There is con­tro­versy on what to do for a patient tak­ing pro­ health care uti­li­za­tion out­comes that mat­ter to patients, pro­vid­
phy­lac­tic-dose LMWH because there is a rel­a­tively low bleed­ing ers, and other stake­hold­ers.
risk, and access to neuraxial anes­the­sia can occur 12 hours after Postpartum, ret­ro­spec­tive cohort data report an increased
the last anti­ co­
ag­u­
lant dose, according to sev­ eral guide­ lines, bleed­ing risk if ther­a­peu­tic-dose anticoagulation is resumed too
includ­ing the Amer­i­can Society of Regional Anesthesia and the soon after deliv­ery. Initiating ther­a­peu­tic-dose LMWH too soon
Society for Obstetric Anesthesia and Perinatology (Table 3).64,65 may lead to a seri­ous post­op­er­a­tive bleed­ing com­pli­ca­tion in
The 2018 ASH guide­line panel sug­gests a spon­ta­ne­ous labor which the patient may be off of anticoagulation for longer, which
over a timed induc­tion of labor for preg­nant patients tak­ing pro­ could then increase the risk of VTE. In a ret­ro­spec­tive cohort study
phy­lac­tic-dose LMWH, but acknowl­edg­ing this deci­sion is also of 232 con­sec­u­tive women on ther­a­peu­tic anticoagulation who
based on indi­vid­ual patient val­ues and pref­er­ences.13 In a ret­ro­ deliv­ered in Que­bec, Canada, between 2003 and 2015, resum­ing

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/559/1851791/559skeith.pdf by guest on 13 December 2021


spec­tive cohort study of 199 patients tak­ing pro­phy­lac­tic-dose ther­a­peu­tic anticoagulation within approx­i­ma­tely 15 hours68 after
LMWH in preg­nancy, approx­i­ma­tely 90% of those in spon­ta­ne­ CD and approximately 9 hours within vag­i­nal deliv­ery was asso­
ous labor were eli­gi­ble for neuraxial anes­the­sia, which did not ci­ated with a higher risk of a com­pos­ite of major hem­or­rhagic
dif­fer based on if the patient was pri­mip­a­rous (first preg­nancy) com­pli­ca­tions (requir­ing trans­fu­sion, hos­pi­tal­i­za­tion, vol­ume
or mul­tip­a­rous.66 In this ret­ro­spec­tive study, there was nota­bly resus­ci­ta­tion, trans­fer to an inten­sive care unit, or sur­gery) and
less time off of anticoagulation for patients in the spon­ta­ne­ous major wound com­pli­ca­tions. Starting with a pro­phy­lac­tic-dose
labor group com­pared with the induc­tion of labor group (last LMWH post­de­liv­ery or delaying ther­a­peu­tic anticoagulation for
LMWH injec­tion to deliv­ery inter­val: 25.8 vs 48.2 hours, respec­ lon­ger appears safer. Practically, for most patients, resum­ing
tively).66 If a patient val­ues access to neuraxial anes­the­sia for pain daily pro­phy­lac­tic-dose LMWH the fol­low­ing day after deliv­ery
con­trol, then other options include a planned induc­tion of labor (~12-24 hours post­de­liv­ery) can be done, such as for case 4.
or stop­ping LMWH early depending on the sce­nario and asso­ci­ I follow the approach of starting with pro­phy­lac­tic-dose LMWH
ated VTE risk. For exam­ple, the last dose of LMWH can be given before esca­lat­ing to ther­a­peu­tic-dose LMWH on day 2 or 3
the day prior to an induc­tion of labor to ensure that at least 12 post­de­liv­ery if needed. For higher-risk cases, such as an acute
hours have passed since the last anti­co­ag­u­lant dose was given VTE within 30 days, this approach should be mod­i­fied and may
for the patient to be eli­gi­ble for neuraxial anes­the­sia. However, in include starting with twice-daily LMWH or intra­ ve­
nous UFH
prac­tice, this is often chal­leng­ing because the induc­tion of labor 6 to 12 hours post­de­liv­ery. Postpartum, either LMWH or vita­min K
tim­ing may not be known until the actual day due to hos­pi­tal antag­o­nists are safe with breastfeeding. Data indi­cate that direct
logis­tics, and the induc­tion of labor dura­tion is often var­i­able and oral anti­co­ag­u­lants cross into breastmilk and should be avoided
can be prolonged in some patients. A dis­cus­sion between mul­ti­ for breastfeeding women until more data are avail­­able.69-70
dis­ci­plin­ary care pro­vid­ers can be help­ful to coor­di­nate logis­tics
and the details of the induc­tion of labor, to pro­vide accu­rate risks Conflict-of-inter­est dis­clo­sure
and ben­e­fits of each approach with the patient. Leslie Skeith has received research funding from CSL Behring,
For patients receiving ther­a­peu­tic-dose LMWH, because of and hon­o­rar­ium from Leo Pharma and Sanofi.
the poten­tial for excess bleed­ing risk dur­ing deliv­ery and delayed
access to neuraxial anes­the­sia of 24 hours after the last LMWH Off-label drug use
dose,64,65 the 2018 ASH guide­line panel rec­om­mends a planned Leslie Skeith: the use of low-molecular-weight heparins is
labor (eg, induc­tion of labor), with the last dose of LMWH given off-label use for the prevention and treatment of VTE during
24 hours prior to induc­tion/deliv­ery.13 If the VTE is not acute (>3-6 pregnancy and the postpartum period. The use of aspirin for pre-
months), then an alter­na­tive approach (albeit with lim­ited data) vention of postpartum VTE is off-label and should only be used in
includes reduc­ing the LMWH dose closer to deliv­ery from a ther­ the setting of a clinical trial.
a­peu­tic dose to a pro­phy­lac­tic dose, to allow for spon­ta­ne­ous
deliv­ery. If the VTE is within 2 weeks of deliv­ery, then using a Correspondence author
more cau­tious anti­co­ag­u­lant reg­i­men may be con­sid­ered to Leslie Skeith, Division of Hematology and Hematological Malig-
min­i­mize time off of anticoagulation, such as starting or switch- nancies, Department of Medicine, Foothills Medical Centre, Uni-
ing to a twice-daily LMWH or arranging admis­sion to the hos­pi­ versity of Calgary, Room C210, 1403-29th Street NW, Calgary
tal for intra­ve­nous UFH. An inferior vena cava (IVC) fil­ter inser­tion T2N 2T9, Alberta, Canada; e-mail: laskeith@ucalgary​­.ca.
may be con­sid­ered if the VTE is within 2 weeks of deliv­ery, but
this is largely based on expert opin­ion. IVC fil­ter inser­tion should
References
­otherwise be avoided when pos­si­ble due to the chal­lenges of 1. Kourlaba G, Relakis J, Kontodimas S, Holm MV, Maniadakis N. A sys­tem­
inser­tion and pos­si­ble migra­tion relat­ing to the gravid uterus and atic review and meta-anal­y­sis of the epi­de­mi­­ol­ogy and bur­den of venous
IVC changes, and the deci­sion should be made in con­sul­ta­tion throm­bo­em­bo­lism among preg­nant women. Int J Gynaecol Obstet.
with a mul­ti­dis­ci­plin­ary spe­cial­ist team. The fail­ure-to-retrieve 2016;132(1):4-10.
2. Heit JA, Kobbervig CE, James AH, Petterson TM, Bailey KR, Melton LJ III.
rate of IVC fil­ters among 80 preg­nant patients was 11.3%, so it is Trends in the inci­dence of venous throm­bo­em­bo­lism dur­ing preg­nancy
impor­tant to remove the IVC fil­ter after deliv­ery once anticoagu- or post­par­tum: a 30-year pop­u­la­tion-based study. Ann Intern Med. 2005;
lation is safely resumed.67 143(10):697-706.

Prakash Singh Shekhawat


VTE pre­ven­tion and man­age­ment in preg­nancy  |  567
3. Pomp ER, Lenselink AM, Rosendaal FR, Doggen CJ. Pregnancy, the post­ bo­lism pro­phy­laxis after cesar­ean deliv­ery in selected guide­lines. J Thromb
par­tum period and prothrombotic defects: risk of venous throm­bo­sis in the Haemost. 2021;19(3):830-838.
MEGA study. J Thromb Haemost. 2008;6(4):632-637. 23. Gassmann N, Viviano M, Righini M, Fontana P, Martinez de Tejada B, Blon-
4. Knight M, Bunch K, Tuffnell D, et  al. Saving Lives, Improving Mothers’ don M. Estimating the risk thresh­olds used by guide­lines to rec­om­mend
Care: les­sons learned to inform mater­nity care from the UK and Ireland post­par­tum thromboprophylaxis. J Thromb Haemost. 2021;19(2):452-
Confidential Enquiries into Maternal Deaths and Morbidity 2016-2018, 459.
published Dec. 2020. https://www.nhsbmenetwork.org.uk/wp-content 24. Middleton P, Shepherd E, Gomersall JC. Venous throm­bo­em­bo­lism pro­phy­
/uploads/2021/03/MBRRACE-UK_Maternal_Report_Dec_2020_v10-1.pdf. laxis for women at risk dur­ing preg­nancy and the early post­na­tal period.
5. Wik HS, Jacobsen AF, Sandvik L, Sandset PM. Prevalence and pre­dic­tors Cochrane Database Syst Rev. 2021;3:CD001689.
for post-throm­botic syn­drome 3 to 16 years after preg­nancy-related venous 25. Sibai BM, Rouse DJ. Pharmacologic thromboprophylaxis in obstet­ rics:
throm­bo­sis: a pop­u­la­tion-based, cross-sec­tional, case-con­trol study. J broader use demands bet­ter data. Obstet Gynecol. 2016;128(4):681-684.
Thromb Haemost. 2012;10(5):840-847. 26. Bates SM, Middeldorp S, Rodger M, James AH, Greer I. Guidance for the
6. Kahn SR, Hirsch AM, Akaberi A, et  al. Functional and exer­cise lim­i­ta­tions treat­ment and pre­ven­tion of obstet­ric-asso­ci­ated venous throm­bo­em­bo­
after a first epi­sode of pul­mo­nary embolism: results of the ELOPE pro­spec­ lism. J Thromb Thrombolysis. 2016;41(1):92-128.
tive cohort study. Chest. 2017;151(5):1058-1068. 27. Blondon M. Thromboprophylaxis after cesar­ean sec­tion: deci­sion anal­y­sis.
7. Hunter R, Noble S, Lewis S, Bennett P. Long-term psy­cho­so­cial impact Thromb Res. 2011;127(suppl 3):S9-S12.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/559/1851791/559skeith.pdf by guest on 13 December 2021


of venous throm­bo­em­bo­lism: a qual­i­ta­tive study in the com­mu­nity. BMJ 28. Kotaska A. Postpartum venous throm­bo­em­bo­lism pro­phy­laxis may cause
Open. 2019;9(2):e024805. more harm than ben­e­fit: a crit­i­cal anal­y­sis of inter­na­tional guide­lines
8. van Es J, Exter PL Den, Kaptein AA, et al. Quality of life after pul­mo­nary embo- through an evi­dence-based lens. BJOG. 2018;125(9):1109-1116.
lism as assessed with SF-36 and PEmb-QoL. Thromb Res. 2013;132:500-505. 29. Rodger M. Pregnancy and venous throm­bo­em­bo­lism: “TIPPS” for risk strat­
9. Kahn SR, Ducruet T, Lamping DL, et al. Prospective eval­u­a­tion of health- i­fi­ca­tion. Hematology Am Soc Hematol Educ Program. 2014;2014(1):387-
related qual­ity of life in patients with deep venous throm­bo­sis. Arch Intern 392.
Med. 2005;165(10):1173-1178. 30. Bates SM, Alonso-Coello P, Tikkinen KA, et  al. Women’s val­ues and pref­
10. Croles FN, Nasserinejad K, Duvekot JJ, Kruip MJ, Meijer K, Leebeek FW. er­ences and health state val­ua ­ ­tions for thromboprophylaxis dur­ing preg­
Pregnancy, thrombophilia, and the risk of a first venous throm­bo­sis: sys­ nancy: a cross-sec­tional inter­view. Thromb Res. 2016;140:22-29.
tem­atic review and Bayes­ian meta-anal­y­sis. BMJ. 2017;359:j4452. 31. Sultan AA, West J, Grainge MJ, et  al. Development and val­i­da­tion of risk
11. Jacobsen AF, Skjeldestad FE, Sandset PM. Incidence and risk pat­terns of pre­dic­tion model for venous throm­bo­em­bo­lism in post­par­tum women:
venous throm­bo­em­bo­lism in preg­nancy and puer­pe­rium-a reg­is­ter-based mul­ti­na­tional cohort study. BMJ. 2016;355:i6253.
case-con­trol study. Am J Obstet Gynecol. 2008;198(2):233.e1-233.e7. 32. Dargaud Y, Rugeri L, Vergnes MC, et al. A risk score for the man­age­ment of
12. Skeith L. Preventing venous throm­bo­em­bo­lism dur­ing preg­nancy and preg­nant women with increased risk of venous throm­bo­em­bo­lism: a multi-
post­par­tum: cross­ing the thresh­old. Hematol Am Soc Hematol Educ Pro- centre pro­spec­tive study. Br J Haematol. 2009;145(6):825-835.
gram. 2017;2017(1):160-167. 33. Lu B, Sultan AA, West J, et  al. External val­i­da­tion of a model to pre­dict
13. Bates SM, Rajasekhar A, Middeldorp S, et al. Amer­i­can Society of Hematology women most at risk of post­par­tum venous throm­bo­em­bo­lism: mater­nity
2018 guide­lines for man­age­ment of venous throm­bo­em­bo­lism: venous throm­ clot risk [published online 8 June 2021]. Thromb Res.
bo­em­bo­lism in the con­text of preg­nancy. Blood Adv. 2018;2(22):3317-3359. 34. Lu MY, Blanchard CT, Oglesby KR, et al. Safety and effi­cacy of a risk-based
14. Amer­i­can College of Obstetricians and Gynecologists’ Committee on Prac- obstet­ric hep­a­rin-based thromboprophylaxis reg­i­men. Am J Obstet Gyne-
tice Bulletins—Obstetrics. ACOG Practice Bulletin No. 196: throm­bo­em­bo­ col. 2021;224(2):S41-S42.
lism in preg­nancy. Obstet Gynecol. 2018;132(1):e1-e17. 35. Rodger MA, Phil­lips P, Kahn SR, James AH, Konkle BA; PROSPER Investi-
15. Bates SM, Greer IA, Middeldorp S, Veenstra DL, Prabulos A-M, Vandvik P-O. gators. Low-molec­u­lar-weight hep­a­rin to pre­vent post­par­tum venous
VTE, thrombophilia, antithrombotic ther­apy, and preg­nancy: Antithrom- throm­bo­em­bo­lism: a pilot randomised pla­cebo-con­trolled trial. Thromb
botic Therapy and Prevention of Thrombosis, 9th ed: Amer­i­can College Haemost. 2015;113(1):212-216.
of Chest Physicians Evidence-Based Clinical Practice Guidelines. Chest. 36. Rodger MA, Phil­lips P, Kahn SR, et al; PROSPER Investigators. Low molec­
2012;141(2, suppl):e691S-e736S. u­lar weight hep­a­rin to pre­vent post­par­tum venous throm­bo­em­bo­lism:
16. No GG. Reducing the Risk of Venous Thromboembolism During Pregnancy a pilot study to assess the fea­si­bil­ity of a ran­dom­ized, open-label trial.
and the Puerperium Pregnancy and the Puerperium. Green-Top Guide- Thromb Res. 2016;142:17-20.
line No. 37a. Royal College of Obstetricians and Gynaecologists; 2015. 37. Schaefer C, Hannemann D, Meister R, et  al. Vitamin K antag­on ­ ists and
https://www.rcog.org.uk/globalassets/documents/guidelines/gtg-37a preg­nancy out­come: a multi-cen­tre pro­spec­tive study. Thromb Haemost.
.pdf. Accessed Nov 4 2021. 2006;95(6):949-957.
17. McLintock C, Brighton T, Chunilal S, et al; Councils of the Society of 38. Beyer-Westendorf J, Tittl L, Bistervels I, et al. Safety of direct oral anti­co­
Obstetric Medicine of Australia and New Zealand; Australasian Society ag­u­lant expo­sure dur­ing preg­nancy: a ret­ro­spec­tive cohort study. Lancet
of Thrombosis and Haemostasis. Recommendations for the pre­ ven­
tion Haematol. 2020;7(12):e884-e891.
of preg­nancy-asso­ci­ated venous throm­bo­em­bo­lism. Aust N Z J Obstet 39. Greer IA, Nelson-Piercy C. Low-molec­u­lar-weight hep­a­rins for thrombo-
Gynaecol. 2012;52(1):3-13. prophylaxis and treat­ment of venous throm­bo­em­bo­lism in preg­nancy: a
18. Chan W-S, Rey E, Kent N-E, et al; VTE in Pregnancy Guideline Working sys­tem­atic review of safety and effi­cacy. Blood. 2005;106(2):401-407.
Group; Society of Obstetricians and Gynecologists of Canada. Venous 40. Rodger MA, Kahn SR, Cranney A, et al; TIPPS inves­ti­ga­tors. Long-term
throm­bo­em­bo­lism and antithrombotic ther­apy in preg­nancy. J Obstet dalteparin in preg­nancy not asso­ci­ated with a decrease in bone min­eral
Gynaecol Can. 2014;36(6):527-553. den­sity: substudy of a ran­dom­ized con­trolled trial. J Thromb Haemost.
19. Palmerola KL, D’Alton ME, Brock CO, Friedman AM. A comparison of 2007;5(8):1600-1606.
­recommendations for pharmacologic thromboembolism prophylaxis after 41. Galambosi P, Hiilesmaa V, Ulander V-M, Laitinen L, Tiitinen A, Kaaja R.
caesarean delivery from three major guidelines. BJOG. 2016;123:2157-2162. Prolonged low-molec­u­lar-weight hep­a­rin use dur­ing preg­nancy and
20. Omunakwe HE, Roberts LN, Patel JP, et al. Re: A comparison of the recom- sub­se­quent bone min­eral den­sity. Thromb Res. 2016;143:122-126.
mendations for pharmacologic thromboembolism prophylaxis after cae- 42. Martillotti G, Boehlen F, Robert-Ebadi H, Jastrow N, Righini M, Blondon M.
sarean delivery from the major guidelines: Impact on thromboprophylaxis Treatment options for severe pul­mo­nary embolism dur­ing preg­nancy and
rates of implementing Royal College of Obstetricians and Gynaecologists’ the post­par­tum period: a sys­tem­atic review. J Thromb Haemost. 2017;15(10):
guidance for reducing the risk of ante- and postnatal venous thromboem- 1942-1950.
bolism. BJOG. 2017;124:831-832. 43. Gándara E, Carrier M, Rodger MA. Management of preg­nancy asso­ci­
21. O’Shaughnessy F, Donnelly JC, Bennett K, et al. Prevalence of postpartum ated venous-throm­bo­em­bo­lism: a sur­vey of prac­tices. Thromb J. 2014;
venous thromboembolish risk factors in an Irish urban obstetric popula- 12:12.
tion. J Thromb Haemost. 2019;17:1875-1885. 44. Lebaudy C, Hulot JS, Amoura Z, et al. Changes in enoxaparin phar­ma­co­ki­
22. Federspiel JJ, Wein LE, Addae-Konadu KL, et al. Venous throm­bo­em­bo­lism net­ics dur­ing preg­nancy and impli­ca­tions for antithrombotic ther­a­peu­tic
inci­dence among patients recommended for phar­ma­co­logic throm­bo­em­ strat­egy. Clin Pharmacol Ther. 2008;84(3):370-377.

568  |  Hematology 2021  |  ASH Education Program


45. Bhutia S, Wong PF. Once ver­sus twice daily low molec­u­lar weight hep­a­rin 60. Knol HM, Schultinge L, Veeger NJ, Kluin-Nelemans HC, Erwich JJ, Meijer
for the ini­tial treat­ment of venous throm­bo­em­bo­lism. Cochrane Database K. The risk of post­par­tum hem­or­rhage in women using high dose of low-
Syst Rev. 2013;(7):1-23. molec­u­lar-weight hep­a­rins dur­ing preg­nancy. Thromb Res. 2012;130(3):334-
46. Lepercq J, Conard J, Borel-Derlon A, et al. Venous throm­bo­em­bo­lism dur­ 338.
ing preg­nancy: a ret­ro­spec­tive study of enoxaparin safety in 624 preg­nan­ 61. Roshani S, Cohn DM, Stehouwer AC, et al. Incidence of post­par­tum haem-
cies. BJOG. 2001;108(11):1134-1140. orrhage in women receiv­ing ther­a­peu­tic doses of low-molec­u­lar-weight
47. Voke J, Keidan J, Pavord S, Spencer NH, Hunt BJ; Brit­ish Society for Hae- hep­ a­
rin: results of a ret­ ro­spec­tive cohort study. BMJ Open. 2011;1(2):​
matology Obstetric Haematology Group. The man­age­ment of ante­na­tal e000257.
venous throm­bo­em­bo­lism in the UK and Ireland: a pro­spec­tive multicentre 62. Santoro R, Iannaccaro P, Prejanò S, Muleo G. Efficacy and safety of the
obser­va­tional sur­vey. Br J Haematol. 2007;139(4):545-558. long-term admin­is­tra­tion of low-molec­u­lar-weight hep­a­rins in preg­nancy.
48. Rey E, Rivard GE. Prophylaxis and treat­ment of throm­bo­em­bolic dis­eases Blood Coagul Fibrinolysis. 2009;20(4):240-243.
dur­ing preg­nancy with dalteparin. Int J Gynaecol Obstet. 2000;71(1):19-24. 63. Tardy B, Chalayer E, Kamphuisen PW, et al; SSC Subcommittee on Con-
49. Barbour LA, Oja JL, Schultz LK. A pro­spec­tive trial that dem­on­strates that trol of Anticoagulation of the ISTH. Definition of bleed­ing events in stud­
dalteparin require­ments increase in preg­nancy to main­tain ther­a­peu­tic lev­ ies eval­u­at­ing pro­phy­lac­tic antithrombotic ther­apy in preg­nant women: a
els of anticoagulation. Am J Obstet Gynecol. 2004;191(3):1024-1029. sys­tem­atic review and a pro­posal from the ISTH SSC. J Thromb Haemost.
50. Jacobsen AF, Qvigstad E, Sandset PM. Low molec­ u­
lar weight hep­ a­
rin 2019;17(11):1979-1988.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/559/1851791/559skeith.pdf by guest on 13 December 2021


(dalteparin) for the treat­ment of venous throm­bo­em­bo­lism in preg­nancy. 64. Horlocker TT, Vandermeuelen E, Kopp SL, Gogarten W, Leffert LR, Benzon
BJOG. 2003;110(2):139-144. HT. Regional anes­the­sia in the patient receiv­ing antithrombotic or throm­
51. Rodie VA, Thomson AJ, Stewart FM, Quinn AJ, Walker ID, Greer IA. Low bo­lytic ther­apy: Amer­i­can Society of Regional Anesthesia and Pain Med-
molec­u­lar weight hep­a­rin for the treat­ment of venous throm­bo­em­bo­lism icine Evidence-Based Guidelines (fourth edi­tion). Reg Anesth Pain Med.
in preg­nancy: a case series. BJOG. 2002;109(9):1020-1024. 2018;43(3):263-309.
52. Smith MP, Norris LA, Steer PJ, Savidge GF, Bonnar J. Tinzaparin sodium 65. Leffert L, Butwick A, Carvalho B, et al; mem­bers of the SOAP VTE Taskforce.
for throm­bo­sis treat­ment and pre­ven­tion dur­ing preg­nancy. Am J Obstet The Society for Obstetric Anesthesia and Perinatology con­sen­sus state­
Gynecol. 2004;190(2):495-501. ment on the anes­thetic man­age­ment of preg­nant and post­par­tum women
53. Ní Ainle F, Wong A, Appleby N, et al. Efficacy and safety of once daily low receiv­ing thromboprophylaxis or higher dose anti­co­ag­u­lants. Anesth
molec­u­lar weight hep­a­rin (tinzaparin sodium) in high risk preg­nancy. Blood Analg. 2018;126(3):928-944.
Coagul Fibrinolysis. 2008;19(7):689-692. 66. Rottenstreich A, Zacks N, Kleinstern G, et  al. Planned induc­tion ver­sus
54. Gib­son PS, Newell K, Sam DX, et al. Weight-adjusted dos­ing of tinzaparin in spon­ta­ne­ous deliv­ery among women using pro­phy­lac­tic anticoagulation
preg­nancy. Thromb Res. 2013;131(2):e71-e75. ther­apy: a ret­ro­spec­tive study. BJOG. 2020;127(10):1241-1248.
55. Nelson-Piercy C, MacCallum P, Mackillop L, et al. Thromboembolic Disease 67. Harris SA, Velineni R, Davies AH. Inferior vena cava fil­ters in preg­nancy: a
in Pregnancy and the Puerperium: Acute Management. Green-Top Guide- sys­tem­atic review. J Vasc Interv Radiol. 2016;27(3):354-360.e8.
line No. 37b. Royal College of Obstetricians and Gynaecologists; 2015. 68. Cote-Poirier G, Bettache N, Cote A-M, et al. Evaluation of complications
56. Romualdi E, Dentali F, Rancan E, et  al. Anticoagulant ther­apy for venous in postpartum women receiving therapeutic anticoagulation. Obstet
throm­bo­em­bo­lism dur­ing preg­nancy: a sys­tem­atic review and a meta- ­Gynecol. 2020;00:1-8.
anal­y­sis of the lit­er­a­ture. J Thromb Haemost. 2013;11(2):270-281. 69. Zhao Y, Arya R, Couchman L, Patel JP. Are apixaban and rivaroxaban dis­trib­
57. Sirico A, Saccone G, Maruotti GM, et al. Low molec­u­lar weight hep­a­rin use uted into human breast milk to clin­i­cally rel­e­vant con­cen­tra­tions? Blood.
dur­ing preg­nancy and risk of post­par­tum hem­or­rhage: a sys­tem­atic review 2020;136(15):1783-1785.
and meta-anal­y­sis. J Matern Fetal Neonatal Med. 2019;32(11):1893-1900. 70. Daei M, Khalili H, Heidari Z. Direct oral anti­co­ag­u­lant safety dur­ing breast-
58. Arbuthnot C, Browne R, Nicole S, Erb SJ, Farrall L, Borg A. A dou­ble cen­tre feeding: a nar­ra­tive review. Eur J Clin Pharmacol. 2021;77(10):1465-1471.
ret­ro­spec­tive study into rates of post­par­tum haemorrhage in women on
low molec­u­lar weight hep­a­rin. Br J Haematol. 2017;176(1):141-143.
59. Galambosi PJ, Kaaja RJ, Stefanovic V, Ulander V-M. Safety of low-molec­u­lar-
weight hep­a­rin dur­ing preg­nancy: a ret­ro­spec­tive con­trolled cohort study. © 2021 by The Amer­i­can Society of Hematology
Eur J Obstet Gynecol Reprod Biol. 2012;163(2):154-159. DOI 10.1182/hema­tol­ogy.2021000291

Prakash Singh Shekhawat


VTE pre­ven­tion and man­age­ment in preg­nancy  |  569
SURVIVORSHIP AFTER ALLOGENEIC HEMATOPOIETIC CELL TRANSPLANT

Psychosocial and financial issues after


hematopoietic cell transplantation

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/570/1851740/570buchbinder.pdf by guest on 13 December 2021


David Buchbinder1 and Nandita Khera2
CHOC Hospital, Orange, CA; and 2College of Medicine, Mayo Clinic in Arizona, Phoenix, AZ
1

With improvement in survival after hematopoietic cell transplantation (HCT), it has become important to focus on sur-
vivors’ psychosocial issues in order to provide patient-centered care across the transplant continuum. The goals of
this article are to describe updates in the literature on certain psychosocial domains (emotional/mental health and
social/financial) in HCT survivors, offer a brief overview of the status of the screening and management of these compli-
cations, and identify opportunities for future practice and research. An evidence-based approach to psychosocial care
can be broken down as primary (promoting health, raising awareness, and addressing risk factors), secondary (screening
and directing early pharmacological and nonpharmacological interventions), and tertiary (rehabilitating, limiting disabil-
ity, and improving quality of life) prevention. Implementing such an approach requires close coordination between mul-
tiple stakeholders, including transplant center staff, referring hematologist/oncologists, and other subspecialists in areas
such as palliative medicine or psychiatry. Innovative models of care that leverage technology can bring these stakehold-
ers together to fulfill unmet needs in this area by addressing barriers in the delivery of psychosocial care.

LEARNING OBJECTIVES
• Understand the burden of psychosocial complications after hematopoietic cell transplantation
• Describe the approach to screening and managing these complications
• Recognize barriers in the delivery of psychosocial care

syndrome at 10 months of age. His parents emigrated from


CLINICAL CASES Mexico without extended family just before his transplant.
J. B. is a 47-year-old non-Hispanic White female who is 6 years Medicaid coverage was adequate, with minimal out-of-
post C antigen mismatched unrelated-donor peripheral pocket expenses or deductibles, but the stress of having
blood hematopoietic cell transplant (HCT) for acute mye- barely enough money for food and necessities was high,
loid leukemia. She had chronic graft-versus-host disease especially during the early posttransplant period. Soon
(GVHD) of the skin, fascia, and joints and is currently on after the HCT, the father ran out of paid time off and was
treatment with prednisone, sirolimus, and extracorporeal forced to take unpaid time off work. The parents shared
photopheresis. She also had multiple other complications, duties during and after transplant, often neglecting their
including a chronic nonhealing ulcer of the left shin, left- own health care needs. A. R. did well from a medical stand-
lower-extremity deep vein thrombosis, chronic renal insuf- point but has lasting cognitive and academic deficits and
ficiency, and a fracture of the left femur due to osteopo- behavioral problems that make it difficult for him to man-
rosis, which then ended up as a mycobacterial infection age school. This has led to a great deal of stress for his par-
of the hip joint after surgery. She got divorced 1 year after ents, especially since they have little social support.
hematopoietic cell transplantation (HCT) and lost her job
3 years after the HCT due to missing work frequently. She
is extremely depressed due to the ongoing medical and Introduction
financial issues and was recently hospitalized for express- J. B. and A. R. illustrate some of the issues faced by 100 000
ing suicidal intent. HCT survivors in the United States, a population projected
A. R. is a 14-year-old Hispanic male who received a matched to increase 5-fold by 2030.1 The cost of cure is the physi-
unrelated-donor bone marrow transplant for Wiskott-Aldrich cal, psychological, and social sequelae of HCT that have a

570 | Hematology 2021 | ASH Education Program


Table 1. Physical, psy­cho­log­i­cal, and social sequelae of HCT psychological test­ing; how­ever, esti­ma­tes are higher when self-
reported (40%–60%).4 A pro­por­tion of HCT sur­vi­vors may expe­
Physical sequelae Psychological sequelae Social sequelae ri­ence per­sis­tent neurocognitive dys­func­tion, while other
Poor phys­i­cal and Depression/anx­i­ety Financial dis­tress sur­vi­vors may expe­ri­ence improve­ments over time. Age, sex,
men­tal health edu­ca­tion, income, cog­ni­tive reserve, type of HCT, and con­di­
tion­ing inten­sity are pre­dic­tive fac­tors for post-HCT cog­ni­tive
Fatigue, sleep, pain Perceived stress Social rein­te­gra­tion
impair­ment. Specific domains (ie, motor func­tion­ing) appear to
Sexual dys­func­tion Adverse cop­ing Return to work be dis­pro­por­tion­ately impaired to a greater extent, whereas
Symptom bur­den Cognitive dys­func­tion Social sup­port/ oth­ers may be spared.10
mar­i­tal prob­lems
Social and finan­cial prob­lems after HCT
Medical com­pli­ca­tions after HCT along with the phys­i­cal and
det­ri­men­tal impact on the qual­ity of life of these sur­vi­vors after psy­cho­log­i­cal prob­lems described above can adversely affect

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/570/1851740/570buchbinder.pdf by guest on 13 December 2021


HCT (Table 1). HCT sur­vi­vors and their care­giv­ers have iden­ti­fied the social well-being as well as the employ­ment and finan­cial
these con­cerns as “what mat­ters to them,” and addressing them sta­tus of sur­vi­vors. In turn, finan­cial hard­ship due to reduced
is an impor­tant com­po­nent for deliv­ery of high-qual­ity care.2 Un- income, loss of employ­ment, and the cost of care for ongo­ing
met needs in emo­tional health and finan­cial bur­den in HCT sur­vi­ active med­i­cal issues can exac­er­bate the stress and worsen the
vors have been recently described.3 psy­cho­log­i­cal prob­lems. Financial tox­ic­ity has emerged as an
The goal of this arti­cle is to describe the updates in the lit­er­ impor­tant com­pli­ca­tion of HCT, as mul­ti­ple stud­ies have dem­
a­ture for the last 5 years for psy­cho­so­cial domains (emo­tional/ on­strated patients’ poor finan­cial well-being after HCT. A lack
men­ tal health and social/finan­ cial) across the HCT tra­jec­ tory of well-val­ i­
dated instru­ ments to mea­ sure social and finan­ cial
describ­ing the bur­den of the prob­lem, with a spe­cial focus on out­comes, the use of mostly small insti­tu­tional stud­ies, and the
the pedi­at­ric/ado­les­cent and young adult (AYA) group. Many absence of lon­gi­tu­di­nal data are some of the gaps in research
sem­ i­
nal stud­ ies conducted ear­ lier are not included. We also for this com­pli­ca­tion of HCT. The prev­a­lence of finan­cial hard­
do not include phys­i­cal and sex­ual func­tion­ing con­cerns and ship ranges from 20% to 70% of HCT sur­vi­vors.4,11-15 In addi­tion
psy­cho­so­cial out­comes for HCT care­giv­ers, though they may to the sociodemographic fac­tors, chronic GVHD in allo­ge­neic
be highly related to the domains of inter­est. We con­clude by HCT sur­vi­vors is a major fac­tor asso­ci­ated with finan­cial stress,
reviewing the cur­rent sta­tus of screen­ing and man­age­ment of as two-thirds of chronic GVHD patients endure finan­cial hard­
these com­pli­ca­tions and oppor­tu­ni­ties for improve­ment, includ­ ship despite health insur­ance, and these are likely to be patients
ing pos­si­ble incor­po­ra­tion of recently tested psy­cho­so­cial inter­ with lower house­hold income.16 Denials and delays in care due to
ven­tions in prac­tice, and high­light the bar­ri­ers and mod­els of insur­ance and loss of cov­er­age are com­mon stress­ors faced by
care to address these com­pli­ca­tions. HCT sur­vi­vors. Financial hard­ship in HCT sur­vi­vors is known to be
asso­ci­ated with poor qual­ity of life, higher per­ceived stress, non-
adherence to treat­ment, higher symp­tom bur­den, and low sat­
Emotional and men­tal health and cog­ni­tive prob­lems is­fac­tion with care but, for­tu­nately, no impact on sur­vival.11,12,16,17
after HCT The return to work (RTW) is an indi­ca­tor of func­tional recov­
Psychological sequelae fol­low­ing allo­ge­neic HCT, the impor­tance ery for trans­plant sur­vi­vors, and the inabil­ity to RTW may be
of which can­ not be underestimated, have been documented asso­ci­ated with finan­cial bur­den. Only about half of the patients
(depres­sion, anx­i­ety, per­ceived stress, adverse cop­ing, and neu- who pre­vi­ously con­trib­uted to house­hold earn­ings may be ­able
rocognitive dys­func­tion). Potential risk fac­tors for psy­cho­log­i­cal to RTW by 2 years after HCT.18 Chronic GVHD is a major fac­tor
sequelae fol­low­ing HCT are numer­ous; how­ever, female gen­der, influ­enc­ing RTW/sick leave, with one-quar­ter of patients with
youn­ger age, not liv­ing with a part­ner, and chronic GVHD appear chronic GVHD reporting being “dis­abled/unable to work” at a
to be salient pre­dic­tors of impair­ment. Among allo­ge­neic HCT median of 2 years post HCT.16,19 Lower phys­i­cal func­tion­ing, mul-
sur­vi­vors, the most fre­quently reported unmet needs are focused timorbidity, being female, and the receipt of periph­eral blood
on psy­cho­log­i­cal sequelae.4 Despite this, few stud­ies of psy­cho­ stem cells for an unre­lated HCT are asso­ci­ated with a lower like­li­
log­i­cal sequelae have been conducted in long-term HCT sur­vi­ hood of RTW post HCT.20-22 A lack of stan­dard guide­lines for RTW
vors in a con­tem­po­rary cohort. These stud­ies are often sub­ject to lead­ing to poor com­mu­ni­ca­tion between pro­vid­ers, patients,
a vari­ety of meth­od­o­log­i­cal lim­i­ta­tions (small or often het­ero­ge­ and employers can be a bar­rier to suc­cess­ful rein­te­gra­tion of
neous sam­ples, a lack of lon­gi­tu­di­nal data with short fol­low-up, patients in the work­place after HCT.23
and the use of var­i­able mea­sures). While a major­ ity of HCT sur­ vi­
vors show rea­ son­able lev­
Depression and anx­i­ety have been reported in 12% to 30% els of social adjust­ment, those with fatigue may be at risk for
of HCT sur­vi­vors.4,5 Even at very dis­tal time points as long as maladjustment and may need effec­tive reha­bil­i­ta­tion strat­e­
>20 years fol­low­ing HCT, depres­sion and anx­i­ety remain a fre­ gies.24 Social rein­te­gra­tion and social sup­port, includ­ing emo­
quent prob­lem.6 While 22% to 43% of HCT sur­vi­vors suf­fer from tional, infor­ma­tional, and logis­ti­cal sup­port, can help mit­i­gate
emo­tional dis­tress, post­trau­matic stress dis­or­der is rel­a­tively the effects of psy­cho­so­cial prob­lems, as illus­trated by stud­ies
infre­quent (3%–13%).7 Importantly, adap­tive or maladaptive cop­ in can­cer patients. One of the pos­i­tive sequelae of over­com­ing
ing strat­e­gies play a cru­cial role in the occur­rence and impact of trau­matic stress­ors caused by life-threat­en­ing dis­ease and inten­
psy­cho­log­i­cal sequelae.8,9 sive treat­ment reported in HCT sur­vi­vors is the phe­nom­e­non of
Estimates sug­gest that 10% to 40% of HCT sur­vi­vors expe­ “post­trau­matic growth.” The roles of emo­tional and instru­men­tal
ri­ence neurocognitive dys­func­tion when mea­sured with neuro- social sup­port, social con­straints, and sup­port from health care
Prakash Singh Shekhawat
Psychosocial com­pli­ca­tions of HCT  |  571
pro­fes­sion­als in pro­mot­ing post­trau­matic growth have been elu­ level, etc), trans­plant-related (con­di­tion­ing reg­i­mens with total
ci­dated fur­ther in the last few years.14,25 body irradiation), and posttransplant fac­ tors such as chronic
health con­di­tions and chronic GVHD. AYA sur­vi­vors of HCT have
a myr­iad of unmet psy­cho­so­cial needs that dif­fer from other HCT
Specific con­sid­er­ations for pedi­at­ric/AYA pop­u­la­tions sur­vi­vors, indi­cat­ing a neces­sity to develop inter­ven­tions spe­cif­i­
Pediatric patients are vul­ner­a­ble to psy­cho­so­cial sequelae after cally tai­lored to them.35
HCT. Among pedi­at­ric HCT sur­vi­vors, esti­ma­tes of phys­i­cal, psy­
cho­log­i­cal, and social dys­func­tion vary due to het­ero­ge­ne­ity in Screening for and inter­ven­tions to help improve
meth­od­o­log­i­cal aspects. Longitudinal eval­u­a­tion of pedi­at­ric psy­cho­so­cial health in HCT sur­vi­vors
HCT sur­vi­vors is vital because the abil­ity to assess spe­cific con­ Most trans­ plant cen­ ters per­
form some form of psy­ cho­so­cial
structs in chil­dren changes over time; how­ever, pro­spec­tive lon­ eval­u­a­tion, includ­ing finan­cial assess­ment of HCT can­di­dates
gi­tu­di­nal data in pedi­at­ric HCT sur­vi­vor­ship care are infre­quent. prior to HCT. Table 2 pro­vi­des exam­ples of some of the screen­
Symptom bur­den is greater and health-related qual­ity of life is ing instru­ments used in clin­i­cal prac­tice, though these may not

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/570/1851740/570buchbinder.pdf by guest on 13 December 2021


poorer when com­pared to healthy peers, though com­pa­ra­ble nec­es­sar­ily be lim­ited to the HCT pop­ul­a­tion. However, there
with con­ven­tion­ally treated can­cer sur­vi­vors.26 Health prob­lems is a lot of var­i­abil­ity in terms of fre­quency and types of these
may pre­vent 16% of sur­vi­vors from attend­ing school; how­ever, the assess­ments before and after HCT at var­io ­ us trans­plant cen­ters.
need for spe­cial edu­ca­tion ser­vices at school was sim­i­lar between Also, the approach when addressing these issues is usu­ally reac­
pedi­at­ric HCT sur­vi­vors and con­ven­tion­ally treated sur­vi­vors.27 tive rather than pro­ac­tive. Increasing aware­ness is vital to iden­
Depression as well as anx­ie ­ ty is often noted among pedi­at­ric HCT tify sur­vi­vors and their fam­ily mem­bers who may ben­e­fit from
sur­vi­vors as com­pared to healthy peers. A recent study found addi­tional in-depth assess­ment and refer­ral for resources for
that 16% of pedi­at­ric HCT sur­vi­vors endorsed depres­sion, where- these issues. Guidelines for post-HCT care rec­om­mend peri­odic
as 10% of sur­vi­vors endorsed anx­i­ety.28 Neurocognitive dys­func­ screen­ing for psy­cho­so­cial dif­fi­cul­ties, espe­cially depres­sion,
tion is prev­a­lent in about 50% of pedi­at­ric HCT sur­vi­vors.28 The after trans­plant, at 6 and 12 months, and annu­ally there­af­ter.36
domain of motor devel­op­ment was affected to the greatest ex- They also rec­om­mend men­tal health pro­fes­sional coun­sel­ing
tent; how­ever, lan­guage, social, and emo­tional devel­op­ment was for those with rec­og­nized def­i­cits and reg­ul­ar assess­ments of
also impaired com­pared to healthy peers. spou­sal/care­giver psy­cho­log­i­cal adjust­ment and fam­ily func­
AYA sur­vi­vors of HCT are a unique pop­u­la­tion that require tion­ing while encour­ag­ing robust sup­port net­works. Universal
spe­cial care con­sid­er­ations. HCT and its sequelae may under­ screen­ing for finan­cial dis­tress should be added to the usual
mine the abil­ity of AYA sur­vi­vors to achieve mile­stones such as assess­ments since it is prev­a­lent and affects patient out­comes.37
grad­u­at­ing from col­lege, selecting a career, and establishing Developing a set of stan­dard patient-reported out­come mea­
employ­ment as well as achiev­ing socio­eco­nomic inde­pen­dence sures to be col­lected at peri­odic time points and to address the
from par­ents, serv­ing as a risk fac­tor for adverse psy­cho­so­cial domains most rel­e­vant to sur­vi­vors is cru­cial. Such mea­sures
out­comes.29,30 Despite the impor­tance of devel­op­ing a greater can help under­stand the pop­u­la­tion-based prev­a­lence of these
under­stand­ing of the AYA expe­ri­ence within the con­text of HCT psy­cho­so­cial issues and screen for prob­lems that would need
sur­vi­vor­ship, few stud­ies have focused on psy­cho­so­cial sequelae inter­ven­tion in real time. Electronic med­i­cal records can be lev­
among AYA sur­vi­vors of HCT. When we com­pare AYA sur­vi­vors er­aged to rou­tinely col­lect a stan­dard­ized set of social, psy­cho­
to healthy peers, dec­re­ments in phys­i­cal and social func­tion­ing log­i­cal, and behav­ioral deter­mi­nants of health to inte­grate this
and health become appar­ent.31,32 AYA sur­vi­vors of HCT report pro­cess with rou­tine clin­i­cal care. Screening rec­om­men­da­tions
feel­ing left behind their peers, which some­times leads them to and pos­si­ble man­age­ment options may also be incor­po­rated
rush back to work and school and take on too many respon­si­ into sur­vi­vor­ship care sum­ma­ries to facil­i­tate the deliv­ery of psy­
bil­i­ties too quickly, resulting in over­whelm­ing demands.33 RTW cho­so­cial care across the tra­jec­tory of HCT care.38 It is inter­est­
and employ­ment issues con­tinue to be major psy­cho­so­cial chal­ ing to note that these indi­vid­ua ­ l­ized care plans as edu­ca­tional
lenges for AYA sur­vi­vors of HCT.34 tools can lead to reduced dis­tress and improve­ment in the men­
Risk fac­tors for adverse psy­cho­so­cial out­comes among AYA tal domain of health-related qual­ity of life, as shown by a recent
sur­ vi­vors of HCT include sociodemographic (age, edu­ ca­
tion ran­dom­ized clin­i­cal trial.39

Table 2. Examples of psy­cho­so­cial and finan­cial screen­ing tools

Scale Description
Psychosocial Assessment of Candidates for Transplant41 Describes psy­cho­so­cial func­tion­ing before organ and HCT
Transplant Evaluation Rating Scale42
Describes psy­cho­so­cial func­tion­ing before organ and HCT
Psychosocial Assessment Tool—Hematopoietic Cell Transplantation43 Describes the fam­ily psy­cho­so­cial risk for fam­i­lies of a child under­go­ing HCT
Stanford Integrated Psychosocial Assessment for Transplantation 44
Measures psy­cho­so­cial read­i­ness for trans­plant
Comprehensive Score for Financial Toxicity45 Describes the finan­cial dis­tress expe­ri­enced by can­cer patients 18 years of
age and older dur­ing the past 7 days
InCharge Financial Distress/Financial Well-Being Scale46 Measures responses to one’s finan­cial state on a con­tin­uum rang­ing from
over­whelm­ing finan­cial dis­tress to no finan­cial dis­tress

572  |  Hematology 2021  |  ASH Education Program


Table 3. Psychosocial inter­ven­tions for HCT sur­vi­vors

Author Sample Study design Intervention Measures Findings


Depression/anx­i­ety
  Copeland n = 20, allo­ge­neic HCT, >18 years Single-arm pro­spec­tive Weekly col­lab­o­ra­tive care Hospital Anxiety and Anxiety decreased and depres­sion
et al47 of age pre-HCT, day 0, +14, meet­ings with HCT cli­ni­cians Depression Scale decreased (pre- to day +60).
+30, +60 and psy­chi­a­trist, review
case and pro­vide phar­ma­co­
logic rec­om­men­da­tions and
psy­cho­so­cial coun­sel­ing refer­rals
  Kim et al48 n = 40, mean age 59.2 years Randomized (inter­ven­tion Digital sto­ry­tell­ing—four 3-min­ute POMS sub­scales for depres­ Perceived social sup­port increased
or infor­ma­tion con­trol) per­sonal/emo­tion­ally rich stories sion and anx­i­ety, PROMIS for inter­ven­tion and decreased for
pre- and post assess­ment about post-HCT care Social Support scale con­trol. Anxiety and depres­sion
improved with time for both.
Coping/stress
  Balck et al49 n = 91, allo­ge­neic and autol­o­gous, Randomized (inter­ven­tion Problem-solv­ing train­ing HADS, SCL-9, Brief Cope, Anxiety, dis­tress, pain, stress were
>18 years of age or con­trol group) pre- Social Problem-Solving lower and active cop­ing bet­ter
HCT, day +11, +23 Inventory-R, Questions with inter­ven­tion. Better ­able to
of Pain, NCCN Distress reduce neg­a­tive ori­en­ta­tion and
Thermometer prob­lem-solve with inter­ven­tion.
  Somers n = 36, allo­ge­neic and autol­o­gous Randomized (inter­ven­tion Mobile pain-cop­ing skills: website Brief pain inven­tory, pain dis­ Intervention with improved pain, self-
et al50 (83%), mean age 56 years or usual treat­ment) with per­son­al­ized mes­sages abil­ity index, pain sub­scale effi­cacy, and on the 2MWT, and
pre- and post assess­ment and pain assess­ment activ­ity. of the chronic pain self-effi­ no improvement in the control
Materials (hand­outs, vid­eos, cacy scale, PROMIS Fatigue group. Changes in pain ­dis­abil­ity
audio files) about pain-cop­ing scale, FACT well-being and fatigue in both groups;
advice from patients and how to scale, 2MWT effect sizes were larger for the
apply pain-cop­ing skills inter­ven­tion group. No changes

Prakash Singh Shekhawat


in pain sever­ity in either group.
  Majhail n = 458, allo­ge­neic (48%) inter­ven­tion Randomized (inter­ven­tion Survivorship care plan CSI, CTXD, Knowledge of Intervention—lower dis­tress scores
et al39 arm, (44%) con­trol arm, median or con­trol) pre- and post Transplant Exposures, at 6 months and greater men­tal
age 59 years for both arms assess­ment (6 months) Health Care Utilization, SF-12 health scores
  Syrjala et al40 n = 755, allo­ge­neic and autol­o­gous Randomized (inter­ven­tion INSPIRE—tai­lored web page with CTXD, SCL-90-R Depression INSPIRE+ prob­lem-solv­ing train­ing
Syrjala et al51 (27%), mean age 51 years or inter­ven­tion + prob­lem- top­ics (1) lift mood, reduce fatigue, Scale, SF-36, FSI more likely to improve in dis­tress
Yi et al52 solv­ing train­ing or con­ boost health; (2) self-care tips and than con­trols. Male, age <40
trol—delayed access to tools; (3) tai­lored care guide­lines; years, Afri­can Amer­i­cans were
inter­ven­tion) pre- and (4) forum for post­ing expe­ri­ences less likely to enroll. Engagement
post assess­ment and for input; (5) resource list did not dif­fer by race, edu­ca­tion,
(6 months) prob­lem-solv­ing—focus on income, rural/urban res­i­dence,
prob­lems and goal set­ting with com­puter expe­ri­ence, donor
psy­chol­o­gists (>4 ses­sions) type, or pres­ence of depres­sion.
Cognitive/devel­op­men­tal
  Ferraro n = 110 Feasibility study of screen­ing MoCA, BACH Neurocognitive screen­ing is
et al53 at pre-HCT, day +100, 2 fea­si­ble to do prior to and post
years, 5 years post HCT trans­plant.
  Hoodin n = 9, allo­ge­neic HCT, mean age Ancillary study and Vorinostat + Tac + MTX for GVHD Cogstate, PHQ-9, GAD-7, Neurocognitive func­tion, depres­
et al54 53 years, con­trol cohorts— fea­si­bil­ity of assessing pro­phy­laxis FACT-General sion, anx­i­ety, quality of life did
con­cur­rent lon­gi­tu­di­nal study vorinostat, cog­ni­tive not dif­fer across time. Modeling—
func­tion, and QOL neurocognitive func­tion in
base­line, day 30, 100, 160 vorinostat patients com­pared to
auto con­trols was equiv­a­lent and

Psychosocial com­pli­ca­tions of HCT  |  573


bet­ter than allo con­trols.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/570/1851740/570buchbinder.pdf by guest on 13 December 2021


Transplant center/ Paent-level Health care system
health care provider • Lack of informaon, • Lack of/inadequate
level knowledge, and skills reimbursement
• Socioeconomic and • Lack of resources and
• Lack of resources

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/570/1851740/570buchbinder.pdf by guest on 13 December 2021


cultural barriers specialists
• Lack of interest and
• Heterogeneity of late • Poor coordinaon of care
compeng priories
complicaons • Compeng priories
• Lack of awareness of
• Logiscal challenges • Inadequate quality
needs and screening and
prevenon guidelines • Different psychosocial assurance and
needs at different mes in accountability
• Lack of tools to address
transplant connuum
psychosocial concerns
• Inadequate
communicaon

Figure 1. Barriers to psychosocial care delivery.

There is a need for inter­ven­tions spe­cific to HCT sur­vi­vors that per­son­al­ized fash­ion and trans­lat­ing the research in psy­cho­so­cial
are evi­dence based and lever­age novel plat­forms to improve inter­ven­tions to actual clin­i­cal prac­tice remains a chal­lenge.
acces­si­bil­ity, with the goal of miti­gat­ing risks and improv­ing out­ To over­come this chal­lenge, we need to develop and imple­
comes fol­low­ing HCT. Table 3 sum­ma­rizes some of the inter­ven­ ment effec­tive mod­els for deliv­er­ing psy­cho­so­cial care. These
tions that have attempted to address the psy­cho­so­cial domains mod­els should be based on the 3-step approach to pre­ven­
in HCT sur­ vi­vors. These inter­ ven­tions were largely aimed at tion of these com­pli­ca­tions. The first step or pri­mary pre­ven­
depres­sion, anx­ie ­ ty, stress, and cop­ing, with only one inter­ven­ tion would include health pro­mo­tion activ­i­ties, includ­ing using
tion focused on cog­ni­tive dys­func­tion. To our knowl­edge, in the trans­ plant as a “teach­ able moment” for pro­ mot­ing health
last 5 years no inter­ven­tions have been published focus­ing on behav­ior change, increas­ing aware­ness, and pre­emp­tively try­
finan­cial hard­ship or social health among HCT sur­vi­vors spe­cif­i­ ing to address risk fac­tors for these com­pli­ca­tions. Secondary
cally, suggesting a concerning gap in the care of HCT sur­vi­vors pre­ven­tion would include screen­ing that, as reviewed above,
and their fam­i­lies. Most inter­ven­tion stud­ies have been by sin­gle should be com­pre­hen­sive, performed mul­ti­ple times post HCT,
insti­tu­tions employing small sam­ple sizes to assess accept­abil­ity and lead to action­able care path­ways pos­si­bly incor­po­rat­ing
as well as fea­si­bil­ity. Most stud­ies were not appro­pri­ately pow- the tested inter­ven­tions in rou­tine care. Tertiary pre­ven­tion tar-
ered to assess effi­cacy and require future eval­u­a­tion within the geted at decreas­ing mor­bid­ity and mor­tal­ity from these com­
con­text of larger ran­dom­ized con­trolled tri­als. pli­ca­tions would include reha­bil­i­ta­tion strat­e­gies and dis­abil­ity
lim­i­ta­tion.
Barriers to psy­cho­so­cial care deliv­ery and mod­els Implementing the above approach requires inte­gra­tion of
to address bar­ri­ers all­the rel­e­vant stake­hold­ers, such as the trans­plant team, pri­
Many bar­ri­ers at dif­fer­ent lev­els need to be over­come to rec­ mary care or oncol­ogy phy­si­cian, psy­chol­o­gist or psy­chi­a­trist,
og­nize and ade­quately treat the psy­cho­so­cial con­cerns of HCT social work­ ers, and finan­ cial coun­ sel­
ors (Table 4). With the
sur­vi­vors in rou­tine clin­i­cal prac­tice (Figure 1). Even if we iden­ increased focus on vir­tual med­i­cine because of the COVID-19
tify HCT sur­vi­vors in need of psy­cho­so­cial sup­port, pro­vid­ing in- pan­demic, there is an oppor­tu­nity to lever­age vir­tual tech­nol­
depth assess­ments with con­nec­tiv­ity to avail­­able resources in a o­gies to deliver well-rounded, risk-strat­i­fied sur­vi­vor­ship care

574  |  Hematology 2021  |  ASH Education Program


Table 4. HCT psy­cho­so­cial pro­vid­ers and roles

Category Role
Transplant phy­si­cian Provide edu­ca­tion to patients and fam­i­lies regard­ing signs of psy­cho­so­cial dis­tress asso­ci­ated with HCT
Recognize signs of psy­cho­so­cial dis­tress and assess for psy­cho­so­cial dis­tress asso­ci­ated with HCT
Transplant nurse Provide edu­ca­tion to patients and fam­i­lies regard­ing signs of psy­cho­so­cial dis­tress asso­ci­ated with HCT
Recognize signs of psy­cho­so­cial dis­tress and assess for psy­cho­so­cial dis­tress asso­ci­ated with HCT
Social worker/case man­ager Educate, rec­og­nize, and assess for psy­cho­so­cial dis­tress.
Facilitate patient and fam­ily adjust­ment to the pro­cess of HCT and its sequelae.
Refer patients and fam­i­lies with dis­tress for spe­cial­ized ser­vices and com­mu­nity resources.
Address a range of psy­cho­so­cial and finan­cial needs (eg, insur­ance ben­e­fits, coor­di­na­tion of care, nav­i­gat­ing
health sys­tem, peer sup­port).
Psychologist Provide con­sul­ta­tion and man­age­ment of psy­cho­so­cial con­cerns iden­ti­fied among HCT patients and fam­i­lies

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/570/1851740/570buchbinder.pdf by guest on 13 December 2021


using var­i­ous psy­cho­ther­a­peu­tic tech­niques.
Assist in the con­trol of symp­toms (eg, anx­i­ety, depres­sion) that may impact patient and fam­ily well-being.
Psychiatrist Diagnose and treat psy­cho­logic and psy­chi­at­ric dis­or­ders that arise dur­ing HCT using psy­cho­phar­ma­co­logic
inter­ven­tions.
Assist in the con­trol of symp­toms (eg, anx­i­ety, depres­sion) that may impact patient and fam­ily well-being.
Chaplain, clergy, pas­to­ral care Assist patients and fam­i­lies with cop­ing, spir­i­tual coun­sel­ing.
Patient finan­cial ser­vices Assist patient and fam­i­lies with prior autho­ri­za­tion, pay­ment plans to put together an appeal for deni­als to help
with costs of care.
Primary care phy­si­cian/pri­mary Work closely with trans­plant team to screen and address psy­cho­so­cial issues and pro­vide sup­port.
hema­tol­o­gist/oncol­o­gist

incor­po­rat­ing psy­cho­so­cial screen­ing and inter­ven­tions. This


can help encour­ age health-related self-effi­ cacy in patients, CLINICAL CASES (Con­tin­ued)
increase health sys­tem capac­ity, and pro­mote adap­tive HCT In addi­tion to the man­age­ment of J. B.’s med­i­cal com­pli­ca­tions,
sur­vi­vor­ship. Online pro­grams may help address some of the a mul­ti­dis­ci­plin­ary team that included her trans­plant phy­si­
bar­ri­ers in pro­vid­ing psy­cho­so­cial care such as dis­tance, vary­ cian, nurse, and social worker; patient finan­cial ser­vices; and
ing needs, and lack of stan­dard fol­low-up care. Recently, the pro­vid­ers from pal­li­a­tive med­i­cine and psy­chi­a­try/psy­chol­ogy
INSPIRE (Inter­net-Based Survivorship Program with Information came together to help her man­age her var­i­ous issues and pro­
and Resources) study found no dif­fer­ences between the study vide coor­di­nated care. Frequent touch points with her using
arms in the pri­mary end point of aggre­gated out­comes of can­ vir­tual vis­its made her feel that her team was engaged in her
cer and treat­ment dis­tress, depres­sive symp­toms, phys­i­cal over­all care. Participating in a sup­port group for chronic GVHD
dys­func­tion, and fatigue, though there was sig­nif­i­cant improve­ patients helped her feel emo­tion­ally supported.For the finan­
ment in dis­tress alone for those in the INSPIRE+ prob­lem-solv­ing cial hard­ship, A. R.’s fam­ily was referred to a local char­i­ta­ble
treat­ment arm.40 orga­ni­za­tion that pro­vided grant funds for rent assis­tance. Due

Table 5. Recommended areas of focus for future research pri­or­i­ties and clin­i­cal prac­tice

Area Focus
Study design and mea­sures Proactively col­lect psy­cho­so­cial and finan­cial health data among HCT patients and fam­i­lies across the entire
tra­jec­tory of trans­plant care (lon­gi­tu­di­nal) using large, rep­re­sen­ta­tive sam­ples.
Increase focus on spe­cial pop­u­la­tions—chil­dren, ado­les­cents, and young adults, older adults, racial/eth­nic minor­ity
patients.
Use well-val­i­dated and stan­dard patient-reported out­come mea­sures.
Screening and assess­ment Develop evi­dence-based rec­om­men­da­tions for uni­ver­sal screen­ing and assess­ment of HCT patients and fam­i­lies
at defined time points across the entire trans­plant tra­jec­tory using a stan­dard set of patient-reported out­come
mea­sures focused on psy­cho­so­cial and finan­cial dis­tress.
Leverage tech­nol­ogy to ensure robust con­nec­tiv­ity to HCT cen­ter and com­mu­nity-based resources.
Rehabilitation and inter­ven­tion Develop and test effec­tive reha­bil­i­ta­tion strat­e­gies, social sup­port (emo­tional, infor­ma­tional, and logis­ti­cal)
inter­ven­tions, and finan­cial sup­port inter­ven­tions.
Employ novel plat­forms for deliv­ery and devel­op­men­tally tai­lored for spe­cial pop­u­la­tions.
Education and aware­ness Raise aware­ness regard­ing psy­cho­so­cial and finan­cial health through var­i­ous means includ­ing edu­ca­tional efforts
directed at all­lev­els includ­ing, but not lim­ited to, patients and fam­i­lies as well as trans­plant care pro­vid­ers and
pol­icy-mak­ers.
Develop met­rics to exam­ine the effec­tive­ness of these mea­sures.
Prakash Singh Shekhawat
Psychosocial com­pli­ca­tions of HCT  |  575
to his cog­ni­tive and aca­demic chal­lenges, A. R. was referred for plant recip­i­ents and their infor­mal care­giv­ers. Biol Blood Marrow Trans-
for­mal neuropsychological eval­u­a­tion. Based upon the detailed plant. 2019;25(1):145-150.
8. Jacobs JM, Fishman S, Sommer R, et  al. Coping and mod­i­fi­able psy­cho­
eval­ua
­ ­tion, the HCT clin­ic
­ al team and A. R.’s par­ents worked so­cial fac­tors are asso­ci­ated with mood and qual­ ity of life in patients
closely with the child’s school, includ­ing his teach­ers and the with chronic graft-ver­ sus-host dis­ease. Biol Blood Marrow Transplant.
admin­is­tra­tion, to develop an appro­pri­ate, indi­vid­u­al­ized edu­ 2019;25(11):2234-2242.
ca­tion plan for A. R. With addi­tional sup­port in the class­room, 9. Kusaka K, Inoguchi H, Nakahara R, et  al. Stress and cop­ing strat­e­gies
among allo­ge­neic haematopoietic stem cell trans­plan­ta­tion sur­vi­vors: a
A. R. was ­able to make mod­est gains in his aca­demic suc­cess.
qual­i­ta­tive study. Eur J Cancer Care (Engl). 2020;29(6):e13307.
10. Sharafeldin N, Bosworth A, Patel SK, et  al. Cognitive func­ tion­
ing
after hema­to­poi­etic cell trans­plan­ta­tion for hema­to­logic malig­nancy:
Future direc­tions results from a pro­spec­tive lon­gi­tu­di­nal study. J Clin Oncol. 2018;36(5):
Table 5 pro­vi­des a suggested out­line of research pri­or­i­ties and 463-475.
11. Abel GA, Albelda R, Khera N, et al. Financial hard­ship and patient-reported
areas of clin­i­cal focus when con­sid­er­ing poten­tial psy­cho­so­cial out­comes after hema­to­poi­etic cell trans­plan­ta­tion. Biol Blood Marrow
risk and finan­cial dis­tress among HCT sur­vi­vors. With advances Transplant. 2016;22(8):1504-1510.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/570/1851740/570buchbinder.pdf by guest on 13 December 2021


in treat­ment prac­tices and sur­vival after HCT, there is an increas­ 12. Hamilton JG, Wu LM, Austin JE, et al. Economic sur­vi­vor­ship stress is asso­ci­
ing rec­og­ni­tion of the adverse impact of trans­plant on psy­cho­ ated with poor health-related qual­ity of life among distressed sur­vi­vors of
hema­to­poi­etic stem cell trans­plan­ta­tion. Psychooncology. 2013;22(4):911-
so­cial aspects of sur­vi­vor­ship, includ­ing the need to proactively
921.
col­lect and inter­vene in patient-reported out­comes after HCT. 13. Abel GA, Albelda R, Salas Coronado DY, et al. Prospective assess­ment of
Multiple ini­tia­tives in the field of HCT are begin­ning to iden­tify famil­ial finan­cial hard­ship after hema­to­poi­etic cell trans­plan­ta­tion. Biol
the gaps in prac­tice and research and rec­og­nize the impor­tance Blood Marrow Transplant. 2015;21(suppl 2):S72.
of inte­grat­ing patient-cen­tered out­come screen­ing and inter­ 14. Jeon M, Yoo IY, Kim S, Lee J. Post-trau­matic growth in sur­vi­vors of allo­ge­neic
hema­to­poi­etic stem cell trans­plan­ta­tion. Psychooncology. 2015;24(8):871-
ven­tions with the goal of improv­ing long-term health out­comes 877.
after HCT.2,4 The role of edu­ca­tional ini­tia­tives to empower and 15. Denzen EM, Majhail NS, Stickney Ferguson S, et  al. Hematopoietic cell
engage patients and their care­giv­ers in improv­ing their psy­cho­ trans­ plan­ ta­
tion in 2020: sum­ mary of year 2 rec­ om­ men­ da­ tions of the
so­cial health is extremely valu­able. A mul­ti­pronged approach to national mar­row donor pro­gram’s sys­tem capac­ity ini­tia­tive. Biol Blood
Marrow Transplant. 2013;19(1):4-11.
address psy­cho­so­cial con­cerns uti­liz­ing all­ rel­e­vant stake­hold­ers
16. Khera N, Hamilton BK, Pidala JA, et al. Employment, insur­ance, and finan­
can go a long way in ensur­ing the long-term suc­cess of this med­ cial expe­ri­ences of patients with chronic graft-ver­sus-host dis­ease in North
i­cally com­pli­cated, resource-inten­sive pro­ce­dure. America. Biol Blood Marrow Transplant. 2019;25(3):599-605.
17. Khera N, Albelda R, Hahn T, et al. Financial hard­ship after hema­to­poi­etic
cell trans­plan­ta­tion: lack of impact on sur­vival. Cancer Epidemiol Biomark-
Conflict-of-inter­est dis­clo­sure ers Prev. 2018;27(3):345-347.
David Buchbinder: no com­pet­ing finan­cial inter­ests to declare. 18. Denzen EM, Thao V, Hahn T, et al. Financial impact of allo­ge­neic hema­to­
Nandita Khera: no com­pet­ing finan­cial inter­ests to declare. poi­etic cell trans­plan­ta­tion on patients and fam­i­lies over 2 years: results
from a mul­ti­cen­ter pilot study. Bone Marrow Transplant. 2016;51(9):1233-
1240.
Off-label drug use
19. Eriksson L, Wennman-Larsen A, Bergkvist K, Ljungman P, Winterling J.
David Buchbinder: nothing to disclose. Important fac­tors asso­ci­ated with sick leave after allo­ge­neic haematopoi-
Nandita Khera: nothing to disclose. etic stem cell trans­plan­ta­tion—a 1-year pro­spec­tive study. J Cancer Surviv.
Published online 8 Jan­ua ­ ry 2021.
20. Lee SJ, Logan B, Westervelt P, et al. Comparison of patient-reported out­
Correspondence comes in 5-year sur­vi­vors who received bone mar­row vs periph­eral blood
Nandita Khera, College of Medicine, Mayo Clinic in Arizona, 5777 E unre­lated donor trans­plan­ta­tion: long-term fol­low-up of a ran­dom­ized clin­
Mayo Blvd, Phoenix, AZ 85054; e-mail: khera​­.nandita@mayo​­.edu. i­cal trial. JAMA Oncol. 2016;2(12):1583-1589.
21. Kirchhoff AC, Leisenring W, Syrjala KL. Prospective pre­dic­tors of return to
work in the 5 years after hema­to­poi­etic cell trans­plan­ta­tion. J Cancer Sur-
References viv. 2010;4(1):33-44.
1. Majhail NS, Tao L, Bredeson C, et  al. Prevalence of hema­to­poi­etic cell 22. Winterling J, Johansson E, Wennman-Larsen A, Petersson LM, Ljungman P,
trans­plant sur­vi­vors in the United States. Biol Blood Marrow Transplant. Alexanderson K. Occupational sta­tus among adult sur­vi­vors fol­low­ing allo-
2013;19(10):1498-1501. SCT. Bone Marrow Transplant. 2014;49(6):836-842.
2. Burns LJ, Abbetti B, Arnold SD, et al. Engaging patients in set­ting a patient- 23. Salit RB, Lee SJ, Burns LJ, et  al. Return-to-work guide­lines and pro­grams
cen­tered out­comes research agenda in hema­to­poi­etic cell trans­plan­ta­ for post-hema­to­poi­etic cell trans­plan­ta­tion sur­vi­vors: an ini­tial sur­vey. Biol
tion. Biol Blood Marrow Transplant. 2018;24(6):1111-1118. Blood Marrow Transplant. 2020;26(8):1520-1526.
3. Hahn T, Paplham P, Austin-Ketch T, et al. Ascertainment of unmet needs and 24. Park J, Wehrlen L, Mitchell SA, Yang L, Bevans MF. Fatigue pre­dicts impaired
par­tic­i­pa­tion in health main­te­nance and screen­ing of adult hema­to­poi­etic social adjust­ment in sur­vi­vors of allo­ge­neic hema­to­poi­etic cell trans­plan­ta­
cell trans­plan­ta­tion sur­vi­vors followed in a for­mal sur­vi­vor­ship pro­gram. tion (HCT). Support Care Cancer. 2019;27(4):1355-1363.
Biol Blood Marrow Transplant. 2017;23(11):1968-1973. 25. Nenova M, DuHamel K, Zemon V, Rini C, Redd WH. Posttraumatic growth,
4. Bevans M, El-Jawahri A, Tierney DK, et  al. National Institutes of Health social sup­port, and social con­straint in hema­to­poi­etic stem cell trans­plant
hema­to­poi­etic cell trans­plan­ta­tion late effects ini­tia­tive: the patient- sur­vi­vors. Psychooncology. 2013;22(1):195-202.
cen­tered out­comes work­ing group report. Biol Blood Marrow Transplant. 26. Yen HJ, Eissa HM, Bhatt NS, et al. Patient-reported out­comes in sur­vi­vors of
2017;23(4):538-551. child­hood hema­to­logic malig­nan­cies with hema­to­poi­etic stem cell trans­
5. Barata A, Gonzalez BD, Sutton SK, et al. Coping strat­e­gies mod­ify risk of plant. Blood. 2020;135(21):1847-1858.
depres­sion asso­ci­ated with hema­to­poi­etic cell trans­plant symp­tom­atol­ 27. Wilhelmsson M, Glosli H, Ifversen M, et al; Nor­dic Society of Pediatric
ogy. J Health Psychol. 2018;23(8):1028-1037. Hematology and Oncology. Long-term health out­comes in sur­vi­vors of
6. Seneviratne AK, Wright C, Lam W, Lipton JH, Michelis FV. Comorbidity pro­ child­hood AML treated with allo­ge­neic HSCT: a NOPHO-AML study. Bone
file of adult sur­vi­vors at 20 years fol­low­ing allo­ge­neic hema­to­poi­etic cell Marrow Transplant. 2019;54(5):726-736.
trans­plan­ta­tion. Eur J Haematol. 2021;106(2):241-249. 28. Luo Y, Yang P, Yang Y, et al. Cognitive and psy­cho­log­i­cal out­comes of pedi­
7. Liang J, Lee SJ, Storer BE, et al. Rates and risk fac­tors for post-trau­matic at­ric allo­ge­neic hema­to­poi­etic stem cell trans­plan­ta­tion sur­vi­vors in a sin­
stress dis­or­der symp­tom­atol­ogy among adult hema­to­poi­etic cell trans­ gle cen­ter in China. Medicine (Baltimore). 2019;98(39):e17307.

576  |  Hematology 2021  |  ASH Education Program


29. Brauer E, Pieters HC, Ganz PA, Landier W, Pavlish C, Heilemann MV. Com- 42. Solh MM, Speckhart D, Sol­o­mon SR, et al. The Transplant Evaluation Rat-
ing of age with can­cer: phys­i­cal, social, and finan­cial bar­ri­ers to inde­pen­ ing Scale pre­dicts over­all sur­vival after allo­ge­neic hema­to­poi­etic stem cell
dence among emerg­ing adult sur­vi­vors. Oncol Nurs Forum. 2018;45(2): trans­plan­ta­tion. Blood Adv. 2020;4(19):4812-4821.
148-158. 43. Pai ALH, Swain AM, Chen FF, et al. Screening for fam­ily psy­cho­so­cial risk in
30. Tremolada M, Bonichini S, Basso G, Pillon M. Perceived social sup­port and pedi­at­ric hema­to­poi­etic stem cell trans­plan­ta­tion with the psy­cho­so­cial
health-related qual­ity of life in AYA can­cer sur­vi­vors and con­trols. Psy- assess­ment tool. Biol Blood Marrow Transplant. 2019;25(7):1374-1381.
chooncology. 2016;25(12):1408-1417. 44. Mishkin AD, Shapiro PA, Reshef R, Lopez-Pintado S, Mapara MY. Standard-
31. Pulewka K, Wolff D, Herzberg PY, et al. Physical and psy­cho­so­cial aspects ized semi-struc­tured psy­cho­so­cial eval­u­a­tion before hema­to­poi­etic stem
of ado­les­cent and young adults after allo­ge­neic hema­to­poi­etic stem-cell cell trans­plan­ta­tion pre­dicts patient adher­ence to post-trans­plant reg­i­­
trans­plan­ta­tion: results from a pro­spec­tive mul­ti­cen­ter trial. J Cancer Res men. Biol Blood Marrow Transplant. 2019;25(11):2222-2227.
Clin Oncol. 2017;143(8):1613-1619. 45. de Souza JA, Yap BJ, Wroblewski K, et al. Measuring finan­cial tox­ic­ity as a
32. Walsh CA, Yi JC, Rosenberg AR, Crouch MV, Leisenring WM, Syrjala KL. clin­i­cally rel­e­vant patient-reported out­come: The val­i­da­tion of the COm-
Factors asso­ci­ated with social func­tion­ing among long-term can­cer sur­vi­ prehensive Score for Financial Toxicity (COST). Cancer. 2017;123(3):476-
vors treated with hema­to­poi­etic stem cell trans­plan­ta­tion as ado­les­cents 484.
or young adults. Psychooncology. 2020;29(10):1579-1586. 46. Prawitz AD, Garman TE, Sorhaindo B, O’Neill B, Kim JI, Drentea P. InCharge
33. Brauer ER, Pieters HC, Ganz PA, Landier W, Pavlish C, Heilemann MV. “From finan­cial dis­tress/finan­cial well-being scale: devel­op­ment, admin­is­tra­tion,

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/570/1851740/570buchbinder.pdf by guest on 13 December 2021


snail mode to rocket ship mode”: ado­les­cents and young adults’ expe­ri­ and score inter­pre­ta­tion. Financial Couns Plan. 2006;17(1):34-49.
ences of returning to work and school after hema­to­poi­etic cell trans­plan­ 47. Copeland AC, Tan X, Nash RP, et al. Collaborative care for depres­sion and
ta­tion. J Adolesc Young Adult Oncol. 2017;6(4):551-559. anx­i­ety in the bone mar­row trans­plant pop­u­la­tion: a pilot fea­si­bil­ity study.
34. Bhatt NS, Brazauskas R, Tecca HR, et al. Post-trans­plan­ta­tion employ­ment Psychooncology. 2021;30(1):118-122.
sta­tus of adult sur­vi­vors of child­hood allo­ge­neic hema­to­poi­etic cell trans­ 48. Kim WS, Langer S, Todd M, et al. Feasibility of a dig­i­tal sto­ry­tell­ing inter­ven­
plant: a report from the Center for International Blood and Marrow Trans- tion for hema­to­poi­etic cell trans­plant patients. J Cancer Educ. Published
plant Research (CIBMTR). Cancer. 2019;125(1):144-152. online 3 Jan­u­ary 2021.
35. Pulewka K, Strauss B, Hochhaus A, Hilgendorf I. Clinical, social, and psy­ 49. Balck F, Zschieschang A, Zimmermann A, Ordemann R. A ran­dom­ized con­
cho-onco­log­i­cal needs of ado­les­cents and young adults (AYA) ver­sus older trolled trial of prob­lem-solv­ing train­ing (PST) for hema­to­poi­etic stem cell
patients fol­low­ing hema­to­poi­etic stem cell trans­plan­ta­tion. J Cancer Res trans­plant (HSCT) patients: effects on anx­i­ety, depres­sion, dis­tress, cop­ing
Clin Oncol. 2021;147(4):1239-1246. and pain. J Psychosoc Oncol. 2019;37(5):541-556.
36. Majhail NS, Rizzo JD, Lee SJ, et al; Center for International Blood and Marrow 50. Somers TJ, Kelleher SA, Dorfman CS, et al. An mHealth pain cop­ing skills
Transplant Research; Amer­i­can Society for Blood and Marrow Transplanta- train­ing inter­ven­tion for hema­to­poi­etic stem cell trans­plan­ta­tion patients:
tion; Euro­pean Group for Blood and Marrow Transplantation; Asia-Pacific devel­op­ment and pilot ran­dom­ized con­trolled trial. JMIR Mhealth Uhealth.
Blood and Marrow Transplantation Group; Bone Marrow Transplant Society 2018;6(3):e66.
of Australia and New Zealand; East Med­i­ter­ra­nean Blood and Marrow Trans- 51. Syrjala KL, Crouch ML, Leisenring WM, et al. Engagement with INSPIRE, an
plantation Group; Sociedade Brasileira de Transplante de Medula Ossea. online pro­gram for hema­to­poi­etic cell trans­plan­ta­tion sur­vi­vors. Biol Blood
Recommended screen­ing and pre­ven­tive prac­tices for long-term sur­vi­ Marrow Transplant. 2018;24(8):1692-1698.
vors after hema­to­poi­etic cell trans­plan­ta­tion. Bone Marrow Transplant. 52. Yi JC, Sullivan B, Leisenring WM, et al. Who enrolls in an online can­cer sur­
2012;47(3):337-341. vi­vor­ship pro­gram? Reach of the INSPIRE ran­dom­ized con­trolled trial for
37. Khera N, Holland JC, Griffin JM. Setting the stage for uni­ver­sal finan­cial hema­to­poi­etic cell trans­plan­ta­tion sur­vi­vors. Biol Blood Marrow Trans-
dis­ tress screen­ ing in rou­ tine can­
cer care. Cancer. 2017;123(21):4092- plant. 2020;26(10):1948-1954.
4096. 53. Ferraro CS, Williams C, Dixon B, Bernhard L, Majhail NS, Hamilton BK.
38. Denzen EM, Preussler JM, Mur­phy EA, et  al. Tailoring a sur­vi­vor­ship care Neurocognitive and phys­i­cal func­tion­ing screen­ing in hema­to­poi­etic
plan: patient and pro­vider pref­er­ences for recip­i­ents of hema­to­poi­etic cell cell trans­plant (HCT) sur­vi­vor­ship clinic. Biol Blood Marrow Transplant.
trans­plan­ta­tion. Biol Blood Marrow Transplant. 2019;25(3):562-569. 2020;26(suppl 3):S365.
39. Majhail NS, Mur­phy E, Laud P, et al. Randomized con­trolled trial of indi­vid­ 54. Hoodin F, LaLonde L, Errickson J, et al. Cognitive func­tion and qual­ity of
u­al­ized treat­ment sum­mary and sur­vi­vor­ship care plans for hema­to­poi­etic life in vorinostat-treated patients after matched unre­lated donor myeloab-
cell trans­plan­ta­tion sur­vi­vors. Haematologica. 2019;104(5):1084-1092. lative con­di­tion­ing hema­to­poi­etic cell trans­plan­ta­tion. Biol Blood Marrow
40. Syrjala KL, Yi JC, Artherholt SB, et al. An online ran­dom­ized con­trolled trial, Transplant. 2019;25(2):343-353.
with or with­out prob­lem-solv­ing treat­ment, for long-term can­cer sur­vi­vors
after hema­to­poi­etic cell trans­plan­ta­tion. J Cancer Surviv. 2018;12(4):560-
570.
41. Hong S, Rybicki L, Corrigan D, et  al. Psychosocial Assessment of Candi-
dates for Transplant (PACT) as a tool for psy­cho­log­i­cal and social eval­u­
a­tion of allo­ge­neic hema­to­poi­etic cell trans­plan­ta­tion recip­i­ents. Bone © 2021 by The Amer­i­can Society of Hematology
Marrow Transplant. 2019;54(9):1443-1452. DOI 10.1182/hema­tol­ogy.2021000292

Prakash Singh Shekhawat


Psychosocial com­pli­ca­tions of HCT  |  577
SURVIVORSHIP AFTER ALLOGENEIC HEMATOPOIETIC CELL TRANSPLANT

Noninfectious complications of hematopoietic


cell transplantation

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/578/1851735/578williams.pdf by guest on 13 December 2021


Kirsten M. Williams
Blood and Marrow Transplant Program, Aflac Cancer and Blood Disorders Center, Emory University School of Medicine, Children’s Healthcare of
Atlanta, Atlanta, GA

Noninfectious lung diseases contribute to nonrelapse mortality. They constitute a spectrum of diseases that can affect
the parenchyma, airways, or vascular pulmonary components and specifically exclude cardiac and renal causes. The dif-
ferential diagnoses of these entities differ as a function of time after hematopoietic cell transplantation. Specific diagno-
sis, prognosis, and optimal treatment remain challenging, although progress has been made in recent decades.

LEARNING OBJECTIVES
• Describe the different pulmonary toxicities that occur after HCT and their timing post HCT as well as risk factors
and recommended treatments
• Describe the workup of lung toxicity and the current challenges to diagnosis and treatment of noninfectious lung
injury after HCT

Introduction Pulmonary differential of Case 1


Noninfectious lung complications after hematopoietic cell The differential diagnosis of noninfectious lung injury early
transplantation (HCT) are uncommon but associated with after HCT largely reflects alveolar injury, many mediated by
significant morbidity and mortality.1 Lung compromise endothelial dysfunction. These early lung injuries include
may be due to intrapulmonary injury to the parenchyma, peri­engraftment respiratory distress syndrome (PERDS),
airways, or vascular structures and excludes that due to capillary leak syndrome (CLS), diffuse alveolar hemorrhage
extrapulmonary, renal, or cardiac causes. Identifying the (DAH), acute interstitial pneumonitis (AIP), or toxic pneu­
correct diagnosis is critical but can be challenging with monitis/injuries, collectively termed idiopathic pneumonia
available tools, though timing after HCT narrows the differ­ syndrome (IPS). Additionally, vascular injury, such as pulmo­
ential. This review thus presents the differential diagnoses nary veno­occlusive disease (PVOD), can lead to respira­
and key findings as a function of time post transplant. tory decline early after HCT (Table 1 and Figure 1). These
diagnoses are associated with respiratory distress, include
radiographic abnormalities, and although pulmonary func­
tion tests (PFTs) are rarely performed, would demonstrate
CLINICAL CASE 1 a restrictive defect. The diagnosis of Case 1 is DAH.
A 9­month­old, who was transplanted for a nonmalignant
condition with a mismatched cord­blood donor after a Diffuse alveolar hemorrhage
reduced­intensity busulfan­based preparative regimen and Hallmarks of this diagnosis include hypoxia, respiratory
had recently engrafted, developed acute hypoxic respira­ distress, and new radiographic infiltrates in the perihilar
tory distress 28 days after HCT, in the setting of sinusoidal and lower lobes, with the diagnosis being confirmed by
obstructive syndrome treated with defibrotide. Fresh blood bloody BAL that does not clear, along with hemosiderin­
was noted on bronchoalveolar lavage (BAL) that did not laden macrophages. The incidence is 2% to 5% in alloge­
clear, as well as a new infiltrate on chest x­ray and rapid neic recipients and 1% in autologous. This patient had risk
progression to respiratory failure. An infectious and cardiac factors for DAH, including status as a cord­blood recipi­
workup was not revealing. Sadly, despite attempting high­ ent, defibrotide treatment, delayed engraftment, though
frequency oscillatory ventilation, this patient succumbed. not sirolimus prophylaxis, total body irradiation (TBI), and
myeloablation.2,3 Patients often present <30 days after HCT

578 | Hematology 2021 | ASH Education Program


Table 1. Diagnoses, key points, and treat­ments of non­in­fec­tious lung injury after HCT

Peak time post HCT/


Diagnosis struc­ture Key find­ings Additional diag­no­sis Proposed treat­ments
DAH <day 30 post HCT Hypoxia, new infil­trate, Hemosiderin-laden Inhaled transexamic acid,
endo­the­lial bloody BAL mac­ro­phages intrapulmonary
recom­bi­nant fac­tor VIIa
PERDS (IPS) Pre-engraft­ment Respiratory dis­tress/ Multilobar infil­trates Steroids/etanercept
Alveolar (from endo­the­lial hyp­oxia
dam­age)
CLS (IPS) Engraftment to day 15 Dyspnea/hyp­oxia, >3% Multilobar infil­trates Steroids/etanercept
Alveolar (from endo­the­lial weight gain
dam­age)

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/578/1851735/578williams.pdf by guest on 13 December 2021


AIP and toxin-related lung 50 days post HCT Fever, dyspnea, cough Ground glass on imag­ing/​
injuries, DPTC Alveolar bilat­eral infil­trates,
restric­tive PFT
IPS (PERDS, CLS, DAH, 45 days post HCT Hypoxia, pul­mo­nary Steroids/etanercept
AIP, DPTC) Alveolar infil­trates
CRS <7 days postcellular Respiratory com­pro­mise, Infiltrates Tociluzumab
ther­apy hyp­oxia
Alveolar
PVOD, PCT, TA-TMA 15-120 days post HCT Hypoxia, dyspnea, vas­cu­lar Sildenafil (PVOD, TA-TMA),
Endothelial occlu­sion, may prog­ress nitric oxide (TA-TMA) pros­
to pul­mo­nary hyper­ten­sion ta­cy­clins, cal­cium chan­nel
Biopsy diag­no­sis block­ers (PVOD) Steroids
(PCT)?
RLD after HCT Day 100-1 year Decreased FEV1, nor­mal PPFE, NSIP inter­sti­tial Etanercept
Alveolar FEV1/VC ratio, fibro­sis on pneu­mo­nia; can include Poor response to ste­roids
CT (upper lobes) other diag­noses
BOS Day 100-1.5 years FEV1 < 75%, ≥10% decline, Newer modal­i­ties: PRM and FAM: inhaled fluticasone,
Airway FEV1/VC LL of the 90% xenon-129 MRI azithromycin, montelukast
CI, absence of infec­tion +1 mg/kg/d pred­ni­sone
and either preexisting with rapid taper; ECP,
cGVHD, air trap­ping by etanercept, GERD tx, nutri­
expi­ra­tory CT or by PFT, or tion, infec­tion pro­phy­laxis
cir­cum­fer­en­tial fibro­sis of
bron­chi­­oles on biopsy
Non-HCT-spe­cific Alveolar (ARDS, PTLD) TRALI/TACO: tem­po­ral A1AT: obstruc­tive dis­ease, TRALI/TACO: sup­port­ive care
com­pli­ca­tions: Airway (A1AT) asso­ci­a­tion blood emphy­sema, PE: anti­co­ag­u­lants
TRALI/TACO, Endothelial (TRALI/ prod­ucts, fever, acute bron­chi­ec­ta­sis ARDS: treat under­ly­ing cause
PE, ARDS, A1AT, TACO, PE) dyspnea PTLD: nod­ules/EBV+ (often anti­mi­cro­bial) and
can­cer/PTLD, PE: dyspnea/hyp­oxia, V/Q+ sup­port­ive care
pneu­mo­tho­rax ARDS: fever, dyspnea, A1AT: A1AT infu­sion, inhaled
hyp­oxia, bilat­eral infil­trates ste­roids
PTLD: rituximab, cel­lu­lar
ther­apy
Pneumothorax: chest tube
EBV, Epstein-Barr virus; ECP, extracorporeal photopheresis; FAM, inhaled fluticasone, azithromycin, and montelukast + 1 mg/kg/d steroid burst and
rapid taper; GERD tx, gastro-esophageal reflux disease treatment; PRM; para­met­ric response map­ping; V/Q , ven­ti­la­tion/per­fu­sion scan.

and do poorly, with <25% of adults and <16% of chil­dren sur­viv­ inhaled transexamic acid. A small case series showed improved
ing a year.4,5 While the path­o­gen­e­sis is incom­pletely under­stood, sur­
vival in patients receiv­ ing intrabronchi recom­ bi­
nant fac­
tor
DAH is thought to reflect accu­mu­lated dam­age from con­di­tion­ VIIa com­pared with his­tor­i­cal patients who received ste­roids,10,11
ing reg­i­mens, aber­rant immune response, and sub­se­quent dam­ which is con­sis­tent with my expe­ri­ence. For patients too sick
age to the endo­the­lium, resulting in hem­or­rhage into alve­o­lar to inter­rupt the ven­ti­la­tory cir­cuit, inhaled transexamic acid, an
spaces. DAH can occur as a result of infec­tions, and thus a full antifibrinolytic agent, can be used with con­ven­tional ven­ti­la­tion.
infec­tious workup is advised (Table 2).6 Treatment his­tor­i­cally We have used this in sev­eral DAH patients with improve­ment
included ste­roids, though mul­ti­ple stud­ies have dem­on­strated (with­out ste­roids), based on published ret­ro­spec­tive data.12 Fi­
a lack of ben­e­fit, and some have shown harm.6-8 Similarly, amino­ nally, extra­cor­po­real mem­brane oxy­gen­a­tion has been rarely
caproic acid and sys­temic recom­bi­nant fac­tor VIIa have not ben­ employed.13 DAH is con­sis­tently diag­nosed, while its eti­­ol­ogy
efited DAH.8,9 In con­trast, small series have supported the use and opti­mal treat­ment are unknown, though local ther­a­pies may
of intrapulmonary admin­is­tra­tion of recom­bi­nant fac­tor VIIa and offer the greatest ben­e­fit, and high-dose ste­roid use has waned.
Prakash Singh Shekhawat
Noninfectious pul­mo­nary tox­ic­ity after HCT  |  579
for ben­e­fit and the rel­at­ive risks of infec­tion and can­cer recur­
rence. Collectively, these AIP- or toxin-induced lung injuries are
uncom­mon com­pli­ca­tions of HCT that would ben­e­fit from spe­
cific diag­nos­tic cri­te­ria and inves­ti­ga­tion into ste­roid-spar­ing
ther­a­peu­tic approaches.

PERDS and CLS


PERDS and CLS lead to alve­ o­lar dam­ age through endo­ the­
lial
injury, with pul­mo­nary edema as a down­stream event. The man­
i­fes­ta­tions include respi­ra­tory dis­tress and hyp­ox­emia, the pres­
ence of multilobar infil­trates by imag­ing, and the absence of infec­
tions, with PERDS typ­i­cally occur­ring within 5 days of engraft­ment,
and some suggesting even a week ear­lier, and CLS occur­ring in

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/578/1851735/578williams.pdf by guest on 13 December 2021


the first 15 days and accom­pa­nied by fluid over­load (>3% weight
gain). Recent data sug­gest PERDS occurs in n ­ early 5% of autol­
o­gous HCTs and 7% of allo­ge­neic HCTs.18,19 ­Several ele­va­tions in
Figure 1. Illustrated HCT noninfectious lung complications in sys­temic cyto­kines have been asso­ci­ated with engraft­ment syn­
terms of structure and peak timing after HCT. The time after drome, includ­ing ST2 protein (an IL-33 receptor), IL-2Rα, tumor
HCT is shown on the x-axis (in months) with the diseases. Light ­necro­sis fac­tor 1 between base­line and day 14, and procalcitonin
blue bubbles, parenchymal processes (CRS; PERDS; CLS, cap­ at day 0.19,20 Engraftment syn­drome is asso­ci­ated with increased
illary leak syndrome from endothelial damage; AIP; rP; DPTS; mor­tal­ity after HCT, though PERDS was not ana­lyzed sep­a­rately.19
RLD). Red vessels, the endothelial processes (DAH; PVOD; PCT; A sim­i­lar inci­dence is reported for CLS, which affected approx­i­
TA-TMA). Green airway, airway disease (BOS). IPS, which is most ma­tely 5% of allo­ge­neic HCT patients in a case series, though all­
often defined early post HCT, includes these diagnoses (defined but 0.6% of these cases were due to sep­sis and asso­ci­ated with
by hypoxia, infiltrates, alveolar injury, and excludes later diagno­ mor­tal­ity.21 A risk fac­tor for CLS may be preexisting vas­cu­lar dys­
ses that often do not present with these features). White box, regulation and cord blood trans­plant.22 PERDS and non­in­fec­tious
pulmonary noninfectious diagnoses not specific to HCT (TRALI; CLS are rarer causes of lung dys­func­tion early after HCT, char­ac­ter­
TACO; COP; PE; PTLD), which can occur throughout the HCT tra­ ized by endo­the­lial dys­func­tion and cyto­kine abnor­mal­i­ties, with
jectory. Above the table are key events occurring during HCT increased mor­tal­ity.
aGVHD and cGVHD.
IPS—a uni­fy­ing diag­no­sis
IPS is an umbrella diag­no­sis that has his­tor­i­cally incor­po­rated
AIP and toxin-related lung injury sev­eral of the above diag­noses. Most def­i­ni­tions have included
Pneumonitis, an inflam­ ma­ tion of the alveoli, can occur early PERDS, CLS, DAH, AIP, and DPTC in this ter­mi­nol­ogy, with diag­
after HCT due to infec­tions, the pre­par­a­tive reg­i­men, or be idi­o­ nos­tic cri­te­ria of non­in­fec­tious, noncardiogenic, nonrenal pul­mo­
pathic. The pre­sen­ta­tion is non­spe­cific, includ­ing fever, dyspnea, nary injury after HCT with asso­ci­ated pul­mo­nary infil­trates and
cough, and ground-glass opac­ i­
ties on imag­ ing. These toxin- hyp­ox­emia due to alve­o­lar injury.23-26 The term is an impor­tant
­induced injuries have had sev­eral terms linked to them, includ­ing uni­fy­ing diag­no­sis, per­mit­ting key clin­i­cal tri­als of rare diag­noses
AIP, radi­a­tion pneu­mo­ni­tis, carmustine pneu­mo­ni­tis, or delayed that col­lec­tively com­prise early non­in­fec­tious lung injury. The
pul­mo­nary tox­ic­ity syn­drome (DPTS from che­mo­ther­apy). Over­ inci­dence of IPS is 3.7% of allo­ge­neic adult HCT patients, with
all, the inci­dence of radi­a­tion and che­mo­ther­apy pneu­mo­ni­tis has a median onset of 43 days, though later onset is described, and
decreased with mod­i­fi­ca­tions to che­mo­ther­apy reg­i­mens and risk fac­tors of myeloablative con­di­tion­ing, high-dose TBI, lower-
the advent of lung blocking and frac­tion­ated TBI.14,15 The recent respi­ra­tory tract viruses pre-HCT, and inborn errors of metab­o­
over­all inci­dence of non­in­fec­tious pneu­mo­ni­tis was 5% one year lism as indi­ca­tion for HCT.25,27 In chil­dren, the most recent data
after HCT, with a median onset of 1.6 months in chil­dren.5 Risk (2014) showed a rate of 6.7% in allo­ge­neic recip­i­ents.28 Approxi­
fac­tors included can­cer indi­ca­tion and myeloablative con­di­tion­ mately 5% of autol­o­gous trans­plant recip­i­ents incur IPS.29 Nota­
ing.5 There was also an asso­ci­a­tion with cord blood trans­plan­ta­ bly, this rate is lower than the rate of the com­pos­ite diag­noses
tion, which may reflect the increased use in TBI in this cohort, com­bined and may reflect the het­ero­ge­ne­ity of dis­or­ders and
though it was not discussed. After myeloablative TBI in adults, patients included under the IPS umbrella. Outcomes remain
the inci­dence of non­in­fec­tious radi­a­tion-induced IP is 7.4%, with poor after allo­ge­neic HCT, with <16% to 30% of chil­dren and
a median onset of 2.5 months, and less after che­mo­ther­apy con­ adults sur­viv­ing a year.25,28,30,31 Many stud­ies have linked ele­vated
di­tion­ing.14 The over­all sur­vival for adults and chil­dren is 50% BAL lev­els of tumor necro­sis growth fac­tor, ST2, and IL-6 to IPS
and 40%, respec­tively.5,14 Reducing lung expo­sure dur­ing TBI has at diag­no­sis.26,32-34 Some stud­ies, though not all­, have shown a
­decreased the rates of radi­a­tion-induced IP fur­ther.16 Other toxin- ben­e­fit from etanercept with cor­ti­co­ste­roids, although less of
medi­ated early lung com­pli­ca­tions can include cyclo­phos­pha­ an impact on long-term sur­vival.26,31,35 The risk of death was high­
mide-induced, eosin­o­philic pneu­mo­nia linked to fludarabine; IP er in patients with prior pul­mo­nary impair­ment, those who re­
or DAH with rituximab; DAH after alemtuzumab; or pneu­mo­ni­tis quired ven­ti­la­tory sup­port, or those who had con­cur­rent renal
after check­point inhib­i­tors.15,17 While ste­roids are often cited for or hepatic injury.25 IPS is a ben­e­fi­cial umbrella diag­no­sis of early
treat­ment of these pneu­mo­ni­tis and toxin-induced injuries, data life-threat­en­ing non­in­fec­tious pul­mo­nary injury after HCT with­
sug­gest that they have fallen out of favor, given the sparce data out con­sen­sus on its opti­mal treat­ment.

580  |  Hematology 2021  |  ASH Education Program


Table 2. Workup con­sid­er­ations of non­in­fec­tious lung injury after HCT

Type Test Considerations


Radiographic CT-inspi­ra­tory Noncontrast suf­fi­cient for most; bron­chi­ec­ta­sis, enlarged
pul­mo­nary ves­sels may be seen in some diag­noses
CT-added expi­ra­tory Valuable to assess air trap­ping (BOS or RLD)
MRI/MRA May iden­tify vas­cu­lar dis­ease
V/Q scan Evaluate for PE
Chest x-ray Less valu­able if air trap­ping is pres­ent
BAL PCP (PJP) PCR PCR tests are increas­ingly valu­able
Explify PCR Explify can detect mul­ti­ple path­o­gens by NGS and may
Respiratory virus PCR detect organ­isms at low lev­els not evi­dent in cul­ture,
Gram stain/cul­ture which may be most valu­able for pretreated patients or
Fungal stain/cul­ture dis­tal lesions

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/578/1851735/578williams.pdf by guest on 13 December 2021


Cytology (sil­ver stain) Blood return that does not clear is diag­nos­tic for DAH
Nocardia cul­ture Visual inspec­tion can iden­tify fun­gal lesions or other
AFB smear/cul­ture indi­ca­tors
CMV PCR Because BAL fluid quan­tity is per kilo­gram and return is
HSV PCR dimin­ished in severe lung injury, it may be impor­tant to
Mycoplasma pneumoniae PCR pri­or­i­tize tests in chil­dren
Legionella pneumophilia cul­ture
Galactomannan
Blood Blood cul­ture Karius uses NGS and can detect lung organ­isms with good
Karius sen­si­tiv­ity, though data sug­gest that some pul­mo­nary
Blood gas infec­tions may be missed (eg, fun­gal nod­ules)
Consider met­he­mo­glo­bin if hyp­oxic and med­i­ca­tions at risk
Urine Histoplasmosis anti­gen
PFTs FEV1, FEV1/VC ratio, RV and RV/TLC, DLCO, Lung clear­ance index may be valu­able 6 min­utes’ walk time
+/−albuterol is a mea­sure of endur­ance for BOS
FEV1 slope of decline can be valu­able for prog­no­sis in BOS
Lung biopsy Pathology, stains, and cul­tures High mor­bid­ity should prompt care­ful con­sid­er­ation of
risk/ben­e­fit anal­y­sis
Echocardiogram/+/−angi­og­ra­phy Pulmonary hyper­ten­sion May also iden­tify peri­car­dial effu­sions
AFB, acid fast bacil­lus; CMV, cyto­meg­a­lo­vi­rus; DLCO, dif­fu­sion capac­ity of lung for car­bon mon­ox­ide; HSV, her­pes sim­plex virus; MRA, mag­netic ­
res­o­nance angi­og­ra­phy; PJP/PCP, Pneumocystis jirovecii pneu­mo­nia (for­merly Pneumocystis carinii pneu­mo­nia); TLC, total lung capac­ity.

Cytokine release syn­drome regard­ing co-inci­dence, and the peak tim­ing dif­fers for these
A new diag­no­sis has emerged related to the admin­is­tra­tion of dis­eases (ear­lier after HCT for SOS and later for PVOD).41 PVOD
genet­i­cally mod­i­fied T cells (chi­me­ric anti­gen recep­tor T-cell is dif­fi­cult to diag­nose, with dyspnea and non­spe­cific find­ings
ther­apy; CAR-T). Cytokine release syn­drome (CRS) can include on imag­ing that even­tu­ally man­i­fest as right ventricular dys­func­
respi­ra­tory com­pro­mise, with infil­trates, hyp­oxia, and endo­the­ tion.42 PVOD has been linked to myeloablative HCT and acute
lial dys­func­tion that closely mim­ics CLS.36-38 A large inter­na­tional graft-­ver­sus-host dis­ease (aGVHD) and other air­way dis­eases.40
study showed that 25% of patients with CAR-T had sig­nif­i­cant Treatment with nitrates may be del­e­te­ri­ous, while cal­cium chan­
pul­mo­nary com­pro­mise.39 Risk fac­tors for CRS include high dis­ nel block­ers, pros­ta­cy­clins, and sildenafil have suggested pos­
ease bur­den, con­cur­rent infec­tions, and lymphodepletion prior si­ble ben­e­fit.42 Defibrotide, an antithrombotic and fibri­no­lytic
to CAR-T.37,38 The role of IL-6 in this pro­cess has been established agent that sta­bi­lizes the endo­the­lium, has yet to be inves­ti­gated
and block­ade (via tocilizumab) has miti­gated tox­ic­ity, some­times for PVOD after HCT, though the mech­a­nism of action and suc­
with ste­roid coad­min­is­tra­tion.37,38 These data beg the ques­tion of cess in SOS would sup­port its use. Rare reports of pul­mo­nary
whether IL-6 block­ade could be valu­able in IPS as well, though as cyto­lytic thrombi (PCT) after HCT appeared in the early 2000s,
yet this treat­ment remains untested. with pre­sen­ta­tion marked by fever, often in patients with ac­
tive aGVHD who had nod­ules on imag­ing and biopsy show­ing
PVOD, pul­mo­nary cyto­lytic thrombi, and trans­plant-­ leu­ko­cytes and baso­philic occlu­sions.43 TA-TMA is a syn­drome
asso­ci­ated throm­botic microangiopathy dis­or­der ­after HCT of endo­the­lial dys­func­tion most com­monly
PVOD dis­ease, PCT, and trans­plant-asso­ci­ated throm­botic micro­ affect­ing the kid­ney and is dif­fi­cult to diag­nose with­out biopsy.
angiopathy (TA-TMA) are endo­the­lial lung com­pli­ca­tions rarely It can lead to pul­mo­nary arte­rial hyper­ten­sion and hyp­ox­emia,
described after HCT that are iden­ti­fied by pathol­ogy and usu­ has been dem­on­strated on biopsy with fibrin occlu­sion of small
ally result in poor out­comes. PVOD is due to inti­mal fibro­sis and pul­mo­nary ves­sels, and has been treated with sildenafil and ni­
venule hyper­tro­phy and obstruc­tion.40 While the path­o­logic tric oxide.44,45 Given the sim­i­lar­i­ties among these diag­noses, it is
find­ings are sim­i­lar to those found in the liver of hepatic VOD unclear if they are sep­a­rate enti­ties or a spec­trum of dis­ease pro­
(sinu­soi­dal obstruc­tive syn­drome; SOS), the lit­er­a­ture is lacking cesses. Collectively, these rare endo­the­lial dis­eases would ben­e­
Prakash Singh Shekhawat
Noninfectious pul­mo­nary tox­ic­ity after HCT  |  581
fit from fur­ther study, refined diag­nos­tic cri­te­ria, and sys­tem­atic 2 has sev­eral risk fac­tors linked to BOS, includ­ing busul­fan con­di­
eval­u­a­tion of treat­ment approaches. tion­ing, viral infec­tions early after HCT, and aGVHD, with periph­
eral blood stem cell donor and ABO incom­pat­i­bil­ity not pres­
Other pul­mo­nary diag­noses after HCT ent.47,50 A recent study sug­gests that pre-HCT viruses may con­fer
HCT recip­i­ents are at risk for pul­mo­nary prob­lems that com­ increased risk as well.27 The cur­rent inci­dence is approx­i­ma­tely 3%
monly occur in crit­i­cal ill­ness. These include inflam­ma­tory con­ in pedi­at­ric and 3% to 6% in adult HCT.1,51-53 A chal­lenge of this dis­
di­tions, acute respi­ra­tory dis­tress syn­drome (ARDS), trans­fu­sion- ease has been its insid­i­ous nature lead­ing to mod­er­ate decline at
asso­ci­ated acute lung injury (TRALI), trans­fu­sion-asso­ci­ated diag­no­sis. The fre­quent use of home devices to detect early de­
cir­cu­la­tory over­load (TACO), and cryp­to­genic orga­niz­ing pneu­ clines in lung func­tion and the use of FEF25-75 as an indi­ca­tor may
mo­ nia (COP; pre­ vi­
ously termed bronchiolitis obliterans and aid in the ear­lier iden­ti­fi­ca­tion of BOS.52,54,55 Newer imag­ing and
orga­niz­ing pneu­mo­nia), which is respon­sive to ste­roids and serum mark­ers show prom­ise for BOS diag­no­sis, includ­ing para­
restric­tive on PFTs and often linked to infec­tions. Other con­di­ met­ric response map­ping (with recent data for machine-learn­ing
tions can con­trib­ute to pul­mo­nary dys­func­tion, includ­ing algo­rithms), xenon-129 mag­netic res­o­nance imag­ing (MRI), and

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/578/1851735/578williams.pdf by guest on 13 December 2021


­alpha-1-antitrypsin defi­ciency (A1AT), pneu­mo­tho­rax, pul­mo­nary serum MMP3, though the data are insuf­fic ­ ient to replace PFTs.56-59
embolism, and, finally, can­cer relapse or posttransplant lymph­ This may be most crit­i­cal for chil­dren who can­not per­form PFTs
oproliferative dis­or­ders (PTLDs). in the future.60 Therapy for BOS con­sis­tently has striven for dis­
ease sta­bi­li­za­tion because the nat­ur­al his­tory of BOS is steady
decline, with severe FEV1 declines asso­ci­ated with highest mor­
tal­ity.61 A pro­spec­tive, multi-insti­tu­tional study of inhaled flutica­
CLINICAL CASE 2 sone, azithromycin, and montelukast with a brief ste­roid burst
One year after matched-sib­ling HCT for relapsed acute leu­ke­ (to 1 mg/kg/d pred­ni­sone equiv­a­lent with rapid taper­ing off in
mia, with busul­fan con­di­tion­ing, com­pli­cated by severe aGVHD 1 month) showed sta­bi­li­za­tion or improve­ment in 64% of patients
and cyto­meg­a­lo­vi­rus reactivation before day 100, a 12-year-old with new-onset BOS at 6 months.62 Immunosuppression with cal­
girl presented with dyspnea on exer­tion and ocu­lar chronic cineurin inhib­i­tor or sirolimus was con­tin­ued. This approach has
GVHD (cGVHD). She under­went lung biopsy, which con­firmed been endorsed by the Euro­pean Society for Blood and Marrow
bronchiolitis obliterans (forced expi­ra­tory vol­ume in 1 sec­ond Transplantation as first-line ther­apy for BOS after HCT.63 One of
[FEV1]/VC ratio >0.7). Although she had mycobacteria, FEV1 these agents, azithromycin, came under scru­tiny due to increased
failed to fully recover with protracted anti­mi­cro­bi­als con­sis­tent relapse rates in a ran­dom­ized trial of azithromycin prior to HCT,64
with biopsy find­ings of bronchiolitis obliterans syn­drome (BOS), which were not con­firmed in a larger study of azithromycin in
improv­ing from 46% to 59% but then plateauing while still on established BOS, though a higher rate of sec­ond­ary neo­plasms
sirolimus treat­ment. She was treated in a trial with montelukast was observed.65 At this time, the ben­e­fits of azithromycin likely
with ris­ing FEV1 (to 73%, with FEV1 of 1.56-1.87 L at 2 years). out­weigh the risks for most BOS patients, though ste­roids and
Now, over a decade later, her FEV1 is 2.08 L, 85% predicted, and other risks for sec­ond­ary neo­plasms should also be min­i­mized
she remains on montelukast and treat­ment for mycobacteria and per­son­al­ized risks eval­u­ated. In patients with ongo­ing pul­mo­
(which recurred after 2 anti­mi­cro­bial ces­sa­tions). nary decline, extra­cor­po­real photopheresis may improve sur­vival,
which builds upon prior ret­ro­spec­tive data of a response rate of
approx­i­ma­tely 60%.47,66 Etanercept has shown some suc­cess for
Pulmonary dif­fer­en­tial in Case 2 BOS (32%), while ruxolitinib has not had an impact on BOS pro­
Late-onset non­in­fec­tious lung dis­ease affects nearly 20% of 100- gres­sion.67 A chal­lenge to eval­u­at­ing ther­a­peu­tic suc­cess is the
day HCT sur­vi­vors.46 Overall risk fac­tors include chest r­adi­a­tion relaps­ing-remit­ting nature of BOS, though rapid declines have
prior to HCT, lung infec­tion within the first 100 days after HCT, and con­sis­tently been asso­ci­ated with poorer out­comes, and the FEV1
a reduced mean forced expi­ra­tory flow at 25% to 75% (FEF25-75) slope can thus guide prog­no­sis.68 Lung trans­plan­ta­tion remains a
at 100 days after HCT.46 BOS is the diag­no­sis of Case 2. final option, and recent data sug­gest improved sur­vival of 80%
at a median of 5 years.69 Overall, the sur­vival of patients with BOS
Bronchiolitis obliterans syn­drome has improved to 40% to 50% at 5 years, which likely reflects the
BOS is the accepted pul­mo­nary man­i­fes­ta­tion of cGVHD, diag­ incor­po­ra­tion of newer ther­a­pies, a reduc­tion of protracted high-
nosed by an FEV1 < 75% predicted and an irre­vers­ible ≥10% decline dose ste­roids, and improve­ments in sup­port­ive care.47 As high­
in <2 years, an FEV1/vital capac­ity (VC) ratio <0.7 or the lower lighted by our case, chil­dren may expe­ri­ence an unique tra­jec­tory
limit (LL) of the 90% CI of the ratio, an absence of infec­tion, and ei­ of recov­ery, which I have observed after sig­nif­i­cant growth and
ther preexisting cGVHD, air trap­ping by expi­ra­tory com­put­er­ized which was recently published in a ret­ro­spec­tive cohort.51
tomog­ra­phy (CT) or by PFT, or cir­cum­fer­en­tial fibro­sis of bron­chi­­
oles on biopsy.47,48 This case high­lights sev­eral impor­tant aspects Restrictive lung dis­ease
of the revised def­i­ni­tion of BOS, includ­ing the abil­ity to diag­nose Restrictive lung dis­ease (RLD) after HCT is less well char­ac­ter­ized.
obstruc­tive dis­ease in chil­dren for whom the LL of nor­mal is far RLD has a much larger dif­fer­en­tial to exclude, such as infec­tions,
higher than 0.7. Now this child could be diag­nosed with­out biopsy. COP, and extrapulmonary con­straints, eg, scle­ro­sis or myo­si­tis.
As the FEV1/VC ratio declines with age, this is also a key mod­i­fi­ca­ Intraparenchymal pathol­ ogy can include elas­ tic fibro­
sis of the
tion for older indi­vid­u­als for whom 0.7 may be a nor­mal value, and pleura or paren­chyma, typ­i­cally in the upper lobes, termed pleu­
a diag­no­sis of BOS is not warranted. In addi­tion, this case high­ ral paren­chy­mal fibroelastosis (PPFE) or non­spe­cific inter­sti­tial
lights the value of BAL, which could have diag­nosed mycobac­ pneu­mo­nia (NSIP). A study of lung explants suggested that late
teria and excluded rare dis­eases, eg, bronchial obliterans.49 Case RLD was due to paren­chy­mal fibro­sis with 20% to 30% of small

582  |  Hematology 2021  |  ASH Education Program


air­ways obstructed (a much lower pro­por­tion than patients with The chang­ing land­scape of lung injury after HCT
BOS, in which 70% were obstructed).70 These RLD find­ings can oc­ Infections and lung injury
cur with many other HCT-asso­ci­ated lung pathol­o­gies, includ­ing The def­i­ni­tion of non­in­fec­tious lung injury is a mov­ing tar­get.
BOS, PVOD, and lym­pho­cytic bronchiolitis.71 Because che­mo­ther­ Polymerase chain reac­ tion (PCR) stud­ ies have improved the
apy can induce PPFE or NSIP, the con­tri­bu­tion of cGVHD has been detec­tion of viral, bac­te­rial, and fun­gal path­o­gens, which has
dif­fi­cult to dis­cern. However, a recent meta-anal­y­sis com­pared been fur­ther enhanced by next-gen­er­a­tion sequenc­ing (NGS)
the inci­dence of PPFE in patients with che­mo­ther­apy or autol­ with high sen­si­tiv­ity and spec­i­fic­ity.78-81 Using such PCR meth­ods
o­gous HCT to those with allo­ge­neic HCT and showed a higher from a ret­ro­spec­tive cohort, 57% of IPS patients had a detect­
rate in allo­ge­neic HCT (71% vs 25%), suggesting that alloimmunity able path­o­gen in the BAL.82 By improv­ing infec­tion diag­nos­tics,
plays a role in this pro­cess.72 A long-term sur­vi­vor pedi­at­ric study the role of infec­tions will be bet­ter elu­ci­dated.
showed an asso­ci­a­tion between RLD and GVHD as well as pre- We may need to mod­ify our approach to the con­sid­er­ation of
HCT pul­mo­nary dys­func­tion.73 There are no diag­nos­tic cri­te­ria for infec­tions with lung injury. Some non­in­fec­tious lung injuries are
RLD after HCT, though these find­ings have been reported: fibro­sis likely late sequelae from an acute infec­tious pro­cess, eg, BOS,

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/578/1851735/578williams.pdf by guest on 13 December 2021


on CT (espe­cially the upper lobes) and reduced dif­fu­sion capac­ity linked to viral infec­tions after HCT, and pro­gres­sion has been
and an increased resid­ual vol­ume (RV)/total lung capac­ity ratio, linked to infec­tions as well.83
a 20% decline in FEV1 with a nor­mal FEV1/VC ratio, and reduced
VC.74,75 From adult series, the inci­dence of RLD would seem rarer Challenges to diag­no­sis, treat­ment, and pre­ven­tion
than BOS, up to 2.6% to 5%.46,74 However, inter­est­ingly, pedi­at­ric There are sig­nif­i­cant diag­nos­tic chal­lenges to non­in­fec­tious
reports of long-term sur­vi­vors (>6 years) sug­gest RLD is com­mon, lung injuries. The diag­ nos­tic cri­te­
ria for BOS and IPS have
in up to 50% of sur­vi­vors, high­light­ing the need for con­sen­sus changed over time, lead­ing to chal­lenges in com­par­ing stud­
diag­nos­tic cri­te­ria.73,76 Little is known to guide treat­ments of RLD; ies, though both have published con­sen­sus cri­te­ria.15,23,24,48,84-86
approx­ i­
ma­tely one-third of patients responded to etanercept, RLD lacks diag­nos­tic cri­te­ria alto­gether. It will be crit­i­cal to
while ruxolitinib did not affect dis­ease.67,77 While newer stud­ies con­tinue to use these stan­dard­ized diag­nos­tic cri­te­ria for these
have granted insight into the var­i­able pathol­ogy of this dis­ease, chal­leng­ing diag­noses and refine with con­sen­sus to per­mit
the clin­i­cal diag­nos­tic cri­te­ria and eti­­ol­ogy remain ill-defined. broad appli­ca­tion.

Figure 2. Flow diagram for workup and diagnosis of noninfectious lung diseases after HCT. Blue box, diseases commonly diagnosed
in the first 100 days after HCT. Gray box, those diseases occurring usually beyond day 100 after HCT. Noninfectious lung injury workup
and diagnoses are in boxes. *Additional pulmonary diagnoses are denoted by stars and listed in the column in which these diagnoses
would be included in the differential. Notably, other processes can exhibit low DLCO, which is often reduced in RLD, but isolated
DLCO reduction should prompt evaluation for vascular diseases of the lung. For the workup of obstructive disease, infection is often
diagnosed, which should prompt repeat testing after treatment to ascertain BOS diagnosis (arrows). bx, biopsy; DLCO, diffusion
capacity of lung for carbon monoxide; ID, infectious disease workup; PE, pulmonary embolism; toxin-IP, toxin associated interstitial
pneumonitis, including that from radiation or chemotherapy.
Prakash Singh Shekhawat
Noninfectious pul­mo­nary tox­ic­ity after HCT  |  583
Similarly, many of these diag­noses lack spe­cific diag­nos­tic Acknowledgments 
cri­te­ria. The endo­the­lial syn­dromes of PERDS, CLS, and TA-TMA Drs. Jennifer Holter Chakrabarty, James George, Gregory Yan­
are par­tic­u­larly plagued by this issue, such that a recent review ik, and Guang-Shing Cheng are acknowl­edged for their expert
made a “plea” for con­sen­sus def­i­ni­tions.86 While there are sub­ coun­sel dur­ing this arti­cle’s prep­a­ra­tion.
tle dif­fer­ences in the find­ings and risk fac­tors of PERDS/CLS and
idi­o­pathic or toxin-related pneu­mo­ni­tis, there is no way to dis­ Conflict-of-inter­est dis­clo­sure
tin­ guish these diag­ noses clin­ i­
cally. It is also unclear whether Kirsten M. Williams: no com­pet­ing finan­cial inter­ests to declare.
these lung injuries result from a final com­mon immu­no­logic path­
way or result from dis­tinct mech­a­nisms. Addressing this crit­i­cal Off-label drug use
ques­tion could enhance our abil­ity to iden­tify bio­mark­ers and Kirsten M. Williams: All drugs recommended in this manuscript
targeted ther­a­pies for these com­po­nents of IPS as well as the constitute off-label use with the exception of tociluzumab for
pul­mo­nary vas­cu­lar diag­noses. CRS. Drugs that are used off label in non-infectious lung treat­
Accurate and non­in­va­sive diag­nos­tic tools that iden­tify lung ments include steroids, etanercept, fluticasone, azithromycin,

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/578/1851735/578williams.pdf by guest on 13 December 2021


diag­noses remain an elu­sive goal. Lung biopsy yields the diag­ montelukast, inhaled transexamic acid, recombinant factor VII.
no­sis in 50% of patients but is asso­ci­ated with mor­bid­ity and
mor­tal­ity.87 BAL yields fewer diag­noses with fewer com­pli­ca­tions Correspondence
and likely a higher yield with newer modal­i­ties (eg, NGS).87 Fig­ Kirsten M. Williams, Blood and Marrow Transplant Program,
ure 2 and Table 2 sum­ma­rize a data-driven workup approach. As Aflac Cancer and Blood Disorders Center, Emory University
is high­lighted in Figure 2, many pro­vid­ers attempt a diag­no­sis School of Medicine, Children’s Healthcare of Atlanta, 1760 Hay­
through non­in­va­sive means (imag­ing, BAL, serum test­ing) and good Dr, 3rd floor W362, Atlanta, GA 30322; e-mail: kirsten​­.marie​
reserve lung biopsy for those patients with non­spe­cific results ­.williams@emory​­.edu.
from these tests. Notably, these tests typ­ic ­ ally require over a
week to yield results, prompting most prac­ti­tion­ers to pre­emp­ References
1. Duque-Afonso J, Ihorst G, Waterhouse M, et al. Impact of lung func­tion on
tively treat broadly for infec­tions and for a non­in­fec­tious diag­
bronchiolitis obliterans syn­drome and out­come after allo­ge­neic hema­to­
no­sis of high sus­pi­cion. Biopsy is still often required to diag­nose poi­etic cell trans­plan­ta­tion with reduced-inten­sity con­di­tion­ing. Biol Blood
vas­cu­lar com­pli­ca­tions, though some cen­ters have tried treat­ Marrow Transplant. 2018;24(11):2277-2284.
ments for the pul­mo­nary VOD or TA-TMA with­out biopsy and 2. Fan K, McArthur J, Morrison RR, Ghafoor S. Diffuse alve­o­lar hem­or­rhage after
pedi­at­ric hema­to­poi­etic stem cell trans­plan­ta­tion. Front Oncol. 2020;10
used response as a method of diag­no­sis (often while awaiting
(9 Sep­tem­ber):1757.
results from the BAL tests). Given the high rate of pneumotho­ 3. Keklik F, Alrawi EB, Cao Q , et al. Diffuse alve­o­lar hem­or­rhage is most often
raxes in patients with BOS, attempting to make this diag­no­sis fatal and is affected by graft source, con­di­tion­ing reg­i­men tox­ic­ity, and
with­out biopsy is advan­ta­geous. Because this remains a clin­i­cal engraft­ment kinet­ics. Haematologica. 2018;103(12):2109-2115.
chal­lenge, hope­fully, imag­ing or blood bio­mark­ers will be valu­ 4. Zhang Z, Wang C, Peters SG, et  al. Epidemiology, risk fac­tors, and out­
comes of dif­fuse alve­o­lar hem­or­rhage after hema­to­poi­etic stem cell trans­
able for non­in­va­sive diag­no­sis in the future.
plan­ta­tion. Chest. 2021;159(6):2325-2333.
The pre­ven­tion of lung injuries after HCT is largely an unat­ 5. Broglie L, Fretham C, Al-Seraihy A, et al. Pulmonary com­pli­ca­tions in pedi­
tain­able end point because of the lack of knowl­edge of the at­ric and ado­les­cent patients fol­low­ing allo­ge­neic hema­to­poi­etic cell
mech­a­nisms under­pin­ning these dis­eases. The reduc­tion of lung trans­plan­ta­tion. Biol Blood Marrow Transplant. 2019;25(10):2024-2030.
6. Majhail NS, Parks K, Defor TE, Weisdorf DJ. Diffuse alve­o­lar hem­or­rhage and
injury through targeted dos­ing (of radi­a­tion or che­mo­ther­apy)
infec­tion-asso­ci­ated alve­o­lar hem­or­rhage fol­low­ing hema­to­poi­etic stem
has dimin­ished the pneu­mo­ni­tis related to these insults. How­ cell trans­plan­ta­tion: related and high-risk clin­i­cal syn­dromes. Biol Blood
ever, most of the non­in­fec­tious lung injuries lack known mod­ Marrow Transplant. 2006;12(10):1038-1046.
i­fia
­ ble incit­ing fac­tors. Further, the lack of objec­tive spe­cific 7. Haider S, Durairajan N, Soubani AO. Noninfectious pul­mo­nary com­pli­ca­
diag­nos­tic cri­te­ria, the rar­ity of these dis­eases, and the rel­a­tive tions of haematopoietic stem cell trans­plan­ta­tion. Eur Respir Rev. 2020;
29(156):190119.
pau­city of ani­mal mod­els that mimic the clin­i­cal con­di­tion have
8. Rathi NK, Tanner AR, Dinh A, et  al. Low-, medium- and high-dose ste­
ham­pered prog­ress in under­stand­ing these dis­eases to develop roids with or with­ out aminocaproic acid in adult hema­ to­
poi­etic SCT
pre­ven­tive strat­e­gies. Finally, the unin­tended con­se­quences of patients with dif­fuse alve­o­lar hem­or­rhage. Bone Marrow Transplant. 2015;
diminishing some known risk fac­tors, eg, aGVHD, can lead to 50(3):420-426.
9. Elinoff JM, Bagci U, Moriyama B, et al. Recombinant human fac­tor VIIa for
increased life-threat­en­ing risks, eg, relapse and infec­tions, con­
alve­o­lar hem­or­rhage fol­low­ing allo­ge­neic stem cell trans­plan­ta­tion. Biol
straining some efforts due to the need to bal­ance these risks.88,89 Blood Marrow Transplant. 2014;20(7):969-978.
10. Baker MS, Diab KJ, Carlos WG, Mathur P. Intrapulmonary recom­bi­nant fac­
tor VII as an effec­tive treat­ment for dif­fuse alve­o­lar hem­or­rhage: a case
Conclusion series. J Bronchology Interv Pulmonol. 2016;23(3):255-258.
11. Park JA, Kim BJ. Intrapulmonary recom­bi­nant fac­tor VIIa for dif­fuse alve­ol­ar
Noninfectious lung injury con­ trib­
utes to nonrelapse mor­ tal­
ity
hem­or­rhage in chil­dren. Pediatrics. 2015;135(1):e216–e220.
and mor­bid­ity. It usu­ally devel­ops in the first year after HCT and 12. O’Neil ER, Schmees LR, Resendiz K, Justino H, Anders MM. Inhaled
can affect the paren­chyma, air­ways, or vas­cu­lar struc­tures. Tim­ tranexamic acid as a novel treat­ment for pul­mo­nary hem­or­rhage in crit­
ing, pre­sen­ta­tion, and imag­ing can aid in distinguishing the diag­ i­
cally ill pedi­ at­
ric patients: an obser­ va­
tional study. Crit Care Explor.
no­sis, and mul­ti­ple diag­noses can coex­ist. However, diag­no­sis 2020;2(1):e0075.
13. Fan K, Hurley C, McNeil MJ, et  al. Case report: man­age­ment approach
and treat­ment are areas of crit­i­cal need. A bet­ter under­stand­ing and use of extra­cor­po­real mem­brane oxy­gen­a­tion for dif­fuse alve­o­lar
of the eti­­ol­ogy and immune dysregulation may inform both new hem­or­rhage after pedi­at­ric hema­to­poi­etic cell trans­plant. Front Pediatr.
diag­nos­tic tools and bet­ter treat­ments to improve out­comes. 2020;8(13 Jan­u­ary):587601.

584  |  Hematology 2021  |  ASH Education Program


14. Chiang Y, Tsai CH, Kuo SH, et al. Reduced inci­dence of inter­sti­tial pneu­ 36. Gavriilaki E, Sakellari I, Gavriilaki M, Anagnostopoulos A. A new era in endo­
mo­ni­tis after allo­ge­neic hema­to­poi­etic stem cell trans­plan­ta­tion using a the­lial injury syn­dromes: tox­ic­ity of CAR-T cells and the role of immu­nity. Int
mod­i­fied tech­nique of total body irra­di­a­tion. Sci Rep. 2016;6:36730. J Mol Sci. 2020;21(11):3886.
15. Ahya VN. Noninfectious acute lung injury syn­dromes early after hema­to­ 37. Lee DW, Santomasso BD, Locke FL, et  al. ASTCT con­sen­sus grad­ing
poi­etic stem cell trans­plan­ta­tion. Clin Chest Med. 2017;38(4):595-606. for cyto­kine release syn­drome and neu­ro­logic tox­ic­ity asso­ci­ated with
16. Durie E, Nicholson E, Anthias C, et al. Determining the inci­dence of inter­ immune effec­tor cells. Biol Blood Marrow Transplant. 2019;25(4):625-638.
sti­tial pneu­mo­ni­tis and chronic kid­ney dis­ease fol­low­ing full inten­sity 38. Maus MV, Alexander S, Bishop MR, et  al. Society for Immunotherapy of
haemopoetic stem cell trans­plant con­di­tioned using a for­ward-planned Cancer (SITC) clin­i­cal prac­tice guide­line on immune effec­tor cell-related
inten­sity mod­u­lated total body irra­di­a­tion tech­nique. Radiother Oncol. adverse events. J Immunother Cancer. 2020;8(2):e001511.
2021;158(May):97-103. 39. Azoulay E, Castro P, Maamar A, et al; Nine-I Investigators. Outcomes in
17. Vande Vusse LK, Madtes DK. Early onset non­in­fec­tious pul­mo­nary syn­ patients treated with chi­me­ric anti­gen recep­tor T-cell ther­apy who were
dromes after hema­to­poi­etic cell trans­plan­ta­tion. Clin Chest Med. 2017;​ admit­ted to inten­sive care (CARTTAS): an inter­na­tional, multicentre, obser­
38(2):​233-248. va­tional cohort study. Lancet Haematol. 2021;8(5):e355–e364.
18. Wieruszewski PM, May HP, Peters SG, et al. Characteristics and out­come 40. Gazourian L, Spring L, Meserve E, et  al. Pulmonary clin­ i­
co­
path­
o­log­

cal
of peri-engraft­ment respi­ra­tory dis­tress syn­drome fol­low­ing autol­o­gous cor­re­la­tion after allo­ge­neic hema­to­poi­etic stem cell trans­plan­ta­tion: an
hema­to­poi­etic cell trans­plant. Ann Am Thorac Soc. 2020;18(6):1013-1019. autopsy series. Biol Blood Marrow Transplant. 2017;23(10):1767-1772.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/578/1851735/578williams.pdf by guest on 13 December 2021


19. Chang L, Frame D, Braun T, et al. Engraftment syn­drome after allo­ge­neic 41. Mandel J, Mark EJ, Hales CA. Pulmonary veno-occlu­ sive dis­ease. Am J
hema­to­poi­etic cell trans­plan­ta­tion pre­dicts poor out­comes. Biol Blood Respir Crit Care Med. 2000;162(5):1964-1973.
Marrow Transplant. 2014;20(9):1407-1417. 42. Bunte MC, Patnaik MM, Pritzker MR, Burns LJ. Pulmonary veno-occlu­sive
20. Shah NN, Watson TM, Yates B, et  al. Procalcitonin and cyto­kine pro­files dis­ease fol­low­ing hema­to­poi­etic stem cell trans­plan­ta­tion: a rare model of
in engraft­ment syn­drome in pedi­at­ric stem cell trans­plan­ta­tion. Pediatr endo­the­lial dys­func­tion. Bone Marrow Transplant. 2008;41(8):677-686.
Blood Cancer. 2017;64(3):e26273. 43. Gulbahce HE, Pambuccian SE, Jessurun J, et al. Pulmonary nod­u­lar lesions
21. Lucchini G, Willasch AM, Dan­iel J, et  al. Epidemiology, risk fac­tors, and in bone mar­row trans­plant recip­i­ents: impact of his­to­logic diag­no­sis on
prog­no­sis of cap­il­lary leak syn­drome in pedi­at­ric recip­i­ents of stem cell patient man­age­ment and prog­no­sis. Am J Clin Pathol. 2004;121(2):205–
trans­plants: a ret­ro­spec­tive sin­gle-cen­ter cohort study. Pediatr Transplant. 210.
2016;20(8):1132-1136. 44. Jodele S, Hirsch R, Laskin B, Davies S, Witte D, Chima R. Pulmonary arte­rial
22. Reikvam H, Grønningsæter IS, Ahmed AB, Hatfield K, Bruserud O. Metabolic hyper­ten­sion in pedi­at­ric patients with hema­to­poi­etic stem cell trans­plant-
serum pro­files for patients receiv­ing allo­ge­neic stem cell trans­plan­ta­tion: asso­ci­ated throm­botic microangiopathy. Biol Blood Marrow Transplant.
the pretransplant pro­file dif­fers for patients with and with­out posttrans­ 2013;19(2):202-207.
plant cap­il­lary leak syn­drome. Dis Markers. 2015;2015(Octo­ber):943430. 45. Nakamura Y, Mitani N, Ishii A, et al. Idiopathic pneu­mo­nia syn­drome with
23. Yanik G, Kitko C. Management of non­in­fec­tious lung injury fol­low­ing hema­ throm­botic microangiopathy-related changes after allo­ge­neic hema­to­poi­
to­poi­etic cell trans­plan­ta­tion. Curr Opin Oncol. 2013;25(2):187-194. etic stem cell trans­plan­ta­tion. Int J Hematol. 2013;98(4):496-498.
24. Clark JG, Hansen JA, Hertz MI, Parkman R, Jensen L, Peavy HH. NHLBI work­ 46. Bergeron A, Chevret S, Peffault de Latour R, et al. Noninfectious lung com­
shop sum­mary: idi­o­pathic pneu­mo­nia syn­drome after bone mar­row trans­ pli­ca­tions after allo­ge­neic haematopoietic stem cell trans­plan­ta­tion. Eur
plan­ta­tion. Am Rev Respir Dis. 1993;147(suppl 6, pt 1):1601-1606. Respir J. 2018;51(5).
25. Wenger DS, Triplette M, Crothers K, et al. Incidence, risk fac­tors, and out­ 47. Williams KM. How I treat bronchiolitis obliterans syn­drome after hema­to­
comes of idi­o­pathic pneu­mo­nia syn­drome after allo­ge­neic hema­to­poi­etic poi­etic stem cell trans­plan­ta­tion. Blood. 2017;129(4):448-455.
cell trans­plan­ta­tion. Biol Blood Marrow Transplant. 2020;26(2):413-420. 48. Williams KM, Chien JW, Gladwin MT, Pavletic SZ. Bronchiolitis obliterans
26. Yanik GA, Ho VT, Levine JE, et  al. The impact of sol­u­ble tumor necro­sis after allo­ge­neic hema­to­poi­etic stem cell trans­plan­ta­tion. JAMA. 2009;302(3):
fac­tor recep­tor etanercept on the treat­ment of idi­o­pathic pneu­mo­nia syn­ 306-314.
drome after allo­ge­neic hema­to­poi­etic stem cell trans­plan­ta­tion. Blood. 49. Yokoi T, Hirabayashi N, Ito M, et al; Nagoya Bone Marrow Transplant Group.
2008;112(8):3073-3081. Broncho-bronchiolitis obliterans as a com­pli­ca­tion of bone mar­row trans­
27. Versluys B, Bierings M, Murk JL, et  al. Infection with a respi­ra­tory virus plan­ta­tion: a clin­i­co­path­o­log­i­cal study of eight autopsy cases. Virchows
before hema­to­poi­etic cell trans­plan­ta­tion is asso­ci­ated with alloimmune- Arch. 1997;431(4):275-282.
medi­ated lung syn­dromes. J Allergy Clin Immunol. 2018;141(2):697-703. 50. Zhou X, O’Dwyer DN, Xia M, et al. First-onset herpesviral infec­tion and lung
e8703e8. injury in allo­ge­neic hema­to­poi­etic cell trans­plan­ta­tion. Am J Respir Crit
28. Sano H, Kobayashi R, Iguchi A, et al. Risk fac­tor anal­y­sis of idi­o­pathic pneu­ Care Med. 2019;200(1):63-74.
mo­nia syn­drome after allo­ge­neic hema­to­poi­etic SCT in chil­dren. Bone 51. Walther S, Rettinger E, Maurer HM, et  al. Long-term pul­mo­nary func­tion
Marrow Transplant. 2014;49(1):38-41. test­ing in pedi­at­ric bronchiolitis obliterans syn­drome after hema­to­poi­etic
29. Afessa B, Abdulai RM, Kremers WK, Hogan WJ, Litzow MR, Peters SG. Risk stem cell trans­plan­ta­tion. Pediatr Pulmonol. 2020;55(7):1725-1735.
fac­tors and out­come of pul­mo­nary com­pli­ca­tions after autol­o­gous hema­to­ 52. Jamani K, He Q , Liu Y, et al. Early post-trans­plan­ta­tion spi­rom­e­try is asso­ci­
poi­etic stem cell trans­plant. Chest. 2012;141(2):442-450. ated with the devel­op­ment of bronchiolitis obliterans syn­drome after allo­
30. Thompson J, Yin Z, D’Souza A, et al. Etanercept and cor­ti­co­ste­roid ther­apy ge­neic hema­to­poi­etic cell trans­plan­ta­tion. Biol Blood Marrow Transplant.
for the treat­ment of late-onset idi­o­pathic pneu­mo­nia syn­drome. Biol Blood 2020;26(5):943-948.
Marrow Transplant. 2017;23(11):1955-1960. 53. Yomota M, Yanagawa N, Sakai F, et  al. Association between chronic
31. Yanik GA, Horowitz MM, Weisdorf DJ, et  al. Randomized, dou­ ble-blind, bac­te­rial air­way infec­tion and prog­no­sis of bronchiolitis obliterans syn­
pla­cebo-con­trolled trial of sol­u­ble tumor necro­sis fac­tor recep­tor: enbrel drome after hema­to­poi­etic cell trans­plan­ta­tion. Medicine (Baltimore).
(etanercept) for the treat­ment of idi­op ­ athic pneu­mo­nia syn­drome after allo­ 2019;98(1):e13951.
ge­neic stem cell trans­plan­ta­tion: Blood and Marrow Transplant Clinical Trials 54. Turner J, He Q , Baker K, et al. Home spi­rom­e­try telemonitoring for early
Network pro­to­col. Biol Blood Marrow Transplant. 2014;20(6):858–864. detec­tion of bronchiolitis obliterans syn­drome in patients with chronic
32. Seo S, Yu J, Jenkins IC, et al. Diagnostic and prog­nos­tic plasma bio­mark­ers graft-ver­sus-host dis­ease. Transplant Cell Ther. 2021;27(7):616.e1-616.e6.
for idi­o­pathic pneu­mo­nia syn­drome after hema­to­poi­etic cell trans­plan­ta­ 55. Abedin S, Yanik GA, Braun T, et al. Predictive value of bronchiolitis obliter­
tion. Biol Blood Marrow Transplant. 2018;24(4):678-686. ans syn­drome stage 0p in chronic graft-ver­sus-host dis­ease of the lung.
33. Bhargava M, Viken KJ, Dey S, et al. Proteome pro­fil­ing in lung injury after Biol Blood Marrow Transplant. 2015;21(6):1127-1131.
hema­to­poi­etic stem cell trans­plan­ta­tion. Biol Blood Marrow Transplant. 56. Liu X, Yue Z, Yu J, et  al. Proteomic char­ac­ter­iza­tion reveals that MMP-
2016;22(8):1383-1390. 3 cor­re­lates with bronchiolitis obliterans syn­drome fol­low­ing allo­ge­
34. Varelias A, Gartlan KH, Kreijveld E, et al. Lung paren­chyma-derived IL-6 pro­ neic hema­to­poi­etic cell and lung trans­plan­ta­tion. Am J Transplant.
motes IL-17A-depen­dent acute lung injury after allo­ge­neic stem cell trans­ 2016;16(8):2342-2351.
plan­ta­tion. Blood. 2015;125(15):2435-2444. 57. Cheng GS, Selwa KE, Hatt C, et  al. Multicenter eval­u­a­tion of para­met­
35. Tizon R, Frey N, Heitjan DF, et al. High-dose cor­ti­co­ste­roids with or with­out ric response map­ ping as an indi­ ca­
tor of bronchiolitis obliterans syn­
etanercept for the treat­ment of idi­o­pathic pneu­mo­nia syn­drome after allo- drome after hema­to­poi­etic stem cell trans­plan­ta­tion. Am J Transplant.
SCT. Bone Marrow Transplant. 2012;47(10):1332-1337. 2020;20(8):2198-2205.

Prakash Singh Shekhawat


Noninfectious pul­mo­nary tox­ic­ity after HCT  |  585
58. Sharifi H, Lai YK, Guo H, et  al. Machine learn­ing algo­rithms to dif­fer­en­ti­ 74. Namkoong H, Ishii M, Mori T, et al. Clinical and radio­log­i­cal char­ac­ter­is­tics
ate among pul­mo­nary com­pli­ca­tions after hema­to­poi­etic cell trans­plant. of patients with late-onset severe restric­tive lung defect after hema­to­poi­
Chest. 2020;158(3):1090-1103. etic stem cell trans­plan­ta­tion. BMC Pulm Med. 2017;17(1):123.
59. Walkup LL, Myers K, El-Bietar J, et al. Xenon-129 MRI detects ven­ti­la­tion def­ 75. Yang JY, Oh SY, Song JW, et al. Restrictive chronic lung func­tion decline
i­cits in pae­di­at­ric stem cell trans­plant patients unable to per­form spi­rom­e­ after haematopoietic stem cell trans­plan­ta­tion. Eur Respir J. 2016;47(1):336–
try. Eur Respir J. 2019;53(5):1801779. 339.
60. Cuvelier GDE, Nemecek ER, Wahlstrom JT, et al. Benefits and chal­lenges 76. L’excel­lent S, Yakouben K, Delclaux C, Dalle JH, Houdouin V. Lung eval­ua ­­
with diag­nos­ing chronic and late acute GVHD in chil­dren using the NIH tion in 10 year sur­vi­vors of pedi­at­ric allo­ge­neic hema­to­poi­etic stem cell
con­sen­sus cri­te­ria. Blood. 2019;134(3):304-316. trans­plan­ta­tion. Eur J Pediatr. 2019;178(12):1833-1839.
61. Kwok WC, Liang BM, Lui MMS, et  al. Rapid ver­sus grad­ual lung func­tion 77. Yanik GA, Mineishi S, Levine JE, et al. Soluble tumor necro­sis fac­tor recep­
decline in bronchiolitis obliterans syn­drome after haematopoietic stem tor: enbrel (etanercept) for sub­acute pul­mo­nary dys­func­tion fol­low­ing
cell trans­plan­ta­tion is asso­ci­ated with sur­vival out­come. Respirology. allo­ge­neic stem cell trans­plan­ta­tion. Biol Blood Marrow Transplant. 2012;
2019;24(5):459-466. 18(7):1044-1054.
62. Williams KM, Cheng GS, Pusic I, et  al. Fluticasone, azithromycin, and 78. Blauwkamp TA, Thair S, Rosen MJ, et al. Analytical and clin­i­cal val­id ­ a­tion
montelukast treat­ment for new-onset bronchiolitis obliterans syn­drome of a micro­bial cell-free DNA sequenc­ing test for infec­tious dis­ease. Nat
after hema­to­poi­etic cell trans­plan­ta­tion. Biol Blood Marrow Transplant. Microbiol. 2019;4(4):663-674.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/578/1851735/578williams.pdf by guest on 13 December 2021


2016;22(4):710-716. 79. Gwinn M, MacCannell D, Armstrong GL. Next-gen­ er­

tion sequenc­ ing of
63. Penack O, Marchetti M, Ruutu T, et  al. Prophylaxis and man­age­ment of infec­tious path­o­gens. JAMA. 2019;321(9):893-894.
graft ver­sus host dis­ease after stem-cell trans­plan­ta­tion for haematolog­ 80. Armstrong AE, Rossoff J, Hollemon D, Hong DK, Muller WJ, Chaudhury
ical malig­nan­cies: updated con­sen­sus rec­om­men­da­tions of the Euro­ S. Cell-free DNA next-gen­er­a­tion sequenc­ing suc­cess­fully detects infec­
pean Society for Blood and Marrow Transplantation. Lancet Haematol. tious path­o­gens in pedi­at­ric oncol­ogy and hema­to­poi­etic stem cell trans­
2020;7(2):e157-e167. plant patients at risk for inva­sive fun­gal dis­ease. Pediatr Blood Cancer.
64. Bergeron A, Chevret S, Granata A, et al; ALLOZITHRO Study Investigators. 2019;66(7):e27734.
Effect of azithromycin on air­ flow decline-free sur­ vival after allo­
ge­ neic 81. Rossoff J, Chaudhury S, Soneji M, et al. Noninvasive diag­no­sis of infec­tion
hema­to­poi­etic stem cell trans­plant: the ALLOZITHRO ran­dom­ized clin­i­cal using plasma next-gen­er­a­tion sequenc­ing: a sin­gle-cen­ter expe­ri­ence.
trial. JAMA. 2017;318(6):557-566. Open Forum Infect Dis. 2019;6(8):ofz327.
65. Cheng GS, Bondeelle L, Gooley T, et al. Azithromycin use and increased 82. Seo S, Renaud C, Kuypers JM, et al. Idiopathic pneu­mo­nia syn­drome after
can­cer risk among patients with bronchiolitis obliterans after hema­to­poi­ hema­to­poi­etic cell trans­plan­ta­tion: evi­dence of occult infec­tious eti­­ol­o­
etic cell trans­plan­ta­tion. Biol Blood Marrow Transplant. 2020;26(2):392– gies. Blood. 2015;125(24):3789-3797.
400. 83. Zinter MS, Hume JR. Effects of hema­to­poi­etic cell trans­plan­ta­tion on the
66. Hefazi M, Langer KJ, Khera N, et  al. Extracorporeal photopheresis pul­mo­nary immune response to infec­tion. Front Pediatr. 2021;9(26 Jan­u­
improves sur­vival in hema­to­poi­etic cell trans­plant patients with bron­ ary):634566.
chiolitis obliterans syn­drome with­out sig­nif­i­cantly impacting mea­sured 84. Panoskaltsis-Mortari A, Griese M, Madtes DK, et al; Amer­i­can Thoracic
pul­mo­nary func­tions. Biol Blood Marrow Transplant. 2018;24(9):1906- Society Committee on Idiopathic Pneumonia Syndrome. An offi­cial Amer­
1913. i­can Thoracic Society research state­ment: non­in­fec­tious lung injury after
67. Bondeelle L, Chevret S, Hurabielle C, et al. Effect of ruxolitinib on lung func­ hema­to­poi­etic stem cell trans­plan­ta­tion: idi­o­pathic pneu­mo­nia syn­
tion after allo­ge­neic stem cell trans­plan­ta­tion. Biol Blood Marrow Trans- drome. Am J Respir Crit Care Med. 2011;183(9):1262-1279.
plant. 2020;26(11):2115-2120. 85. Klein OR, Cooke KR. Idiopathic pneu­mo­nia syn­drome fol­low­ing hema­to­
68. Cheng GS, Storer B, Chien JW, et al. Lung func­tion tra­jec­tory in bronchi­ poi­etic stem cell trans­plan­ta­tion. J Pediatr Intensive Care. 2014;3(3):147–
olitis obliterans syn­drome after allo­ge­neic hema­to­poi­etic cell trans­plant. 157.
Ann Am Thorac Soc. 2016;13(11):1932-1939. 86. Pagliuca S, Michonneau D, Sicre de Fontbrune F, et al. Allogeneic reac­tiv­ity-
69. Kliman DS, Kotecha SR, Abelson DC, Snell GI, Glanville AR, Ma DDF. Favor­ medi­ated endo­the­lial cell com­pli­ca­tions after HSCT: a plea for con­sen­sual
able out­ come of lung trans­ plan­ta­
tion for severe pul­ mo­nary graft ver­ def­i­ni­tions. Blood Adv. 2019;3(15):2424-2435.
sus host dis­ease: an Aus­tra­lian mul­ti­cen­ter case series. Transplantation. 87. Chellapandian D, Lehrnbecher T, Phil­lips B, et al. Bronchoalveolar lavage
2019;103(12):2602-2607. and lung biopsy in patients with can­cer and hema­to­poi­etic stem-cell trans­
70. Verleden SE, McDonough JE, Schoemans H, et  al. Phenotypical diver­sity plan­ta­tion recip­i­ents: a sys­tem­atic review and meta-anal­y­sis. J Clin Oncol.
of air­way mor­phol­ogy in chronic lung graft vs. host dis­ease after stem cell 2015;33(5):501-509.
trans­plan­ta­tion. Mod Pathol. 2019;32(6):817-829. 88. Williams KM, Inamoto Y, Im A, et al. National Institutes of Health con­sen­sus
71. Takeuchi Y, Miyagawa-Hayashino A, Chen F, et  al. Pleuroparenchymal devel­op­ment pro­ject on cri­te­ria for clin­i­cal tri­als in chronic graft-ver­sus-
fibroelastosis and non-spe­cific inter­sti­tial pneu­mo­nia: fre­quent pul­mo­nary host dis­ease, I: the 2020 Etiology and Prevention Working Group report.
sequelae of haematopoietic stem cell trans­ plan­
ta­
tion. Histopathology. Transplant Cell Ther. 2021;27(6):452-466.
2015;66(4):536-544. 89. Kitko CL, Pidala J, Schoemans HM, et  al. National Institutes of Health
72. Higo H, Miyahara N, Taniguchi A, Maeda Y, Kiura K. Cause of pleuroparen­ con­sen­sus devel­op­ment pro­ject on cri­te­ria for clin­i­cal tri­als in chronic
chymal fibroelastosis fol­low­ing allo­ge­neic hema­to­poi­etic stem cell trans­ graft-ver­sus-host dis­ease, IIa: the 2020 Clinical Implementation and Early
plan­ta­tion. Respir Investig. 2019;57(4):321-324. Diagnosis Working Group report. Transplant Cell Ther. 2021;27(7):545-557.
73. Madanat-Harjuoja LM, Valjento S, Vettenranta K, Kajosaari M, Dyba T, Taski­
nen M. Pulmonary func­tion fol­low­ing allo­ge­neic stem cell trans­plan­ta­tion in
child­hood: a ret­ro­spec­tive cohort study of 51 patients. Pediatr Transplant. © 2021 by The Amer­i­can Society of Hematology
2014;18(6):617-624. DOI 10.1182/hema­tol­ogy.2021000293

586  |  Hematology 2021  |  ASH Education Program


SURVIVORSHIP AFTER ALLOGENEIC HEMATOPOIETIC CELL TRANSPLANT

Infectious complications and vaccines


Per Ljungman

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/587/1851732/587ljungman.pdf by guest on 13 December 2021


Division of Hematology, Department of Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden; and Department of Cellular Therapy and
Allogeneic Stem Cell Transplantation, Karolinska Comprehensive Cancer Center, Karolinska University Hospital, Huddinge, Stockholm, Sweden

Infections are a major cause of morbidity and can result in mortality in long-term survivors after allogeneic hematopoietic
cell transplantation. Chronic graft-versus-host disease and delayed immune reconstitution are recognized risk factors.
Different strategies must be utilized depending on the individual patient’s situation but include prolonged antimicrobial
prophylaxis and vaccination. Some important infections due to pathogens preventable by vaccination are pneumococci,
influenza, varicella-zoster virus, and SARS-CoV-2. Despite the fact that such recommendations have been in place for
decades, implementation of these recommendations has been reported to be poor.

LEARNING OBJECTIVES
• Learn about important pathogens causing disease in allogeneic HCT survivors
• Increase awareness of the benefits and risks of vaccination of allogeneic HCT survivors

Introduction to the emergency room with a high fever that had lasted
Infections are major causes of morbidity and can result in 12 hours. He had a history of moderate chronic GVHD and
mortality in long-term survivors (more than 1 year) after was still on tacrolimus. His wife reported that during the
allogeneic hematopoietic stem cell transplantation (HCT). last hour at home he had become lethargic. At examination
Table 1 shows some important pathogens in this population. he was unconscious, was febrile, and had a blood pressure
The risk is increased with active chronic graft-versus-host of 79/50. He was admitted. C-reactive protein was 150 and
disease (GVHD) due to both incomplete immune reconsti- the white blood cell count was 11.5. A computed-tomogra-
tution and ongoing immunosuppressive therapy. Although phy scan was performed but showed no pathology. Blood
the risk for nonrelapse mortality has decreased over time,1 cultures were taken and a lumbar puncture performed. The
in a retrospective analysis infections represented 20.7% of latter showed a cell count of 70, prominently consisting of
all deaths. Furthermore, infection was the second most fre- neutrophils. A gram stain showed gram-positive cocci, and
quent cause of death in patients still alive 1 year after HCT.2 he was started on ceftriaxone.
Two pandemics during the last 12 years have illustrated the
vulnerability of this population. The 2009 H1N1 influenza
“swine flu” pandemic and the recent COVID-19 pandemic Bacterial infections
both caused significant mortality in the allogeneic-HCT Bacterial infections are the most common type of infection
population in long-term survivors. It is to be expected that causing mortality in both 1- and 5-year survivors after allo-
new infections will appear from time to time and that those geneic HCT.2 The cause of infections occurring late after
who have undergone allogeneic HCT might be more vul- transplant is frequently not identified, especially if occur-
nerable than the general population. Therefore, awareness ring away from transplant centers, but bacterial infections
and preventive measures are indicated. likely represent many of these “unknown” infections. The
incidence of invasive pneumococcal disease (IPD) after
allogeneic HCT has been reported to be 80 times that in a
healthy population,3 can occur many years after allogeneic
HCT, and has a high mortality. The risk is increased in pa-
CLINICAL CASE tients with chronic GVHD. Vaccination is an effective way
A 63-year-old man who had undergone an allogeneic of reducing IPD in both young children and older adults.
unrelated-donor transplantation 3 years previously came Other vaccine-preventable bacterial infections that might
Prakash Singh Shekhawat
Infectious complications and vaccines | 587
Table 1.  Some impor­tant path­o­gens in allo­ge­neic HCT the sec­ond in 5-year sur­vi­vors after allo­ge­neic HCT.2 Late infec­
sur­vi­vors tions occur with Pneumocystis jirovecii, espe­cially in patients with
delayed immune recon­sti­tu­tion, but can also occur with other
Bacteria Pneumococci fungi, par­tic­ul­arly in the set­tings of relapse of the under­ly­ing dis­
Hib ease or severe chronic GVHD. Pneumocystis pro­phy­laxis is there­
fore indi­cated. Ibrutinib has been asso­ci­ated with an increased
Meningococci
risk of inva­sive fun­gal infec­tion, and a recent small case series of
Viruses Influenza A and B patients treated for chronic GVHD illus­trates this risk.9 Ruxolitinib
SARS-CoV-2 has also been asso­ci­ated with inva­sive fun­gal infec­tions. Systemic
anti­fun­gal pro­phy­laxis should be con­sid­ered in patients with se-
Respiratory syncytial virus
vere chronic GVHD receiv­ing inten­sive immu­no­sup­pres­sion.
Varicella-zoster virus
Hepatitis B, C, and E

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/587/1851732/587ljungman.pdf by guest on 13 December 2021


Fungi Pneumocystis jirovecii
CLINICAL CASE (Con­tin­ued)
Endemic myco­ses
The patient was treated with ceftriaxone and improved; how­
ever, he had devel­oped hear­ing loss and some addi­tional neu­
cause severe late infec­tions are Haemophilus influenzae type ro­log­i­cal def­i­cit. Blood and CSF cul­tures showed growth of
B (Hib) and menin­go­cocci. Multiresistant bac­te­ria can col­o­nize Streptococcus pneumoniae. Questioning his wife pro­vided the
allo­ge­neic HCT patients, and it has been reported that patients infor­ma­tion that his phy­si­cians had recommended vac­ci­na­tion
becom­ing long-term car­ri­ers have an increased risk for late mor­ against pneu­mo­cocci after the HCT, but it had never been per-
tal­ity.4 Some cen­ters give anti­bac­te­rial pro­phy­laxis pri­mar­ily formed.
targeting pneu­mo­cocci to patients with chronic GVHD. No con­
trolled data exist regard­ing this approach, and what agent to
use and its effi­cacy is likely to depend on the anti­bac­te­rial resis­ Vaccines
tance sit­u­a­tion of pneu­mo­cocci in the com­mu­nity. Table 2 shows cur­rent rec­om­men­da­tions for vac­ci­na­tion of HCT
recip­i­ents. These can be broadly divided into com­mon vac­cine-
Viral infec­tions pre­vent­able infec­tions caus­ing severe dis­ease after allo­ge­neic
Viral infec­tions have been reported to be the sec­ond most com­ HCT and vac­cines against rare but severe infec­tions when a broad
mon cause of infec­tious dis­ease mor­tal­ity in long-term sur­vi­vors.2 immu­nity in the pop­u­la­tion is desir­able. Examples of the for­mer
Community-acquired respi­ra­tory virus infec­tions are impor­tant are pneu­mo­coc­cal infec­tions, influ­enza, and COVID-19, while
causes of severe infec­tion occur­ring years after an allo­ge­neic exam­ples of the lat­ter are tet­a­nus and diph­the­ria. In addi­tion,
HCT, espe­cially in patients with delayed immune recon­sti­tu­tion. travel vac­cines are indi­cated for trans­plant recip­i­ents want­ing to
The most impor­tant of these infec­tions until recently have been travel to areas where they might encoun­ter region­ally com­mon
caused by influ­enza A and B, para­in­flu­enza virus, and respi­ra­ impor­tant path­og ­ ens, such as yel­low fever or Jap­a­nese enceph­
tory syn­cy­tial virus. However, COVID-19 emerged dur­ing the last a­li­tis. This review discusses some exam­ples of the 3 sit­u­a­tions.
year and has become a major cause of late mor­tal­ity after HCT.5,6 An impor­tant aspect of vac­ci­nat­ing allo­ge­neic HCT recip­i­ents
Ljungman et al reported from the European Society for Blood is safety. Currently avail­­able data with nonlive vac­cines sup­port
and Marrow Transplantation reg­is­try a mor­tal­ity of 28.7% in pa- a high level of safety with side effects sim­i­lar to those in a nor­mal
tients diag­nosed with COVID-19 at 1 to 2 years after allo­ge­neic pop­u­la­tion. Knowledge about risks with new vac­cine plat­forms
HCT and 20.9% in patients at >2 years after the pro­ce­dure.6 Shar- such as those used in the recently licensed COVID-19 vac­cines
ma et al reported in a Center for International Blood and Marrow is less and will require con­tin­ued and care­ful sur­veil­lance. Live
Transplant Research anal­y­sis a mor­tal­ity of 15.6% in patients with atten­u­ated vac­cines can cause vac­cine-induced dis­ease, which
COVID-19 more than 1 year after HCT.5 It is likely, how­ever, that can be fatal in severely immunosuppressed indi­vid­ua ­ ls. Careful
both these stud­ies have a selec­tion bias in that milder infec­tions weighing of risk/ben­e­fit is there­fore needed.
in patients with­out comorbidities have been undi­ag­nosed. Although inter­na­tional rec­om­men­da­tions regard­ing vac­ci­
Other impor­ tant viral infec­ tions in long-term sur­ vi­
vors are na­tion have existed for sev­eral years,10,11 sev­eral reports state
reactivated var­i­cella-zoster virus (VZV) and chronic hep­a­ti­tis virus that the implementation is poor, as illus­trated by the clin­i­cal
infec­tions. Reactivated VZV infec­tion can be severe even dur­ing case.12-14 The rea­sons vary, includ­ing com­pli­ance, unclear respon­
appro­pri­ate anti­vi­ral pro­phy­laxis, espe­cially if the patient is still si­bil­i­
ties, and reluc­tance to immu­ nize patients with GVHD or
receiv­ing immu­no­sup­pres­sion.7 Patients infected with hep­a­ti­tis B ongo­ing immu­no­sup­pres­sion. Data clearly sug­gest, how­ever,
virus or who have infected donors should receive pro­phy­laxis a that patients with GVHD should be vac­ci­nated with nonlive vac­
least dur­ing the time of ongo­ing immu­no­sup­pres­sion.8 Hepati- cines, although it must be rec­og­nized that the immune response
tis E virus has emerged as a poten­tial cause of liver cir­rho­sis in might be lower.
patients on chronic immu­no­sup­pres­sion, such as solid-organ and
allo­ge­neic HCT trans­plant recip­i­ents. Further stud­ies are needed. Pneumococcal vac­cines
Pneumococcal poly­sac­cha­ride vac­cine is a poor inducer of an
Fungal infec­tions immune response, espe­cially in patients with chronic GVHD. On
Invasive fun­gal infec­tions were reported to be the third most fre­ the other hand, stud­ies with the pneu­mo­coc­cal con­ju­gate vac­
quent cause of infec­tious dis­ease mor­tal­ity in 1-year sur­vi­vors but cines (PCV) showed that they could induce strong and dura­ble

588  |  Hematology 2021  |  ASH Education Program


Table 2.  Recommended vac­cines after allo­ge­neic HCT

Vaccine Recommendation Comments


Nonlive vac­cines
Tetanus tox­oid + diph­the­ria tox­oid Yes Three doses (DT) starting 6 mo after trans­plan­ta­tion
Inactive influ­enza Yes Seasonal, begin­ning 4-6 mo after trans­plan­ta­tion depending on sea­son
Inactivated polio­vi­rus Yes Three doses starting 6 (-12) mo after trans­plan­ta­tion
Conjugated Hib Yes Three doses starting 6 (-12) mo after trans­plan­ta­tion
Pneumococcal con­ju­gate Yes Three doses starting 3 (-6) mo after trans­plan­ta­tion; booster at 12 mo in patients
with chronic GVHD
Pneumococcal poly­sac­cha­ride Yes Booster at 12 mo in patients with­out GVHD no ear­lier than 8 weeks after

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/587/1851732/587ljungman.pdf by guest on 13 December 2021


con­ju­gate
Acellular per­tus­sis Yes Children <7 starting 6 (-12) mo after trans­plan­ta­tion
Hepatitis B virus Yes In countries where it is recommended to the gen­eral pop­u­la­tion, starting 6 (-12)
mo after trans­plan­ta­tion
Papillomavirus Yes As in the gen­eral pop­u­la­tion, starting ear­li­est 6-12 mo after trans­plan­ta­tion;
three doses
Meningococcal con­ju­gate Yes As in the gen­eral pop­u­la­tion, starting 6 mo after trans­plan­ta­tion; two doses
Recombinant zoster vac­cine Can be con­sid­ered Limited data after stem cell trans­plan­ta­tion
Vaccines against COVID-19 Yes Limited data but risk/ben­e­fit favors vac­ci­na­tion
Live vac­cines
MMR Individual con­sid­er­ation Children and sero­neg­a­tive adults, not before 24 mo after HCT; not to be given
to patients with GVHD
Varicella Individual con­sid­er­ation Seronegative patients, not before 24 mo after BMT; not to be given in patients
with GVHD
Live zoster Not recommended
DT, diph­the­ria and tet­a­nus tox­oids; MMR, mea­sles, mumps, and rubella. Adapted from Cordonnier et al.10

immune responses. The cur­rent rec­om­men­da­tions are to start after HCT but could be ini­ti­ated as early as 4 months after HCT
vac­ci­na­tion with 3 doses of PCV at 3 to 4 months after HCT fol- dur­ing a com­mu­nity out­break. Children 6 months to 8 years of
lowed by either a pneu­mo­coc­cal poly­sac­cha­ride vac­cine V23 age should be given a sec­ond dose. The live atten­u­ated influ­
dose in patients with­out or a fourth PCV dose in patients with enza vac­cine should not be used in HCT recip­i­ents. Family mem­
chronic GVHD.10,11 The intro­duc­tion of PCV in the man­age­ment bers and hos­pi­tal staff should receive influ­enza vac­cine, thereby
strat­egy has reduced the risk of IPD after allo­ge­neic HCT.15 The reduc­ing the risk for trans­mis­sion.10,11
long-term retainment of antibodies against pneu­ mo­cocci has Several stud­ ies have shown poorer immune sero­ log­i­
cal
been stud­ ied after vary­ing vac­cine sched­ ules. However, the responses after vac­ci­na­tion in allo­ge­neic HCT recip­i­ents, both
major­ity received at least 3 PCV doses, as cur­rently recommend- adults and chil­ dren, com­ pared to healthy con­ trols. The time
ed. Fifty per­cent of patients were protected against all 7 ana­lyzed after trans­plan­ta­tion is the most impor­tant fac­tor for vac­cine
sero­types at a median of 9.3 years after trans­plan­ta­tion while 70% effi­cacy, with patients vac­ci­nated later responding bet­ter. Two-
were protected against 5 of 5 sero­types.16 A lack of pro­tec­tive an- dose vac­cine sched­ules and the use of adjuvanted vac­cines have
tibodies was asso­ci­ated with chronic GVHD, relapse of under­ly­ been stud­ied with vary­ing results.
ing malig­nancy, and cord blood trans­plan­ta­tion. This shows that Despite sub­op­ti­mal sero­log­i­cal responses, there might be
seroprotection ought to be reg­u­larly assessed to define the need clin­i­cal effec­tive­ness of vac­ci­na­tion since pro­tec­tive anti­body
for booster doses. New con­ju­gated vac­cines cov­er­ing addi­tional lev­els against severe influ­enza dis­ease are poorly defined. Kumar
sero­types are in devel­op­ment. et al showed in a pro­spec­tive cohort study that influ­enza vac­
ci­na­tion dur­ing the rel­e­vant sea­son reduced the risk of severe
Hib dis­eases such as pneu­mo­nia and the need for admis­sion to an
Immunization against Hib is recommended after HCT. However, inten­sive care unit.17 Piñana et al showed in a sim­i­larly designed
nontypable Haemophilus influenzae strains has become more cohort study that influ­enza vac­ci­na­tion reduced the risk of lower-
com­mon, and there is cur­rently no vac­cine against these strains. respi­ra­tory tract influ­enza and hos­pi­tal admis­sions.18 Thus, there
is a sub­stan­tial clin­i­cal ben­e­fit to reg­u­lar influ­enza vac­ci­na­tion of
Influenza vac­cine HCT recip­i­ents.
Influenza A and B infec­tions can be severe and life-threat­en­ing,
and fatal infec­tions can occur sev­eral years after HCT. Therefore, COVID-19 vac­cines
yearly influ­enza vac­ci­na­tion with the inactivated vac­cine is rec- COVID-19 is asso­ci­ated with sub­stan­tial mor­bid­ity and mor­tal­ity
ommended for all­allo­ge­neic HCT recip­i­ents starting at 6 months after allo­ge­neic HCT. Thus, vac­ci­na­tion is indi­cated but must be
Prakash Singh Shekhawat
Infectious com­pli­ca­tions and vac­cines  | 589
eval­u­ated regard­ing the risk of pos­si­bly induc­ing GVHD or other effect of vac­ci­na­tion varies between dif­fer­ent stud­ies, with a
immune acti­va­tion phe­nom­ena. Currently, no infor­ma­tion is avail­­ seem­ingly higher response rate in adults than in chil­dren.
able addressing these issues in allo­ge­neic HCT recip­i­ents. Data
in patients with hema­to­log­i­cal malig­nan­cies, espe­cially chronic Travel vac­cines
lymphocytic leukemia and mul­ ti­
ple mye­ loma, and after solid- Vaccines are avail­­able for HCT patients liv­ing in cer­tain areas
organ trans­plan­ta­tion sug­gest lower rates of response than in of the world or trav­el­ing to areas where cer­tain infec­tions are
healthy con­trols.19-24 It is cur­rently unclear how to inter­pret the endemic. Nonlive vac­ cines include those against tick-borne
results of anti­body assays in severely immu­no­com­pro­mised indi­vid­ enceph­a­li­tis32,33 and Jap­a­nese enceph­a­li­tis.34 The response to
u­als such as HCT patients regard­ing pro­tec­tion against COVID-19. hep­a­ti­tis A vac­cine was shown to be poor in both pre­vi­ously
Thus, the con­tin­ued use of pro­tec­tive mea­sures is recommended. sero­neg­a­tive and sero­pos­i­tive indi­vid­u­als.35 Yellow fever is en-
demic in some areas of the world, and the only vac­cine is live
atten­u­ated. In a ret­ro­spec­tive French mul­ti­cen­ter study, 21 pa-
HPV vac­cine
tients who received yel­low fever vac­ci­na­tion a median of 39
HCT patients are prone to develop pap­il­lo­ma­vi­rus-driven com­

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/587/1851732/587ljungman.pdf by guest on 13 December 2021


months after HCT dem­on­strated a high response rate with­out
pli­ca­tions such as cer­vi­cal dys­pla­sia. Stratton et al showed that
documented side effects.36 For patients who live in or must vis-
strong immune responses can be elicited by the quad­ri­va­lent
it areas where yel­low fever is endemic, immu­ni­za­tion could be
human pap­il­lo­ma­vi­rus (HPV) vac­cine with­out sig­nif­i­cant side ef-
con­sid­ered at least 2 years after HCT and if the patient is with­out
fects.25 It would be log­i­cal to vac­ci­nate HPV-sero­neg­a­tive indi­
GVHD or ongo­ing immu­no­sup­pres­sion.
vid­u­als to pre­vent acqui­si­tion of HPV, but whether vac­ci­na­tion of
HCT recip­i­ents already infected with HPV would result in a clin­
i­cal ben­efi
­ t is unknown. This could be a topic for future stud­ies. Conflict-of-inter­est dis­clo­sure
Per Ljungman: research funding, con­sul­tancy, lec­tur­ing: Pfizer.

Varicella vac­cine Off-label drug use


Primary var­i­cella can be severe after HCT. The existing vac­cine Per Ljungman: nothing to disclose.
is live and atten­ua ­ ted and should not be used soon after HCT. A
sero­neg­a­tive patient should, if pos­si­ble, be immu­nized before Correspondence
trans­plan­ta­tion, pro­vid­ing that enough time can elapse after Per Ljungman, Department of Cellular Therapy and Allogeneic
the vac­ci­na­tion. Vaccination of sero­neg­a­tive fam­ily mem­bers is Stem Cell Transplantation, M75, Karolinska University H­ ospital,
recommended. A few uncon­trolled stud­ies have reported that Hud­ dinge, SE-14186 Stockholm, Sweden; e-mail: per​­
.ljungman
var­i­cella vac­ci­na­tion is safe and can result in sero­con­ver­sion @ki​­.se.
if performed more than 2 years after HCT in patients with­out
chronic GVHD or ongo­ing immu­no­sup­pres­sion. References
A high pro­por­tion of HCT patients develop her­pes zoster that 1. Penack O, Peczynski C, Mohty M, et  al. How much has allo­ge­neic stem
occa­sion­ally becomes severe. Both live and nonlive recom­bi­nant cell trans­plant-related mor­tal­ity improved since the 1980s? A ret­ro­spec­tive
anal­y­sis from the EBMT. Blood Adv. 2020;4(24):6283-6290.
vac­cines exist. Issa et al gave 1 dose of live atten­u­ated zoster
2. Styczyński J, Tridello G, Koster L, et al; Infectious Diseases Working Party.
vac­cine to 58 allo­ge­neic HCT recip­i­ents and noted no sig­nif­i­cant Death after hema­to­poi­etic stem cell trans­plan­ta­tion: changes over cal­en­
side effects.26 However, fatal infec­tions have been documented dar year time, infec­tions and asso­ci­ated fac­tors. Bone Marrow Transplant.
after autol­o­gous HCT. Due to the proven effi­cacy and safety of 2020;55(1):126-136.
acy­clo­vir pro­phy­laxis, vac­ci­na­tion with the live zoster vac­cine is 3. van Aalst M, Lötsch F, Spijker R, et al. Incidence of inva­sive pneu­mo­coc­cal
dis­ease in immu­no­com­pro­mised patients: a sys­tem­atic review and meta-
not recommended dur­ing at least the first few years after HCT. anal­y­sis. Travel Med Infect Dis. 2018;24(July-August):89-100.
The recently licensed recom­ bi­
nant zoster vac­ cine showed 4. Heidenreich D, Kreil S, Jawhar M, et  al. Course of col­on ­ i­za­tion by mul­ti­
high immu­no­ge­nic­ity and decreased the rate of her­pes zoster in drug-resis­tant organ­isms after allo­ge­neic hema­to­poi­etic cell trans­plan­ta­
autol­o­gous HCT recip­i­ents.27 It has also been stud­ied in allo­ge­neic tion. Ann Hematol. 2018;97(12):2501-2508.
5. Sharma A, Bhatt NS, St Martin A, et al. Clinical char­ac­ter­is­tics and out­comes
HCT recip­i­ents but not in a con­trolled trial. Camargo et al dem­
of COVID-19 in haematopoietic stem-cell trans­ plan­ta­tion recip­ i­
ents: an
on­strated that 2 doses of recombinant zoster vaccine were less obser­va­tional cohort study. Lancet Haematol. 2021;8(3):e185-e193.
immu­no­genic in allo­ge­neic HCT recip­i­ents com­pared to autol­o­ 6. Ljungman P, de la Camara R, Mikulska M, et  al. COVID-19 and stem cell
gous recip­i­ents and that VZV reactivations occurred.28 Baumrin et trans­plan­ta­tion: results from an EBMT and GETH mul­ti­cen­ter pro­spec­tive
al showed that a 2-dose reg­i­men was safe and tol­er­a­ble, with high sur­vey. Leukemia. 2021:1-10. Published online 2 June 2021.
7. Baumrin E, Cheng MP, Kanjilal S, Ho VT, Issa NC, Baden LR. Severe her­pes
rates of mild to mod­er­ate local and sys­temic side effects but with­ zoster requir­ing intra­ve­nous anti­vi­ral treat­ment in allo­ge­neic hema­to­poi­
out increas­ing the rates of GVHD; how­ever, VZV reactivations were etic cell trans­plan­ta­tion recip­i­ents on stan­dard acy­clo­vir pro­phy­laxis. Biol
seen after vac­ci­na­tion.29 Additional stud­ies are there­fore needed. Blood Marrow Transplant. 2019;25(8):1642-1647.
8. Mallet V, van Bömmel F, Doerig C, et al; 5th Euro­pean Conference on Infec-
tions in Leukaemia. Management of viral hep­a­ti­tis in patients with haema-
Measles vac­cine
tological malig­nancy and in patients under­go­ing haemopoietic stem cell
The major­ity of allo­ge­neic HCT patients will become sero­neg­ trans­plan­ta­tion: rec­om­men­da­tions of the 5th Euro­pean Conference on
a­tive to mea­sles dur­ing extended fol­low-up. There are docu- Infections in Leukaemia (ECIL-5). Lancet Infect Dis. 2016;16(5):606-617.
mented cases of fatal mea­sles in BMT recip­i­ents.30,31 The loss of 9. Kaloyannidis P, Ayyad A, Bahaliwah Z, et al. Ibrutinib for ste­roid refrac­tory
immu­nity is more rapid in patients pre­vi­ously vac­ci­nated against chronic graft-ver­sus-host dis­ease: ther­a­peu­tic effi­ciency can be lim­ited
by increased risk of fun­gal infec­tion. Bone Marrow Transplant. 2021;56(8):​
mea­sles com­pared to those hav­ing expe­ri­enced nat­u­ral infec­ 2034-2037.
tion. Vaccination can only be con­sid­ered in a patient with­out 10. Cordonnier C, Einarsdottir S, Cesaro S, et al; Euro­pean Conference on Infec-
chronic GVHD or ongo­ing immu­no­sup­pres­sion. The reported tions in Leukaemia Group. Vaccination of haemopoietic stem cell trans­plant

590  |  Hematology 2021  |  ASH Education Program


recip­i­ents: guide­lines of the 2017 Euro­pean Conference on Infections in Leu- 24. Boyarsky BJ, Chiang TP, Ou MT, et  al. Antibody response to the Janssen
kaemia (ECIL 7). Lancet Infect Dis. 2019;19(6):e200-e212. COVID-19 vac­cine in solid organ trans­plant recip­i­ents. Transplantation.
11. Rubin LG, Levin MJ, Ljungman P, et al; Infectious Diseases Society of Amer- 2021;105(8):e82-e83.
ica. 2013 IDSA clin­i­cal prac­tice guide­line for vac­ci­na­tion of the immu­no­ 25. Stratton P, Battiwalla M, Tian X, et  al. Immune response fol­low­ing quad­
com­pro­mised host. Clin Infect Dis. 2014;58(3):e44-100. ri­va­lent human pap­il­lo­ma­vi­rus vac­ci­na­tion in women after hema­to­poi­etic
12. Dyer G, Gilroy N, Brice L, et al. A sur­vey of infec­tious dis­eases and vac­ci­ allo­ge­neic stem cell trans­plant: a nonrandomized clin­i­cal trial. JAMA Oncol.
na­tion uptake in long-term hema­to­poi­etic stem cell trans­plant sur­vi­vors in 2020;6(5):696-705.
Australia. Transpl Infect Dis. 2019;21(2):e13043. 26. Issa NC, Marty FM, Leblebjian H, et al. Live atten­u­ated var­i­cella-zoster vac­
13. Ariza-Heredia EJ, Gulbis AM, Stolar KR, et al. Vaccination guide­lines after cine in hema­to­poi­etic stem cell trans­plan­ta­tion recip­i­ents. Biol Blood Mar-
hema­to­poi­etic stem cell trans­plan­ta­tion: prac­ti­tion­ers’ knowl­edge, row Transplant. 2014;20(2):285-287.
­atti­tudes, and gap between guide­lines and clin­i­cal prac­tice. Transpl Infect 27. Bastidas A, de la Serna J, El Idrissi M, et al; Zoster Efficacy Study in Patients
Dis. 2014;​16(6):878-886. Undergoing HCT Study Group Collaborators. Effect of recom­bi­nant zoster
14. Gouveia-Alves F, Gouveia R, Ginani VC, et  al. Adherence and immune vac­cine on inci­dence of her­pes zoster after autol­o­gous stem cell trans­plan­
response to revaccination fol­low­ing hema­to­poi­etic stem cell trans­plan­ta­ ta­tion: a ran­dom­ized clin­i­cal trial. JAMA. 2019;322(2):123-133.
tion at a pedi­at­ric onco-hema­tol­ogy ref­er­ence cen­ter. Transpl Infect Dis. 28. Camargo JF, Lin RY, Natori Y, et al. Reduced immu­no­ge­nic­ity of the adju-
2018;20(4):e12903. vanted recom­bi­nant zoster vac­cine after hema­to­poi­etic cell trans­plant: a

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/587/1851732/587ljungman.pdf by guest on 13 December 2021


15. Roberts MB, Bak N, Wee LYA, et al. Clinical effec­tive­ness of con­ju­gate pneu­ pilot study. Blood Adv. 2020;4(19):4618-4622.
mo­coc­cal vac­ci­na­tion in hema­to­poi­etic stem cell trans­plan­ta­tion recip­i­ 29. Baumrin E, Izaguirre NE, Bausk B, et  al. Safety and reactogenicity of the
ents. Biol Blood Marrow Transplant. 2020;26(2):421-427. recom­bi­nant zoster vac­cine after allo­ge­neic hema­to­poi­etic cell trans­plan­ta­
16. Robin C, Bahuaud M, Redjoul R, et al. Antipneumococcal seroprotection tion. Blood Adv. 2021;5(6):1585-1593.
years after vac­ci­na­tion in allo­ge­neic hema­to­poi­etic cell trans­plant recip­i­ 30. Kaplan LJ, Daum RS, Smaron M, McCarthy CA. Severe mea­sles in immu­no­
ents. Clin Infect Dis. 2020;71(8):e301-e307. com­pro­mised patients. JAMA. 1992;267(9):1237-1241.
17. Kumar D, Ferreira VH, Blumberg E, et  al. A 5-year pro­spec­tive mul­ti­cen­ 31. Nakano T, Shimono Y, Sugiyama K, et  al. Clinical fea­tures of mea­sles in
ter eval­u­a­tion of influ­enza infec­tion in trans­plant recip­i­ents. Clin Infect Dis. immu­no­com­pro­mised chil­dren. Acta Paediatr Jpn. 1996;38(3):212-217.
2018;67(9):1322-1329. 32. Harrison N, Grabmeier-Pfistershammer K, Graf A, et  al. Humoral immune
18. Piñana JL, Pérez A, Montoro J, et  al. Clinical effec­ tive­ ness of influ­ enza response to tick-borne enceph­a­li­tis vac­ci­na­tion in allo­ge­neic blood and
vac­ci­na­tion after allo­ge­neic hema­to­poi­etic stem cell trans­plan­ta­tion: a mar­row graft recip­i­ents. NPJ Vaccines. 2020;5(1):67.
cross-​sec­tional, pro­spec­tive, obser­va­tional study. Clin Infect Dis. 2019;​ 33. Einarsdottir S, Nicklasson M, Veje M, et al. Vaccination against tick-borne
68(11):1894-1903. enceph­a­li­tis (TBE) after autol­o­gous and allo­ge­neic stem cell trans­plan­ta­
19. Herishanu Y, Avivi I, Aharon A, et  al. Efficacy of the BNT162b2 mRNA tion. Vaccine. 2021;39(7):1035-1038.
COVID-19 vac­cine in patients with chronic lym­pho­cytic leu­ke­mia. Blood. 34. Assawawiroonhakarn S, Apiwattanakul N, Pakakasama S, et al. Immunoge-
2021;137(23):3165-3173. nicity of vero cell cul­ture-derived Jap­a­nese enceph­a­li­tis vac­cine in pedi­at­
20. Waissengrin B, Agbarya A, Safadi E, Padova H, Wolf I. Short-term safety ric and young hema­to­poi­etic stem cell trans­plan­ta­tion recip­i­ents. Pediatr
of the BNT162b2 mRNA COVID-19 vac­cine in patients with can­cer treated Infect Dis J. 2021;40(3):264-268.
with immune check­point inhib­i­tors. Lancet Oncol. 2021;22(5):581-583. 35. Adati EM, da Silva PM, Sumita LM, et al. Poor response to hep­a­ti­tis A vac­ci­
21. Pimpinelli F, Marchesi F, Piaggio G, et al. Fifth-week immu­no­ge­nic­ity and na­tion in hema­to­poi­etic stem cell trans­plant recip­i­ents. Transpl Infect Dis.
safety of anti-SARS-CoV-2 BNT162b2 vac­cine in patients with mul­ti­ple mye­ 2020;22(3):e13258.
loma and mye­lo­pro­lif­er­a­tive malig­nan­cies on active treat­ment: pre­lim­i­nary 36. Sicre de Fontbrune F, Arnaud C, Cheminant M, et al. Immunogenicity and
data from a sin­gle insti­tu­tion. J Hematol Oncol. 2021;14(1):81. safety of yel­low fever vac­cine in allo­ge­neic hema­to­poi­etic stem cell trans­
22. Monin L, Laing AG, Muñoz-Ruiz M, et al. Safety and immu­no­ge­nic­ity of one plant recip­i­ents after with­drawal of immu­no­sup­pres­sive ther­apy. J Infect
ver­sus two doses of the COVID-19 vac­cine BNT162b2 for patients with can­ Dis. 2018;217(3):494-497.
cer: interim anal­y­sis of a pro­spec­tive obser­va­tional study. Lancet Oncol.
2021;22(6):765-778.
23. Marion O, Del Bello A, Abravanel F, et al. Safety and immu­no­ge­nic­ity of anti-
SARS-CoV-2 mes­sen­ger RNA vac­cines in recip­i­ents of solid organ trans­ © 2021 by The Amer­i­can Society of Hematology
plants. Ann Intern Med. 2021:M21-1341. Published online 25 May 2021. DOI 10.1182/hema­tol­ogy.2021000294

Prakash Singh Shekhawat


Infectious com­pli­ca­tions and vac­cines  | 591
THE CHANGING LANDSCAPE OF α - AND β -THALASSEMIA DIAGNOSIS AND TREATMENT

Advances in the management of α-thalassemia


major: reasons to be optimistic

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/592/1851406/592horvei.pdf by guest on 13 December 2021


Paulina Horvei,1 Tippi MacKenzie,2 and Sandhya Kharbanda1
Division of Pediatric Allergy, Immunology and Bone Marrow Transplantation, and 2Division of Pediatric Surgery and Fetal Treatment Center, UCSF
1

Benioff Children’s Hospital, University of California, San Francisco, CA

α-Thalassemia major (ATM) is a severe disease resulting from deletions in all 4 copies of the α-globin gene. Although it is
usually fatal before birth, the advent of in utero transfusions has enabled survival of a growing number of children. Post-
natal therapy consists of chronic transfusions or stem cell transplantation, similar to patients with β-thalassemia major.
In this review, we discuss the experience with postnatal stem cell transplantation in patients with ATM, as well as the
ongoing phase 1 clinical trial of in utero stem cell transplantation for this condition.

LEARNING OBJECTIVES
• Review HCT for patients with ATM
• Discuss optimal conditioning regimens for HCT in patients with ATM

CLINICAL CASE patterns such that in California 1 in 10 000 newborns have


A pregnant woman of Southeast Asian ancestry under- a clinically significant α-thalassemia, and the rate of ATM
went routine prenatal ultrasound and was found to have a is 0.2 per 100 000 state births.2 α-Thalassemia has varying
fetus with hydrops fetalis, manifesting as ascites and pla- degrees of severity depending on the number of deleted or
centamegaly. She and her partner were both known to mutated genes and remaining functional α-globin genes.
be carriers of 2 α-globin deletions, giving them a 25% Carriers of this condition can have (a) 1 α-globin gene
chance of a pregnancy with deletion in all 4 α-globin genes deleted/inactivated in each chromosome (α−/α−), known
(α-thalassemia major [ATM]). Given the concern for ATM as the trans form of the α-thalassemia trait (common in
in the fetus, the couple were counseled regarding their people of African descent), or b) 2 missing/inactivated
options for the pregnancy, including pregnancy termina- α-globin genes on the same chromosome (αα/−−), known
tion or fetal therapy with intrauterine transfusions (IUTs). as the cis form of the α-thalassemia trait (common in peo-
They opted for IUTs, which were performed starting at ple of Asian descent).
23 weeks of gestation and repeated every 3 weeks until There are 2 clinically significant forms: ATM, also known
birth. The baby was born at term and stayed in the hospi- as α0-thalassemia or hemoglobin (Hb) Bart’s hydrops feta-
tal for 2 weeks. Postnatal transfusions were given every 3 lis, caused by deletion/inactivation of all 4 α-globin genes
weeks. Neurological development was normal at the time (−/−), and HbH disease, most frequently caused by deletion/
of testing at 8 months. inactivation of 3 α-globin genes (−/−α).3

Introduction Normal and abnormal developmental Hb


Epidemiology and genetics and pathology of hydrops
α-Thalassemia is a recessively inherited hemoglobinopathy Fetal oxygen exchange is accomplished by embryonic Hb
caused by mutations in the α-globin genes located on the (composed of 2 zeta and 2 gamma chains, or ξ2γ2) until 2
short arm of chromosome 16. It is one of the most common months of gestation (Figure 1); thereafter, fetal Hb (α2γ2)
monogenic disorders in the world, affecting approximately performs this function.1 A reduction of α-globin results in
5% of the population; prevalence is highest in China, South- the aggregation of γ-globin tetramers in utero (Hb Bart’s)
east Asia, the Middle East, India, and Africa.1 There is a rising and after birth, of β-globin tetramers (HbH). Both Hb Bart’s
incidence in the Western United States due to immigration and HbH have increased oxygen affinity, resulting in poor

592 | Hematology 2021 | ASH Education Program


Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/592/1851406/592horvei.pdf by guest on 13 December 2021
Figure 1. Hb switching at the human α- and β-globin loci. Green, embryonic globins; blue, fetal globins; red, adult globins. Primitive
erythropoiesis, derived from the yolk sac, is characterized by the expression of ζ-globin (from the α-globin locus) and ɛ-globin (from
the β-globin locus). These are silenced at approximately 8 weeks’ gestation. α-Globin then accounts for the entirety of the transcrip-
tional output from the α-globin locus. At the β-globin locus, there is a switch to fetal globin (γ-globin) during fetal life and then a sec-
ond switch to the adult β-globin. The predominant type of Hb corresponding to each developmental stage is shown below. Adapted
with permission from King and Higgs 2018.4

oxy­gen deliv­ery; they cause the pre­ma­ture destruc­tion of mature IUTs and impact on clin­i­cal out­comes
red blood cells (RBCs), lead­ing to hemo­ly­sis, and dam­age matur­ Given the sever­ity of ATM and the lack of ther­a­peu­tic options
ing ery­throid pre­cur­sors, lead­ing to inef­fec­tive eryth­ro­poi­e­sis.5 uni­ ver­sally avail­­
able until recently, par­ ents have most often
While most patients with a 3 α-glo­bin gene dele­tion do not have elected to undergo ter­ mi­na­
tion of preg­ nancy. However, IUTs
severe symp­toms, some patients with nondeletional muta­tions, are now com­monly performed for mul­ti­ple fetal ane­mias and
such as Hb Constant Spring, require chronic trans­fu­sions and are increas­ingly used to treat fetuses with ATM. Although the
may ben­e­fit from stem cell trans­plan­ta­tion.6 ini­tial IUT can treat the crit­i­cally low ane­mia, sub­se­quent IUTs
ATM is the most severe form: patients have a pre­ na­tal are gen­ er­ally given every 3 weeks to main­ tain an appro­ pri­
onset of non­im­mune hydrops fetalis as a result of heart fail­ure ate Hb level.1 Most fetal cen­ters start IUTs at 18 weeks of ges­
induced by severe ane­mia. Embryonic Hb Portland (ξ2γ2) is ta­tional age and fol­low pro­to­cols sim­i­lar to those for treat­ment
the only func­tional oxy­gen-car­ry­ing Hb in these infants, which of isoimmunization,8 with low com­pli­ca­tion rates (1.2% per pro­
is short-lived, as the ξ-glo­bin gene is then silenced, switch- ce­ dure, 3.3% per fetus).9 Since most fetuses are diag­ nosed
ing to the non­ func­
tional Hb Bart’s. Extramedullary eryth­ ro­ after 18 weeks, the most com­mon approach is an intra­ve­nous
poi­e­sis with marked hepatosplenomegaly and an enlarged infu­sion through the umbil­i­cal vein. The pro­to­col is to trans­fuse
pla­centa are com­mon. With few excep­tions, ATM is fatal in O neg­a­tive blood with a high hemat­o­crit so that the ane­mia
utero or shortly after birth with­out IUTs to treat the ane­mia. may be corrected with­out vol­ume overloading the fetus. Several
In untreated patients, hydrops can lead to mater­nal com­pli­ reports indi­cate that this fetal ther­apy pro­vi­des ben­e­fits dur­ing
ca­tions such as ane­mia, polyhydramnios, pre­term labor, and the peri­na­tal and neo­na­tal period: it reverses ane­mia, fetal growth
pre­eclamp­sia.3 restric­tion, and hydrops; decreases pre­term deliv­er­ies; and con­
trib­utes to infants hav­ing higher Apgar scores and a shorter dura­
Prenatal diag­no­sis and coun­sel­ing tion of neo­na­tal ven­ti­la­tion com­pared to untreated patients.5
Couples who are car­ri­ers for α-glo­bin dele­tions and are at risk Several case series have also dem­on­strated a long-term pos­i­tive
of hav­ing a child with ATM should receive genetic coun­sel­ing impact on growth and devel­op­ment.1,5,10,11 After birth, infants with
to review the nat­u­ral his­tory of the dis­or­der and dis­cuss pre­ ATM con­tinue to require trans­fu­sions, using a pro­to­col sim­i­lar to
na­tal genetic test­ing, repro­duc­tive options, and pre­na­tal and patients with β-thal­as­se­mia major (BTM). Given the improv­ing
post­na­tal treat­ment options and their risks. During preg­nancy, sur­vival of patients with ATM, par­ents can be given the option of
a defin­i­tive fetal diag­no­sis can be obtained using chorionic vil­ fetal ther­apy dur­ing a non­di­rec­tive pre­na­tal coun­sel­ing ses­sion.
lus sam­pling, amnio­cen­te­sis, or fetal blood sam­pling.7 Given the
neces­sity of IUTs to enable sur­vival, early diag­no­sis (pref­er­a­bly Allogeneic hema­to­poi­etic stem cell trans­plan­ta­tion
prior to the onset of hydrops fetalis) is impor­tant. Although some for ATM
infants have, report­edly, sur­vived with­out IUTs, they are almost Allogeneic hema­to­poi­etic stem cell trans­plan­ta­tion (HCT) is cur­
always born pre­term and have neo­na­tal com­pli­ca­tions due to rently the only avail­­able cura­tive treat­ment option for patients
the effects of untreated fetal hyp­oxia.5 with ATM. The under­ly­ing prin­ci­ple of allo­ge­neic HCT in ATM is
Prakash Singh Shekhawat
Advances in ther­a­pies for α-thal­as­se­mia major  |  593
sim­i­lar to that in other hemo­glo­bin­op­a­thies and involves the and 86% vs 82%, respec­tively).23 Evidence also sup­ports that
replace­ment of recip­i­ent hema­to­poi­etic stem cells (HSCs) with patients who receive a trans­plant before 2 years have the best
donor HSCs, resulting in the pro­duc­tion of donor-derived RBCs out­comes, with a 2-year OS and EFS of 95% and 93%.24
that do not har­bor the α-thal­as­se­mia muta­tion. Traditional myeloablative con­di­tion­ing reg­i­mens such as
In 1998 a 21-month-old girl with ATM received a bone mar­row Bu/Cy typ­i­cally result in sustained donor mye­loid engraft­ment;
(BM) HCT from a matched sib­ling donor (MSD) after con­di­tion­ing how­ever, they are asso­ci­ated with sig­nif­i­cant poten­tial tox­ic­ity
with busul­fan (Bu), cyclo­phos­pha­mide (Cy), and horse-derived that can result in organ dys­func­tion, such as VOD and treat­ment-
antithymocyte glob­u­lin (hATG). Graft-ver­sus-host dis­ease (GVHD) related mor­tal­ity (TRM). Therefore, the use of reduced-tox­ic­ity
pro­phy­laxis included meth­o­trex­ate (MTX) and cyclo­spor­ine con­di­tion­ing (RTC) reg­i­mens, with the goal of min­i­miz­ing toxic-
A (CSA). Neutrophil engraft­ment was detected at day +17. No ities with­out jeopardizing engraft­ment, is an appeal­ing option
major HCT-related com­pli­ca­tions devel­oped. Hb lev­els remained and a con­tin­ued sub­ject of inves­ti­ga­tion in the field. Studies done
>10 g/dL with­ out blood trans­ fu­sions despite the pres­ ence of in patients with high-risk BTM showed that sub­sti­tu­tion of Flu for
resid­ ual host HSCs.12 Since this first case published in 1998, Cy or Bu/Flu-based con­di­tion­ing reg­i­mens led to myeloablation
14 other patients who received an HCT for ATM have been but were asso­ci­ated with decreased tox­ic­ity com­pared to the

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/592/1851406/592horvei.pdf by guest on 13 December 2021


shared with the sci­en­tific com­mu­nity (Table 1): 5 patients (33%) con­ven­tional Bu/Cy reg­i­mens, hence the term RTC.22,25 Further
did not receive IUTs, and all­were born pre­ma­ture, with lower strides toward improved safety pro­files of RTC reg­i­mens have
Apgar scores at 1 and 5 min­utes, had evi­dence of hydrops been made with the use of phar­ma­co­ki­netic (PK) model-based
(3 mild, 2 severe), and required lon­ger inten­sive neo­na­tal care. dos­ing of con­di­tion­ing agents, which allows for reduced tox­ic­ity
Ten patients (66%) received IUTs and con­ se­quently had less while maintaining the reg­i­men’s effi­cacy. The drug’s expo­sure,
hydrops and were born at term (50%) or late pre­term. Despite mea­sured as a cumu­la­tive area under the curve (cAUC) over the
some patients expe­ri­enc­ing a chal­leng­ing ini­tial neo­na­tal course, treat­ment course, can be adjusted in real time using ther­a­peu­tic
with chronic trans­ fu­
sions, inten­sive care, and close fol­ low-up drug mon­i­tor­ing. A drug expo­sure below a cer­tain thresh­old car­
they were ­able to be bridged to trans­plant. Ten patients (66%) ries an increased risk of graft fail­ure, whereas an expo­sure above
received HCT at an age ≤24 months, with the youn­gest being the desired range is asso­ci­ated with an increased risk of organ
5 months of age. In total, 16 HCTs were performed; 2 patients had tox­ic­ity. In a study com­par­ing PK-targeted Bu and Flu and con­
graft fail­ure, 1 of which was sal­vaged with a sec­ond trans­plant, ven­tional Bu and Cy, the for­mer approach was shown to be less
while the other con­tin­ued on chronic trans­fu­sions. In 5 patients toxic while maintaining effi­cacy in pedi­at­ric HCT patients.26 In
the choice of con­di­tion­ing reg­i­men was not reported (NR). Of sev­eral non­ma­lig­nant dis­eases, includ­ing thal­as­se­mia, this expo­
the remaining, the major­ ity received myeloablative con­ di­
tion­ sure could be fur­ther reduced due to the accept­abil­ity of mixed
ing, which in 8 patients (53%) was a Bu-based reg­i­men along with but sta­ble mye­loid chi­me­rism, which often leads to ther­a­peu­tic
other agents, includ­ing fludarabine (Flu) and Cy. With respect to ben­e­fit. More recently, our group has shown that a less toxic
types of donors and stem cell source, 4 patients received an MSD and even lower expo­sure to Bu with a cAUC of 70 mg·h/L was
(3 BM, 1 NR), 1 received related mismatched umbil­i­cal cord blood ade­ quate for dis­ ease cor­rec­ tion and achieve­ ment of dura­ ble
cells (UCB), 4 received matched unre­lated donor stem cells (1 BM, full or sta­ble mixed mye­loid engraft­ment (chi­me­rism of >20%) in
3 periph­eral blood stem cells [PBSCs]), 4 got mismatched unre­ pedi­at­ric non­ma­lig­nant con­di­tions, with rare cases of severe Bu-
lated donor HSCs (2 UCB, 2 NR), and 1 patient received αβ T-cell- related tox­ic­ity and GVHD.27 Further, stud­ies aiming to under­
depleted haploidentical PBSCs. Only 1 patient, who was 13 years stand the rela­tion­ship between graft rejec­tion and Flu dose in
old at the time of trans­plant—the oldest patient reported—died chil­dren under­go­ing HCT have been conducted to opti­mize the
from trans­plant-related com­pli­ca­tions. Considering those patients immu­no­sup­pres­sive ele­ment of con­di­tion­ing based on which
that engrafted, all­became trans­ fu­
sion inde­
pen­ dent. Chimerism a Flu cAUC of 16 to 20 mg·h/L is deemed safe and effec­tive
data were reported in 13 patients, and approx­im ­ a­tely half of the to over­come the immune bar­rier while show­ing no ben­e­fit of
patients (6/13) were found to have mixed chi­me­rism and became increased expo­sure with respect to sur­vival.28,29 In a recent study
trans­fu­sion inde­pen­dent, sim­i­lar to those with full chi­me­rism. by Contreras et al,30 chil­dren receiv­ing RTC for non­ma­lig­nant
From those patients with known iron over­ load prior to HCT HCT, a total of 62 patients, were transplanted (23% had a diag­
(9 total; 1 mild, 4 mod­er­ate, 4 severe), only 1 patient was reported no­sis of hemo­glo­bin­op­a­thies, 7, BTM, and 7, sickle cell dis­ease
to develop severe and 1 patient to develop mild veno-occlu­sive [SCD]). Using targeted Bu and Flu in com­bi­na­tion with alemtu-
dis­ease of the liver (VOD). Of the patients who suc­ cess­ fully zumab (2012-2018), the cumu­la­tive inci­dence of graft fail­ure was
engrafted and sur­vived, 6/13 (53%) had no devel­op­men­tal delays 6.9% (none occurred in patients with an under­ly­ing diag­no­sis of
reported (66% of whom had pre­vi­ously received IUTs), and 46% hemo­glo­bin­op­a­thies), the 3-year cumu­la­tive inci­dence of TRM
(6/13) had a mild delay. For 1 patient this was NR.1,12-20 was 3.4%, and the 3-year OS was 96.6%, with a low inci­dence of
In con­trast to ATM, the results of allo­ge­neic HCT for a closely acute GVHD (aGVHD) grade 2 to 4 and chronic GVHD (7% and
related hemo­glo­bin­op­a­thy, BTM, are widely reported and well 5%, respec­tively). These results sug­gest that the use of targeted
stud­ied in large patient pop­u­la­tions. Patients transplanted after Bu and Flu offers a well-tol­er­ated option for chil­dren with non­
the year 2000 have had an out­stand­ing 2-year over­all sur­vival ma­lig­nant dis­or­ders to achieve sustained engraft­ment with a low
(OS) and event-free sur­vival (EFS) with the use of MSD (93% and inci­dence of trans­plant-related com­pli­ca­tions.30
85%, respec­tively) and BM (91% and 83%, respec­tively).22 How- The thresh­old chi­me­rism needed to ren­der trans­fu­sion inde­
ever, in recent years the use of Bu-based myeloablative reg­i­mens pen­dence fol­low­ing trans­plan­ta­tion in patients with thal­as­se­
have sig­nif­i­cantly improved out­comes with alter­na­tive donors, mias is not clearly known. However, the lim­ited data in BTM
and they are now com­pa­ra­ble to those of MSD (5-year OS and sug­gest that com­plete donor chi­me­rism is not essen­tial for sus-
EFS for MSD and HLA-matched unre­lated donors of 89% vs 87% tained engraft­ment or for achiev­ing trans­fu­sion inde­pen­dence,

594  |  Hematology 2021  |  ASH Education Program


Table 1. Summary of patients receiv­ing HCT for ATM

Conditioning reg­i­men and


Age at HCT Donor source GVHD pro­phy­laxis Toxicity and com­pli­ca­tions Outcomes Authors
21 months MSD BM Bu, Cy, hATG, MTX, CSA None Neutrophil engraft­ment at Chik et al12
day +17. Stable mixed
chi­me­rism (75%-90%).
Transfusion inde­pen­dent.
20 months 5/6 sib­ling UCB (α-thal trait) Bu, Cy, hATG, MTX, CSA HHV-6 vire­mia, grade 2 skin Neutrophil engraft­ment Zhou et al14
aGVHD, ITP at day +26. 100% donor
chi­me­rism. Transfusion
inde­pen­dent.
23 months MSD BM (α-thal and HbE trait) Cy, TBI (1400 cGy), MTX None Neutrophil engraft­ment at Thornley et al15
day +26. Stable mixed chi­
me­rism (66%). Transfusion
inde­pen­dent.
24 months MSD Bu, Flu, ATG, TLI Mild VOD Initially engrafted but then Joshi et al16
had graft fail­ure at 7
months after HCT. On
chronic trans­fu­sions with
che­la­tion ther­apy.
8 months, 18 months #1: 4/6 unre­lated UCB #1 HCT: Bu, Flu, TLI, CSA, EBV + PTLD #1 HCT: graft fail­ure and on Yi et al17
mismatched at B and DRB; MMF; #2 HCT: Bu, Flu, ATG, chronic trans­fu­sions.
#2 MUD 6/6 PBSCT, CD34+ TBI (200 cGy), CSA, MMF #2 HCT: neu­tro­phil engraft­
selec­tion ment at day +18. Stable
mixed chi­me­rism (90%-
97%). Transfusion

Prakash Singh Shekhawat


inde­pen­dent.
44 months 5/6 unre­lated UCB Bu, Cy, ATG Grade 2 skin aGVHD, 100% donor chi­me­rism. Gumuscu et al13
can­dida sep­sis Transfusion inde­pen­dent.
19 months MSD BM NR NR Neutrophil engraft­ment at Pongtanakul et al20
day +17. Initially, 100%
donor at day +25 then sta­
ble mixed chi­me­rism (41%
donor at day +112) with sta­
ble Hb of 9.1. Discontinued
immu­no­sup­pres­sion and
received DLI × 5. Converted
to 100% donor chi­me­rism.
Transfusion inde­pen­dent.
5 months MUD 10/10 BM Bu, Flu, ATG, MTX, CSA Severe VOD Neutrophil engraft­ment at Elsaid et al19
day +25. Stable mixed
chi­me­rism. Transfusion
inde­pen­dent.
13 years 9/10 MMUD NR (RIC) Invasive fun­gal infec­tion, Died 10 months post trans­ Pecker et al20
grade 4 skin and gut GVHD plant from trans­plant-
related com­pli­ca­tions.
10 years 9/10 MMUD NR (RIC) None 100% donor chi­me­rism. Pecker et al.20
Transfusion inde­pen­dent.

Advances in ther­a­pies for α-thal­as­se­mia major  |  595


Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/592/1851406/592horvei.pdf by guest on 13 December 2021
Table 1. (continued)

Conditioning reg­i­men and


Age at HCT Donor source GVHD pro­phy­laxis Toxicity and com­pli­ca­tions Outcomes Authors
12 months MSD NR NR Transfusion inde­pen­dent by Kreger et al1
18 months.

596  |  Hematology 2021  |  ASH Education Program


24 months NR NR NR Transfusion inde­pen­dent by Kreger et al1
age 3.
22 months MUD 12/12 PBSCT Bu, Cy, hATG, MTX, CSA Grade 2 skin and grade 3 gut Neutrophil engraft­ment at Chan et al18
aGVHD, HHV-7 vire­mia day +15. 100% donor chi­
me­rism. Transfusion inde­
pen­dent.
28 months MUD PBSC HU, azacitadine, Cy, Bu, TT, Grade 2 skin aGVHD. HHV-7 Neutrophil engraft­ment at Chan et al18
Flu, rATG, MTX, MMF, CSA and EBV vire­mia, cen­tral day +11. Mixed chi­me­rism
line sep­sis (98%). Transfusion inde­
pen­dent.
60 months AB TCD haploidentical PBSC HU, azacitadine, Cy, TT, Fu, Grade 2 skin and gut aGVHD. Neutrophil engraft­ment at Chan et al18
Treo, rATG Klebsiella bac­ter­emia. day +13. Mixed chi­me­rism
(99%). Transfusion inde­
pen­dent.
AB TCD, αβ T-cell depleted; cGVHD, chronic graft-ver­sus-host dis­ease; DRB, HLA DR isotype, beta chain; DLI, donor lym­pho­cyte infu­sions; EBV, Epstein-Barr virus; HbE, hemoglobin E;
HHV, human her­pes­vi­rus; HU, hydroxy­urea; ITP, immune throm­bo­cy­to­pe­nia; Mel, mel­pha­lan; MMF, mycophenolate mofetil; MUD, matched unre­lated donor; MMUD, mismatched unre­lated donor;
PBSCT, periph­eral blood stem cell trans­plant; PTLD, post-transplant proliferative disease; rATG, rab­bit-derived ATG; RIC, reduced-inten­sity con­di­tion­ing; TBI, total body irra­di­a­tion; TLI, total
lym­phoid irra­di­a­tion; Treo, treosulfan; TT, thio­tepa.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/592/1851406/592horvei.pdf by guest on 13 December 2021


Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/592/1851406/592horvei.pdf by guest on 13 December 2021

Figure 2. Prenatal screening for ATM. (1) This tool is not a replacement for referral to genetic counseling, which may happen at
any time in this pathway. Genetic counseling provides guidance for genetic testing and management options for families with
­pregnancies at risk for severe forms of thalassemia. (2) The sensitivity and specificity are not definitive, and not all carriers will be
detected by this screening. (3) The American College of Obstetricians and Gynecologists recommends that all pregnant women
have a CBC with a ­ ssessment of MCV. (4) Rare mutations, such as nondeletional α-thalassemia and others, may not be captured in this
­algorithm. In high-risk cases, or where Hb electrophoresis is abnormal, consultation with a genetic counselor and/or h ­ ematologist
is ­recommended. (5) The presence of HbA2 > 3.5 does not exclude a coexisting α0-thalassemia trait. In individuals of Southeast
Asian, ­Filipino, or ­Chinese descent who have microcytic hypochromic anemia, perform α-globin gene deletion and common vari-
ant studies irrespective of HbA2 level. CBC, complete blood count; CVS, chorionic villus sampling; HPLC, high-performance liquid
­chromatography; MCA, ­middle c ­ erebral artery; MCH, mean corpuscular hemoglobin; MCV, mean corpuscular volume; MoM, multiples
of the median; PCR, ­polymerase chain reaction; PSV, peak systolic velocity; PUBS, percutaneous umbilical blood sampling. Adapted
Prakash Singh Shekhawat
with permission from the University of California, San Francisco hemoglobinopathy screening algorithm.
Advances in ther­a­pies for α-thal­as­se­mia major  |  597
and this is thought to be due to inef­fec­tive eryth­ro­poi­e­sis, near-com­plete Hb replace­ment (donor Hb >90%), ulti­mately
which allows the donor RBCs to have a sur­vival advan­tage. The correcting SCD and β-thal­as­se­mia phe­no­types.36
per­sis­tence of resid­ual host cells at >25% early fol­low­ing trans­ There have been mul­ti­ple pre­vi­ous reports of IUHCT in mul­
plant (<60 days) was shown to be a risk for graft rejec­tion, while ti­ple dis­ease set­tings, includ­ing in fetuses with ATM with low
a level <10% was deemed low risk for rejec­tion.31 Patients with engraft­ment.37,38 However, none of these pre­vi­ous reports has
per­sis­tent mixed chi­me­rism (PMC), defined as a sta­ble mixed used the strat­egy of a high dose of mater­nal cells, infused intra­
chi­me­rism for >2 years, were a ­ ble to remain trans­fu­sion inde­ ve­nously, which is the pro­to­col of our ongo­ing clin­i­cal trial for
pen­dent even with >25% resid­ual host cells, and their graft was ATM. Of note, engraft­ment has been seen in the uniquely per­mis­
func­tional and suf­fi­cient to lead to trans­fu­sion inde­pen­dence.31 sive set­ting of immunodeficiencies such as bare lym­pho­cyte syn­
This is fur­ther supported by evi­dence from split chi­me­rism in drome and severe com­bined immu­no­de­fi­ciency.39,40 We chose
patients with PMC, where Andreani et  al.32 observed that the to start our clin­i­cal trial in patients with ATM since they require
pro­por­tion of donor-derived cells was equally dis­trib­uted in the in IUTs for sur­vival so there is no addi­tional pro­ce­dural risk from
dif­fer­ent cell lines, both in the periph­eral blood and BM, with the trans­plan­ta­tion pro­to­col. If the pro­to­col is safe, it could be
the excep­tion of the eryth­ro­cyte com­part­ment. Despite the applied to other hemo­glo­bin­op­a­thies or other dis­eases that can

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/592/1851406/592horvei.pdf by guest on 13 December 2021


pres­ence of few donor-engrafted nucle­ated cells, the eryth­ be treated with HCT, such as Fanconi ane­mia or inborn errors
ro­cytes were almost com­pletely donor in ori­gin (80%-100%). of metab­o­lism. Given the impor­tance of cell source, dose, and
This enrich­ment of donor RBCs in the blood was not observed other var­i­ables in engraft­ment, it is vital to per­form IUHCT in the
in ery­throid pre­cur­sors from the mar­row, suggesting that the con­text of well-designed clin­i­cal tri­als.41
inef­fec­tive eryth­ro­poi­e­sis pre­sum­ably respon­si­ble for this phe­
nom­e­non works at a later stage of ery­throid devel­op­ment.32 Conclusions and future direc­tions
Consequently, since ATM is also char­ac­ter­ized by inef­fec­tive Although ATM was pre­vi­ously seen as a fatal dis­ease, there are
eryth­ro­poi­e­sis due to the imbal­ance of α and β chains,5 it is now mul­ ti­
ple reports of patients who have sur­ vived to birth
likely that com­plete donor chi­me­rism is not crit­i­cal, and PMC with excel­lent qual­ity of life after IUTs. Since these patients still
would be suf­fi­cient to ren­der trans­fu­sion inde­pen­dence fol­low­ need chronic trans­fu­sions after birth, it is impor­tant to con­tinue
ing allo­ge­neic HCT.31 to develop strat­eg ­ ies for a defin­i­tive cure using HCT. Postnatal
HCT can be cura­tive in ATM and can be safely performed using
Rationale for in utero trans­plan­ta­tion in patients with ATM RTC, espe­cially with PK model-based dos­ing to main­tain effi­cacy
In utero HSC trans­plan­ta­tion (IUHCT) is a prom­is­ing ther­apy for while min­i­miz­ing tox­ic­ity. It should be performed early in life to
dis­eases that can be treated by post­na­tal HCT. IUHCT takes decrease the risk of TRM and other com­pli­ca­tions. In the future,
advan­tage of the unique immune sta­tus in the fetus to allow nongenotoxic anti­body-medi­ated con­di­tion­ing reg­i­mens could
engraft­ment of transplanted cells with­out con­di­tion­ing.33 Our fur­ther improve safety and effi­cacy.42 IUHCT is another prom­is­
team is cur­rently performing a phase 1 clin­i­cal trial of IUHCT ing approach that we are test­ing in a clin­i­cal trial. For IUHCT, it
(NCT02986698) using mater­nal stem cells as the donor: this is impor­tant to use mater­nal stem cells to take advan­tage of the
strat­egy takes advan­tage of 2 aspects of mater­nal-fetal biol­ preexisting tol­er­ance between the mother and fetus. This strat­
ogy resulting from the nat­u­ral traf­fick­ing of cells between the egy will likely still require a post­na­tal “boost” trans­plan­ta­tion,
mother and the fetus: first, the pres­ence of mater­nal cells in the and defin­ing safe and effec­tive pro­to­cols to reduce trans­plant-
fetus results in fetal tol­er­ance to noninherited mater­nal anti­gens related com­pli­ca­tions will be impor­tant. In the future, gene ther­
dur­ing preg­nancy34; sec­ond, the mater­nal immune sys­tem can apy or gene editing approaches may also be devel­ oped for
medi­ate rejec­tion of third-party cells transplanted into the fetus these patients. Several strat­e­gies devel­oped for β-thal­as­se­mia
in mouse mod­els.35 Thus, trans­plan­ta­tion of mater­nal cells pro­ could be employed, includ­ing ex vivo lentiviral gene ther­apy to
vi­des the highest like­li­hood of suc­cess. We are cur­rently enroll­ replace α-glo­bin or gene editing to intro­duce α-glo­bin into a
ing patients with ATM for IUHCT between 18 and 26 weeks of geno­mic safe har­bor. Given the improved sur­vival of patients
ges­ta­tion (Figure 2). It is impor­tant to note that although there with ATM as a result of in IUTs, increas­ing num­bers of patients
are immune advan­tages to transplanting mater­nal stem cells with ATM will require chronic care: improved HCT or gene ther­
into the fetus, the cur­rent lack of an appro­pri­ate con­di­tion­ing apy options could be trans­ for­
ma­tional in enabling defin­i­
tive
reg­i­men to cre­ate space in the hema­to­poi­etic niche results in cures.
low lev­els of engraft­ment in most ani­mal mod­els unless a very
high dose of cells is infused. Therefore, the most likely sce­nario Conflict-of-inter­est dis­clo­sure
is that the pre­na­tal trans­plan­ta­tion is part of a 2-step pro­to­col Paulina Horvei: no com­pet­ing finan­cial inter­ests to declare.
in which the in utero trans­plant achieves enough engraft­ment Tippi MacKenzie: sci­en­tific advi­sory board mem­ber: Acrigen.
to sus­tain tol­er­ance to mater­nal anti­gens after birth followed Sandhya Kharbanda: no com­pet­ing finan­cial inter­ests to declare.
by a post­na­tal “boost” using mater­nal HSC with an RTC reg­
i­men with min­i­mal or no immunoablation, improv­ing engraft­ Off-label drug use
ment to clin­i­cally mean­ing­ful lev­els. If suc­cess­ful, this pro­to­col Paulina Horvei: nothing to disclose.
would over­come the lack of donor avail­abil­ity as well as offer an Tippi MacKenzie: nothing to disclose.
improved safety pro­file com­pared to tra­di­tional post­na­tal HCT. Sandhya Kharbanda: nothing to disclose.
Indeed, prom­is­ing out­comes have been seen in murine mod­els
of SCD and BTM in which IUHCT resulted in the devel­op­ment Correspondence
of donor-spe­cific tol­er­ance and PMC. A post­na­tal boost was Sandhya Kharbanda, Division of Pediatric Allergy, Immunolo-
given, and chi­me­rism was fur­ther enhanced to high lev­els with gy and Bone Marrow Transplantation, UCSF Benioff Children’s

598  |  Hematology 2021  |  ASH Education Program


Hospital, University of California, 1975 4th St, San Francisco, CA reg­i­men according to a new risk strat­i­fi­ca­tion. Biol Blood Marrow Trans-
94158; e-mail: sandhya​­.kharbanda@ucsf​­.edu. plant. 2014;20(12):2066-2071.
23. Li C, Mathews V, Kim S, et  al. Related and unre­lated donor trans­plan­ta­
tion for β-thal­as­se­mia major: results of an inter­na­tional sur­vey. Blood Adv.
References 2019;3(17):2562-2570.
1. Kreger EM, Singer ST, Witt RG, et  al. Favorable out­comes after in utero 24. Baronciani D, Angelucci E, Potschger U, et al. Hemopoietic stem cell trans­
trans­fu­sion in fetuses with alpha thal­ as­
se­mia major: a case series and plan­ta­tion in thal­as­se­mia: a report from the Euro­pean Society for Blood
review of the lit­er­a­ture. Prenat Diagn. 2016;36(13):1242-1249. and Bone Marrow Transplantation Hemoglobinopathy Registry, 2000-
2. Feuchtbaum L, Carter J, Dowray S, Currier RJ, Lorey F. Birth prev­a­lence of 2010. Bone Marrow Transplant. 2016;51(4):536-541.
dis­or­ders detect­able through new­born screen­ing by race/eth­nic­ity. Genet 25. Sheth V, Grisariu S, Avni B, et al. Fludarabine-based reduced tox­ic­ity yet
Med. 2012;14(11):937-945. myeloablative con­di­tion­ing is effec­tive and safe par­tic­u­larly in patients
3. Tamary H, Dgany O. Alpha-Thalassemia. 2005 Nov 1 [updated 2020 Oct 1]. with high-risk thal­as­se­mia under­go­ing allo­ge­neic trans­plan­ta­tion. Pediatr
GeneReviews® [Internet]. Seattle (WA): University of Washington, Seattle; Blood Cancer. 2018;65(11):e27312.
1993–2021. 26. Bartelink IH, van Reij EM, Gerhardt CE, et  al. Fludarabine and expo­sure-
4. King AJ, Higgs DR. Potential new approaches to the management of the targeted busul­fan com­pares favor­ably with busul­fan/cyclo­phos­pha­mide-
Hb Bart’s hydrops fetalis syndrome: the most severe form of α-thalassemia. based reg­i­mens in pedi­at­ric hema­to­poi­etic cell trans­plan­ta­tion: maintaining
Hematology Am Soc Hematol Educ Program. 2018 Nov 30;2018(1):353-360.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/592/1851406/592horvei.pdf by guest on 13 December 2021


effi­cacy with less tox­ic­ity. Biol Blood Marrow Transplant. 2014;20(3):
5. Songdej D, Babbs C, Higgs DR; Bart’s Hydrops Fetalis Syndrome Inter- 345-353.
national Consortium. An inter­na­tional reg­is­try of sur­vi­vors with Hb Bart’s 27. Apsel Winger B, Shukla P, Kharbanda S, et  al. The rela­tion­ship between
hydrops fetalis syn­drome. Blood. 2017;129(10):1251-1259. busul­fan expo­sure and achieve­ment of sustained donor mye­loid chi­me­rism
6. Lal A, Goldrich ML, Haines DA, Azimi M, Singer ST, Vichinsky EP. Heteroge- in patients with non-malig­nant dis­or­ders. Transplant Cell Ther. 2021;27(3):
neity of hemo­glo­bin H dis­ease in child­hood. N Engl J Med. 2011;364(8):710- 258.e1-258.e6.
718. 28. Long-Boyle JR, Green KG, Brunstein CG, et al. High fludarabine expo­sure and
7. Amer­i­can College of Obstetricians and Gynecologists Committee on Obstet- rela­tion­ship with treat­ment-related mor­tal­ity after nonmyeloablative hema­
rics. ACOG Practice Bulletin No. 78: hemo­glo­bin­op­a­thies in preg­nancy. to­poi­etic cell trans­plan­ta­tion. Bone Marrow Transplant. 2011;46(1):20-26.
Obstet Gynecol. 2007;109(1):229-237. 29. Ivaturi V, Dvorak CC, Chan D, et  al. Pharmacokinetics and model-based
8. Society for Maternal-Fetal Medicine; Mari G, Norton M, Stone J, et al. Soci- dos­ing to opti­mize fludarabine ther­apy in pedi­at­ric hema­to­poi­etic cell
ety for Maternal-Fetal Medicine (SMFM) Clinical Guideline #8: the fetus trans­plant recip­i­ents. Biol Blood Marrow Transplant. 2017;23(10):1701-1713.
at risk for ane­mia—diag­no­sis and man­age­ment. Am J Obstet Gynecol. 30. Contreras CF, Long-Boyle JR, Shimano KA, et al. Reduced tox­ic­ity con­di­
2015;212(6):697-710. tion­ing for non­ma­lig­nant hema­to­poi­etic cell trans­plants. Biol Blood Mar-
9. Zwiers C, Lindenburg ITM, Klumper FJ, de Haas M, Oepkes D, Van Kamp IL. row Transplant. 2020;26(9):1646-1654.
Complications of intra­uter­ine intra­vas­cu­lar blood trans­fu­sion: les­sons 31. Andreani M, Testi M, Lucarelli G. Mixed chi­me­rism in haemoglobinopa-
learned after 1678 pro­ce­dures. Ultrasound Obstet Gynecol. 2017;50(2): thies: from risk of graft rejec­tion to immune tol­er­ance. Tissue Antigens.
180-186. 2014;83(3):137-146.
10. Chan WY, Leung AW, Luk CW, Li RC, Ling AS, Ha SY. Outcomes and morbid- 32. Andreani M, Testi M, Battarra M, Lucarelli G. Split chi­me­rism between nucle­
ities of patients who sur­vive haemoglobin Bart’s hydrops fetalis syn­drome: ated and red blood cells after bone mar­row trans­plan­ta­tion for haemoglo-
20-year ret­ro­spec­tive review. Hong Kong Med J. 2018;24(2):107-118. binopathies. Chimerism. 2011;2(1):21-22.
11. Zhang HJ, Amid A, Janzen LA, et  al. Outcomes of haemoglobin Bart’s 33. Witt R, MacKenzie TC, Peranteau WH. Fetal stem cell and gene ther­apy.
hydrops fetalis fol­low­ing intra­uter­ine trans­fu­sion in Ontario, Canada. Arch Semin Fetal Neonatal Med. 2017;22(6):410-414.
Dis Child Fetal Neonatal Ed. 2021;106(1):51-56. 34. Mold JE, Michaëlsson J, Burt TD, et al. Maternal allo­an­ti­gens pro­mote the
12. Chik KW, Shing MM, Li CK, et al. Treatment of hemo­glo­bin Bart’s hydrops devel­ op­ ment of tolerogenic fetal reg­ u­
la­
tory T cells in utero. Science.
with bone mar­row trans­plan­ta­tion. J Pediatr. 1998;132(6):1039-1042. 2008;322(5907):1562-1565.
13. Gumuscu B, Thompson EI, Grovas AC, Zach TL, Warkentin PI, Coccia PF. Suc- 35. Nijagal A, Wegorzewska M, Jarvis E, Le T, Tang Q , MacKenzie TC. Maternal
cessful unre­lated cord blood trans­plan­ta­tion for homo­zy­gous α-thal­as­se­mia. T cells limit engraft­ment after in utero hema­to­poi­etic cell trans­plan­ta­tion in
J Pediatr Hematol Oncol. 2013;35(7):570-572. mice. J Clin Invest. 2011;121(2):582-592.
14. Zhou X, Ha SY, Chan GC, et al. Successful mismatched sib­ling cord blood 36. Peranteau WH, Hayashi S, Abdulmalik O, et al. Correction of murine hemo­
trans­plant in Hb Bart’s dis­ease. Bone Marrow Transplant. 2001;28(1):105- glo­bin­op­a­thies by pre­na­tal tol­er­ance induc­tion and post­na­tal nonmye-
107. loablative allo­ge­neic BM trans­plants. Blood. 2015;126(10):1245-1254.
15. Thornley I, Lehmann L, Ferguson WS, Davis I, Forman EN, Guinan EC. 37. Cowan MJ, Golbus M. In utero hema­to­poi­etic stem cell trans­plants for
Homozygous alpha-thal­as­se­mia treated with intra­uter­ine trans­fu­sions and inherited dis­eases. Am J Pediatr Hematol Oncol. 1994;16(1):35-42. Accessed
post­na­tal hema­to­poi­etic stem cell trans­plan­ta­tion. Bone Marrow Trans- 23 Octo­ber 2021. https:­/­/pubmed­.ncbi­.nlm­.nih­.gov­/7906103­/
plant. 2003;32(3):341-342. 38. Hayward A, Ambruso D, Battaglia F, et al. Microchimerism and tol­er­ance
16. Joshi DD, Nickerson HJ, McManus MJ. Hydrops fetalis caused by homo­zy­ fol­low­ing intra­uter­ine trans­plan­ta­tion and trans­fu­sion for alpha-thal­as­se­mia
gous alpha-thal­as­se­mia and Rh anti­gen alloimmunization: report of a sur­vi­ -1. Fetal Diagn Ther. 1998;13(1):8-14.
vor and lit­er­a­ture review. Clin Med Res. 2004;2(4):228-232. 39. Touraine JL, Raudrant D, Royo C, et al. In-utero trans­plan­ta­tion of stem cells
17. Yi JS, Moertel CL, Baker KS. Homozygous alpha-thal­as­se­mia treated with in bare lym­pho­cyte syn­drome. Lancet. 1989;1(8651):1382.
intra­uter­ine trans­fu­sions and unre­lated donor hema­to­poi­etic cell trans­ 40. Flake AW, Roncarolo MG, Puck JM, et  al. Treatment of X-linked severe
plan­ta­tion. J Pediatr. 2009;154(5):766-768. com­bined immu­no­de­fi­ciency by in utero trans­plan­ta­tion of pater­nal
18. Chan WYK, Lee PPW, Lee V, et  al. Outcomes of allo­ge­neic trans­plan­ta­ bone mar­row. N Engl J Med. 1996;335(24):1806-1810.
tion for hemo­glo­bin Bart’s hydrops fetalis syn­drome in Hong Kong. Pediatr 41. MacKenzie TC, David AL, Flake AW, Almeida-Porada G. Consensus state­
Transplant. 2021;25(6):e14037. ment from the first inter­na­tional con­fer­ence for in utero stem cell trans­
19. Elsaid MY, Capitini CM, Diamond CA, Porte M, Otto M, DeSantes KB. Suc- plan­ta­tion and gene ther­apy. Front Pharmacol. 2015;6(10 Feb­ru­ary):15.
cessful matched unre­lated donor stem cell trans­plant in hemo­glo­bin 42. Palchaudhuri R, Saez B, Hoggatt J, et  al. Non-genotoxic con­di­tion­ing
Bart’s dis­ease. Bone Marrow Transplant. 2016;51(11):1522-1523. for hema­to­poi­etic stem cell trans­plan­ta­tion using a hema­to­poi­etic-cell-
20. Pongtanakul B, Sanpakit K, Chongkolwatana V, Viprakasit V. Normal cog­ni­ spe­cific inter­nal­iz­ing immunotoxin. Nat Biotechnol. 2016;34(7):738-745.
tive func­tion­ing in a patient with Hb Bart’s hydrops suc­cess­fully cured by
hema­to­poi­etic SCT. Bone Marrow Transplant. 2014;49(1):155-156.
21. Pecker LH, Guerrera MF, et al. Homozygous α-thalassemia: Challenges sur-
rounding early identification, treatment, and cure. Pediatr Blood Cancer.
2017 Jan;64(1):151-155.
22. Anurathapan U, Pakakasama S, Mekjaruskul P, et al. Outcomes of thal­as­se­
mia patients under­go­ing hema­to­poi­etic stem cell trans­plan­ta­tion by using © 2021 by The Amer­i­can Society of Hematology
a stan­dard myeloablative ver­sus a novel reduced-tox­ic­ity con­di­tion­ing DOI 10.1182/hema­tol­ogy.2021000295
Prakash Singh Shekhawat
Advances in ther­a­pies for α-thal­as­se­mia major  |  599
THE CHANGING LANDSCAPE OF α- AND β-THALASSEMIA DIAGNOSIS AND TREATMENT

β-Thalassemia: evolving treatment options


beyond transfusion and iron chelation

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/600/1851627/600langer.pdf by guest on 13 December 2021


Arielle L. Langer1 and Erica B. Esrick2
Division of Hematology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA; and 2Division of Hematology/Oncology, Boston
1

Children’s Hospital, Harvard Medical School, Boston, MA

After years of reliance on transfusion alone to address anemia and suppress ineffective erythropoiesis in β-thalassemia,
many new therapies are now in development. Luspatercept, a transforming growth factor–β inhibitor, has demonstrated
efficacy in reducing ineffective erythropoiesis, improving anemia, and possibly reducing iron loading. However, many
patients do not respond to luspatercept, so additional therapeutics are needed. Several medications in development
aim to induce hemoglobin F (HbF): sirolimus, benserazide, and IMR-687 (a phosphodiesterase 9 inhibitor). Another group
of agents seeks to ameliorate ineffective erythropoiesis and improve anemia by targeting abnormal iron metabolism in
thalassemia: apotransferrin, VIT-2763 (a ferroportin inhibitor), PTG-300 (a hepcidin mimetic), and an erythroferrone anti-
body in early development. Mitapivat, a pyruvate kinase activator, represents a unique mechanism to mitigate ineffective
erythropoiesis. Genetically modified autologous hematopoietic stem cell transplantation offers the potential for lifelong
transfusion independence. Through a gene addition approach, lentiviral vectors have been used to introduce a β-globin
gene into autologous hematopoietic stem cells. One such product, betibeglogene autotemcel (beti-cel), has reached
phase 3 trials with promising results. In addition, 2 gene editing techniques (CRISPR-Cas9 and zinc-finger nucleases) are
under investigation as a means to silence BCL11A to induce HbF with agents designated CTX001 and ST-400, respectively.
Results from the many clinical trials for these agents will yield results in the next few years, which may end the era of
relying on transfusion alone as the mainstay of thalassemia therapy.

LEARNING OBJECTIVES
• Understand the strengths and limitations of luspatercept for the treatment of TDT and NTDT
• Understand the potential benefits and toxicity of genetically modified autologous HSCT for TDT
• Describe therapies under development for the treatment of TDT and NTDT

After many years without novel disease-modifying ther- of approximately 9.5g/dL is indicated because of end-organ
apeutics, numerous agents are now in development for damage. However, she has been unable to tolerate the
β-thalassemia. We review therapies that have been recently transfusion volumes, as attempts at transfusion frequently
approved or are in development for transfusion-dependent trigger a heart failure exacerbation and acute worsening of
thalassemia (TDT) and non-transfusion-dependent thalasse- her pulmonary pressures. She is started on luspatercept,
mia (NTDT) β-thalassemia using 4 patient cases (Table 1). which leads to a rise in hemoglobin to 9.2 to 10.1g/dL with
drops below this range only in the setting of acute illness.

Luspatercept is a recombinant fusion protein that blocks


CLINICAL CASE 1: USE OF LUSPATERCEPT transforming growth factor-β (TGF-β) superfamily ligands
A 54-year-old woman with congestive heart failure, parox- and promotes late-stage erythroid maturation, resulting
ysmal atrial fibrillation on warfarin, and pulmonary hyper- in effective reduction in transfusion requirements in some
tension due to β-thalassemia intermedia has a hemoglobin patients with TDT.1 In a randomized, placebo-controlled
ranging from 7.5 to 8.5g/dL without packed red blood cell trial, 21.4% of patients met the primary end point of at
(RBC) transfusion. Transfusion to maintain hemoglobin nadir least a 33% reduction in transfusion volume during weeks

600 | Hematology 2021 | ASH Education Program


Table 1. Current lim­i­ta­tions of thal­as­se­mia care dence of throm­bo­sis may com­pound other risk fac­tors, such as
prior sple­nec­tomy, and this may affect the risk-ben­e­fit ratio of
Limitation Potential solu­tions ini­ti­at­ing luspatercept. In case 1, the patient was already on life­
Regular trans­fu­sion appoint­ments Reduce trans­fu­sion require­ment long anticoagulation because of atrial fibril­la­tion, so this risk was
Stem cell trans­plan­ta­tion miti­gated. Finally, published data pri­mar­ily address the TDT pop­
u­la­tion; forth­com­ing results will help clar­ify how best to tai­lor the
Iron over­load Reduce trans­fu­sion require­ment
Block intes­ti­nal iron absorp­tion use of luspatercept for patients with NTDT.
Escalate che­la­tion Further fol­low-up will be required to deter­mine if luspater-
Iron che­la­tor tox­ic­ity Reduce trans­fu­sion require­ment
cept may improve iron over­load, although pre­lim­i­nary results
Block intes­ti­nal iron absorp­tion are prom­is­ing.6 Early find­ings that fer­ri­tin and liver iron con­cen­
Additional che­la­tion options tra­tion (LIC) improved in both those who met the pri­mary end
Lack of allo­ge­neic stem cell donor Genetically mod­i­fied autol­o­gous point and those who did not sug­gest that reduced iron load­
trans­plan­ta­tion ing from the gut is likely playing a role, rather than just reduced
Nontransplant ther­a­pies that

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/600/1851627/600langer.pdf by guest on 13 December 2021


trans­fu­sion bur­den. If this bears out, it would rep­re­sent a sig­
pre­clude desir­abil­ity of trans­plant nif­i­cant addi­tional ben­e­fit, par­tic­u­larly for indi­vid­u­als in poor
iron bal­ance. Although luspatercept rep­re­sents an excit­ing new
13 through 24 com­pared with 4.5% receiv­ing pla­cebo (P < .001). ther­a­peu­tic option and is the first novel approved ther­apy in
This equated to a least mean square dif­fer­ence in trans­fu­sion many years, cau­tion is merited in set­ting expec­ta­tions, as most
bur­den of 1.35 fewer RBC trans­fu­sions in weeks 13 through 24 patients will not be ­able to forgo trans­fu­sion, and increas­ing the
in the TDT pop­u­la­tion. Follow-up data show trans­fu­sion reduc­ inter­val between trans­fu­sions is often not prac­ti­cally fea­si­ble or
tion con­tin­ued at 48 weeks and up to 4.8 years into treat­ment.2,3 appeal­ing since vis­its every 3 weeks are required for this sub­cu­
Common side effects included head­ache, myal­gia, and bone ta­ne­ous med­i­ca­tion. As such, a great unmet need remains.
pain. Although rare, 8 of 223 (3.6%) patients in the luspatercept
arm had throm­bo­em­bo­lism com­pared with 1 of 109 patients in
the pla­cebo arm (Table 2).1
In the NTDT pop­u­la­tion, data from a phase 1/2 nonrandomized
trial showed an increase in hemo­glo­bin of at least 1.5 g/dL in 58% CLINICAL CASE 2: AUTOLOGOUS GENETIC THERAPIES
of patients receiv­ing higher dose lev­els.4 A ran­dom­ized, pla­cebo- A 17-year-old boy with TDT on a sta­ble trans­fu­sion reg­i­men and
con­trolled trial for luspatercept in patients with NTDT has fin­ished in good iron bal­ance wishes to con­sider cura­tive intent ther­apy.
accrual, and pre­lim­i­nary results showed hemo­glo­bin of at least He is con­sid­er­ing his goals for adult­hood, includ­ing attend­ing
1.5 g/dL in 52.1% in the luspatercept arm com­pared with 0% in col­lege that is not near a large med­i­cal cen­ter, and is inter­ested
the pla­cebo arm and no throm­bo­em­bolic events in either arm in avoiding fre­quent med­i­cal and trans­fu­sion appoint­ments. He
(NCT03342404).5 has no avail­­able donor for allo­ge­neic stem cell trans­plan­ta­tion.
Case 1 dem­on­strates sev­eral strengths and lim­i­ta­tions of luspa- He opts to enroll in a clin­i­cal trial of genet­i­cally mod­i­fied autol­
tercept. This med­i­ca­tion is most likely to be use­ful in patients for o­gous hema­to­poi­etic stem cell trans­plan­ta­tion (HSCT) using a
whom a mod­er­ate increase in hemo­glo­bin or a mod­er­ate reduc­ len­ti­vi­rus vec­tor.
tion in trans­fu­sion vol­ume would be impactful. This may be the
case because of dif­fi­culty with vol­ume sta­tus, as was the case in
this patient; for patients with alloimmunization for whom obtain- Patients may desire a cura­tive intent ther­apy given the long-
ing RBC units is more dif­fi­cult; or for patients whose qual­ity of term impact on mor­bid­ity, mor­tal­ity, and qual­ity of life asso­ci­
life would improve by spend­ing less time at trans­fu­sion appoint­ ated with chronic trans­fu­sion ther­apy. Until recently, the only
ments. The risk of throm­bo­em­bo­lism should be con­sid­ered when cura­tive option was allo­ge­neic HSCT. Allogeneic HSCT has pro­
ini­ti­at­ing this med­i­ca­tion; although low in clin­i­cal tri­als, the inci­ vided a cure to many patients with TDT, with the best out­comes

Table 2. Luspatercept con­sid­er­ations1-6

Strengths Limitations
Subcutaneous admin­is­tra­tion Does not require intra­ve­nous access Requires infu­sion cen­ter visit every 3 weeks
Reduction of trans­fu­sion in TDT 21.4% had >33% reduc­tion Most patients with­out sig­nif­i­cant reduc­tion
7.6% had >50% reduc­tion Majority still have vis­its every 3 weeks
Increase in hemo­glo­bin in NTDT 52.1% had 1.5-g/dL increase Many patients with­out sig­nif­i­cant increase
Requires increased appoint­ment bur­den
Not yet approved by reg­u­la­tory agencies
Risk of throm­bo­em­bo­lism Rates low: 8 in 223 (3.6%) in trial Patients with thal­as­se­mia already at increased
risk of throm­bo­em­bo­lism, espe­cially if prior
sple­nec­tomy
Impact on iron load­ing Preliminary data showed lower fer­ri­tin and LIC Additional data needed to ver­ify find­ings
in some patients, includ­ing some who did
not meet pri­mary end point
Prakash Singh Shekhawat
Evolving ther­a­pies in β-thal­as­se­mia  |  601
Table 3. Gene ther­apy tri­als for β-thal­as­se­mia

602  |  Hematology 2021  |  ASH Education Program


Also in devel­op­ment for
Therapy Gene mod­i­fi­ca­tion tech­nique Genetic mech­a­nism Phase of devel­op­ment sickle cell dis­ease? Comments
Betibeglogene autotemcel Lentiviral vec­tor Insertion of mod­i­fied β-glo­bin Phase 3 tri­als Yes Reopened after tem­po­rary halt due
(beti-cel; for­merly to AML cases in sickle cell patients
BB305)8-11,17,18
GLOBE12 Lentiviral vec­tor Insertion of human β-glo­bin Phase 1/2 trial com­pleted No Intrabone admin­is­tra­tion
CTX00114,15 CRISPR-Cas9 Targets BCL11A enhancer → Phase 1/2 trial Yes
HbF induc­tion
ST-40016 ZFN Targets BCL11A enhancer → Phase 1/2 trial No
HbF induc­tion

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/600/1851627/600langer.pdf by guest on 13 December 2021


reported for matched sib­ling donors and recip­i­ents who under­ Case 2 dem­on­strates the clin­i­cal demand for cura­tive intent
went trans­plan­ta­tion in teen­age years.7 However, the risk of ther­apy and its poten­tial impact on qual­ity of life as well as
acute and chronic com­pli­ca­tions and the lack of avail­­able donors health. Although genet­i­cally mod­i­fied autol­o­gous HSCT may
have lim­ited its use. offer a more fea­si­ble and less toxic option for cura­tive intent
Ex vivo mod­i­fi­ca­tion of CD34+ hema­to­poi­etic stem cells using ther­apy com­pared with allo­ge­neic HSCT, lim­i­ta­tions are likely to
dif­fer­ent tech­niques has opened the pos­si­bil­ity of autol­o­gous remain. HSCT of any kind is a costly ther­apy and not likely to be
HSCT as a cura­tive ther­apy, thereby elim­i­nat­ing the need for a avail­­able in low-resource set­tings. In addi­tion to mon­e­tary costs,
donor as well as the risks spe­cific to allo­ge­neic HSCT, such as under­go­ing autol­o­gous HSCT requires a sig­nif­i­cant time com­
graft-ver­sus-host dis­ease. In this con­text, genet­i­cally mod­i­fied mit­ment and inter­rup­tion of other life events. The poten­tial for
autol­o­gous HSCT is an area of active research with sig­nif­i­cant reduced fer­til­ity or infer­til­ity due to expo­sure to myeloablative
advance­ments in the past few years. alkylator con­di­tion­ing is an impor­tant con­sid­er­ation for many
One ex vivo gene ther­apy, betibeglogene autotemcel (beti- patients. Finally, 2 cases of acute mye­ loid leu­
ke­
mia (AML) in
cel; for­merly LentiGlobin), uses a lentiviral vec­tor to add a mod­ patients with sickle cell dis­ease treated with beti-cel have raised

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/600/1851627/600langer.pdf by guest on 13 December 2021


i­fied β-glo­bin gene into CD34+ cells for infu­sion by autol­o­gous con­cerns.17,18 Evaluation sug­gests that nei­ther case was caused
HSCT after myeloablative con­di­tion­ing. In phase 1/2 tri­als, beti- by inser­tional muta­gen­e­sis. To date, no patients with thal­as­se­
cel treat­ment led to trans­fu­sion inde­pen­dence (TI) in 12 of 13 mia have devel­ oped myelodysplastic syn­ drome or AML after
non-β0/β0 geno­type and 3 of 9 β0/β0 geno­type patients with genet­i­cally mod­i­fied autol­o­gous HSCT, but addi­tional data will
TDT, or 68% over­all.8 The remaining patients had a reduc­tion in be nec­es­sary before final con­clu­sions may be drawn about what
trans­fu­sion vol­ume. Toxicity was pri­mar­ily related to busul­fan fac­tors influ­ence malig­nancy risk.
used for HSCT con­ di­tion­
ing. The orig­ i­
nal beti-cel trial estab-
lished the poten­tial for genet­i­cally mod­i­fied autol­o­gous HSCT,
although most patients with the β0/β0 geno­type did not achieve
TI. Preliminary data from 2 phase 3 tri­als of beti-cel for β0/β0
(NCT03207009) and non-β0/β0 (NCT02906202) patients have CLINICAL CASE 3: TARGETING ABNORMAL IRON
dem­on­strated TI at >12 months of fol­low-up in 88% of 34 eval- METABOLISM
uable patients, includ­ing 24 of 27 non-β0/β0 and 6 of 7 β0/β0 A 34-year-old man with NTDT is concerned about ongo­ing iron
patients, with no addi­tional toxicities noted.9 Preliminary reports over­load. His hemo­glo­bin aver­ages 8.4 g/dL, and he receives
of long-term fol­low up of 44 patients with >2 years of fol­low-up trans­fu­sion a few times per year. His LIC is esti­mated at 8 mg
after beti-cel treat­ment (23-76 months) showed a dura­ble hemo­ per gram of dry weight on mag­netic res­o­nance imag­ing. In
glo­bin response, reduc­tion in iron bur­den, and favor­able safety addi­tion to inten­si­fy­ing his iron che­la­tion, he inquires whether
pro­file.10 Initial eval­u­a­tion of 27 pedi­at­ric patients treated with there are any ther­a­pies that may raise his hemo­glo­bin or reduce
beti-cel, 16 of whom were under 12 years old, showed a com­pa­ his iron absorp­tion. He is open to par­tic­i­pa­tion in clin­i­cal tri­als.
ra­ble TI rate of 84.6% (Table 3).11
Another phase 1/2 trial uses a lentiviral vec­tor (the GLOBE
vec­tor) to add a β-glo­bin gene to autol­o­gous hema­to­poi­etic Currently, there are no established ther­a­pies to address this
stem cells, with intrabone admin­is­tra­tion of the trans­duced cells. patient’s ane­mia. However, sev­eral med­i­ca­tions in devel­op­ment
Three of 4 evaluable pedi­at­ric patients achieved TI, whereas 3 may improve ane­mia in patients with NTDT that also tar­get iron
adult patients had a reduced trans­fu­sion bur­den with­out achiev­ metab­o­lism. Apotransferrin has been shown to upregulate hep-
ing TI.12 At pres­ent, a sub­se­quent trial is not reg­is­tered. cidin and downregulate trans­ fer­
rin recep­ tor 1.19,20 This leads to
In con­trast to the gene addi­tion approach, another strat­egy decreased iron absorp­tion from the gut,19 as well as poten­tially less
for autol­o­gous genetic ther­a­pies for β-hemo­glo­bin­op­a­thies is to car­diac iron load­ing.21 The cor­rec­tion of path­o­logic iron metab­o­
decrease BCL11A expres­sion in order to induce expres­sion of γ- lism led to more effec­tive eryth­ro­poi­e­sis in mouse mod­els.20 These
glo­bin and hemo­glo­bin F (HbF). The first trial targeting BCL11A pre­clin­i­cal find­ings have spurred a phase 2 trial of intra­ve­nous apo-
uses a lentiviral vec­tor, BCH-BB694, to intro­duce short hair­pin transferrin every 2 weeks in patients with β-thal­as­se­mia intermedia
RNA to decrease BCL11A expres­sion.13 Six patients with sickle cell that seeks to raise hemo­glo­bin and reduce trans­fu­sion require­
dis­ease who received BCH-BB694 dem­on­strated safety and fea­ ments (NCT03993613).
si­bil­ity and had sub­stan­tial induc­tion of HbF. This vec­tor has not Similarly, a ferroportin inhib­i­tor (VIT-2763) has begun a phase 2
been eval­u­ated in patients with TDT. Trials are cur­rently open for trial (NCT04364269) after dem­on­strat­ing improve­ment in ane­mia
patients with thal­as­se­mia, in whom a gene editing tech­nique, and dysregulated iron metab­o­lism in a mouse model of thal­as­
either CRISPR-Cas9 or zinc-fin­ger nucle­ase (ZFN), is employed se­mia.22 Like the apotransferrin trial, the VIT-2753 is focused on
to dis­rupt an ery­throid enhancer of BCL11A. CTX001 is a CRISPR- cor­rec­tion of ane­mia but includes sec­ond­ary out­comes mea­sures
Cas9–mod­i­fied autol­o­gous HSCT prod­uct being inves­ti­gated of iron overloading. Notably, VIT-2753 is admin­is­tered orally. In
in TDT as well as sickle cell dis­ease (NCT03655678). Preliminary con­sid­er­ing treat­ment for patients with NTDT, this is par­tic­u­larly
results from the first 10 patients treated with CTX001 for TDT impactful for qual­ity of life, as many of these patients do not oth­
showed sub­stan­tial HbF induc­tion, broad dis­tri­bu­tion of HbF, er­wise have recur­ring infu­sion cen­ter appoint­ments (Table 4).
and achieve­ment of TI in all­patients.14,15 Toxicity was related to Hepcidin itself has also been a tar­get for drug devel­op­ment,
busul­fan con­di­tion­ing. A phase 1/2 study of ST-400, an autol­o­ because increas­ing hepcidin has been shown to both reduce
gous HSCT prod­uct in which the BCL11A enhancer is disrupted iron absorp­tion and ame­lio­rate inef­fec­tive eryth­ro­poi­e­sis.23 A
by a ZFN, is cur­rently under way (NCT03432364), but only very hepcidin mimetic, PTG-300, has com­pleted accrual for a phase 2
pre­lim­i­nary data on the first 2 patients have been reported.16 study, includ­ing both NTDT and TDT arms (NCT03802201). A
Prakash Singh Shekhawat
Evolving ther­a­pies in β-thal­as­se­mia  |  603
Table 4. Novel med­i­ca­tions for β-thal­as­se­mia

Route and fre­quency Current tar­get patient In devel­op­ment for


Medication Mechanism of action of admin­is­tra­tion Phase of devel­op­ment pop­u­la­tion sickle cell dis­ease? Comments
1,5
Luspatercept TGF-β inhib­i­tor Subcutaneous, every EMA and FDA approved NTDT, TDT No Full data for NTDT pend­ing;
3 weeks in use for MDS
Sotatercept36,37 TGF-β inhib­i­tor Subcutaneous, every Development halted NTDT, TDT No In devel­op­ment for pul­mo­nary
3 weeks hyper­ten­sion

604  |  Hematology 2021  |  ASH Education Program


Sirolimus25-28 mTOR inhib­i­tor; HbF Oral, daily Phase 2 tri­als TDT No In use for other dis­or­ders
induc­tion
Benserazide31,32 HbF induc­tion Oral, daily Phase 1 trial NTDT Yes In use for Parkinson dis­ease
IMR-68729,30 PDE-9 inhib­i­tor; HbF Oral, daily Phase 2 trial NTDT, TDT Yes
induc­tion
Apotransferrin19-21 Hepcidin upregulation Intravenous, every Phase 2 trial NTDT No
2 weeks
VIT-276322 Ferroportin inhib­i­tor Oral, one or twice daily Phase 2 trial NTDT Yes
PTG-30023 Hepcidin mimetic Subcutaneous, once Phase 2 tri­als NTDT, TDT No In devel­op­ment for
weekly hemo­chro­ma­to­sis and
poly­cy­the­mia vera
Mitapivat33-35 Pyruvate kinase acti­va­tor Oral, twice daily Phase 3 tri­als NTDT, TDT Yes In devel­op­ment for pyru­vate
kinase defi­ciency
Ruxolitinib38 JAK 1/2 inhib­i­tor Oral, twice daily Development halted TDT No Spleen size reduc­tion;
no change in trans­fu­sion
EMA, Euro­pean Medicines Agency; FDA, Food and Drug Administration; JAK, Janus-asso­ci­ated kinase; MDS, myelodysplastic syn­drome; mTOR, mech­a­nis­tic tar­get of rapamycin.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/600/1851627/600langer.pdf by guest on 13 December 2021


recent mouse study of an erythroferrone anti­ body show­ ing Case 4 is a reminder that even in well-man­aged patients,
improved ane­mia as well as iron homeo­sta­sis pro­vi­des another there is sig­nif­i­cant room for improve­ment in care. All 4 patients
poten­tial ther­a­peu­tic tar­get in the iron reg­u­la­tion path­way.24 presented high­light the nuanced needs of patients with thal­as­
Case 3 empha­sizes the large unmet need of ther­a­pies for se­mia and the impor­tance of tai­lor­ing ther­a­peu­tic choices to
NTDT and the appeal of a ther­apy that may also address non- the indi­vid­ual as new treat­ments emerge. Although the med­i­
transfusional hemosiderosis in these patients. These tri­als not ca­tions and inter­ven­tions reviewed here are grounded in strong
only high­light the role of altered iron metab­o­lism in inef­fec­tive pre­clin­i­cal research on thal­as­se­mia, the results of these tri­als are
eryth­ro­poi­e­sis but also pro­vide sev­eral prom­is­ing ther­a­peu­tic needed to under­stand which agents will be clin­i­cally impact-
tar­gets for ame­lio­rat­ing ane­mia. ful and which agents will not be fruit­ful for patient care. If early
stud­ies are borne out, it is likely that both patients with TDT
and NTDT will have sev­eral options beyond trans­fu­sion and iron
che­la­tion in the com­ing years.
CLINICAL CASE 4: FETAL HEMOGLOBIN INDUCTION
Conflict-of-inter­est dis­clo­sure

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/600/1851627/600langer.pdf by guest on 13 December 2021


AND OTHER MECHANISMS
Arielle L. Langer: no com­pet­ing finan­cial inter­ests to declare.
A 21-year-old woman with TDT and in good iron bal­ance recently Erica B. Esrick: steering com­ mit­tee (con­ sul­ting) for blue­bird
tried luspatercept with only a min­i­mal reduc­tion in trans­fu­sion bio and research funding to insti­tu­tion from Celgene (site for
bur­den. She inquires if there are other ther­ a­
pies that might luspatercept trial).
reduce her trans­fu­sion require­ment. She is not inter­ested in allo­
ge­neic or genet­i­cally mod­i­fied autol­o­gous HSCT, as she is wor­ Off-label drug use
ried about the upfront risk and dis­rup­tion in her life, because Arielle L. Langer: nothing to disclose. The majority of the drugs
she is doing quite well over­all on her cur­rent trans­fu­sion and discussed are not yet approved.
che­la­tion reg­i­men. Erica B. Esrick: nothing to disclose. The majority of the drugs
discussed are not yet approved.

Several agents cur­rently in clin­i­cal tri­als could pro­vide good Correspondence


options for this patient. Sirolimus, the mech­a­nis­tic tar­get of Erica B. Esrick, Division of Hematology/Oncology, Bos­ton Children’s
rapamycin inhib­i­tor, is being inves­ti­gated as a method of increas­ Hospital, 300 Longwood Avenue, Bos­ton, MA 02115; e-mail: erica​
ing HbF expres­sion. Sirolimus appears to upregulate the expres­ ­.esrick@chil­drens​­.harvard​­.edu.
sion of HbF in ery­throid cell cul­tures derived from patients with
β-thal­as­se­mia, as well as sickle cell patients, and may increase References
clear­ance of α-glo­bin in RBC pre­cur­sors.25-28 The lat­ter find­ing 1. Cappellini MD, Viprakasit V, Taher AT, et al; BELIEVE Investigators. A phase 3
may have the poten­tial to reduce inef­fec­tive eryth­ro­poi­e­sis sep­ trial of luspatercept in patients with trans­fu­sion-depen­dent β-thal­as­se­mia.
N Engl J Med. 2020;382(13):1219-1231.
a­rate from HbF induc­tion. In this con­text, 2 ongo­ing phase 2 tri­als
2. Taher AT, Porter JB, Hermine O, et al. Fewer red blood cell trans­fu­sion units
of sirolimus in patients with TDT (NCT03877809, NCT04247750) and vis­its across base­line trans­fu­sion bur­den lev­els in patients with beta-
will track mark­ers of inef­fec­tive eryth­ro­poi­e­sis in addi­tion to the thal­as­se­mia treated with luspatercept in the phase 3 believe trial. Paper
pri­mary end point of HbF induc­tion. presented at the 26th Congress of the Euro­pean Hematology Association
Another path­way for HbF induc­tion may be phos­pho­di­es­ter­ (EHA), June 9-17, 2021. Abstract 325024.
3. Piga A, Tartaglione I, Gamberini R, et  al. Long-term effi­cacy and safety
ase 9 inhi­bi­tion with IMR-687.29,30 A phase 2 trial of IMR-687 for both out­comes in the phase 2 study of luspatercept in Β-thal­as­se­mia. Paper
patients with TDT and NTDT is cur­rently under way (NCT04411082). presented at the 26th Congress of the Euro­pean Hematology Association
Benserazide, which is cur­rently used in com­bi­na­tion with levo­ (EHA), June 9-17, 2021. Abstract EP1304.
dopa for the treat­ment of Parkinson dis­ease, showed prom­is­ 4. Piga A, Perrotta S, Gamberini MR, et al. Luspatercept improves hemo­glo­
bin lev­els and blood trans­fu­sion require­ments in a study of patients with
ing induc­tion of HbF, although the study was not of thal­as­se­mia
β-thal­as­se­mia. Blood. 2019;133(12):1279-1289.
mod­els,31 but it did not induce HbF in patients on this med­i­ca­tion 5. Taher AT, Cappellini MD, Kattamis A, et al. The BEYOND study: results
for Parkinson dis­ease.32 A phase 1 trial of benserazide in patients of a phase 2, dou­ble-blind, ran­dom­ized, pla­cebo-con­trolled mul­ti­cen­ter
with NTDT is ongo­ing (NCT04432623). study of luspatercept in adult patients with non-tranfusion depen­dent
Mitapivat (for­merly AG-348), an allo­ste­ric acti­va­tor RBC-spe­cific beta-thal­as­se­mia. Paper presented at the 26th Congress of the Euro­pean
Hematology Association (EHA), June 9-17, 2021. Abstract S101.
pyru­ vate kinase, rep­ re­
sents a dis­ tinct and novel mech­ a­
nism. 6. Hermine O, Cappellini MD, Taher AT, et al. Longitudinal effect of luspater-
RBC-spe­cific pyru­vate kinase acti­va­tion increases aden­o­sine tri­ cept treat­ment on iron over­load and iron che­la­tion ther­apy (ICT) in adult
phos­phate syn­the­sis, as well as reduces the pro­duc­tion of reac­tive patients (Pts) with β-thal­as­se­mia in the believe trial. Blood. 2020;136(suppl 1):
oxy­gen spe­cies and con­cen­tra­tion of 2,3-diphosphoglycerate.33 47-48.
7. Baronciani D, Angelucci E, Potschger U, et al. Hemopoietic stem cell trans­
Preliminary results of a phase 2 trial of mitapivat in α and β NTDT
plan­ta­tion in thal­as­se­mia: a report from the Euro­pean Society for Blood
showed a response, defined as an increase in hemo­glo­bin of at and Bone Marrow Transplantation Hemoglobinopathy Registry, 2000-
least 1 g/dL in 16 of 20 patients, includ­ing all 5 patients with α- 2010. Bone Marrow Transplant. 2016;51(4):536-541.
thal­as­se­mia.34 Phase 3 stud­ies of mitapivat in TDT (NCT04770779) 8. Thompson AA, Walters MC, Kwiatkowski J, et al. Gene ther­apy in patients
and NTDT (NCT04770753) patients are under way.35 Like the phase with trans­fu­sion-depen­dent β-thal­as­se­mia. N Engl J Med. 2018;378(16):1479-
1493.
2 trial, these tri­als include patients with α-thal­as­se­mia, a patient 9. Locatelli F, Kwiatkowski JL, Walters MC, et al. Betibeglogene autotemcel in
pop­ u­la­
tion that is under­ served but out­ side the scope of this patients with trans­fu­sion-depen­dent β-thal­as­se­mia: updated results from
review. HGB-207 (NORTHSTAR-2) and HGB-212 (NORTHSTAR-3). Paper presented

Prakash Singh Shekhawat


Evolving ther­a­pies in β-thal­as­se­mia  |  605
at the 26th Congress of the Euro­pean Hematology Association (EHA), June 25. Fibach E, Bianchi N, Borgatti M, et al. Effects of rapamycin on accu­mu­la­
9-17, 2021. Abstract S266. tion of alpha-, beta- and gamma-glo­bin mRNAs in ery­throid pre­cur­sor cells
10. Yannaki E, Locatelli F, Kwiatkowski JL, et  al. Betibeglogene autotemcel from beta-thal­as­sae­mia patients. Eur J Haematol. 2006;77(5):437-441.
gene ther­apy for the treat­ment of trans­fu­sion-depen­dent β-thal­as­se­mia: 26. Mischiati C, Sereni A, Lampronti I, et al. Rapamycin-medi­ated induc­tion of
updated long-term effi­cacy and safety results. Paper presented at the 26th gamma-glo­bin mRNA accu­mu­la­tion in human ery­throid cells. Br J Haema-
Congress of the Euro­pean Hematology Association (EHA), June 9-17, 2021. tol. 2004;126(4):612-621.
Abstract S267. 27. Pecoraro A, Troia A, Calzolari R, et al. Efficacy of rapamycin as inducer of Hb
11. Kulozik AE, Thuret I, Kwiatkowski JL, et al. Interim results of betibeglogene F in pri­mary ery­throid cul­tures from sickle cell dis­ease and β-thal­as­se­mia
autotemcel gene ther­apy in pedi­at­ric patients with trans­fu­sion-depen­dent patients. Hemoglobin. 2015;39(4):225-229.
Β-thal­as­se­mia (TDT) treated in the phase 3 NORTHSTAR-2 and NORTHSTAR-3 28. Lechauve C, Keith J, Khandros E, et  al. The autophagy-acti­vat­ing kinase
stud­ies. Paper presented at the 26th Congress of the Euro­pean Hematology ULK1 medi­ates clear­ance of free α-glo­bin in β-thal­as­se­mia. Sci Transl Med.
Association (EHA), June 9-17, 2021. Abstract EP1301. 2019;11(506):eaav4881.
12. Marktel S, Scaramuzza S, Cicalese MP, et al. Intrabone hema­to­poi­etic stem 29. Almeida CB, Traina F, Lanaro C, et al. High expres­sion of the cGMP-spe­cific
cell gene ther­apy for adult and pedi­at­ric patients affected by trans­fu­sion- phos­pho­di­es­ter­ase, PDE9A, in sickle cell dis­ease (SCD) and the effects
depen­dent ß-thal­as­se­mia. Nat Med. 2019;25(2):234-241. of its inhi­bi­tion in ery­throid cells and SCD neu­tro­phils. Br J Haematol.
13. Esrick EB, Lehmann LE, Biffi A, et al. Post-tran­scrip­tional genetic silenc­ 2008;142(5):836-844.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/600/1851627/600langer.pdf by guest on 13 December 2021


ing of BCL11A to treat sickle cell dis­ease. N Engl J Med. 2021;384(3):205- 30. McArthur JG, Svenstrup N, Chen C, et al. A novel, highly potent and selec­
215. tive phos­pho­di­es­ter­ase-9 inhib­i­tor for the treat­ment of sickle cell dis­ease.
14. Frangoul H, Altshuler D, Cappellini MD, et al. CRISPR-Cas9 gene editing for Haematologica. 2020;105(3):623-631.
sickle cell dis­ease and β-thal­as­se­mia. N Engl J Med. 2021;384(3):252-260. 31. Pace BS, Perrine S, Li B, et  al. Benserazide race­mate and enan­tio­mers
15. Locatelli F, Ailinca-Luchian S, Bobruff Y, et al. CTX001 for trans­fu­sion-depen­dent induce fetal glo­bin gene expres­sion in vivo: stud­ies to guide clin­i­cal devel­
β-thal­as­se­mia: safety and effi­cacy results from the ongo­ing CLIMB THAL-111 op­ment for beta thal­as­se­mia and sickle cell dis­ease. Blood Cells Mol Dis.
study of autol­o­gous CRISPR-CAS9-mod­i­fied CD34+ hema­to­poi­etic stem and 2021;89:102561.
pro­gen­i­tor cells. Paper presented at the 26th Congress of the Euro­pean Hema- 32. Santos MEHP, Olops L, Vendrame F, et al. Benserazide as a poten­tial novel
tology Association (EHA), June 9-17, 2021. Abstract EP733. fetal hemo­glo­bin inducer: an obser­va­tional study in non-car­ri­ers of hemo­
16. Smith AR, Schiller GJ, Vercellotti GM, et al. Preliminary results of a phase 1/2 glo­bin dis­or­ders. Blood Cells Mol Dis. 2021;87:102511.
clin­i­cal study of zinc fin­ger nucle­ase-medi­ated editing of BCL11A in autol­ 33. Kung C, Hixon J, Kosinski PA, et al. AG-348 enhances pyru­vate kinase activ­
o­gous hema­to­poi­etic stem cells for trans­fu­sion-depen­dent beta thal­as­se­ ity in red blood cells from patients with pyru­vate kinase defi­ciency. Blood.
mia. Blood. 2019;134(suppl 1):3544-3544. 2017;130(11):1347-1356.
17. Hsieh MM, Bonner M, Pierciey FJ, et al. Myelodysplastic syn­drome unre­lated 34. Kuo KHM, Layton DM, Lal A, et al. Results from a phase 2, open-label, mul­ti­
to lentiviral vec­tor in a patient treated with gene ther­apy for sickle cell dis­ cen­ter study of the oral pyru­vate kinase activatorr mitapivat in adults with
ease. Blood Adv. 2020;4(9):2058-2063. non–trans­fu­sion-depen­dent alpha- or beta-thal­as­se­mia. Paper presented
18. blue­bird bio Provides updated find­ings from reported case of acute mye­loid at the 26th Congress of the Euro­pean Hematology Association (EHA), June
leu­ke­mia (AML) in LentiGlobin for Sickle Cell Disease (SCD) Gene Therapy 9-17, 2021. Abstract S267.
Program—blue­bird bio, Inc. Accessed 6 July 2021. 35. Kuo KHM, Layton DM, Al-Samkari H, et al. ENERGIZE and ENERGIZE-T: two
19. Gelderman MP, Baek JH, Yalamanoglu A, et al. Reversal of hemo­chro­ma­to­sis phase 3, ran­dom­ized, dou­ble-blind, pla­cebo-con­trolled stud­ies of mitapi-
by apotransferrin in non-trans­fused and trans­fused Hbbth3/+ (het­ero­zy­gous vat in adults with non–trans­fu­sion-depen­dent or trans­fu­sion-depen­dent
B1/B2 glo­bin gene dele­tion) mice. Haematologica. 2015;100(5):611-622. alpha- or beta-thal­as­se­mia. Paper presented at the 26th Congress of the
20. Li H, Choesang T, Bao W, et al. Decreasing TfR1 expres­sion reverses ane­mia Euro­pean Hematology Association (EHA), June 9-17, 2021. Abstract PB1805.
and hepcidin sup­pres­sion in β-thal­as­se­mic mice. Blood. 2017;129(11):1514- 36. Cappellini MD, Porter J, Origa R, et  al. Sotatercept, a novel transforming
1526. growth fac­tor β ligand trap, improves ane­mia in β-thal­as­se­mia: a phase II,
21. Garbowski MW, Evans P, Vlachodimitropoulou E, Hider R, Porter JB. open-label, dose-find­ing study. Haematologica. 2019;104(3):477-484.
Residual eryth­ro­poi­e­sis pro­tects against myo­car­dial hemosiderosis in 37. Humbert M, McLaughlin V, Gibbs JSR, et al; PULSAR Trial Investigators.
trans­fu­sion-depen­dent thal­as­se­mia by low­er­ing labile plasma iron via tran­ Sotatercept for the treat­ment of pul­mo­nary arte­rial hyper­ten­sion. N Engl J
sient gen­er­a­tion of apotransferrin. Haematologica. 2017;102(10):1640-1649. Med. 2021;384(13):1204-1215.
22. Manolova V, Nyffenegger N, Flace A, et al. Oral ferroportin inhib­i­tor ame­ 38. Taher AT, Karakas Z, Cassinerio E, et  al. Efficacy and safety of ruxolitinib
lio­rates inef­fec­tive eryth­ro­poi­e­sis in a model of β-thal­as­se­mia. J Clin Invest. in reg­u­larly trans­fused patients with thal­as­se­mia: results from a phase 2a
2019;130(1):491-506. study. Blood. 2018;131(2):263-265.
23. Schmidt PJ, Flem­ing MD. Modulation of hepcidin as ther­apy for pri­mary and
sec­ond­ary iron over­load dis­or­ders: pre­clin­i­cal mod­els and approaches.
Hematol Oncol Clin North Am. 2014;28(2):387-401.
24. Arezes J, Foy N, McHugh K, et  al. Antibodies against the erythroferrone
N-ter­mi­nal domain pre­vent hepcidin sup­pres­sion and ame­lio­rate murine © 2021 by The Amer­i­can Society of Hematology
thal­as­se­mia. Blood. 2020;135(8):547-557. DOI 10.1182/hema­tol­ogy.2021000313

606  |  Hematology 2021  |  ASH Education Program


THE CHANGING LANDSCAPE OF α - AND β -THALASSEMIA DIAGNOSIS AND TREATMENT

Optimal strategies for carrier screening


and prenatal diagnosis of α- and β-thalassemia

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/607/1851620/607mensah.pdf by guest on 13 December 2021


Cheryl Mensah1 and Sujit Sheth2
Division of Hematology and Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY; and 2Division of Hematology and Oncology,
1

Department of Pediatrics, Weill Cornell Medicine, New York, NY

The thalassemias are inherited quantitative disorders of hemoglobin synthesis with a significant worldwide burden, which
result in a wide spectrum of disease from the most severe transfusion-dependent form to the mildest asymptomatic
carrier state. In this article, we discuss the importance of carrier, prenatal, and newborn screening for thalassemia. We
examine the rationale for who should be screened and when, as well as the current methodology for screening. Deficien-
cies in the newborn screening program are highlighted as well. With the advent of inexpensive and rapid genetic testing,
this may be the most practical method of screening in the future, and we review the implications of population-based
implementation of this strategy. Finally, a case-based overview of the approach for individuals with the trait as well as
prospective parents who have a potential fetal risk of the disease is outlined.

LEARNING OBJECTIVES
• Understand the current guidelines and recommendations for screening and diagnosis of thalassemia syndromes

increasing awareness of the disease, with the potential for


CLINICAL CASE reducing transmission. Current newborn screening identi-
A 24-year-old woman of Thai descent is planning to start fes affected individuals with the more common, but not
a family with her partner, who is of Chinese descent. She all, severe α- and β-thalassemias, but most carriers may
recalls being told she is anemic all her life and has been on not be identifed in this manner. In time, with advances in
iron supplements off and on for many years, without correc- technology for genetic testing, the platform for screening
tion of her anemia. She recalls that her mother has the same may include direct gene analysis using diagnostic chips or
medical history. Her internist refers her to hematology for whole-genome or exome sequencing, providing a more
testing, and she is diagnosed with β-thalassemia trait. She complete genetic picture and allowing for more defnitive
is then referred to genetics with her partner (a recent immi- carrier identifcation. A brief overview of pathophysiology
grant who has never been tested) for testing and counsel- and a rational approach to testing and screening are exam-
ing regarding the risk of thalassemia in their offspring. ined based on the why, who, when, and how.

Pathophysiology
Introduction Several hundred mutations have been described in the
The thalassemias are a group of genetic diseases with a α- and β-globin gene clusters, and the resulting imbalance
high prevalence and signifcant morbidity. The broad range between the complementary globins results in intramed-
of clinical manifestations and complications, as well as ullary apoptosis of the maturing erythroid precursors and
high burden of disease, from the quality-of-life as well as leads to the hallmark ineffective erythropoiesis (IE).1 These
fnancial standpoint, underscores the importance of min- genotypes are summarized in Figure 1.
imizing its prevalence and optimizing outcomes in those Mutations or deletions of the α-globin genes result in
who are affected, through early diagnosis and compre- an excess of γ-globin in the fetus. When all 4 α-genes are
hensive care. Identifcation of individuals with mild disease deleted, HbF cannot be produced in the second trimes-
or carriers, prenatal screening, and counseling are key to ter and results in severe anemia and hydrops fetalis. When

Prakash Singh Shekhawat


Optimal strategies for thalassemia screening | 607
Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/607/1851620/607mensah.pdf by guest on 13 December 2021
Figure 1. Thalassemia syndromes. Adapted from Sheth and Thein.1

3 genes are deleted or mutated, there is for­ma­tion of tet­ra­mers of require reg­u­lar trans­fu­sions but may still develop man­i­fes­ta­tions
γ-glo­bin (hemo­glo­bin Barts [HbBarts]) ini­tially and β-glo­bin (HbH) sec­ond­ary to the IE such as extramedullary hema­to­poi­e­sis, bone
later. When both β-glo­bin genes are mutated, there is decreased changes, and vas­cu­lar dis­ease, also lead­ing to sig­nif­i­cant mor­
or absent HbA pro­duc­tion in the third tri­mes­ter and pro­gres­sive bid­ity.1,2 Treatment and mon­i­tor­ing are cum­ber­some, are expen­
ane­mia post­na­tally. The char­ac­ter­iza­tion of com­pound het­ero­zy­ sive, and sig­nif­i­cantly affect qual­ity of life. Thus, the thal­as­se­mia
gotes with β-thal­as­se­mia and HbE muta­tions leads to a sim­i­lar syn­dromes have a heavy bur­den of dis­ease and are good can­di­
pic­ture but with the addi­tional pres­ence of HbE. dates for screen­ing.
As a result of IE and ane­mia, clin­i­cal man­i­fes­ta­tions vary, with
the most severely affected indi­vid­u­als requir­ing life­long reg­u­ Scope of the prob­lem
lar trans­fu­sions and iron che­la­tion, with poten­tial for a vari­ety About 1.5% of the world’s pop­u­la­tion car­ries a thal­as­se­mia gene
of com­pli­ca­tions.1,2 Less severely affected indi­vid­u­als may not muta­tion, approx­i­mat­ing 80 to 90 mil­lion peo­ple world­wide

608  |  Hematology 2021  |  ASH Education Program


Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/607/1851620/607mensah.pdf by guest on 13 December 2021
Figure 2. Thalassemia distribution map showing prevalence of thalassemia syndromes.

(Figure 2).3,4 The car­rier state is found in Africa, around the Med­i­ ment of ane­mia, and start trans­fu­sions at the appro­pri­ate time,
ter­ra­nean, and the Middle East, but most affected indi­vid­u­als live min­i­miz­ing the risk for com­pli­ca­tions.
in the Indian sub­con­ti­nent, Southeast Asia, and China.1,3,4 How-
ever, chang­ing immi­gra­tion pat­terns have led to a world­wide
Who should be screened
dis­tri­bu­tion of the dis­ease.5
Individuals—based on demo­graph­ics and his­tory
Adults and chil­dren of Asian, Afri­can, or Med­i­ter­ra­nean ances­try,
Why we should screen
with a fam­ily his­tory of thal­as­se­mia, fam­ily his­tory of life­long ane­
Screening for thal­as­se­mia syn­dromes is impor­tant in 3 main set­
mia, and those with a his­tory of micro­cytic ane­mia unre­spon­sive
tings: for the cor­rect diag­no­sis and man­age­ment of ane­mia, pre­
to iron sup­ple­men­ta­tion or micro­cytic ane­mia with­out iron defi­
na­tal coun­sel­ing for par­ents at risk of hav­ing an affected child,
ciency, should be screened for thal­as­se­mia.6
and new­born screen­ing for early diag­no­sis.6
Thalassemia car­ri­ers are com­monly misdiagnosed with iron-
defi­ciency ane­mia and often receive prolonged courses of iron, Prospective par­ents—pre­na­tal screen­ing
occa­sion­ally par­en­ter­ally. Appropriate screen­ing would obvi- The Amer­i­can College of Obstetrics and Gynecology (ACOG)
ate unnec­es­sary use of iron in car­ri­ers and mon­i­tor­ing pre­vents rec­og­nizes that patients with Afri­can, Med­i­ter­ra­nean, and South-
sequelae and chronic com­pli­ca­tions in those with thal­as­se­mia east Asian ances­try are at increased risk for hemo­glo­bin­op­a­
intermedia. thies, includ­ing sickle cell and thal­as­se­mia.7 As a result of more
Prenatal screen­ing can be used to iden­tify adult car­ri­ers inter­ wide­spread mixing of pop­u­la­tions, lim­it­ing screen­ing to these
ested in starting fam­i­lies. For known car­ri­ers, screen­ing of part­ners ancestries may not be suf­fi­cient, and broader guide­lines are
leads to pre­na­tal risk assess­ment and fam­ily plan­ning coun­sel­ing. nec­es­sary.8
Pregnant women diag­nosed with thal­as­se­mia trait should have Prospective par­ents who are iden­ti­fied as thal­as­se­mia car­ri­
part­ner screen­ing to assess fetal risk, par­tic­u­larly for fetuses with ers should receive pre­na­tal genetic test­ing with coun­sel­ing. If
hydrops fetalis and still­birth, and receive edu­ca­tion and coun­sel­ one part­ner is con­firmed to be a car­rier, the other should be
ing.7 Earlier diag­no­sis of hydrops fetalis and still­birth can mit­i­gate exten­sively tested. Couples who are both thal­as­se­mia car­ri­ers
the men­tal and phys­i­cal con­se­quences for preg­nant moth­ers. can be offered in vitro fer­til­iza­tion and pre­im­plan­ta­tion genetic
Newborn screen­ing pro­grams allow early iden­ti­fi­ca­tion of diag­no­sis.7 For expec­tant par­ents, antepartum genetic test­ing
affected infants, who can be mon­i­tored closely for the devel­op­ can be offered prior to deliv­ery along with coun­sel­ing.

Prakash Singh Shekhawat


Optimal strat­e­gies for thal­as­se­mia screen­ing  |  609
Newborns less than the nor­mal range—approx­i­ma­tely 3% to 5%—and may
At-risk new­borns should be screened for thal­as­se­mia syn­dromes.6 be flagged as pos­si­ble β-thal­as­se­mia.6 Individuals with β0/HbE
However, this poses tech­ni­cal chal­lenges because the meth­od­ol­ may have no HbA and only have HbF and HbE, whereas those
ogy cur­rently used for new­born hemo­glo­bin­op­a­thy screen­ing (in with β+/HbE will have some HbA. HbA2, which is ele­vated in car­
all­50 states in the United States) is spe­cif­i­cally designed to detect ri­ers, is pro­duced in very small quan­ti­ties in the fetus (0.5%) and
sickle hemo­glo­bin. The detec­tion of HbBarts (tet­ra­mers of γ-glo­bin) is not typ­i­cally reported on the new­born screen.1,2
on the new­ born screen would facil­ i­
tate the early diag­ no­sis of
α-thal­as­se­mia trait or HbH dis­ease, but this requires hemo­glo­bin Newborn screen­ing in the United States. There is cur­rently no
quan­ti­ta­tion, not performed uni­ver­sally. The most severe form of stan­dard for new­born screen­ing across the United States, and
β-thal­as­se­mia, β0/β0-thal­as­
se­mia, could be diag­ nosed by the rec­om­men­da­tions for test­ing vary from state to state.12 In 2017,
absence of HbA on the new­born screen. Less severe forms of β-thal­ the Amer­ic ­ an Academy of Pediatrics reviewed new­born hemo­
as­se­mia could also be detected but not usu­ally the trait. In regions glo­bin­op­a­thy screen­ing across the 50 states.13 Thirty-one states
where thal­as­se­mia is more com­mon, par­tic­u­larly Asia, implement- recommended ane­mia screen­ing beyond rou­tine care or sickle

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/607/1851620/607mensah.pdf by guest on 13 December 2021


ing new­born screen­ing meth­od­ol­ogy to be ­able to detect thal­as­se­ cell screen­ing. Five of the 31 states recommended hemo­glo­bin
mia would be an impor­tant strat­egy for early diag­no­sis, coun­sel­ing, elec­tro­pho­re­sis and 4 recommended hema­tol­ogy refer­ral.13 Only
and reg­u­lar com­pre­hen­sive care where appro­pri­ate. 1 rec­om­mend ini­tial genetic test­ing. The other 19 states do not
have thal­as­se­mia screen­ing rec­om­men­da­tions beyond rou­tine
Infants care. Twelve of these 19 states reported α-thal­as­se­mia trait with
The Amer­i­can Academy of Pediatrics had a longstanding rec­om­ no addi­tional fol­low-up, and 7 did not report α-thal­as­se­mia or
men­da­tion for a screen­ing hemo­glo­bin/hemat­o­crit for all­ infants car­rier sta­tus at all­.13
at 9 months of age.9 To pre­vent iron defi­ciency, iron-for­ti­fied for­ Initial new­born screen­ing in most states is by high-per­for­mance
mula is now recommended, and screen­ing is done at 12 months of liq­uid chro­ma­tog­ra­phy (HPLC) or iso­elec­tric focus­ing (IEF) on a
age for for­mula-fed infants. Severe thal­as­se­mia, β0/β0-thal­as­se­mia, spot of blood. These tests detect hemo­glo­bin var­i­ants and may
usu­ally pres­ents well before this age with pro­gres­sive ane­mia and pro­vide lim­ited data to sup­port diag­no­sis of a thal­as­se­mia syn­
other clin­i­cal man­i­fes­ta­tions. However, more inter­me­di­ate forms, drome. HPLC, either by cat­ion exchange or reverse phase, quan-
includ­ing β+/β+-thal­as­se­mia, HbE/β0-thal­as­se­mia, or HbH dis­ease, tifies hemo­glo­bin var­i­ants such as hemo­glo­bin A, A2, F, S, H, and
may have few clin­i­cal man­i­fes­ta­tions besides mod­er­ate ane­mia and Barts.13,14 However, dif­fer­ent hemo­glo­bin var­i­ants may coelute,
may be missed until the 9- to 12-month hemo­glo­bin/hemat­o­crit mak­ing it dif­fi­cult to dis­tin­guish one var­i­ant from another. Thus,
is done. Screening these infants would be impor­tant to estab­ HPLC is used for ini­tial screen­ing and is followed by a con­fir­ma­tory
lish a diag­no­sis, pre­clude unnec­es­sary iron sup­ple­men­ta­tion, and test, either IEF or molec­u­lar test­ing.10 IEF is more labor inten­sive but
begin mon­i­tor­ing and com­pre­hen­sive care to opti­mize out­comes identifies and quantifies hemo­glo­bin var­i­ants more pre­cisely than
and min­i­mize mor­bid­ity. stan­dard hemo­glo­bin elec­tro­pho­re­sis.13 Molecular test­ing includes
poly­mer­ase chain reac­tion (PCR)–based test­ing or DNA sequenc­
How we screen ing.1 Allele-spe­cific PCR-based meth­ods are used to detect point
Newborn thal­as­se­mia screen­ing muta­tions in α- and β-glo­bin genes. Gap PCR is used to detect
With the high prev­a­lence and wide dis­tri­bu­tion of hemo­glo­bin­ dele­tions, α dupli­ca­tions, and deletional hemo­glo­bin var­i­ants.1
op­a­thies, test­ing for these was incor­po­rated into the new­born DNA sequenc­ing is more expen­sive and more com­pre­hen­sive and
screen.6 The nor­mal new­born at term has approx­i­ma­tely 80% can be used to diag­nose pre­vi­ously uniden­ti­fied muta­tions.1
to 90% HbF and 10% to 20% HbA, with trace amounts of other There is no nation­wide stan­dard of reporting HbBarts per­cent­
hemo­glo­bins such as HbA2, which are typ­i­cally not reported. age to pro­vid­ers, patients, or their fam­i­lies.10 A Centers for Dis-
ease Control and Prevention sur­vey found that HbBarts reporting
α-Thalassemia new­born screen­ing.  α-Thalassemia syn­dromes varies among new­born screen­ing pro­grams, and some pro­grams
com­pat­i­ble with life can be detected on new­born screen­ing. do not report quan­ti­ta­tive HbBarts or rec­om­mend fol­low-up.13
Fetuses with 3 α-gene dele­ tions or 2 α-gene dele­ tions and
Thalassemia screen­ing in chil­dren and adults
1 α-gene with a Constant Spring muta­tion (or other nondeletion-
Prospectively screen­ing all­patients for thal­as­se­mia is nei­ther cost-
al α muta­tion) will pro­duce excess unbound γ-glo­bin, which will
effec­tive nor indi­cated because iron defi­ciency is rel­a­tively much
result in the for­ma­tion of γ-tet­ra­mers, des­ig­nated HbBarts, iden­
more com­mon and explains a micro­cytic ane­mia. Establishing the
ti­fi­able on new­born screen­ing. The amount of HbBarts cor­re­lates
diag­no­sis of thal­as­se­mia trait is rel­e­vant, and avoid­ance of unnec­
with the sever­ity of α-thal­as­se­mia, although not in all­instances,
es­sary iron sup­ple­men­ta­tion is impor­tant. Individuals with an inter­
and can be detected antepartum and at birth.9,10 Newborns with
me­di­ate-sever­ity thal­as­se­mia syn­drome with IE have com­pli­ca­tions
2 α-gene dele­tions may have 3% to 6% HbBarts at birth, and a
that could be prevented if com­pre­hen­sive care is pro­vided early.
silent car­rier miss­ing only 1 α-gene may have 1% to 4% at birth.11
In the fol­low­ing sit­u­a­tions, screen­ing for thal­as­se­mia is
recommended:
β-Thalassemia new­born screen­ing. Fetuses with β0/β0 muta­
tions will pro­duce no HbA and will have only HbF on the new­ a. Family his­tory of thal­as­se­mia—trait or dis­ease
born screen, only oth­er­wise seen in extreme pre­ma­tu­rity, where b. Microcytic ane­mia with a neg­a­tive his­tory for iron defi­ciency—
β-glo­bin expres­sion has not begun yet. Thus, ges­ta­tional age is ade­quate die­tary iron intake, absence of blood loss
an impor­tant con­sid­er­ation, and nor­mal ranges should be estab- c. Persistent micro­ cytic ane­ mia despite an ade­ quate trial of
lished at var­i­ous ges­ta­tional ages for eas­ier inter­pre­ta­tion. Fe- sup­ple­men­tal iron
tuses with 1 β0-muta­tion and 1 β+-muta­tion or 2 β+-muta­tions will Testing of such indi­vid­u­als can be approached in a step­
have some HbA on the new­born screen. If this is quan­ti­fied, it is wise man­ner (Figure 3).

610  |  Hematology 2021  |  ASH Education Program


Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/607/1851620/607mensah.pdf by guest on 13 December 2021

Figure 3. Ideal algorithm for thalassemia screening, developed to screen for thalassemia syndromes in adults based on history,
physical examination, and laboratory parameters.

Prenatal screen­ing deter­min­ing fetal risk and is based on hemo­glo­bin elec­tro­pho­


ACOG rec­om­mends that women with a low mean corpuscular re­sis or genetic test­ing as appro­pri­ate for β- and α-thal­as­se­mia,
volume (MCV) should have serum fer­ri­tin assess­ment. Those with respec­tively.8 Couples who are both iden­ti­fied as car­ri­ers should
nor­mal lev­els and micro­cytic ane­mia should have hemo­glo­bin elec­ receive genetic coun­ sel­
ing to deter­ mine risk of an affected
tro­pho­re­sis test­ing.7 For patients with a nor­mal hemo­glo­bin elec­ fetus, and fam­ily deci­sion sup­port should be pro­vided. The most
tro­pho­re­sis and Asian ances­try, genetic test­ing for α-thal­as­se­mia severe form of α-thal­as­se­mia—dele­tion of all 4 α-genes—results
is recommended. In each sit­ u­
a­tion,Prakash
part­
ner test­
ing is key
Singh Shekhawat to in hydrops fetalis, with severe ane­mia (hemo­glo­bin rang­ing from

Optimal strat­e­gies for thal­as­se­mia screen­ing  |  611


3-8 g/dL), organomegaly, and edema, usu­ally lead­ing to intra­uter­ genetic test­ing becomes more widely avail­­able, tech­ni­cally
ine fetal demise from heart fail­ure, with pre­term labor, still­birth, eas­ier to per­form, and cheaper, mov­ing test­ing to a genome/
and a det­ri­men­tal effect on mater­nal health.1,11 Prenatal screen­ing exome or chip tech­nol­ogy would be most effec­tive in defin­
would iden­tify the at-risk fetus, and amnio­cen­te­sis or chorionic vil­ i­tively iden­ti­fy­ing indi­vid­u­als with thal­as­se­mia. We rec­og­
lus sam­pling would con­firm the diag­no­sis.11 In a suspected preg­ nize this may be a future-ori­ ented goal and a not widely
nancy, Dopp­ler assess­ments of cere­bral ves­sel flow veloc­i­ties or fea­si­ble approach. Until uni­ver­sal genetic test­ing can be per-
direct fetal blood sam­pling to quan­tify the ane­mia could be used. formed, we rec­om­mend the opti­mal strat­egy of performing
If intra­uter­ine trans­fu­sions are insti­tuted in time, such fetuses a hemo­ glo­
bin elec­ tro­
pho­ re­sis, HPLC, or IEF on all­infants;
may sur­vive but would be trans­fu­sion depen­dent post­na­tally— reporting the hemo­glo­bin var­i­ants, espe­cially HbBarts, and
des­ig­nated α-thal­as­se­mia major.11 unsta­ble hemo­glo­bin var­i­ants; and quan­ti­fy­ing HbA. Additions
It is impor­tant to keep in mind that 2 α-gene dele­tions are not to the new­born screen are jus­ti­fied if a treat­ment intro­duced
uncom­mon in Afri­can ances­try indi­vid­u­als, but these muta­tions early would reduce mor­bid­ity and mor­tal­ity. For thal­as­se­mia,
tend to occur on oppo­site chro­mo­somes (trans con­fig­u­ra­tion), this remains con­tro­ver­sial, and pro­spec­tive data dem­on­strat­

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/607/1851620/607mensah.pdf by guest on 13 December 2021


and hence the risk of hav­ing a fetus with all 4 genes deleted is ing the ben­e­fits of this approach could pro­vide a ratio­nale.
lower than in indi­vid­u­als with East Asian ances­try, in whom the . We rec­om­mend screen­ing all­ indi­vid­u­als (adults and chil­dren)
4
2 gene dele­tions are more com­monly on the same chro­mo­some with appro­pri­ate his­to­ries, cer­tain phys­i­cal exam­i­na­tion find­
(cis con­fig­u­ra­tion). For women who are iden­ti­fied as hav­ing ings, and micro­cytic ane­mia for thal­as­se­mia using the algo­
β-thal­as­se­mia trait or HbS trait, part­ner test­ing of hemo­glo­bin rithm in Figure 3.
frac­tion­ation is imper­a­tive to assess fetal risk for β-thal­as­se­mia
or sickle cell dis­ease (HbS-β thal).1

Preimplantation genetic test­ing.  Preconception, ACOG rec­om­


mends genetic test­ing and coun­sel­ing for cou­ples at high risk for CLINICAL CASE (Con­tin­ued)
thal­as­se­mia as noted.7 For cou­ples inter­ested in avoiding elec­ The cou­ple is at risk for an affected child given that the pro­
tive ter­mi­na­tion, in vitro fer­til­iza­tion and pre­im­plan­ta­tion genetic spec­tive mother has β-thal­as­se­mia trait, and the pro­spec­tive
diag­no­sis can be offered.7 For cou­ples who were not diagnosed father is of Chi­nese descent. He should have car­rier test­ing for
as high risk for thalassemia prior to preg­nancy, DNA test­ing via β-thal­as­se­mia trait. If he is a car­rier, then the poten­tial fetus is
chronic vil­lus sam­pling or amnio­cen­te­sis should be offered. Coun- at risk of hav­ing the dis­ease. Preimplantation diag­no­sis should
seling should be offered in all­such sit­u­a­tions.7 This approach has be offered before con­cep­tion, or test­ing of the fetus should be
resulted in a major reduc­tion in the birth rate of infants with thal­as­ offered early in the preg­nancy. Some fam­i­lies may choose nei­
se­mia through­out the Med­i­ter­ra­nean region and the Middle East, ther option.
as well as in parts of the Indian sub­con­ti­nent and Southeast Asia.
Novel approaches con­tinue to be explored in an attempt to avoid
the use of inva­sive pro­ce­dures such as chronic vil­lus sam­pling and Conclusion
amnio­cen­te­sis, includ­ing harvesting fetal DNA from fetal cells in Due to increas­ingly chang­ing demo­graph­ics and migra­tion from
mater­nal blood or mater­nal plasma.15,16 areas where thal­as­se­mia is more prev­a­lent to those where it is less
so, the prev­a­lence of thal­as­se­mia is chang­ing. It has become an
Suggested approach for screen­ ing of indi­
vid­
u­als at risk, all­ impor­tant health issue to iden­tify patients with thal­as­se­mia syn­
ages.  As the demo­graph­ics con­tinue to change glob­ally and in dromes and thal­as­se­mia trait to be ­able to not only pro­vide early
the United States, a com­pre­hen­sive approach for early diag­no­sis com­pre­hen­sive care but also avoid unnec­es­sary inter­ven­tions.
will improve the health of patients and min­i­mize risk. We sug­gest Early diag­no­sis can pre­pare at-risk cou­ples, as well as iden­tify
the fol­low­ing, starting pre­con­cep­tion and through adult­hood: at-risk fetuses and new­borns. Newborn screen­ing for hemo­glo­bin
1. Screening and iden­ti­fi­ca­tion of thal­as­se­mia car­rier sta­tus and dis­or­ders, pioneered with a sickle cell focus, has come a long way,
coun­sel­ing early for all­pro­spec­tive moth­ers at risk based but there is still room for advances to achieve the goal of opti­mal
on eth­nic­ity, fam­ily his­tory, med­i­cal his­tory, and lab­o­ra­tory screen­ing for thal­as­se­mia and estab­lish­ment of an early diag­no­sis
param­e­ters. Confirmatory genetic test­ing of both part­ners to opti­mize man­age­ment.
and genetic coun­sel­ing must be offered and, if avail­­able, pre­
im­plan­ta­tion genetic diag­no­sis when appro­pri­ate. Acknowledgments
2. If a preg­nancy has already occurred in a mother at risk, the C.M and S.S. have received funding from the Centers for Disease
same screen­ing should be done, and if thal­as­se­mia trait is Control and Prevention and the Health Resources and Services
con­firmed as described pre­vi­ously, part­ner test­ing and ge- Administration.
netic coun­sel­ing are indi­cated. Testing of the fetus should be
offered if both par­ents are car­ri­ers or if the father’s sta­tus can­ Conflict-of-inter­est dis­clo­sure
not be ascertained. More advanced tech­niques such as fetal Cheryl Mensah has served as a consultant to Bluebird bio and
DNA in mater­nal blood need to be tested more exten­sively to Chiesi. She currently serves on adjudication committee for CRISPR/
avoid more inva­sive test­ing pro­ce­dures. If the fetus is found Vertex for thalassemia.
to be affected, appro­pri­ate coun­sel­ing should be pro­vided. Sujit Sheth is a con­sul­tant to Agios, Bluebird bio, Bristol Myers
3. Newborn screen­ing should be stan­dard­ized to include hemo­ Squibb, and Chiesi and serves on a clin­i­cal trial steering com­mit­
glo­bin char­ac­ter­iza­tion and frac­tion­ation for all­. Ideally, as tee for CRISPR/Vertex CTX001 for thal­as­se­mia.

612  |  Hematology 2021  |  ASH Education Program


Off-label drug use 8. Shang X, Xu X. Update in the genet­ics of thal­as­se­mia: what cli­ni­cians need
to know. Best Pract Res Clin Obstet Gynaecol. 2017;39:3-15.
Cheryl Mensah: nothing to disclose.
9. Kohli-Kumar M. Screening for ane­mia in chil­dren: AAP rec­om­men­da­tions—a
Sujit Sheth: nothing to disclose. cri­tique. Pediatrics. 2001;108(3):e56.
10. Bender M, Yusuf C, Davis T, et al. Newborn screen­ing prac­tices and alpha-
Correspondence thal­as­se­mia detec­tion—United States, 2016. MMWR Morb Mortal Wkly
Sujit Sheth, Division of Pediatric Hematology Oncology, Weill Rep. 2020;69:1269–1272.
11. Vichinsky EP. Alpha thal­as­se­mia major—new muta­tions, intra­uter­ine man­
Cornell Medical College, 525 E 68th Street, P695, New York, NY
age­ment, and out­comes. Hematology Am Soc Hematol Educ Program.
10065; e-mail: shethsu@med​­.cornell​­.edu. 2009;2009(1):35–41.
12. Benson JM, Therrell BL. History and cur­rent sta­tus of new­born screen­ing for
References hemo­glo­bin­op­a­thies. Semin Perinatol. 2010;34(2):134-144.
1. Sheth S, Thein S. Thalassemia: a dis­or­der of glo­bin syn­the­sis. In: Kaushan- 13. Fogel BN, Nguyen HLT, Smink G, Sekhar DL. Variability in state-based rec­
sky K, Prchal JT, Burns LJ, Lichtman MA, Levi M, Linch DC, eds. Williams om­men­da­tions for man­age­ment of alpha thal­as­se­mia trait and silent car­rier
Hematology. 10th ed. McGraw-Hill; 2021. detected on the new­born screen. J Pediatr. 2018;195(2, pt 2):283-287.
2. Taher AT, Weatherall DJ, Cappellini MD. Thalassaemia. Lancet. 2018;391(10116): 14. Hoppe CC. Newborn screen­ing for non-sick­ling hemo­glo­bin­op­a­thies.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/607/1851620/607mensah.pdf by guest on 13 December 2021


155-167. Hematology Am Soc Hematol Educ Program. 2009;2009(1):19-25.
3. Modell B, Darlison M. Global epi­de­mi­­ol­ogy of haemoglobin dis­or­ders and 15. Cheung M-C, Goldberg JD, Kan YW. Prenatal diag­no­sis of sickle cell ane­
derived ser­vice indi­ca­tors. Bull World Health Organ. 2008;86(6):480-487. mia and thal­as­se­mia by anal­y­sis of fetal cells in mater­nal blood. Nat Genet.
4. Williams TN, Weatherall DJ. World dis­tri­bu­tion, pop­u­la­tion genet­ics, and 1996;14:264-268.
health bur­den of the hemo­glo­bin­op­a­thies. Cold Spring Harb Perspect Med. 16. Hung ECW, Chiu RWK, Lo YMD. Detection of cir­cu­lat­ing fetal nucleic acids:
2012;2(9):a011692. a review of meth­ods and appli­ca­tions. J Clin Pathol. 2009;62(4):308-313.
5. Sayani FA, Kwiatkowski JL. Increasing prev­a­lence of thal­as­se­mia in America:
impli­ca­tions for pri­mary care. Ann Med. 2015;47(7):592-604.
6. Centers for Disease Control and Prevention. Hemoglobinopathies: Current
Practices for Screening, Confirmation and Follow-up. Association of Public
Health Laboratories; 2015.
7. ACOG Committee on Obstetrics. ACOG Practice Bulletin No. 78: hemo­glo­ © 2021 by The Amer­i­can Society of Hematology
bin­op­a­thies in preg­nancy. Obstet Gynecol. 2007;109(1):229-237. DOI 10.1182/hema­tol­ogy.2021000296

Prakash Singh Shekhawat


Optimal strat­e­gies for thal­as­se­mia screen­ing  |  613
THE COVID CRASH: LESSONS LEARNED FROM A WORLD ON PAUSE

How to recognize and manage


COVID-19-associated coagulopathy

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/614/1851465/614gerber.pdf by guest on 13 December 2021


Gloria F. Gerber and Shruti Chaturvedi
Division of Hematology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD

COVID-19 is frequently associated with abnormalities on coagulation testing and a coagulopathy driven by inflamma-
tion, intravascular coagulation activation, and microvascular thrombosis. Elevated D-dimer is the most common finding
and is a predictor of adverse outcomes including thrombosis, critical illness, and death. Although COVID-19-associated
coagulopathy has some similarities to disseminated intravascular coagulation, the platelet count is usually preserved,
coagulation times are usually normal or minimally prolonged, and thrombosis is more common than bleeding, at least
in noncritically ill patients. Bleeding is uncommon but may be a significant problem in critically ill patients, includ-
ing those who may develop a consumptive coagulopathy with frank disseminated intravascular coagulation and those
on extracorporeal membrane oxygenation. Blood product support to correct coagulopathy is reserved for bleeding
patients or those requiring invasive procedures. Current recommendations suggest that all hospitalized patients should
receive at least a prophylactic dose of anticoagulation. Results from a multiplatform randomized clinical trial suggest
that therapeutically dosed anticoagulation may improve outcomes, including the need for organ support and mortality
in moderately ill patients but not in those requiring critical care. The results of ongoing trials evaluating the impact of
different antithrombotic strategies (therapeutic agents and intensity) on COVID-19 outcomes are eagerly awaited and
are expected to have important implications for patient management. We also discuss COVID-19 vaccine-associated
cytopenias and bleeding as well as vaccine-induced thrombotic thrombocytopenia, in which thrombosis is associated
with thrombocytopenia, elevated D-dimer, and, frequently, hypofibrinogenemia.

LEARNING OBJECTIVES
• Recognize the coagulation derangements commonly encountered in COVID-19 associated with outcomes includ-
ing thrombosis, need for critical care support, and mortality
• Appropriately monitor and manage coagulation abnormalities in hospitalized patients with COVID-19
• Recognize and manage the rare complications of COVID-19 vaccination-associated thrombocytopenia with bleed-
ing and vaccine-induced thrombotic thrombocytopenia (VITT)

Introduction cause mortality.5,6 In critically ill patients, this coagulopathy


The COVID-19 pandemic has contributed to significant may evolve into frank DIC. While less common than labora-
mortality and morbidity across the world. Despite unprec- tory coagulopathy or thrombosis, bleeding occurs in up to
edented public health efforts including the development of 5% of hospitalized patients and has important implications
multiple effective vaccines, the pandemic continues to rage for management.3 In this review we discuss our approach
in many parts of the world, with novel viral variants leading to COVID-19-associated coagulopathy (CAC) and bleeding.
to recurrent outbreaks. While COVID-19 is a predominantly
respiratory illness, hemostatic and thrombotic complica-
tions are common.1,2 Early in the pandemic, a high preva-
lence of thromboembolic complications was observed in CLINICAL CASE
patients with COVID-19.3,4 Many patients exhibit laboratory A 51-year-old woman with hypertension and asthma pre-
evidence of a coagulopathy reminiscent of, but not identi- sented with fever, cough, and dyspnea; polymerase chain
cal to, disseminated intravascular coagulation (DIC), which reaction for SARS-CoV-2 was positive, and she was hospital-
is associated with thrombosis risk, critical illness, and all- ized due to the need for supplemental oxygen. Laboratory

614 | Hematology 2021 | ASH Education Program


Table 1.  Similarities and dif­fer­ences between CAC the fact that intra­vas­cu­lar coag­u­la­tion acti­va­tion and micro­vas­cu­
and DIC/SIC lar throm­bo­sis are rel­a­tively local­ized to the pul­mo­nary cir­cu­la­tion
in CAC versus the more sys­temic acti­va­tion of coag­u­la­tion in DIC.
CAC DIC SIC Additionally, fibri­no­ly­sis is suppressed in CAC, which may con­trib­
Platelet count ↑↓ ↓↓ ↓ ute to the prothrombotic phe­no­type.11,12 Iba et al have pro­posed
diag­nos­tic cri­te­ria for CAC (Table 2).13
D-dimer ↑ ↑↑ ↑
Thrombocytopenia and PT pro­lon­ga­tion are less com­mon in
PT/aPTT ↔↑ ↑↑ ↑ COVID-19 but pre­dict poor clin­i­cal out­comes. In an anal­y­sis of 201
Fibrinogen ↑ ↓↓ ↓ hos­pi­tal­ized patients from China, prolonged PT on admis­sion was
asso­ci­ated with the devel­op­ment of acute respi­ra­tory dis­tress
Antithrombin ↔ ↓ ↓
syn­drome (ARDS; haz­ard ratio, 1.56 [1.32-1.83]; P < .001).14 Inter-
VWF ↑ ↑ ↑ estingly, throm­bo­cy­to­pe­nia was pres­ent in only 18.8% of these
FVIII ↑ ↑↓ ↑ patients com­pared to >50% in patients with ARDS from other

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/614/1851465/614gerber.pdf by guest on 13 December 2021


LA/aPL + - - infec­tious causes. Compared to non-COVID ARDS, COVID-19 is
asso­ci­ated with higher PT, anti­throm­bin, fibrin­o­gen, and plate­let
Thrombosis ↑↑ ↑ ↑
count and lower aPTT and D-dimer lev­els.15 In a meta-anal­y­sis of 9
Bleeding ↔↑ ↑↑ ↑ stud­ies includ­ing 1779 COVID-19 patients, the plate­let count was
sig­nif­i­cantly lower in patients with severe dis­ease and patients
who died.16 Similarly, a marked decrease in plasma fibrin­og ­ en has
test­ing on admis­sion revealed ele­vated dimerized plas­min frag­ been reported shortly before death in a num­ber of patients.5 An
ment D (D-dimer; 1.4 mg/L [0.00-0.49]) and fibrin­o­gen (345 mg/dL). early study by Tang et al noted a higher prev­a­lence of DIC by
Platelet count (238  ×  109/L) and pro­throm­bin time (PT; 10.8 sec­ International Society on Thrombosis and Haemostasis (ISTH) cri­
onds) were nor­mal. Activated par­tial throm­bo­plas­tin time (aPTT) te­ria on admis­sion in nonsurvivors vs sur­vi­vors (71.4% vs 0.6%).
was prolonged (43 sec­onds [23.1-30.9]). However, sub­se­quent stud­ies report a much lower inci­dence
of DIC at ini­tial pre­sen­ta­tion. For exam­ple, none of the 150 con­
sec­u­tive patients with COVID-19 and ARDS and only 2.2% of 388
CAC con­sec­u­tive patients hos­pi­tal­ized with COVID-19 in Milan met the
Laboratory find­ings in CAC cri­te­ria for DIC at pre­sen­ta­tion.15,17 Thus, overt DIC is uncom­mon
Coagulation derange­ments suggesting hyper­co­ag­u­la­bil­ity are in COVID-19 but may develop in crit­i­cally ill patients.3
com­mon in COVID-19. Markedly ele­vated D-dimer is the most com­ Based on the spec­trum of coag­u­la­tion abnor­mal­i­ties in COVID-
mon find­ing and is asso­ci­ated with adverse out­comes, includ­ing 19 that par­al­lels dis­ease sever­ity, three stages of COVID-19 coagu-
throm­bo­sis, the need for crit­i­cal care sup­port, and over­all mor­tal­ lopathy have been pro­posed (Figure 1).18 In stage 1, patients exhibit
ity.3,5,7 Elevated D-dimer lev­els cor­re­late with lev­els of inflam­ma­tory mild symp­toms and mild local­ized inflam­ma­tion and ele­vated D-
mark­ers, includ­ing C-reac­tive pro­tein, sed­i­men­ta­tion rate, procal- dimer. Stage 2 is char­ac­ter­ized by more severe symp­toms requir­
citonin, and fer­ri­tin, supporting an asso­ci­a­tion with the inflam­ ing sup­ple­men­tal oxy­gen, with pul­mo­nary inflam­ma­tion and intra­
ma­tory state of COVID-19.3 However, fibrin­o­gen lev­els are usu­ally vas­cu­lar coag­u­la­tion acti­va­tion (ele­vated D-dimer, fibrin­o­gen).
ele­vated/nor­mal, a plate­let count <100  ×  109/L is uncom­mon,3,8 and Patients who prog­ress to stage 3 require crit­i­cal care sup­port and
the PT is nor­mal at pre­sen­ta­tion in the major­ity of patients. Thus, dem­on­strate severe sys­temic inflam­ma­tion and coagulopathy
in most cases CAC is dis­tinct from the pro­to­typ­i­cal con­sump­tive with mark­edly ele­vated D-dimer and fibrin­o­gen, prolonged PT,
coagulopathies, such as sep­sis-induced coagulopathy (SIC) and and throm­bo­cy­to­pe­nia. A sub­set of these patients devel­ops overt
DIC (Table 1).9,10 Moreover, CAC is less likely to be asso­ci­ated with DIC that may be due to COVID-19 or bac­te­rial super­in­fec­tion and
bleed­ing than throm­bo­sis. These dif­fer­ences are prob­a­bly due to is asso­ci­ated with a high risk of bleed­ing and death.

Table 2.  Proposed cri­te­ria for CAC com­pared with ISTH cri­te­ria for SIC and DIC

Proposed CAC cri­te­ria13 ISTH SIC cri­te­ria10 ISTH DIC cri­te­ria9


≥2 of the fol­low­ing: ≥4 points: ≥5 points:
1) Platelet count <150 × 109/L 1) Platelet count, × 109/L 1) Platelet count, × 109/L
2) D-dimer ele­va­tion >2 × ULN   <150 but ≥100 = 1  <100 = 1
3) PT prolonged >1s or INR >1.2   <100  =  2  <50 = 2
4) Presence of micro and/or macrovascular thrombosis 2) INR 2) D-dimer
**Risk fac­tors for the devel­op­ment of CAC include ele­vated fibrin­o­gen and    >1.2 but ≤1.4 = 1     increased <5 × ULN = 2
VWF and pres­ence of lupus anti­co­ag­u­lant or antiphospholipid antibodies   >1.4 = 2    ≥5 × ULN = 3
3) SOFA score 3) PT
  1 = 1    prolonged ≥3 s but
  ≥2 = 2 <6 s = 1
   prolonged ≥6 s = 2
4) Fibrinogen
   ≤1.0 g/L = 1
INR, international normalized ratio; SOFA, Sequential Organ Failure Assessment; ULN, upper limit of nor­mal.
Prakash Singh Shekhawat
COVID-19 coagulopathy | 615
Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/614/1851465/614gerber.pdf by guest on 13 December 2021
Figure 1.  Stages of CAC. The spectrum of coagulation abnormalities in COVID-19 parallels disease severity. In stage 1, patients exhibit
mild symptoms and evidence of mild localized inflammation and coagulopathy with elevations in D-dimer approximately 2 to 3 times
the upper limit of normal. Stage 2 is characterized by more severe symptoms requiring supplemental oxygen with pulmonary inflam-
mation and intravascular coagulation activation in the lungs as well as some systemic involvement. This is reflected in further D-dimer
elevation to 3 to 6 times the upper limit of normal. Patients progressing to stage 3 require critical care with respiratory and other
organ support and demonstrate severe systemic inflammation and coagulopathy with markedly elevated D-dimer and fibrinogen,
prolonged PT, thrombocytopenia, and high incidence of thromboembolism. A proportion of these patients will develop overt DIC as
a result of their COVID-19 illness or other superimposed infections.18

Antiphospholipid antibodies in COVID-19 Other coag­u­la­tion param­e­ters


The aPTT is prolonged in some patients with COVID-19. A like- In addi­tion to the com­monly eval­u­ated lab­o­ra­tory param­e­ters
ly expla­na­tion is the pres­ence of a lupus anti­co­ag­u­lant (LA) or discussed above, ele­vated coag­u­la­tion fac­tor VIII (FVIII) and von
antiphospholipid antibodies (aPL). In a series of 56 hos­pi­tal­ized Willebrand fac­ tor (VWF) are fre­ quent find­ ings in hos­pi­tal­
ized
patients with COVID-19, LA was detected in 44.6%; 8.9% also COVID-19 patients and cor­re­late with ele­va­tions in inflam­ma­tory
had either anticardiolipin (aCL) or anti-beta-2-gly­co­pro­tein anti- mark­ers.15,21-23 Nonsurvivors have lower anti­throm­bin lev­els than
bodies.19 Both tra­di­tional aPL (such as immu­no­glob­u­lin [Ig] G and sur­vi­vors (84% vs 91%), but lev­els gen­er­ally do not drop below
IgM aCL) and var­io ­ us “noncriteria” aPL (includ­ing anti-PS/PT as 80%.5,15,22 In a study com­par­ing 48 inten­sive care unit (ICU) to
well as IgA isotypes of aCL and anti-beta-2-gly­co­pro­tein) have 30 gen­eral ward patients, fibrinolytic pathway parameters, includ­
been detected in COVID-19. Whether these are tran­sient aPL, as ing plas­min­o­gen acti­va­tor inhib­i­tor, tis­sue plas­min­o­gen acti­va­tor,
reported in other viral infec­tions,20 or per­sis­tent aPL, more likely and throm­bin activatable fibri­no­ly­sis inhib­i­tor, were sig­nif­i­cantly
to be asso­ci­ated with long-term throm­botic risk, is unknown. higher in the ICU patients.22 Viscoelastic test­ing (thromboelas-
Moreover, LA assays must be interpreted with cau­tion in crit­i­ tography, rotational thromboelastometry) reveals increased clot
cally ill patients due to poten­tial confounding by anti­co­ag­u­lant strength with con­tri­bu­tions of both ele­vated fibrin­o­gen and plate­
ther­apy. High lev­els of acute phase reac­tants such as C-reac­tive lets.21,24 Fibrinolysis is also suppressed.11,12 Thrombin gen­er­a­tion is
pro­tein can also cause false pos­i­tives by prolonging in vitro clot­ nor­mal to increased even in patients receiv­ing pro­phy­lac­tic anti-
ting times. Prolonged aPTT due to LA may neces­si­tate the use of coagulation.25 However, the clin­ic ­ al util­ity of these assays is uncer­
the anti-factor Xa assay for mon­i­tor­ing hep­a­rin ther­apy. tain, and they are not recommended out­side of a research set­ting.

616  |  Hematology 2021  |  ASH Education Program


Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/614/1851465/614gerber.pdf by guest on 13 December 2021
Figure 2.  Pathogenesis of CAC. SARS-CoV-2 binds the angiotensin-converting enzyme 2 receptor on respiratory epithelium and endo-
thelial cells. Viral replication and shedding lead to pneumocyte and endothelial cell apoptosis, triggering an inflammatory response,
which in a portion of patients leads to pathogenic cytokine storm. These cytokines and endothelial injury result in procoagulant
changes, including the release of VWF and FVIII and the upregulation of TF, P-selectin, and fibrinogen. Anticoagulant proteins such as
endothelial protein C receptor, antithrombin, and thrombomodulin are downregulated. Fibrinolytic impairment also contributes to
hypercoagulability. Neutrophil activation and the release of neutrophil extracellular traps further stimulate thrombosis, and platelets
in COVID-19 may exhibit marked hyperreactivity. SARS-CoV-2 spike proteins may directly amplify complement, and contribute to vas-
cular injury and microthrombosis. The coagulation derangements observed in COVID-19 are a direct result of microthrombosis, which
is initially localized to the lungs, causing elevations in D-dimer and fibrinogen. As the disease progresses, more systemic inflammation
and the activation of coagulation lead to thrombocytopenia and prolongation of clotting times. In some patients this may progress
to a frank consumptive coagulopathy.

Pathogenesis of CAC (4) effects on coag­u­la­tion and fibri­no­lytic sys­tems: hyp­oxia and
Thromboinflammation is cen­tral to the path­o­phys­i­­ol­ogy of CAC, vaso­con­stric­tion induce the expres­sion of TF and plas­min­o­gen
which is driven by a com­bi­na­tion of direct effects of viral infec­tion acti­va­tor inhibitor disrupting the bal­ance between coag­u­la­tion
on pul­mo­nary vas­cu­lar endo­the­lium, a local and sys­temic inflam­ and fibri­no­ly­sis.29 Patients with COVID-19 have increased throm­bin
ma­tory response, and cross talk between the inflam­ma­tory and and plas­min gen­er­a­tion poten­tial and an increase in fibrin for­ma­
coag­u­la­tion path­ways (Figure 2). Pathogenic mech­a­nisms con­trib­ tion,25 con­sis­tent with other reports of decreased fibri­no­ly­sis.11,12
ut­ing to CAC have been reviewed in detail by Iba et al13: (1) direct Perturbation of the bal­ance between throm­bin gen­er­a­tion (fibrin
viral effects and inflam­ma­tion: SARS-CoV-2 enters respi­ra­tory epi­ clot for­ma­tion) and plas­min gen­er­a­tion (fibri­no­lytic path­ways) is
the­lium and endo­the­lium via the angiotensin-converting enzyme 2 crit­i­cal to the path­o­phys­i­­ol­ogy of COVID-19.11,21 In severe COVID-19,
recep­tor. Direct endo­the­lial effects include procoagulant changes the lag time to throm­ bin, plas­ min, and fibrin for­ ma­tion is in-
such as sur­face adhe­sion mol­e­cule expres­sion and the release of creased, suggesting a loss in coag­u­la­tion-acti­vat­ing mech­a­nisms
VWF and FVIII. Viral rep­li­ca­tion and shed­ding trig­ger an inflam­ma­ in severe dis­ease.25 While coag­u­la­tion acti­va­tion is ini­tially restrict-
tory response, or “cyto­kine storm.” Proinflammatory cyto­kines IL- ed to the lungs and man­i­fests as an increase in D-dimer, severe
1β, IL-6, and tumor necro­ sis fac­ tor upregulate procoagulant dis­ease is char­ac­ter­ized by sys­temic inflam­ma­tion and dis­sem­i­
mol­e­cules, includ­ing tis­sue fac­tor (TF), P-selectin, fibrin­o­gen, FVIII, nated microthrombosis caus­ing a con­sump­tive coagulopathy.
and VWF; (2) neu­tro­phil extra­cel­lu­lar traps: neu­tro­phil acti­va­tion
and release of neu­tro­phil extra­cel­lu­lar traps pro­mote plate­let acti­ Bleeding in COVID-19
va­tion, phosphatidylserine expo­sure, acti­va­tion of fac­tor V and Bleeding is infre­quent in the set­ting of CAC, which has a pre­dom­
fac­tor XI, and throm­bin gen­er­a­tion26; (3) com­ple­ment acti­va­tion: i­nantly prothrombotic phe­no­type. Helms et al reported bleed­
SARS-CoV-2 spike pro­teins directly acti­vate com­ple­ment,27 and ing com­pli­ca­tions in 2.7% of 150 patients with COVID-related
com­ple­ment depo­si­tion and microthrombosis are observed in the ARDS.15 Among 2773 hos­pi­tal­ized COVID-19 patients from New
lungs, skin, kid­neys, and hearts of patients with severe dis­ease28; York, major bleed­ing was reported in 1.9% not on ther­a­peu­tic
Prakash Singh Shekhawat
COVID-19 coagulopathy | 617
anticoagulation and in 3% of those on ther­a­peu­tic anticoagula- tably, this rec­om­men­da­tion is iden­ti­cal to that for other acute-
tion.30 However, in an anal­y­sis of 400 patients hos­pi­tal­ized with ly ill inpa­tients. In the absence of sig­nif­i­cant renal impair­ment,
COVID-19 in Bos­ton, Al-Samkari et al found a higher over­all bleed­ we encour­age the use of low-molec­u­lar-weight hep­a­rin (LMWH)
ing rate of 4.8%; major bleed­ing occurred in 2.3%, includ­ing 5.6% over unfractionated hep­ ar­in (UFH) to avoid pos­ si­
ble hep­a­
of crit­i­cally ill patients, and was asso­ci­ated with a plate­let count rin resis­tance with UFH. If UFH is used, we sug­gest mon­i­tor­ing
<150 × 109/L at admis­sion (Odds Ratio 2.90 [1.05-7.99]).3 Prelimi- anti-factor Xa lev­els due to poten­tial confounding of the aPTT due
nary data from a multiplatform, ran­dom­ized con­trolled trial found to ele­vated FVIII and fibrin­o­gen or LAs. Further, anticoagulation
a 0.9% and 1.9% rate of major bleed­ing in noncritically ill par­ rec­om­men­da­tions may need to be mod­i­fied for patients with
tic­i­pants on pro­phy­lac­tic or ther­a­peu­tic anticoagulation, respec­ severe throm­bo­cy­to­pe­nia, bleed­ing risk, and extremes of body
tively.31 Among crit­ i­
cally ill patients, how­ ever, major bleed­ ing weight. Early in the pan­demic, there were reports of throm­bo­sis
occurred in 3.1% on ther­ap ­ eu­tic anticoagulation and 2.4% on despite stan­dard thromboprophylaxis. Based on these reports
thromboprophylaxis.32 and extreme lab­o­ra­tory derange­ments, cli­ni­cians fre­quently
Among patients with COVID-19 ARDS treated with extra­cor­ esca­lated anticoagulation to inter­me­di­ate or even ther­a­peu­tic

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/614/1851465/614gerber.pdf by guest on 13 December 2021


po­real mem­brane oxy­gen­at­ ion (ECMO), major bleed­ing occurs in inten­sity. However, whether a higher inten­sity of anticoagulation
>40%, includ­ing intra­cra­nial bleed­ing in up to 12%, which is sig­nif­ reduces mor­bid­ity or mor­tal­ity with­out an unac­cept­able increase
i­cantly higher than the 2% to 6% bleed­ing rate reported in non- in bleed­ing risk remains uncer­tain and is being eval­u­ated in mul­
COVID-19 ECMO cohorts.33,34 DIC, shear stress-induced acquired ti­ple tri­als.
von Willebrand syn­drome, COVID-19-asso­ci­ated endothelitis, and Preliminary data from a multiplatform, ran­ dom­ized con­
inten­sive anticoagulation are impli­cated in severe bleed­ing on trolled trial com­pris­ing 3 global stud­ies (REMAP-CAP, ATTACC,
ECMO.34 The anticoagulation required for ECMO and the acti­va­ and ACTIV-4A) com­par­ing the effects of stan­dard-dose vs full-
tion of coag­u­la­tion from arti­fi­cial sur­faces may con­found inter­pre­ dose hep­a­rin on the pri­mary out­come, a com­pos­ite of 21-day
ta­tion of coag­u­la­tion stud­ies. Careful inter­pre­ta­tion is required “organ sup­port-free” days and in-hos­pi­tal mor­tal­ity, were
to bal­ance anticoagulation, bleed­ing, and throm­bo­sis in patients recently made avail­­ able as pre­ prints.31,32 The enroll­
ment of
receiv­ing ECMO. mod­er­ately ill patients was stopped early for supe­ri­or­ity; ther­
a­peu­tic anticoagulation reduced the require­ment for organ
Management of CAC sup­ port and mor­ tal­
ity (OR 1.29 [1.04-1.61]) irrespective of
Which lab­o­ra­tory param­e­ters should be eval­u­ated in CAC? base­line D-dimer.31 Meanwhile, the enroll­ment of crit­i­cally ill
In out­pa­tients with COVID-19, there is cur­rently insuf­fic ­ ient evi­ patients was halted due to futil­ity, with no decrease in mor­tal­
dence to rec­om­mend mon­i­tor­ing of coag­u­la­tion mark­ers, includ­ ity and a higher rate of major bleed­ing with ther­a­peu­tic anti-
ing D-dimer, plate­let count, PT, and fibrin­o­gen, since these are coagulation.32 These appar­ently dis­crep­ant results sug­gest
unlikely to have an impact on man­ age­ment. For hos­ pi­tal­
ized that per­haps ther­a­peu­tic hep­a­rin works best if started early.
patients, we agree with the interim rec­om­men­da­tions from the The INSPIRATION study reported that in crit­i­cally ill patients
ISTH and the Amer­i­can Society of Hematology to seri­ally mon­i­ with COVID-19, inter­me­di­ate- vs pro­phy­lac­tic-dose LMWH did
tor D-dimer lev­els, plate­let count, PT, and fibrin­o­gen since these not result in a decrease in the com­pos­ite end point of throm­
help with risk strat­i­fi­ca­tion.35 Worsening of these param­e­ters may bo­sis, extra­cor­po­real mem­brane oxy­gen­a­tion, or mor­tal­ity
war­rant more aggres­sive crit­i­cal care sup­port and con­sid­er­ation within 30 days.38 The mul­ti­na­tional prag­matic RAPID COVID
of inves­ti­ga­tional ther­a­pies. D-dimer ele­va­tion is non­spe­cific and COAG trial (NCT04362085) is eval­u­at­ing ther­a­peu­tic vs pro­
need not prompt imag­ing for venous throm­bo­em­bo­lism (VTE) phy­lac­tic doses of LMWH or UFH in hos­pi­tal­ized, noncritically
in the absence of clin­i­cal symp­toms or signs of VTE.36 However, ill COVID-19 patients with ele­vated D-dimer on the com­pos­ite
increas­ing D-dimer in the set­ting of decreas­ing C-reac­tive pro­ out­come of ICU admis­sion, non­in­va­sive pos­i­tive pres­sure ven­
tein may war­rant screen­ing for VTE. ti­la­tion, mechan­i­cal ven­ti­la­tion, and death at 28 days. The
ACTION trial, a prag­matic trial at 31 sites in Brazil, com­pared
Role of blood prod­uct trans­fu­sion in-hos­pi­tal ther­a­peu­tic anticoagulation with rivaroxaban
There is no evi­dence that blood com­po­nent ther­apy to cor­rect (92%) or enoxaparin followed by rivaroxaban until day 30 vs
abnor­mal lab­o­ra­tory param­e­ters improves out­comes in the ab- pro­phy­lac­tic-dose enoxaparin or UFH in hos­pi­tal­ized patients
sence of bleed­ing. In patients with bleed­ing or those need­ing with ele­vated D-dimer. There was no dif­fer­ence in the pri­mary
inva­sive pro­ce­dures, we sug­gest trans­fu­sion of plate­lets to main­ effi­cacy out­come of time to death and dura­tion of hos­pi­tal­i­za­
tain a plate­let count ≥50 × 109/L, cryoprecipitate if fibrin­og ­ en is tion and sup­ple­men­tal oxy­gen use through 30 days but sig­nif­
<150 mg/dL, and plasma for an inter­na­tional nor­mal­ized ratio ≥1.8 i­cantly more bleed­ing in the rivaroxaban group.39 Pending the
after correcting fibrin­ o­
gen. Four fac­ tor prothrombin complex final results of these and other stud­ies, the Amer­i­can Society
concentrate (25 units/kg) may be used instead of plasma in pa- of Hematology guide­line panel sug­gests pro­phy­lac­tic over
tients with severe coagulopathy and bleed­ing to avoid vol­ume inter­me­di­ate or ther­a­peu­tic dos­ing in mod­er­ately and severely
over­load that can worsen respi­ra­tory sta­tus. ill patients with COVID-19 with­out con­firmed or suspected
VTE. Higher-inten­ sity anticoagulation may be pre­ ferred in
Anticoagulation mod­er­ately ill (non-ICU, stage 2) patients deemed at high
As with other coagulopathies, treat­ment of the under­ly­ing con­ throm­botic and low bleed­ing risk. Therapeutically dosed anti-
di­tion is the only defin­i­tive solu­tion. Based on the asso­ci­a­tion of coagulation is appro­pri­ate for patients with preexisting indi­
D-dimer and other coag­u­la­tion mark­ers with severe dis­ease, mor­ ca­tions such as atrial fibril­la­tion and mechan­i­cal heart valves
tal­ity, and throm­bo­sis, at least pro­phy­lac­tic-dose anticoagulation and, if needed, to main­tain extra­cor­po­real cir­cuits and vas­cu­
is recommended in hos­pi­tal­ized patients with COVID-19.1,36,37 No- lar access devices.

618  |  Hematology 2021  |  ASH Education Program


COVID-19 vac­cines: cytopenias, bleed­ing, and throm­bo­sis Summary
The COVID-19 vac­ cines have proven to be highly safe and Inflammation, intra­vas­cu­lar coag­u­la­tion acti­va­tion, and micro­vas­
effec­tive and remain the most prom­is­ing tool in containing the cu­lar throm­bo­sis cause coag­u­la­tion derange­ments in COVID-19.
COVID-19 pan­demic. Cases of immune throm­bo­cy­to­pe­nia and Elevated D-dimer and fibrin­ o­
gen lev­ els are the most com­ mon
bleed­ing, includ­ing life-threat­en­ing hem­or­rhage, have been diag­ find­ing and are pre­dic­tive of adverse out­comes, includ­ing the
nosed after expo­sure to the mRNA-based vac­cines pro­duced by need for crit­i­cal care and mor­tal­ity. In con­trast to DIC, throm­bo­
Moderna (mRNA-1273) and Pfizer-BioNTech (BNT162b2).40 Fortu- cy­to­pe­nia is rare, and the PT/aPTT are usu­ally nor­mal or min­i­mally
nately, the major­ity of these respond to immune throm­bo­cy­to­ prolonged. Some patients exhibit aPTT pro­lon­ga­tion due to aPL.
pe­nia-directed treat­ments such as intra­ve­nous Ig and cor­ti­co­ste­ Bleeding, while rare, may occur in crit­i­cally ill patients that develop
roids.40 It is unclear whether this rep­re­sents a causal rela­tion­ship a con­sump­tive coagulopathy and those on ECMO. Blood-prod­uct
or a coin­ci­den­tal asso­ci­a­tion; how­ever, a recent anal­y­sis from the sup­port to cor­rect the coagulopathy is reserved for bleed­ing pa-
Vaccine Adverse Event Reporting System reported that the inci­ tients or those requir­ing inva­sive pro­ce­dures. The results of ongo­
dence of throm­bo­cy­to­pe­nia after either mRNA vac­cine was 0.8 ing tri­als eval­u­at­ing the impact of higher-inten­sity anticoagulation

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/614/1851465/614gerber.pdf by guest on 13 December 2021


per mil­lion doses, which is no higher than the back­ground rate of on COVID-19 out­comes are eagerly awaited. Until then, all­hos­pi­
immune throm­bo­cy­to­pe­nia (3.3 per 100 000 adults).41 tal­ized patients should receive stan­dard thromboprophylaxis.
More recently, rare cases of a vac­ cine-induced throm­
botic throm­bo­cy­to­pe­nia (VITT) were reported after admin­is­ Acknowledgments
tra­tion of the ade­no­vi­ral vec­tor vac­cines (ChAdOx1 nCoV-19 This work was supported by National Institutes of Health National
and Ad26.COV2.S).42 The esti­ mated inci­ dence is 1 case per Heart, Lung, and Blood Institute grant K99HL150594 (to S.C) and
100,000 expo­ sures with a slight female pre­ pon­der­ ance, and T32HL007525 (to G.F.G.) and an Amer­i­can Society of Hematology
it occurs more com­ monly in youn­ ger (<55-60 years) indi­ Scholar Award (to S.C.).
vid­ u­
als, although this could reflect the demo­ graph­ ics of
indi­vid­u­als vac­ci­
nated early. Occurring 4 to 30 days post­ Conflict-of-inter­est dis­clo­sure
vac­ci­na­tion, VITT is asso­ci­ated throm­bo­cy­to­pe­nia and Glo­ria F. Gerber: no com­pet­ing finan­cial inter­ests to declare.
throm­bo­sis, par­tic­u­larly at unusual sites such as the cere­bral or Shruti Chaturvedi: no com­pet­ing finan­cial inter­ests to declare.
splanch­nic cir­cu­la­tions, along with high lev­els of plate­let-acti­vat­
ing IgG antibodies that rec­og­nize plate­let fac­tor 4 (PF4) in the Off-label drug use
absence of hep­a­rin. Anti-PF4/hep­a­rin antibodies are detect­able Glo­ria F. Gerber: the off-label use of prothrombin complex
by enzyme-linked immu­no­sor­bent assays, but rapid immu­no­as­ ­concentrate is discussed.
say may yield false-neg­a­tive results. Functional assays should be Shruti Chaturvedi: the off-label use of prothrombin complex
performed in the pres­ence and absence of PF4. The stan­dard ­concentrate is discussed.
sero­to­nin release assay may yield false-neg­a­tive results as VITT
antibodies exhibit PF4-depen­dent activ­ity and may dem­on­strate Correspondence
decreased reac­tiv­ity in the pres­ence of hep­a­rin. Patients with Shruti Chaturvedi, Johns Hopkins University, Ross Research Bldg,
VITT fre­quently have a con­sump­tive coagulopathy with ele­vated Rm 1025, 720 Rutland Ave, Baltimore, MD 21205; e-mail: schatur3@
D-dimer and hypofibrinogenemia. VITT is asso­ci­ated with high jhmi​­.edu.
mor­ tal­
ity, with cere­ bral hem­ or­
rhage as the lead­ ing cause of
death. Treatment with a nonheparin anti­co­ag­u­lant and intra­ve­ References
1. Iba T, Levy JH, Connors JM, Warkentin TE, Thachil J, Levi M. Managing
nous Ig is recommended, with plate­let trans­fu­sions reserved for throm­bo­sis and car­dio­vas­cu­lar com­pli­ca­tions of COVID-19: answer­ing the
bleed­ing. While much is still to be learned about the inci­dence, ques­tions in COVID-19-asso­ci­ated coagulopathy. Expert Rev Respir Med.
path­o­gen­e­sis, and opti­mal man­age­ment of VITT, given the mil­ 2021;15(8):1003-1011.
li­ons of indi­vid­u­als vac­ci­nated world­wide, these hema­to­logic 2. Iba T, Levy JH, Levi M, Connors JM, Thachil J. Coagulopathy of coronavirus
dis­ease 2019. Crit Care Med. 2020;48(9):1358-1364.
com­pli­ca­tions are van­ish­ingly rare and should not hin­der vac­ci­ 3. Al-Samkari H, Karp Leaf RS, Dzik WH, et  al. COVID-19 and coag­u­la­tion:
na­tion efforts. bleed­ing and throm­botic man­i­fes­ta­tions of SARS-CoV-2 infec­tion. Blood.
2020;136(4):489-500.
4. Nopp S, Moik F, Jilma B, Pabinger I, Ay C. Risk of venous throm­bo­em­bo­lism
in patients with COVID-19: a sys­tem­atic review and meta-anal­y­sis. Res Pr
CLINICAL CASE (Con­t in­u ed) Thromb Haemost. 2020;4(7):1178-1191.
5. Tang N, Li D, Wang X, Sun Z. Abnormal coag­u­la­tion param­e­ters are asso­
Our patient had find­ings typ­i­cal of CAC with an ele­vated d-dimer
ci­ated with poor prog­no­sis in patients with novel coronavirus pneu­mo­nia.
and fibrin­o­gen with nor­mal plate­lets and PT. An LA was detected, J Thromb Haemost. 2020;18(4):844-847.
account­ ing for her prolonged aPTT. She received a pro­ phy­ 6. Zhou F, Yu T, Du R, et  al. Clinical course and risk fac­tors for mor­tal­ity of
lac­tic dose of LMWH in addi­tion to dexa­meth­a­sone. D-dimer adult inpa­tients with COVID-19 in Wuhan, China: a ret­ro­spec­tive cohort
trended up to a peak of 4.1 mg/dL, and plate­let count dropped study. Lancet. 2020;395(10229):1054-1062.
7. Zhang L, Yan X, Fan Q , et al. D-dimer lev­els on admis­sion to pre­dict in-hos­pi­tal
to 118 × 109/L on day 5 of hos­pi­tal­i­za­tion. PT remained nor­mal. mor­tal­ity in patients with Covid-19. J Thromb Haemost. 2020;18(6):1324-
Oxygen require­ments peaked at 4 L by nasal can­nula on day 9 1329.
and grad­u­ally improved over the fol­low­ing week. No throm­botic 8. Guan WJ, Ni ZY, Hu Y, et al; China Medical Treatment Expert Group for
events occurred. She was discharged home with­out out­pa­tient Covid-19. Clinical char­ac­ter­is­tics of coronavirus dis­ease 2019 in China. N
Engl J Med. 2020;382(18):1708-1720.
thromboprophylaxis on day 15. Repeat test­ ing to deter­
mine
9. Taylor FB Jr, Toh CH, Hoots WK, Wada H, Levi M; Scientific Subcommit-
whether her LA is per­sis­tent was pend­ing at discharge. tee on Disseminated Intravascular Coagulation (DIC) of the International
Society on Thrombosis and Haemostasis (ISTH). Towards def­i­ni­tion, clin­i­cal
Prakash Singh Shekhawat
COVID-19 coagulopathy | 619
and lab­o­ra­tory cri­te­ria, and a scor­ing sys­tem for dis­sem­i­nated intra­vas­cu­ 28. Magro C, Mulvey JJ, Berlin D, et al. Complement asso­ci­ated micro­vas­cu­lar
lar coag­u­la­tion. Thromb Haemost. 2001;86(5):1327-1330. injury and throm­bo­sis in the path­o­gen­e­sis of severe COVID-19 infec­tion: a
10. Ding R, Wang Z, Lin Y, Liu B, Zhang Z, Ma X. Comparison of a new cri­te­ria report of five cases. Transl Res. 2020;220(June):1-13.
for sep­sis-induced coagulopathy and International Society on Thrombosis 29. Connors JM, Levy JH. COVID-19 and its impli­ca­tions for throm­bo­sis and anti-
and Haemostasis dis­sem­i­nated intra­vas­cu­lar coag­u­la­tion score in crit­i­cally coagulation. Blood. 2020;135(23):2033-2040.
ill patients with sep­sis 3.0: a ret­ro­spec­tive study. Blood Coagul Fibrinoly- 30. Paranjpe I, Fuster V, Lala A, et al. Association of Treatment Dose Anticoagu-
sis. 2018;29(6):551-558. lation With In-Hospital Survival Among Hospitalized Patients With COVID-
11. Hardy M, Michaux I, Lessire S, et al. Prothrombotic dis­tur­bances of hemo­ 19. J Am Coll Cardiol. 2020;76(1):122-124.
sta­sis of patients with severe COVID-19: a pro­spec­tive lon­gi­tu­di­nal obser­ 31. Lawler PR, Goligher EC, et al. Therapeutic anticoagulation in non-crit­i­cally
va­tional study. Thromb Res. 2021;197(Jan­u­ary):20-23. ill patients with Covid-19. medRxiv. Preprint posted online 17 May 2021.
12. Wright FL, Vogler TO, Moore EE, et  al. Fibrinolysis shut­down cor­re­la­tion 32. Zarychanski R. Therapeutic anticoagulation in crit­i­cally ill patients with
with throm­bo­em­bolic events in severe COVID-19 infec­tion. J Am Coll Surg. Covid-19—pre­lim­i­nary report. medRxiv. Preprint posted online 12 March
2020;231(2):193-203.e1203e1. 2021.
13. Iba T, Warkentin TE, Thachil J, Levi M, Levy JH. Proposal of the def­i­ni­tion for 33. Schmidt M, Hajage D, Lebreton G, et al; Groupe de Recherche Clinique en
COVID-19-asso­ci­ated coagulopathy. J Clin Med. 2021;10(2):191. Reanimation et Soins intensifs du Patient en Insuffisance Respiratoire aigue
14. Wu C, Chen X, Cai Y, et al. Risk fac­tors asso­ci­ated with acute respi­ra­tory (GRC-RESPIRE) Sorbonne Université; Paris-Sorbonne ECMO-COVID Inves-

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/614/1851465/614gerber.pdf by guest on 13 December 2021


dis­tress syn­drome and death in patients with coronavirus dis­ease 2019 tigators. Extracorporeal mem­brane oxy­gen­a­tion for severe acute respi­ra­
pneu­mo­nia in Wuhan, China. JAMA Intern Med. 2020;180(7):934. tory dis­tress syn­drome asso­ci­ated with COVID-19: a ret­ro­spec­tive cohort
15. Helms J, Tacquard C, Severac F, et al; CRICS TRIGGERSEP (Clinical Research study. Lancet Respir Med. 2020;8(11):1121-1131.
in Intensive Care and Sepsis Trial Group for Global Evaluation and Research 34. Lebreton G, Schmidt M, Ponnaiah M, et al; Paris ECMO-COVID-19 Investi-
in Sepsis) Group. High risk of throm­bo­sis in patients with severe SARS- gators. Extracorporeal mem­brane oxy­gen­a­tion net­work orga­ni­sa­tion and
CoV-2 infec­tion: a mul­ti­cen­ter pro­spec­tive cohort study. Intensive Care clin­i­cal out­comes dur­ing the COVID-19 pan­demic in Greater Paris, France:
Med. 2020;46(6):1089-1098. a multicentre cohort study. Lancet Respir Med. 2021;9(8):851-862.
16. Lippi G, Plebani M, Henry BM. Thrombocytopenia is asso­ci­ated with severe 35. Thachil J, Tang N, Gando S, et  al. ISTH interim guid­ance on rec­og­ni­tion
coronavirus dis­ease 2019 (COVID-19) infec­tions: a meta-anal­y­sis. Clin Chim and man­ age­ ment of coagulopathy in COVID-19. J Thromb Haemost.
Acta. 2020;506(July):145-148. 2020;18(5):1023-1026.
17. Lodigiani C, Iapichino G, Carenzo L, et al; Humanitas COVID-19 Task 36. Cuker A, Tseng EK, Nieuwlaat R, et  al. Amer­i­can Society of Hematology
Force. Venous and arte­rial throm­bo­em­bolic com­pli­ca­tions in COVID-19 2021 guide­ lines on the use of anticoagulation for thromboprophylaxis
patients admit­ted to an aca­demic hos­pi­tal in Milan, Italy. Thromb Res. in patients with COVID-19. Blood Adv. 2021;5(3):872-888.
2020;191(July):9-14. 37. Barrett CD, Moore HB, Yaffe MB, Moore EE. ISTH interim guid­ance on rec­
18. Thachil J, Cushman M, Srivastava A. A pro­ posal for stag­ ing COVID-19 og­ni­tion and man­age­ment of coagulopathy in COVID-19: a com­ment. J
coagulopathy. Res Pract Thromb Haemost. 2020;4(5):731-736. Thromb Haemost. 2020;18(8):2060-2063.
19. Harzallah I, Debliquis A, Drenou B. Lupus anti­co­ag­u­lant is fre­quent in patients 38. Sadeghipour P, Talasaz AH, Rashidi F, et al; INSPIRATION Investigators.
with Covid-19. J Thromb Haemost. 2020;18(8):2064-2065. Effect of inter­me­di­ate-dose vs stan­dard-dose pro­phy­lac­tic anticoagulation
20. Abdel-Wahab N, Talathi S, Lopez-Olivo MA, Suarez-Almazor ME. Risk of on throm­botic events, extra­cor­po­real mem­brane oxy­gen­a­tion treat­ment,
devel­op­ing antiphospholipid antibodies fol­low­ing viral infec­tion: a sys­tem­ or mor­tal­ity among patients with COVID-19 admit­ted to the inten­sive care
atic review and meta-anal­y­sis. Lupus. 2018;27(4):572-583. unit: the INSPIRATION ran­dom­ized clin­i­cal trial. JAMA. 2021;325(16):1620-
21. Panigada M, Bottino N, Tagliabue P, et al. Hypercoagulability of COVID-19 1630.
patients in inten­sive care unit: a report of thromboelastography find­ings and 39. Lopes RD, de Barros E, Silva PGM, Furtado RHM, et al; ACTION Coalition
other param­e­ters of hemo­sta­sis. J Thromb Haemost. 2020;18(7):1738-1742. COVID-19 Brazil IV Investigators. Therapeutic ver­sus pro­phy­lac­tic anticoagu-
22. Nougier C, Benoit R, Simon M, et al. Hypofibrinolytic state and high throm­ lation for patients admit­ted to hos­pi­tal with COVID-19 and ele­vated D-dimer
bin gen­er­a­tion may play a major role in SARS-COV2 asso­ci­ated throm­bo­ con­cen­tra­tion (ACTION): an open-label, multicentre, randomised, con­trolled
sis. J Thromb Haemost. 2020;18(9):2215-2219. trial. Lancet. 2021;397(10291):2253-2263.
23. Blasi A, von Meijenfeldt FA, Adelmeijer J, et al. In vitro hyper­co­ag­u­la­bil­ity 40. Lee EJ, Cines DB, Gernsheimer T, et al. Thrombocytopenia fol­low­ing Pfizer
and ongo­ing in vivo acti­va­tion of coag­u­la­tion and fibri­no­ly­sis in COVID-19 and Moderna SARS-CoV-2 vac­ci­na­tion. Am J Hematol. 2021;96(5):534-537.
patients on anticoagulation. J Thromb Haemost. 2020;18(10):2646-2653. 41. Welsh KJ, Baumblatt J, Chege W, Goud R, Nair N. Thrombocytopenia
24. Ranucci M, Ballotta A, Di Dedda U, et  al. The procoagulant pat­tern of includ­ing immune throm­bo­cy­to­pe­nia after receipt of mRNA COVID-19 vac­
patients with COVID-19 acute respi­ ra­
tory dis­tress syn­drome. J Thromb cines reported to the Vaccine Adverse Event Reporting System (VAERS).
Haemost. 2020;18(7):1747-1751. Vaccine. 2021;39(25):3329-3332.
25. Bouck EG, Denorme F, Holle LA, et al. COVID-19 and sep­sis are asso­ci­ated 42. Arepally GM, Ortel TL. Vaccine-induced immune throm­botic throm­bo­
with dif­fer­ent abnor­mal­i­ties in plasma procoagulant and fibri­no­lytic activ­ cy­to­pe­nia: what we know and do not know. Blood. 2021;138(4):293-298.
ity. Arterioscler Thromb Vasc Biol. 2021;41(1):401-414.
26. Middleton EA, He XY, Denorme F, et al. Neutrophil extra­cel­lu­lar traps con­
trib­ute to immunothrombosis in COVID-19 acute respi­ra­tory dis­tress syn­
drome. Blood. 2020;136(10):1169-1179.
27. Yu J, Yuan X, Chen H, Chaturvedi S, Braunstein EM, Brodsky RA. Direct acti­
va­tion of the alter­na­tive com­ple­ment path­way by SARS-CoV-2 spike pro­ © 2021 by The Amer­i­can Society of Hematology
teins is blocked by fac­tor D inhi­bi­tion. Blood. 2020;136(18):2080-2089. DOI 10.1182/hema­tol­ogy.2021000297

620  |  Hematology 2021  |  ASH Education Program


THE COVID CRASH: LESSONS LEARNED FROM A WORLD ON PAUSE

COVID-19 and thrombosis: searching for evidence


Bright Thilagar,1 Mohammad Beidoun,2 Ruben Rhoades,3 and Scott Kaatz1

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/621/1852155/621thilagar.pdf by guest on 13 December 2021


Division of Hospital Medicine, Henry Ford Health System, Detroit, MI; 2Department of Internal Medicine, Henry Ford Health System, Detroit, MI;
1

Division of Hematology, Thomas Jefferson University, Philadelphia, PA


3

Early in the pandemic, COVID-19-related increases in rates of venous and arterial thromboembolism were seen. Many
observational studies suggested a benefit of prophylactic anticoagulation for hospitalized patients using various dosing
strategies. Randomized trials were initiated to compare the efficacy of these different options in acutely ill and critically
ill inpatients as the concept of immune-mediated inflammatory microthrombosis emerged. We present a case-based
review of how we approach thromboembolic prophylaxis in COVID-19 and briefly discuss the epidemiology, the patho-
physiology, and the rare occurrence of vaccine-induced thrombotic thrombocytopenia.

LEARNING OBJECTIVES
• Appreciate the incidence, risk factors, and pathophysiology of thrombosis in COVID­19 infection
• Accurately prescribe anticoagulation prophylaxis before, during, and after hospitalization
• Be able to recognize and diagnose vaccine­induced immune thrombotic thrombocytopenia

P <.001).4 Increased arterial thrombotic events were also


CLINICAL CASE reported during hospitalization, with a prevalence of 3.9%
A 62­year­old physician had been rounding on the for coronary artery events and 1.6% for stroke.1 Acute
COVID­19 ward early in the pandemic (April 2020) and con­ limb ischemia was reported in 0.3% to 1% of hospital­
tracted the disease. After 1 week, he became hypoxic and ized patients, predominantly affecting men, with 18% of
was admitted to the general medical floor, requiring 3L patients suffering limb loss.5
nasal oxygen. He had no major comorbidities and received Rates of postdischarge VTE in COVID­19 patients, how­
prophylactic­dose low­molecular­weight heparin (LMWH). ever, were not as high. In a cohort from California, VTE
rates in COVID­19 patients were no higher than patients
testing negative for COVID­19 (1.8% vs 2.2%; P =.16).6 A
Introduction pre­/postpandemic comparison of postdischarge VTE did
COVID­19 infection clearly increases the risk of thrombotic not show a statistically significant risk of VTE in COVID­19
events for hospitalized patients, but rates of reported inci­ patients (odds ratio, 1.6; 95% CI, 0.77-3.1).7
dence have varied. A meta­analysis of retrospective studies
involving 64 503 patients showed that deep vein thrombo­ Pathophysiology of thrombosis in COVID-19 infection
sis (DVT) had an overall prevalence of 11.2% and pulmonary The coagulopathy of COVID­19 infection is complex, involv­
embolism (PE) of 7.8% in those needing hospitalization.1 ing an interplay between endothelial cell injury, inflammation,
Pooled rates of venous thromboembolism (VTE) were and coagulation. Infection of pulmonary alveolar cells causes
higher in the intensive care unit (ICU) setting (27.9% vs 7.1% severe endothelial injury and is marked by a local inflamma­
in the ward).2 Studies screening patients for VTE reported tory infiltrate and microthrombi.8 Microthrombi in alveolar
a prevalence rate of 25.2% compared to a rate of 12.7% capillaries and vascular congestion were present in nearly
in those testing only symptomatic patients.1 When more 45% of patients who died from acute respiratory complica­
than 95% of the hospitalized patients received pharmaco­ tions due to COVID­19.9 Alveolar capillary microthrombi were
logic VTE prophylaxis, rates were lower, at 3.1% for non­ 9 times more prevalent in those who died from COVID­19
ICU patients and 7.6% for ICU patients.3 Venous or arterial infection compared to similar severe H1N1 infections, and
thrombosis was independently associated with higher increased thrombotic complications were mostly noted in
mortality risk (hazard ratio [HR], 1.82; 95% CI, 1.54-2.15; those with longer hospital stays.8 In severe infection, this
Prakash Singh Shekhawat
COVID­19 and thrombosis: searching for evidence | 621
Table 1. Risk fac­tors to remem­ber in COVID-19-related throm­bo­sis4,37

COVID-19 infec­tion COVID-19 (ade­no­vi­rus) vec­tor vac­cines


Older age (>75) Middle age (18-49)
Male Female
Obesity Platelet count <150
Hypertension Splanchnic vein and cere­bral vein throm­bo­sis reported in higher rates
Prior car­diac dis­ease
Active can­cer or recent anti­can­cer treat­ment
Elevated D-dimer, FDP, LA
VTE in lower-extrem­ity deep veins and PE com­monly seen

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/621/1852155/621thilagar.pdf by guest on 13 December 2021


FDP, fibrin deg­ra­da­tion prod­ucts.

immune response can be exag­ger­ated and cause sys­temic hyper­ increases the like­li­hood of devel­op­ment of hep­a­rin-induced
inflammation, marked by high lev­ els of proinflammatory cyto­ throm­bo­cy­to­pe­nia (HIT), another poten­tial eti­­ol­ogy of VTE in
kines.10,11 The down­stream effects of endo­ the­
lial cell acti­
va­tion these patients. Rates of HIT in severe COVID-19 infec­tion are
and a sys­temic inflam­ma­tory response include a hyper­co­ag­u­la­ble var­i­able across stud­ies, with rates rang­ing from 0.16% in all­
state and both micro- and macrovascular throm­bo­sis, with mul­ti­ admit­ted patients to 8.1% in an ICU pop­u­la­tion. Anti-plate­let
ple inter­re­lated mech­a­nisms. fac­tor 4 (PF4) antibodies are fre­quently detected even in the
absence of clin­i­cal HIT,21,22 and 1 group has dem­on­strated non-
Activation of endo­the­lial cells, plate­lets, hep­a­rin-depen­dent plate­let-acti­vat­ing immune complexes that
and leu­ko­cytes may con­trib­ute to throm­bo­sis.23
Direct infec­tion of vas­cu­lar endo­the­lial cells with resultant apo­pto­
sis has been described in an autopsy series of COVID-19 patients.12 Antiphospholipid antibodies
This, along with com­ple­ment acti­va­tion,13 con­trib­utes to a local A final poten­ tial mech­ a­
nism for throm­ bo­sis in patients with
inflam­ma­tory reac­tion and fur­ther endo­the­lial cell acti­va­tion, with COVID-19 is the devel­op­ment of antiphospholipid antibodies,
the expres­sion of tis­sue fac­tor, release of von Willebrand fac­tor an appeal­ ing the­ory given the hyperinflammatory response
(VWF), and decreased syn­the­sis of nitric oxide and pros­ta­cy­clin,14 and poten­tial for anti­body pro­duc­tion. As many as 90% of crit­
all­ of which pro­mote coag­u­la­tion. An increase in VWF cou­pled i­cally ill patients have been found to be pos­i­tive for a lupus
with a rel­a­tive imbal­ance in ADAMTS-13 has been dem­on­strated anti­co­ag­u­lant (LA).24,25 The clin­ic
­ al sig­nif­i­cance of this is unclear,
in patients with COVID-19 of vary­ing sever­ity,15 and ele­vated VWF as an asso­ci­a­tion with throm­bo­sis has not been con­sis­tently
anti­gen and low ADAMTS-13 activ­ity seem to cor­re­late with the dem­on­strated,25 assays may be sus­ cep­ ti­
ble to inter­ fer­
ence
devel­op­ment of VTE.16 Platelet acti­ va­
tion increases in severe by C-reac­tive pro­tein and unfractionated hep­a­rin, and rates
COVID-19 infec­tion, trig­ger­ing increased tis­sue fac­tor expres­sion of anticardiolipin and anti-β2-gly­co­pro­tein seem to be sig­nif­
and coag­u­la­tion.14,17,18 Similarly, neu­tro­phil acti­va­tion increases, with i­cantly lower.24-26 However, 1 study at a ter­tiary care cen­ter in
the release of neu­tro­phil extra­cel­lu­lar traps that pro­mote throm­bo­ the US found a sig­nif­i­cant asso­ci­a­tion between LA pos­i­tiv­ity
sis in these patients.19 and venous or arte­rial throm­bo­sis.26

Coagulation and fibri­no­ly­sis


The increase in tis­ sue fac­ tor expres­sion on a vari­ ety of cells
induces extrin­sic coag­u­la­tion path­way acti­va­tion, while the CLINICAL CASE (Con­tin­ued)
release of fac­tor VIII from dam­aged endo­the­lium, NETosis, and
plate­let and com­ple­ment acti­va­tion all­ con­trib­ute to intrin­sic Our patient was a real one and in fact is the senior author of this
path­way acti­va­tion. These events result in throm­bin gen­er­a­ arti­cle. He received pro­phy­lac­tic-dose enoxaparin while on the
tion and the for­ma­tion of a fibrin clot. COVID-19 infec­tion is also med­i­cal floor.
marked by high lev­els of fibrin­o­gen,10,11 an acute phase reac­tant,
and the inhi­bi­tion of fibri­no­lytic path­ways. Patients with severe
infec­tion and shut­down of fibri­no­ly­sis—as mea­sured by a lack Anticoagulation pro­phy­laxis
of clot lysis on vis­co­elas­tic test­ing—were found to have greater When choos­ing a pro­phy­lac­tic approach, the risk of bleed­ing must
dimerized plas­min frag­ment D (D-dimer) and fibrin­o­gen lev­els be con­sid­ered, par­tic­u­larly if the plate­let count is <50 000, and
and a sig­nif­i­cantly greater inci­dence of clin­i­cal VTE (Table 1).20 mechan­i­cal pro­phy­laxis is likely the best approach until the bleed­
ing risk abates. The dose and choice of anti­co­ag­u­lant also depend
Heparin-induced throm­bo­cy­to­pe­nia on renal func­ tion, and it may be rea­ son­ able to give “obe­ sity-
Given the pro­pen­sity for throm­bo­sis, patients admit­ted with adjusted” doses—eg, enoxaparin 40 mg twice daily or a daily total
COVID-19 are fre­quently man­aged with anticoagulation pro­phy­ dose of 0.5 mg/kg based on lim­ited evi­dence.27 Of note, obe­sity-
laxis. This, com­bined with the sys­temic inflam­ma­tory response, adjusted pro­phy­lac­tic dos­ing dif­fers from inter­me­di­ate dos­ing.

622  |  Hematology 2021  |  ASH Education Program


Noncritically ill patients aban (20 mg or renally adjusted 15 mg daily) for 30 days or stan­dard
The Amer­ i­
can Society of Hematology (ASH) draft guide­ VTE pro­phy­laxis.31 Clinically unsta­ble patients (10%) ran­dom­ized to
lines (updated 8 Feb­ru­ary 2021) sug­gest pro­phy­lac­tic over rivaroxaban first received enoxaparin or unfractionated hep­a­rin
inter­me­di­ate- or ther­a­peu­tic-dose anticoagulation for patients (1 patient) followed by rivaroxaban when clin­i­cally sta­ble. Patients
with acute (not crit­i­cal) ill­ness and acknowl­edge pend­ing results ran­dom­ized to stan­dard pro­phy­laxis primarily received enoxaparin
of the com­bined anal­y­sis of the REMAP-CAP, ACTIV-4, and ATTAC (84%), and 13% con­tin­ued postdischarge pro­phy­laxis at their cli­ni­
multiplatform ran­dom­ized con­trolled tri­als (mpRCT).28 These cian’s dis­cre­tion. The hier­ar­chi­cal anal­y­sis of time to death, dura­
3 mpRCTs har­mo­nized their pro­to­cols to accel­er­ate the bat­tle tion of hos­pi­tal­i­za­tion, and dura­tion of sup­ple­men­tal oxy­gen was
against COVID-19 infec­tion and reported their pooled results in performed using the win ratio, and there was no dif­fer­ence in effi­
sep­a­rate papers for crit­i­cally ill and noncritically ill patients.29,30 cacy with a win ratio of 0.86; P  =  .40. There was also no dif­fer­ence in
The first patient was ran­dom­ized on 21 April 2020, and these each com­po­nent of this com­pos­ite out­come or throm­bo­em­bolic
tri­
als were stopped on 22 Jan­ u­
ary 2021. The pre­ print report event. However, major or clin­i­cally rel­e­vant non­ma­jor bleed­ing
appeared on 17 May 2021 when the prespecified supe­ri­or­ity stop­ was increased with ther­a­peu­tic anticoagulation (8%) com­pared to

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/621/1852155/621thilagar.pdf by guest on 13 December 2021


ping rule thresh­old was attained.30 The pri­mary anal­y­sis pop­u­la­ stan­dard dos­ing (2%) with a rel­a­tive risk of 3.64 ( P  =  .001).
tion had 2219 par­tic­i­pants with con­firmed COVID-19 who did not Until the 3 mpRCTs are fully published, it is rea­son­able to use
require ICU-level organ sup­port, which was defined as high-flow either ther­a­peu­tic or pro­phy­lac­tic doses of LMWH (pre­ferred if
oxy­gen, mechan­i­cal ven­ti­la­tion (inva­sive or non­in­va­sive), vaso­pres­ renal func­tion is accept­able) or hep­a­rin. Direct oral anti­co­ag­u­
sors, or inotropes. They were ran­dom­ized in an open-label man­ lants (DOACs) should not be used unless there is another indi­ca­
ner to ther­a­peu­tic-dose hep­a­rin (94.7% received LMWH, mostly tion, such as atrial fibril­la­tion.
enoxaparin) or usual care thromboprophylaxis (71.7% low dose
and 26.5% inter­me­di­ate dose) for up to 14 days. The pri­mary out­ How we treat noncritically ill hos­pi­tal­ized patients
come of sur­vival to hos­pi­tal dis­charge with­out organ sup­port (as
defined above) through 21 days occurred in 76.4% of par­tic­i­pants We dis­cuss the risks and ben­e­fits of pro­phy­lac­tic vs ther­
receiv­ing the usual care and increased by 4.6% with ther­a­peu­tic- a­peu­tic doses of anticoagulation and their impact on the
dose anticoagulation, with a median adjusted odds ratio of 1.29% need for organ sup­port in noncritically ill patients hos­pi­tal­
and a 99% prob­a­bil­ity of the ther­a­peu­tic dose being effec­tive. The ized with COVID-19. We use ther­a­peu­tic dos­ing of LMWH
prob­a­bil­ity of sur­vival to hos­pi­tal dis­charge with ther­a­peu­tic-dose or unfractionated hep­a­rin based on these early results and
hep­a­rin was 87.1%, with a median abso­lute improve­ment of 1.3%. eagerly await peer-reviewed pub­ li­
ca­
tion and guide­ line
Major bleed­ing occurred in 1.9% and 0.9% of ther­a­peu­tic-dose and updates.
usual care par­tic­i­pants, respec­tively. A prespecified anal­y­sis based
on D-dimer lev­els showed a slightly bet­ter prob­a­bil­ity of supe­ri­or­
ity in patients with high lev­els (97.3%) vs low lev­els (92.9%).
The ACTION trial ran­dom­ized 615 hos­pi­tal­ized patients (who CLINICAL CASE (Con­tin­ued)
were pri­mar­ily sta­ble and not crit­i­cally ill) in 31 Brazilian sites with Oxygen require­ ment increased for our patient, and a high
ele­vated D-dimer lev­ els above the upper limit of nor­ mal from flow was needed with trans­fer to the ICU, where LMWH was
24 June 2020 to 26 Feb­ru­ary 2021 to ther­a­peu­tic doses of rivarox­ increased to inter­me­di­ate dose. Several days later, intu­ba­tion

Figure 1. ASH recommendations for VTE prophylaxis in COVID patients with critical illness. Reproduced with permission from
Cuker et al.28
Prakash Singh Shekhawat
COVID-19 and throm­bo­sis: searching for evi­dence  |  623
and mechan­i­cal ven­ti­la­tion were required. Meanwhile, the Intermediate dose.  Among 600 ran­dom­ized COVID-19 patients in
patient’s father was also crit­i­cally ill with COVID-19, and his cli­ the ICU, 562 were included in the pri­mary anal­y­sis of the INSPIRATION
ni­cians used ther­a­peu­tic-dose LMWH: 2 patients with the same trial.32 Intermediate enoxaparin doses of 1 mg/kg once daily vs
last name, 1 floor apart, with dif­fer­ent doses to pre­vent the 40 mg daily (with adjust­ment for weight and cre­at­i­nine clear­ance)
same com­pli­ca­tions—all­ were searching for evi­dence. were com­pared for the pri­mary com­pos­ite effi­cacy out­come of
venous or arte­rial throm­bo­sis, treat­ment with extra­cor­po­real
mem­brane oxy­gen­a­tion, or mor­tal­ity. No dif­fer­ences were noted in
Critically ill patients 30-day out­comes between inter­me­di­ate and pro­phy­lac­tic doses
ASH guide­lines sug­gest pro­phy­lac­tic- over inter­me­di­ate-dose (45.7% vs 44.1%; odds ratio, 1.06; P   =   .70). Major bleed­ing occurred
anticoagulation in crit­i­cally ill patients, and rec­om­men­da­tions in 2.5% and 1.4% of patients who received inter­ me­ di­
ate and
for ther­a­peu­tic dose vs pro­phy­lac­tic dose are forthcoming ­pro­phy­lac­tic dos­ing, respec­tively, and the risk dif­fer­ence failed
(Figure 1). to meet the noninferiority mar­gin. Severe throm­bo­cy­to­pe­nia
occurred in 6 patients, all­of whom received an inter­me­di­ate dose.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/621/1852155/621thilagar.pdf by guest on 13 December 2021


Therapeutic dose.  Data for crit­i­cally ill par­tic­i­pants ran­dom­ized
while receiv­ing organ sup­port as pre­vi­ously described in the How we treat crit­i­cally ill patients
3 mpRCTs were posted on 12 March 2021.29 This prespecified
cohort stopped recruit­ ment for crit­ i­
cally ill patients on 19 We use pro­phy­lac­tic- and not inter­me­di­ate- or ther­a­peu­tic-
Decem­ber 2020 because of futil­ity. The pri­mary out­come of sur­ dose LMWH or hep­a­rin in crit­i­cally ill patients with COVID-19.
vival until dis­charge and the median num­ber of organ-sup­port-
free days was 3 in those ran­dom­ized to ther­ap ­ eu­tic-dose LMWH
or hep­a­rin and 5 days with the usual care (41% pro­phy­lac­tic-
dose and 51% inter­me­di­ate-dose LMWH or hep­a­rin; ­adjusted CLINICAL CASE (Con­tin­ued)
odds ratio, 0.87; 95% cred­i­ble inter­val, 0.70-1.08; like­li­hood of After a cou­ple of weeks in the ICU with respi­ra­tory fail­ure and
not achiev­ing mean­ing­ful rel­at­ ive improve­ment of at least 20%, sep­tic shock, our patient was extubated but suf­fered a small PE
99.8%). while his cen­tral line was being removed. He was treated for

Table 2. List of selected anticoagulation tri­als in COVID-19

Patient Trial name and


pop­u­la­tion trial iden­ti­fier Intervention Comparison Outcome Status
Hospitalized ACTION Trial 31
Therapeutic rivaroxaban Usual care thromboprophylaxis Mortality, length of stay, and oxy­gen use Published
(non-ICU) or LMWH
ATTACC30 Therapeutic hep­a­rin Usual care thromboprophylaxis Hospital dis­charge with­out need for Preprint
NCT04372589 or LMWH organ sup­port
ACTIV-4a30 Therapeutic hep­a­rin Usual care thromboprophylaxis Hospital dis­charge with­out need for Preprint
NCT04505774 or LMWH organ sup­port
REMAP-CAP30 Therapeutic hep­a­rin Usual care thromboprophylaxis Hospital dis­charge with­out need for Preprint
NCT02735707 or LMWH organ sup­port
Hospitalized ATTACC29 Therapeutic hep­a­rin Usual care thromboprophylaxis Hospital dis­charge with­out need for Preprint
(ICU) NCT04372589 or LMWH organ sup­port
ACTIV-4a29 Therapeutic hep­a­rin Usual care thromboprophylaxis Hospital dis­charge with­out need for Preprint
NCT04505774 or LMWH organ sup­port
REMAP-CAP29 Therapeutic hep­a­rin Usual care thromboprophylaxis Hospital dis­charge with­out need for Preprint
NCT02735707 or LMWH organ sup­port
INSPIRATION32 Intermediate-dose Prophylactic-dose hep­a­rin or Composite of VTE, arte­rial throm­bo­sis, Published
NCT04486508 hep­a­rin or LMWH LMWH or ECMO
Post dis­charge ACTIV-4 Prophylactic-dose Placebo Composite out­come of symp­tom­atic DVT, Recruiting
Convalescent apixaban PE, other VTE, ische­mic stroke, acute
NCT04650087 MI, other ATE, and all­-cause mor­tal­ity
Outpatient ACTIV-4c Prophylactic-dose Placebo Composite of symp­tom­atic DVT or PE, Recruiting
NCT04498273 apixaban, ther­a­peu­tic- ATE, MI, ische­mic stroke, hos­pi­tal­i­za­tion
dose apixaban, ASA for car­dio­vas­cu­lar/pul­mo­nary events,
and all­-cause mor­tal­ity
PREVENT-HD Prophylactic-dose Placebo Composite of symp­tom­atic VTE, MI, Recruiting
NCT04508023 rivaroxaban ische­mic stroke, sys­temic embolism,
acute limb ische­mia, all­-cause
hos­pi­tal­i­za­tion, and all­-cause mor­tal­ity
ASA, aspi­rin; ECMO, extra­cor­po­real mem­brane oxy­gen­a­tion; MI, myo­car­dial infarc­tion.

624  |  Hematology 2021  |  ASH Education Program


3 months with a DOAC, which pre­cluded the deci­sion regard­ As discussed pre­vi­ously, rates of VTE are rel­a­tively low post
ing post-hos­pi­tal dis­charge anti­co­ag­u­lant pro­phy­laxis. hos­pi­tal­i­za­tion; how­ever, mit­i­ga­tion of arte­rial and VTE along
with reduc­tions in all­-cause mor­tal­ity deserves inves­ti­ga­tion. A
pro­spec­tive reg­is­try of 4906 patients with COVID-19 were con­
Postdischarge pro­phy­laxis tacted a mean of 92 days after dis­charge, and 1.55% devel­oped
This patient had symp­toms of PE with acute chest pain and VTE, arte­rial throm­bo­em­bolic events (ATE) occurred in 1.71%,
wors­ened hyp­ox­emia and was sta­ble for com­puted tomog­ra­ and all­-cause mor­tal­ity was 4.83%. There was a 46% rel­a­tive
phy, ren­der­ing a straight­for­ward workup. We only pur­sue diag­ reduc­tion in the com­pos­ite out­come of VTE, ATE, and mor­tal­ity
nos­ing VTE based on clin­i­cal sus­pi­cion and do not screen or (P = .0046) in the mul­ti­var­i­able anal­y­sis.34 The ACTIV-4b National
use D-dimer thresh­olds to screen in accor­dance with guid­ance Institutes of Health-spon­sored trial is inves­ti­gat­ing whether apix­
state­ments.33 aban at pro­phy­lac­tic or ther­a­peu­tic doses and low-dose aspi­rin,

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/621/1852155/621thilagar.pdf by guest on 13 December 2021

Figure 2. VITT diagnostic flowchart from ISTH interim guidance published 20 April 2021. aPTT, activated partial thromboplastin time;
IVIG, intravenous immunoglobulin, PT, prothrombin time. Reproduced with permission from the ISTH.41
Prakash Singh Shekhawat
COVID-19 and throm­bo­sis: searching for evi­dence  |  625
com­pared to pla­cebo, can reduce these com­pli­ca­tions after hos­ On pre­sen­ta­tion, lab­o­ra­tory test­ing gen­er­ally shows vary­
pi­tal­i­za­tion. The stud­ies are sum­ma­rized in Table 2. ing lev­els of throm­bo­cy­to­pe­nia, high D-dimer lev­els, and low
The US Food and Drug Administration has approved rivarox­ fibrin­o­gen lev­els.35,38,39 Diagnostic test­ing for PF4 antibodies via
aban and betrixaban for postdischarge VTE pro­phy­laxis; how­ enzyme-linked immu­no­sor­bent assay (ELISA) results is one of
ever, betrixaban is not com­mer­cially avail­­able. The pro­phy­lac­tic the more sen­si­tive test­ing modal­i­ties for VITT, while the rapid
dose of 10 mg of rivaroxaban for a total of 31 to 39 days has been HIT tests such as the chemi­lu­mi­nes­cent immunoassay and latex
approved but should not be used in patients with cre­a­tine clear­ immunoturbidometric assay are not sen­ si­
tive and can yield
ance less than 30 mL/min, with drug-drug interactions and high false-neg­a­tive results.38 Principles of man­age­ment mir­ror those
bleeding-risk conditions.27 of aHIT, with the goal of inhibiting Fcγ recep­tor-medi­ated plate­
let acti­va­tion through the use of intra­ve­nous immune glob­u­lin
How we treat patients after dis­charge (with suggested daily dos­ing of 0.5-1 g/kg of ideal body weight
for at least 2 days) and the use of either oral or paren­tal nonhep­
We only use postdischarge pro­phy­laxis in selected patients arin anti­co­ag­u­lants.37 (See Figure 2 for man­age­ment sug­ges­tions

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/621/1852155/621thilagar.pdf by guest on 13 December 2021


in accor­dance with US Food and Drug Administration from the International Society on Thrombosis and Hemostasis
approval for med­i­cally ill patients with­out increased bleed­ [ISTH].)40 Vaccination with Ad26.COV2.S car­ries a warn­ing about
ing risk using a DOAC. the poten­tial devel­op­ment of VITT, espe­cially in women aged 18
to 49.36
“Prehospitalization” pro­phy­laxis
The trial results above sug­gest that higher doses of anticoag­ Conclusion
ulation are not effec­tive late in the dis­ease course, and it is a The throm­bo­em­bolic bur­den of COVID-19 seems to be decreas­
rea­son­able hypoth­e­sis that anticoagulation very early in the dis­ ing as the pan­demic evolves, and a mul­ti­tude of mech­an ­ isms
ease may be ben­e­fi­cial to decrease immuno-micro pul­mo­nary are pur­ported to explain the ele­vated risk with this infec­tion.
throm­bo­sis. Two tri­als with DOACs are under­way to help answer Key to under­stand­ing the tim­ing, dos­ing, and patient set­ting for
this ques­tion (Table 2). pro­phy­laxis is an appre­ci­a­tion of immune-induced inflam­ma­tory
microthrombosis. Robust ran­dom­ized tri­als are the best tools to
How we treat non­hos­pi­tal­ized out­pa­tients under­stand the appro­pri­ate use of anticoagulation to pre­vent
throm­bo­em­bolic com­pli­ca­tions, and the final results of many
We do not use anticoagulation pro­phy­laxis early in COVID- ongo­ing stud­ies are eagerly awaited.
19 for patients who do not require hos­pi­tal­i­za­tion.
Conflict-of-inter­est dis­clo­sure
Postvaccination: vac­cine-induced immune throm­botic Bright Thilagar: no competing financial interest to declare.
throm­bo­cy­to­pe­nia Mohammad Beidoun: no competing financial interest to declare.
The approval and wide­spread admin­is­tra­tion of vac­cines for Ruben Rhoades: no competing financial interest to declare.
COVID-19 brought yet another ele­ment into the dis­cus­sion of Scott Kaatz: research funding: Janssen, BMS, Osmosis R ­ esearch,
COVID-19 and throm­bo­sis. Vaccine-induced immune throm­botic National Institutes of Health; con­sul­tancy: Janssen, BMS, ­Alexion/
throm­bo­cy­to­pe­nia (VITT) or throm­bo­sis with throm­bo­cy­to­pe­nia Portola, Novartis, CSL Behring, Gilead.
syndrome is the name given to the throm­botic com­pli­ca­tions
cur­rently being reported across the world fol­low­ing the admin­
Off-label drug use
is­tra­tion of the ade­no­vi­rus vec­tor vac­cines (ChAdOx1 nCoV-19
Bright Thilagar: nothing to disclose.
[AstraZeneca/COVIDSHIELD] and Ad26.COV2.S [Johnson and
Mohammad Beidoun: nothing to disclose.
Johnson/Janssen]) against SARS-CoV-2.35 The under­ly­ing path­
Ruben Rhoades: nothing to disclose.
o­phys­i­­ol­ogy of VITT is not fully under­stood. One hypoth­e­sis is
Scott Kaatz: nothing to disclose.
that the polyanionic con­stit­u­ents of the ade­no­vi­rus vec­tor vac­
Scott Kaatz’s institution received research support from Osmosis
cines cause the for­ma­tion of antibodies to PF4. These antibodies
Research for one of the multi-platform trials using therapeutic
against PF4 then induce plate­let acti­va­tion, caus­ing both throm­
dose anticoagulation prophylaxis.
bo­cy­to­pe­nia and throm­bo­sis. VITT is sim­i­lar to auto­im­mune hep­
a­rin-induced throm­bo­cy­to­pe­nia (aHIT), with the pres­ence of
anti-PF4 polyanion antibodies induc­ing plate­let acti­va­tion in the Correspondence
absence of and inde­pen­dently of hep­a­rin expo­sure; how­ever, Scott Kaatz, Division of Hospital Medicine, CFP 413, 2799 W
VITT and aHIT dif­fer in the dis­tri­bu­tion of the thrombi.36,37 Grand Blvd, Detroit, MI 48202; e-mail: skaatz1@hfhs​­.org.
VITT gen­er­ally pres­ents between 4 to 30 days after ini­tial vac­
ci­na­tion with either ade­no­vi­rus vec­tor vac­cine.35 Early reports References
1. Tan BK, Mainbourg S, Friggeri A, et al. Arterial and venous throm­bo­em­bo­
showed that VITT pri­mar­ily presented in youn­ger females. Data
lism in COVID-19: a study-level meta-anal­y­sis. Thorax. Published online 23
on the inci­dence range from 1 case per 26 000 to 1 case per Feb­ru­ary 2021.
127 000 doses of ChAdOx1 nCoV-19 vac­cine admin­is­tered and 2. Jiménez D, García-Sanchez A, Rali P, et al. Incidence of VTE and bleed­ing
1 case per 500 000 doses of the Ad26.COV2.S. No spe­cific risk among hos­pi­tal­ized patients with coronavirus dis­ease 2019: a sys­tem­atic
fac­tors have been iden­ti­fied. In VITT, throm­bo­sis tends to man­i­ review and meta-anal­y­sis. Chest. 2021;159(3):1182-1196.
3. Al-Samkari H, Karp Leaf RS, Dzik WH, et  al. COVID-19 and coag­u­la­tion:
fest in rare sites, most nota­bly in the cere­bral venous sinuses and bleed­ing and throm­botic man­i­fes­ta­tions of SARS-CoV-2 infec­tion. Blood.
in the splanch­nic and hepatic veins.35,38 2020;136(4):489-500.

626  |  Hematology 2021  |  ASH Education Program


4. Bilaloglu S, Aphinyanaphongs Y, Jones S, Iturrate E, Hochman J, Berger 26. Reyes Gil M, Barouqa M, Szymanski J, Gonzalez-Lugo JD, Rahman S, Billett
JS. Thrombosis in hos­pi­tal­ized patients with COVID-19 in a New York City HH. Assessment of lupus anti­co­ag­u­lant pos­i­tiv­ity in patients with coronavi­
Health System. JAMA. 2020;324(8):799-801. rus dis­ease 2019 (COVID-19). JAMA Netw Open. 2020;3(8):e2017539.
5. Etkin Y, Conway AM, Silpe J, et  al. Acute arte­ rial throm­
bo­ em­bo­lism in 27. Anticoagulation Forum, Inc. Venous throm­bo­em­bo­lism (VTE) pro­phy­
patients with COVID-19 in the New York City Area. Ann Vasc Surg. 2021 laxis in acutely ill med­i­cal patients. Accessed 19 Sep­tem­ber 2021. https://
(Jan­u­ary);70:290-294. acforum-excellenceorg/Resource-Center/resource_files/1702-2021-03-
6. Roubinian NH, Dusendang JR, Mark DG, et al. Incidence of 30-day venous 03-142440pdf.
throm­bo­em­bo­lism in adults tested for SARS-CoV-2 infec­tion in an inte­grated 28. Cuker A, Tseng EK, Nieuwlaat R, et  al. Amer­i­can Society of Hematology
health care sys­tem in Northern California. JAMA Intern Med. 2021;181(7):997- 2021 guide­lines on the use of anticoagulation for thromboprophylaxis in
1000. patients with COVID-19. Blood Adv. 2021;5(3):872-888.
7. Roberts LN, Whyte MB, Georgiou L, et al. Postdischarge venous throm­bo­ 29. Zarychanski R. Therapeutic anticoagulation in crit­i­cally ill patients with
em­bo­lism fol­low­ing hos­pi­tal admis­sion with COVID-19. Blood. 2020;136(11): COVID-19—pre­lim­i­nary report. medRxiv. Preprint posted online 12 March
1347-1350. 2021.
8. Ackermann M, Verleden SE, Kuehnel M, et  al. Pulmonary vas­cu­lar endo­ 30. Lawler PR, Goligher EC, Berger JS, et  al. Therapeutic anticoagulation
thelialitis, throm­bo­sis, and angio­gen­e­sis in COVID-19. N Engl J Med. in non-crit­ i­
cally ill patients with COVID-19. N Engl J Med. 2021;385(9):
2020;383(2):120-128. 790-802.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/621/1852155/621thilagar.pdf by guest on 13 December 2021


9. Menter T, Haslbauer JD, Nienhold R, et  al. Postmortem e ­ xam­

na­tion of 31. Lopes RD, de Barros E, Silva PGM, Furtado RHM, et al; ACTION Coalition
COVID-19 patients reveals dif­fuse alve­o­lar dam­age with severe ­cap­il­lary COVID-19 Brazil IV Investigators. Therapeutic ver­sus pro­phy­lac­tic antico­
con­ges­tion and var­ie­gated find­ings in lungs and other organs ­suggesting agulation for patients admit­ted to hos­pi­tal with COVID-19 and ele­vated
vas­cu­lar dys­func­tion. Histopathology. 2020;77(2):198-209. D-dimer con­cen­tra­tion (ACTION): an open-label, multicentre, randomised,
10. Tang N, Li D, Wang X, Sun Z. Abnormal coag­u­la­tion param­e­ters are asso­ con­trolled trial. Lancet. 2021;397(10291):2253-2263.
ci­ated with poor prog­no­sis in patients with novel coronavirus pneu­mo­nia. 32. Sadeghipour P, Talasaz AH, Rashidi F, et al; INSPIRATION Investigators.
J Thromb Haemost. 2020;18(4):844-847. Effect of inter­me­di­ate-dose vs stan­dard-dose pro­phy­lac­tic anticoagulation
11. Ranucci M, Ballotta A, Di Dedda U, et  al. The procoagulant pat­tern of on throm­botic events, extra­cor­po­real mem­brane oxy­gen­a­tion treat­ment,
patients with COVID-19 acute respi­ra­tory dis­ tress syn­drome. J Thromb or mor­ tal­
ity among patients with COVID-19 admit­ ted to the inten­ sive
Haemost. 2020;18(7):1747-1751. care unit: the INSPIRATION ran­dom­ized clin­i­cal trial. JAMA. 2021;325(16):
12. Varga Z, Flammer AJ, Steiger P, et al. Endothelial cell infec­tion and endo­ 1620-1630.
theliitis in COVID-19. Lancet. 2020;395(10234):1417-1418. 33. Spyropoulos AC, Levy JH, Ageno W, et al; Subcommittee on Perioperative,
13. Yu J, Yuan X, Chen H, Chaturvedi S, Braunstein EM, Brodsky RA. Direct acti­ Critical Care Thrombosis, Haemostasis of the Scientific, Standardization
va­tion of the alter­na­tive com­ple­ment path­way by SARS-CoV-2 spike pro­ Committee of the International Society on Thrombosis and Haemosta­
teins is blocked by fac­tor D inhi­bi­tion. Blood. 2020;136(18):2080-2089. sis. Scientific and Standardization Committee com­mu­ni­ca­tion: clin­i­cal
14. Canzano P, Brambilla M, Porro B, et al. Platelet and endo­the­lial acti­va­tion as guid­ance on the diag­no­sis, pre­ven­tion, and treat­ment of venous throm­
poten­tial mech­a­nisms behind the throm­botic com­pli­ca­tions of COVID-19 bo­em­bo­lism in hos­pi­tal­ized patients with COVID-19. J Thromb Haemost.
patients. JACC Basic Transl Sci. 2021;6(3):202-218. 2020;18(8):1859-1865.
15. Mancini I, Baronciani L, Artoni A, et al. The ADAMTS13-von Willebrand fac­tor 34. Giannis D, Allen SL, Tsang J, et  al. Postdischarge throm­bo­em­bolic out­
axis in COVID-19 patients. J Thromb Haemost. 2021;19(2):513-521. comes and mor­tal­ity of hos­pi­tal­ized patients with COVID-19: the CORE-19
16. Delrue M, Siguret V, Neuwirth M, et al. von Willebrand fac­tor/ADAMTS13 axis reg­is­try. Blood. 2021;137(20):2838-2847.
and venous throm­bo­em­bo­lism in mod­er­ate-to-severe COVID-19 patients. 35. Cines DB, Bussel JB. SARS-CoV-2 vac­ cine-induced immune throm­ botic
Br J Haematol. 2021;192(6):1097-1100. throm­bo­cy­to­pe­nia. N Engl J Med. 2021;384(23):2254-2256.
17. Hottz ED, Azevedo-Quintanilha IG, Palhinha L, et al. Platelet acti­va­tion and 36. MacNeil JR, Su JR, Broder KR, et al. Updated rec­om­men­da­tions from the
plate­let-mono­cyte aggre­gate for­ma­tion trig­ger tis­sue fac­tor expres­sion in advi­sory com­ mit­
tee on immu­ ni­
za­
tion prac­tices for use of the Janssen
patients with severe COVID-19. Blood. 2020;136(11):1330-1341. (Johnson and Johnson) COVID-19 vac­cine after reports of throm­bo­sis with
18. Althaus K, Marini I, Zlamal J, et al. Antibody-induced procoagulant plate­lets throm­bo­cy­to­pe­nia syn­drome among vac­cine recip­i­ents—United States,
in severe COVID-19 infec­tion. Blood. 2021;137(8):1061-1071. April 2021. MMWR Morb Mortal Wkly Rep. 2021;70(17):651-656.
19. Middleton EA, He XY, Denorme F, et al. Neutrophil extra­cel­lu­lar traps con­ 37. Greinacher A, Thiele T, Warkentin TE, et al. Thrombotic throm­bo­cy­to­pe­nia
trib­ute to immunothrombosis in COVID-19 acute respi­ra­tory dis­tress syn­ after ChAdOx1 nCov-19 vac­ci­na­tion. N Engl J Med. 2021;384(22):2092-2101.
drome. Blood. 2020;136(10):1169-1179. 38. See I, Su JR, Lale A, et al. US case reports of cere­bral venous sinus throm­bo­
20. Wright FL, Vogler TO, Moore EE, et  al. Fibrinolysis shut­down cor­re­la­tion sis with throm­bo­cy­to­pe­nia after Ad26.COV2.S vac­ci­na­tion, March 2 to April
with throm­bo­em­bolic events in severe COVID-19 infec­tion. J Am Coll Surg. 21, 2021. JAMA. 2021;325(24):2448-2456.
2020;231(2):193-203.e1203e1. 39. Scully M, Singh D, Lown R, et al. Pathologic antibodies to plate­let fac­tor 4
21. Delrue M, Siguret V, Neuwirth M, et al. Contrast between prev­a­lence of HIT after ChAdOx1 nCoV-19 vac­ci­na­tion. N Engl J Med. 2021;384(23):2202-2211.
antibodies and con­firmed HIT in hos­pi­tal­ized COVID-19 patients: a pro­spec­ 40. Nazy I, Sachs UJ, Arnold DM, et al. Recommendations for the clin­i­cal and
tive study with clin­i­cal impli­ca­tions. Thromb Haemost. 2021;121(7):971-975. lab­o­ra­tory diag­no­sis of VITT against COVID-19: com­mu­ni­ca­tion from the
22. Daviet F, Guervilly C, Baldesi O, et al. Heparin-induced throm­bo­cy­to­pe­nia ISTH SSC sub­com­mit­tee on plate­let immu­nol­ogy. J Thromb Haemost.
in severe COVID-19. Circulation. 2020;142(19):1875-1877. 2021;19(6):1585-1588.
23. Nazy I, Jevtic SD, Moore JC, et  al. Platelet-acti­vat­ing immune complexes
iden­ti­fied in crit­i­cally ill COVID-19 patients suspected of hep­a­rin-induced
throm­bo­cy­to­pe­nia. J Thromb Haemost. 2021;19(5):1342-1347.
24. Bowles L, Platton S, Yartey N, et al. Lupus anti­co­ag­u­lant and abnor­mal coag­
u­la­tion tests in patients with COVID-19. N Engl J Med. 2020;383(3):288-290.
25. Siguret V, Voicu S, Neuwirth M, et  al. Are antiphospholipid antibod­
ies asso­ci­ated with throm­botic com­pli­ca­tions in crit­i­cally ill COVID-19 © 2021 by The Amer­i­can Society of Hematology
patients? Thromb Res. 2020;195(Novem­ber):74-76. DOI 10.1182/hema­tol­ogy.2021000298

Prakash Singh Shekhawat


COVID-19 and throm­bo­sis: searching for evi­dence  |  627
THE COVID CRASH: LESSONS LEARNED FROM A WORLD ON PAUSE

Passive immune therapies:


another tool against COVID-19

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/628/1852150/628estcourt.pdf by guest on 13 December 2021


Lise J. Estcourt
NHS Blood and Transplant, John Radcliffe Hospital, Oxford, UK

Passive immune therapy consists of several different therapies, convalescent plasma, hyperimmune globulin, and severe
acute respiratory syndrome coronavirus 2 (SARS-CoV-2) neutralizing monoclonal antibodies. Although these treatments
were not part of any pandemic planning prior to coronavirus disease 2019 (COVID-19), due to the absence of high-quality
evidence demonstrating benefit in other severe respiratory infections, a large amount of research has now been per-
formed to demonstrate their benefit or lack of benefit in different patient groups. This review summarizes the evidence
up to July 2021 on their use and also when they should not be used or when additional data are required. Vaccination
against SARS-CoV-2 is the most important method of preventing severe and fatal COVID-19 in people who have an intact
immune system. Passive immune therapy should only be considered for patients at high risk of severe or fatal COVID-19.
The only therapy that has received full regulatory approval is the casirivimab/imdevimab monoclonal cocktail; all other
treatments are being used under emergency use authorizations. In Japan, it has been licensed to treat patients with mild
to moderate COVID-19, and in the United Kingdom, it has also been licensed to prevent infection.

LEARNING OBJECTIVES
• Summarize current evidence on the use of passive immune therapy to prevent infection—whether it reduces risk
of death or hospitalization
• Summarize current evidence on the use of passive immune therapy for high-risk patients who have mild COVID-19
symptoms—risk of death
• Can state whether passive immune therapy reduces all-cause mortality for hospitalized patients and whether any
subgroups will beneft

(1854-1917) was awarded the frst Nobel Prize in Medicine


CLINICAL CASE 1 “for his work on serum therapy, especially its applica-
A 74-year-old man (case) had a test for severe acute respiratory tion against diphtheria.”1 Today, passive antibody therapy
syndrome coronavirus 2 (SARS-CoV-2) after a family gathering involves treatment with polyclonal antibodies derived from
for the christening of his granddaughter. He had had the test humans (convalescent plasma [CP] or hyperimmune glob-
because he was a close contact for the index case at the chris- ulin), animals (antisera), or antigen-specifc monoclonal
tening. His 49-year-old nephew (index case) had been admit- antibodies (mAbs).2 In this review, I focus on CP and neu-
ted to hospital the day after the christening and was found to tralizing mAbs and the current evidence for their use. There
be SARS-CoV-2 positive. The nephew had had some symp- are no published trials on the use of hyperimmune globulin
toms at the time of the christening but had not thought that in COVID-19.3
these were due to coronavirus disease 2019 (COVID-19). The CP after infection, particularly after severe illness, may
74-year-old man self-isolated with his wife while awaiting the contain high levels of polyclonal pathogen-specifc anti-
results of the test. bodies. These antibodies may confer passive immunity to
recipients and in viral diseases are thought to have their
main action via neutralization of viral particles.4 Collection
Introduction and transfusion of CP can occur rapidly after the onset
Passive antibody therapy is one of the oldest treatments of a pandemic, with collection of plasma occurring from
for infectious diseases that is still in use. Emil von Behring 14 days after a patient has recovered from the infection.

628 | Hematology 2021 | ASH Education Program


Table 1. Comparison between dif­fer­ent pas­sive anti­body ther­a­pies

Characteristic CP Human hyper­im­mune glob­u­lin mAb


Speed of pro­duc­tion after start of Rapid—weeks Slow—months Slowest—months
pan­demic Can be pro­duced once patients Needs time to col­lect plasma from Need to iden­tify poten­tial
have recov­ered from infec­tion a large num­ber of peo­ple who antibodies that would be use­ful
(14 to 28 days after recov­ery) have recov­ered from infec­tion to develop as a mAb and then
man­u­fac­ture anti­body
Can adapt to viral var­i­ants Yes Yes—more slowly than CP No
Number of anti–SARS-CoV-2 Many—poly­clonal Many—poly­clonal One to 2 antibodies
antibodies prod­uct con­tains
Amount of anti­body contained Very var­i­able. Variability can be Fixed amount of total anti­body Fixed amount of neu­tral­iz­ing SARS-
within the prod­uct reduced by using mini-pools— CoV-2 neu­tral­iz­ing anti­body

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/628/1852150/628estcourt.pdf by guest on 13 December 2021


used in Argentina but not all­
countries allowed to pro­duce
mini-pools
Route of admin­is­tra­tion Intravenous Subcutaneous, intra­mus­cu­lar, or Subcutaneous, intra­mus­cu­lar, or
intra­ve­nous intra­ve­nous
Derived from blood* Yes Yes No
Availability Able to be pro­duced in any Not ­able to be pro­duced in every Limited sup­ply due to com­plex
coun­try in which peo­ple have coun­try but manufactur­ing manufactur­ing require­ments
recov­ered from the infec­tion require­ments are not com­plex
and are a­ ble to pro­duce plasma
Cost Relatively cheap—$100 to $200 More expen­sive than CP but Expensive—$1000s per dose
per dose cheaper than mAb—may be Cannot be afforded by low- and
Can be used in low- and mid­dle- ­able to be pro­duced locally, mid­dle-income countries
income countries in low- and mid­dle-income Can only be pro­duced at a lim­ited
countries num­ber of manufactur­ing sites
*Any prod­uct derived from human blood requires viral test­ing to ensure that the trans­fused prod­uct does not cause a trans­fu­sion-trans­mit­ted infec­
tion. In high-income countries with good screen­ing sys­tems, trans­fu­sion-trans­mit­ted infec­tion is very rare. In the United Kingdom, there were no
trans­fu­sion-trans­mit­ted infec­tions reported to the national hemovigilance sys­tem in 2020 (https:​­/​­/www​­.shotuk​­.org​­/wp​­-content​­/uploads​­/myimages​
­/TTI​­-Supplementary​­-material​­-2020​­.pdf).

The anti­body response in CP donors adapts with the virus, either that mono­clo­nal cock­tails are effec­tive and less likely to lead
due to donors becom­ing infected with new var­i­ants of the virus to resis­tance than use of a sin­gle mono­clo­nal. However, even
or due to vac­ci­na­tion after an ini­tial nat­u­ral infec­tion. How- a mono­clo­nal cock­tail could become inef­fec­tive in the future,
ever, the lev­els of neu­tral­iz­ing anti­body can vary sig­nif­i­cantly as shown by the pro­ spec­tive map­ ping of viral var­ i­
ants that
from 1 unit to the next, and a min­i­mum thresh­old of anti­body is detected a poten­tial muta­tion (E406W) that could escape neu­
required within each unit to ensure that the trans­fu­sion con­tains tral­i­za­tion by both com­po­nents of the casirivimab/imdevimab
a suf­fi­cient level of neu­tral­iz­ing anti­body (Table 1). mono­clo­nal cock­tail, as well as both com­po­nents of the bam-
Hyperimmune glob­u­lin is cur­rently used to pro­tect vul­ner­ lanivimab/etesevimab cock­tail (Table 2).7 Monoclonal ther­apy is
a­ble indi­vid­u­als from other viral infec­tions, includ­ing var­i­cella expen­sive and requires very spe­cial­ized manufactur­ing units. It is
zoster.5 It is pro­duced by pooling thou­sands of dona­tions from there­fore a treat­ment that most low- and mid­dle-income coun-
peo­ple who have recov­ered from an infec­tion or have been vac­ tries can­not afford and will find more dif­fi­cult to man­u­fac­ture
ci­nated against an infec­tion and have high lev­els of antibodies. locally. CP can be pro­duced in many countries and is much more
It pro­duces a con­sis­tent prod­uct that always has a defined level afford­able.8 Therefore, when think­ing about whether to use pas­
of anti­body within it. However, it takes time to pro­duce, so it sive immu­ni­za­tion ther­apy, con­sid­er­ation needs to be made not
can­not be used as early in a pan­demic. It will also evolve with just on its effec­tive­ness but also on its acces­si­bil­ity.
changes with the viral var­i­ant but not as rap­idly as CP.
Neutralizing mono­ clo­ nal anti­ body ther­ apy can be derived Prophylactic ther­apy
from humans who have had an infec­tion, or been vac­ci­nated, or The main­stay of pre­vent­ing infec­tion in peo­ple with an intact
from human­ized mice that have been exposed to SARS-CoV-2 immune sys­tem is vac­ci­na­tion. Active immu­ni­za­tion against SARS-
anti­gens.2 Monoclonal anti­body pro­duc­tion can iden­tify antibod- CoV-2 has been shown to be effec­tive at pre­vent­ing severe and
ies with a high level of neu­tral­iz­ing activ­ity and can be pro­duced fatal COVID-19, as well as reduc­ing the risk of symp­tom­atic COVID-
with­out the need for blood donors. mAbs will, how­ever, not 19.9 In the United States, by the end of June 2021, ­vac­ci­na­tion had
adapt to viral var­i­ants, and so over time, the virus can become averted an esti­mated 279 000 deaths and up to 1.25 mil­lion
resis­tant to the mono­clo­nal anti­body. This has already hap­pened hos­pi­tal­i­za­tions.10 However, vac­ci­na­tion is not as effec­tive in
with the SARS-CoV-2 virus (Table 2). Monotherapy with bam- patients with an impaired immune sys­tem, either due to immu­
lanivimab has had its emer­gency use autho­ri­za­tion (EUA) with­ no­sup­pres­sive ther­apy or an under­ly­ing dis­ease that affects the
drawn due to devel­op­ment of viral resis­tance.6 This may mean immune sys­tem. Patients with hema­to­logic ­malig­nan­cies mount
Prakash Singh Shekhawat
Passive immune ther­a­pies: tool against COVID-19  |  629
Table 2. Different types of mAb in clin­i­cal use

Clinical In clin­i­cal use Competent


Route of trial (out­side of Viral resis­tance author­ity
Name of mono­clo­nal Site of action admin­is­tra­tion results* clin­i­cal tri­als) detected approval
Bamlanivimab Blocks bind­ing of SARS- IV Yes Previously—FDA Yes EUA (now revoked
CoV-2 to the ACE2 EUA revoked Marked—gamma (P.1) in United States)
recep­tor by targeting April 2021 and beta (B.1.351)
the RBD on the spike VoCs
pro­tein of SARS-CoV-2 Modest—delta (B.1.617.2)
VoC
Bamlanivimab plus Blocks bind­ing of IV Yes Yes—use paused Yes EUA†
etesevimab SARS-CoV-2 to the in United Marked—gamma (P.1)
ACE2 recep­tor by States and beta (B.1.351) VoC

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/628/1852150/628estcourt.pdf by guest on 13 December 2021


targeting dif­fer­ent but Modest—Delta (B.1.617.2)
overlapping epi­topes VoC
on the spike pro­tein
RBD of SARS-CoV-2
Casirivimab plus Blocks bind­ing of SARS- IV or S/C Yes Yes Yes—only to casirivimab Yes—MHRA and
imdevimab CoV-2 to the ACE2 Marked—beta (B.1.351) Jap­a­nese
recep­tor by targeting VoC reg­u­la­tory
dif­fer­ent epi­topes on agency†
the spike pro­tein RBD
of SARS-CoV-2 (do not
over­lap)
Sotrovimab Blocks bind­ing of SARS- IV, trialing IM Yes Yes No EUA†
CoV-2 to the ACE2
recep­tor by targeting
an epi­tope on the RBD
of the spike pro­tein
that is con­served
between SARS-CoV
and SARS-CoV-2
Regdanvimab Blocks bind­ing of SARS- IV Yes Yes Yes EUA‡
CoV-2 to the ACE2 Marked—beta (B.1.351)
recep­tor by targeting VoC
an epi­tope on the RBD
of the spike pro­tein of
SARS-CoV-2
*Results avail­­able in Table 2 for all­-cause mor­tal­ity and need for hos­pi­tal­i­za­tion (by day 30) in Table 3 for out­pa­tients.
†EUA in the United States as well as other countries. In the United States, the indi­ca­tion is for mild or mod­er­ate COVID-19 not requir­ing oxy­gen ther­apy.
‡EUA in South Korea and Indonesia, but indi­ca­tions for use dif­fer.
FDA, Food and Drug Administration (United States); IM, intra­mus­cu­lar; IV, intra­ve­nous; MHRA, Medicines and Healthcare prod­ucts Regulatory Agency
(United Kingdom); RBD, recep­tor-bind­ing domain; S/C, sub­cu­ta­ne­ous; VoC, var­i­ant of con­cern.

blunted anti­ body responses to SARS-CoV-2 vac­ ci­


na­tion, and dif­fer­ence in mor­tal­ity (Figure 1). However, bamlanivimab is not
those most at risk appear to be patients who are actively treated effec­tive against many of the viral var­i­ants cur­rently in cir­cu­la­
with Bruton tyro­sine kinase inhib­i­tors, ruxolitinib, venetoclax, or tion. Ongoing stud­ies of CP, hyper­im­mune glob­u­lin, and mAbs
anti-CD20 anti­body ther­a­pies.11 Consideration there­ fore needs assessing high-risk pro­phy­laxis will be assessed within liv­ing sys­
to be given to pre­vent­ing infec­tion in peo­ple who are unvac­ci­ tem­atic reviews, so addi­tional infor­ma­tion may be avail­­able over
nated or par­tially vac­ci­nated and are at high risk of devel­op­ing the next few months.23,24 It is espe­cially impor­tant to know their
severe and fatal COVID-19 due to comorbidities or those who addi­tional impact over and above ­vac­ci­na­tion for those indi­vid­
have been fully vac­ci­nated but are unable to mount an effec­ u­als who have a poor response to vac­ci­na­tion, for exam­ple, the
tive immune response to vac­ci­na­tion. There have been 2 com­ immunosuppressed pop­u­la­tion.11
pleted tri­als assessing mono­clo­nal ther­apy12,13 (Table 3), with 1
trial assessing the effect of 1.2 g of the casirivimab/imdevimab
cock­tail (sub­cu­ta­ne­ously via 4 injec­tions) given to house­hold
con­tacts of a pos­i­tive case.13 However, most of these indi­vid­ CLINICAL CASE (Con­tin­ued)
u­als were not at high risk of severe dis­ease, with fewer than The patient and his wife received the results of the poly­mer­
10% older than 65 years and only 1% with immune sup­pres­sion. ase chain reac­tion test the next day, and both had tested pos­i­
The other trial used bamlanivimab, which showed a ben­e­fit in tive for SARS-CoV-2. In total, 15 of the 45 peo­ple who attended
nurs­ing home res­i­dents who received the inter­ven­tion in pro­ the chris­ten­ing tested pos­i­tive for SARS-CoV-2 within the days
gres­sion to mod­er­ate or severe dis­ease but no evi­dence of a fol­
low­ing the chris­ ten­
ing. He started to develop symp­ toms

630  |  Hematology 2021  |  ASH Education Program


Table 3. Completed ran­dom­ized con­trolled tri­als of anti-SARS-CoV-2 mAb ther­apy

Number Need for hos­pi­


ran­dom­ized Viral var­i­ants tal­i­za­tion at 30
up to data Number Patient con­sid­ered in Mortality at 30 days or death
Study Country Intervention(s) cut­off ana­lyzed pop­u­la­tion Primary out­come Type of anal­y­sis ana­ly­ses* days (GRADE)† (GRADE)†
Prophylaxis
O’Brien et al‡13 Moldova, S/C casirivimab/ 753 1505 Age ≥12 years Incidence of Interim No NR NR
NCT04452318 Romania, imdevimab 1.2 g Household con­tact symp­tom­atic Planned Recruited
United (0.6 g + 0.6 g) of con­firmed COVID-19 recruit­ment ??-Jan­u­ary 2021
States SARS-CoV-2 case 3750
Placebo 752
SARS-CoV-2 PCR
and anti­body
neg­a­tive

BLAZE-212 United States Bamlanivimab 588 966 Residents and Incidence of Final No RR, 0.83 NR
NCT04497987 (4.2 g) staff at 74 skilled COVID-19— Recruited 95% CI,
nurs­ing viro­log­i­cal or August- 0.25-2.70
Placebo 587
and assisted liv­ing clin­i­cal Novem­ber (low)
facil­i­ties 2020
Within 7 days
of a reported
con­firmed SARS-
CoV-2 case
Outpatients (asymp­tom­atic)
O’Brien et al14 United States S/C casirivimab/ 155 311 Age ≥12 years Incidence of Interim July 2020 to NR RR, 0.14
NCT04452318‡ imdevimab 1.2 g Confirmed SARS- symp­tom­atic Planned Jan­u­ary 2021 95% CI, 0.01-2.76

Prakash Singh Shekhawat


(0.6 g + 0.6 g) CoV-2 COVID-19 recruit­ment (low)
SARS-CoV-2 3750
Placebo 156
anti­body
neg­a­tive
Asymptomatic
Outpatients (mild dis­ease)
Weinreich et al Chile, Mexico, Casirivimab/ 838 4057 Adult Clinical—COVID- Interim No RR, 1.02 RR, 0.30
(phase 3)15 Romania, imdevimab 1.2 g Confirmed SARS- related Planned Recruited 95% CI, 0.06- 95% CI, 0.13-0.68
NCT04425629‡ United (0.6 g + 0.6 g) CoV-2 hos­pi­tal­i­za­tion recruit­ment Sep­tem­ber 16.22 (low)
States ≤7 days from onset or death 6420 2020 to (low)
symp­toms (day 29) Subgroup— Jan­u­ary 2021
Casirivimab/ 1529 RR, 0.33 RR, 0.29
≥1 risk fac­tor anti­body
imdevimab 2.4 g 95% CI, 0.01- 95% CI, 0.17-0.48
COVID-19 neg­a­tive
(1.2 g + 1.2 g) 3.94 (mod­er­ate)
Mild symp­toms
(low)
(WHO 2-3)
Placebo 1500 Excluded — —
Hospitalized (any
rea­son)
Vaccinated or plan
to be vac­ci­nated

Passive immune ther­a­pies: tool against COVID-19  |  631


Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/628/1852150/628estcourt.pdf by guest on 13 December 2021
Table 3. (continued)

Number Need for hos­pi­


ran­dom­ized Viral var­i­ants tal­i­za­tion at 30
up to data Number Patient con­sid­ered in Mortality at 30 days or death
Study Country Intervention(s) cut­off ana­lyzed pop­u­la­tion Primary out­come Type of anal­y­sis ana­ly­ses* days (GRADE)† (GRADE)†
COMET-ICE‡16 Brazil, Sotrovimab (0.5 g) 291 583 Adult Clinical—dis­ease Interim No RR, 0.33 RR, 0.14
NCT04545060 Canada, Confirmed SARS- pro­gres­sion Stopped Recruited 95% CI, 0.01- 95% CI,
Placebo 292
Spain, CoV-2 recruit­ment August 2020 8.18 0.04-0.48
United ≤5 days from onset due to to March (low) (low)
States symp­toms effi­cacy 1057 2021
>55 years or par­tic­i­pants
comorbidities
Mild symp­toms
(WHO 2-3)
Excluded
Likely to require
hos­pi­tal­i­za­tion
≤24 h
Likely to die ≤7
days

632  |  Hematology 2021  |  ASH Education Program


Severely immu­no­
com­pro­mised
Vaccinated or plan
to be vac­ci­nated
within 4 weeks
BLAZE-117 Puerto Rico, Bamlanivimab 104 577 Adult Virological—viral Interim No No events RR, 0.17
NCT04427501 United (0.7 g) Confirmed SARS- clear­ance Planned Recruited June- (low) 95% CI, 0.02-1.33
States CoV-2 recruit­ment Sep­tem­ber (low)
Mild symp­toms 3160 2020
Bamlanivimab 109 RR, 0.32
(WHO 2-3)
(2.8 g) 95% CI, 0.07-1.47
Excluded
(low)
Requires oxy­gen
Bamlanivimab 104 ther­apy RR, 0.34
(7.0 g) Any seri­ous 95% CI,
con­com­i­tant 0.08-1.56
sys­temic dis­ease (low)
Pregnant or
Bamlanivimab/ 114 breastfeeding See updated See updated
etesevimab results results below
(2.8 g + 2.8 g) below
Placebo 161 — —
BLAZE-118 United States Bamlanivimab/ 518 1035 Adult Clinical—COVID- Interim No RR, 0.05 RR, 0.30
NCT04427501 etesevimab Confirmed SARS- 19–related Planned Sep­tem­ber- 95% CI, 95% CI, 0.16-0.59
(2.8 g + 2.8 g) CoV-2 hos­pi­tal­i­za­tion recruit­ment Decem­ber 0.00-0.81 (low)
Mild symp­toms (acute care 3160 2020 (low)
Placebo 517
(WHO 2 to 3) for ≥24 hours)
Excluded or death from
Requires oxy­gen any cause by
ther­apy day 29
Any seri­ous
con­com­i­tant
sys­temic dis­ease
Pregnant or
breastfeeding

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/628/1852150/628estcourt.pdf by guest on 13 December 2021


Table 3. (continued)

Number Need for hos­pi­


ran­dom­ized Viral var­i­ants tal­i­za­tion at 30
up to data Number Patient con­sid­ered in Mortality at 30 days or death
Study Country Intervention(s) cut­off ana­lyzed pop­u­la­tion Primary out­come Type of anal­y­sis ana­ly­ses* days (GRADE)† (GRADE)†
Eom et al‡19 Romania, Regdanvimab 101 327 Adult Clinical—time Interim No No events RR, 0.45
NCT04602000 South 0.04 g/kg Confirmed SARS- to clin­i­cal Planned Recruited (low) 95% CI, 0.14-1.42
Korea, CoV-2 recov­ery recruit­ment Octo­ber- (low)
Spain, ≤7 days from onset Virological—viral 1172 Decem­ber
Regdanvimab 103 RR, 0.56
United symp­toms clear­ance 2020
0.08 g/kg 95% CI, 0.19-1.60
States Excluded
(low)
Hospitalized
Placebo 103 —
Weinreich et al Chile, Mexico, Casirivimab/ 92 275 Adult Virological—viral Interim No NR RR, 0.43
(phase 1/2)20 Romania, imdevimab 2.4 g Confirmed SARS- clear­ance Recruited 95% CI, 0.08-2.19
NCT04425629 United (1.2 g + 1.2 g) CoV-2 June- August (low)
States ≤7 days from onset 2020
Casirivimab/ 90 RR, 0.21
symp­toms
imdevimab 8 g 95% CI, 0.02-1.79
Excluded
(4 g + 4 g) (low)
Hospitalized
Placebo 93 —
Inpatients (mod­er­ate or severe dis­ease)
RECOVERY‡21 United Casirivimab/ 4839 9185 Any age Clinical— Complete No RR, 0.94 NA
NCT04381936 Kingdom imdevimab 8 g Suspected or con­ all-cause Subgroup— Recruited 95% CI, 0.87-
(4 g + 4 g) firmed COVID-19 mor­tal­ity anti­body Sep­tem­ber 1.02
Median 9 days day 28 neg­a­tive 2020 to May (mod­er­ate)
Standard care 4946

Prakash Singh Shekhawat


symp­tom onset 2021
>90% requir­ing
oxy­gen ther­apy
(WHO score ≥5)
ACTIV-322 Denmark, Bamlanivimab (7 g) 314 Inpatients, Adult Clinical—time Interim anal­y­sis, No RR, 1.39 NA
NCT04501978 India, mod­er­ate Confirmed SARS- to sustained bamlanivimab Recruited 95% CI, 0.07-
Poland, dis­ease CoV-2 recov­ery arm stopped August to 1.47
Singapore, ≤12 days from onset for futil­ity, Octo­ber (low)
Spain, symp­toms recruit­ment 2020
Switzerland, Excluded ongo­ing for
United Requiring organ other arms
Kingdom, sup­port
United Pregnant/breast
States feed­ing
*Was the type of virus (eg, alpha, beta, gamma, delta) detected at base­line taken into con­sid­er­ation as a sub­group anal­y­sis?
†GRADE assess­ment—assesses cer­tainty of the evi­dence. High cer­tainty: very con­fi­dent that the true effect lies close to that of the esti­mate of the effect. Moderate cer­tainty: mod­er­ately
con­fi­dent in the effect esti­mate: The true effect is likely to be close to the esti­mate of the effect, but there is a pos­si­bil­ity that it is sub­stan­tially dif­fer­ent. Low cer­tainty: con­fi­dence in the effect
esti­mate is lim­ited: the true effect may be sub­stan­tially dif­fer­ent from the esti­mate of the effect. Very low cer­tainty: very lit­tle con­fi­dence in the effect esti­mate: The true effect is likely to be
sub­stan­tially dif­fer­ent from the esti­mate of effect.
‡Pre–peer review.
NA, not applicable; NR, not reported; PCR, poly­mer­ase chain reac­tion; WHO, World Health Organization.

Passive immune ther­a­pies: tool against COVID-19  |  633


Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/628/1852150/628estcourt.pdf by guest on 13 December 2021
Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/628/1852150/628estcourt.pdf by guest on 13 December 2021
Figure 1. SARS-CoV-2 mAb vs placebo or standard care—all-cause mortality at 28 days or hospital discharge.

of fever and a dry cough the day after his pos­it­ive test, but by the para­med­ics and taken to the local emer­gency room. In
his symp­toms were mild, and he treated the fever with parac- the emer­gency room, he was given ste­roids and admit­ted to
etamol and rested at home. the hos­pi­tal for oxy­gen ther­apy.

Treatment with asymp­tom­atic infec­tion Treatment with mod­er­ate or severe symp­toms


or mild symp­toms Two tri­als have assessed mono­clo­nal anti­body use in mod­er­ately
Vaccination is known to decrease the risk of pro­ gres­
sion to or severely unwell patients; nei­ther showed a ben­e­fit for patients
mod­er­ate or severe COVID-19 dis­ease, although it has less of an over­all (Figure 1), but there was a ben­e­fit for patients who had not
impact on the devel­op­ment of asymp­tom­atic or mild symp­toms. yet devel­oped a detect­able anti­body response (Figure 3). Those
All of the cur­rent evi­dence for pas­sive immune ther­apy is prior to patients who did not develop an anti­body response had a much
vac­ci­na­tion, or patients who were vac­ci­nated or about to be vac­ higher mor­tal­ity rate than those who did not.19,21 This is partly
ci­nated were excluded from the tri­als. Data from Public Health because patients who have a delayed anti­body response are
England have shown that vac­ci­na­tion decreases the risk of hos­ older and have more comorbidities. This does mean that reduc­
pi­tal­i­za­tion by over 90%, even with the delta var­i­ant. It is there­ ing mor­tal­ity in this group will mean a larger abso­lute reduc­tion
fore more dif­fi­cult to know the real addi­tional effect of pas­sive in mor­tal­ity; for exam­ple, based on the RECOVERY data, 54 lives
immune ther­apy over and above vac­ci­na­tion (Tables 3 and 4). per 1000 patients treated would be saved (95% CI, 24-80 lives
In the stud­ies that assessed bamlanivimab alone or regdan- saved per 1000 patients treated). Some SARS-CoV-2 var­i­ants are
vimab, there were no deaths in any of the study arms; there­ resis­ tant to bamlanivimab monotherapy, whereas the cur­ rent
fore, any effect on all­-cause mor­tal­ity can­not be assessed.42 The major var­i­ants in cir­cu­la­tion are still sen­si­tive to neu­tral­i­za­tion by
only study that showed a reduc­tion in mor­tal­ity was the BLAZE-1 the casirivimab/imdevimab cock­tail. However, this may change
trial17 arm that used the bamlanivimab/etesevimab mono­ clo­ with the devel­op­ment of new var­i­ants.7 As var­i­ant screen­ing can­
nal cock­tail (Figure 1); the casirivimab/imdevimab cock­tail trial not be done in real time, pas­sive anti­body treat­ments need to
showed a trend in the direc­tion of effect, but it was not clin­i­cally be used that are effec­tive against all­cur­rent var­i­ants in a par­tic­
sig­nif­i­cant,15,20 nor was the effect of high-dose CP (Figure 2).3,8 u­lar region of the world. CP use does not show a ben­e­fit over­all
Therefore, although it is sug­ges­tive that pas­sive immune ther­ for patients with mod­er­ate or severe COVID-19.29,30,39 CP has also
apy if given early could save lives and the reduce risk of severe been assessed in anti­body-neg­at­ive patients in 2 major tri­als,
dis­ease, addi­tional data are required before it can be used in RECOVERY and REMAP-CAP.29,39 This shows a sim­i­lar trend in the
rou­tine prac­tice. None of the stud­ies have performed a cost- direc­tion of a pos­si­ble effect in CP, but it does not reach sta­
effec­tive­ness anal­y­sis, but it is likely that those patients who do tis­ti­cal sig­nif­i­cance (risk ratio [RR], 0.93; 95% CI, 0.86-1.01). This
not respond to vac­ci­na­tion and are at high risk of severe or fatal may partly be because CP is a much more var­i­able prod­uct, with
COVID-19 are the group that will dem­on­strate a ben­e­fit. some units hav­ing much lower anti­body lev­els than oth­ers, so
even if a min­i­mum titer is used within the tri­als, this may not have
been suf­fi­cient. Several tri­als have shown an effect in a sub­group
of par­tic­i­pants who received a higher titer prod­uct.8,32
CLINICAL CASE (Con­t in­u ed)
Seven days after the chris­ ten­
ing, he devel­ oped increas­ ing
short­ness of breath and called an ambu­lance. On exam­i­na­tion
by the para­med­ics, he was pyrex­ial (tem­per­a­ture 38.9°C), had a CLINICAL CASE (Con­tin­ued)
respi­ra­tory rate of 25 breaths per min­ute, and was hyp­oxic with Over the next 2 days, the 74-year-old man con­tin­ued to dete­ri­o­
an oxy­gen sat­u­ra­tion of 91% on room air. He was given oxy­gen rate and was admit­ted to inten­sive care for non­in­va­sive ven­ti­la­tory

634  |  Hematology 2021  |  ASH Education Program


Table 4. Completed ran­dom­ized con­trolled tri­als of CP and hyper­im­mune glob­u­lin ther­apy

Viral var­i­ants
Randomized Number Neutralizing anti­body con­sid­ered in
Study Country Intervention(s) per arm ana­lyzed Patient pop­u­la­tion Primary out­come titer ana­ly­ses*
Prophylaxis
No com­pleted stud­ies
Outpatients (asymp­tom­atic)
No com­pleted stud­ies
Outpatients (mild dis­ease)
Libster et al8 Argentina 250 mL CP on 80 160 Age >74 or 65 to 74 and Development of severe Anti–S IgG SARS-CoV-2 No
NCT04479163 day 1 comorbidity dis­ease—defined as (COVIDAR IgG) Recruited
Confirmed SARS-CoV-2 RR ≥30 breaths/min min­i­mum titer 1:1000 June-Octo­ber 2020
Saline (Placebo) 80
Mild ill­ness, not requir­ing or oxy­gen
hos­pi­tal­i­za­tion sat­u­ra­tions <93%
≤48 h from symp­tom onset on air
Inpatients (mod­er­ate dis­ease (WHO 4 or 5)
Agarwal et al25 India Two doses 235 464 Adult Composite all­- NAb not used to select No
CTRI/2020/04/024775 200 mL CP, Confirmed SARS-CoV-2 cause mor­tal­ity plasma, tested at Recruited
24 h apart, SpO2 ≤93% and RR >24/min or pro­gres­sion to end of study—63% April-July 2020
pref­er­a­bly or PaO2/FiO2 200-300 severe dis­ease of donors had NAb
dif­fer­ent Excluded (PaO2/FiO2 <100) titer >1:20 with
donors Critically ill (PaO2/FiO2 within day 28 median titer 1:40
<200 or shock requir­ing
Standard care 229
vaso­pres­sors)

Prakash Singh Shekhawat


Simonovich et al26 Argentina 10-15 mL/kg 228 333 Adult Clinical sta­tus at day Anti–S IgG SARS-CoV-2 No
NCT04383535 Mini-pools (5-10 Confirmed SARS-CoV-2, 30 ordi­nal categories (COVIDAR IgG) Recruited
donors) requir­ing hos­pi­tal­i­za­tion, 1: death Median titer of 1:3200 May-August 2020
pneu­mo­nia, plus SpO2 2: inva­sive ven­ti­la­tory (IQR, 1:800 to
Saline (pla­cebo) 105
<93% or PaO2/FiO2 <300 sup­port 1:3200)
Excluded 3: hos­pi­tal­ized with
MV or NIV sup­ple­men­tal
oxy­gen require­ments
4: hos­pi­tal­ized with­out
sup­ple­men­tal
oxy­gen require­ments
5: discharged with­out
full return of base­line
phys­i­cal func­tion
6: discharged with full
return of base­line
phys­i­cal func­tion

Passive immune ther­a­pies: tool against COVID-19  |  635


Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/628/1852150/628estcourt.pdf by guest on 13 December 2021
Table 4. (continued)

Viral var­i­ants
Randomized Number Neutralizing anti­body con­sid­ered in
Study Country Intervention(s) per arm ana­lyzed Patient pop­u­la­tion Primary out­come titer ana­ly­ses*
27
Avendaño-Solà et al† Spain 250-300 mL CCP 38 81 Adult Proportion of patients NAb not avail­­able No
NCT04345523 on day 1 Confirmed SARS-CoV-2 in cat­e­gory 5, 6, to select plasma, Recruited
Radiological changes or or 7 of 7-cat­e­gory all­ dona­tion on April-July 2020
Standard care 43
clin­i­cal fea­tures plus SpO2 COVID-19 ordi­nal sub­se­quent test­ing
<94%, <12-day symp­tom scale at day 15 had VMNT-ID50 > 1:80
onset
Excluded
MV, high-flow O2
AlQahtani et al28 Bahrain Two doses 20 40 Age ≥21 Requirement for NR No
NCT04356534 200 mL CCP, Confirmed SARS-CoV-2 ven­ti­la­tion Recruited
24 h apart Pneumonia, plus SpO2 <92% (NIV or MV) April-June 2020
or PaO2/FiO2 <300
Standard care 20
Excluded
MV or MOF
Inpatients (mod­er­ate or severe dis­ease—WHO 4 to 7)

636  |  Hematology 2021  |  ASH Education Program


RECOVERY 202129 United Two doses 5795 11 558 Any age Clinical—all­-cause Minimum Euroimmun 6 Partially
NCT04381936 Kingdom 275 mL CCP, Suspected or con­firmed mor­tal­ity day 28 Used sur­ro­gate
24 h apart COVID-19 of ran­dom­ized
Median 9 days symp­tom before/after
Standard care 5763
onset Decem­ber 2020
>90% requir­ing oxy­gen for WT/alpha
ther­apy (WHO score ≥5) var­i­ant
Recruited
May 2020 to
Jan­u­ary 2021
CONCOR-1†30 Brazil, Canada, 500 mL CCP 614 921 Adult Intubation or death Viral neu­tral­iz­ing No
NCT04348656 United Confirmed SARS-CoV-2 by day 30 antibodies titer Recruited
Standard care 307
States Requiring oxy­gen ther­apy >1:160 or antibodies May 2020 to
(WHO score ≥5) against the RBD Jan­u­ary 2021
Excluded of the SARS-CoV-2
Symptoms >12 days spike pro­tein titer
MV >1:100
O’Donnell et al31 Brazil, United 200-250 mL CCP 150 223 Adult Clinical sta­tus at Anti–SARS-CoV-2 No
NCT04359810 States Confirmed SARS-CoV-2 day 28 fol­low­ing antispike IgG Recruited
200-250 mL 73
Requiring oxy­gen ther­apy ran­dom­i­za­tion anti­body titer ≥1:400 April-Novem­ber
non­im­mune
(WHO score ≥5) (7-point ordi­nal scale 2020
plasma
based on WHO)

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/628/1852150/628estcourt.pdf by guest on 13 December 2021


Table 4. (continued)

Viral var­i­ants
Randomized Number Neutralizing anti­body con­sid­ered in
Study Country Intervention(s) per arm ana­lyzed Patient pop­u­la­tion Primary out­come titer ana­ly­ses*
CAPSID†32 Germany CCP (250- 53 105 Age 18-75 years Composite end point Median PRNT50 No
NCT04433910 325 mL) on Confirmed SARS-CoV-2 of sur­vival and no neu­tral­i­za­tion titer Recruited
days 1, 3, and 5 RR >30 or requir­ing oxy­gen lon­ger fulfilling 1:160 (IQR, 1:80 to August-Decem­ber
ther­apy (WHO score ≥5) cri­te­ria of severe 1:320) 2020
Standard care, 52
COVID-19 (day 21)
cross­over to
CCP at day 14
if not improved
Gharbharan et al33 The 300 mL CCP on 43 86 Adult Mortality until Minimum of PRNT50 No
NCT04342182 Netherlands day 1 Confirmed SARS-CoV-2 dis­charge or titer of ≥1:80 Recruited
within 96 h max­i­mum of 60 days April-June 2020
Standard care 43
Excluded
Patients on MV >96 h
Ray et al†34 India Two doses 40 80 Adult All-cause mor­tal­ity at Euroimmun ≥1.5
CTRI/2020/05/025209 200 mL CCP, Confirmed SARS-CoV-2 30 days
24 h apart RR >30, SpO2 <90%,
PaO2/FiO2 <300
Standard care 40
Excluded
preg­nant, MV
Bennett-Guerrero et al35 United States 480 mL (2 units) 59 74 Adult Number of days patient Ideally >1:320, but No
NCT04344535 CCP Confirmed SARS-CoV-2 remained ven­ti­la­tor meet­ing min­i­mum Recruited
Excluded free (up to 28 days) titer per FDA April-August 2020
480 mL (2 units) 15
Pregnant/breastfeeding
non­im­mune

Prakash Singh Shekhawat


plasma
Pouladzadeh et al36 Iran 500 mL CCP 30 60 Confirmed SARS-CoV-2 Improvement in the NR No
IRCT20200310046736N1 WHO score >4 lev­els of cyto­kine Recruited
Standard care 30
Excluded storm indi­ces March-May 2020
Pregnant/breastfeeding
Comorbidities (eg, heart,
liver, kid­ney dis­ease)
Smokers
Hamdy Salman and Ail Egypt 250 mL CP on 15 30 Adult At least 50% NAb not used to select No
Mohamed37 day 1 Confirmed SARS-CoV-2 improve­ment of the plasma Recruited
NCT04530370 2 or more of RR ≥24, SpO2 sever­ity of ill­ness at June-August 2020
Saline (pla­cebo) 15
≤93%, PaO2/FiO2 <300, any time dur­ing 5-
pul­mo­nary infil­trates day study period
Excluded
MOF, sep­tic shock
Bajpai et al†38 India Two doses 14 29 Age 18-65 years Proportion of patients Variable No
NCT04346446 250 mL CCP, Confirmed SARS-CoV-2 remaining free Recruited
24 h apart Pneumonia, plus SpO2 <93% of mechan­i­cal April-May 2020
or PaO2/FiO2 <300 ven­ti­la­tion day 7
Nonimmune 15
Excluded
plasma
Comorbidities (kid­ney,
heart or liver dis­ease,
COPD)

Passive immune ther­a­pies: tool against COVID-19  |  637


Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/628/1852150/628estcourt.pdf by guest on 13 December 2021
Table 4. (continued)

Viral var­i­ants
Randomized Number Neutralizing anti­body con­sid­ered in
Study Country Intervention(s) per arm ana­lyzed Patient pop­u­la­tion Primary out­come titer ana­ly­ses*
Inpatients (severe dis­ease—WHO 6 to 7)
REMAP-CAP†39 Australia, Two doses 1084 2000 Adult Organ sup­port–free Minimum Euroimmun 6 No
NCT02735707 Canada, 275 mL CCP, Confirmed SARS-CoV-2 days at day 21 Recruited
United 24 h apart ≤48 h since admis­sion to ICU May 2020 to
Kingdom, Jan­u­ary 2021
Standard care 916
United
States
Gonzalez et al†40 Mexico Two doses 130 90 Adult (16-90 years) Duration of NAb not used to select No
NCT04381858 200 mL CCP, Suspected or con­firmed hos­pi­tal­i­za­tion plasma Recruited
24 h apart COVID-19 All-cause mor­tal­ity May-Octo­ber 2020
Severe respi­ra­tory fail­ure day 28
IVIg 0.3 g/kg 60
daily for 5 days
Li et al41 China 4-13 mL/kg of CP 52 103 Confirmed SARS-CoV-2 Time to clin­i­cal Minimum of S-RBD– No
ChiCTR2000029757 Excluded improve­ment spe­cific IgG of 1:640 Recruited

638  |  Hematology 2021  |  ASH Education Program


Standard care 51
high-titer S-RBD–spe­cific (patient dis­charge or (approx­i­ma­tely Feb­ru­ary-April 2020
IgG (≥1:640) reduc­tion 2 points equiv­a­lent to NAb
on 6-point dis­ease of 1:40)
sever­ity scale)
*Was the type of virus (e.g., alpha, beta, gamma, delta, etc.) detected at base­line taken into con­sid­er­ation as a sub­group anal­y­sis?
†Pre–peer review.
CCP, COVID-19 con­va­les­cent plasma; COPD, chronic obstruc­tive pul­mo­nary dis­ease; FDA, Food and Drug Administration; FiO2, fraction of inspired oxygen; ICU, inten­sive care unit; IQR,
interquartile range; IVIg, intra­ve­nous immu­no­glob­u­lin; MOF, multiorgan fail­ure; MV, mechan­i­cal ven­ti­la­tion; NAb, neu­tral­iz­ing anti­body; NIV, non-inva­sive ven­ti­la­tion; PaO2, partial pressure of
oxygen; PRNT50, 50% reduction in plaque count using the plaque reduction neutralization test; RR, respiratory rate; SpO2, oxygen saturation; VMNT, virus microneutralization test; WT, wild type.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/628/1852150/628estcourt.pdf by guest on 13 December 2021


Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/628/1852150/628estcourt.pdf by guest on 13 December 2021
Figure 2. CP vs placebo or standard care—all-cause mortality at 28 days or hospital discharge.

Figure 3. SARS-CoV-2 mAb vs placebo or standard care—all-cause mortality at 28 days or hospital discharge by antibody status
at baseline.

Prakash Singh Shekhawat


Passive immune ther­a­pies: tool against COVID-19  |  639
sup­port. An emer­gency use access request for CP was made, and The virus con­tin­ues to change, and so although treat­ments
he received CP on day 10 after the chris­ten­ing. may be effec­tive against cur­rent SARS-CoV-2 viral var­ia ­nts of
con­cern, this may not be true in the future. Passive immune ther­
a­pies will either have to be very broad spec­trum or adapt with
Treatment of crit­i­cally unwell par­tic­i­pants the virus.
Fewer tri­ als have spe­ cif­i­
cally assessed inter­ ven­
tions for the
crit­i­cally unwell patients (requir­ing respi­ra­tory or car­dio­vas­cu­ Conflict-of-inter­est dis­clo­sure
lar organ sup­port) with an inten­sive care level of care (Tables 1 Lise J. Estcourt: author on Cochrane liv­ing sys­tem­atic reviews
and 2). Several tri­ als exclude patients requir­ ing mechan­ i­
cal of mono­clo­nal ther­ap
­ ies and CP. Investigator on the RECOVERY
ven­ti­la­tion or organ sup­port of any type. One of the tri­als that and REMAP-CAP tri­als.
has focused on this patient group is the REMAP-CAP trial.39 It
showed no ben­e­fit of CP over­all (Figure 2), but a prespecified Off-label drug use
sub­group showed poten­tial ben­e­fit of CP. This trial, based on Lise J. Estcourt: nothing to declare.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/628/1852150/628estcourt.pdf by guest on 13 December 2021


Bayes­ian sta­tis­tics, showed an 89.9% pos­te­rior prob­a­bil­ity of
ben­e­fit in this sub­group. This was a broad group of immunosup-
Correspondence
pressed patients based on the Acute Physiology and Chronic
Lise J. Estcourt, NHS Blood and Transplant, Level 2, John Radcliffe
Health Evaluation (APACHE) score def­i­ni­tion of immu­no­sup­pres­
Hospital, Oxford, OX3 9BQ , UK; e-mail: lise​­.estcourt@nhsbt​­.nhs​
sion.39 However, as it was a small sub­group, addi­tional research
­.uk.
is required to con­firm whether or not this is a true find­ing. The
RECOVERY trial did include patients receiv­ing non­in­va­sive and
inva­sive ven­ti­la­tion, and no evi­dence of a dif­fer­ence was seen References
1. The Nobel Prize. Emil von Behring Nobel Lecture, Serum Therapy in Ther-
for those patients receiv­ing inva­sive (RR, 0.71; 95% CI, 0.35-1.47; apeutics and Medical Science. Accessed 1 June 2021. https:­/­/www­.nobel
70 par­tic­i­pants) or non­in­va­sive ven­ti­la­tion (RR, 0.86; 95% CI, prize­.org­/prizes­/medicine­/1901­/behring­/lecture­/
0.68-1.08; 673 par­tic­i­pants), but the CIs are wide. 2. Taylor PC, Adams AC, Hufford MM, De La Torre I, Winthrop K, Gottlieb RL.
Neutralizing mono­clo­nal antibodies for treat­ment of COVID-19. Nat Rev
Immunol. 2021;21(6):382-393.
3. Piechotta V, Iannizzi C, Chai KL, et  al. Convalescent plasma or hyper­im­
CLINICAL CASE (Con­t in­u ed) mune immu­no­glob­u­lin for peo­ple with COVID-19: a liv­ing sys­tem­atic
review. Cochrane Database Syst Rev. 2021(5):CD013600.
Unfortunately, the 74-year-old man con­tin­ued to dete­ri­o­rate and 4. Casadevall A, Pirofski L-A. The con­va­les­cent sera option for containing
sub­se­quently required inva­sive ven­ti­la­tion. Fourteen days after COVID-19. J Clin Invest. 2020;130(4):1545-1548.
the chris­ten­ing, he died due to COVID-19. In total, 5 of the guests 5. Lachiewicz AM, Srinivas ML. Varicella-zoster virus post-expo­sure man­age­
ment and pro­phy­laxis: a review. Prev Med Rep. 2019;16:101016.
at the chris­ten­ing were admit­ted to hos­pi­tal, includ­ing all 4 of the 6. Food and Drug Administration. Revocation let­ter bamlanivimab 04162021.
grand­par­ents of the baby and the uncle (the index case), who had 2021. Accessed 1 June 2021. https://www.fda.gov/news-events/press-an
dia­be­tes and hyper­ten­sion. Both grand­fa­thers died of COVID-19. nouncements/coronavirus-covid-19-update-fda-revokes-emergen
cy-use-authorization-monoclonal-antibody-bamlanivimab
7. Starr TN, Greaney AJ, Addetia A, et  al. Prospective map­ ping of viral
muta­ tions that escape antibodies used to treat COVID-19. Science.
Key points (see Visual abstract) 2021;371(6531):850-854.
Up to now (July 2021), most of the evi­dence is based on tri­als 8. Libster R, Pérez Marc G, Wappner D, et al; Fundación INFANT–COVID-19
Group. Early high-titer plasma ther­apy to pre­vent severe Covid-19 in older
performed prior to the intro­duc­tion of vac­ci­na­tion. Vaccination adults. N Engl J Med. 2021;384(7):610-618.
is likely to be the most cost-effec­tive strat­egy for pre­vent­ing 9. Haas EJ, Angulo FJ, McLaughlin JM, et al. Impact and effec­tive­ness of mRNA
infec­tion in the gen­eral pop­u­la­tion, includ­ing healthy indi­vid­u­ BNT162b2 vac­ cine against SARS-CoV-2 infec­ tions and COVID-19 cases,
als with high-risk expo­sure (eg, health care work­ers). There are hospitalisations, and deaths fol­low­ing a nation­wide vac­ci­na­tion cam­paign
in Israel: an obser­va­tional study using national sur­veil­lance data. Lancet.
insuf­fi­cient data on the out­comes of pro­phy­lac­tic pas­sive anti­
2021;397(10287):1819-1829.
body ther­apy in immunosuppressed indi­vid­u­als. 10. Galvani A, Moghadas SM, Schneider EC. Deaths and hos­ pi­tal­i­
za­
tions
Passive immune ther­apy (mono­clo­nal ther­apy and CP) may averted by rapid U.S. vac­ci­na­tion roll­out. The Commonwealth Fund, Issue
be ben­ e­fi­
cial for high-risk patients who have mild COVID-19 Briefs. Accessed 15 July 2021. https:­/­/www­.commonwealthfund­.org­/publi
symp­toms, but more data are required. Some countries, includ­ cations­/issue­-briefs­/2021­/jul­/deaths­-and­-hospitalizations­-averted­-rapid­
us­-vaccination­-rollout
ing the United States, are using pas­sive mono­clo­nal ther­apy for 11. Maneikis K, Šablauskas K, Ringelevičiūtė U, et  al. Immunogenicity of the
this indi­ca­tion under EUA. BNT162b2 COVID-19 mRNA vac­cine and early clin­i­cal out­comes in patients
High-dose pas­sive immune ther­apy (casirivimab/imdevimab) with haematological malig­ nan­cies in Lithuania: a national pro­ spec­ tive
reduces all­-cause mor­tal­ity for hos­pi­tal­ized patients who have cohort study. Lancet Haematol. 2021;8(8):e583-e592.
12. Cohen MS, Nirula A, Mulligan MJ, et al; BLAZE-2 Investigators. Effect of bam-
not yet devel­oped a detect­able anti­body response (SARS-CoV-2
lanivimab vs pla­cebo on inci­dence of COVID-19 among res­i­dents and staff
IgG anti­body). This mono­clo­nal cock­tail has just been approved of skilled nurs­ing and assisted liv­ing facil­it­ies: a ran­dom­ized clin­i­cal trial.
by the Medicines and Healthcare prod­ucts Regulatory Agency JAMA. 2021;326(1):46-55.
based on this evi­dence, but indi­ca­tions for its use are cur­rently 13. O’Brien MP, Forleo-Neto E, Musser BJ, et al. Subcutaneous REGEN-COV anti­
not avail­­able. body com­bi­na­tion for Covid-19 pre­ven­tion. N Engl J Med. 2021;385:1184-
1195.
CP may be ben­e­fi­cial for immunosuppressed indi­vid­u­als who 14. O’Brien MP, Forleo-Neto E, Sarkar N, et al. Subcutaneous REGEN-COV anti­
are severely or crit­i­cally unwell, but more data are required. body com­bi­na­tion in early SARS-CoV-2 infec­tion. medRxiv. 2021.

640  |  Hematology 2021  |  ASH Education Program


15. Weinreich DM, Sivapalasingam S, Norton T, et  al. REGEN-COV anti­body ized con­trolled, open-label trial [published online 9 Sep­tem­ber 2021].
com­bi­na­tion and out­comes in out­pa­tients with Covid-19 [published online Nat Med.
29 Sep­tem­ber 2021]. N Engl J Med. 31. O’Donnell MR, Grinsztejn B, Cummings MJ, et  al. A ran­dom­ized dou­ble-
16. Gupta A, Gonzalez-Rojas Y, Juarez E, et al. Early Covid-19 treat­ment with blind con­trolled trial of con­va­les­cent plasma in adults with severe COVID-
SARS-CoV-2 neu­tral­iz­ing anti­body sotrovimab. medRxiv. 2021. 19. J Clin Invest. 2021;131(13):e150646.
17. Gottlieb RL, Nirula A, Chen P, et al. Effect of bamlanivimab as monotherapy 32. Körper S, Weiss M, Zickler D, et  al. High dose con­va­les­cent plasma in
or in com­bi­na­tion with etesevimab on viral load in patients with mild to COVID-19: results from the ran­dom­ized trial CAPSID. medRxiv. 2021.
mod­er­ate COVID-19: a ran­dom­ized clin­i­cal trial. JAMA. 2021;325(7):632-644. 33. Gharbharan A, Jordans CCE, GeurtsvanKessel C, et  al. Effects of potent
18. Dougan M, Nirula A, Azizad M, et al. Bamlanivimab plus etesevimab in mild neu­tral­iz­ing antibodies from con­va­les­cent plasma in patients hos­pi­tal­ized
or mod­er­ate Covid-19 [published online 14 July 2021]. N Engl J Med. for severe SARS-CoV-2 infec­tion. Nat Commun. 2021;12(1):3189.
19. Eom JS, Ison M, Streinu-Cercel A, et al. Efficacy and safety of CT-P59 plus 34. Ray Y, Paul SR, Bandopadhyay P, et al. Clinical and immu­no­log­i­cal ben­e­fits
stan­dard of care: a phase 2/3 ran­dom­ized, dou­ble-blind, pla­cebo-con­ of con­va­les­cent plasma ther­apy in severe COVID-19: insights from a sin­gle
trolled trial in out­pa­tients with mild-to-mod­er­ate SARS-CoV-2 infec­tion cen­ter open label randomised con­trol trial. medRxiv. 2020.
[published online 16 March 2021]. Res Square. 35. Bennett-Guerrero E, Romeiser JL, Tal­bot LR, et al. Severe acute respi­ra­tory
20. Weinreich DM, Sivapalasingam S, Norton T, et al; Trial Investigators. REGN- syn­drome coronavirus 2 con­va­les­cent plasma ver­sus stan­dard plasma in
COV2, a neu­tral­iz­ing anti­body cock­tail, in out­pa­tients with Covid-19. N coronavirus dis­ease 2019 infected hos­pi­tal­ized patients in New York: a dou­

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/628/1852150/628estcourt.pdf by guest on 13 December 2021


Engl J Med. 2021;384(3):238-251. ble-blind ran­dom­ized trial. Crit Care Med. 2021;49(7):1015-1025.
21. Horby PW, Mafham M, Peto L, et al. Casirivimab and imdevimab in patients 36. Pouladzadeh M, Safdarian M, Eshghi P, et al. A ran­dom­ized clin­i­cal trial eval­
admit­ted to hos­pi­tal with COVID-19 (RECOVERY): a randomised, con­trolled, u­at­ing the immu­no­mod­u­la­tory effect of con­va­les­cent plasma on COVID-19-
open-label, plat­form trial. medRxiv. 2021. related cyto­kine storm [published online 10 April 2021]. Intern Emerg Med.
22. ACTIV-3/TICO LY-CoV555 Study Group. A neu­tral­iz­ing mono­clo­nal anti­body 37. Hamdy Salman O, Ail Mohamed HS. Efficacy and safety of trans­ fus­ ing
for hos­pi­tal­ized patients with Covid-19. N Engl J Med. 2021;384(10):905-914. plasma from COVID-19 sur­vi­vors to COVID-19 vic­tims with severe ill­ness:
23. Hirsch C, Valk SJ, Piechotta V, et al. SARS-CoV-2-neutralising mono­clo­nal a dou­ble-blinded con­trolled pre­lim­i­nary study. Egypt J Anaesth. 2020;​
antibodies to pre­vent COVID-19 [published online May 20, 2021]. Cochrane 36(1):264-272.
Database Syst Rev. 38. Bajpai M, Kumar S, Maheshwari A, et  al. Efficacy of con­va­les­cent plasma
24. Valk SJ, Piechotta V, Kimber C, et al. Convalescent plasma and hyper­im­ ther­apy com­pared to fresh fro­zen plasma in severely ill COVID-19 patients:
mune immu­no­glob­u­lin to pre­vent infec­tion with SARS-CoV-2. Cochrane a pilot ran­dom­ized con­trolled trial. medRxiv. 2020.
Database Syst Rev. 2021;1:CD013802. 39. The REMAP-CAP Investigators. Convalescent plasma in crit­i­cally ill patients
25. Agarwal A, Mukherjee A, Kumar G, Chatterjee P, Bhatnagar T, Malhotra P. with Covid-19. medRxiv. 2021.
Convalescent plasma in the man­age­ment of mod­er­ate Covid-19 in adults in 40. Gonzalez JLB, González Gámez M, Mendoza Enciso EA, et al. Efficacy and
India: open label phase II multicentre randomised con­trolled trial (PLACID safety of con­va­les­cent plasma and intra­ve­nous immu­no­glob­u­lin in crit­i­cally
Trial). BMJ. 2020:m3939. ill COVID-19 patients: a con­trolled clin­i­cal trial. medRxiv. 2021.
26. Simonovich VA, Burgos Pratx LD, Scibona P, et al; PlasmAr Study Group. A 41. Li L, Zhang W, Hu Y, et al. Effect of con­va­les­cent plasma ther­apy on time to
ran­dom­ized trial of con­va­les­cent plasma in Covid-19 severe pneu­mo­nia. N clin­i­cal improve­ment in patients with severe and life-threat­en­ing COVID-
Engl J Med. 2021;384(7):619-629. 19: a ran­dom­ized clin­i­cal trial. JAMA. 2020;324(5):460-470.
27. Avendaño-Solá C, Ramos-Martínez A, Muñez-Rubio E, et  al. Convalescent 42. Kreuzberger N, Hirsch C, Chai KL, et al. SARS-CoV-2-neutralising mono­clo­
plasma for COVID-19: a mul­ti­cen­ter, ran­dom­ized clin­i­cal trial. medRxiv. 2020. nal antibodies for treat­ment of COVID-19. Cochrane Database Syst Rev.
28. AlQahtani M, Abdulrahman A, Almadani A, et  al. Randomized con­trolled 2021;9:CD013825.
trial of con­va­les­cent plasma ther­apy against stan­dard ther­apy in patients
with severe COVID-19 dis­ease. Sci Rep. 2021;11(1):9927.
29. RECOVERY Collaborative Group. Convalescent plasma in patients admit­
ted to hos­ pi­
tal with COVID-19 (RECOVERY): a randomised con­ trolled,
open-label, plat­form trial. Lancet. 2021;397(10289):2049-2059.
30. Bégin P, Callum J, Jamula E, et al. Convalescent plasma for hos­pi­tal­ized © 2021 by The Amer­i­can Society of Hematology
patients with COVID-19 and the effect of plasma antibodies: a ran­dom­ DOI 10.1182/hema­tol­ogy.2021000299

Prakash Singh Shekhawat


Passive immune ther­a­pies: tool against COVID-19  |  641
UPDATE IN GRAFT- VERSUS - HOST DISEASE

Acute GVHD: think before you treat


Laura F. Newell1 and Shernan G. Holtan2

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/642/1852146/642newell.pdf by guest on 13 December 2021


Knight Cancer Institute, Hematology and Medical Oncology, Oregon Health and Science University, Portland, OR; and 2Division of Hematology,
1

Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN

The treatment of acute graft-versus-host disease (aGVHD) has become more nuanced in recent years with the develop-
ment of improved risk classification systems and a better understanding of its complex, multisystem pathophysiology.
We review contemporary approaches to the risk stratification and initial treatment of aGVHD, including ongoing clinical
trials. We summarize the findings that led to the first US Food and Drug Administration approval for steroid-refractory
aGVHD (SR-aGVHD), ruxolitinib, as well as some of the challenges clinicians still face in treating SR-aGVHD. Finally, we
discuss the evaluation and management of steroid-dependent aGVHD, which affects approximately one-third of patients
who have long-term, waxing and waning symptoms distinct from chronic GVHD. Future clinical trials for aGVHD treat-
ment may identify steroid-sparing approaches for patients who have a high likelihood of response and approaches to
improve tissue repair and dysbiosis for those unlikely to respond to immunosuppression alone.

LEARNING OBJECTIVES
• Identify risk­stratifed approaches to the initial management of aGVHD
• Identify potential contributing factors for persistent or recurring symptoms after the initial treatment of aGVHD

Introduction rolimus and methotrexate for GVHD prophylaxis. Day 11


Recent advancements have improved overall survival and methotrexate was 50% dose reduced, and leucovorin
decreased the incidence of grades 3 and 4 acute graft­ rescue was added for severe mucositis. On day 21, he
versus­host disease (aGVHD) after allogeneic hematopoi­ developed a patchy maculopapular skin rash involving
etic cell transplantation (HCT), but substantial challenges a body surface area (BSA) of <20% and was started on
remain.1-3 High­dose corticosteroids are standard therapy 0.1% triamcinolone topical cream. On day 25, he devel­
for aGVHD, but this approach does not consider individ­ oped fever, nausea and vomiting, and worsening rash
ual factors and may lead to over­ or undertreatment. In involving his face, anterior/posterior torso, and lower
this review we describe current risk­adapted approaches extremities to his knees (60% BSA). He had stage 3 skin,
to aGVHD management, highlighting opportunities for stage 1 upper­gastrointestinal (GI), and overall grade 2
improvement. Minnesota standard­risk aGVHD.6,7 Treatment included
Sixty years elapsed between the description of sec­ 2 mg/kg/d prednisone and continued tacrolimus. A com­
ondary disease as an immunologic complication of murine plete response (CR) by day 28 of therapy was sustained
bone marrow transplantation (1959) and the frst US Food at 8 weeks. Although steroids were discontinued by day
and Drug Administration (FDA) approval for SR­aGVHD 52 of therapy, he had hyperglycemia requiring insulin and
treatment, ruxolitinib (2019).4,5 With a deeper understand­ became cushingoid during treatment.
ing of the pathophysiology of aGVHD, including a need for
tissue repair and reversal of dysbiosis, clinical advance­
ments should be tested at a much faster pace. Risk-based assessment and initial treatment
Clinical severity-based risk assessment
The grading criteria for aGVHD are well established for the
skin, liver, and intestinal tract, with higher grades associated
CLINICAL CASE with worse transplant outcomes.7 However, discrepancies
A 47­year­old man with acute myeloid leukemia under­ have remained in clinical staging across centers, which can
went matched unrelated donor HCT after myeloablative influence multicenter trial outcome reporting.8 In order to
busulfan and cyclophosphamide conditioning with tac­ standardize staging, consensus guidelines were developed

642 | Hematology 2021 | ASH Education Program


at the University of Michigan and sub­ se­ quently tested in the able to cli­ni­cians, novel mark­ers may yet be devel­oped, and the
mul­ ti­
cen­
ter Mount Sinai Acute GVHD International Consortium role of bio­mark­ers remains an area of active research inves­ti­ga­
(MAGIC).9,10 These guide­lines pro­vide more pre­cise def­i­ni­tions for tion. The assess­ment of bio­marker-based risk strat­i­fi­ca­tion and
aGVHD organ stag­ing as well as con­fi­dence lev­els for diag­no­sis mon­i­tor­ing should con­tinue in the con­text of pro­spec­tive clin­i­cal
(con­firmed, prob­a­ble, pos­si­ble, neg­a­tive) based on supporting tri­als.
evi­dence (eg, his­to­logic con­fir­ma­tion, clin­i­cal action). Experts from
the Euro­pean Society for Blood and Marrow Transplantation, the Risk-adapted ini­tial treat­ment
National Institutes of Health, and the Center for International Blood For patients with grade 2a man­i­fes­ta­tions of aGHVD (defined as
and Marrow Transplant Research Task Force have recommended upper-GI symp­toms, stool out­put <1 L/d, rash <50% BSA, with­
the MAGIC cri­te­ria for the diag­no­sis and scor­ing of aGVHD.11 out hepatic involve­ment), treat­ment with lower-dose ste­roids
Simpler clas­si­fi­ca­tion schemes may fur­ther delin­eate aGVHD (0.5 mg/kg/d vs 1.0 mg/kg/d) has been shown to be effec­tive
risk in order to test per­son­al­ized treat­ment strat­e­gies in patients with­out increas­ing the risk of sec­ond­ary immu­no­sup­pres­sion.18
with dif­fer­ing organ stag­ing who are likely to have sim­i­lar out­ However, for patients with grade 2b or higher man­i­fes­ta­tions

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/642/1852146/642newell.pdf by guest on 13 December 2021


comes. The Minnesota aGVHD risk score incor­ po­rates sites (defined as stool vol­ ume ≥1  L/d, rash ≥50% BSA, or hepatic
of organ involve­ment and stage of dis­ease to prog­nos­ti­cate a involve­ment), treat­ment with lower-dose ste­roids (1.0 mg/kg/d
response to ini­tial aGVHD ther­apy and the risk of transplant- vs 2.0 mg/kg/d) was asso­ci­ated with an increased like­li­hood of
related mor­tal­ity.6,12 A web-based cal­cu­la­tor is avail­­able online to requir­ing sec­ond­ary immu­no­sup­pres­sive ther­apy.
deter­mine the Minnesota aGVHD score (https:​­/​­/redcap​­.ahc​­.umn​ Recently, the BMT CTN reported (trial 1501) a ran­dom­ized
­.edu​­/surveys​­/​­?s=bNmFhseJIf). Together, MAGIC stag­ing and the phase 2 study test­ing the ste­roid-free ini­tial treat­ment of Minne­
Minnesota score are used in the risk assess­ment of patients with sota stan­dard risk aGVHD (N = 127) with sirolimus vs pred­ni­sone.19
newly diag­nosed aGVHD in many cur­rent clin­i­cal tri­als. In this study, the day-28 response rate was sim­i­lar, at 65% for
sirolimus (90% CI, 54%-76%) vs 73% (90% CI, 64%-82%) for pred­
Biomarker-based risk assess­ment ni­sone. However, patients on the sirolimus arm had fewer side
In addi­tion to opti­miz­ing clin­i­cal GVHD scor­ing sys­tems, efforts effects from ther­apy and an improved patient-reported qual­ity
to improve the deter­mi­na­tion of sever­ity and like­li­hood of re­ of life, suggesting that ste­roid-free treat­ment of stan­dard-risk
sponse have focused on iden­ti­fy­ing periph­eral blood bio­mark­ aGVHD is fea­si­ble. A note of cau­tion is that <10% of the par­tic­
ers. Criteria and phases of bio­marker devel­op­ment, as well as i­pants enrolled in BMT CTN 1501 had lower-GI GVHD, and thus
the dif­fer­ent types of bio­mark­ers for acute and chronic GVHD, we still do not know with con­fi­dence how sirolimus ther­apy
have recently been reviewed by Adom et  al.13 Because some would per­form as the upfront ther­apy in this patient pop­u­la­tion.
avail­­able bio­mark­ers used to diag­nose aGVHD are con­founded Additionally, it is unclear how widely these results have been
by organ dam­age or infec­tions, they are of lim­ited use in aGVHD adopted across clin­i­cal cen­ters.
risk assess­ment. Patients with a favor­able like­li­hood of response to aGVHD
The MAGIC group iden­ti­fied 2 serum bio­mark­ers of GVHD treat­ment may be can­di­dates for ste­roid-spar­ing approaches; in
that in com­bi­na­tion pre­dict severe GVHD and nonrelapse mor­ con­trast, those patients at highest risk of fatal aGVHD may be
tal­ity (NRM): a sup­pres­sor of tumor­i­gen­e­sis, a mem­ber of the can­di­dates for novel agents. GVHD treat­ment tri­als within the
inter­leu­kin 1 recep­tor fam­ily and the sol­u­ble recep­tor for inter­ last 5 years incor­po­rat­ing risk-assess­ment algo­rithms are sum­
leu­ kin 33, and regenerating islet-derived 3-alpha, pro­ duced ma­rized in Table 1.
by dam­aged Paneth cells with anti­mi­cro­bial and epi­the­lial-
pro­tec­tive prop­er­ties.14 In a mul­ti­cen­ter val­i­da­tion study, the
MAGIC algo­rithm prob­a­bil­ity (MAP) and clin­i­cal responses were
deter­mined at the time of treat­ment ini­ti­a­tion and again after 4
weeks of ther­apy. Using the ini­tial MAP, patients could be cat­ CLINICAL CASE
e­go­rized by Ann Arbor (AA) score,1-3 each with a dis­tinct risk of A 61-year-old woman with a his­tory of acute mye­loid leu­ke­mia
NRM; after 4 weeks, changes in the MAP fur­ther refined the esti­ in sec­ond com­plete remis­sion under­went fludarabine and mel­
ma­tion of NRM risk in all­AA groups. Importantly, the MAP was pha­lan con­di­tioned matched unre­lated (male) donor peripheral
found to more accu­rately pre­dict 6-month NRM than a change blood stem cell transplant. She received GVHD pro­phy­laxis with
in clin­i­cal symp­toms. tacrolimus and meth­o­trex­ate. Her trans­plant course was com­pli­
Additional bio­mark­ers of inflam­ma­tion and tis­sue dam­age cated by neutropenic fever, pro­tein/cal­o­rie mal­nu­tri­tion requir­
have been inves­ ti­
gated. Amphiregulin (AREG), an epi­ der­mal ing total par­en­teral nutri­tion (TPN), and grade 1 mucositis. On
growth fac­tor (EGF)-like mol­e­cule involved in type 2 immune day 11 she devel­oped a maculopapular skin rash involv­ing 36%
responses and tis­sue repair, is another novel bio­marker asso­ci­ BSA, and her anti­bi­ot­ics were changed due to con­cern about
ated with GVHD risk. Elevated lev­els of AREG have been iden­ti­ a pos­si­ble drug reac­tion. On day 14 she devel­oped increas­ing
fied in late aGVHD.15 Additionally, AREG lev­els have been shown diar­rhea with a neg­a­tive infec­tious workup. On day 23, she had
to fur­ther refine the Minnesota aGVHD risk score, using sam­ples a rash with 30% BSA involve­ ment, nau­
sea, eme­sis, and diar­
col­lected at aGVHD diag­no­sis as part of the Blood and Marrow rhea (<1500 mL/d). She was diag­nosed with stage 2 skin, stage 1
Transplant Clinical Trials Network (BMT CTN) 0302 and 0802.16 upper-GI, stage 2 lower-GI, and over­all grade 3 Minnesota high-
An AREG thresh­old of ≥33 pg/mL iden­ti­fied patients with a lower risk aGVHD and started on 2 mg/kg/d meth­yl­pred­nis­o­lone. Her
like­li­hood of day-28 response and a higher 2-year NRM. Patho­ skin rash improved; how­ever, she con­tin­ued to have nau­sea and
logically ele­vated AREG lev­els decline over time in patients who watery diar­rhea (1000-1500 mL/d) progressing to ileus. Her hos­pi­
respond to aGVHD ther­apy.17 Although bio­marker test­ing is avail­­ tal course was also com­pli­cated by cytomegalovirus reactivation
Prakash Singh Shekhawat
Acute GVHD  | 643
Table 1. First-line risk-adapted treat­ment tri­als for aGVHD

Transplant eli­gi­
Study num­ber GVHD risk cat­e­gory Study type/inter­ven­tion Treatment tar­get Study sta­tus Primary out­come mea­sures
bil­ity cri­te­ria
Standard-risk aGVHD
NCT02806947 Standard-risk (Minnesota Randomized, mul­ti­cen­ter, phase 2 mTOR inhib­i­tor Any donor type, Completed Day-28 CR/PR rates sim­i­lar
BMT CTN risk), pre­vi­ously untreated Sirolimus vs pred­ni­sone 2 mg/kg con­di­tion­ing for sirolimus vs pred­ni­sone
1501(19) aGVHD requir­ing sys­temic Child, adult, (64.8% vs 73%)
ther­apy, older-adult Sirolimus asso­ci­ated with
AAscore 1-2 bio­marker ages reduced ste­roid expo­
assess­ment (performed sure and hyper­gly­ce­
at ran­dom­i­za­tion) mia, improved QOL, and
increased risk for TMA
NCT03846479 Low-risk aGVHD, Minnesota Single arm, monotherapy JAK1 inhib­i­tor Any donor type, Recruiting Day-28 Minnesota stan­dard-risk
stan­dard risk, AA score 1 Itacitinib con­di­tion­ing clin­i­cal cri­te­ria (CR, PR, TRM)
Ages 12-75 Day-28 AA score 1
years
NCT04144036 Standard-risk aGVHD, Single-cen­ter nonrandomized Humanized mono­clo­nal Any donor type, Recruiting Day-28 SAE, MTD of
Minnesota risk, AA score phase 1 anti­body bind­ing to con­di­tion­ing neihulizumab, plasma Cmax
1-2 Neihulizumab dose esca­la­tion CD162 (PSGL-1) Ages ≥18 years

644  |  Hematology 2021  |  ASH Education Program


High-risk aGVHD
NCT02133924 High-risk aGVHD, Multicenter, phase 2 Humanized recom­bi­nant Any donor type, Recruiting Day-28 CR
AA score 3 Natalizumab + pred­ni­sone anti­body bind­ing to con­di­tion­ing
alpha-4 integrin Ages ≥18 years
NCT0252502917 Arm 1: high-risk aGVHD Nonrandomized phase ½ uhCG, which may pro­ Any donor type, Active, not Phase 1: MTD of uhCG/EGF in
(Minnesota risk, or Standard of care IST + Pregnyl mote immune tol­er­ con­di­tion­ing recruiting com­bi­na­tion with stan­dard IST
AA score 3, or AREG ance, pro­vi­des source Ages 0-76 years Phase 2: day-28 CR, PR, MR,
≥33 pg/mL) of EGF for epi­the­lial and no response
Arm 2a: ste­roid-depen­dent repair (Pregnyl®)
aGVHD
Arm 2b: SR-aGVHD
NCT03158896 De novo high-risk aGVHD Phase 1, dose esca­la­tion Immunosuppressive Ages 18-75 Active, not Day-45 treat­ment-related SAE
SR-aGVHD Umbilical cord-derived, ex vivo cul­ prop­er­ties: sup­press years recruiting
tured, and expanded Wharton’s pro­lif­er­a­tion of acti­vated
Jelly MSC T cells, increase pro­duc­
tion of reg­u­la­tory T cells,
shift immune response
toward tol­er­ance
NCT04061876 High-risk grade 2-4 aGVHD Randomized, phase 2 JAK1/2 inhib­i­tor Any donor type Recruiting Day-28 ORR
(based on ST2, REG3α, or Ruxolitinib + cor­ti­co­ste­roids vs Ages 14-65
other exper­i­ment object) cor­ti­co­ste­roids alone years
NCT04167514 High-risk aGVHD requir­ing Randomized, dou­ble-blind, pla­cebo- Serine pro­te­ase inhib­i­tor Any donor type, Recruiting Day-28 CR, PR
cor­ti­co­ste­roids (first-line con­trolled, mul­ti­cen­ter phase 3 con­di­tion­ing
ther­apy) AAT + cor­ti­co­ste­roids vs cor­ti­co­ste­ Ages ≥18 years
roids alone
NCT04291261 High-risk aGVHD, AA score Single-arm phase 2 Induces apo­pto­sis of Any donor type, Active, not Day-28 CR
2-3 ECP with methoxsalen +2 mg/kg mono­nu­clear cells con­di­tion­ing recruiting
cor­ti­co­ste­roids Ages ≥18 years
NCT04397367 Grade 2-4 or high-risk aGVHD Nonrandomized JAK1/2 inhib­i­tor Any donor type Recruiting Day-28 CR
(based on ST2, REG3α, or Ruxolitinib + cor­ti­co­ste­roids Ages 14-65
other exper­i­ment object) years
AAT, alpha-1 antitrypsin; ECP, extra­cor­po­real photopheresis; IST, immu­no­sup­pres­sive ther­apy; JAK1, Janus kinase 1; MR, mixed response; MSC, mes­en­chy­mal stem cell; MTD, max­i­mum tol­er­ated dose;
mTOR, mech­a­nis­tic tar­get of rapamycin; PR, par­tial response; PSGL-1, P-selectin gly­co­pro­tein 1; QOL, qual­ity of life; REG3α, regenerating islet-derived 3-alpha; SAE, seri­ous adverse event; ST2, sup­
pres­sor of tumor­i­gen­e­sis; TMA, throm­botic microangiopathy; TRM, trans­plant-related mor­tal­ity.
Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/642/1852146/642newell.pdf by guest on 13 December 2021
treated with ganciclovir, a vancomycin-resis­tant Enterococcus Other exper­ i­
men­tal approaches to SR-aGVHD with vary­
faecium uri­ nary tract infec­tion treated with linezolid, and BK ing mech­ a­
nisms of action are being tested in clin­ i­
cal tri­
als,
cys­ti­tis pal­li­ated with pyridium. The plan was to start ruxolitinib recently sum­ma­rized by Abedin and Hamadani and Martin.20,25
after insur­ance approval and with the con­trol of cytomegalovirus With admi­ra­ble humil­ity in his “How I Treat” pub­li­ca­tion, Dr
reactivation; how­ever, she was never a ­ ble to start ther­apy. On Martin admit­ted, “Truth be told, I do not know how to treat
day 32 post-HCT, she devel­oped a pro­duc­tive cough, hypo­ten­ SR-aGVHD.” This state­ment under­scores the tre­men­dous dif­fi­
sion, and tachy­car­dia and required oxy­gen and was trans­ferred culty in over­com­ing this largely fatal dis­ease despite decades
to the inten­sive care unit for the man­age­ment of sep­tic shock. of intense study. Truth be told, we do not fully under­stand the
Labs showed pan­cy­to­pe­nia and new lung infil­trates. She was path­o­phys­i­­ol­ogy of SR-aGVHD. Most novel ther­a­peu­tics have
given high-flow oxy­gen, nor­epi­neph­rine, and broad-spec­trum been potent immu­no­sup­pres­sants, yet that may not be the cor­
anti­bi­ot­ics. There was some ini­tial improve­ment, but on day 36 rect or only approach. In a pre­clin­i­cal model, there were no dif­
she devel­oped atrial fibril­la­tion with rapid ven­tric­u­lar response, fer­ences in T-cell expan­sion, acti­va­tion, or enhanced cyto­kine
was started on an amiodarone drip, and devel­oped respi­ra­tory pro­duc­tion in donor T cells in mice with respon­sive vs refrac­

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/642/1852146/642newell.pdf by guest on 13 December 2021


dis­tress. Her respi­ra­tory sta­tus wors­ened, and she died on day 41. tory aGVHD.26 Recently, murine SR-aGVHD was described as
driven by inter­leu­kin 22-depen­dent dysbiosis, with a pro­tec­tive
role of CXCR3hi mono­nu­clear phago­cytes in reduc­ing bac­te­rial
trans­lo­ca­tion.27 Gene expres­sion stud­ies from human colo­rec­tal
SR-aGVHD biop­sies showed that human SR-aGVHD is char­ac­ter­ized by tis­
Definition of SR-aGVHD sue response to dam­age, cel­lu­lar stress, and mac­ro­phage accu­
The EBMT-NIH-CIBMTR Task Force suggested the fol­low­ing def­i­ mu­la­tion, not T-cell pro­lif­er­a­tion.28 Collectively, these recent
ni­tions for SR-aGVHD or ste­roid-resis­tant aGVHD:1 pro­gres­sion of stud­ies sug­gest that future ther­a­peu­tic efforts in SR-aGVHD,
aGVHD within 3 to 5 days of treat­ment with ≥2 mg/kg/d pred­ni­sone in addi­tion to targeting the ini­tial T-cell-medi­ated dam­age and
equiv­a­lent, or2 fail­ure to improve with 5 to 7 days of treat­ment, or inflam­ma­tion, might also con­sider stud­ies of agents designed
incom­plete response after more than 28 days of immu­no­sup­pres­ to enhance tis­sue repair and to cor­rect dysbiosis while try­ing to
sive ther­apy includ­ing ste­roids.3,11 SR-aGVHD has also been rec­og­ avoid broad immu­no­sup­pres­sion and its inher­ent risks of infec­
nized as (a) wors­en­ing GVHD man­i­fes­ta­tions in patients receiv­ing tion17,29-34. Recently described tar­gets such as CD83 sug­gest this
≥1 mg/kg/d pred­ni­sone equiv­a­lent ≥2 days prior to ste­roid dose may be fea­si­ble.35
taper­ing; (b) per­sis­tent grade 2 to 4 GVHD with­out improve­ment
≥7 days dur­ing con­tin­ued treat­ment with >0.4 mg/kg/d pred­ni­
sone equiv­a­lent, or (c) ini­tial improve­ment followed by exac­er­
ba­tion ≥3 days dur­ing ste­roid taper at any dose of >0.4 mg/kg/d
pred­ni­sone equiv­a­lent.20 In prac­tice, it can be very chal­leng­ing to CLINICAL CASE
know how long to wait before adding a sec­ond-line agent. A 23-year-old man under­went a myeloablative, matched unre­
lated donor peripheral blood stem cell transplant for high-risk
Treatment options for SR-aGVHD T-cell acute lym­pho­blas­tic leu­ke­mia and was diag­nosed with
In May 2019, the FDA approved the first treat­ment for SR-aGVHD: stage 2 skin, stage 3 GI, over­all Minnesota high-risk aGVHD.
ruxolitinib, an inhib­i­tor of Janus kinase 1 and 2, for pedi­at­ric and He was treated with high-dose ste­ roids, uri­
nary-derived
adult patients 12 years of age or older.21 FDA approval was based human chorionic gonad­o­tro­pin (uhCG), and EGF in clin­i­cal trial
upon the REACH-1 trial (NCT02953678, study INCB 18424-271), a NCT02525029. He achieved a CR at 4 weeks after the ini­ti­a­tion
phase 2, mul­ti­cen­ter, open-label, sin­gle-arm study of 71 patients, of ther­apy, which was sustained at 8 weeks. Over the ensu­ing
49 of whom had grades 2 to 4 SR-aGVHD (includ­ ing patients 2 years, he had 6 hos­pi­tal­i­za­tions with a recur­rence of nau­sea,
who failed ste­roid treat­ment with or with­out receiv­ing addi­tional vomiting, and diar­rhea. With each epi­sode he was tested for
GVHD ther­apy).21 A starting dose of ruxolitinib, 5 mg twice daily, infec­tions (1 epi­sode was due to norovirus) and other com­pli­ca­
was admin­is­tered with meth­yl­pred­nis­o­lone.5 At day 28, the over­ tions, yet he would tran­siently improve with a mod­est increase
all response rate (ORR) was 55% (83% for grade 2, 41% for grade in pred­ni­sone dose. At 3 years posttransplant, he was read­
3, and 43% for grade 4 aGVHD), includ­ing 27% with a CR, with a mitted with a body mass index of 16 and recal­ci­trant anorexia
median time to first response of 7 days (range, 6-49). Ruxolitinib and diar­rhea. Repeat intes­ti­nal biop­sies showed vil­lous blunt­
was fur­ther inves­ti­gated in a phase 3, mul­ti­cen­ter, open-label ran­ ing, but the patient and donor absence of HLA-DQ2/8 ruled
dom­ized trial com­par­ing the effi­cacy of ruxolitinib vs inves­ti­ga­tor’s out celiac dis­ease. Fecal elas­tase was mildly low at 168 µg/g of
choice of ther­apy in patients 12 years of age and older with SR- stool, sug­ges­tive of mild/mod­er­ate pan­cre­atic exo­crine insuf­
aGVHD (REACH-2, NCT02913261).22 Day-28 ORR was higher in the fi­ciency. Fecal calprotectin was mildly ele­vated at 72.8 mg/kg,
ruxolitinib group vs the con­trol group (62% vs 39%, P < .001), with suggesting the pres­ ence of inflam­ ma­tion/neu­ tro­
phils in the
34% and 19% CR, respec­tively; dura­ble day-56 ORR was also higher intes­ti­
nal lumen (nor­ mal, 0-49.9  mg/kg). Plasma AREG was
in the ruxolitinib group vs the con­trol group (40% vs 22%, P < .001). mildly ele­vated at 29.1 pg/mL, suggesting tis­sue dam­age and
Ruxolitinib was asso­ci­ated with a higher inci­dence of throm­bo­cy­ mild sys­temic inflam­ma­tion (nor­mal, <5 pg/mL; active severe
to­pe­nia and a mod­est increase in ane­mia and cyto­meg­a­lo­vi­rus intes­ti­nal aGVHD is typ­i­cally >33  pg/ml). Plasma cit­rul­line was
infec­tion. In the REACH-1 study, the median time to death or new low at 0.9 µmol/dL (nor­mal, 1.3-6.0 µmol/dL; val­ues <1 µmol/dL
aGVHD ther­apy was 5.7 months; in the REACH-2 study, median are pre­dic­tive of total vil­lous atro­phy).36 D-xylose absorp­tion
fail­ure-free sur­vival was 5.0 months. Previous series have shown
21,22
was low, with a serum level of 7 pg/mL at 2 hours (nor­mal, 32-
that most patients die from SR-aGVHD within 6 months of diag­no­ 58  pg/mL), suggesting mal­ ab­sorp­tion. Budesonide and TPN
sis, and only 25% to 30% of patients sur­ Prakash Singh
vive beyond Shekhawat
2 years. 23,24
were resumed. After a week­long hos­pi­tal­i­za­tion, he was dis­

Acute GVHD  | 645


Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/642/1852146/642newell.pdf by guest on 13 December 2021
Figure 1. Considerations for therapy of GVHD in the first line and beyond.

charged from the hos­pi­tal tol­er­at­ing some oral intake but likely Conflict-of-inter­est dis­clo­sure
to be depen­dent on TPN long-term given the find­ings of low Shernan G. Holtan: advi­sor: Incyte, Generon.
enterocyte mass (low cit­rul­line) and mal­ab­sorp­tion (low D-xy­ Laura F. Newell: no conflicts to disclose.
lose absorp­tion).
Off-label drug use
Laura F. Newell: no off-label drug use.
Steroid-depen­dent aGVHD Shernan G. Holtan: Off-label drug use for SGH: Urinary-derived hu­
This patient high­lights a com­mon clin­i­cal sce­nario of a prolonged man chorionic gonadotropin has Orphan Drug Designation from
aGVHD course of wax­ ing and wan­ ing symp­ toms. “Steroid- the Food and Drug Administration for the treatment of acute GVHD.
depen­dent” aGVHD can be defined as (a) only achiev­ing a par­
tial (not com­plete) response to ste­roids after 8 weeks, (b) still Correspondence
requir­ing >10 mg/m2 pred­ni­sone after 8 weeks or any pred­ni­sone Shernan G. Holtan, Division of Hematology, Oncology, and Trans­
at all­after 10 weeks, or (c) a flare of aGVHD symp­toms requir­ing plantation, Department of Medicine, University of Minnesota,
at least a 25% increase in pred­ni­sone dose.37 Steroid-depen­dent 55455 Minneapolis, MN; e-mail: sgholtan@umn​­.edu.
aGVHD is expe­ri­enced by 31% of patients with aGVHD.37 While it
is not asso­ci­ated with increased mor­tal­ity, it may be asso­ci­ated References
with mor­bid­ity and a prolonged health care bur­den, as this case 1. Hahn T, McCarthy PL Jr, Hassebroek A, et  al. Significant improve­ment in
sur­vival after allo­ge­neic hema­to­poi­etic cell trans­plan­ta­tion dur­ing a period
descrip­tion high­lights. This patient had high-risk aGVHD with an
of sig­nif­i­cantly increased use, older recip­i­ent age, and use of unre­lated
ini­tial response to high-dose cor­ti­co­ste­roids and uhCG/EGF but donors. J Clin Oncol. 2013;31(19):2437-2449.
sub­se­quently flared, suggesting that one or more under­ly­ing 2. Gooley TA, Chien JW, Pergam SA, et al. Reduced mor­tal­ity after allo­ge­
causes had not been com­pletely addressed by his ini­tial ther­apy. neic hema­to­poi­etic-cell trans­plan­ta­tion. N Engl J Med. 2010;363(22):2091-
2101.
Figure 1 shows the dif­fer­en­tial diag­noses and con­sid­er­ations
3. Khoury HJ, Wang T, Hemmer MT, et  al. Improved sur­ vival after acute
in eval­u­at­ing aGVHD. An increase in immu­no­sup­pres­sion may graft-ver­sus-host dis­ease diag­no­sis in the mod­ern era. Haematologica.
not be the right treat­ment for all­patients with wors­en­ing symp­ 2017;102(5):958-966.
toms. Several ques­tions must be answered in the com­ing years 4. van Bekkum D, Vos O, Weyzen WW. The path­o­gen­e­sis of the sec­ond­ary
to improve out­comes. Can bio­mark­ers be used repeat­edly over dis­ease after for­eign bone mar­row trans­plan­ta­tion in x-irra­di­ated mice.
Blood. 2020;135(20):1739-1749.
weeks to months as a guide to taper­ing immu­no­sup­pres­sion?
5. Jagasia M, Perales MA, Schroeder MA, et al. Ruxolitinib for the treat­ment of
Which patients need dif­fer­ent modes of sup­port­ive care (eg, ste­roid-refrac­tory acute GVHD (REACH1): a mul­ti­cen­ter, open-label phase 2
reme­di­at­ion of dysbiosis vs tis­sue dam­age), and can this even trial. Blood. 2020; 135(20):1739-1749.
be dis­tin­guished bio­log­i­cally? How long should adjunct repair- 6. MacMillan ML, Robin M, Harris AC, et al. A refined risk score for acute graft-
ver­sus-host dis­ease that pre­dicts response to ini­tial ther­apy, sur­vival, and
based ther­ap ­ ies such as uhCG/EGF be con­tin­ued to achieve
trans­plant-related mor­tal­ity. Biol Blood Marrow Transplant. 2015;21(4):761-
max­i­mal muco­sal healing? What other tar­gets of aGVHD (eg, 767.
the endo­the­lium) should be treated? Additional clin­i­cal tri­als are 7. Przepiorka D, Weisdorf D, Martin P, et  al. 1994 con­sen­sus con­fer­ence on
urgently needed to address these ques­tions. acute GVHD grad­ing. Bone Marrow Transplant. 1995;15(6):825-828.

646  |  Hematology 2021  |  ASH Education Program


8. Weisdorf DJ, Hurd D, Carter S, et al. Prospective grad­ing of graft-ver­sus- 23. Xhaard A, Rocha V, Bueno B, et al. Steroid-refrac­tory acute GVHD: lack of
host dis­ease after unre­lated donor mar­row trans­plan­ta­tion: a grad­ing algo­ long-term improved sur­vival using new gen­er­a­tion anticytokine treat­ment.
rithm ver­sus blinded expert panel review. Biol Blood Marrow Transplant. Biol Blood Marrow Transplant. 2012;18(3):406-413.
2003;9(8):512-518. 24. Rashidi A, DeFor TE, Holtan SG, Blazar BR, Weisdorf DJ, MacMillan ML. Out­
9. Levine JE, Hogan WJ, Harris AC, et al. Improved accu­racy of acute graft- comes and pre­dic­tors of response in ste­roid-refrac­tory acute graft-ver­sus-
ver­sus-host dis­ease stag­ing among mul­ti­ple cen­ters. Best Pract Res Clin host dis­ease. Biol Blood Marrow Transplant. 2019;25(11):2297-2302.
Haematol. 2014;27(3-4):283-287. 25. Abedin S, Hamadani M. Experimental phar­ma­ceu­ti­cals for ste­roid-refrac­
10. Harris AC, Young R, Devine S, et  al. International, mul­ti­cen­ter stan­dard­i­ tory acute graft-ver­sus-host dis­ease. J Exp Pharmacol. 2020;12:549-557.
za­tion of acute graft-ver­sus-host dis­ease clin­i­cal data col­lec­tion: a report eCollection 2020.
from the Mount Sinai Acute GVHD International Consortium. Biol Blood 26. Toubai T, Rossi C, Tawara I, et al. Murine mod­els of ste­roid refrac­tory graft-
Marrow Transplant. 2016;22(1):4-10. ver­sus-host dis­ease. Sci Rep. 2018;8(1):12475.
11. Schoemans HM, Lee SJ, Ferrara JL, et al; EBMT (Euro­pean Society for Blood 27. Song Q , Wang X, Wu X, et al. IL-22-depen­dent dysbiosis and mono­nu­clear
and Marrow Transplantation) Transplant Complications Working Party and phago­cyte deple­tion con­trib­ute to ste­roid-resis­tant gut graft-ver­sus-host
the “EBMT−NIH (National Institutes of Health)−CIBMTR (Center for Interna­ dis­ease in mice. Nat Commun. 2021;12(1):805.
tional Blood and Marrow Transplant Research) GvHD Task Force.” EBMT- 28. Holtan SG, Shabaneh A, Betts BC, et al. Stress responses, M2 mac­ro­phages,
NIH-CIBMTR task force posi­tion state­ment on stan­dard­ized ter­mi­nol­ogy and a dis­tinct micro­bial sig­na­ture in fatal intes­ti­nal acute graft-ver­sus-host

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/642/1852146/642newell.pdf by guest on 13 December 2021


and guid­ ance for graft-ver­ sus-host dis­ ease assess­ ment. Bone Marrow dis­ease. Nature. 2015;528(7583):560-564.
Transplant. 2018;53(11):1401-1415. 29. Lindemans CA, Calafiore M, Mertelsmann AM, et  al. Interleukin-22 pro­
12. MacMillan ML, DeFor TE, Holtan SG, Rashidi A, Blazar BR, Weisdorf DJ. Val­ motes intes­ti­nal-stem-cell-medi­ated epi­the­lial regen­er­a­tion. Nature. 2015;
idation of Minnesota acute graft-ver­sus-host dis­ease risk score. Haemato- 528(7583):560-564.
logica. 2020;105(2):519-524. 30. Bruce DW, Stefanski HE, Vincent BG, et  al. Type 2 innate lym­phoid cells
13. Adom D, Rowan C, Adeniyan T, Yang J, Paczesny S. Biomarkers for allo­ge­neic treat and pre­vent acute gas­tro­in­tes­ti­nal graft-ver­sus-host dis­ease. J Clin
HCT out­comes. Front Immunol. 2020;11:673. Invest. 2017;127(5):1813-1825.
14. Srinagesh HK, Özbek U, Kapoor U, et al. The MAGIC algo­rithm prob­a­bil­ity is 31. Norona J, Apostolova P, Schmidt D, et al. Glucagon-like pep­tide 2 for intes­ti­
a val­i­dated response bio­marker of treat­ment of acute graft-ver­sus-host dis­ nal stem cell and Paneth cell repair dur­ing graft-ver­sus-host dis­ease in mice
ease. Blood Adv. 2019;3(23):4034-4042. and humans. Blood. 2020;136(12):1442-1455.
15. Holtan SG, Khera N, Levine JE, et al. Late acute graft-ver­sus-host dis­ease: 32. Michonneau D, Latis E, Curis E, et al. Metabolomics anal­y­sis of human acute
a pro­spec­tive anal­y­sis of clin­i­cal out­comes and cir­cu­lat­ing angio­genic fac­ graft-ver­sus-host dis­ease reveals changes in host and microbiota-derived
tors. Blood. 2016;128(19):2350-2358. metab­o­lites. Nat Commun. 2019;10(1):5695.
16. Holtan SG, DeFor TE, Panoskaltsis-Mortari A, et al. Amphiregulin modifies 33. Beckman MF, Morton DS, Bahrani Mougeot F, Mougeot JC. Allogenic stem
the Minnesota acute graft-ver­sus-host dis­ease risk score: results from BMT cell trans­plant-asso­ci­ated acute graft ver­sus host dis­ease: a com­pu­ta­tional
CTN 0302/0802. Blood Adv. 2018;2(15):1882-1888. drug dis­cov­ery text min­ing approach using oral and gut microbiome sig­
17. Holtan SG, Hoeschen AL, Cao Q , et al. Facilitating res­o­lu­tion of life-threat­ na­tures. Support Care Cancer. 2021;29(4):1765-1779.
en­ing acute GVHD with human chorionic gonad­o­tro­pin and epi­der­mal 34. Rashidi A, Ebadi M, Shields-Cutler RR, et al. Early E. casseliflavus gut col­
growth fac­tor. Blood Adv. 2020;4(7):1284-1295. o­ni­za­tion and out­comes of allo­ge­neic hema­to­poi­etic cell trans­plan­ta­tion.
18. Mielcarek M, Furlong T, Storer BE, et al. Effectiveness and safety of lower PLoS One. 2019;14(8):e0220850.
dose pred­ni­sone for ini­tial treat­ment of acute graft-ver­sus-host dis­ease: a 35. Shrestha B, Walton K, Reff J, et al. Human CD83-targeted chi­me­ric anti­gen
ran­dom­ized con­trolled trial. Haematologica. 2015;100(6):842-848. recep­tor T cells pre­vent and treat graft-ver­sus-host dis­ease. J Clin Invest.
19. Pidala J, Hamadani M, Dawson P, et al. Randomized mul­ti­cen­ter trial of siro­ 2020;130(9):4652-4662.
limus vs pred­ni­sone as ini­tial ther­apy for stan­dard-risk acute GVHD: the 36. Crenn P, Vahedi K, Lavergne-Slove A, Cynober L, Matuchansky C, Messing
BMT CTN 1501 trial. Blood. 2020;135(2):97-107. B. Plasma cit­rul­line: a marker of enterocyte mass in vil­lous atro­phy-asso­ci­
20. Martin PJ. How I treat ste­roid-refrac­tory acute graft-ver­sus-host dis­ease. ated small bowel dis­ease. Gastroenterology. 2003;124(5):1210-1219.
Blood. 2020;135(19):1630-1638. 37. El Jurdi N, Rayes A, MacMillan ML, et  al. Steroid-depen­dent acute GVHD
21. Przepiorka D, Luo L, Subramaniam S, et  al. FDA approval sum­mary: rux­ after allo­ge­neic hema­to­poi­etic cell trans­plan­ta­tion: risk fac­tors and clin­i­cal
olitinib for treat­ment of ste­roid-refrac­tory acute graft-ver­sus-host dis­ease. out­comes. Blood Adv. 2021;5(5):1352-1359.
Oncologist. 2020;25(2):e328-e334.
22. Zeiser R, von Bubnoff N, Butler J, et al; REACH2 Trial Group. Ruxolitinib for
glu­co­cor­ti­coid-refrac­tory acute graft-ver­sus-host dis­ease. N Engl J Med. © 2021 by The Amer­i­can Society of Hematology
2020;382(19):1800-1810. DOI 10.1182/hema­tol­ogy.2021000300

Prakash Singh Shekhawat


Acute GVHD  | 647
UPDATE IN GRAFT- VERSUS - HOST DISEASE

Updates in chronic graft-versus-host disease


Betty K. Hamilton

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/648/1851934/648hamilton.pdf by guest on 13 December 2021


Blood and Marrow Transplant Program, Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH

Chronic graft-versus-host disease (GVHD) is the leading cause of late morbidity and mortality after allogeneic hema-
topoietic cell transplantation. Symptoms and manifestations of chronic GVHD are heterogeneous and pleomorphic,
and there are no standard treatments beyond corticosteroids. Therapy is typically prolonged, and chronic GVHD and
its treatment are associated with adverse effects that have a significant impact on long-term quality of life and func-
tional status. Several advances have been made over the last 2 decades to define the diagnosis of chronic GVHD as
well as its severity and response criteria for clinical trials. Further understanding into the biologic mechanisms of the
development of chronic GVHD has led to the investigation of several novel immunomodulatory and targeted therapies.
Multi-institutional collaboration and pharmaceutical support in the development of therapies based on sound biologic
mechanisms and clinical trials with defined end points and responses have led to several promising agents on the hori-
zon of approval for treatment of chronic GVHD. This article reviews advances in our knowledge of chronic GVHD and its
biologic framework to improve approaches to prevention and treatment.

LEARNING OBJECTIVES
• Describe biologic phases and mechanisms of chronic GVHD development
• Describe novel approaches to chronic GVHD prevention and treatment
• Understand the continued need for high-quality multicenter clinical trials based on biologic rationale and using
chronic GVHD end points

CLINICAL CASE ing tacrolimus. The patient was very reluctant to consider
A 64-year-old male with a history of relapsed acute mye- systemic corticosteroids or other medications. He devel-
logenous leukemia in second complete remission under- oped worsening fatigue, anorexia, shortness of breath
went a reduced-intensity matched unrelated-donor (pleural effusions), lower-extremity swelling, dysphagia,
peripheral blood stem cell transplant. Graft-versus-host and anemia/thrombocytopenia. Additional workup to rule
disease (GVHD) prophylaxis consisted of tacrolimus and out other etiologies was consistent with a GVHD flare. He
mycophenolate mofetil (MMF). In the setting of tapering was restarted on 0.5mg/kg prednisone with plans for a
immunosuppression, 4 months after transplant he devel- quick taper and started on ibrutinib. He had an excellent
oped worsening fatigue, anorexia, skin rash, dry eyes, and response; his counts improved and symptoms resolved. He
dry mouth. Skin biopsy and ocular evaluation confrmed was able to taper off prednisone without a flare in GVHD.
chronic GVHD. Tacrolimus was increased to therapeutic He remains on low-dose tacrolimus and ibrutinib.
levels, and he was started on 1mg/kg prednisone. Topical
therapies including dexamethasone oral rinses and ocu-
lar agents were used, including scleral lenses. The GVHD Introduction
symptoms resolved on steroids; however, he had several Allogeneic hematopoietic cell transplantation (HCT) is
adverse effects to steroids, such as hyperglycemia, irrita- the only potentially curative therapy for many high-risk
bility, edema, and cytomegalovirus reactivation. Systemic hematologic malignancies, metabolic and immunode-
corticosteroids were subsequently successfully tapered fciencies, and bone marrow failure syndromes, and the
off. The patient was eager to get off all immunosuppres- number of transplant procedures continues to increase.
sion, but again, after a taper of tacrolimus, he had a flare of Despite signifcant advances in donor selection, condi-
GVHD with oral, eye, and skin involvement. There was some tioning regimens, and supportive care, chronic GVHD
improvement with maximizing topical therapy and increas- remains the leading cause of late morbidity and mortal-

648 | Hematology 2021 | ASH Education Program


ity.1,2 The inci­dence of chronic GVHD ranges from 30% to 50%, Tissue dam­age to the thy­mus and the sec­ond­ary lym­phoid
depending on GVHD pro­phy­laxis reg­i­mens and the donor graft organs pre­dis­poses patients to sub­se­quent chronic GVHD. The
source.1 Chronic GVHD, once established, is a het­ero­ge­neous sec­ ond phase is thus char­ ac­
ter­ized by thy­ mic injury, which
and pleo­ mor­phic syn­drome in which most patients have at allows for the emer­gence of and the selec­tion of auto- and allo-
least 3 involved organs, and treat­ment typ­i­cally requires the reactive T-cell pop­u­la­tions.13 A loss of cen­tral and periph­eral tol­
prolonged (median 2-3.5 years) use of immu­ no­sup­ pres­ sive er­ance leads to impaired or defi­cient reg­u­la­tory T (Treg) and
agents.3 Chronic GVHD and its treat­ment are thus often asso­ci­ B cells.15 Donor T fol­lic­u­lar helper (Tfh) cells have been shown to
ated with late mor­tal­ity as well as sev­eral adverse effects with expand in the sec­ond­ary lym­phoid organs and pro­mote the sur­
sig­nif­i­cant impact on long-term qual­ity of life and func­tional vival, expan­sion, and dif­fer­en­ti­a­tion of donor B cells into aber­rant
sta­tus among HCT sur­vi­vors.2,4 anti-host-immu­no­glob­u­lin-pro­duc­ing plasma cells via cyto­kines
The National Institutes of Health (NIH) chronic GVHD pro­jects such as IL-21 and B-cell acti­vat­ing fac­tor.16 Chronic inflam­ma­tion
of 2005 and 2014 have pro­vided a crit­i­cal frame­work for sub­ is thought to be maintained by Th17 cells that have escaped
stan­tial advances in the trans­plant field over the last decade,5,6 immune reg­u­la­tion.12

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/648/1851934/648hamilton.pdf by guest on 13 December 2021


outlining stan­dard guide­lines and def­i­ni­tions for chronic GVHD The third phase is marked by aber­rant tis­sue repair. Activated
diag­no­sis, grad­ing, and response cri­te­ria for clin­i­cal tri­als.7,8 Fur- mac­ro­phages depen­dent on IL-17 and col­ony-stim­u­lat­ing fac­tor
ther refine­ment of response def­i­ni­tions has enhanced the reli­ (CSF-1) have been shown to play an impor­tant role in this by
abil­ity and prac­ti­cal util­ity of these mea­sures in clin­i­cal tri­als, pro­duc­ing transforming growth fac­ tor β and plate­ let-derived
ulti­mately lead­ing to the first US Food and Drug Administration growth fac­tor alpha.17 This leads to fibro­blast acti­va­tion, result-
(FDA) approval of treat­ment for chronic GVHD.9 In 2020, the NIH ing in chronic GVHD man­ i­
fes­
ta­tions such as sclero­ derma or
Chronic GVHD Consensus Conference fur­ ther set a for­ ward- bronchiolitis obliterans. Thus, it is increas­ingly clear that sub­clin­
think­ing agenda to address gaps and needs in future research i­cal path­o­genic pro­cesses crit­i­cal to the devel­op­ment of chronic
over the next 3 to 7 years. This arti­ cle sum­ ma­ rizes recent GVHD begin long before the spe­cific clin­i­cal man­i­fes­ta­tions
advances in our under­stand­ing of chronic GVHD and its (1) eti­­ become evi­dent.
ol­ogy and pre­ven­tion, (2) devel­op­ment and diag­no­sis, and (3)
novel treat­ments. Chronic GVHD pre­ven­tion
Calcineurin inhib­i­tor (CNI)-based pro­phy­laxis, typ­i­cally in com­bi­
Etiology and pre­ven­tion na­tion with meth­o­trex­ate (MTX) or MMF, has been stan­dard prac­
Advances in our under­stand­ing of chronic GVHD have led to the tice over the past 3 decades for GVHD pre­ven­tion.18 Although
rec­og­ni­tion of sev­eral donor, recip­i­ent, and trans­plant fac­tors these reg­i­mens may have rel­a­tive ben­e­fit in lim­it­ing acute GVHD,
that con­trib­ute to its ini­ti­a­tion and devel­op­ment. Established they are not effec­tive against chronic GVHD and, par­a­dox­i­cally,
clin­i­cal risk fac­tors include growth fac­tor-mobi­lized periph­eral may sup­port its devel­op­ment by blocking thy­mic cen­tral tol­er­
blood stem cells, mismatched or unre­lated donor grafts, female- ance and Treg func­tion.19-21 The need for improve­ments in GVHD
to-male trans­plan­ta­tion, older recip­i­ent age, and acute GVHD pre­ven­tion strat­e­gies has been rec­og­nized as a pri­or­ity by the
his­tory.10 Preclinically, although no murine model ade­ quately field,22 and sev­eral novel strat­e­gies and end points have been
mim­ics the clin­i­cal spec­trum of chronic GHVD,11 bio­logic insights inves­ti­gated.
from mul­ti­ple mod­els with dis­tinct pathophysiologies have laid Although sev­eral pro­phy­laxis strat­e­gies have been shown
a foun­da­tion lead­ing to fur­ther clin­i­cal stud­ies and new treat­ to decrease the inci­dence of chronic GVHD, their effects on
ment par­a­digms. Several detailed and com­pre­hen­sive reviews immune recon­ sti­
tu­tion and dis­ease relapse remain prob­ lem­
of chronic GVHD biol­ogy and pre­clin­i­cal mod­els already exist atic. Because donor-derived effec­tor T cells are crit­i­cal ini­ti­a­
and are out­side the scope of this arti­cle.12-14 Key ele­ments in the tors of chronic GVHD, approaches to deplete T cells from the
path­o­phys­i­­ol­ogy, how­ever, will be briefly sum­ma­rized here to hema­to­poi­etic cell graft have been inves­ti­gated, dem­on­strat­ing
pro­vide a frame­work for the updates on novel ther­a­peu­tic tar­ sig­nif­i­cant reduc­tions in the inci­dence and sever­ity of chronic
gets for pre­ven­tion and treat­ment. GVHD. While the ex vivo removal of T cells by the pos­i­tive selec­
tion of CD34+ cells has become the pre­dom­i­nant approach in
Etiology and path­o­gen­e­sis clin­i­cal use,23 other novel approaches, such as αβ-T-cell graft
Chronic GVHD is a result of com­plex and dynamic mech­a­nisms deple­tion, have dem­on­strated sig­nif­i­cant reduc­tions in mod­er­
and can be thought to occur within 3 bio­logic phases: (1) early ate and severe chronic GVHD.24 Although T-cell deple­tion has
inflam­ma­tion due to tis­sue injury, (2) thy­mic injury and T- and B- not been shown to increase relapse risk, it is asso­ci­ated with
cell dysregulation, and (3) tis­sue repair and fibro­sis (Figure 1).14 delayed immune recon­sti­tu­tion and higher rates of infec­tion-
Although these pro­cesses are often overlapping and depen­dent, related mor­tal­ity,23 which was recently con­firmed in a phase 3
they do not nec­es­sar­ily need to occur in sequence, or even occur mul­ti­cen­ter Blood and Marrow Transplant Clinical Trials Network
at all­. As with acute GVHD, exper­i­men­tal mod­els sug­gest that the (BMT CTN) trial.25 In vivo T-cell deple­tion with antithymocyte
ini­tial phase of chronic GVHD also begins with dam­age of host tis­ glob­ u­lin (ATG) has also been effec­ tive in reduc­ ing the inci­
sues by pretransplant con­di­tion­ing and the sub­se­quent release of dence of chronic GVHD. Several phase 3 stud­ies have con­firmed
inflam­ma­tory cyto­kines. Damage to gut tis­sues and the release of reduc­tions in chronic GVHD with ATG, with sub­se­quent supe­
micro­bial con­tents result in the acti­va­tion of anti­gen-presenting rior qual­ity of life and GVHD-free sur­vival.26-28 However, con­tro­
cells, and inflam­ma­tory cyto­kines stim­u­late the acti­va­tion of do- versy remains on its effect on over­all sur­vival due to increases
nor alloreactive T cells, driv­ing helper T (Th1)/T-cyto­toxic 1 (Tc1), in relapse and nonrelapse mor­tal­ity, as well as opti­mal dos­ing,
and Th17/Tc17 dif­fer­en­ti­a­tion and expan­sion of effec­tor T cells, for­mu­la­tion, and depen­dency on lym­pho­cyte count at the time
caus­ing fur­ther cyto­tox­ic­ity to host tar­get cells.12 of admin­is­tra­tion.28
Prakash Singh Shekhawat
Chronic graft-ver­sus-host dis­ease | 649
Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/648/1851934/648hamilton.pdf by guest on 13 December 2021
Figure 1.  Proposed biologic phases of chronic GVHD. A few novel therapeutic approaches targeting biologic pathways are highlight-
ed. Reproduced with permission from the Cleveland Clinic Center for Medical Art and Photography.

The role of donor B cells in chronic GVHD biol­ogy has also set­ting and has shown itself to be a poten­tially effec­tive strat­
pro­vided a strong ratio­nale for the eval­u­a­tion of B-cell deple­ egy for both acute and chronic GVHD pre­ven­tion.
tion strat­eg­ ies for chronic GVHD pre­ven­tion. Prior stud­ies have The opti­ mal GVHD pro­ phy­ laxis reg­

men for the pre­ ven­
incor­po­rated mono­clo­nal anti-B-cell ther­apy as part of the con­ tion of chronic GVHD remains elu­sive. A bench­mark anal­y­sis of
di­tion­ing reg­im­ en or the peritransplant period with the intent novel GVHD pre­ven­tion was conducted through the Center for
of reduc­ing relapse of B-cell malig­nan­cies.29 Although sig­nif­i­cant International Blood and Marrow Transplant Research to select
deple­tion of B cells and reduced rates of chronic GVHD have prom­is­ing agents and new, clin­i­cally mean­ing­ful out­come mea­
been reported,30 con­cerns over the delayed recon­sti­tu­tion of sure­ ments for fur­ ther inves­
ti­
ga­tion through the BMT CTN.33
B-cell immu­nity and excess infec­tions remain a con­cern. Several com­pos­ite GVHD end points were explored, includ­ing
Posttransplant cyclo­ phos­ pha­
mide (PTCy) is a promising chronic GVHD relapse-free sur­vival (CRFS), which includes sur­
approach used suc­ cess­fully to pre­
vent GVHD in the HLA-​ vival with­out devel­op­ment of chronic GVHD, dis­ease relapse, or
mismatched/haploidentical set­ting.31 Recent work has pro­ death, and GVHD relapse-free sur­vival (GRFS), which includes
posed that the effi­cacy of PTCy is due to the reduc­tion of sur­vival with­out acute grade 3 to 4 GVHD, chronic GVHD, dis­
alloreactive CD4+ effec­tor T-cell pro­lif­er­a­tion, impaired func­ ease relapse, or death. The results of these ana­ly­ses led to the
tion­ al­
ity of sur­ viv­
ing CD4+ and CD8+ effec­ tor T cells, and devel­op­ment of 2 mul­ti­cen­ter BMT CTN clin­i­cal tri­als. PROG-
pref­er­en­tial recov­ery of CD4+ Treg cells.32 PTCy has been inves­ RESS 1 (BMT CTN 1203) was a ran­dom­ized phase 2, 3-arm trial
ti­gated both with and with­out CNI in the fully HLA-matched inves­ti­gat­ing bortezomib/Tac/MTX, maraviroc/Tac/MTX, and

650  |  Hematology 2021  |  ASH Education Program


PTCy/Tac/MMF in reduced-inten­sity HCT, eval­u­at­ing GRFS as Treatment of established chronic GVHD
a pri­mary end point. PTCy was asso­ci­ated with supe­rior GRFS Given the het­ero­ge­neous man­i­fes­ta­tions of established chron-
and had the low­est inci­dence of chronic GVHD requir­ing immu­ ic GVHD, clear guide­lines for treat­ment are lacking. High-dose
no­sup­pres­sion com­pared to other arms.34 This led to the ongo­ cor­ti­co­ste­roids, typ­i­cally 0.5 to 1 mg/kg/d, remain the first-line
ing BMT CTN trial 1703 (NCT03959241) phase 3 trial com­par­ing stan­dard treat­ment for chronic GVHD.3 While there is no cur­
PTCy/Tac/MMF with Tac/MTX, with the pri­mary end point of rent evi­dence to sup­port any ben­e­fit from addi­tional agents
GRFS, poten­tially establishing a new stan­dard of care for GVHD in the up-front set­ting, they are often con­sid­ered in severe
pre­ven­ tion. PROGRESS 2 (BMT CTN 1301) was a ran­ dom­ ized cases. The response rate to ste­roids alone is about 50%, with
phase 3, open-label, 3-arm trial com­par­ing CNI-free approaches more than half of patients requir­ing sec­ond-line ther­apy with-
of CD34 selec­tion, PTCy, and Tac/MTX, with a pri­mary end point in 2 years. A num­ber of sec­ond-line agents of vary­ing effi­cacy
of CRFS. While there were no sta­tis­ti­cally sig­nif­i­cant dif­fer­ences and toxicities have been inves­ ti­
gated over the past sev­ eral
in CRFS between the 3 approaches, CD34+ selec­tion was found decades, high­light­ing the het­ero­ge­ne­ity in prac­tice, var­i­abil­
to be asso­ci­ated with worse over­all sur­vival, driven by increased ity in response rates, and chal­lenges in deter­min­ing the best

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/648/1851934/648hamilton.pdf by guest on 13 December 2021


infec­tion-related mor­tal­ity.25 treat­ment options.43 Corticosteroids thus remain the stan­dard
despite con­tinu­ing to be asso­ci­ated with sev­eral short- and
Other novel strat­e­gies long-term toxicities, includ­ ing infec­tion, myop­ a­
thy, edema,
As we work to fur­ther under­stand the devel­op­ment of GVHD, we hyper­gly­ce­mia, bone loss, avas­cu­lar necro­sis, cat­a­racts, and
now rec­og­nize sev­eral addi­tional poten­tial tar­gets. For exam­ple, sleep/mood dis­tur­bances. In clin­i­cal prac­tice, top­i­cal ther­a­pies
instead of blan­ket B- or T-cell deple­tion, pre­ven­tion strat­e­gies and sup­port­ive mea­sures to min­i­mize these adverse effects are
to block the devel­op­ment of path­o­genic B cells, or to inhibit of crit­i­cal impor­tance.44
Tfh cells, may bet­ter pre­vent chronic GVHD with­out affect­ing The ideal treat­ment for chronic GVHD con­tin­ues to be elu­
other out­comes. Further, strat­e­gies to aug­ment Tregs, as have sive. Goals of ther­apy are sev­eral in num­ber but should include
been reported with novel agents such as IL-2,35 ruxolitinib,36 symp­tom bur­den reduc­tion and improve­ment of qual­ity of life,
hypomethylating agents,37 extra­cor­po­real photopheresis,38 pro- pre­ven­tion of pro­gres­sion and inflam­ma­tory activ­ity, pre­ven­tion
teasome inhib­i­tors,39 and adop­tive trans­fer of Tregs,40 may need of fibro­sis and dis­abil­ity, pres­er­va­tion of response to allow for
addi­tional explo­ra­tion. Table 1 pro­vi­des a sum­mary of sev­eral with­drawal of immu­no­sup­pres­sion, repair and mod­u­la­tion of the
recently published and ongo­ ing stud­ ies that include chronic immune sys­tem, and, ulti­mately, improve­ment of chronic GVHD
GVHD as a pri­mary or as part of a pri­mary com­pos­ite end point. mor­tal­ity.
With these aims in mind, the focus of novel chronic GVHD
Development and diag­no­sis treat­ ment has thus shifted from the use of broad, long-term
While the 2005 and 2014 NIH Chronic GVHD Consensus Con- immu­no­sup­pres­sion toward the inves­ti­ga­tion of immu­no­mod­u­
ference papers established and refined stan­dard def­i­ni­tions for la­tory agents that tar­get path­ways rel­e­vant to the path­o­phys­i­­ol­
chronic GVHD diag­no­sis,5,6 the 2020 Chronic GVHD Consen- ogy of the dis­ease (Figure 1). Several mul­ti­cen­ter phase 2 and 3
sus Conference fur­ther high­lighted sev­eral impor­tant con­sid­ clin­i­cal tri­als have now inves­ti­gated mul­ti­ple agents in the treat­
er­ations in the devel­op­ment and diag­no­sis of chronic GVHD, ment of up-front or ste­roid-refrac­tory chronic GVHD, and a few
focus­ ing espe­ cially on the neces­ sity of early rec­og­ni­tion.41 recently reported tri­als are briefly reviewed here.
These include the need for ear­lier clin­i­cal iden­ti­fi­ca­tion by both Ibrutinib is a small-molec­u­lar tyro­sine kinase inhib­i­tor target-
pro­vid­ers and patient/care­giv­ers, poten­tially with the use of ing Bruton’s tyro­sine kinase (BTK), which plays an impor­tant role
tech­nol­ogy such as telehealth and apps, as well as the need in B-cell recep­tor sig­nal­ing and acti­va­tion of B cells and T cells.
for bet­ter iden­ti­fi­ca­tion of pre­ced­ing symp­toms or bio­mark­ A phase 2 trial of ibrutinib in 42 patients with ste­roid-refrac­tory
ers asso­ci­ated with sub­se­quent more severe/mor­bid forms chronic GVHD led to the first FDA approval of a sec­ond-line ther­
of chronic GVHD—includ­ing bio­mark­ers from blood and tis­ apy for chronic GVHD, dem­on­strat­ing an over­all response rate
sue or find­ings from imag­ing or func­tional test­ing. Trials spe­ of 67%. Approximately 71% of respond­ers con­tin­ued to show
cif­i­cally inves­ti­gat­ing the devel­op­ment of chronic GVHD with a response for at least 20 weeks, with responses gen­ er­
ally
increased mon­i­tor­ing using blood, fluid, tis­sue, imag­ing, and observed across all­organ sys­tems involved.9 Although the num­
patient-reported symp­toms at time points through­out trans­ ber of patients treated on this trial was rel­a­tively small, the FDA
plant to detect early signs of dis­ease are cur­rently ongo­ing approval of this drug high­lights the suc­cess of multi-insti­tu­tional
(NCT04372524, NCT04188912). col­lab­o­ra­tion and phar­ma­ceu­ti­cal sup­port in devel­op­ing a trial
The early iden­ti­fi­ca­tion of symp­toms or sub­clin­i­cal indi­ca­tors based on pre­clin­i­cal data and clear bio­logic mech­a­nisms, with
of chronic GVHD is nec­es­sary for the pro­posed approach of pre­ defined clin­i­cal end points.
emp­tive treat­ment of chronic GVHD,42 poten­tially allowing for Ruxolitinib is a JAK 1/2 inhib­i­tor that suppresses T-cell acti­va­
the pre­ven­tion of the more severe and mor­bid phe­no­types of tion by inhibiting cyto­kine recep­tor-medi­ated sig­nal­ing for a vari­
chronic GVHD. In a pre­ven­ta­tive/pre­emp­tive approach with rit- ety of proinflammatory cyto­kines and is cur­rently FDA approved
uximab posttransplant, a mod­est decrease in chronic GVHD and for the treat­ment of ste­roid-refrac­tory acute GVHD.36 A phase 3
ste­roid-requir­ing chronic GVHD was found.30 Ideally, future pre­ study of ruxolitinib vs best avail­­ able ther­apy (BAT) in ste­ roid-
emp­tive approaches will have the advan­tage of directing treat­ refrac­tory chronic GVHD was recently reported,45 dem­on­strat­ing
ment at patients who have been iden­ti­fied as at greatest risk, an over­all response rate of 50% com­pared to 26% in the BAT arm.
allowing for early, more targeted and sub­se­quently less long- Failure-free sur­vival was sig­nif­i­cantly lon­ger for ruxolitinib-treated
term, dam­ag­ing ther­apy. patients, and symp­tom improve­ment was greater with ruxolitinib

Prakash Singh Shekhawat


Chronic graft-ver­sus-host dis­ease | 651
Table 1.  Recently published and ongo­ing pre­ven­tion stud­ies with chronic GVHD as a pri­mary end point

Phase Prevention reg­i­men Primary end point/out­come Reference or NCT num­ber


Phase 3 ATG vs no ATG with CSA/MTX Chronic GVHD at 2 years: 32.2% (ATG) vs 68.7% (no ATG), P < .001 26
Phase 3 ATG vs no ATG with CNI/MTX Freedom from sys­temic immu­no­sup­pres­sion with­out resump­tion 27
up to 12 months: 37% (ATG) vs 16% (no ATG), OR 4.25, P = .0006
Phase 3 ATG vs no ATG with Tac/MTX Moderate-severe chronic GVHD-free sur­vival: 48% (ATG) vs 44% 28
(no ATG), P = .47
Phase 2 PTCy/CSA Chronic GVHD requir­ing sys­temic treat­ment: 16% (95% CI, 5%- 48
28%)
Phase 2 PTCy/Tac/MMF vs bortezomib/Tac/MTX GRFS: HR 0.72, P = .04 (PTCy/Tac/MMF) vs HR 0.98, 49
vs maraviroc/Tac/MTX P = .92 (bortezomib/Tac/MTX) vs HR 1.10, P = .49
(maraviroc/Tac/MTX)

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/648/1851934/648hamilton.pdf by guest on 13 December 2021


Phase 2 Rituximab (D100, 6, 9, 12 months) post-HCT Chronic GVHD: 48%, chronic GVHD requir­ing ste­roids: 31% 30
Phase 3 CD34 selec­tion vs PTCy vs Tac/MTX CRFS: 60.2% (CD34), 60.3% (PTCy), 56.6% (Tac/MTX), P = .72 25
Phase 1/2 Ixazomib/Tac/Sir Moderate/severe chronic GVHD NCT03225417
Phase 2 Palifermin Chronic GVHD NCT02356159
Phase 2 Obinutuzumab Chronic GVHD requir­ing ste­roids NCT02867384
Phase 2 Ixazomib/CNI/MTX Acute or chronic GVHD NCT03082677
Phase 2 PTCy/Tac/MMF GRFS NCT03128359
Phase 2 PTCy/Sir/MMF Extensive chronic GVHD NCT03192397
Phase 2 Tocilizumab/Tac/MTX GRFS NCT03699631
Phase 2 Ex vivo deple­tion of CD45RA +
GRFS NCT03779854
cells/Tac/MTX
Phase 3 Atorvastatin/Tac/MTX Chronic GVHD NCT03066466
Phase 3 PTCy/Tac/MMF vs Tac/MTX GRFS NCT03959241
CSA, cyclosporine.

com­pared to BAT (24% vs 11%). Based on the results of this study, Despite our improved under­stand­ing of chronic GVHD, it
ruxolitinib has been granted pri­or­ity review by the FDA for the remains a com­plex dis­ease marked by sig­nif­i­cant immune dys­
treat­ment of chronic GVHD, and an FDA deci­sion on drug approval func­tion, dis­abil­ity, and toxicities, and chronic GVHD-related
is expected before the end of the year. non-relapse mortality keeps ris­ing over time.47 Practically, the
Belumosudil is an oral selec­ tive rho-asso­ ci­
ated coiled-coil choice of first- and sec­ond-line ther­apy con­tin­ues to depend on
kinase 2 (ROCK) inhib­i­tor targeting Tfh-cell gen­er­a­tion and thus phy­si­cian pref­er­ences and clin­i­cal expe­ri­ence, patient dis­ease
B-cell dif­fer­en­ti­at­ion and sur­vival. Results of the phase 2 trial his­tory and comorbidities, cost/avail­abil­ity of treat­ment, and
of belumosudil in ste­ roid-refrac­ tory chronic GVHD were also access to clin­i­cal tri­als. As we prog­ress away from broad immu­
recently reported,46 dem­on­strat­ing an over­all response rate of no­ sup­pres­sion, there remains a need to inves­ ti­gate agents
74% in heavily pretreated patients. Responses were seen across that tar­get the bio­logic path­ways rel­e­vant to the path­o­phys­
all­affected organs, and of respond­ers, 49% maintained response i­­ol­ogy of dis­ease and chronic GVHD symp­toms; to ulti­mately
for at least 20 weeks. Failure-free sur­vival was 77% at 6 months. under­stand which drug will work when. The iden­ti­fi­ca­tion and
Based on the results of this study, belumosudil has also been use of blood and tis­sue bio­mark­ers, imag­ing, and other novel
granted pri­or­ity review by the FDA. approaches can bet­ter iden­tify spe­cific clin­i­cal and bio­logic
The need to inves­ti­gate other novel ther­a­pies either targeted phe­no­types to dif­fer­en­ti­ate chronic GVHD and work toward a
toward fibro­sis or already being devel­oped for other fibrotic dis­ more tai­lored, per­son­al­ized ther­apy for patients.
eases has been rec­og­nized. For exam­ple, pirefenidone, which Reflecting back to our clin­i­cal case, this sce­nario dem­on­
inhib­its transforming growth fac­tor β sig­nal­ing, is cur­rently FDA strates sev­eral aspects of chronic GVHD devel­op­ment, evo­lu­
approved for idi­o­pathic pul­mo­nary fibro­sis and has been shown tion, and treat­ment chal­lenges and high­lights the impor­tance of
in mouse mod­els to restore pul­mo­nary func­tion and reverse lung under­stand­ing the goals of ther­apy from both a patient and pro­
fibro­sis. Its study in chronic GVHD-related bronchiolitis obliter- vider per­spec­tive. While broad immu­no­sup­pres­sion may reduce
ans is ongo­ing (NCT03315741). Axatilimab is a mono­clo­nal anti­ symp­ tom bur­ den and decrease inflam­ ma­tory activ­ ity in the
body against CSF-1, which reg­u­lates mono­cyte dif­fer­en­ti­a­tion short term, there remains a need to fur­ther inves­ti­gate targeted
into mac­ro­phages pro­mot­ing fibro­sis, and has been shown to agents to pre­serve func­tion, pro­vide a prolonged response to
have poten­tial effi­cacy in heavily pretreated chronic GVHD. A allow for the with­drawal of med­i­ca­tions, pre­vent dis­abil­ity, and
phase 2 study fur­ther eval­ua ­ t­ing this is ongo­ing (NCT03604692). ulti­mately improve qual­ity of life and over­all mor­tal­ity. The con­
Table 2 sum­ma­rizes sev­eral other up-front and ste­roid-refrac­tory tin­ued under­stand­ing of bio­logic mech­a­nisms through multi-
chronic GVHD tri­als. insti­tu­tional col­lab­o­ra­tive pre­clin­i­cal studies, clin­i­cal tri­als with

652  |  Hematology 2021  |  ASH Education Program


Table 2.  Active ongo­ing stud­ies for sys­temic treat­ment in up-front or ste­roid-refrac­tory chronic GVHD

Drug mech­a­nism/
Phase Treatment Indication Enrollment Primary end point NCT num­ber
tar­get
Pilot Itacitinib JAK inhib­i­tor Steroid refrac­tory 40 ORR at 6 months NCT04200365
Phase 2 Itacitinib and ECP JAK inhib­i­tor, Treg Up-front 58 ORR at 24 weeks; NCT0446182
dose-lim­it­ing tox­ic­ity
Phase 1 SHR0302 JAK inhib­i­tor Up-front 30 Adverse events NCT04146207
Phase 1 Abatacept T-cell costim block­ Steroid refrac­tory 22 Maximum tol­er­ated dose NCT01954979
ade
Phase Itacitinib JAK inhib­i­tor Up-front 431 Dose deter­mi­na­tion; NCT03584516
2/3 ORR at 6 months

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/648/1851934/648hamilton.pdf by guest on 13 December 2021


Phase 2 Mesenchymal stem cells Immunomodulatory Up-front 152 ORR at 12 weeks NCT04692376
Phase 2 IL-2 and ECP Treg Steroid refrac­tory 24 ORR at 4 weeks NCT03007238
Phase 1/2 Ofatumumab CD20 B-cell anti­body Up-front 44 ORR at 6 months; max­i­ NCT010680965
mum tol­er­ated dose
Phase 2 KD025 (belumosudil) ROCK inhib­i­tor Steroid refrac­tory 166 ORR at 6 months NCT03640481
Phase 2 Acalabrutinib BTK inhib­i­tor Recurrent/pro­gres­sive 50 Best response at 6 NCT04198922
months
Phase 1/2 Glasdegib Hedgehog sig­nal­ing Sclerosis/ste­roid- 21 Maximum tol­er­ated dose NCT03415867
inhib­i­tor refrac­tory
Phase 2 Ibrutinib/rituximab BTK inhib­i­tor/CD20 Up-front 35 Off immu­no­sup­pres­sion NCT04235036
B-cell anti­body at 8 weeks
Phase 1 Leflunomide Immunomodulatory Steroid depen­dent 24 Dose-lim­it­ing tox­ic­ity NCT04212416
Phase 2 Axalitimab CSF-1 inhib­i­tor Steroid refrac­tory 210 ORR at 6 months NCT04710576
Phase 2 Ibrutinib BTK inhib­i­tor Up-front 40 ORR at 6 months NCT04294641
Phase 1/2 Baricitinib JAK inhib­i­tor Steroid refrac­tory 31 Safety and effi­cacy NCT02759731
Phase 3 Ruxolitinib JAK inhib­i­tor Steroid refrac­tory 330 ORR at day 168 NCT03112603
ECP, extracorporeal photopheresis; ORR, overall response rate; ROCK, rho-asso­ci­ated coiled-coil kinase.

care­ful cor­rel­a­tive stud­ies of blood, fluid, tis­sue sam­ples, imag­ 3. Flowers ME, Martin PJ. How we treat chronic graft-ver­sus-host dis­ease.
ing, and patient-reported symp­toms—are nec­es­sary to achieve Blood. 2015;125(4):606-615.
4. Wingard JR, Majhail NS, Brazauskas R, et  al. Long-term sur­vival and late
a more per­son­al­ized approach to the pre­ven­tion and treat­ment deaths after allo­ge­neic hema­to­poi­etic cell trans­plan­ta­tion. J Clin Oncol.
of chronic GVHD. 2011;29(16):2230-2239.
5. Filipovich AH, Weisdorf D, Pavletic S, et al. National Institutes of Health
con­sen­sus devel­op­ment pro­ject on cri­te­ria for clin­i­cal tri­als in chronic
Conflict-of-inter­est dis­clo­sure graft-​ ver­
sus-host dis­ ease, I: Diagnosis and Staging Working Group
Betty K. Hamilton: con­sul­tancy: Syndax, Equilium. report. Biol Blood Marrow Transplant. 2005;11(12):945-956.
6. Jagasia MH, Greinix HT, Arora M, et al. National Institutes of Health con­sen­
sus devel­op­ment pro­ject on cri­te­ria for clin­i­cal tri­als in chronic graft-ver­
Off-label drug use sus-host dis­ease, I: the 2014 Diagnosis and Staging Working Group report.
Betty K. Hamilton: none. Biol Blood Marrow Transplant. 2015;21(3):389-401.e1.
7. Pavletic SZ, Martin P, Lee SJ, et al; Response Criteria Working Group. Mea-
suring ther­a­peu­tic response in chronic graft-ver­sus-host dis­ease: National
Correspondence Institutes of Health con­sen­sus devel­op­ment pro­ject on cri­te­ria for clin­i­cal
Betty K. Hamilton, Blood and Marrow Transplant Program, Hema- tri­als in chronic graft-ver­sus-host dis­ease, IV: Response Criteria Working
tology and Medical Oncology, Taussig Cancer Institute, Cleve- Group report. Biol Blood Marrow Transplant. 2006;12(3):252-266.
land Clinic, 9500 Euclid Ave, Cleveland, OH 44195; e-mail: ham- 8. Lee SJ, Wolff D, Kitko C, et al. Measuring ther­a­peu­tic response in chronic
graft-ver­sus-host dis­ease. National Institutes of Health con­sen­sus devel­
iltb2@ccf​­.org.
op­ment pro­ject on cri­te­ria for clin­i­cal tri­als in chronic graft-ver­sus-host
dis­ease, IV: the 2014 Response Criteria Working Group report. Biol Blood
References Marrow Transplant. 2015;21(6):984-999.
1. Arai S, Arora M, Wang T, et al; Graft-ver­sus-Host Disease Working Com- 9. Miklos D, Cutler CS, Arora M, et al. Ibrutinib for chronic graft-ver­sus-host
mittee of the CIBMTR. Increasing inci­dence of chronic graft-ver­sus-host dis­ease after fail­ure of prior ther­apy. Blood. 2017;130(21):2243-2250.
dis­ease in allo­ge­neic trans­plan­ta­tion: a report from the Center for Inter- 10. Flowers ME, Inamoto Y, Carpenter PA, et al. Comparative anal­y­sis of risk
national Blood and Marrow Transplant Research. Biol Blood Marrow Trans­ fac­tors for acute graft-ver­sus-host dis­ease and for chronic graft-ver­sus-
plant. 2015;21(2):266-274. host dis­ease according to National Institutes of Health con­sen­sus cri­te­ria.
2. Pidala J, Kurland B, Chai X, et  al. Patient-reported qual­ity of life is asso­ Blood. 2011;117(11):3214-3219.
ci­ated with sever­ity of chronic graft-ver­sus-host dis­ease as mea­sured by 11. Zeiser R, Blazar BR. Preclinical mod­els of acute and chronic graft-ver­sus-
NIH cri­te­ria: report on base­line data from the Chronic GVHD Consortium. host dis­ease: how pre­dic­tive are they for a suc­cess­ful clin­i­cal trans­la­tion?
Blood. 2011;117(17):4651-4657. Prakash Singh Shekhawat Blood. 2016;127(25):3117-3126.

Chronic graft-ver­sus-host dis­ease | 653


12. MacDonald KP, Hill GR, Blazar BR. Chronic graft-ver­sus-host dis­ease: bio­log­ 32. Nunes NS, Kanakry CG. Mechanisms of graft-ver­sus-host dis­ease pre­ven­
i­cal insights from pre­clin­i­cal and clin­i­cal stud­ies. Blood. 2017;129(1):13-21. tion by post-trans­plan­ta­tion cyclo­phos­pha­mide: an evolv­ing under­stand­
13. Zeiser R, Blazar BR. Pathophysiology of chronic graft-ver­sus-host ­dis­ease ing. Front Immunol. 2019;10(Novem­ber):2668.
and ther­a­peu­tic tar­gets. N Engl J Med. 2017;377(26):2565-2579. 33. Pasquini MC, Logan B, Jones RJ, et al. Blood and mar­row trans­plant clin­ic ­ al
14. Cooke KR, Luznik L, Sarantopoulos S, et al. The biol­ogy of chronic graft-ver­ tri­als net­work report on the devel­op­ment of novel end­points and selec­
sus-host dis­ease: a task force report from the National Institutes of Health tion of prom­is­ing approaches for graft-ver­sus-host dis­ease pre­ven­tion tri­
con­sen­sus devel­op­ment pro­ject on cri­te­ria for clin­i­cal tri­als in chronic graft- als. Biol Blood Marrow Transplant. 2018;24(6):1274-1280.
ver­sus-host dis­ease. Biol Blood Marrow Transplant. 2017;23(2):211-234. 34. Bolanos-Meade J, Reshef R, Fraser RB, et  al. Novel approaches for graft-
15. Khoder A, Sarvaria A, Alsuliman A, et  al. Regulatory B cells are enriched ver­ sus-host dis­ ease (GVHD) pro­ phy­ laxis: pri­ mary results of prog­ ress I
within the IgM mem­ory and tran­si­tional sub­sets in healthy donors but are mul­ti­cen­ter trial of matched allo­ge­neic hema­to­poi­etic cell trans­plan­ta­
defi­cient in chronic GVHD. Blood. 2014;124(13):2034-2045. tion (alloHCT) using reduced inten­sity con­di­tion­ing (RIC) BMT CTN 1203.
16. Sarantopoulos S, Stevenson KE, Kim HT, et al. High lev­els of B-cell acti­ Abstract presented at: BMT Tandem Scientific Meeting; 25 Feb­ru­ary 2018;
vat­ing fac­tor in patients with active chronic graft-ver­sus-host dis­ease. Salt Lake City, UT. Late Breaking Abstracts. Accessed 30 August 2021.
Clin Cancer Res. 2007;13(20):6107-6114. https:​­/​­/bmt​­.confex​­.com​­/tandem​­/2018​­/meetingapp​­.cgi​­/Paper​­/11767.
17. Alexander KA, Flynn R, Lineburg KE, et al. CSF-1-depen­dent donor-derived 35. Matsuoka K, Koreth J, Kim HT, et al. Low-dose inter­leu­kin-2 ther­apy restores
mac­ro­phages medi­ate chronic graft-ver­sus-host dis­ease. J Clin Invest. reg­u­la­tory T cell homeo­sta­sis in patients with chronic graft-ver­sus-host dis­

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/648/1851934/648hamilton.pdf by guest on 13 December 2021


2014;124(10):4266-4280. ease. Sci Transl Med. 2013;5(179):179ra43.
18. Hamilton BK, Liu Y, Hemmer MT, et al. Inferior out­comes with cyclo­spor­ine 36. Zeiser R, Socié G. The devel­op­ment of ruxolitinib for glu­co­cor­ti­coid-refrac­
and mycophenolate mofetil after myeloablative allo­ge­neic hema­to­poi­etic tory acute graft-ver­sus-host dis­ease. Blood Adv. 2020;4(15):3789-3794.
cell trans­plan­ta­tion. Biol Blood Marrow Transplant. 2019;25(9):1744-1755. 37. Goodyear OC, Dennis M, Jilani NY, et al. Azacitidine aug­ments expan­sion of
19. Sorror ML, Leisenring W, Deeg HJ, Martin PJ, Storb R. Twenty-year fol­low- reg­u­la­tory T cells after allo­ge­neic stem cell trans­plan­ta­tion in patients with
up of a con­trolled trial com­par­ing a com­bi­na­tion of meth­o­trex­ate plus acute mye­loid leu­ke­mia (AML). Blood. 2012;119(14):3361-3369.
cyclo­spor­ine with cyclo­spor­ine alone for pro­phy­laxis of graft-ver­sus-host 38. Bruserud O, Tvedt TH, Paulsen PQ , et  al. Extracorporeal photopheresis
dis­ease in patients admin­is­tered HLA-iden­ti­cal mar­row grafts for leu­ke­mia. (photochemotherapy) in the treat­ment of acute and chronic graft ver­
Biol Blood Marrow Transplant. 2005;11(10):814-815. sus host dis­ease: immu­no­log­i­cal mech­a­nisms and the results from clin­i­cal
20. Jenkins MK, Schwartz RH, Pardoll DM. Effects of cyclo­spor­ine A on T cell stud­ies. Cancer Immunol Immunother. 2014;63(8):757-777.
devel­op­ment and clonal dele­tion. Science. 1988;241(4873):1655-1658. 39. Blanco B, Pérez-Simón JA, Sánchez-Abarca LI, et  al. Treatment with bor-
21. Zeiser R, Nguyen VH, Beilhack A, et al. Inhibition of CD4+CD25+ reg­ul­a­tory tezomib of human CD4+ T cells pre­serves nat­ur­al reg­u­la­tory T cells and
T-cell func­tion by calcineurin-depen­dent inter­leu­kin-2 pro­duc­tion. Blood. allows the emer­gence of a dis­tinct sup­pres­sor T-cell pop­u­la­tion. Haema­
2006;108(1):390-399. tologica. 2009;94(7):975-983.
22. Ferrara JL, Anasetti C, Stadtmauer E, et al. Blood and Marrow Transplant 40. Di Ianni M, Falzetti F, Carotti A, et  al. Tregs pre­vent GVHD and pro­mote
Clinical Trials Network State of the Science Symposium 2007. Biol Blood immune recon­sti­tu­tion in HLA-haploidentical trans­plan­ta­tion. Blood. 2011;
Marrow Transplant. 2007;13(11):1268-1285. 117(14):3921-3928.
23. Tamari R, Oran B, Hilden P, et al. Allogeneic stem cell trans­plan­ta­tion for 41. Kitko CL, Pidala J, Schoemans HM, et  al. National Institutes of Health
advanced myelodysplastic syn­drome: com­par­i­son of out­comes between con­sen­sus devel­op­ment pro­ject on cri­te­ria for clin­i­cal tri­als in chronic
CD34+ selected and unmod­i­fied hema­to­poi­etic stem cell trans­plan­ta­tion. graft-ver­sus-host dis­ease, IIa: the 2020 Clinical Implementation and Early
Biol Blood Marrow Transplant. 2018;24(5):1079-1087. Diagnosis Working Group report: NIH cGVHD WG2a. Transplant Cell Ther.
24. de Witte MA, Janssen A, Nijssen K, et  al. αβ T-cell graft deple­tion for 2021;27(7):545-557.
allo­ge­neic HSCT in adults with hema­to­log­i­cal malig­nan­cies. Blood Adv. 42. Pidala J, Kitko C, Lee SJ, et al. National Institutes of Health con­sen­sus devel­
2021;5(1):240-249. op­ment pro­ject on cri­te­ria for clin­i­cal tri­als in chronic graft-ver­sus-host dis­
25. Pasquini M, Luznik L, Logan B, et  al. Calcineurin inhib­i­tor-free graft-ver­ ease, IIb: the 2020 Preemptive Therapy Working Group report. Transplant
sus-host dis­ease (GVHD) pro­phy­laxis in hema­to­poi­etic cell trans­plan­ta­tion Cell Ther. 2021;27(8):632-641.
(HCT) with myeloablative con­di­tion­ing reg­i­mens (MAC) and HLA-matched 43. Wolff D, Schleuning M, von Harsdorf S, et al. Consensus con­fer­ence on
donors: results of the BMT CTN 1301 PROGRESS II trial [ab­stract]. T ­ ransplant clin­i­cal prac­tice in chronic GVHD: sec­ond-line treat­ment of chronic graft-
Cell Ther. 2021;27(3 suppl). TCT abstract LBA1. Accessed 30 August 2021. ver­sus-host dis­ease. Biol Blood Marrow Transplant. 2011;17(1):1-17.
https:​­/​­/www​­.hematologyandoncology​­.net​­/supplements​­/highlights​­-in​ 44. Carpenter PA, Kitko CL, Elad S, et  al. National Institutes of Health con­
­-graft​­-vs​­-host​­-disease​­-from​­-the​­-2021​­-transplantation​­-cellular​­-therapy​­-tct​ sen­sus devel­op­ment pro­ject on cri­te­ria for clin­i­cal tri­als in chronic graft-
­-meetings​­-of​­-the​­-astct​­-and​­-the​­-cibmtr​­/. ver­sus-host dis­ease, V: the 2014 Ancillary Therapy and Supportive Care
26. Kröger N, Solano C, Wolschke C, et al. Antilymphocyte glob­u­lin for pre­ven­ Working Group report. Biol Blood Marrow Transplant. 2015;21(7):1167-1187.
tion of chronic graft-ver­sus-host dis­ease. N Engl J Med. 2016;374(1):43-53. 45. Zeiser RP, Ram N, Hasmi R, et al. Ruxolitinib (RUX) vs best avail­­able ther­apy
27. Walker I, Panzarella T, Couban S, et al; Cana­ dian Blood and Marrow (BAT) in patients with ste­roid-refrac­tory/ste­roid-depen­dent chronic graft-
Trans​plant Group. Pretreatment with anti-thy­mo­cyte glob­u­lin ver­sus no vs-host dis­ease (cGVHD): pri­mary find­ings from the phase 3, ran­dom­ized
anti-thy­mo­cyte glob­u­lin in patients with haematological malig­nan­cies REACH3 study. Blood. 2020;136(suppl 1):22-24.
under­go­ing haemopoietic cell trans­plan­ta­tion from unre­lated donors: 46. Cutler C, Lee SJ, Arai S, et  al. Belumosudil for chronic graft-ver­sus-host
a randomised, con­trolled, open-label, phase 3, multicentre trial. Lancet dis­ease (cGVHD) after 2 or more prior lines of ther­apy: the Rockstar Study
Oncol. 2016;17(2):164-173. (KD025-213). Blood. 2020;136(suppl 1):45-46.
28. Soiffer RJ, Kim HT, McGuirk J, et  al. Prospective, ran­dom­ized, dou­ble- 47. DeFilipp Z, Onstad L, Arai S, et  al. Non-relapse mor­tal­ity among patients
blind, phase III clin­i­cal trial of anti-T-lym­pho­cyte glob­u­lin to assess impact diag­nosed with chronic graft-ver­sus-host dis­ease: an updated anal­y­sis from
on chronic graft-ver­sus-host dis­ease-free sur­vival in patients under­go­ing the chronic GVHD con­sor­tium. Transplant Cell Ther. 2021;27(3):S79-S80.
HLA-matched unre­lated myeloablative hema­to­poi­etic cell trans­plan­ta­ 48. Mielcarek M, Furlong T, O’Donnell PV, et al. Posttransplantation cyclo­phos­
tion. J Clin Oncol. 2017;35(36):4003-4011. pha­mide for pre­ven­tion of graft-ver­sus-host dis­ease after HLA-matched
29. Laport GG, Wu J, Logan B, et al; Blood and Marrow Transplant Clinical Trials mobi­lized blood cell trans­plan­ta­tion. Blood. 2016;127(11):1502-1508.
Network. Reduced-inten­sity con­di­tion­ing with fludarabine, cyclo­phos­pha­ 49. Bolaños-Meade J, Reshef R, Fraser R, et al. Three pro­phy­laxis ­reg­im ­ ens (tac-
mide, and high-dose rituximab for allo­ge­neic hema­to­poi­etic cell trans­plan­ rolimus, mycophenolate mofetil, and cyclo­phos­pha­mide; t­acrolimus,
ta­tion for fol­lic­u­lar lym­phoma: a phase two mul­ti­cen­ter trial from the Blood meth­o­trex­ate, and bortezomib; or tacrolimus, meth­o­trex­ate, and maraviroc)
and Marrow Transplant Clinical Trials Network. Biol Blood Marrow Trans­ ver­sus tacrolimus and meth­o­trex­ate for pre­ven­tion of graft-ver­sus-host dis­
plant. 2016;22(8):1440-1448. ease with haemopoietic cell trans­plan­ta­tion with reduced-inten­sity con­di­
30. Cutler C, Kim HT, Bindra B, et al. Rituximab pro­phy­laxis pre­vents cor­ti­co­ tion­ing: a randomised phase 2 trial with a non-randomised con­tem­po­ra­ne­ous
ste­roid-requir­ing chronic GVHD after allo­ge­neic periph­eral blood stem cell con­trol group (BMT CTN 1203). Lancet Haematol. 2019;6(3):e132-e143.
trans­plan­ta­tion: results of a phase 2 trial. Blood. 2013;122(8):1510-1517.
31. Luznik L, Jalla S, Engstrom LW, Iannone R, Fuchs EJ. Durable engraft­ment
of major his­to­com­pat­i­bil­ity com­plex-incom­pat­i­ble cells after nonmyeloab-
lative con­di­tion­ing with fludarabine, low-dose total body irra­di­a­tion, and © 2021 by The Amer­i­can Society of Hematology
posttransplantation cyclo­phos­pha­mide. Blood. 2001;98(12):3456-3464. DOI 10.1182/hema­tol­ogy.2021000301

654  |  Hematology 2021  |  ASH Education Program


UPDATE IN GRAFT- VERSUS - HOST DISEASE

What else do I need to worry about when treating


graft-versus-host disease?

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/655/1851925/655jawahri.pdf by guest on 13 December 2021


Areej El-Jawahri
Hematology­Oncology, Massachusetts General Hospital, Boston, MA

Graft-versus-host disease (GVHD) is the main cause of morbidity and mortality in allogeneic hematopoietic stem cell
transplant survivors. Patients with acute and chronic GVHD often endure substantial symptom burden and quality of
life (QOL) and functional impairments. Living with GVHD affects multiple domains of patient-reported QOL, physical
functioning, and psychological well-being. Patients describe living with GVHD as a life-altering “full-time job” requiring
unique knowledge, personal growth, and resilient coping strategies. Managing the supportive care needs of patients
living with GVHD must include (1) monitoring of patient-reported QOL and symptom burden; (2) routine screening for
psychological distress and implementing therapeutic strategies to treat depression, anxiety, and posttraumatic stress
symptoms; (3) a systematic review of care needs by a multidisciplinary team experienced in managing transplant-related
complications and organ-specific GVHD symptoms; and (4) ensuring optimal prevention and management of infection
complications in this highly immunocompromised population. Improving the QOL in patients with GVHD requires a mul-
tidisciplinary approach with emphasis on aggressive symptom management, psychological coping, and promoting phys-
ical activity and rehabilitation in this population living with immense prognostic uncertainty and struggling to adapt to
this difficult and unpredictable illness.

LEARNING OBJECTIVES
• Understand the impact of GVHD on quality of life, functional status, symptom burden, and psychological distress
in allogeneic HCT survivors
• Develop strategies to address the supportive care needs of patients with GVHD

The supportive care needs of patients


CLINICAL CASE with acute GVHD
Emma is a 39­year­old woman, a middle school teacher, Acute GVHD is a common complication of allogeneic
who was initially diagnosed with acute myeloid leukemia hematopoietic stem cell transplant (HCT), which has a sig­
and achieved a complete remission after induction che­ nificant impact on patient­reported quality of life (QOL),
motherapy. Subsequently, she underwent a myeloablative physical functioning, and clinical outcomes.1,2 Classic acute
matched unrelated donor peripheral stem cell transplan­ GVHD typically occurs within the first 100 days posttrans­
tation during a 4­week hospitalization. Two weeks after plant and predominantly affects the skin, liver, and gastro­
discharge, she developed diarrhea, prompting a hospital intestinal tract.1,2 Recent advances in the assessment and
readmission. She was diagnosed with acute skin and gas­ management of acute GVHD rely on identification and risk
trointestinal graft­versus­host disease (GVHD) and initiated stratification of patients based on clinical staging of the
first­line therapy with methylprednisolone 2mg/kg/d. She disease and blood biomarkers.3­5 Despite these advances,
had a complete response within 16 days of therapy and there are substantial gaps in our understanding of the sup­
was discharged from the hospital 3 weeks later. Due to portive care needs of this population, particularly when
prolonged corticosteroid exposure, Emma developed ste­ it comes to their QOL and overall functioning.6 Patients
roid myopathy and weakness, and she also struggled with with acute GVHD struggle with substantial symptom bur­
some lower extremity edema during the first few months den, including nausea, anorexia, diarrhea, and decline in
following her rehospitalization. functional status.6 As in Emma’s case, treatment of acute
GVHD typically involves high­dose corticosteroid therapy
Prakash Singh Shekhawat
Supportive care for GVHD | 655
along with other immu­no­sup­pres­sive agents, which may fur­ther and fecal microbiota trans­plan­ta­tion, are cur­rently being tested
increase the risk of com­pli­ca­tions and symp­toms such as infec­ in clin­i­cal tri­als.18
tions and weak­ness. In a ran­ dom­ ized clin­

cal trial, the inte­ gra­
tion of spe­ cialty
Studies com­pre­hen­sively exam­in­ing the sup­port­ive care pal­ li­
at­ive care has been shown to improve a wide range of
needs of patients with acute GVHD are lacking in part due to patient-reported out­comes, includ­ing QOL, symp­tom bur­den,
(1) the lack of a valid and reli­able tool spe­cific for symp­toms of fatigue, depres­sion, anx­i­ety, and post­trau­matic stress symp­toms
acute GVHD; (2) the need for fre­quent patient-reported assess­ both acutely dur­ing HCT hos­pi­tal­i­za­tion and up to 6 months
ments, which can place sub­stan­tial bur­den on this acutely ill post-HCT.19,20 Although these data are not spe­cific to patients
pop­u­la­tion; and (3) lack of robust stud­ies cor­re­lat­ing the objec­ with acute GVHD, admis­sion for acute GVHD is a rea­son­able trig­
tive response cri­te­ria with clin­i­cally mean­ing­ful changes in ger for spe­cialty pal­li­a­tive care given the role of pal­li­a­tive care in
QOL and acute GVHD symp­toms.6 Nonetheless, mul­ti­ple stud­ man­ag­ing com­plex symp­toms, facil­i­tat­ing effec­tive cop­ing, cul­
ies have shown that acute GVHD is asso­ci­ated with a decline ti­vat­ing prog­nos­tic aware­ness, and addressing the psy­cho­log­i­
in patient-reported QOL, phys­i­cal func­tion­ing, role func­tion­ing, cal and exis­ten­tial dis­tress in patients liv­ing with a seri­ous ill­ness.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/655/1851925/655jawahri.pdf by guest on 13 December 2021


social func­tion­ing, men­tal health, and gen­eral health.7-11 As high­ Although com­pre­hen­sive geri­at­ric assess­ments have not been
lighted in Emma’s clin­i­cal case, patients with acute GVHD often tested in patients with acute GVHD, they focus on numer­ous
spend prolonged peri­ods in the hos­pi­tal, which fur­ther con­trib­ domains that are affected by patients with acute GVHD. A com­
utes to their social iso­la­tion and psy­cho­log­i­cal dis­tress. Given pre­hen­sive geri­at­ric assess­ment model of care that incor­po­rates
the extent of trauma and iso­la­tion these patients expe­ri­ence dur­ a focus on func­tional capac­ity, cog­ni­tion, mood, polypharmacy,
ing their ill­ness course, stud­ies are also needed to exam­ine their social sup­port, finan­cial con­cerns, and nutri­tion, as well as over­
exis­ten­tial dis­tress and demor­al­iza­tion. all goals of care, may be a ben­e­fic ­ ial frame­work to address the
When car­ing for patients with acute GVHD, cli­ni­cians should needs of this pop­u­la­tion.
at min­i­mum screen for psy­cho­log­i­cal dis­tress. Prior work has
shown the fea­si­bil­ity of patient-reported out­comes mon­i­tor­ing
and psy­cho­so­cial dis­tress screen­ing in allo­ge­neic HCT recip­i­
ents.12,13 Studies inte­ grat­ing serial patient-reported out­ comes
CLINICAL CASE (Con­tin­ued)
mon­i­tor­ing dur­ing the acute GVHD course will help cli­ni­cians
guide clin­i­cal care in addressing the unmet needs of this pop­u­la­ Emma recov­ered from her acute GVHD symp­toms and con­tin­ued
tion, as well as mon­i­tor poten­tial response to ther­apy. In addi­tion, to make prog­ress toward her HCT recov­ery. At 8 months post-
clin­i­cal tri­als focused on the man­age­ment of acute GVHD must HCT, Emma devel­oped symp­toms of chronic GVHD affect­ing her
also incor­po­rate serial patient-reported out­come assess­ments skin, mouth, eyes, and vagina while taper­ing her immu­no­sup­pres­
that allow for robust exam­i­na­tion of response to ther­apy and sion ther­apy. Overall, she devel­oped mod­er­ate chronic GVHD
com­pli­ca­tions related to prolonged ste­roid use such as weak­ based on the num­ber of organ sys­tems involved. She received
ness, func­tional decline, fatigue, insom­nia, and psy­cho­log­i­cal treat­ment with sys­temic cor­ti­co­ste­roids, tacrolimus, ruxolitinib,
dis­tress. Moreover, max­i­mal sup­port­ive care mea­sures, includ­ing and ibrutinib. Given the sever­ity of her symp­toms, Emma has not
top­i­cal treat­ments for skin GVHD as well as anti­di­ar­rheal ther­apy been ­able to return to work. She also strug­gles to man­age her
for patients with gas­tro­in­tes­ti­nal GVHD, should be implemented daily rou­tine with top­i­cal ther­a­pies for her oral, ocu­lar, skin, and
rou­tinely in clin­i­cal prac­tice. Given the poten­tial risk of func­tional vag­i­nal GVHD.
decline, phys­i­cal ther­apy and reha­bil­i­ta­tion are also essen­tial
com­po­nents to max­i­mize the QOL and func­tion­ing of patients
with acute GVHD.14,15 Cumulative expo­sure to high-dose cor­ti­co­ QOL and symp­tom bur­den in chronic GVHD
ste­roids is asso­ci­ated with adverse side effects, includ­ing infec­ Chronic GVHD is a debil­ i­
tat­
ing immu­ no­logic syn­
drome that
tion com­pli­ca­tions, avas­cu­lar necro­sis, oste­o­po­ro­sis, edema, attacks mul­ti­ple organs and is the major cause of post-HCT mor­
and ste­roid myop­a­thy.16 In fact, the inci­dence of ste­roid myop­a­ bid­ity and mor­tal­ity.21-25 Patients with chronic GVHD strug­gle to
thy in patients with acute GVHD is as high as 41%.17 Studies have man­age their ill­ness, which often results in sub­stan­tial phys­i­cal
shown that patients with acute GVHD have base­ line impair­ symp­toms, func­tional lim­i­ta­tions, and impaired QOL.9,21,22,26,27
ments in their func­tion, which are wors­ened within 14 days of Treatment of chronic GVHD, such as cor­ti­co­ste­roids, is lim­ited
receiv­ing cor­ti­co­ste­roid ther­apy.14 These find­ings under­score the in effi­cacy and has sig­ nif­i­
cant toxicities, thus con­trib­
ut­
ing to
poten­tial need for early super­vised con­sul­ta­tion with phys­i­cal the unpre­dict­able tra­jec­tory of this ill­ness and its unre­lent­ing
ther­apy, occu­pa­tional ther­apy, and phys­i­cal med­i­cine and reha­ course.27 Studies have shown that the sever­ity of chronic GVHD
bil­i­ta­tion to closely mon­i­tor and inter­vene to improve phys­i­cal is asso­ci­ated with the extent of QOL, phys­i­cal, and func­tional
func­tion­ing in this pop­u­la­tion.14 As in Emma’s case, patients can impair­ments seen in this pop­u­la­tion.28 The Lee Symptom Scale
also develop some lower extrem­ity edema and fluid reten­tion is a reli­able and valid mea­sure for chronic GVHD symp­toms that
due to cor­ti­co­ste­roid use, poor nutri­tion and hypoalbuminemia, cor­re­lates with National Institutes of Health cri­te­ria as well as
and their inflam­ma­tory state from acute GVHD. Prompt man­age­ patient-reported chronic GVHD sever­ity.29
ment of these com­pli­ca­tions and close atten­tion to nutri­tional Given the global and mul­ ti­
di­
men­
sional impact of chronic
needs in this pop­u­la­tion can improve patients’ over­all QOL and GVHD in allo­ge­neic HCT sur­vi­vors, patients with chronic GVHD
func­tion­ing. Notably, recent stud­ies have shown an asso­ci­a­tion should be reviewed by a team expe­ri­enced in man­ag­ing trans­
between the gut microbiota and the risk of devel­op­ing acute plant-related com­pli­ca­tions and the sup­port­ive care needs of
GVHD as well as over­all trans­plant out­comes.18 Future ther­a­pies this pop­u­la­tion.30 Furthermore, trans­plant cen­ters should estab­
for acute GVHD, includ­ing probiotics, nutri­tional sup­ple­ments, lish a clin­i­cal net­work of spe­cial­ists with an inter­est in chronic

656  |  Hematology 2021  |  ASH Education Program


GVHD to allow for mul­ti­dis­ci­plin­ary man­age­ment.30 Assessment also be a cause of psy­cho­log­i­cal dis­tress in patients liv­ing with
of QOL and chronic GVHD symp­toms is recommended for all­ chronic GVHD and deal­ing with an unre­lent­ing ill­ness. Although
patients liv­ing with chronic GVHD.28 Improving QOL in this pop­ stud­ies have focused on finan­ cial dis­
tress in allo­
ge­
neic HCT
u­la­tion requires a mul­ti­dis­ci­plin­ary approach with a focus on recip­i­ents,40 future assess­ment of the extent of finan­cial dis­tress
aggres­sive symp­tom man­age­ment, psy­cho­log­i­cal cop­ing, and and tox­ic­ity is needed for patients with chronic GVHD along with
pro­mot­ing phys­i­cal activ­ity and reha­bil­i­ta­tion.30 con­sid­er­ation for finan­cial nav­i­ga­tion pro­grams and other evi­
dence-based inter­ven­tions to reduce finan­cial tox­ic­ity and psy­
Psychological dis­tress in patients with chronic GVHD cho­log­i­cal dis­tress in this pop­u­la­tion.
Given the phys­ i­
cal symp­ tom bur­ den and uncer­ tainty about
the ill­ness course in patients with chronic GVHD, psy­cho­log­i­ Organ-spe­cific sup­port­ive care man­age­ment
cal dis­tress is prev­a­lent in this pop­u­la­tion, with 50% and 30% of for chronic GVHD
patients with chronic GVHD reporting depres­sion and anx­i­ety Management of organ-spe­ cific issues is a core com­ po­
nent
symp­toms, respec­tively.31,32 Notably, self-reported depres­ sion of opti­ mal sup­ port­ ive GVHD care that entails top­ i­
cal ther­

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/655/1851925/655jawahri.pdf by guest on 13 December 2021


symp­toms have been asso­ci­ated with lower sur­vival in patients apy and other inter­ven­tions directed at organ spe­cific–con­trol
with chronic GVHD.28 In prior stud­ies, the use of adap­tive cop­ of symp­toms.30,41 The National Institutes of Health Consensus
ing strat­e­gies was asso­ci­ated with lower psy­cho­log­i­cal dis­tress Development Project and the Brit­ish Society for Bone Marrow
in patients with chronic GVHD.29 Coping style is a mod­i­fi­able Transplantation have both insti­tuted rec­om­men­da­tions for the
con­struct that, if altered to be more adap­tive in the con­text of sup­port­ive care and man­age­ment of organ-spe­cific com­pli­ca­
a chronic med­i­cal stressor, could poten­tially change the tra­jec­ tions of chronic GVHD.42 We review the spe­cific ther­a­pies for
tory of QOL and psy­cho­log­i­cal dis­tress in patients with chronic the most com­mon chronic GVHD symp­toms, includ­ing skin, oral,
GVHD.32 Therefore, psy­cho­log­i­cal inter­ven­tions, such as cog­ ocu­lar, and gen­i­tal man­i­fes­ta­tions of chronic GVHD.
ni­tive behav­ioral ther­apy, to pro­mote adap­ta­tion and enhance
effec­tive cop­ing have a prom­is­ing poten­tial for improv­ing QOL Skin chronic GVHD
and mood in this pop­u­la­tion.33-39 Supportive care for skin chronic GVHD focuses on pre­ven­tion
Similar to acute GVHD, screen­ing for psy­cho­so­cial dis­tress and man­age­ment of chronic GVHD symp­toms, includ­ing pruritus,
and prompt refer­ral to sup­port­ive care ser­vices such as psy­chol­ rash, pain, dyspigmentation, lim­ited range of motion, ero­sions,
ogy, psy­chi­a­try, and/or social work for pro­ac­tive man­age­ment ulcer­a­tions, and super­in­fec­tions (Table 1).
of depres­sion and anx­i­ety are crit­i­cal to opti­miz­ing the qual­ity Sun pro­tec­tion is impor­tant to reduce the risk of ultra­vi­o­let
of care for patients with chronic GVHD.31,32 In addi­tion to ther­apy radi­a­tion caus­ing an exac­er­ba­tion in skin chronic GVHD symp­
with a licensed clin­i­cal social worker or psy­chol­o­gist, patients toms as well as risk of sec­ond­ary skin can­cers.43 Photopro­
may ben­e­fit from a phar­ma­co­logic approach for mood man­age­ tection mea­sures include sun avoid­ance, pro­tec­tive cloth­ing,
ment with anti­de­pres­sants or anxi­o­lyt­ics. Financial dis­tress can phys­i­cal sun blocks, and high-potency sun­screens that pro­

Table 1. Supportive care rec­om­men­da­tions for chronic GVHD of the skin

Preventative mea­sures
  Photoprotection (UVA and UVB block­ade)
  Avoidance of sun expo­sure
  Use of sun­screens (≥SPF 20 with broad-spec­trum UVA and UVB pro­tec­tion)
Treatment
  Intact skin
     Symptomatic treat­ments with emol­lients and anti­pru­ritic agents
     Topical cor­ti­co­ste­roids
     Light ther­apy (PUVA, UVAI, UVB, nar­row­band UVB)
     Topical calcineurin inhib­i­tors (pimecrolimus, tacrolimus)
  Erosion and ulcer­a­tions
     Wound dress­ings and debride­ment
    Control of edema
  Sclerotic man­i­fes­ta­tion with joint stiff­ness or con­trac­tures
     Deep mus­cle/fas­cial mas­sage to improve range of motion
     Referral to phys­i­cal ther­apy, occu­pa­tional ther­apy, or phys­i­cal med­i­cine and reha­bil­i­ta­tion
     Daily stretching exer­cises to improve range of motion
     Strengthening, iso­tonic, iso­met­ric, and isokinetic exer­cises
  Other
     Dyspigmentation: trial of depigmenting creams containing hydro­qui­none, top­i­cal tre­tin­oin, or cor­ti­co­ste­roids
     Hair loss: dermovate scalp lotion once or twice daily
     Xerosis: reg­u­lar mois­tur­iz­ers
     Pruritus: tepid water rather than hot water for bath­ing, top­i­cal cor­ti­co­ste­roids, oral anti­his­ta­mines, doxepin, or gabapentin
PUVA, psoralen + ultraviolet light; SPF, sun pro­tec­tion fac­tor; UVA, ultraviolet light therapy.
Prakash Singh Shekhawat
Supportive care for GVHD  |  657
tect against both ultra­vi­o­let A (UVA) and ultra­vi­o­let B (UVB) Oral GVHD
radi­a­tion. Routine lubri­ca­tion of dry skin with emol­lients may Oral GVHD involv­ing the mouth and oral mucosa has 3 com­
decrease pruritus and enhance skin integ­rity.42 Topical ste­roids po­nents: muco­sal involve­ment, sal­i­vary gland involve­ment,
and emol­lients can be used to treat nonsclerotic skin lesions and scle­rotic involve­ment of the mouth and surrounding tis­
with­out ero­sions or ulcer­a­tions. Mid-strength top­i­cal ste­roids sues.30,41,42 Oral chronic GVHD can result in dry­ness, pain, ody­
(eg, tri­am­cin­o­lone 0.1% cream or ointment) can be used to nophagia, and taste impair­ment. New oral lesions should also
treat skin areas from the neck down. Higher-potency top­i­cal be eval­u­ated for sec­ond­ary can­cers given that they are more
ste­roids such as clobetasol 0.5% can be used in skin areas that com­mon in allo­ge­neic HCT recip­i­ents. Table 2 sum­ma­rizes man­
are refrac­tory to mid-strength top­i­cal treat­ment. In unre­spon­ age­ment strat­e­gies for oral chronic GVHD.30,41,42 Treatment of
sive cases, short-term occlu­sion of mid-strength ste­roids with oral cav­ity chronic GHVD is mainly focused on using cor­ti­co­ste­
damp tow­els (“wet wraps”) increases skin hydra­tion and ste­ roid rinses to reduce inflam­ma­tion in the oral cav­ity. Dexameth­
roid pen­e­tra­tion.30,41,42 Lower-potency top­i­cal cor­ti­co­ste­roids asone or other cor­ti­co­ste­roid rinse for­mu­la­tions are held and
such as top­ic ­ al hydro­cor­ti­sone 1.0% to 2.0% are pre­ferred swished in the mouth for approx­i­ma­tely 5 min­utes as often as

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/655/1851925/655jawahri.pdf by guest on 13 December 2021


for long-term use on the face, axil­lae, and groin. Other adju­ 4 to 6 times per day. For patients with severe symp­toms, top­i­
vant treat­ments that can be par­tic­u­larly help­ful for pruritus cal anal­ge­sia with vis­cous lido­caine can also be help­ful. Tacro­
related to GVHD include top­i­cal hydro­cor­ti­sone/pramoxine or limus rinses are also an alter­na­tive that can be effec­tive.30,41,42
menthol-based cream/lotions, as well as sys­temic anti­his­ta­ Tacrolimus ointment is espe­cially use­ful for treat­ment of lip
mines or the tri­cy­clic agent doxepin.30,41,42 Other inter­ven­tions GVHD involve­ment. Intralesional injec­tions with tri­am­cin­o­lone
such as top­i­cal calcineurin inhib­i­tors have been reported to can also be help­ful for dis­crete oral lesions that fail to respond
improve ery­thema and pru­ri­tus. For patients with ulcerated to top­i­cal ther­apy.
skin lesions, wound dress­ings main­tain a moist envi­ron­ment Patients with chronic GVHD affect­ing the sal­i­vary gland often
that enhances repair of the epi­the­lium. Protective films can report dry mouth and sensitivities to hot, cold, spicy, and acidic
also be use­ ful to pre­ vent break­ down of com­ pro­mised but foods. Patients may also develop mucoceles, which often pres­
nonulcerated skins. Topical anti­ mi­
cro­
bi­als may also have ent as pain­less blis­ters on the pal­ate.30,41,42 Supportive care mea­
some util­ity to pre­vent recur­rent infec­tions. For patients with sures for dry mouth include fre­quent liq­uid intake, use of sal­i­vary
major wounds related to skin GVHD, mul­ti­dis­ci­plin­ary exper­ stim­u­lants such as sugar-free gum, oral mois­tur­iz­ing agents, and
tise from der­ ma­ tol­
ogy, plas­tic sur­gery, and wound care is saliva sub­sti­tutes. Mouth dry­ness can increase the risk of car­ies
often needed. and tooth decay, which can be prevented with top­i­cal fluo­ride
Patients with scle­ro­sis affect­ing the skin and sub­cu­ta­ne­ous use. In patients with severe symp­toms, treat­ment with cho­lin­
tis­sue, includ­ing fas­cia, joints, and the mus­cu­lo­skel­e­tal sys­tem, er­gic ago­nists such as cevimeline or pilo­car­pine may enhance
strug­gle with sub­stan­tial func­tional impair­ments depending on sal­i­vary secre­tions.30,41,42
the sever­ity of the scle­ro­sis.30,41,42 Sclerotic fea­tures are one of
the most fre­quent and most dif­fi­cult man­i­fes­ta­tions of chronic Ocular GVHD
GVHD. Often patients with scle­ rotic skin GVHD require pro­ Ocular GVHD may pres­ent with acute con­junc­ti­val inflam­ma­tion,
longed treat­ment with sys­temic ther­a­pies, includ­ing cor­ti­co­ste­ pseudomembranous con­junc­ti­vi­tis, or ker­a­to­con­junc­ti­vi­tis sicca
roids, which fur­ther causes mus­cu­lar atro­phy, osteopenia, and syn­drome (or dry eye syn­drome). Keratoconjunctivitis sicca syn­
func­tional lim­i­ta­tions. Table 1 high­lights sup­port­ive care inter­ drome is the most com­mon pre­sen­ta­tion of chronic ocu­lar GVHD
ven­tions to address the needs of patients with scle­rotic chronic and is diag­nosed by the pres­ence of symp­toms, tear pro­duc­tion
GVHD of the skin.30,41,42 aver­ag­ing ≤5 mm (Schirmer’s test), and clin­i­cal signs of ker­a­ti­
tis.30,41,42 Most treat­ments of ocu­lar GVHD are aimed at reliev­ing
dry eye symp­toms, includ­ing burn­ing, irri­ta­tion, pain, for­eign
body sen­sa­tion, blurred vision, pho­to­pho­bia, and exces­sive tear­
Table 2. Supportive care rec­om­men­da­tions for oral chronic ing. Table 3 sum­ma­rizes the rec­om­men­da­tions for sup­port­ive
GVHD care man­age­ment of ocu­lar GVHD.
Preservative-free arti­fi­cial tears are often used in patients
Mild to mod­er­ate muco­sal dis­ease with chronic GVHD to increase lubri­ca­tion and reduce ocu­lar
  Localized appli­ca­tion of high-potency top­i­cal cor­ti­co­ste­roids symp­toms.30,41,42 Oral med­i­ca­tions can also be used to increase
  Generalized appli­ca­tion of upper mid-strength top­i­cal cor­ti­co­ste­roid lubri­ca­tion by stim­u­lat­ing aque­ous tear flow with selec­tive
  Topical anal­ge­sics mus­ca­rinic ago­nists such as cevimeline or pilo­car­pine. Man­
Moderate to severe muco­sal dis­ease agement of ocu­lar GVHD also depends on decreas­ing evap­
  Localized appli­ca­tion of upper mid-strength top­i­cal cor­ti­co­ste­roids o­ra­tion.30,41,42 Warm com­ pres­sors and lid care can max­ i­
mize
  Topical appli­ca­tion of tacrolimus 0.1% ointment meibomian gland out­put that pro­duces the oil layer of the tear
  Intralesional ther­apy with high-potency ste­roids for refrac­tory lesions film, which pro­tects against evap­o­ra­tion. Certain pro­tec­tive
  Topical appli­ca­tion of cyclo­spor­ine rinses eye wear such as mois­ture cham­ber googles may also help
  Oral phototherapy
decrease evap­o­ra­tion.30,41,42 Doxycycline is often pre­scribed for
Salivary gland dis­ease patients with ocu­lar GVHD to treat rosacea blepharitis, thereby
  Home fluo­ride ther­apy reduc­ing the inflam­ma­tion of the lid. Severe cases may also
  Frequent water sip­ping and saliva sub­sti­tutes ben­e­fit from scleral lenses, but these are avail­­able only in a few
  Salivary stim­u­lants (sugar-free gum, sugar-free candy) spe­cial­ized cen­ters. To decrease drain­age from the ocu­lar sur­
  Sialogogues: cevimeline, pilo­car­pine
faces, tem­po­rary and per­ma­nent occlu­sions of the tear ducts

658  |  Hematology 2021  |  ASH Education Program


Table 3. Supportive care rec­om­men­da­tions for ocu­lar chronic sur­face inflam­ma­tion, but these are often avail­­able only in spe­
GVHD cial­ized cen­ters.30,41,42

Topical and oral ther­a­pies Vulvar and vag­i­nal GVHD


  Artificial tears, pre­ser­va­tive free Vulvar and vag­i­nal chronic GVHD pres­ents with muco­sal abnor­
  Viscous ointment at bed­time/vis­cous tears dur­ing the day mal­i­ties that can sub­se­quently evolve into scle­rotic changes.30,41,42
  Cyclosporine and top­i­cal ste­roid eye drops Patients with vulvovaginal chronic GVHD pres­ent with symp­toms
  Oral agents such as cevimeline and pilo­car­pine of vag­i­nal dry­ness, dyspareunia, and dys­uria. If left untreated, vul­
  Doxycycline to reduce inflam­ma­tion
vovaginal chronic GVHD can result in scle­ro­sis of the vul­var and
Surgical vag­i­nal tis­sues, resulting in changes such as agglu­ti­na­tion of the
  Punctal occlu­sion (tem­po­rary using sil­i­cone plugs) cli­to­ral hood, narrowing of the introitus, and short­en­ing of the
  Permanent occlu­sion using ther­mal cau­tery vag­i­nal canal. Table 4 sum­ma­rizes the sup­port­ive care and man­
  Superficial debride­ment of fil­a­men­tary ker­a­ti­tis age­ment strat­e­gies for patients with vulvovaginal GVHD.30,41,42
  Partial tarsorrhaphy

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/655/1851925/655jawahri.pdf by guest on 13 December 2021


Itching and irri­ta­tion can be relieved by the appli­ca­tion of
Eye wear/envi­ron­men­tal strat­egy emol­lients to the exter­nal gen­i­ta­lia. Water-based lubri­cants
  Occlusive eye wear (mois­ture cham­ber gog­gles) can also be used in the vagina to reduce itching and irri­ta­tion.
  Lid care/warm com­pres­sors/humid­i­fied envi­ron­ment Patients with vulvovaginal GVHD often also have low estra­diol
  Bandage con­tact lens lev­els and vag­i­nal atro­phy.30,41,42 Topical estro­gen ther­apy with
Treatments not widely avail­­able or with­out the use of vag­i­nal dila­tors can be extremely effec­tive
  Autologous serum eye drops in the absence of abso­lute con­tra­in­di­ca­tions. For treat­ment of
  Gas-per­me­able con­tact lens (scleral lens pros­the­sis) vulvovaginal GVHD symp­toms, top­i­cal treat­ment with ultra-high-
potency cor­ti­co­ste­roid treat­ment rep­re­sents the main ther­apy,
Table 4. Supportive care rec­om­men­da­tions for vul­var although top­i­cal calcineurin ointments can be used in this pop­
and vag­i­nal chronic GVHD u­la­tion as well.30,41,42

Vulvar dis­com­fort
Late effects and infec­tion pro­phy­laxis and vac­ci­na­tions
for patients with chronic GVHD
  Avoid mechan­i­cal and chemical irri­tants
Patients with chronic GVHD are at high risk of late effects, includ­­
  Cleanse gen­i­tal area with warm water, allow air cir­cu­la­tion, and
wipe front to back ing skel­e­tal com­pli­ca­tions, sec­ond­ary can­cers, car­dio­vas­cu­lar
  Sparing use of sim­ple emol­lients to vulva dis­ease, and throm­bo­em­bolic events.44,45 Due to both chronic
  Water-based lubri­cants GVHD and its treat­ ment, patients often develop met­ a­
bolic
Vulvovaginal symp­toms due to low estro­gen sta­tus com­pli­ca­tions, includ­ing hyper­ten­sion, hyper­lip­id­emia, dia­be­
tes, and met­a­bolic syn­drome.44,45 These, in turn, increase the
  Topical estro­gen with or with­out dila­tor ther­apy
risk of car­dio­vas­cu­lar events. Careful atten­tion to car­dio­vas­cu­lar
Topical ther­apy for vulvovaginal GVHD risk fac­tors and these com­pli­ca­tions is nec­es­sary in pro­vid­ing
  High- and ultra-high-potency cor­ti­co­ste­roids high-qual­ity sur­vi­vor­ship care for this pop­u­la­tion.44,45 Patients
  ○ Clobetasol gel 0.05% (vagina) with chronic GVHD are highly immu­no­com­pro­mised with def­
  ○ Betamethasone dipropionate augmented gel (vagina) or ointment
i­cits in mac­ro­phage func­tion, immu­no­glob­u­lin pro­duc­tion, and
(vulva)
  ○ Tacrolimus ointment 0.1% (vulva) T-cell func­tion.30,41,42 A review of infec­tion pro­phy­laxis and vac­
ci­na­tion strat­e­gies for patients with chronic GVHD is beyond
Surgical ther­apy
the scope of this chap­ter, but these issues are well outlined in
  Surgery for stric­tures com­pre­hen­sive guide­lines for the pre­ven­tion of oppor­tu­nis­
tic infec­tions fol­low­ing HCT published by the Centers for Dis­
may help patients with mod­er­ate to severe ocu­lar dis­ease.30,41,42 ease Control and Prevention, the Infectious Diseases Society
In addi­tion to these mea­ sures, patients with ocu­ lar sur­face of America, and the Amer­i­can Society for Blood and Marrow
inflam­ma­tion may ben­e­fit from the judi­cious use of top­i­cal ste­ Transplantation.46 Table 5 out­lines strat­e­gies for mon­i­tor­ing and
roid ther­apy to reduce inflam­ma­tion. Topical cyclo­spor­ine is man­age­ment of other chronic GVHD com­pli­ca­tions, includ­ing
also com­monly used. Autologous tears can also reduce ocu­lar pul­mo­nary GVHD.

Table 5. Supportive care rec­om­men­da­tions for pul­mo­nary chronic GVHD

   Pulmonary func­tion test and high-res­o­lu­tion expi­ra­tory phase chest com­put­er­ized tomog­ra­phy to assess for chronic GVHD of the lung
   Routine mon­i­tor­ing of pul­mo­nary func­tion test in high-risk pop­u­la­tion is warranted
   Systemic ther­apy with cor­ti­co­ste­roid and other chronic GVHD agents
   Macrolides, inhaled ste­roids, and leu­ko­tri­ene inhib­i­tors are help­ful as an adjunc­tive ther­apy (ie, FAM ther­apy)
   Consideration of use of intra­ve­nous immu­no­glob­u­lins, par­tic­u­larly those with low IgG lev­els
   Adequate vac­ci­na­tions against pneu­mo­coc­cus and sea­sonal influ­enza
   Referral to pulmonology and con­sid­er­ation for pul­mo­nary reha­bil­i­ta­tion
FAM, fluticasone, azithromycin, and montelukast.
Prakash Singh Shekhawat
Supportive care for GVHD  |  659
10. Chiodi S, Spinelli S, Ravera G, et al. Quality of life in 244 recip­i­ents of allo­
CLINICAL CASE (Con­t in­u ed) ge­neic bone mar­row trans­plan­ta­tion. Br J Haematol. 2000;110(3):614-619.
11. Worel N, Biener D, Kalhs P, et  al. Long-term out­come and qual­ity of life
Despite her grat­i­tude that she is still alive and ­able to spend of patients who are alive and in com­plete remis­sion more than two years
qual­ity time with her chil­dren, Emma often mourns the immense after allo­ge­neic and syn­ge­neic stem cell trans­plan­ta­tion. Bone Marrow
losses she has expe­ri­enced as a result of her ill­ness and its impact Transplant. 2002;30(9):619-626.
on her day-to-day life. She also strug­gles with the uncer­tainty 12. Shaw BE, Brazauskas R, Millard HR, et al. Centralized patient-reported out­
come data col­lec­tion in trans­plan­ta­tion is fea­si­ble and clin­i­cally mean­ing­
regard­ing her future health and prog­no­sis. She is grate­ful for her
ful. Cancer. 2017;123(23):4687-4700.
trans­plant cli­ni­cians who often val­i­date her emo­tional strug­gles, 13. Lee SJ, Loberiza FR, Antin JH, et  al. Routine screen­ing for psy­cho­so­cial
allow her the space to dis­cuss her emo­tional jour­ney with this ill­ dis­tress fol­low­ing hema­to­poi­etic stem cell trans­plan­ta­tion. Bone Marrow
ness, and pro­vide sup­port­ive resources, includ­ing psy­cho­so­cial Transplant. 2005;35(1):77-83.
coun­sel­ing to pro­cess this dif­fi­cult ill­ness. 14. Ngo-Huang A, Yadav R, Bansal S, et al. An explor­atory study on phys­i­cal func­
tion in stem cell trans­plant patients under­go­ing cor­ti­co­ste­roid treat­ment
for acute graft-ver­sus-host-dis­ease. Am J Phys Med Rehabil. 2021;100(4):
402-406.

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/655/1851925/655jawahri.pdf by guest on 13 December 2021


15. Hamada R, Kondo T, Murao M, et al. Effect of the sever­ity of acute graft-ver­
Summary sus-host dis­ease on phys­i­cal func­tion after allo­ge­neic hema­to­poi­etic stem
Living with GVHD affects mul­ti­ple domains of patient-reported cell trans­plan­ta­tion. Support Care Cancer. 2020;28(7):3189-3196.
QOL, phys­i­cal func­tion­ing, and psy­cho­log­i­cal well-being. Patients 16. Schäcke H, Döcke WD, Asadullah K. Mechanisms involved in the side
describe liv­ing with GVHD as a life-alter­ing “full-time job” requir­ effects of glu­co­cor­ti­coids. Pharmacol Ther. 2002;96(1):23-43.
17. Lee HJ, Oran B, Saliba RM, et al. Steroid myop­a­thy in patients with acute
ing unique knowl­edge, per­sonal growth, and resil­ient cop­ing
graft-ver­sus-host dis­ease treated with high-dose ste­roid ther­apy. Bone
strat­eg
­ ies. Management of GVHD must include opti­mal sup­port­ Marrow Transplant. 2006;38(4):299-303.
ive care mea­sures to address GVHD symp­toms, pro­mote effec­ 18. Yoshioka K, Kakihana K, Doki N, Ohashi K. Gut microbiota and acute
tive cop­ing, and reduce psy­cho­log­i­cal and exis­ten­tial dis­tress graft-ver­sus-host dis­ease. Pharmacol Res. 2017;122:90-95.
19. El-Jawahri A, LeBlanc T, VanDusen H, et al. Effect of inpa­tient pal­li­a­tive care
in a pop­u­la­tion liv­ing with immense prog­nos­tic uncer­tainty and
on qual­ity of life 2 weeks after hema­to­poi­etic stem cell trans­plan­ta­tion: a
strug­gling to adapt to a dif­fi­cult and unpre­dict­able ill­ness. ran­dom­ized clin­i­cal trial. JAMA. 2016;316(20):2094-2103.
20. El-Jawahri A, Traeger L, Greer JA, et al. Effect of inpa­tient pal­li­a­tive care dur­
ing hema­to­poi­etic stem-cell trans­plant on psy­cho­log­i­cal dis­tress 6 months
Conflict-of-inter­est dis­clo­sure after trans­plant: results of a ran­dom­ized clin­i­cal trial. J Clin Oncol. 2017;
Areej El-Jawahri: no com­pet­ing finan­cial inter­ests to declare. 35(32):3714-3721.
21. Duell T, van Lint MT, Ljungman P, et al. Health and func­tional sta­tus of long-
term sur­vi­vors of bone mar­row trans­plan­ta­tion. EBMT Working Party on
Off-label drug use Late Effects and EULEP Study Group on Late Effects. Euro­pean Group for
Areej El-Jawahri: nothing to disclose. Blood and Marrow Transplantation. Ann Intern Med. 1997;126(3):184-192.
22. Socié G, Stone JV, Wingard JR, et al. Long-term sur­vival and late deaths
after allo­ge­neic bone mar­row trans­plan­ta­tion. Late Effects Working Com­
Correspondence mittee of the International Bone Marrow Transplant Registry. N Engl J Med.
Areej El-Jawahri, Hematology-Oncology, Mas­sa­chu­setts General 1999;341(1):14-21.
Hospital, 55 Fruit St, Yawkey 9E, Bos­ton, MA 02114; e-mail:ael- 23. Sullivan KM, Witherspoon RP, Storb R, et al. Prednisone and aza­thi­op ­ rine
jawahri@part­ners​­.org. com­pared with pred­ni­sone and pla­cebo for treat­ment of chronic graft-v-
host dis­ease: prog­nos­tic influ­ence of prolonged throm­bo­cy­to­pe­nia after
allo­ge­neic mar­row trans­plan­ta­tion. Blood. 1988;72(2):546-554.
References 24. Wingard JR, Piantadosi S, Vogelsang GB, et  al. Predictors of death from
1. Holtan SG, Pasquini M, Weisdorf DJ. Acute graft-ver­sus-host dis­ease: a bench- chronic graft-ver­sus-host dis­ease after bone mar­row trans­plan­ta­tion.
to-bed­side update. Blood. 2014;124(3):363-373. Blood. 1989;74(4):1428-1435.
2. Choi S, Reddy P. Graft-ver­sus-host dis­ease. Panminerva Med. 2010;52(2): 25. Loughran TP Jr, Sullivan K, Morton T, et al. Value of day 100 screen­ing stud­
111-124. ies for predicting the devel­op­ment of chronic graft-ver­sus-host dis­ease
3. MacMillan ML, Robin M, Harris AC, et al. A refined risk score for acute graft- after allo­ge­neic bone mar­row trans­plan­ta­tion. Blood. 1990;76(1):228-234.
ver­sus-host dis­ease that pre­dicts response to ini­tial ther­apy, sur­vival, and 26. Sutherland HJ, Fyles GM, Adams G, et  al. Quality of life fol­low­ing bone
trans­plant-related mor­tal­ity. Biol Blood Marrow Transplant. 2015;21(4):761- mar­row trans­plan­ta­tion: a com­par­i­son of patient reports with pop­u­la­tion
767. norms. Bone Marrow Transplant. 1997;19(11):1129-1136.
4. Levine JE, Braun TM, Harris AC, et al. A prog­nos­tic score for acute graft- 27. Lee SJ, Flowers MED. Recognizing and man­ag­ing chronic graft-ver­sus-host
ver­sus-host dis­ ease based on bio­ mark­ ers: a multicentre study. Lancet dis­ease. Hematology. 2008;2008(1):134-141.
Haematol. 2015;2(1):e21-e29. 28. Pidala J, Kurland B, Chai X, et  al. Patient-reported qual­ity of life is asso­
5. Major-Monfried H, Renteria AS, Pawarode A, et al. MAGIC bio­mark­ers pre­ ci­ated with sever­ity of chronic graft-ver­sus-host dis­ease as mea­sured by
dict long-term out­comes for ste­roid-resis­tant acute GVHD. Blood. 2018; NIH cri­te­ria: report on base­line data from the Chronic GVHD Consortium.
131(25):2846-2855. Blood. 2011;117(17):4651-4657.
6. Patel SS, Lapin B, Majhail NS, Hamilton BK. Patient-reported out­comes in 29. Lee Sk, Cook EF, Soiffer R, Antin JH. Development and val­i­da­tion of a scale
acute graft-ver­sus-host dis­ease: opti­miz­ing patient care and clin­i­cal trial to mea­sure symp­toms of chronic graft-ver­sus-host dis­ease. Biol Blood Mar-
end­points. Bone Marrow Transplant. 2020;55(8):1533-1539. row Transplant. 2002;8(8):444-452.
7. Kurosawa S, Yamaguchi T, Mori T, et al. Patient-reported qual­ity of life after 30. Dignan FL, Scarisbrick JJ, Cor­nish J, et al; Haemato-oncol­ogy Task Force of
allo­ge­neic hema­to­poi­etic cell trans­plan­ta­tion or che­mo­ther­apy for acute Brit­ish Committee for Standards in Haematology; Brit­ish Society for Blood
leu­ke­mia. Bone Marrow Transplant. 2015;50(9):1241-1249. and Marrow Transplantation. Organ-spe­cific man­age­ment and sup­port­ive
8. Lee SJ, Kim HT, Ho VT, et al. Quality of life asso­ci­ated with acute and chronic care in chronic graft-ver­sus-host dis­ease. Br J Haematol. 2012;158(1):62-78.
graft-ver­sus-host dis­ease. Bone Marrow Transplant. 2006;38(4):305-310. 31. El-Jawahri A, Pidala J, Khera N, et al. Impact of psy­cho­log­i­cal dis­tress on
9. Syrjala KL, Chapko MK, Vitaliano PP, Cummings C, Sullivan KM. Recovery qual­ity of life, func­tional sta­tus, and sur­vival in patients with chronic graft-
after allo­ge­neic mar­row trans­plan­ta­tion: pro­spec­tive study of pre­dic­tors ver­sus-host dis­ease. Biol Blood Marrow Transplant. 2018;24(11):2285-2292.
of long-term phys­i­cal and psy­cho­so­cial func­tion­ing. Bone Marrow Trans- 32. Jacobs JM, Fishman S, Sommer R, et al. Coping and mod­i­fi­able psy­cho­so­
plant. 1993;11(4):319-327. cial fac­tors are asso­ci­ated with mood and qual­ity of life in patients with

660  |  Hematology 2021  |  ASH Education Program


chronic graft-ver­sus-host dis­ease. Biol Blood Marrow Transplant. 2019; 40. Abel GA, Albelda R, Khera N, et al. Financial hard­ship and patient-reported
25(11):2234-2242. out­comes after hema­to­poi­etic cell trans­plan­ta­tion. Biol Blood Marrow
33. Traeger L, Greer JA, Fernandez-Robles C, Temel JS, Pirl WF. Evidence-based Transplant. 2016;22(8):1504-1510.
treat­ment of anx­i­ety in patients with can­cer. J Clin Oncol. 2012;30(11):1197- 41. Couriel DR. Ancillary and sup­port­ive care in chronic graft-ver­sus-host dis­
1205. ease. Best Pract Res Clin Haematol. 2008;21(2):291-307.
34. Duncan M, Moschopoulou E, Herrington E, et al; SURECAN Investigators. 42. Carpenter PA, Kitko CL, Elad S, et  al. National Institutes of Health Con­
Review of sys­tem­atic reviews of non-phar­ma­co­log­i­cal inter­ven­tions to sensus Development Project on Criteria for Clinical Trials in Chronic Graft-
improve qual­ity of life in can­cer sur­vi­vors. BMJ Open. 2017;7(11):e015860. ver­sus-Host Disease: V. The 2014 ancil­lary ther­apy and sup­port­ive care
35. DuHamel KN, Mosher CE, Winkel G, et al. Randomized clin­i­cal trial of tele­ work­ing group report. Biol Blood Marrow Transplant. 2015;21(7):1167-1187.
phone-admin­is­tered cog­ni­tive-behav­ioral ther­apy to reduce post-trau­ 43. Kitajima T, Imamura S. Graft-ver­sus-host reac­tion enhanced by ultra­vi­o­let
matic stress dis­or­der and dis­tress symp­toms after hema­to­poi­etic stem-cell radi­a­tion. Arch Dermatol Res. 1993;285(8):499-501.
trans­plan­ta­tion. J Clin Oncol. 2010;28(23):3754-3761. 44. Carpenter PA. Late effects of chronic graft-ver­sus-host dis­ease. Best Pract
36. Stanton AL, Luecken LJ, MacKinnon DP, Thompson EH. Mechanisms in psy­ Res Clin Haematol. 2008;21(2):309-331.
cho­so­cial inter­ven­tions for adults liv­ing with can­cer: oppor­tu­nity for inte­ 45. Majhail NS. Long-term com­pli­ca­tions after hema­to­poi­etic cell trans­plan­ta­
gra­tion of the­ory, research, and prac­tice. J Consult Clin Psychol. 2013;81(2): tion. Hematol Oncol Stem Cell Ther. 2017;10(4):220-227.
318-335. 46. Centers for Disease Control and Prevention; Infectious Disease Society of

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/655/1851925/655jawahri.pdf by guest on 13 December 2021


37. Bower JE. Cancer-related fatigue—mech­ a­
nisms, risk fac­ tors, and treat­ America; Amer­i­can Society of Blood and Marrow Transplantation. Guide­
ments. Nat Rev Clin Oncol. 2014;11(10):597-609. lines for pre­vent­ing oppor­tu­nis­tic infec­tions among hema­to­poi­etic stem
38. Gudenkauf LM, Antoni MH, Stagl JM, et al. Brief cog­ni­tive-behav­ioral and cell trans­plant recip­i­ents. MMWR Recomm Rep. 2000;49(RR-10):1-125, CE1-7.
relax­a­tion train­ing inter­ven­tions for breast can­cer: a ran­dom­ized con­
trolled trial. J Consult Clin Psychol. 2015;83(4):677-688.
39. Duijts SF, van Beurden M, Oldenburg HS, et al. Efficacy of cog­ni­tive behav­
ioral ther­apy and phys­i­cal exer­cise in alle­vi­at­ing treat­ment-induced men­o­
pausal symp­toms in patients with breast can­cer: results of a ran­dom­ized, © 2021 by The Amer­i­can Society of Hematology
con­trolled, mul­ti­cen­ter trial. J Clin Oncol. 2012;30(33):4124-4133. DOI 10.1182/hema­tol­ogy.2021000302

Prakash Singh Shekhawat


Supportive care for GVHD  |  661
WHAT ’ S NEW IN PLASMA CELL DISORDERS ?

Advances in MGUS diagnosis, risk stratification,


and management: introducing myeloma-defining
genomic events

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/662/1851672/662landgren.pdf by guest on 13 December 2021


Ola Landgren
Myeloma Program, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL

In the 1960s, Dr Jan Waldenström argued that patients who had monoclonal proteins without any symptoms or evidence
of end-organ damage represented a benign monoclonal gammopathy. In 1978, Dr Robert Kyle introduced the concept of
“monoclonal gammopathy of undetermined significance” (MGUS) given that, at diagnosis, it was not possible with avail-
able methods (ie, serum protein electrophoresis to define the concentration of M-proteins and microscopy to determine
the plasma cell percentage in bone marrow aspirates) to determine which patients would ultimately progress to multiple
myeloma. The application of low-input whole-genome sequencing (WGS) technology has circumvented previous prob-
lems related to volume of clonal plasma cells and contamination by normal plasma cells and allowed for the interrogation
of the WGS landscape of MGUS. As discussed in this chapter, the distribution of genetic events reveals striking differences
and the existence of 2 biologically and clinically distinct entities of asymptomatic monoclonal gammopathies. Thus,
we already have genomic tools to identify “myeloma-defining genomic events,” and consequently, it is reasonable to
consider updating our preferred terminologies. When the clinical field is ready to move forward, we should be able to
consolidate current terminologies—from current 7 clinical categories: low-risk MGUS, intermediate-risk MGUS, high-risk
MGUS, low-risk smoldering myeloma, intermediate-risk smoldering myeloma, high-risk smoldering myeloma, and multi-
ple myeloma—to future 3 genomic-based categories: monoclonal gammopathy, early detection of multiple myeloma (in
which myeloma-defining genomic events already have been acquired), and multiple myeloma (patients who are already
progressing and clinically defined cases). Ongoing investigations will continue to advance the field.

LEARNING OBJECTIVES
• Review of current clinical risk scores for myeloma precursor conditions, which are limited indirect measures of
disease burden/activity
• Show how low-input WGS reveals striking differences and existence of 2 biologically and clinically distinct entities

CLINICAL CASE confrmed normal complete blood count and compre-


In the beginning of January 2016, a 46-year-old previously hensive metabolic panel, whereas a serum protein elec-
healthy male lawyer was applying for a new life insurance trophoresis (SPEP) revealed evidence of a monoclonal
policy. As part of the application process, the insurance band in the gamma region and immunofxation showed
company required a standard health questionnaire and immunoglobulin G (IgG) κ isotype. The concentration of
laboratory blood tests. The results from the blood work the monoclonal (M)–protein was determined to 0.9 g/dL.
showed a normal complete blood count and a normal Quantitative immunoglobulins showed normal IgG, immu-
comprehensive metabolic panel with the exception of an noglobulin A (IgA), and immunoglobulin M (IgM) levels.
elevated total protein of 8.9 g/dL (normal reference range, Also, serum free light chains (FLCs) were evaluated,
6.0–8.3 g/dL). The patient was referred to a hematologist, and they showed normal levels. Given the low concen-
who ordered repeat laboratory blood tests, including tration of the M-protein, the IgG isotype, the absence
additional serum immune assays, to further investigate of an abnormal FLC ratio, and normal IgA (150 mg/dL)
the elevated total protein levels. The repeated results or IgM (173 mg/dL) concentrations, per current clinical

662 | Hematology 2021 | ASH Education Program


­guide­lines,1 the ­hema­tol­o­gist recommended no bone mar­row rou­tine workup with a clin­i­cal exam and base­line echo­car­dio­
biopsy and no imag­ing. Instead, the patient was recommended gram and elec­tro­car­dio­gram.
to repeat the bloodwork in 6 months. The patient was given
the diag­no­sis of mono­clo­nal gammopathy of unde­ter­mined
sig­nif­i­cance (MGUS), and he was told that the life­time risk of Because most MGUS cases will never prog­ress, sta­tis­ti­cally
pro­gres­sion to mul­ti­ple mye­loma (MM) was very low. speak­ing, this patient case illus­trates a rel­a­tively uncom­mon sit­u­
When the patient returned for a fol­low-up visit in July 2016, all­ a­tion with pro­gres­sion to MM within a few years from ini­tial MGUS
the results were vir­tu­ally unchanged. Per cur­rent clin­i­cal guide­ diag­no­sis. However, most phy­si­cians who mon­i­tor large num­bers
lines,1 the hema­tol­o­gist now recommended fol­low-up annu­ally of patients with MGUS have seen these kinds of cases in their
with repeated bloodwork. When the patient returned in July clinic. This is reflec­tive of the fol­low­ing 2 facts: (1) most cases
2017, the IgG κ M-pro­tein had increased to 1.1 g/dL, whereas all­ with MGUS remain sta­ble over time, but at the same time, (2) all­
other results were sim­i­lar to the year before. The patient had MM cases are con­sis­tently pre­ceded by MGUS (but the major­ity
no symp­toms. In July 2018, he returned for a fol­low-up visit, and do not know they had MGUS prior to MM because no test­ing was

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/662/1851672/662landgren.pdf by guest on 13 December 2021


the bloodwork now revealed an IgG κ M-pro­tein of 1.4 g/dL. done).2 When it comes to cur­rent clin­i­cal risk scores to pre­dict
The FLC κ lev­els were slightly ele­vated at 5.3 mg/dL (nor­mal pro­gres­sion from MGUS to MM, it is impor­tant to empha­size that
ref­er­ence range, 0.33-1.94 mg/dL), FLC λ was 0.8 mg/dL (nor­ today’s risk scores only pro­vide the aver­age risk of pro­gres­sion
mal ref­er­ence range, 0.57-2.63 mg/dL), and the FLC ratio was for all­indi­vid­u­als with a given score. None of the avail­­able risk
6.63 (nor­mal ref­er­ence range, 0.26-1.65). Also, the IgM con­cen­ scores pro­vide the abso­lute risk of pro­gres­sion for an indi­vid­
tra­tion was decreased at 25 mg/dL (nor­mal ref­er­ence range, ual patient. Furthermore, cur­rent clin­i­cal risk scores are based
37-286 mg/dL), whereas IgA was within the nor­mal ref­er­ence on tumor bur­den, and they are unable to detect progressors
inter­val at 78 mg/dL (61-356 mg/dL). Given the grad­ual wors­ among MGUS cases with lower dis­ease bur­den. Indeed, cur­rent
en­ing of serum immune mark­ers, the hema­tol­o­gist discussed clin­i­cal risk scores show that indi­vid­u­als with higher dis­ease bur­
with the patient that he may choose to undergo a bone mar­ den (higher plasma cell per­cent­age and/or higher serum immune
row biopsy and imag­ing to rule out MM, but the patient asked marker con­cen­tra­tions) on aver­age have a higher prob­a­bil­ity of
his doc­tor if it was totally nec­es­sary since he felt com­pletely progressing com­pared with the aver­age indi­vid­ual with lower
healthy, ate healthy food, and exer­cised sev­eral days every dis­ease bur­den. Although lower dis­ease bur­den has lower risk of
week. After a lon­ger dis­cus­sion, the patient and the hema­tol­ pro­gres­sion, just like the above patient case, there are indi­vid­u­als
o­gist decided to hold off with addi­tional test­ing and con­tinue with lower dis­ease bur­den progressing to MM. Conversely, many
with annual lab­o­ra­tory results. indi­vid­u­als with higher dis­ease bur­den will never prog­ress.
In June 2019, about 3.5 years after ini­tial diag­no­sis of MGUS, There is an unmet clin­i­cal need for bet­ter assays allowing
the patient came for a visit to review his annual bloodwork. the phy­si­cian to deter­mine the indi­vid­ual patient’s risk of pro­
Now the SPEP showed an IgG κ M-pro­tein of 1.6 g/dL. The FLC κ gres­sion to MM. We do not yet have any such established assays
lev­els were again ele­vated now at 8.6 mg/dL (nor­mal ref­er­ence avail­­able in the clinic. The best tools we have today include lon­
range, 0.33-1.94 mg/dL), FLC λ was 0.5 mg/dL (nor­mal ref­er­ence gi­tu­di­nal mon­i­tor­ing and reassessments, as discussed in detail in
range, 0.57-2.63 mg/dL), and the FLC ratio was 17.2 (nor­mal ref­ this chap­ter. This review addresses cur­rent sta­tus of sci­ence and
er­ence range, 0.26-1.65). The IgM con­cen­tra­tion was fur­ther clin­i­cal man­age­ment, novel and emerg­ing tech­nol­o­gies, recent
decreased at 18 mg/dL (nor­mal ref­er­ence range, 37-286 mg/dL), dis­cov­er­ies, and future direc­tions.
and IgA was decreased at 40 mg/dL (61-356 mg/dL). Hemoglo-
bin was slightly decreased at 13.2 g/dL (nor­mal ref­er­ence range, Initial obser­va­tions and emer­gence of dif­fer­ent
13.5-17.5  g/dL), whereas all­ other lab­o­ra­tory results (­includ­ing schools of thought
cal­cium, cre­at­i­nine, albu­min, lac­tate ­dehy­dro­ge­nase, and β2- Early dis­cov­ery work focus­ing on abnor­mal serum pro­teins was
microglobulin) were nor­mal. The hema­tol­o­gist recommended pioneered by Dr Jan Waldenström and col­leagues. In 1944, he
the patient to undergo a bone mar­row biopsy and a pos­i­tron presented his paper “Incipient Myelomatosis or ‘Essential’ Hyper-
emis­sion tomog­ra­phy/com­puted tomog­ra­phy to rule out MM. globulinemia With Fibrinogenopenia—A New Syndrome?” in
Immunohistochemistry staining of the core biopsy spec­i­men which he described 3 patients with refrac­tory ane­mia and bleed­
showed 30% κ light chain–restricted plasma cells in the bone ing ten­dency whose sera exhibited a very high vis­cos­ity. Walden-
mar­row. Also, whole-body pos­i­t ron emis­sion tomog­ra­phy/ ström spec­u­lated that a spe­cial glob­u­lin frac­tion was the cause
com­puted tomog­ra­phy revealed a 1.1-cm diam­e­ter lytic lesion of the increased vis­cos­ity, and by ultra­cen­tri­fu­ga­tion stud­ies,
in ileum on the right side of the pel­vis with an standardized he was ­able to dem­on­strate that the sera of 2 of these patients
uptake value (SUV) of 6.7, a 1.2-cm diam­e­ter lytic lesion in the contained mac­ro­glob­u­lins (ie, plasma glob­u­lins of high molec­u­
left femur with a SUV of 8.2, and 2 small (<5 mm diam­e­ter) lar weight)—sub­se­quently referred to as “Waldenström’s mac­ro­
lytic lesions in the left fifth and sixth ribs with SUVs of 2.5 and glob­u­li­ne­mia.”3 Together with Dr Carl-Bertil Laurell—a world-class
3.1, respec­tively. Fluorescence in situ hybrid­iza­tion and sin­gle- clin­i­cal chem­ist—Waldenström started explor­ing con­di­tions with
nucle­o­tide poly­mor­phism array test­ing of the bone mar­row gammaglobulin derange­ments in a sys­tem­atic man­ner, as well as
aspi­rate did not cap­ture any high-risk char­ac­ter­is­tics. Ten- clin­i­cal cor­re­lates of mono­clo­nal and poly­clonal gammopathies.4
color flow cytom­e­try of the bone mar­row aspi­rate con­firmed Through their trans­la­tional research efforts, they delin­eated the
the immunophenotype of κ light chain–restricted plasma occur­rence and clin­i­cal sig­nif­i­cance of so-called mono­clo­nal
cells expressing CD56 and CD117 but neg­a­tive for CD20. The com­po­nents. Gradually, obser­va­tions from the lab­o­ra­tory cou­
patient was diag­ nosed with stan­ dard-risk MM and started pled with clin­i­cal data led to the emer­gence of 2 major schools
com­bi­na­tion ther­apy 2 weeks later, after he had under­gone of thought. In the 1960s, Waldenström pro­posed that there were
Prakash Singh Shekhawat
Myeloma pre­cur­sor con­di­tions  |  663
patients who had mono­clo­nal (M)–pro­teins with­out any symp­ less than 60%) plasma cells in the bone mar­row; then, based on
toms or evi­dence of end-organ dam­age, representing a benign cur­rent cri­te­ria, the diag­no­sis would be smol­der­ing mye­loma.1,9
mono­clo­nal gammopathy (MG).5-8 Waldenström was of the firm Using a com­bi­na­tion of serum-based pro­tein assays (SPEP,
belief that benign MG was unre­lated to MM. Conversely, the alter­ immunofixation, and serum FLC assays), MGUS cases can be
nate opin­ion was that some patients with asymp­tom­atic mono­ clas­si­fied based on the isotype of M-pro­teins pres­ent. So-called
clo­nal pro­teins nev­er­the­less progressed over time to MM and non-IgM MGUS is the most com­mon type and is defined by the
that it was impor­tant to not term the pro­cess entirely benign. pres­ence of IgG, IgA, and, rarely, immu­no­glob­u­lin D or immu­no­
In 1978, Dr Robert Kyle published his obser­va­tions from a ret­ro­ glob­u­lin E M-pro­teins.10 IgM MGUS is defined by the pres­ence of
spec­tive chart review of all­indi­vid­u­als (N = 241) diag­nosed with a IgM M-pro­teins.11 Light chain MGUS is defined by an abnor­mal FLC
MG at the Mayo Clinic prior to Jan­u­ary 1, 1971. In brief, among the ratio, indi­cat­ing an excess of mono­clo­nal FLCs in the absence of
241 cases, he found that after a 5-year fol­low-up period, (1) the M-pro­teins.12 Non-IgM MGUS and light chain MGUS are caused by
M-pro­tein remained sta­ble in 137 (57%) patients; (2) the M-pro­tein mono­clo­nal bone mar­row plasma cells, and they are pre­cur­sors
increased by 50% or more in 22 (9%) patients; (3) onset of MM, of MM.2 IgM MGUS is com­monly caused by mono­clo­nal lymph-

Downloaded from http://ashpublications.org/hematology/article-pdf/2021/1/662/1851672/662landgren.pdf by guest on 13 December 2021


Waldenström mac­ro­glob­u­li­ne­mia, or amy­loid light chain amy­ oplasmacytic cells and is a pre­cur­sor to other lymphoprolifera-
loid­osis occurred in 27 (11%) patients; and (4) 55 (23%) patients tive dis­or­ders, most nota­bly Waldenström mac­ro­glob­u­li­ne­mia,
died with­out 5-year fol­low-up serum.6 When he com­pared the chronic lym­pho­cytic leu­ke­mia, and, only in very rare instances
mean M-pro­tein con­cen­tra­tion and per­cent bone mar­row plasma (<0.5%), MM.11 In addi­tion, MGUS of all­types, espe­cially λ light
cell infil­tra­tion at base­line for the 4 groups, there were no dif­fer­ chain MGUS, can pre­cede amy­loid light chain amy­loid­osis.13 It
ences (mean con­cen­tra­tions were 1.6-1.8 g/dL and mean plasma should be noted that in the gen­eral MM pop­u­la­tion, around 80%
cell infil­tra­tion in bone mar­row aspi­rates was 3%-4% across the of patients have an M-pro­tein and approx­i­ma­tely 20% have light
4 groups).6 Based on these small num­bers, Kyle6 observed that chain secre­ tory MM (ie, with­ out evi­
dence of an M-pro­ tein).14
among indi­vid­u­als with an M-pro­tein and who later devel­oped Among patients with MM who have an M-pro­tein, most of them
MM, the size of the mono­clo­nal peak increased along with symp­ also have an abnor­mal FLC ratio caused by over­pro­duc­tion of
toms prior to onset of MM. Therefore, Kyle argued in his sem­i­nal either κ or λ FLCs.14 Among patients diag­nosed with MGUS with
paper from 1978 that “mono­clo­nal gammopathy of u ­ nde­ter­mined an M-spike, around 30% also have an abnor­mal FLC ratio.15
sig­nif­i­cance” (MGUS) is a bet­ter term because one can­not tell
whether the mono­clo­nal pro­tein will remain unchanged or Epidemiologic stud­ies: novel insights
whether the patient has an evolv­ ing MM. The word unde­ter­ In ret­ro­spec­tive stud­ies with long-term fol­low-up, Kyle et al10
mined was used to reflect that, at diag­no­sis, it was not pos­si­ble and oth­ers16 have reported 0.5% to 1.0% annual aver­age risk of
with avail­­able meth­ods (ie, SPEP to define the con­cen­tra­tion of pro­gres­sion from MGUS to MM. Importantly, most ret­ro­spec­tive
M-pro­teins and micros­copy to deter­mine the plasma cell per­ stud­ies seek­ing to iden­tify risk fac­tors for pro­gres­sion are based
cent­age in bone mar­row aspi­rates) to deter­mine which patients on sta­tis­ti­cal mod­els using risk fac­tor data from a sin­gle time
would ulti­mately prog­ress to MM. point (usu­ally the ini­tial workup).10,16 Data from this kind of mod­
el­ing have been used to develop clin­i­cal con­sen­sus guide­lines
Diagnostic cri­te­ria and types of abnor­mal serum pro­teins recommending annual periph­ eral blood mon­ i­tor­
ing of serum
The cur­rent def­i­ni­tion of MGUS is char­ac­ter­ized by the pres­ence pro­tein mark­ers and other assays for patients with inter­me­di­ate-
of M-pro­teins or an abnor­mal FLC ratio in periph­eral blood.1 For risk and high-risk MGUS.1 Inspired by the obser­va­tions by Kyle
an indi­vid­ual to be diag­nosed with MGUS, per cur­rent def­i­ni­ in 1978,6 a few smaller ret­ro­spec­tive stud­ies17,18 have pro­posed
tions, the con­cen­tra­tion of the mono­clo­nal spike (M-spike) has evolv­ing changes in M-pro­tein lev­els are asso­ci­ated with pro­
to be less than 3 g/dL, and for an indi­vid­ual to be diag­nosed gres­sion to MM. However, in clin­i­cal prac­tice, most patients are
with light chain MGUS, the FLC ratio has to be abnor­mal (nor­ typ­i­cally coun­seled based on their risk pro­file cap­tured at ini­tial
mal ref­er­ence for κ/λ FLC ratio, 0.26-1.65), but the involved/ workup. Regarding cases with light chain MGUS, there is only lim­
unin­volved ratio has to be less than 100.9 It should be empha­ ited infor­ma­tion avail­­able regard­ing the risk of pro­gres­sion from
sized that ele­vated FLC con­cen­tra­tions are not unique to plasma light chain MGUS to light chain MM, and con­se­quently, clin­i­cal
cell dis­or­ders, and a clin­i­cal inter­pre­ta­tion of the results is always guide­lines for this con­di­tion are lacking.12 However, the big­gest
required. For exam­ple, ele­vated FLC lev­els can be reflec­tive of lim­i­ta­tion of defin­ing risk in this fash­ion is lead-time bias because
under­ly­ing renal insuf­fi­ciency, auto­im­mune con­di­tions, sys­temic there are no pri­ mary care screen­ ing guide­ lines. Therefore,
inflam­ma­tion, infec­tion, and other causes. Furthermore, if the patients are often found to have MGUS after an inci­den­tal rou­tine
indi­vid­ual undergoes a bone mar­row biopsy, based on cur­rent lab­o­ra­tory find­ing (eg, as part of the workup for ele­vated total
diag­ nos­tic cri­te­ria, the plasma cell involve­ ment of the bone pro­tein). Alternatively, patients may be diag­nosed with MGUS
mar­row must be less than 10%.1 Last, the clin­i­cal workup must after a workup for non-mye­loma-defin­ing signs/symp­toms (eg,
be neg­a­tive for evi­dence of end-organ dam­age from plasma unex­plained periph­eral neu­rop­at­hy or slight increase in serum
cell dys­cra­sia, hyper­cal­ce­mia, ane­mia, renal fail­ure, lytic bone cre­at­i­nine).
lesions, or mul­ti­ple (2 or more) focal lesions in the skel­et­on by In 2009, the first pro­spec­tive pop­ul­a­tion-based MGUS screen­
mag­netic res­o­nance imag­ing.9 If 1 or more of those abnor­mal­ ing study was published. Using stored periph­eral blood sam­
i­ties are iden­ti­fied, unless there is another expla­na­tion for the ples that were col­lected as part of the large (N = 77 469) National
abnor­mal­ity (eg, ane­mia due to bleed­ing, renal fail­ure due to Cancer Institute Prostate, Lung, Colorectal, and Ovarian Cancer
car­dio­vas­cu­lar dis­ease), then the patient would be diag­nosed Screening Trial (NCI PLCO), this large study was a ­ ble to show
with MM. If none of the above-listed abnor­mal­i­ties are pres­ent, that MM is con­sis­tently pre­ceded by the MGUS pre­cur­sor stage.2
but the M-spike is 3 g/dL or more and/or there is over 10% (but This impor­tant obser­va­tion defin­i­tively links MM to its ­pre­cur­sor

664  |  Hematology 2021  |  ASH Education Program


dis­ease. This study has set the stage for future inves­ti­ga­tions limit of nor­mal; 1-2 points). For the first time, a scor­ing sys­tem
seek­ing

You might also like