You are on page 1of 699

6888_IFC 25/10/18 2:31 PM Page 2

Antigen-Antibody Characteristic Chart*


ANTIGENS
Antigen RBC Antigen Antigen Antigen
Antigen Antigen ISBT Freq. % Expression Distrib. Demonstrates Modification
System Name Name W B at Birth Plasma/RBC Dosage Enzyme/Other
**D RH1 85 92 strong RBC only no Enz. ↑

**C RH2 68 27 strong RBC only yes Enz. ↑


**E RH3 29 22 strong RBC only yes Enz. ↑

**c RH4 80 97 strong RBC only yes Enz. ↑


**e RH5 98 99 strong RBC only yes Enz. ↑
Rh
ce/f RH6 64 92 strong RBC only no Enz. ↑
Ce RH7 70 27 strong RBC only no Enz. ↑
Cw RH8 1 rare strong RBC only yes Enz. ↑

G RH12 86 92 strong RBC only no Enz. ↑


V RH10 1 30 strong RBC only no Enz. ↑

VS RH20 1 32 strong RBC only no Enz. ↑ AET → ZZAP →


K KEL1 9 rare strong RBC only occ Enz. → AET+ ↓ ZZAP ↓++

k KEL2 98.8 100 strong RBC only occ Enz. → AET+ ↓ ZZAP ↓++
Kpa KEL3 2 rare strong RBC only occ Enz. → AET+ ↓ ZZAP ↓++

Kell Kpb KEL4 99.9 100 strong RBC only occ Enz. → AET+ ↓ ZZAP ↓++
Jsa KEL6 .01 20 strong RBC only occ Enz. → AET+ ↓ ZZAP ↓++

Jsb KEL7 99.9 99 strong RBC only occ Enz. → AET+ ↓ ZZAP ↓++
†Kx — 99.9 99.9 weak RBC low occ Enz. → AET+ ↓ ZZAP ↓++

FYa FY1 65 10 strong RBC only yes Enz. ↓ AET ↓ ZZAP ↓

FYb FY2 83 23 strong RBC only yes Enz. ↓ AET ↓ ZZAP ↓


FY3 100 32 strong RBC only no Enz. → AET → ZZAP →
Duffy
FY5 100 32 expressed RBC only no Enz. → AET → ZZAP →
(on cord cells)
•FY6 100 32 expressed RBC only no Enz. ↓ AET → ZZAP ↓
(on cord cells)

*This chart is to be used for general information only. Please refer to the appropriate chapter for more detailed information.
AET = 2-aminoethylisthiouronium bromide; ↑ = enhanced reactivity; → = no effect; ↓ = depressed reactivity; occ = occasionally; CGD = chronic granulomatious disease;
HDN = hemolytic disease of the newborn; HTR = hemolytic transfusion reaction; NRBC = non-red blood cell; RBC = red blood cell; WBC = white blood cell; ZZAP = dithiothreitol
plus papain.
• No human antibody to FY6 has been reported.
† It has been found that Kx is inherited independently of the Kell system; consequently it is no longer referred to as K15.
**Frequency in Asians: D 99%, C 93%, E 39%, c 47%, e 96%
6888_IFC 25/10/18 2:31 PM Page 3

ANTIBODIES
Immunoglobin Clinical
Serology Comp. Class Optimum Significance
Stimulation Saline AHG Binding IgM IgG Temperature HTR HDN Comments
RBC occ yes no occ yes warm yes yes Very rarely IgA anti-D may be
produced; however, this is
invariably with IgG.
RBC occ yes no occ yes warm yes yes
RBC/NRBC occ yes no occ yes warm yes yes Anti-E may occur without
obvious immune stimulation.
RBC occ yes no occ yes warm yes yes
RBC occ yes no occ yes warm yes yes Warm autoantibodies may appear
to have anti-e-like specificity.
RBC occ yes no occ yes warm yes yes
RBC occ yes no occ yes warm yes yes
RBC/NRBC occ yes no occ yes warm yes yes Anti-Cw may occur without
obvious immune stimulation.
RBC occ yes no occ yes warm yes yes
RBC occ yes no occ yes warm yes yes Antibodies to V and VS can present
problems in the black population
where the antigen frequencies are in
the order of 30 to 32.
RBC occ yes no occ yes warm yes yes
RBC occ yes some occ yes warm yes yes Some antibodies to Kell antigens
have been reported to react poorly
in low ionic media.
RBC no yes no rarely yes warm yes yes
RBC no yes no no yes warm yes yes Kell system antigens are destroyed
by AET and by ZZAP.
RBC rarely yes no rarely yes warm yes yes
RBC rarely yes no rarely yes warm yes yes Anti-K1 has been reported to occur
following certain bacterial infections.
RBC no yes no no yes warm yes yes
RBC no yes no occ yes warm yes yes The lack of Kx expression on RBCs
and WBCs has been associated with
the McLeod phenotype and CGD.
RBC rare yes some rare yes warm yes yes Fya and Fyb antigens are destroyed by
enzymes. Fy (a–b–) cells are resistant
to invasion by P. vivax merozoites, a
malaria-causing parasite.
RBC rare yes some rare yes warm yes yes
RBC no yes rarely no yes warm yes yes FY3 and 5 are not destroyed by
enzymes.
RBC no yes ? no yes warm FY5 may be formed by interaction of
Rh and Duffy gene products.
FY6 was described using a mono-
clonal antibody which reacts
with most human red cells except
Fy(a–b–). This antigen is responsi-
ble for susceptibility of cells to
penetration by P. vivax.
(Continued on inside back cover)
6888_FM_i-xvi 31/10/18 10:54 AM Page i

Modern
Blood Banking
& Transfusion
Practices
Seventh Edition

Denise Harmening, PhD, MT(ASCP)


Director of the Online Masters of Science in Clinical
Laboratory Management
Professor, Department of Medical Laboratory
Science
College of Health Sciences
Rush University
Chicago, Illinois, USA
6888_FM_i-xvi 31/10/18 10:54 AM Page ii

F. A. Davis Company
1915 Arch Street
Philadelphia, PA 19103
www.fadavis.com

Copyright © 2019 by F. A. Davis Company

Copyright © 2019 by F. A. Davis Company. All rights reserved. This product is protected by copyright. No part of it may be reproduced, stored in a
retrieval system, or transmitted in any form or by any means, electronic, mechanical, photocopying, recording, or otherwise, without written permission
from the publisher.

Printed in the United States of America

Last digit indicates print number: 10 9 8 7 6 5 4 3 2 1

Senior Acquisitions Editor: Christa Fratantoro


Manager of Content Development: George W. Lang
Developmental Editor: Karen Gulliver
Art and Design Manager: Carolyn O’Brien
Content Project Manager: Megan Suermann

As new scientific information becomes available through basic and clinical research, recommended treatments and drug therapies undergo changes.
The author(s) and publisher have done everything possible to make this book accurate, up to date, and in accord with accepted standards at the time of
publication. The author(s), editors, and publisher are not responsible for errors or omissions or for consequences from application of the book, and make
no warranty, expressed or implied, in regard to the contents of the book. Any practice described in this book should be applied by the reader in accordance
with professional standards of care used in regard to the unique circumstances that may apply in each situation. The reader is advised always to check
product information (package inserts) for changes and new information regarding dose and contraindications before administering any drug. Caution is
especially urged when using new or infrequently ordered drugs.

Library of Congress Cataloging-in-Publication Data

Names: Harmening, Denise, editor.


Title: Modern blood banking & transfusion practices / [edited by] Denise
Harmening.
Other titles: Modern blood banking and transfusion practices
Description: Seventh edition. | Philadelphia : F.A. Davis Company, [2019] |
Includes bibliographical references and index.
Identifiers: LCCN 2018036883 (print) | LCCN 2018037766 (ebook) | ISBN
9780803694620 | ISBN 9780803668881 (pbk.)
Subjects: | MESH: Blood Banks—methods | Blood Transfusion—methods | Blood
Grouping and Crossmatching
Classification: LCC RM172 (ebook) | LCC RM172 (print) | NLM WH 460 | DDC
615/.39—dc23
LC record available at https://lccn.loc.gov/2018036883

Authorization to photocopy items for internal or personal use, or the internal or personal use of specific clients, is granted by F.A. Davis Company for users
registered with the Copyright Clearance Center (CCC) Transactional Reporting Service, provided that the fee of $.25 per copy is paid directly to CCC, 222
Rosewood Drive, Danvers, MA 01923. For those organizations that have been granted a photocopy license by CCC, a separate system of payment has been
arranged. The fee code for users of the Transactional Reporting Service is: 978-0-8036-6888-1/19 0 + $.25.
6888_FM_i-xvi 31/10/18 10:54 AM Page iii

To all students—full-time, part-time, past, present,


and future—who have touched and will continue to
touch the lives of so many educators. . . .
It is to you this book is dedicated in the hope of
inspiring an unquenchable thirst for knowledge!
6888_FM_i-xvi 31/10/18 10:54 AM Page iv
6888_FM_i-xvi 31/10/18 10:54 AM Page v

Foreword

Blood groups were discovered more than a century ago and This impressive legacy is a testament to the success of her
have a rich and varied history, including being recognized as approach and experience as an educator, administrator, re-
the first markers of human diversity used to study popula- searcher, and author, and is also reflected in her membership
tion migration and admixture. The study of blood groups on the “Medscape” Pathology Editorial Advisory Board.
has also made important contributions to biology: for in- She has gathered a group of experienced scientists and
stance, the Colton blood antigens are carried on a membrane teachers who, along with herself, cover all the important
protein called aquaporin, which was the first water channel areas of blood transfusion science. The book is divided into
discovered in mammals. Rh-associated glycoprotein (RhAG) five sections. The chapters included in Part I, “Fundamental
was the first mammalian ammonia transporter recognized, Concepts,” provide a firm base on which the student can
and the Kidd blood group protein was the first urea trans- learn the practical and technical importance of the other
porter discovered. All have related protein family members chapters. The chapters in Part II, “Blood Groups and Sero-
present in many tissues and organs. Modern Blood Banking logic Testing,” and Part III, “Transfusion Practices,” provide
& Transfusion Practice is an introduction to the role of blood information for medical technologists without overwhelm-
groups in transfusion therapy, in which blood groups are the ing them with esoteric and clinical details. Part IV covers
core of compatible blood transfusion. leukocyte antigens and relationship (parentage) testing. The
Today, the Internet has truly facilitated access to new chapters in Part V, “Quality Management and Compliance,”
knowledge in all areas of science, and the field of transfusion complete the scope of transfusion science. Although de-
medicine is no exception! Now, with knowledge mobile on signed primarily for medical technologists, the book is also
every device, we are in an age of information explosion. well suited to pathology residents, hematology fellows, and
However, to take advantage of the continual flow of new others who want to review principles of blood banking and
information generated by blood transfusion scientists and to transfusion practices.
apply it to everyday work in the blood bank, requires a solid Dr. Harmening indicates that this seventh edition will be
knowledge base. It can be difficult to select one book that her last revision. I speak for the profession in commending
covers all the information that technologists in training need and thanking her for the major contribution she has made
to know about blood transfusion science to build that foun- to the education of blood bankers. Importantly, the princi-
dation. It requires a well-thought-out systematic presenta- ples outlined here will remain; some details may change and
tion of information for training. Dr. Denise Harmening has new blood group antigens will be recognized, but the basic
produced that single volume, now in its seventh edition! foundation will remain relevant in this “collectors” edition.

CONNIE M. WESTHOFF, SBB, PhD


Executive Scientific Director
Immunohematology and Genomics
New York Blood Center

v
6888_FM_i-xvi 31/10/18 10:54 AM Page vi
6888_FM_i-xvi 31/10/18 10:54 AM Page vii

Preface

This book is designed to provide the medical technologist, basic concepts of genetics, immunology, and molecular
blood bank specialist, and resident with a concise and thor- biology. In Part II, four chapters focus on blood groups and
ough guide to transfusion practices and immunohematology. four on routine blood bank practices, which include “The
This text, a perfect “crossmatch” of theory and practice, pro- Antiglobulin Test,” “Detection and Identification of Anti-
vides the reader with a working knowledge of routine blood bodies,” “Pretransfusion Testing,” and “Blood Bank Testing
banking. Forty-four contributors from across the country Technologies and Automation.”
have shared their knowledge and expertise in 29 comprehen- Part III, “Transfusion Practices,” includes a chapter titled
sive chapters. More than 500 illustrations and tables facilitate “Cellular Therapy in the Transplant Setting” and covers the
the comprehension of difficult concepts not routinely illus- more traditional topics of donor screening, transfusion-
trated in other texts. In addition, color plates provide a means transmitted diseases, component preparation, transfusion
for standardizing the reading of agglutination reactions. therapy, transfusion reactions, and apheresis. Certain clinical
Several features of this textbook offer great appeal to situations that are particularly relevant to blood banking are
students and educators, including chapter outlines and also discussed in this section, including hemolytic disease of
educational objectives at the beginning of each chapter; case the fetus and newborn, autoimmune hemolytic anemias, and
studies, review questions, and summary charts at the end of tissue banking.
each chapter; and an extensive and convenient glossary for The human leukocyte antigen (HLA) system and relation-
easy access to definitions of blood bank terms. ship testing are discussed in Part IV of the book. In Part V,
A blood group antigen-antibody characteristic chart is “Quality Management and Compliance,” a chapter titled
provided on the inside cover of the book to aid in retention “Patient Blood Management” is discussed. The chapters on
of the vast amount of information presented, and serves as a quality management, transfusion safety and federal regula-
review of the characteristics of the blood group systems. tory requirements, laboratory information systems, and legal
Original, comprehensive step-by-step illustrations of ABO and ethical considerations complete the scope of practice for
forward and reverse grouping help the student to master this transfusion services.
important testing, which represents the foundation of blood This book is a culmination of the tremendous efforts of
banking. many dedicated professionals who participated in this proj-
The seventh edition is divided into the following sections: ect by donating their time and expertise because they care
about the blood bank profession. The book’s intention is to
• Part I: Fundamental Concepts
foster improved patient care by providing the reader with a
• Part II: Blood Groups and Serologic Testing
basic understanding of modern blood banking and transfu-
• Part III: Transfusion Practices
sion practices. The seventh edition is designed to generate
• Part IV: Leukocyte Antigens and Relationship Testing
an unquenchable thirst for knowledge in all medical labo-
• Part V: Quality Management and Compliance
ratory scientists, blood bankers, and practitioners, whose
In Part I, the introduction to the historical aspects of red education, knowledge, and skills provide the public with
blood cell and platelet preservation serves as a prelude to the excellent health care.

DENISE HARMENING, PhD, MT(ASCP)

vii
6888_FM_i-xvi 31/10/18 10:54 AM Page viii

Editorial Reviewers

We especially thank our editorial reviewers for assisting with manuscript, art, and page proof review.
Regina M. Leger, MSQA, MT(ASCP)SBB, CMQ/OE(ASQ) Gale Suwe, BS, MT(ASCP)SBB
Director, Specialist in Blood Banking (SBB) Program Instructor, Specialist in Blood Bank Technology Program
Lead Technologist, Special Immunohematology Lab Rush University
American Red Cross Blood Services Chicago, IL USA
Southern California Region
Pomona, California, USA

Justin Richard Rhees, MS, MLS(ASCP)CM, SBBCM


Assistant Professor, Medical Laboratory Sciences
Weber State University
Ogden, Utah, USA

viii
6888_FM_i-xvi 31/10/18 10:54 AM Page ix

Contributors

Robert W. Allen, PhD Judy Ellen Ciaraldi, BS, MT(ASCP)SBB, CQA(ASQ)


Chair & Director School of Forensic Sciences Consumer Safety Officer
Center for Health Sciences Division of Blood Applications
Oklahoma State University Office of Blood Research and Review
Tulsa, Oklahoma, USA Center for Biologics Evaluation and Research
U.S. Food and Drug Administration
Lucia M. Berte, MA, MT(ASCP)SBB, DLM; Rockville, Maryland, USA
COA(ASQ)CMQ/OE
President Julie L. Cruz, MD, FACP
Laboratories Made Better! P.C. Medical Director
Quality Systems Consultant Versiti-Indiana Blood Center
Broomfield, Colorado, USA Indianapolis, Indiana, USA

Maria P. Bettinotti, PhD, dip. ABHI Meghan Delaney, DO, MPH


Assistant Professor of Medicine Chief, Pathology and Laboratory Medicine
Director, Immunogenetics Laboratory Children’s National Health System
Johns Hopkins George Washington University Medical School
Baltimore, MD, USA Washington, DC, USA

Susanne Bishop, MS, MLS(ASCP)CM SBBCM Helene DePalma, MS, MT(ASCP)SBB, CQA(ASQ)
Assistant Professor Associate Professor
Division of Medical Laboratory Science City University of New York-York College
College of Allied Health Professionals Jamaica, New York, USA
University of Nebraska Medical Center
Omaha, Nebraska, USA Laurie Gillard, MS, MLS(ASCP)CM SBB
Director of the Specialist in Blood Banking Program
Theresa Bostock, MS, MT(ASCP)SBB Assistant Professor, Department of Medical Laboratory Science
Laboratory Director Rush University
Orange Regional Medical Center Chicago, Illinois, USA
Middletown, New York, USA
Ralph E. B. Green B. App. Sci, FAIMLS, MACE
Michele R. Brown, PhD, MS, MLS(ASCP)CM SBBCM Formerly, Associate Professor
Assistant Professor & Program Director, Healthcare Simulation Discipline and Program Leader
Department of Health Services Administration, School of Health Discipline of Laboratory Medicine
Professions School of Medical Sciences
Simulation Associate, Office of Interprofessional Simulation for RMIT University
Innovative Clinical Practice Melbourne, Australia
University of Alabama
Birmingham, Alabama, USA Steven F. Gregurek, MD
Transfusion Medicine Medical Director
Lorraine Caruccio, PhD, MT(ASCP)SBB Valley Health System
Formerly, National Institutes of Health Las Vegas, Nevada, USA
Rockville, Maryland, USA
Denise Harmening, PhD, MT(ASCP)
JoAnn Christensen, MS, MT(ASCP)SBB Director of the Online Masters in Clinical Laboratory Management
Director, IRL East Area 2 Professor, Department of Medical Laboratory Science
NY-Penn, NE PA, and Greater Alleghenies Regions College of Health Sciences
American Red Cross Rush University
West Henrietta, New York, USA Chicago, Illinois, USA

ix
6888_FM_i-xvi 31/10/18 10:54 AM Page x

x Contributors

Elizabeth A. Hartwell, MD, MT(ASCP)SBB Richard H. McBride, Jr, MS, MT(ASCP)SBB


Associate Professor, Department of Pathology and Immunology Chief, Blood and Plasma Branch
Baylor College of Medicine Division of Blood Components and Devices
Associate Medical Director Office of Blood Research and Review
Baylor St. Luke’s Medical Center Center for Biologics Research and Review
Houston, Texas, USA Food and Drug Administration
Silver Spring, Maryland, USA
Virginia C. Hughes, PhD, MLS(ASCP)SBB
Associate Professor Matthew Montgomery, MD
Department of Medical Laboratory Sciences Assistant Medical Director
College of Health Sciences Hoxworth Blood Center
University of Delaware University of Cincinnati
Newark, Delaware, USA Cincinnati, Ohio, USA

Melanie S. Kennedy, MD Gerald P. Morris, MD, PhD


Clinical Associate Professor Emeritus Department of Pathology
Department of Pathology University of California, San Diego
College of Medicine San Diego, California, USA
The Ohio State University
Columbus, Ohio, USA Angela K. Novotny, MT(ASCP)SBBCM
Manager, Blood Bank
Scott A. Koepsell, MD, PhD The University of Texas Medical Branch
Department of Pathology and Microbiology Galveston, Texas, USA
University of Nebraska Medical Center
Omaha, Nebraska, USA Donna L. Phelan, BA, MT(HEW), CHS(ASHI)
Technical Supervisor
Barbara Kraj, PhD, MLS(ASCP)CM MBCM HLA Laboratory
Associate Professor and Program Director Barnes-Jewish Hospital
School of Medical Diagnostics and Translational Science St. Louis, Missouri, USA
Old Dominion University
Norfolk, VA, USA Mark D. Pool, MD
Assistant Professor of Pathology, Rush Medical College
Regina M. Leger, MSQA, MT(ASCP)SBB, CMQ/OE(ASQ) Blood Center Medical Director, Rush University Medical Center
Director, Specialist in Blood Banking (SBB) Program Chicago, Illinois, USA
Lead Technologist, Special Immunohematology Lab
American Red Cross Blood Services Justin Richard Rhees, MS, MLS (ASCP)CM, SBBCM
Southern California Region Assistant Professor
Pomona, California, USA Medical Laboratory Sciences
Weber State University
Nadine Lerret, PhD, MLS(ASCP)CM Ogden, Utah, USA
Assistant Professor, Clinical Chemistry Course Director
Department of Medical Laboratory Science Karen Rodberg, MBA, MT(ASCP)SBB
Rush University Director, Reference Services
Chicago, Illinois, USA American Red Cross Blood Services
Southern California Region
Beth L. Manning, BS, MT(ASCP)SBBCM Pomona, California, USA
Marshfield Clinic Health System
Transfusion Services Kathleen Sazama, MD, JD, MS, MT(ASCP)
Marshfield, Wisconsin, USA Formerly, Chief Medical Officer
LifeSouth Community Blood Centers, Inc.
Holli Mason, MD Gainesville, Florida, USA
Director, Transfusion Medicine and Serology
Director, Pathology Residency Training Program
Harbor UCLA Medical Center
Associate Clinical Professor
David Geffen School of Medicine at UCLA
Torrance, California, USA
6888_FM_i-xvi 31/10/18 10:54 AM Page xi

Contributors xi

Jayanna Slayten, MS, MT(ASCP)SBBCM Heather M. Vaught, BS, MLS(ASCP)CM SBBCM


Blood Bank Supervisor Director, Transfusion Medicine Laboratory
Indiana University Health Blood Bank Indiana University Health Pathology Laboratory
Indianapolis, Indiana, USA Indianapolis, Indiana, USA
And Adjunct Faculty, University of Texas Medical Branch
Specialist in Blood Banking Program Phyllis S. Walker, MS, MT(ASCP)SBB
Galveston, Texas USA Formerly, Manager, Immunohematology Reference Laboratory
Blood Centers of the Pacific
Phyllis Straff, MS(ISM), MT(ASCP) San Francisco, California, USA
Laboratory Information Systems Project Manager
Dept of Pathology and Laboratory Medicine Elizabeth F. Williams, MHS, MT(ASCP)CM, SBB
Rush University, College of Health Sciences Associate Professor, Clinical Educational Coordinator
Chicago, Illinois, USA Department of Clinical Laboratory Sciences
School of Allied Health Professions
Gale Suwe, BS, MT(ASCP)SBB LSUHSC Health Sciences Center
Instructor New Orleans, Louisiana, USA
Rush University
Specialist in Blood Banking Program Laurie Wolf, MLS(ASCP)SBBCM
Chicago, Illinois, USA Transfusion Service Manager, Pathology and Laboratory Medicine
The University of Kansas Health System
Ann Tiehen, MT(ASCP)SBB Kansas City, Kansas USA
Formerly, Education Coordinator
North Shore University Health System Jules G. Zinni, MLS(ASCP)CM MBCM
Evanston Hospital Medical Technologist, Blood Bank
Department of Pathology and Laboratory Medicine Northwestern Memorial Hospital
Evanston, Illinois, USA Chicago, Illinois, USA, USA

Kathleen S. Trudell, MLS(ASCP)CM SBBCM


Clinical Coordinator, Immunohematology
Division of Medical Laboratory Science
College of Allied Health Professions
University of Nebraska Medical Center
Omaha, Nebraska, USA
6888_FM_i-xvi 31/10/18 10:54 AM Page xii

Reviewers

Leslie M. Allshouse, MEd, MBA, MLS(ASCP)CM Phyllis Ingham, EdD, Med, MT(ASCP)
Program Director and Senior Instructor, Medical Laboratory Science Program Program Director, Clinical Laboratory Technology/Phlebotomy
University of Delaware West Georgia Technical College
Newark, Delaware, USA Waco, Georgia, USA

Karen Bowers, RT, MEd Rebecca Lawson, MS, MLS(ASCP)CM, AHI(AMT)


Instructor, Medical Laboratory Technology Science Program Staff Associate, Health Sciences and Distance Learning Division
College of New Caledonia Adjunct Instructor, Clinical Laboratory Technologies
Prince George, British Columbia, Canada SUNY Broome Community College
Binghamton, New York, USA
Maritza Cintron, MPA, BS
Technician Program Director, Medical Laboratory Teresita Padilla-Beneviedes, MSc, PhD
Marion Technical College Instructor and Faculty Diversity Scholar, Biochemistry and Molecular Pharmacology
Ocala, Florida, USA University of Massachusetts
Worcester, Massachusetts, USA
Christina Camillo, MS, MLS(ASCP)CM
Instructor and Clinical Coordinator, Health Sciences Art Phillips, MS, MT(ASCP) BB
Salisbury University Adjunct Lecturer, Biology–Medical Technology Program
Salisbury, Maryland, USA The College of Staten Island (CUNY)
Staten Island, New York, USA
Guyla Evans, PhD, MLS(ASCP)CM, SC(ASCP)CM
Clinical Assistant Professor, Clinical Laboratory Science Kal Randhawa, MLT, Med
East Carolina University Faculty, Medical Laboratory Science
Greenville, North Carolina, USA British Columbia Institute of Technology
Barnaby, British Columbia, Canada
Candice Suzanne Grayson, MS, MA, MLS(ASCP)CM
Program Director, Medical Laboratory Technology Program Carol Rentas, PhD, MEd, MT(ASCP)
Community College of Baltimore County Director, Undergraduate MLS Programs, Integrative Health Sciences
Baltimore, Maryland, USA George Washington University
Ashburn, Virginia, USA
Andrea Hoffmann Fontenot, MLS(ASCP)SBBCM
Assistant Professor, Medical Laboratory Technician Program Justin Richard Rhees, MS MLS(ASCP)CM, SBBCM
Delgado Community College Assistant Professor, Medical Laboratory Sciences
New Orleans, Louisiana, USA Weber State University
Ogden, Utah, USA
Kerry Harbert, MA, MT(ASCP)
Associate Professor, Medical Laboratory Science Ann Christine Ridder, MBA, MLS(ASCP)CM
West Virginia University Clinical Education Program Director
Morgantown, West Virginia, USA School of Clinical Laboratory Science
Augusta Health
Hillary Harris, MLS(ASCP)
Fishersville, Virginia, USA
Medical Laboratory Scientist
Indiana University Health University Hospital, Brenda Schoffstall, PhD, MT(ASCP)
Indianapolis, Indiana, USA Associate Professor, Biology
Barry University
Elizabeth Hart, MA, CLS
Miami, Florida, USA
Senior Lecturer, Medical Laboratory Science
University of Massachusetts Dartmouth
Dartmouth, Massachusetts, USA

xii
6888_FM_i-xvi 31/10/18 10:54 AM Page xiii

Reviewers xiii

Judith A. Seidel, MT(ASCP)SBB Lorraine Torres, EdD, MT(ASCP)


Clinical Instructor, Clinical Laboratory Science Program, Academic Affairs Program Director, Clinical Laboratory Science Program
Indiana University Health The University of Texas at El Paso
Indianapolis, Indiana, USA El Paso, Texas, USA

Kelly D. Shirley, MAEd, MLS(ASCP)CM SBBCM Kathleen Winker, MT(ASCP)


Education Coordinator, School of Clinical Laboratory Science Program Director, MLT – Health and Public Safety
Carolinas College of Health Sciences Blackhawk Technical College
Charlotte, North Carolina, USA Monroe, Wisconsin, USA

Tasha T. Simpson MSc, C (ASCP)CM, MLS(ASCPi)CM Jeff Wolz, MA, MT(ASCP)


Assistant Professor, Medical Laboratory Sciences Director, MLS Program
Andrews University Arizona State University
Berrien Springs, Michigan, USA Phoenix, Arizona, USA

James Kyle Taylor, PhD, MLS(ASCP)CM


Department Head, Medical and Clinical Laboratory Sciences
Auburn University at Montgomery
Montgomery, Alabama, USA
6888_FM_i-xvi 31/10/18 10:54 AM Page xiv

Contents

Part I 17 Adverse Effects of Blood Transfusion .................................. 373


Fundamental Concepts
18 Apheresis ....................................................................................................... 396
1 Red Blood Cell and Platelet Preservation:
Historical Perspectives and Current Trends ........................ 1 19 Cellular Therapy in the Hematopoietic
Transplant Setting .................................................................................. 417
2 Basic Genetics .............................................................................................. 24
20 Hemolytic Disease of the Fetus and
3 Fundamentals of Immunology ...................................................... 45 Newborn (HDFN) .................................................................................... 427
4 Concepts in Molecular Biology .................................................... 77 21 Autoimmune Hemolytic Anemias ............................................ 441

Part II 22 Tissue Banking as a Part of the Transfusion


Service .............................................................................................................. 476
Blood Groups and Serologic Testing

5 The Antiglobulin Test .......................................................................... 103 Part IV


Leukocyte Antigens and Relationship Testing
6 The ABO Blood Group System .................................................... 119
23 The HLA System ...................................................................................... 497
7 The Rh Blood Group System ........................................................ 149
24 Relationship Testing ............................................................................. 515
8 Blood Group Terminology and Common
Blood Groups: The Lewis, P, I, MNS, Kell,
Duffy, Kidd, and Lutheran .............................................................. 173 Part V
Quality Management and Compliance
9 Uncommon Blood Groups ............................................................... 212
25 Quality Management in the Blood Bank ........................... 531
10 Detection and Identification of Antibodies .................... 232
26 Patient Blood Management .......................................................... 552
11 Pretransfusion Testing ....................................................................... 256
27 Transfusion Safety and Federal Regulatory
12 Blood Bank Testing Technologies Requirements ............................................................................................. 574
and Automation ....................................................................................... 268
28 Laboratory Information Systems
Part III in the Blood Bank .................................................................................. 590
Transfusion Practices 29 Medicolegal and Ethical Aspects of Providing
Blood Collection and Transfusion Services ..................... 607
13 Donor Selection ....................................................................................... 281

14 Transfusion-Transmitted Diseases ........................................... 307 Answer Key .............................................................................................................. 617

15 Component Preparation ................................................................... 333


Glossary ...................................................................................................................... 629

Index .............................................................................................................................. 653


16 Transfusion Therapy ............................................................................ 355

xiv
6888_FM_i-xvi 31/10/18 10:54 AM Page xv

Procedures Available on DavisPlus

The following procedures can be found on the textbook’s companion website at DavisPlus.

Related Chapter Procedure


Chapter 5: The Antiglobulin Test • Procedure 5–1: Direct Antiglobulin Test
• Procedure 5–2: Indirect Antiglobulin Test

Chapter 6: The ABO Blood Group System • Procedure 6–1: Determination of the Secretor Property

Chapter 10: Detection and Identification • Procedure 10–1: Plasma Inhibition Studies
of Antibodies

Chapter 11: Pretransfusion Testing • Procedure 11–1: Preparation of Washed “Dry” Button of RBCs for Serologic Tests
• Procedure 11–2: Model One-Tube-Per-Donor-Unit Crossmatch Procedure
• Procedure 11–3: Saline Replacement Procedure

Chapter 21: Autoimmune Hemolytic Anemias • Procedure 21–1: Use of Thiol Reagents to Disperse Autoagglutination
• Procedure 21–2: Cold Autoadsorption
• Procedure 21–3: Prewarm Technique for Testing Serum Containing Cold Agglutinins
• Procedure 21–4: Adsorption of Cold Autoantibodies With Rabbit Erythrocyte Stroma
• Procedure 21–5: Dissociation of IgG by Chloroquine
• Procedure 21–6: Digitonin-Acid Elution
• Procedure 21–7: Autologous Adsorption of Warm Reactive Autoantibodies Application
• Heat and Enzyme Method
• ZZAP Method
• Procedure 21–8: Demonstration of Drug-Induced Immune Complex Formation
• Procedure 21–9: Detection of Antibodies to Penicillin or Cephalothin
• Procedure 21–10: EDTA/Glycine Acid (EGA) Method to Remove Antibodies From RBCs
• Procedure 21–11: Separation of Transfused From Autologous RBCs by Simple
Centrifugation: Reticulocyte Harvesting

Also available at DavisPlus (http://davisplus.fadavis.com/): Polyagglutination, by Phyllis S. Walker, MS, MT(ASCP)SBB.

xv
6888_FM_i-xvi 31/10/18 10:54 AM Page xvi
Fundamental Concepts

Chapter 1
Red Blood Cell and Platelet Preservation:
Historical Perspectives and Current Trends
Denise M. Harmening, PhD, MT(ASCP) and Michelle R. Brown, PhD, MS,
MLS(ASCP)CMSBBCM

Introduction Formation of O Type RBCs Current Trends in Platelet Preservation


Historical Overview Blood Pharming Research
Current Status RBC Substitutes Development of Platelet Substitutes
RBC Biology and Preservation Tissue Engineering of RBCs Revisiting Approaches for Storage of
RBC Membrane Platelets at 1°c to 6°C
Platelet Preservation
Metabolic Pathways The Platelet Storage Lesion Frozen Platelets
RBC Preservation Clinical Use of Platelets Summary Chart
Anticoagulant Preservative Solutions Platelet Testing and Quality Control Review Questions
Additive Solutions Monitoring References
Freezing and Rejuvenation Measurement of Viability and
Current Trends in RBC Preservation Functional Properties of Stored
Research Platelets
Improved Additive Solutions Platelet Storage and Bacterial
Contamination
Procedures to Reduce and Inactivate
Pathogens Pathogen Reduction for Platelets

OBJECTIVES
1. List the major developments in the history of transfusion medicine.
2. Describe several biological properties of red blood cells (RBCs) that can affect post-transfusion survival.
3. Identify the metabolic pathways that are essential for normal RBC function and survival.
4. Define the hemoglobin-oxygen dissociation curve, including how it is related to the delivery of oxygen to tissues by
transfused RBCs.
5. Explain how transfusion of stored blood can cause a shift to the left of the hemoglobin-oxygen dissociation curve.
6. State the temperature for storage of RBCs in the liquid state.
Continued
6888_Ch01_001-023 29/10/18 4:38 PM Page 2

2 PART I Fundamental Concepts

OBJECTIVES—cont’d
7. Define storage lesion and list the associated biochemical changes.
8. Explain the importance of 2,3-diphosphoglycerate (2,3-DPG) levels in transfused blood, including what happens to levels
post-transfusion and which factors are involved.
9. Name the approved anticoagulant preservative solutions, explain the function of each ingredient, and state the maximum
storage time for RBCs collected in each.
10. Name the additive solutions licensed in the United States, list the common ingredients, and describe the function of each
ingredient.
11. Explain how additive solutions are used and list their advantages.
12. Explain rejuvenation of RBCs.
13. List the name and composition of the FDA-approved rejuvenation solution and state the storage time following rejuvenation.
14. Define the platelet storage lesion.
15. Describe the indications for platelet transfusion and the importance of the corrected count increment (CCI).
16. Explain the storage requirements for platelets.
17. Explain the swirling phenomenon and its significance.
18. List the two major reasons why routine platelet storage is limited to 5 days in the United States.
19. List the various ways that blood banks in the United States meet the FDA regulation requiring that blood establishments
and transfusion services must assure that the risk of bacterial contamination of platelets is adequately controlled using FDA
approved or cleared devices.
20. Explain the use and advantages of platelet additive solutions (PASs), and name two that are approved for use in the
United States.

Introduction first example of blood preservation research. Karl Landsteiner


in 1901 discovered the ABO blood groups and explained
People have always been fascinated by blood: Ancient the serious reactions that occur in humans as a result of
Egyptians bathed in it, aristocrats drank it, authors and incompatible transfusions. His work in the beginning of the
playwrights used it as themes, and modern humanity trans- 20th century won a Nobel Prize.
fuses it. The road to an efficient, safe, and uncomplicated Next came devices designed for performing the transfu-
transfusion has been difficult, but great progress has been sions. Edward E. Lindemann was the first to succeed. He
made. This chapter reviews the historical events leading to carried out vein-to-vein transfusion of blood by using
the current status of how blood is stored. A review of red multiple syringes and a special cannula for puncturing the
blood cell (RBC) biology serves as a building block for the vein through the skin. However, this time-consuming, com-
discussion of red blood cell preservation, and a brief plicated procedure required many skilled assistants. It was
description of platelet metabolism sets the stage for review- not until Unger designed his syringe-valve apparatus that
ing the platelet storage lesion. Current trends in red blood transfusions from donor to patient by an unassisted physi-
cell and platelet preservation research are presented for cian became practical.
the inquisitive reader. An unprecedented accomplishment in blood transfusion
was achieved in 1914 when Hustin reported the use of
Historical Overview sodium citrate as an anticoagulant solution for transfusions.
Later, in 1915, Lewisohn determined the minimum amount
In 1492, blood was taken from three young men and given of citrate needed for anticoagulation and demonstrated its
to the stricken Pope Innocent VII in the hope of curing nontoxicity in small amounts. Transfusions became more
him; unfortunately, all four died. Although the outcome of practical and safer for the patient.
this event was unsatisfactory, it is the first time a blood The development of preservative solutions to enhance the
transfusion was recorded in history. The path to successful metabolism of the RBCs followed. Glucose was evaluated as
transfusions that is so familiar today is marred by many early as 1916, when Rous and Turner introduced a citrate-
reported failures, but our physical, spiritual, and emotional dextrose solution for the preservation of blood. However, the
fascination with blood is primordial. Why did success elude function of glucose in RBC metabolism was not understood
experimenters for so long? until the 1930s. Therefore, the common practice of using
Clotting was the principal obstacle to overcome. Attempts glucose in the preservative solution was delayed. World War II
to find a nontoxic anticoagulant began in 1869, when Braxton stimulated blood preservation research because the demand
Hicks recommended sodium phosphate. This was perhaps the for blood and plasma increased. During World War II,
6888_Ch01_001-023 29/10/18 4:38 PM Page 3

Chapter 1 Red Blood Cell and Platelet Preservation: Historical Perspectives and Current Trends 3

the pioneer work of Dr. Charles Drew on developing tech- blood drives conducted at their place of work, school, and
niques in blood transfusion and blood preservation led to church, as well as at community and hospital-based blood
the establishment of a widespread system of blood banks.1 centers. Volunteer donors are not paid and provide nearly all
In February 1941, Dr. Drew was appointed director of of the blood used for transfusion in the United States.
the first American Red Cross blood bank at Presbyterian Traditionally, the amount of whole blood in a unit has been
Hospital.1 The pilot program Dr. Drew established became 450 mL ± 10% of blood (1 pint). More recently, 500 mL ± 10%
the model for the national volunteer blood donor program of blood is being collected.5 These units are collected from
of the American Red Cross.1 donors with a minimum hematocrit of 38%.5 Modified plastic
In 1943, Loutit and Mollison of England introduced the for- collection systems are used when collecting 500 mL of blood,
mula for the preservative acid-citrate-dextrose (ACD). Efforts with the volume of anticoagulant preservative solution being
in several countries resulted in the landmark publication of the increased from 63 to 70 mL. The total blood volume of most
July 1947 issue of the Journal of Clinical Investigation, which adults is 10 to 12 pints, and donors can replenish the fluid lost
devoted nearly a dozen papers to the topic of blood preserva- from the 1-pint donation in 24 hours. The donor’s red blood
tion. Hospitals responded immediately, and in 1947 blood cells are replaced within 1 to 2 months after donation.4 A
banks were established in many major cities of the United volunteer donor can donate whole blood every 8 weeks. (Refer
States; subsequently, transfusion became commonplace. to Chapter 13 on Donor Selection.)
The daily occurrence of transfusions led to the discovery Units of the whole blood collected can be separated into
of numerous blood group systems. Antibody identification three components: packed RBCs, platelets, and plasma. In
surged to the forefront as sophisticated techniques were recent years, less whole blood has been used to prepare
developed. The interested student can review historic events platelets because of the increased utilization of apheresis
during World War II in Kendrick’s Blood Program in World platelets. Hence, many units are converted only into RBCs
War II, Historical Note.2 In 1957, Gibson introduced an im- and plasma. The plasma can be converted by cryoprecipita-
proved preservative solution called citrate-phosphate-dextrose tion to a clotting factor concentrate that is rich in fibrinogen.
(CPD), which was less acidic and eventually replaced ACD A unit of whole blood–prepared RBCs may be stored for
as the standard preservative used for blood storage. 21 to 42 days, depending on the anticoagulant preservative
Frequent transfusions and the massive use of blood soon solution used when the whole blood unit is collected and
resulted in new problems, such as circulatory overload. whether a preserving solution is added to the separated
Component therapy has helped these problems. In the past, RBCs. Donated blood is free. However, there is a cost asso-
a single unit of whole blood could serve only one patient. ciated with collection, testing, processing, storing, and ship-
With component therapy, however, one unit may be used for ping of the blood components. The donation process
multiple transfusions. Today, health-care providers can select consists of three predonation steps. Donors receive the
the specific component for their patient’s particular needs following (Box 1–1):
without risking the inherent hazards of whole blood trans-
1. Educational reading materials
fusions. Health-care providers can transfuse only the re-
2. A donor health history questionnaire
quired fraction in the concentrated form, decreasing the
3. An abbreviated physical examination
possibility of overloading the circulatory system. Appropriate
blood component therapy now provides more effective treat-
ment and more complete use of blood products. Extensive
use of blood during this period, coupled with component BOX 1–1
separation, led to increased comprehension of erythrocyte The Donation Process
metabolism and a new awareness of the problems associated
with RBC storage. Step 1: Educational Materials
Educational material (such as the AABB pamphlet “An Important
Current Status Message to All Blood Donors”) that contains information on the risks
of infectious diseases transmitted by blood transfusion, including
the symptoms and signs of AIDS, is given to each prospective donor
AABB, formerly the American Association of Blood Banks, to read.
estimates that 6.8 million volunteers donate blood each year.
Based on the 2015 National Blood Collection and Utilization Step 2: The Donor Health History Questionnaire
Survey (NBCUS) approximately 12.6 million units of red A uniform donor history questionnaire, designed to ask questions
blood cells (RBCs) were collected, and around 11.4 million that protect the health of both the donor and the recipient, is given
to every donor. The health history questionnaire is used to identify
were transfused.3 This represents a decline of 11.6% and donors who have been exposed to diseases that can be transmitted
13.9%, respectively since 2013.4 With an aging population in blood (e.g., variant Creutzfeldt-Jakob, West Nile virus, malaria,
and advances in medical treatments requiring transfusions, babesiosis, or Chagas disease).
the demand for blood and blood components is expected to Step 3: The Abbreviated Physical Examination
continue to be high. It is estimated that one in three people
The abbreviated physical examination for donors includes blood
will need blood at some point in their lifetime.4 These units pressure, pulse, and temperature readings; hemoglobin or hemat-
are donated by fewer than 10% of healthy Americans who ocrit level; and the inspection of the arms for skin lesions.
are eligible to donate each year.4 Volunteers can donate at
6888_Ch01_001-023 29/10/18 4:38 PM Page 4

4 PART I Fundamental Concepts

The donation process, especially steps 1 and 2, has been RBC Membrane
carefully modified over time to allow for the rejection of
donors who may transmit transfusion-associated disease to The RBC membrane represents a semipermeable lipid bilayer
recipients. For a more detailed description of donor screen- supported by a mesh-like protein cytoskeleton structure
ing and processing, refer to Chapter 13. (Fig. 1–1).7 Phospholipids, the main lipid components of the
The nation’s blood supply is safer than it has ever been be- membrane, are arranged in a bilayer structure comprising
cause of the donation process and extensive laboratory testing the framework in which globular proteins traverse and
of blood. Current infectious disease screening tests performed move. Proteins that extend from the outer surface and span
on each unit of donated blood are listed in Table 1–1. For a the entire membrane to the inner cytoplasmic side of the
more detailed description of transfusion-associated disease, RBC are termed integral membrane proteins. Beneath the
refer to Chapter 14. lipid bilayer, a second class of membrane proteins, called
peripheral proteins, is located and limited to the cytoplasmic
RBC Biology and Preservation surface of the membrane forming the RBC cytoskeleton.7

Three areas of RBC biology are crucial for normal erythrocyte


survival and function: Advanced Concepts
1. Normal chemical composition and structure of the RBC Both proteins and lipids are organized asymmetrically
membrane within the RBC membrane. Lipids are not equally distrib-
2. Hemoglobin structure and function uted in the two layers of the membrane. The external layer
3. RBC metabolism6 is rich in glycolipids and choline phospholipids.7 The
internal cytoplasmic layer of the membrane is rich in amino
Defects in any or all of these areas will result in RBCs
phospholipids.7 The biochemical composition of the RBC
surviving fewer than the normal 120 days in circulation.
membrane is approximately 52% protein, 40% lipid, and
8% carbohydrate.6
As mentioned previously, the normal chemical compo-
sition and the structural arrangement and molecular inter-
Table 1–1 Current Donor Screening Tests actions of the erythrocyte membrane are crucial to the
for Infectious Diseases normal length of RBC survival of 120 days in circulation.
In addition, they maintain a critical role in two important
Test Date Test Required
RBC characteristics: deformability and permeability.8
Syphilis 1950s
Deformability
Hepatitis B surface antigen (HBsAg) 1971
Hepatitis B core antibody (anti-HBc) 1986 To remain viable, normal RBCs must also remain flexible,
deformable, and permeable. The loss of adenosine triphos-
Hepatitis C virus antibody (anti-HCV) 1990 phate (ATP) (energy levels) leads to a decrease in the phos-
Human immunodeficiency virus 19921 phorylation of spectrin and, in turn, a loss of membrane
antibodies (anti-HIV-1/2) deformability.6 An accumulation or increase in deposition
of membrane calcium also results, causing an increase in
Human T-cell lymphotropic virus 19972 membrane rigidity and loss of pliability.8 These cells are at
antibody (anti-HTLV-I/II)
a marked disadvantage when they pass through the small
Human immunodeficiency virus 1999 (3 to 5 µm in diameter) sinusoidal orifices of the spleen,
(HIV-1) NAT* an organ that functions in extravascular sequestration, and
Hepatitis C virus (HCV) NAT* 1999 removal of aged, damaged, or less deformable RBCs or
fragments of their membrane. The loss of RBC membrane
West Nile virus NAT 2004 is exemplified by the formation of spherocytes (cells with
Trypanosoma cruzi antibody 2007 a reduced surface-to-volume ratio; Fig. 1–2) and bite cells,
(anti-T. cruzi) in which the removal of a portion of membrane has left a
permanent indentation in the remaining cell membrane
Hepatitis B virus (HBV) NAT 2009 (Fig. 1–3). The survival of these forms is also shortened.
Babesia microti antibody and NAT 2012
(recommended) Permeability
Zika virus NAT 2016 The permeability properties of the RBC membrane and the
active RBC cation transport prevent colloid hemolysis and
NAT = nucleic acid amplification testing
*Initially under IND starting in 1999. control the volume of the RBCs. Any abnormality that
1Anti-HIV-1 testing implemented in 1985. increases permeability or alters cationic transport may
2Anti-HTLV testing implemented in 1988.
6888_Ch01_001-023 29/10/18 4:38 PM Page 5

Chapter 1 Red Blood Cell and Platelet Preservation: Historical Perspectives and Current Trends 5

I = integral proteins
Spectrin P = peripheral proteins
ankyrin-band 3
Phospholipids interaction
Fatty acid
chains GP-C Membrane
F-actin surface

I Lipid
I I I bilayer

GP-B 3 3 GP-A
7
P 2.1
4.2 6 P
P Membrane
P
cytoskeleton
Adducin Protein 4.1
Spectrin dimer-dimer
Alpha chain Spectrin- Ankyrin interaction
Spectrin Beta chain actin-4.1-adducin
interaction

Figure 1–1. Schematic illustration of red blood cell membrane depicting the composition and arrangement of RBC membrane proteins. GP-A = glycophorin A;
GP-B = glycophorin B; GP-C = glycophorin C; G = globin. Numbers refer to pattern of migration of SDS (sodium dodecyl sulfate) polyacrylamide gel pattern stained with
Coomassie brilliant blue. Relations of protein to each other and to lipids are purely hypothetical; however, the positions of the proteins relative to the inside or outside of the
lipid bilayer are accurate. (Note: Proteins are not drawn to scale and many minor proteins are omitted.) (Reprinted with permission from Harmening, DH: Clinical Hematology
and Fundamentals of Hemostasis, 5th ed., FA Davis, Philadelphia, 2009.)

decrease RBC survival. The RBC membrane is freely per-


meable to water and anions.9 Chloride (Cl–) and bicarbon-
ate (HCO3–) can traverse the membrane in less than a
second. It is speculated that this massive exchange of ions
occurs through a large number of exchange channels
located in the RBC membrane. The RBC membrane is rela-
tively impermeable to cations such as sodium (Na+) and
potassium (K+).
RBC volume and water homeostasis are maintained by
controlling the intracellular concentrations of sodium and
potassium.9 The erythrocyte intracellular-to-extracellular
ratios for Na+ and K+ are 1:12 and 25:1, respectively.6 The
300 cationic pumps, which actively transport Na+ out of
Figure 1–2. Spherocytes. the cell and K+ into the cell, require energy in the form of
ATP. Calcium (Ca2+) is also actively pumped from the inte-
rior of the RBC through energy-dependent calcium-
ATPase pumps.6 Calmodulin, a cytoplasmic calcium-binding
protein, is speculated to control these pumps and to pre-
vent excessive intracellular Ca2+ buildup, which changes
the shape and makes it more rigid.6 When RBCs are
ATP-depleted, Ca2+ and Na+ are allowed to accumulate
intracellularly, and K+ and water are lost, resulting in a
dehydrated rigid cell that is subsequently sequestered by
the spleen, resulting in a decrease in RBC survival.9

Metabolic Pathways

The RBCs’ metabolic pathways that produce ATP are mainly


anaerobic because the function of the RBC is to deliver
oxygen, not to consume it. Because the mature erythrocyte
Figure 1–3. “Bite” cells. has no nucleus and there is no mitochondrial apparatus
6888_Ch01_001-023 29/10/18 4:38 PM Page 6

6 PART I Fundamental Concepts

for oxidative metabolism, energy must be generated almost (2,3-DPG). The amount of 2,3-DPG found within RBCs
exclusively through the breakdown of glucose. has a significant effect on the affinity of hemoglobin
for oxygen and therefore affects how well RBCs function
post-transfusion.
Advanced Concepts
RBC metabolism may be divided into the anaerobic gly- Hemoglobin-Oxygen Dissociation Curve
colytic pathway and three ancillary pathways that serve
to maintain the structure and function of hemoglobin Hemoglobin’s primary function is gas transport: oxygen
(Fig. 1–4): the pentose phosphate pathway, the methemo- delivery to the tissues and carbon dioxide (CO2) excretion.
globin reductase pathway, and the Luebering-Rapoport One of the most important controls of hemoglobin affinity
shunt. All of these processes are essential if the erythrocyte for oxygen is the RBC organic phosphate 2,3-DPG. The
is to transport oxygen and to maintain critical physical unloading of oxygen by hemoglobin is accompanied by
characteristics for its survival. Glycolysis generates about widening of a space between β chains and the binding of
90% of the ATP needed by the RBC. Approximately 10% 2,3-DPG on a mole-for-mole basis, with the formation of
is provided by the pentose phosphate pathway. The methe- anionic salt bridges between the chains.10 The resulting
moglobin reductase pathway is another important pathway conformation of the deoxyhemoglobin molecule is known
of RBC metabolism, and a defect can affect RBC post- as the tense (T) form, which has a lower affinity for oxygen.6
transfusion survival and function. Another pathway that When hemoglobin loads oxygen and becomes oxyhemo-
is crucial to RBC function is the Luebering-Rapoport globin, the established salt bridges are broken, and β chains
shunt. This pathway permits the accumulation of an im- are pulled together, expelling 2,3-DPG. This is the relaxed
portant RBC organic phosphate, 2,3-diphosphoglycerate (R) form of the hemoglobin molecule, which has a higher

PHOSPHOGLUCONATE
PATHWAY
(oxidative)
H2O2
GP
EMBDEN-MEYERHOF
PATHWAY GSH GSSG
(non-oxidative)
Glucose GR
ATP
HK NADP NADPH
ADP
Glucose 6-P 6-P-Gluconate
G-6-PD
GPI 6-PGD CO2
Fructose 6-P Pentose-P
ATP PFK
ADP
Fructose 1,6-diP
METHEMOGLOBIN A
REDUCTASE
PATHWAY Glyceraldehyde DHAP
LUEBERING-RAPAPORT
Hemoglobin NAD PATHWAY
R GAPD
Methemoglobin NADH
HK Hexokinase 1,3-diP-Glycerate DPGM
GPI Glucose-6-phosphate isomerase ADP 2,3-diP-Glycerate
PFK Phosphofructokinase PGK DPGP
ATP
A Aldolase 3-P-Glycerate
TPI Triose phosphate isomerase
GAPD Glyceraldehyde-3-phosphate dehydrogenase PGM
PGM Phosphoglycerate mutase
E Enolase 2-P-Glycerate
PK Pyruvate kinase
LDH Lactic dehydrogenase E
DPGM Diphosphoglyceromutase P-Enolpyruvate
DPGP Diphosphoglycerate phosphatase
ADP
G-6-PD Glucose-6-phosphate dehydrogenase PK
6-PGD 6-Phosphogluconate dehydrogenase ATP
GR Glutathione reductase Pyruvate
GP Glutathione peroxidase NADH
LDH
DHAP Dihydroxyacetone-P NAD
PGK Phosphoglycerate kinase Lactate
R NADH-methemoglobin reductase

Figure 1–4. Red blood cell metabolism. (Reprinted with permission from Hillman, RF, and Finch, CA: Red Cell Manual, 7th ed., FA Davis, Philadelphia, 1996.)
6888_Ch01_001-023 29/10/18 4:38 PM Page 7

Chapter 1 Red Blood Cell and Platelet Preservation: Historical Perspectives and Current Trends 7

affinity for oxygen.6 These allosteric changes that occur as are much less efficient because only 12% of the oxygen
the hemoglobin loads and unloads oxygen are referred to can be released to the tissues.6 Multiple transfusions of
as the respiratory movement. The dissociation and binding 2,3-DPG–depleted stored blood can shift the oxygen dis-
of oxygen by hemoglobin are not directly proportional to sociation curve to the left.10
the partial pressure of oxygen (pO2) in its environment but
instead exhibit a sigmoid-curve relationship, known as the
hemoglobin-oxygen dissociation curve (Fig. 1–5). RBC Preservation
The shape of this curve is very important physiologically
The goal of blood preservation is to provide viable and func-
because it permits a considerable amount of oxygen to be
tional blood components for patients requiring blood trans-
delivered to the tissues with a small drop in oxygen tension.
fusion. RBC viability is a measure of in vivo RBC survival
For example, in the environment of the lungs, where the
following transfusion. Because blood must be stored from the
pO2 tension, measured in millimeters of mercury (mm Hg),
time of donation until the time of transfusion, the viability of
is nearly 100 mm Hg, the hemoglobin molecule is almost
RBCs must be maintained during the storage time as well.
100% saturated with oxygen. As the RBCs travel to the
The U.S. Food and Drug Administration (FDA) requires an
tissues, where the pO2 drops to an average of 40 mm Hg
average 24-hour post-transfusion RBC survival of more than
(mean venous oxygen tension), the hemoglobin saturation
75%.11 In addition, the FDA mandates that red blood cell
drops to approximately 75% saturation, releasing about
integrity be maintained throughout the shelf-life of the stored
25% of the oxygen to the tissues.6 This is the normal
RBCs. This is assessed as free hemoglobin less than 1% of
situation of oxygen delivery at a basal metabolic rate. The
total hemoglobin.11 These two criteria are used to evaluate
normal position of the oxygen dissociation curve depends
new preservation solutions and storage containers. To deter-
on three different ligands normally found within the RBC:
mine post-transfusion RBC survival, RBCs are taken from
H+ ions, CO2, and organic phosphates. Of these three
healthy subjects, stored, and then labeled with radioisotopes,
ligands, 2,3-DPG plays the most important physiological
reinfused to the original donor, and measured 24 hours after
role. Normal hemoglobin function depends on adequate
transfusion. Despite FDA requirements, the 24-hour post-
2,3-DPG levels in the RBC. In situations such as hypoxia,
transfusion RBC survival at outdate can be less than 75% and
a compensatory shift to the right of the hemoglobin-
in critically ill patients is often less than 75%.12,13
oxygen dissociation curve alleviates the tissue oxygen
To maintain optimum viability, blood is stored in the liquid
deficit. This rightward shift of the curve, mediated by in-
state between 1°C and 6°C for a specific number of days, as
creased levels of 2,3-DPG, decreases hemoglobin’s affinity
determined by the preservative solution(s) used. The loss of
for the oxygen molecule and increases oxygen delivery to
RBC viability has been correlated with the storage lesion, which
the tissues. A shift to the left of the hemoglobin-oxygen
is associated with various biochemical changes14 (Table 1–2).
dissociation curve results, conversely, in an increase in
hemoglobin-oxygen affinity and a decrease in oxygen de-
livery to the tissues. With such a dissociation curve, RBCs
Advanced Concepts
Because low 2,3-DPG levels profoundly influence the oxy-
gen dissociation curve of hemoglobin,14 DPG-depleted
100
Normal
90 “Left-shifted”

“Right-shifted”
80 Table 1–2 RBC Storage Lesion
Oxyhemoglobin (% saturation)

↑Abn Hb
70 ↑pH Characteristic Change Observed
↓DPG ↓pH
↓Temp ↑DPG
60 Viable cells (%) Decreased
↓P50 ↑Temp
↑P50
50 Glucose Decreased
P50
40 ATP Decreased

30 Lactic acid Increased

20 pH Decreased

10 2,3-DPG Decreased
Normal P50 = 28 mm Hg
0 Oxygen dissociation curve Shift to the left (increase in
0 10 20 30 40 50 60 70 80 90 100 hemoglobin and oxygen affinity;
less oxygen delivered to tissues)
PO2 (mm Hg)
Plasma K+ Increased
Figure 1–5. Hemoglobin-oxygen dissociation curve. (Reprinted with permission
from Harmening, DH: Clinical Hematology and Fundamentals of Hemostasis, Plasma hemoglobin Increased
5th ed., FA Davis, Philadelphia, 2009.)
6888_Ch01_001-023 29/10/18 4:38 PM Page 8

8 PART I Fundamental Concepts

RBCs may have an impaired capacity to deliver oxygen to


Advanced Concepts
the tissues. As RBCs are stored, 2,3-DPG levels decrease,
with a shift to the left of the hemoglobin-oxygen dissocia- It is interesting to note that blood stored in all CPD preser-
tion curve, and less oxygen is delivered to the tissues. It is vatives also becomes depleted of 2,3-DPG by the second
well accepted, however, that 2,3-DPG is re-formed in stored week of storage. The reported pathophysiological effects of
RBCs after in vivo circulation, resulting in restored oxygen the transfusion of RBCs with low 2,3-DPG levels and
delivery. The rate of restoration of 2,3-DPG is influenced by increased affinity for oxygen include an increase in cardiac
the acid-base status of the recipient, the phosphorus meta- output, a decrease in mixed venous (pO2) tension, or a
bolism, the degree of anemia, and the overall severity of the combination of these.18 The physiological importance of
disorder.6 It has been reported that within the first hour after these effects is not easily demonstrated. This is a complex
transfusion, most RBC clearance occurs.13 Approximately mechanism with numerous variables involved that are
220 to 250 mg of iron are contained in one RBC unit.15 beyond the scope of this text.
Therefore, rapid RBC clearance of even 25% of a single unit Stored RBCs regain the ability to synthesize 2,3-DPG after
of blood delivers a massive load of hemoglobin iron to the transfusion, but levels necessary for optimal hemoglobin-
monocyte and macrophage system, potentially producing oxygen delivery are not reached immediately. Approxi-
harmful effects.12 mately 24 hours are required to restore normal levels of
Despite the biochemical, structural, and functional 2,3-DPG after transfusion.18 The 2,3-DPG concentrations
changes that occur to RBCs during storage, there is no after transfusion have been reported to reach normal levels
significant difference in rates of death between patients as early as 6 hours post-transfusion.18 Most of these
who were transfused with only fresh blood versus studies have been performed on normal, healthy individu-
those patients who were transfused with the oldest blood als. However, evidence suggests that, in the transfused
available.16 subject whose capacity is limited by an underlying physio-
logical disturbance, even a brief period of altered oxygen
hemoglobin affinity is of great significance.12 It is quite
Anticoagulant Preservative Solutions clear now that 2,3-DPG levels in transfused blood are
important in certain clinical conditions. Some studies
Table 1–3 lists the approved anticoagulant preservative demonstrate that myocardial function improves following
solutions for whole blood and RBC storage at 1°C to 6°C. transfusion of blood with high 2,3-DPG levels during
The addition of various chemicals, along with the approved cardiovascular surgery.18 Several investigators suggest that
anticoagulant preservative CPD, was incorporated in an the patient in shock who is given 2,3-DPG–depleted eryth-
attempt to stimulate glycolysis so that ATP levels were better rocytes in transfusion may have already strained the com-
maintained.17 One of the chemicals, adenine, incorporated pensatory mechanisms to their limits.18,19–21 Perhaps for
into the CPD solution (CPDA-1) increases ADP levels, this type of patient, the poor oxygen delivery capacity of
thereby driving glycolysis toward the synthesis of ATP. 2,3-DPG–depleted cells makes a significant difference in
CPDA-1 contains 0.25 mM of adenine plus 25% more recovery and survival.
glucose than CPD. Adenine-supplemented blood can be It is apparent that many factors may limit the viability
stored at 1°C to 6°C for 35 days; the other anticoagulants are of transfused RBCs. One of these factors is the plastic
approved for 21 days. Table 1–4 lists the various chemicals material used for the storage container. The plastic must
used in anticoagulant solutions and their functions during be sufficiently permeable to CO2 in order to maintain
the storage of red blood cells. higher pH levels during storage. Glass storage containers
are no longer used in the United States. Currently, the
majority of blood is stored in polyvinyl chloride (PVC)
Table 1–3 Approved Anticoagulant plastic bags. One issue associated with PVC bags relates
Preservative Solutions to the plasticizer di(ethylhexyl)-phthalate (DEHP), which
is used in the manufacture of the bags. It has been found
Storage to leach from the plastic into the lipids of the plasma
Name Abbreviation Time (Days) medium and RBC membranes of the blood during storage.
Acid citrate-dextrose ACD-A 21 However, its use or that of alternative plasticizers that
(formula A)* leach are important because they have been shown to sta-
bilize the RBC membrane and therefore reduce the extent
Citrate-phosphate CPD 21
dextrose of hemolysis during storage. Another issue with PVC is
its tendency to break at low temperatures; therefore, com-
Citrate-phosphate- CP2D 21 ponents frozen in PVC bags must be handled with care.
double-dextrose In addition to PVC, polyolefin containers, which do not
Citrate-phosphate- CPDA-1 35 contain DEHP, are available for some components, and
dextrose-adenine latex-free plastic containers are available for recipients
with latex allergies.5
*ACD-A is used for apheresis components.
6888_Ch01_001-023 29/10/18 4:38 PM Page 9

Chapter 1 Red Blood Cell and Platelet Preservation: Historical Perspectives and Current Trends 9

Table 1–4 Chemicals in Anticoagulant Solutions


Chemical Function Present In
ACD-A CPD CP2D CPDA-1

Citrate (sodium citrate/citric acid) Chelates calcium; prevents clotting. X X X X

Monobasic sodium phosphate Maintains pH during storage; necessary for maintenance X X X X


of adequate levels of 2,3-DPG.

Dextrose Substrate for ATP production (cellular energy). X X X X

Adenine Production of ATP (extends shelf-life from 21 to 35 days). X

ACD-A = acid citrate-dextrose (formula A); CPD = citrate-phosphate dextrose; CP2D = citrate phosphate double dextrose; CPDA-1 = citrate-phosphate-dextrose-adenine;
2,3-DPG = 2,3-diphosphoglycerate; ATP = adenosine triphosphate

Additive Solutions The additive solution is contained in a satellite bag and


is added to the RBCs after most of the plasma has been ex-
Additive solutions (AS) are preserving solutions that are added pressed. All three additives contain saline, adenine, and glu-
to the RBCs after removal of the plasma with or without cose. AS-1, AS-5, and AS-7 also contain mannitol, which
platelets. Additive solutions are now widely used. One of the protects against storage-related hemolysis, whereas AS-3
reasons for their development is that removal of the plasma contains citrate and phosphate for the same purpose.22 All
component during the preparation of packed RBCs removed of the additive solutions are approved for 42 days of storage
much of the nutrients needed to maintain RBCs during storage. for packed RBCs.22 Table 1–5 lists the currently approved
This was dramatically observed when high-hematocrit RBCs additive solutions.
were prepared. The influence of removing substantial amounts
of adenine and glucose present originally in, for example, the
CPDA-1 anticoagulant preservative solution, led to a decrease Advanced Concepts
in viability, particularly in the last 2 weeks of storage.16
Table 1–6 shows the biochemical characteristics of RBCs
Packed RBCs prepared from whole blood units collected
stored in the additive solutions after 42 days of stor-
in primary anticoagulant preservative solutions can be rela-
age.22,23,24 Additive system RBCs are used in the same way
tively void of plasma with high hematocrits, which causes
as traditional RBC transfusions. Blood stored in additive
the units to be more viscous and difficult to infuse, especially
solutions is now routinely given to newborn infants and
in emergency situations. Additive solutions (100 mL to the
pediatric patients, although some clinicians still prefer
packed RBCs prepared from a 450-mL blood collection) also
CPDA-1 RBCs because of their concerns about one or more
overcome this problem. Additive solutions reduce hemato-
of the constituents in the additive solutions.25
crits from around 65% to 80% to around 55% to 65% with a
None of the additive solutions maintain 2,3-DPG
volume of approximately 300 to 400 mL.22 The ability to
throughout the storage time. As with RBCs stored only with
pack RBCs to fairly high hematocrits before adding additive
primary anticoagulant preservatives, 2,3-DPG is depleted
solution also provides a means to harvest greater amounts
by the second week of storage.23
of plasma with or without platelets. Box 1–2 summarizes the
benefits of RBC additive solutions.
Currently, four additive solutions are licensed in the
United States:
1. Adsol (AS-1; Fenwal Inc.) Table 1–5 Additive Solutions in Use
2. Nutricel (AS-3; Haemonetics Corporation) in North America
3. Optisol (AS-5; Terumo Corporation)
4. SOLX (AS-7; Haemonetics Corporation) Storage
Name Abbreviation Time (Days)
Adsol (Fenwal AS-1 42
Inc.)
BOX 1–2
Nutricel (Haemonetics AS-3 42
Benefits of RBC Additive Solutions Corporation)
• Extends the shelf-life of RBCs to 42 days by adding nutrients
Optisol (Terumo AS-5 42
• Allows for the harvesting of more plasma and platelets from Corporation)
the unit
• Produces a packed RBC of lower viscosity that is easier to infuse SOLX (Haemonetics) AS-7 42
6888_Ch01_001-023 29/10/18 4:38 PM Page 10

10 PART I Fundamental Concepts

Table 1–6 Red Blood Cell Additives: Biochemical Characteristics


AS-1 AS-3 AS-5 AS-7
Storage period (days) 42 42 42 42

pH (measured at 37°C) 6.6 6.5 6.5 6.6

24-hour survival*(%) 83 85.1 80 80

ATP (% initial) 68 67 68.5 91%

2,3-DPG (% initial) 6 6 5 **1.5 µmol/L/g Hb

Hemolysis (%) 0.5 0.7 0.6 0.3

*Survival studies reported are from selected investigators and do not include an average of all reported survivals.
**Reported by researchers using different units

Freezing and Rejuvenation Currently, the FDA licenses frozen RBCs for a period of
10 years from the date of freezing; that is, frozen RBCs may
RBC Freezing be stored up to 10 years before thawing and transfusion.26
RBC freezing is primarily used for autologous units and the Once thawed, these RBCs demonstrate function and viability
storage of rare blood types. Autologous transfusion (auto near those of fresh blood. Experience has shown that 10-year
meaning “self”) allows individuals to donate blood for storage periods do not adversely affect viability and
their own use to meet their needs for blood transfusion function.27 Table 1–8 lists the advantages and disadvantages
(see Chapter 16, “Transfusion Therapy”). of RBC freezing.
The procedure for freezing a unit of packed RBCs is not
complicated. It involves the addition of a cryoprotective agent
to RBCs that are less than 6 days old. Glycerol is used most Advanced Concepts
commonly and is added to the RBCs slowly with vigorous Transfusion of frozen cells must be preceded by a deglyc-
shaking, thereby enabling the glycerol to permeate the RBCs. erolization process; otherwise, the thawed cells would be
The cells are then rapidly frozen and stored in a freezer. The accompanied by hypertonic glycerol when infused, and
usual storage temperature is below –65°C, although storage RBC lysis would result. Removal of glycerol is achieved by
(and freezing) temperature depends on the concentration systematically replacing the cryoprotectant with decreasing
of glycerol used.23 Two concentrations of glycerol have concentrations of saline. The usual protocol involves wash-
been used to freeze RBCs: a high-concentration glycerol ing with 12% saline, followed by 1.6% saline, with a final
(40% weight in volume [wt/vol]) and a low-concentration wash of 0.2% dextrose in normal saline.5 A commercially
glycerol (20% wt/vol) in the final concentration of the cryo- available cell-washing system, such as those manufactured
preservative.23 Most blood banks that freeze RBCs use the by several companies, has traditionally been used in the
high-concentration glycerol technique. deglycerolizing process. Excessive hemolysis is monitored
Table 1–7 lists the advantages of the high-concentration by noting the hemoglobin concentration of the wash su-
glycerol technique in comparison with the low-concentration pernatant. Osmolality of the unit should also be monitored
glycerol technique. See Chapter 15 for a detailed to ensure adequate deglycerolization. Traditionally, because
description of the RBC freezing procedure.

Table 1–7 Advantages of High-Concentration Glycerol Technique Over Low-Concentration


Glycerol Technique
Advantage High Glycerol Low Glycerol
1. Initial freezing temperature –80°C –196°C

2. Need to control freezing rate No Yes

3. Type of freezer Mechanical Liquid nitrogen

4. Maximum storage temperature –65°C –120°C

5. Shipping requirements Dry ice Liquid nitrogen

6. Effect of changes in storage temperature Can be thawed and refrozen Critical


6888_Ch01_001-023 29/10/18 4:38 PM Page 11

Chapter 1 Red Blood Cell and Platelet Preservation: Historical Perspectives and Current Trends 11

Table 1–8 Advantages and Disadvantages 3. Development of procedures to convert A, B, and AB type
of RBC Freezing RBCs to O type RBCs
4. Development of methods to produce RBCs through
Advantages Disadvantages bioengineering (blood pharming)
Long-term storage (10 years) A time-consuming process 5. Development of RBC substitutes

Maintenance of RBC viability Higher cost of equipment and


and function materials Improved Additive Solutions
Low residual leukocytes and Storage requirements (–65°C) Research is being conducted to develop improved additive
platelets
solutions for RBC preservation. One reason for this is
Removal of significant amounts Higher cost of product because longer storage periods could improve the logistics
of plasma proteins of providing RBCs for clinical use.

Procedures to Reduce and Inactivate Pathogens

Research is being conducted to develop procedures that


a unit of blood is processed in an open system (one in
would reduce the level of or inactivate residual viruses, bac-
which sterility is broken) to add the glycerol (before freezing)
teria, and parasites in RBC units. One objective is to develop
or the saline solutions (for deglycerolization), the outdating
robust procedures that could possibly inactivate all
period of thawed RBCs stored at 1°C to 6°C has been
pathogens that may be present, including new and emergent
24 hours.23 Generally, RBCs in CPD or CPDA-1 anticoag-
viruses. Amustaline (S-303) pathogen reduction system is
ulant preservatives or additive solutions are glycerolized
currently being studied and has demonstrated adequate post-
and frozen within 6 days of whole blood collection.23
Closed-system devices have been developed that allow transfusion viability according to FDA criteria.28 This nucleic
acid-targeted pathogen inactivation technology was devel-
the glycerolization and deglycerolization processes to be
oped to reduce the risk of transfusion-transmitted infectious
performed under sterile conditions.27 RBCs prepared from
disease with RBC transfusions.29 Areas of concern that must
450-mL collections and frozen within 6 days of blood col-
be addressed before pathogen inactivation technologies are
lection with CPDA-1 can be stored after thawing at 1°C to
approved for use with RBCs in the United States are potential
6°C for up to two weeks when prepared in a closed
toxicity, immunogenicity, cellular function, and cost. Cur-
system.23
rently, the FDA has not approved any Pathogen Reduction
Technology (PRT) for use with RBCs.29
RBC Rejuvenation
Rejuvenation of RBCs is the process by which ATP and 2,3- Formation of O Type RBCs
DPG levels are restored or enhanced by metabolic alter-
ations. Currently, FDA-approved rejuvenation solution The inadequate supply of O type RBC units that is periodi-
cally encountered can hinder blood centers and hospital
contains phosphate, inosine, and adenine.22 Rejuvenated
blood banks in providing RBCs for specific patients. Re-
RBCs may be prepared up to three days after expiration
search over the last 30 years has been evaluating how A and
when stored in CPD, CPDA-1, and AS-1 storage solu-
B type RBCs can be converted to O type RBCs, the universal
tions.22 Currently, rejuvenated RBCs must be washed be-
donor.30 The use of enzymes that remove the carbohydrate
fore infusion to remove the inosine (which may be toxic)
moieties of the A and B antigens is the mechanism for form-
and transfused within 24 hours or frozen for long-term
ing O type RBCs.30 The enzymes are removed by washing
storage.22 The rejuvenation process is expensive and time-
after completion of the reaction time.
consuming, thus it is not used often; however, the process
is invaluable for preserving selected autologous and rare
units of blood for later use. Blood Pharming

Creating RBCs in the laboratory (blood pharming) is an-


Current Trends in RBC Preservation other area of research that has the potential to increase the
Research amount of blood available for transfusion. In 2008, the De-
fense Advanced Research Projects Agency (DARPA) awarded
Arteriocyte, a bioengineering company, a contract to develop
Advanced Concepts
a system for producing O-negative RBCs on the battlefield.31
Research and development in RBC preparation and preser- The company, which uses proprietary technology (NANEX)
vation is being pursued in five areas: to turn hematopoietic stem cells (HSCs) from umbilical
1. Development of improved additive solutions cords into type O, Rh-negative RBCs, sent its first shipment
2. Development of procedures to reduce and inactivate the of the engineered blood to the FDA for evaluation in 2010.31
level of pathogens that may be in RBC units FDA approval is required before human trials can begin.
Cultured RBCs generated from in vitro hematopoietic stem
6888_Ch01_001-023 29/10/18 4:38 PM Page 12

12 PART I Fundamental Concepts

cells has been reported as well.32 However, this has not Table 1–9 Phases of Testing
proven practical for routine transfusion. The challenges
associated with blood pharming are scalability or large-scale Phase Description of Testing
production and cost-effectiveness. Preclinical In vivo and animal testing.

RBC Substitutes Phase I Researchers test drug in a small group of people


(20 to 80) for the first time to evaluate its safety,
Scientists have been searching for a substitute for blood for determine a safe dosage range, and identify side
effects.
over 150 years. Blood substitutes continue to be of interest
because of their potential to alleviate shortages of donated Phase II The drug is given to a larger group of people
blood. In the 1980s, safety concerns about HIV led to re- (100 to 300) to see if it is effective and to further
newed interest in finding a substitute for human blood; and evaluate its safety.
more recently, the need for blood on remote battlefields has Phase III The drug is given to large groups of people
heightened that interest.33 The U.S. military is one of the (1,000 to 3,000) to confirm its effectiveness, monitor
strongest advocates for the development of blood substitutes, side effects, compare it to commonly used treat-
which it supports through its own research and partnerships ments, and collect information that will allow the
drug to be used safely.
with private-sector companies.33 Today the search continues
for a safe and effective oxygen carrier that could eliminate Phase IV Postmarketing studies to gather additional informa-
many of the problems associated with blood transfusion, such tion about the drug’s risks, benefits, and optimal use.
as the need for refrigeration, limited shelf-life, compatibility,
immunogenicity, transmission of infectious agents, and short-
ages. Box 1–3 lists the potential benefits of artificial oxygen
carriers. Since RBC substitutes are drugs, they must go Hemoglobin-Based Oxygen Carriers
through extensive testing in order to obtain FDA approval. HBOC commercial development focused on “oxygen thera-
Safety and efficacy must be demonstrated through clinical peutic” indications to provide immediate oxygenation until
trials. Table 1–9 outlines the different phases of testing. medical or surgical interventions could be initiated. Early
Current research on blood substitutes is focused on two trauma trials with HemAssist® (BAXTER), Hemopure®
areas: hemoglobin-based oxygen carriers (HBOCs) and per- (HbO2Therapeutics), and PolyHeme® (NORTHFIELD
fluorocarbons (PFCs).34,35 Originally developed to be used Laboratories) for resuscitating hypotensive shock all failed
in trauma situations such as accidents, combat, and surgery, due to the safety concerns of cardiac issues and increased
RBC substitutes have, until recently, fallen short of meeting mortality.
requirements for these applications.34 Despite years of re- Although several HBOCs have progressed to phase II and
search, RBC substitutes are still not in routine use today. III clinical trials, currently none have been approved for
South Africa, Mexico, and Russia are the only countries clinical use in humans in the United States.36,37 A 2008 meta-
in which blood substitutes are approved for clinical use. analysis of 16 clinical trials involving 3,711 patients and five
None have received FDA approval for clinical use in the different HBOCs found a significantly increased risk of death
United States, although specific products are still in phase and myocardial infarction associated with the use of
III clinical trials. HBOCs.37 As a result, in 2008, the Food and Drug Adminis-
tration (FDA) put all HBOC trials in the United States on
clinical hold due to the unfavorable outcomes.35 However,
BOX 1–3 Hemopure (HBOC-201) and PolyHeme are still in phase III
Potential Benefits of Artificial Oxygen Carriers clinical trials in the United States and Europe.38 Hemopure
was approved for clinical use in South Africa in 2001 to treat
• Abundant supply
adult surgical patients who are anemic, and in Russia for
• Readily available for use in prehospital settings, battlefields, and
remote locations
acute anemia.34 Many HBOCs have been researched; how-
• Can be stockpiled for emergencies and warfare
ever, the majority have been discontinued due to complica-
• No need for typing and crossmatching
tions of cardiac toxicity, gastrointestinal distress, neurotoxicity,
• Available for immediate infusion
renal failure, and increased mortality.34 Table 1–10 summa-
• Extended shelf-life (1 to 3 years)
rizes some of the many HBOCs developed.
• Can be stored at room temperature
However, some experts believe that HBOCs hold more
• Free of bloodborne pathogens
promise than PFCs.33,39
• At full oxygen capacity immediately
Table 1–11 lists the advantages and disadvantages of
• Do not prime circulating neutrophils, reducing the incidence of
HBOCs.
multiorgan failure
Perfluorocarbons
• Can deliver oxygen to tissue that is inaccessible to RBCs
• Have been accepted by Jehovah’s Witnesses Perfluorocarbons are synthetic hydrocarbon structures in
• Could eventually cost less than units of blood which all hydrogen atoms have been replaced with fluorine.
They are chemically inert, are excellent gas solvents, and
6888_Ch01_001-023 29/10/18 4:38 PM Page 13

Chapter 1 Red Blood Cell and Platelet Preservation: Historical Perspectives and Current Trends 13

Table 1–10 Hemoglobin-Based Oxygen Carriers


Product Manufacturer Chemistry/Source History/Status
HemAssist (DCLHb) Baxter Diaspirin cross-linked Hgb from First HBOC to advance to phase III clini-
outdated human RBCs cal trials in United States. Removed
from production because of increased
mortality rates.

PolyHeme (SFH-P) Northfield Laboratories Polymerized and pyridoxalated Underwent phase II/III clinical trials in
human Hgb United States. Did not obtain FDA
approval.

Hemopure (HBOC-201) HbO2 Polymerized bovine Hgb Still in phase II/III clinical trials in
[hemoglobin glutamer – 250 (bovine)] United States and Europe. Approved
Therapeutics
for use in South Africa (2001) to
treat adult surgical patients who
are anemic, and in Russia for acute
anemia.

Oxyglobin HbO2 Polymerized bovine Hgb Approved by the FDA and the European
Medicines Agency (EMA) to treat
Therapeutics
canine anemia in veterinary use.

MP4OX Sangart Polyethylene glycol (PEG) attached to In phase II trials in United States;
the surface of Hgb from human RBCs phase III in Europe.
Hemospan (MP4)
Terminated development and opera-
tions in December 2013.

HemoLink Hemosol Purified human Hgb from outdated Abandoned due to cardiac toxicity.
RBCs, cross-linked and polymerized

HemoTech HemoBioTech Derived from bovine Hgb Limited clinical trial outside the United
States.

Table 1–11 Advantages and Disadvantages treatment of traumatic brain injury in Switzerand and
Israel.39 Refer to Table 1–12 for further details and review of
of Hemoglobin-Based Oxygen
PFCs, and Table 1–13 for the advantages and disadvantages
Carriers
of perfluorochemicals.
Advantages Disadvantages
Tissue Engineering of RBCs
Long shelf-life Short intravascular half-life

Very stable Possible toxicity Research into large-scale production of RBCs from stem cells
(blood pharming) seems to have more promise and is receiving
No antigenicity (unless bovine) Increased O2 affinity more attention and funding than are blood substitutes. RBCs
No requirement for blood typing Increased oncotic effect have been cultured in-vitro for many years and have been suc-
procedures cessfully tested in animal models. However, there are limita-
tions in the number of RBCs that can be cultured from one unit
of blood and the associated costs of these expensive cultures.
By culturing stem cells in the presence of the essential
carry O2 and CO2 by dissolving them. Because of their small cytokines, stem cell factor, and erythropoietin, unilineage
size (about 0.2 µm in diameter), they are able to pass production of erythroblasts has been achieved.39 Culture of
through areas of vasoconstriction and deliver oxygen to tis- cells in expansion medium and subsequently in maturation
sues that are inaccessible to RBCs.39 PFCs have been under medium has shown progress of in-vitro erythroid expansion.39
investigation as possible RBC substitutes since the 1970s. The general consensus for producing RBCs in-vitro is a precul-
Fluosol (Green Cross Corp.) was approved by the FDA in ture of hematopoietic stem cells (HSC) for erythroid progenitor
1989 but was removed from the market in 1994 due to clini- cells, with a subsequent generation of high numbers of ery-
cal shortcomings and poor sales. Other PFCs have proceeded throblasts. This is followed by a erythroid maturation phase in
to clinical trials. Perftoran West Ltd is in clinical use in the presence of a feeder layer to facilitate progression to a ma-
Russia and Mexico.39 Two others are no longer under devel- ture RBC.39 Maturation without a feeder layer has also been
opment, and one (Oxycyte, Oxygen Biotherapeutics Inc.) is reported and this development is necessary if tissue engineer-
currently being investigated as an oxygen therapeutic for ing RBCs is to become used for transfusion therapy.39
6888_Ch01_001-023 29/10/18 4:38 PM Page 14

14 PART I Fundamental Concepts

Table 1–12 Perfluorocarbons


Fluosol-DA Green Cross Corporation of Japan The first and only oxygen-carrying blood substitute ever to receive approval
from the FDA for human clinical use in the United States. Approved in 1989;
discontinued in 1994 because of clinical shortcomings and poor sales.

Oxygent Alliance Pharmaceutical Corporation Phase III trial in Europe completed; phase III trial in United States terminated
due to adverse effects. Development stopped due to lack of funding.

Oxycyte Originally Synthetic Blood International; Shift in research from use as RBC substitute to other medical applications. Cur-
name changed to Oxygen Biotherapeutics rently in phase II trials in Switzerland for treatment of traumatic brain injury.
in 2008

Perftoran Perftoran Approved for use in Russia and Mexico.

PHER-O2 Sanguine Corporation Under evaluation for transfusion, therapy for heart attack and stroke.

Table 1–13 Advantages and Disadvantages Maintaining pH was determined to be a key parameter for
retaining platelet viability in vivo when platelets were stored
of Perfluorochemicals
at 20°C to 24°C.41 The loss of platelet quality during storage
Advantages Disadvantages is known as the platelet storage lesion. During storage, a
varying degree of platelet activation occurs that results in
Biological inertness Adverse clinical effects
release of some intracellular granules and a decline in ATP
Lack of immunogenicity High O2 affinity and ADP. 41
The reduced oxygen tension (pO2) in the plastic platelet
Easily synthesized Retention in tissues
storage container results in an increase in the rate of glycol-
Requirement for O2 administration ysis by platelets to compensate for the decrease in ATP re-
when infused generation from the oxidative (TCA) metabolism. This
Deep-freeze storage temperatures increases glucose consumption and causes an increase in lac-
tic acid that must be buffered. This results in a fall in pH.
During the storage of platelet concentrates (PCs) in plasma,
the principal buffer is bicarbonate. When the bicarbonate
Platelet Preservation
buffers are depleted during platelet concentrate storage, the
Approximately 2.4 million platelet units are distributed and pH rapidly falls to less than 6.2, which is associated with a
2.2 million platelet transfusions are administered yearly in loss of platelet viability. In addition, when pH falls below 6.2,
the United States.3 Platelets are involved in the blood coag- the platelets swell and there is a disk-to-sphere transforma-
ulation process and are given to treat or prevent bleeding. tion in morphology that is associated with a loss of mem-
They are given either therapeutically to stop bleeding or pro- brane integrity.40 The platelets then become irreversibly
phylactically to prevent bleeding. Better availability and swollen, aggregate together, or lyse, and when infused, will
management of platelet inventory has been a goal of blood not circulate or function. This change is irreversible when
banks for many years. The financial impact of outdated and the pH falls to less than 6.2.40 During storage of platelet con-
returned platelet units is the primary reason to find a way to centrates, the pH will remain stable as long as the production
improve inventory management. Increasing the storage time of lactic acid does not exceed the buffering capacity of the
during platelet preservation is one way to reduce the number plasma or other storage solution. Table 1–14 summarizes
of outdated platelet units. With the limit of five days of stor- platelet changes during storage (the platelet storage lesion).
age for platelet concentrates, approximately 20% to 30% of It should be noted that except for change in pH, the effect of
the platelet inventory is discarded either by the blood sup- in vitro changes on post-transfusion platelet survival and
plier or the hospital blood bank.4 function is unknown, and some of the changes may be re-
versible upon transfusion.42
The Platelet Storage Lesion Generally, the quality-control measurements required by
various accreditation organizations for platelet concentrates
Platelet storage still presents one of the major challenges to include platelet concentrate volume, platelet count, pH of the
the blood bank because of the limitations of storing platelets. unit, and residual leukocyte count if claims of leukoreduction
In the United States, platelets are stored at 20°C to 24°C with are made.43 In addition, immediately before distribution to
maintaining continuous gentle agitation throughout the stor- hospitals, a visual inspection is made that often includes an
age period of 5 days. Agitation has been shown to facilitate assessment of platelet swirl (no visible aggregation).43 The
oxygen transfer into the platelet bag and oxygen consump- absence of platelet swirling is associated with the loss of
tion by the platelets. The positive role for oxygen has been membrane integrity during storage, resulting in the loss of
associated with the maintenance of platelet component pH.40 discoid shape with irreversible sphering.44 Box 1–4 lists the
6888_Ch01_001-023 29/10/18 4:38 PM Page 15

Chapter 1 Red Blood Cell and Platelet Preservation: Historical Perspectives and Current Trends 15

Table 1–14 The Platelet Storage Lesion apheresis (apheresis platelets). Currently, greater than 92%
of platelet transfusions are from apheresed platelets and
Characteristic Change Observed about 8% are pools of whole blood-derived platelets (WBD).3
Lactate Increased Platelets still remain the primary means of treating throm-
bocytopenia, even though therapeutic responsiveness varies
pH Decreased according to patient status and platelet storage conditions.43
ATP Decreased (See Chapter 15 for the methods for preparing platelet con-
centrates.) One unit of whole blood-derived platelet concen-
Morphology scores change Decreased trate contains ≥5.5 × 1010 platelets suspended in 40 to
from discoid to spherical 70 mL of plasma.45 These platelets may be provided as a
(loss of swirling effect)
single unit or as pooled units; however, pooled units only
Degranulation Increased have a shelf life of 4 hours. Apheresis platelets contain
((β-thromboglobulin, ≥3.0 × 1011 in one unit which is the therapeutic equivalent
platelet factor 4) of 4 to 6 units of whole blood-derived platelets.45 There are
Platelet activation markers Increased a number of containers used for 5-day storage of whole
(P-selectin [CD62P] or CD63) blood–derived (WBD) and apheresis platelets. Box 1-5
lists the factors to be considered when using 5-day plastic
Platelet aggregation Drop in responses to some agonists
storage bags.
Additive solutions may be used for storage of apheresis
platelets. In the United States, platelets are being stored in a
100% plasma medium, unless a platelet additive solution is
BOX 1–4
used. Two platelet additive solutions (PAS) are FDA ap-
In Vitro Platelet Assays Correlated With proved, PAS-C (Intersol) and PAS-F (Isoplate), for the storage
In Vivo Survival of apheresis platelets for 5 days.46 The PASs are designed to
support platelets during storage in reduced amounts of resid-
• pH
ual plasma. With the addition of a PAS, residual plasma is
• Shape change
reduced to 35% with both InterSol and Isoplate.46 One
• Hypotonic shock response
advantage is that this approach provides more plasma for
• Lactate production
fractionation. In addition, there are data indicating that
• pO2
optimal additive solutions may improve the quality of
platelets during storage, reduce adverse effects associated
with transfusion of plasma, and promote earlier detection of
in vitro platelet assays that have been correlated with in vivo bacteria.46 Box 1–6 lists the advantages of using a platelet
survival. additive solution for platelet storage.
Clinical Use of Platelets Platelet Testing and Quality Control Monitoring
Platelet components are effectively used to treat bleeding as- For component testing, the FDA Guidance document rec-
sociated with thrombocytopenia, a marked decrease in ommends: 1) Actual platelet yield (volume × platelet count)
platelet number, or dysfunctional platelets. That is, platelets must be determined after each platelet collection; 2) Weight/
are transfused when there is a quantitative or qualitative de- volume conversion is necessary to determine the volume
fect with the patient’s platelets. The efficacy of the platelet of each platelet collection; and 3) Bacterial contamination
transfusion is usually estimated from the corrected count
increment (CCI) of platelets measured after transfusion.22
The corrected count increment (CCI) is a calculated measure
of patient response to platelet transfusion that adjusts for the BOX 1–5
number of platelets infused and the size of the recipient,
based upon body surface area (BSA).22 Factors to Be Considered When Using 5-Day Plastic
Storage Bags
CCI = (postcount – precount) × BSA/platelets transfused
• Temperature control of 20°C to 24°C is critical during platelet
where postcount and precount are platelet counts (/µL) after preparation and storage.
and before transfusion, respectively. BSA is the patient body • Careful handling of plastic bags during expression of platelet-poor
surface area (meter2) and platelets transfused is the number plasma helps prevent the platelet button from being distributed
of administered platelets (× 1011).22 The CCI is usually de- and prevents removal of excess platelets with the platelet-poor
plasma.
termined 10 to 60 minutes after transfusion. It should be
• Residual plasma volumes recommended for the storage of platelet
noted that the CCI does not evaluate or assess function of concentrates from whole blood (45 to 65 mL).
the transfused platelets.23 • For apheresis platelets, the surface area of the storage bags needs
Today, platelets are prepared as concentrates from whole to allow for the number of platelets that will be stored.
blood (whole blood-derived platelet concentrates) and by
6888_Ch01_001-023 29/10/18 4:38 PM Page 16

16 PART I Fundamental Concepts

BOX 1–6 Table 1–15 Performance Criteria for


Platelet Concentrate Collections
Advantages of Using Platelet Additive Solutions
Test Recommended Result
• Optimizes platelet storage in vitro
• Saves plasma for other purposes (e.g., transfusion or fractionation) Actual platelet yield of ≥ 3.0 × 1011
• Facilitates ABO-incompatible platelet transfusions transfusable component
• Reduces plasma-associated transfusion side effects, such as febrile pH ≥ 6.2
and allergic reactions, and may reduce risk of transfusion-related
acute lung injury (TRALI) Percent component ≥ 85% component retention if
• Improves effectiveness of photochemical pathogen reduction retention performed*
technologies
Residual WBC Count** Single collection: < 5.0 × 106
• Potentially improves bacterial detection
Double collection: Collection: < 8.0 × 106
or Components: < 5.0 × 106
Triple collection: Collection: < 1.2 × 107
or Components: < 5.0 × 106
testing as specified by the storage container manufacturer.45
In terms of Quality Control (QC), the FDA recommends *Or per the container/automated blood cell separator device manufacturer’s
the following as a part of your QC: 1) “define a plan for non- specifications
selectively identifying collections to be tested. This should **The stratified recommended results should ensure that the individual transfusable
units will be < 5.0 × 106 even with a 25% error in equilibration of the volume for
ensure testing of components collected on each individual double and triple collections.
automated blood cell separator device, each collection type, Modified from: Guidance for Industry and FDA Review Staff: Collection of Platelets
and each location.”45 2) “define sampling schemes for actual by Automated Methods (December 2007), available at: https://www.fda.gov/
platelet yield (including volume determination) and pH, downloads/BiologicsBloodVaccines/GuidanceComplianceRegulatoryIn formation/
Guidances/Blood/UCM062946.pdf
and residual WBC.” (The platelet yield of the collection and
designation of single, double, or triple PC should be made
prior to performing the residual WBC count QC.) 3) “test
actual platelet yield (platelet count times the volume) and to 24°C has been associated with the retention of post-
pH at the maximum allowable storage time for the container transfusion platelet viability and has been the key issue that
system used (or representing the dating period).” “In addi- has been addressed to improve conditions for storage at this
tion, actual platelet yield and pH testing may be conducted temperature.47
on one storage container of a double or triple collection.” Retaining platelet function during storage is also an issue.
4) “include the residual WBC count for leukocyte-reduced Function is defined as the ability of viable platelets to
collections, if manufacturing leukocyte-reduced products.” respond to vascular damage in promoting hemostasis.
5) “describe the criteria for investigation of failures during
QC, and have a method to document all calculations and test Platelet Storage and Bacterial Contamination
results.”45
Table 1-15 lists the Performance Criteria for Platelet Con- The major concern associated with storage of platelets at
centrate Collections. 20°C to 24°C is the potential for bacterial growth if the pre-
pared platelets contain bacteria because of contamination at
Measurement of Viability and Functional the phlebotomy site or if the donor has an unrecognized
Properties of Stored Platelets bacterial infection.48 Environmental contamination during
processing and storage is another potential, though less com-
Viability indicates the capacity of platelets to circulate after mon, source of bacteria. Room temperature storage and the
infusion without premature removal or destruction. Platelets presence of oxygen provide a good environment for bacterial
have a life span of 8 to 10 days after release from megakaryo- proliferation. Sepsis due to contaminated platelets is the
cytes.6 Storage causes a reduction in this parameter, even most common infectious complication of transfusion.49
when pH is maintained. Viability of stored platelets is deter- According to the FDA, bacterial contamination was the third
mined by measuring pretransfusion and post-transfusion leading cause of transfusion-associated fatality in 2015.50 It
platelet counts and expressing the difference based on the is estimated that 1:2,000 to 1:5,000 units of platelets are con-
number of platelets transfused (CCI). taminated with bacteria.51 An estimated 10% of patients
The observation of the swirling phenomenon (absence transfused with a bacterially contaminated platelet unit
of aggregation) caused by discoid platelets when placed in develop life-threatening sepsis which is often fatal.51 In 2002,
front of a light source has been used to obtain a semiqual- the College of American Pathologists added a requirement
itative evaluation of the retention of platelet viability prop- that laboratories have a method to screen platelets for
erties in stored units.43 The extent of shape change and the bacterial contamination, and the AABB introduced a similar
hypotonic shock response in in vitro tests appears to pro- requirement in 2004. Blood establishments and transfusion
vide some indication about the retention of platelet viability services must assure that the risk of bacterial contamination
properties.43 The maintenance of pH during storage at 20°C of platelets is adequately controlled using FDA approved
6888_Ch01_001-023 29/10/18 4:38 PM Page 17

Chapter 1 Red Blood Cell and Platelet Preservation: Historical Perspectives and Current Trends 17

or cleared devices or other adequate and appropriate meth- transfusion but reduces the shelf-life of the platelets to
ods found acceptable for this purpose by the FDA. [21 CFR 4 hours because they are prepared in an open system.45 The
606.145(a)] FDA defines “Prestorage pooled WBD platelets” as single
Commercial systems such as BacT/ALERT (bioMérieux) units of WBD platelets pooled and tested ≥24 hours after col-
and eBDS (Pall Corp.) have been approved by the FDA for lection, and stored in an FDA-cleared container for extended
screening platelets for bacterial contamination. These are pool storage for up to 5 days (consistent with the container
both culture-based systems.48 As the level of bacteria in the package insert). “Poststorage pooled WBD platelets” repre-
platelets at the time of collection can be low, samples are sent stored single units of WBD platelets that are pooled
not taken until after at least 24 hours of storage.48 This pro- within 4 hours prior to transfusion.45
vides time for any bacteria present to replicate to detectable In 2005, the FDA approved the use of prestorage-pooled
levels. BacT/ALERT measures bacteria by detecting a platelets prepared by AcrodoseTM PL System.45 Acrodose
change in carbon dioxide levels associated with bacterial platelets are pooled ABO-matched, leukoreduced WBD platelets
growth.48 This system provides continuous monitoring of that have been cultured and are ready for transfusion.54 Acro-
the platelet sample–containing culture bottles, which are dose systems are the only platelet pooling systems available
held for the shelf-life of the platelet unit or until a positive in the United States that provide a clinically equivalent
reaction is detected. The eBDS system measures the oxygen alternative to apheresis platelets.54 Because they are pro-
content of the air within the sample pouch for 18 to duced in a closed system, they can be stored for 5 days from
30 hours following incubation.52 A decrease in oxygen level collection. They provide a therapeutic dose equivalent to
indicates the presence of bacteria. BacT/ALERT and eBDS, apheresis platelets and at a lower cost, but they expose the
which are used for screening platelets in the United States, recipient to multiple donors. A recent study comparing
have documented good sensitivity and specificity; however, transfusion reactions from prestorage-pooled platelets,
false-negative test results have been documented.52 With apheresis platelets, and poststorage-pooled WBD platelets
both culture systems, the need to delay sampling and the found no difference in reaction rates among the different
requirement for incubation delay entry of the platelet prod- products.55 Prestorage-pooled platelets may prove to be a
ucts into inventory. Box 1–7 lists the disadvantages associ- useful adjunct to apheresis platelets, which are often in short
ated with the use of culture methods for the detection of supply, and may lead to improved utilization of WBD
bacterial contamination of platelets. platelets.54
The practice of screening platelets for bacterial contami- In November 2009, the FDA approved the first rapid test
nation has reduced, but not eliminated, the transfusion of to detect bacteria in platelets—the Pan Genera Detection
contaminated platelet products. False-negative cultures can (PGD) test (Verax Biomedical).56 The PGD test, which was
occur when bacteria are present in low numbers and when previously approved by the FDA for testing leukocyte-
the pathogen is a slow-growing organism. reduced platelets as an adjunct to culture, is an immunoas-
Because current bacterial screening methods are not 100% say that detects lipoteichoic acids on gram-positive bacteria
sensitive, they must be supplemented by other precautions, and lipopolysaccharides on gram-negative bacteria. Both
such as the donor interview and proper donor arm disinfec- aerobes and anaerobes are detected. A sample of only
tion. Another precaution is the diversion of the first aliquot 500 µL is required.56 Following pretreatment, the sample
(about 20 to 30 mL) of collected blood into a separate but is loaded into a disposable plastic cartridge with built-in
connected diversion pouch.53 This procedure minimizes the controls that turn from yellow to blue-violet when the test
placement of skin plugs, the most common source of bacte- is ready to be read, in approximately 20 minutes. A pink
rial contamination, into the WBD platelet products. bar in either the gram-positive or gram-negative test
In view of the ability to test for bacterial contamination window indicates a positive result. The PGD test can be
and the use of diversion pouches and sterile docking instru- performed by transfusion services just prior to release of
ments, there is now interest in storing pools of platelets up platelet products.52 The optimum time for sampling is at
to the outdate of the individual concentrates. Traditionally, least 72 hours after collection.56
four to six WBD platelets are pooled into a single bag by Transfusion services must either obtain their platelets
the transfusion service just prior to issue. This facilitates from a collection facility that performs an approved test for
bacterial contamination or they must perform an approved
test themselves.52 At this time, the approved tests are bacte-
BOX 1–7 rial culture or the Verax PGD test.48
The PGD rapid test has a sensitivity of 99.3% and a speci-
Disadvantages of Culture Methods for Detection ficity of 60%.56 While it is essential that the rapid test is sen-
of Bacterial Contamination of Platelets sitive enough to detect the majority of platelets containing
• Product loss due to sampling bacteria, a high false-positive rate could lead to discarding
• Delay in product release, further reducing already short shelf-life units of platelet that would be suitable for transfusion. The
• False-negative results false positive rate of 0.51% with the PGD test is a valid con-
• Cost cern because platelets are a blood product in short supply.52
• Logistical problems of culturing WBD platelets Despite sensitive methods to detect bacteria in platelets,
septic transfusion reactions still occur.
6888_Ch01_001-023 29/10/18 4:38 PM Page 18

18 PART I Fundamental Concepts

Pathogen Reduction for Platelets guidance of March 2016 includes a recommendation for
secondary testing of platelets prior to issuing, unless the
Procedures have been developed to treat platelet components platelets had been stored for 3 or fewer days; or the prod-
to reduce or inactivate any residual pathogens (bacteria, uct had been treated with an approved pathogen reduction
viruses, parasites, and protozoa) that may be present. The device.52 It is interesting to note that INTERCEPT platelets
term pathogen inactivation (PI) is the process of treating the were phased into routine use in Switzerland in 2011.59
blood component, and the components themselves are re- The FDA defines primary testing of platelets as the
ferred to as being pathogen reduced (PR).48 PR/PI procedures initial/first-time testing of a platelet component to detect
could potentially add an additional level of safety by protect- the presence of bacterial contamination.48 Testing is
ing against unknown and newly emerging pathogens. In 2014, conducted early in the storage period of platelets using a
the FDA approved a pathogen reduction technology for culture-based device or other adequate or appropriate
apheresis platelets stored for up to 5 days as a measure to method found acceptable by the FDA. Secondary testing of
reduce the risk of bacterial contamination of platelets.57 platelets is any additional test of a platelet component to
Currently, the INTERCEPT® Blood System is the only FDA detect the presence of bacterial contamination in a unit
approved technology for the manufacture of certain pathogen- that previously showed no bacterial contamination upon
reduced apheresis platelet and plasma products.57 primary testing.48 Secondary testing of platelets is usually
The INTERCEPT system (Cerus Corp.), uses amotos- conducted late in the storage period and may be conducted
alen that is activated by ultraviolet A (UVA) light and binds with either a culture-based bacterial detection device, or a
to the nucleic acid base pairs of pathogens, preventing rapid bacterial detection device acceptable to the FDA.48
replication.58 The targeting of nucleic acids is possible Secondary testing is currently optional and conducted with
because platelets, like RBCs, do not contain functional a rapid test.52
nucleic acids. INTERCEPT platelet components can be Table 1-16 lists the bacterial detection devices currently
used with platelets stored in 100% plasma or plasma and available in the United States.
PAS.58 Pathogen inactivation technology reduces the risk
of transfusion-transmitted infections. For 5-day apheresis
platelets, the requirements to control the bacterial contam- Advanced Concepts
ination risk are currently met by either culturing the prod- Dating of apheresis platelets up to 7 days is currently available
uct at least 24 hours after collection, or pathogen-reducing in the United States.48 In order for blood centers to extend
it within 24 hours after collection, using an FDA-cleared the dating period from 5 to 7 days, the platelets must be
and approved device. [21 CFR 606.145(a)] The FDA’s draft

Table 1–16 Bacterial Detection Devices Currently Available in the United States
Bacterial Testing
Methodology Device Indication Platelet Product Tested
Culture-based bacterial bioMerieux Detection of bacteria as a quality Apheresis platelets
detection devices BacT/ALERT control (QC) test

Detection of bacteria as a QC test Pools of single units of whole blood derived


platelets (WBDP)

Detection of bacteria as a QC test Single units of WBDP

Haemonetics eBDS Detection of bacteria as a QC test Apheresis platelets suspended in plasma

Detection of bacteria as a QC test WBDP suspended in plasma

Rapid bacterial detection Verax PGD test Safety measure Apheresis platelets suspended in plasma or
devices PAS-C/plasma

Pre-storage pools suspended in plasma

Detection of bacteria Post-storage WBDP pools suspended in plasma

Single units of WBDP suspended in plasma

Immunetics BacTx Detection of bacteria as a QC test Apheresis platelets suspended in plasma

Detection of bacteria as a QC test Post-storage WBD pools suspended in plasma

Taken from: Appendix A, Blood Products Advisory Committee Meeting FDA White Oak Campus, Silver Spring, MD November 30 - December 1, 2017. Available from: https://www.fda
.gov/downloads/AdvisoryCommittees/CommitteesMeetingMaterials/BloodVaccinesandOtherBiologics/BloodProductsAdvisoryCommittee/UCM587085.pdf
6888_Ch01_001-023 29/10/18 4:38 PM Page 19

Chapter 1 Red Blood Cell and Platelet Preservation: Historical Perspectives and Current Trends 19

collected in an FDA-approved 7-day platelet storage con- complex biochemistry and physiology. Besides having a long
tainer. In addition 1) “the 7-day container is labeled with shelf-life, platelet substitutes appear to have reduced poten-
a requirement to test every product with a bacterial detec- tial to transmit pathogens as a result of the processing
tion device cleared by FDA as a “safety measure test” and procedures. A number of different approaches have been
2) the platelets are cultured at least 24 hours after collec- utilized.60 One approach with the potential for providing
tion with an FDA-cleared device, and secondarily retested clinically useful products is the use of lyophilization.61
with a bacterial detection test labeled as a ‘safety measure,’ Two products prepared from human platelets are in pre-
proximate to the time of transfusion.” The FDA’s current clinical testing. One preparation uses washed platelets
review practice is to permit labeling of tests for bacterial treated with paraformaldehyde, with subsequent freezing in
detection in platelets for transfusion as a “safety measure” 5% albumin and lyophilization.62 These platelets on rehy-
when clinical studies have shown benefit for detection of dration have been reported to have hemostatic effectiveness
contamination not revealed by previous bacterial testing in different animal models. A second method involves the
and where clinical specificity was determined. In the freeze-drying of trehalose-loaded platelets.63 Platelet-like
United States the Verax PGD test is the only rapid bacterial nanoparticles are also being investigated as hemostatic
detection device cleared as a “safety measure” test and is agents since the shape and surface chemistry are similar to
used to extend dating to day 7. For transfusion services that of circulating platelets.64
who want to extend platelet dating beyond 5 days, the
FDA recommends performing secondary testing on Revisiting Approaches for Storage of Platelets
apheresis platelets previously cultured or PRT-treated, or at 1°C to 6°C
on single units of WBD platelets previously cultured, using
Although storage of platelets at 1°C to 6°C was discontinued
a device cleared by the FDA as a “safety measure.”48 Trans-
many years ago, there has been an interest in developing
fusion Services may extend the dating period beyond
ways to overcome the storage lesion that occurs at 1°C to
day 5 through day 6 or day 7, after registering with the
6°C.65 The rationale for the continuing effort reflects con-
FDA and listing the blood products.48
cerns about storing and shipping platelets at 20°C to 24°C,
especially the chance for bacterial proliferation. Refrigeration
Current Trends in Platelet Preservation would significantly reduce the risk of bacterial contamina-
Research tion, allowing for longer storage. Many approaches have
been attempted without success, although early results
showed some promise. The approaches primarily involve
Advanced Concepts adding substances to inhibit cold-induced platelet activation,
Research and development in platelet preservation is being as this is thought to be the key storage lesion.65 Spherical
pursued in many directions, including the following: platelets, manifested as a result of cold storage, have been
assumed to be a trigger for low viability. The specific ap-
1. Development of better platelet additive solutions proach involves and requires the enzymatic galactosylation
2. Development of more procedures to reduce and inacti- of cold-stored platelets to modify specifically one type of
vate the level of pathogens that may be in platelet units membrane protein.66
3. Development of platelet substitutes
4. New approaches for storage of platelets at 1°C to 6°C Frozen Platelets
5. Development of processes to cryopreserve platelets
Although considered a research technique and not licensed
Only the last three will be mentioned briefly.
by the FDA, frozen platelets are used occasionally in the
United States as autologous transfusions for patients who
Development of Platelet Substitutes are refractory to allogeneic platelets. Platelets are collected
by apheresis, the cryopreservative dimethyl sulfoxide
In view of the short shelf-life of liquid-stored platelet prod- (DMSO) is added, and the platelets are frozen at –80°C. The
ucts, there has been a long-standing interest in developing frozen platelets can be stored for up to 2 years. Prior to
platelet substitute products that maintain hemostatic func- transfusion, the platelets are thawed and centrifuged to
tion. Platelet substitutes are in the early stages of develop- remove the DMSO. Although in vivo recovery after transfu-
ment. It is understood that platelet substitutes may have use sion is only about 33%, the platelets seem to function
only in specific clinical situations because platelets have a effectively.65
6888_Ch01_001-023 29/10/18 4:38 PM Page 20

20 PART I Fundamental Concepts

SUMMARY CHART
 Each unit of whole blood collected contains approxi-  Rejuvesol is the only FDA-approved rejuvenation
mately 450 mL of blood and 63 mL of anticoagulant solution used in some blood centers to regenerate ATP
preservative solution or approximately 500 mL of blood and 2,3-DPG levels before RBC freezing.
and 70 mL of anticoagulant preservative solution.  Rejuvenation is used primarily to salvage O type and
 A donor can give blood every 8 weeks. rare RBC units that are outdated or used with specific
 Glycolysis generates approximately 90% of the ATP anticoagulant preservative solution up to 3 days past
needed by RBCs, and 10% is provided by the pentose outdate.
phosphate pathway.  Research is being conducted to improve on the current
 Seventy-five percent post-transfusion survival of RBCs additive solutions.
is necessary for a successful transfusion.  In 2016, INTERCEPT was approved by the FDA for
 ACD, CPD, and CP2D are approved preservative solu- pathogen reduction of platelets in 100% plasma.
tions for storage of RBCs at 1°C to 6°C for 21 days, and  RBC substitutes under investigation include hemoglobin-
CPDA-1 is approved for 35 days. based oxygen carriers and perfluorocarbons.
 Additive solutions (Adsol, Nutricel, Optisol, SOLX)  Two platelet additive solutions, InterSol and Isoplate,
are approved in the United States for RBC storage for have been approved for use in the United States.
42 days. Additive solution RBCs have been shown to  The FDA has approved the use of 7-day platelets as
be appropriate for neonates and pediatric patients. long as specific criteria are met.
 RBCs can be frozen for 10 years from the date of freez-
ing if they are glycerolized and frozen within 6 days of
whole blood collection in CPD or CPDA-1.

Review Questions 4. The majority of platelets transfused in the United States


today are:
1. What is the maximum volume of blood that can be a. Whole blood–derived platelets prepared by the
collected from a 110-lb donor, including samples for platelet-rich plasma method.
processing? b. Whole blood–derived platelets prepared by the buffy
a. 450 mL coat method.
b. 500 mL c. Apheresis platelets.
c. 525 mL d. Prestorage-pooled platelets.
d. 550 mL 5. Which of the following anticoagulant preservatives
2. How often can a blood donor donate whole blood? provides a storage time of 35 days at 1°C to 6°C for units
a. Every 24 hours of whole blood and prepared RBCs if an additive solution
b. Once a month is not added?
c. Every 8 weeks a. ACD-A
d. Twice a year b. CP2D
c. CPD
3. When RBCs are stored, there is a “shift to the left.” This d. CPDA-1
means:
a. Hemoglobin-oxygen affinity increases, owing to an 6. What are the current storage time and storage temper-
increase in 2,3-DPG. ature for platelet concentrates and apheresis platelet
b. Hemoglobin-oxygen affinity increases, owing to a components?
decrease in 2,3-DPG. a. 5 days at 1°C to 6°C
c. Hemoglobin-oxygen affinity decreases, owing to a b. 5 days at 24°C to 27°C
decrease in 2,3-DPG. c. 5 days at 20°C to 24°C
d. Hemoglobin-oxygen affinity decreases, owing to an d. 7 days at 22°C to 24°C
increase in 2,3-DPG.
6888_Ch01_001-023 29/10/18 4:39 PM Page 21

Chapter 1 Red Blood Cell and Platelet Preservation: Historical Perspectives and Current Trends 21

7. RBCs can be frozen for: 15. The INTERCEPT pathogen reduction system uses which
a. 12 months. of the following methods?
b. 1 year. a. Riboflavin and UV light
c. 5 years. b. Amotosalen and UV light
d. 10 years. c. Solvent/detergent treatment
d. Irradiation
8. Whole blood and RBC units are stored at what
temperature?
References
a. 1°C to 6°C
b. 20°C to 24°C 1. Parks D. Charles Richard Drew, MD 1904–1950. J Natl Med
c. 37°C Assoc. 1979;71:893-95.
2. Kendrick DB. Blood program in World War II [historical note].
d. 24°C to 27°C Washington (DC): Washington Office of Surgeon General,
9. Additive solutions are approved for storage of red blood Department of Army; 1964. p. 1-23.
3. Ellingson KD, Sapiano MRP, Haas KA, Savinkina AA, Baker
cells for how many days? ML, Chung KW, et al. Continued decline in blood collection
a. 21 and transfusion in the United States–2015. Transfusion. 2017
b. 42 Jun; 57(suppl 2):1588-98.
c. 35 4. Whitaker B, Rajbhandary S, Kleinman S, Harris A, Kamani N.
Trends in United States blood collection and transfusion:
d. 7 results from the 2013 AABB Blood Collection, Utilization,
10. One criterion used by the FDA for approval of new and Patient Blood Management Survey. Transfusion. 2016;
Sep;56(9):2173-83.
preservation solutions and storage containers is an aver- 5. Fung MK, Eder AF, Spitalnik SL, Westhoff CM. Technical
age 24-hour post-transfusion RBC survival of more than: manual. 19th ed. AABB, 2017.
a. 50%. 6. Harmening DM. Clinical hematology and fundamentals of
b. 60%. hemostasis. 5th ed. Philadelphia: FA Davis; 2009.
7. Himbert S, Alsop RJ, Rose M, Hertz L, Dhaliwal A, Moran-
c. 65%. Mirabal JM, et al. The molecular structure of human red
d. 75%. blood cell membranes from highly oriented, solid supported
multi-lamellar membranes. Sci Rep. 2017 Jan 3;7:39661.
11. What is the lowest allowable pH for a platelet compo- doi: 10.1038/srep39661.
nent at outdate? 8. Alsop RJ, Maria Schober R, Rheinstädter MC. Swelling of phos-
a. 6 pholipid membranes by divalent metal ions depends on the
b. 5.9 location of the ions in the bilayers. Soft Matter. 2016
Aug 10;12(32):6737-48. doi: 10.1039/c6sm00695g.
c. 6.8 9. Alshalani A and Acker JP. Red blood cell membrane water per-
d. 6.2 meability increases with length of ex vivo storage. Cryobiology.
2017 Jun;76:51-58. doi: 10.1016/j.cryobiol.2017.04.009. Epub
12. Which of the following occurs during storage of red 2017 Apr 26. PMID: 28456565
blood cells? 10. Wither M, Dzieciatkowska M, Nemkov T, Strop P, D’Alessandro
a. pH decreases A, Hansen KC. Hemoglobin oxidation at functional amino
b. 2,3-DPG increases acid residues during routine storage of red blood cells. Trans-
fusion. 2016 Feb;56(2):421-6. doi: 10.1111/trf.13363. Epub
c. ATP increases 2015 Oct 1.
d. plasma K+ decreases 11. Dumont LJ, AuBuchon JP. Evaluation of proposed FDA criteria
for the evaluation of radiolabeled red cell recovery trials. Trans-
13. Which of the following is approved for bacterial detec- fusion. 2008;48:1053-60.
tion specific to extending the expiration of apheresed 12. Hod EA, Zhang N, Sokol SA, Wojczyk BS, Francis RO, Ansaldi D,
platelets to 7 days? et al. Transfusion of red blood cells after prolonged storage pro-
a. BacT/ALERT duces harmful effects that are mediated by iron and inflamma-
tion. Blood. 2010;115:4284-92.
b. eBDS 13. Luten M, Roerdinkholder-Stoelwinder B, Schaap NP, de Grip
c. Gram stain WJ, Bos HJ, Bosman GJ, et al. Survival of red blood cells after
d. Pan Genera Detection (PGD) test transfusion: a comparison between red cell concentrates of dif-
ferent storage periods. Transfusion. 2008;48:1478-85.
14. Which of the following is the most common cause of 14. D’Alessandro A, Gray AD, Szczepiorkowski ZM, Hansen K,
bacterial contamination of platelet products? Herschel LH, Dumont LJ. Red blood cell metabolic responses
a. Entry of skin plugs into the collection bag to refrigerated storage, rejuvenation, and frozen storage. Trans-
fusion. 2017 Apr;57(4):1019-30.
b. Environmental contamination during processing 15. Ozment CP, Turi JL. Iron overload following red blood cell
c. T in the donor transfusion and its impact on disease severity. Biochim Biophys
d. Incorrect storage temperature Acta. 2009;1790:694-701.
6888_Ch01_001-023 29/10/18 4:39 PM Page 22

22 PART I Fundamental Concepts

16. Heddle NM, Cook RJ, Arnold DM, Liu Y, Barty R, Crowther hemoglobin-based oxygen carriers. Clinics (Sao Paulo).
MA, et al. Effect of short-term vs. long-term blood storage 2009;64(8):803-13.
on mortality after transfusion. N Engl J Med. 2016;375: 34. Tao Z, Ghoroghchian PP. Microparticle, nanoparticle, and stem
1937-45. cell-based oxygen carriers as advanced blood substitutes.
17. Sun K, D’Alessandro A, Xia Y. Purinergic control of red blood Trends in Biotechnology, September 2014, Vol. 32, No. 9.
cell metabolism: novel strategies to improve red cell storage. 35. Keipert PE. Hemoglobin-based oxygen carrier (HBOC) devel-
Blood Transfusion 2017;15(6):535-42. opment in trauma: previous regulatory challenges, lessons
18. Simon TL, Snyder EL, Stowell CP, et al, editors. Rossi’s principles learned, and a path forward. Adv Exp Med Biol. 2017;977:
of transfusion medicine. 4th ed. Malden, MA: Wiley-Blackwell; 343-50.
2009. 36. Mackenzie CF1, Dubé GP, Pitman A, Zafirelis M. Users guide
19. Vandromme MJ, et al. Transfusion and pneumonia in the to pitfalls and lessons learned about HBOC-201 during clinical
trauma intensive care unit: an examination of the temporal trials, expanded access, and clinical use in 1,701 patients.
relationship. J Trauma. 2009;67:97-101. Shock. 2017 Oct 25. doi: 10.1097/SHK.0000000000001038.
20. Weinberg JA, McGwin G Jr, Vandromme MJ, Marques MB, [Epub ahead of print] .
Melton SM, Reiff DA, et al. Duration of red cell storage influences 37. Natanson C, Kern SJ, Lurie P, Banks SM, Wolfe, SM. Cell-free
mortality after trauma. J Trauma. 2010 Dec;69(6):1427-31; dis- hemoglobin-based blood substitutes and risk of myocardial
cussion 1431-2. doi: 10.1097/TA.0b013e3181fa0019. infarction and death. JAMA. 2008;99:2304-12.
21. Offner PJ, Moore EE, Biffl WL, Johnson JL, Silliman CC. 38. Dubé GP, Pitman AN, Mackenzie CF. Relative efficacies of
Increased rate of infection associated with transfusion of HBOC-201 and polyheme to increase oxygen transport com-
old blood after severe injury. Arch Surg. 2002;137:711-16. pared to blood and crystalloids. “2017 Military Supplement.”
22. Guidance for Industry: Circular of Information for the Use of Shock. 2017 Nov 14. doi: 10.1097/SHK.0000000000001058.
Human Blood and Blood Products. AABB, America’s Blood Cen- [Epub ahead of print]
ters, American Red Cross (ARC), and the Armed Services Blood 39. Van Veen T, Hunt JA. Tissue engineering red blood cells: a ther-
Program (ASBP), November, 2013. Available from: https://www apeutic. J Tissue Eng Regen Med. 2015; 9:760-70.
.fda.gov/biologicsbloodvaccines/guidancecomplianceregulatory 40. Mittal K, Kaur R. Platelet storage lesion: an update. Asian J
information/guidances/blood/ucm364565.htm Transfus Sci. 2015 Jan-Jun;9(1):1-3.
23. McCullough J. Preparation, storage, and characteristics of 41. Kreuger, AL, Caram-Deelder, C, Jacobse, J, Kerkhoffs, J, Bom,
blood components and plasma derivatives. In: McCullough J, JG, Middelburg, RA. Effect of storage time of platelet products
editor. Transfusion medicine. 4th ed. Hoboken: Wiley & Sons, on clinical outcomes after transfusion: a systematic review and
2016. meta-analyses. Vox Sang. 2017 112:291-300.
24. D’Alessandro A, Nemkov T, Kelher M, West FB, Schwindt RK, 42. Horvath M, Eichelberger B, Koren D, et al. Function of platelets
Banerjee A, et al. Routine storage of red blood cell (RBC) units in apheresis platelet concentrates and in patient blood after
in additive solution-3: a comprehensive investigation of the transfusion as assessed by Impact-R. Transfusion. 2010;50:
RBC metabolome. Transfusion. 2015 Jun;55(6):1155-68. doi: 1036-42.
10.1111/trf.12975. Epub 2014 Dec 30. PMID: 25556331 43. Mallhi RS, Kumar S, Philip J. A comparative assessment of
25. Sloan SR. Neonatal transfusion review. Paediatr Anaesth. quality of platelet concentrates prepared by Buffy coat poor
2011;21(1):25–30. platelet concentrate method and aphaeresis derived platelet
26. U.S. Department of Health and Human Services, Food and concentrate methods. Indian J Hematology Blood Transfusion.
Drug Administration. e-CFR Electronic Code of Federal Reg- 2015;31(4):453-59.
ulations, Title 21—Food and Drugs, Blood Products 600–680. 44. Smethurst PA. Aging of platelets stored for transfusion.
Washington (DC): U.S. Government Printing Office; May, Platelets. 2016 Sep;27(6):526-34. doi: 10.3109/09537104.
2018. 2016.1171303. Epub 2016 May 2. Review. PMID: 27135140
27. Valeri CR, Ragno G, Pivacek LE, Srey R, Hess JR, Lippert LE, 45. Guidance for Industry and FDA Review Staff: Collection of
et al. A multicenter study of in-vitro and in-vivo values in Platelets by Automated Methods (December 2007), available
human RBCs frozen with 40% (wt/vol) glycerol and stored after at: https://www.fda.gov/downloads/BiologicsBloodVaccines/
deglycerolization for 15 days at 4°C in AS-3: assessment of RBC GuidanceComplianceRegulatory In formation/Guidances/Blood/
processing in the ACP 215. Transfusion. 2001;41:933-39. UCM062946.pdf
28. Cancelas JA, Gottschall JL, Rugg N, Graminske S, Schott MA, 46. Greco CA, Zhang JG, Kalab M, Yi QL, Ramirez-Arcos SM,
North A, et al. Red blood cell concentrates treated with the Gyongyossy-Issa MI. Effect of platelet additive solution on bac-
amustaline (S-303) pathogen reduction system and stored for terial dynamics and their influence on platelet quality in stored
35 days retain post-transfusion viability: results of a two-centre platelet concentrates. Transfusion. 2010 Nov;50(11):2344-52.
study. Vox Sang. 2017;112:210-18. doi: 10.1111/j.1537-2995.2010.02726.x.
29. Brixner V, Kiessling AH, Madlener K, Müller MM, Leibacher J, 47. Talukdar B, Chakraburty S, Hazra R, et al. Quality assessment
Dombos S, et al. Red blood cells treated with the amustaline of platelet concentrates: a comparative study using three dif-
(S-303) pathogen reduction system: a transfusion study in ferent methods. International Journal of Biomedical Research.
cardiac surgery. Transfusion. 2018 Apr;58(4):905-16. doi: 2017;8(4):194-99.
10.1111/trf.14528. 48. U.S. Department of Health and Human Services Food and Drug
30. Kwan DH, Constantinescu I, Chapanian R, Higgins MA, Administration Center for Biologics and Evaluation and Re-
Kötzler MP, Samain E, et al. Toward efficient enzymes for search. Bacterial Risk Control Strategies for Blood Collection
the generation of universal blood through structure-guided Establishments and Transfusion Services to Enhance the Safety
directed evolution. J Am Chem Soc. 2015 May 6;137(17): and Availability of Platelets for Transfusion. 2016.
5695-705. doi: 10.1021/ja5116088. 49. Benjamin RJ, Braschler T, Weingand T, Corash LM. Hemovigi-
31. Arteriocyte Cellular Therapies Medical Systems [now Isto lance monitoring of platelet septic reactions with effective bac-
Biologics]. St. Louis; 2017 [cited 2011 Aug 30]. Blood Pharm- terial protection systems. Transfusion. 2017;57(12):2946-57.
ing. Available from: www.arteriocyte.com. doi: 10.1111/trf.14284 [doi].
32. Lee N. The Lancet technology: pharming blood. Lancet. 50. Fatalities Reported to FDA Following Blood Collection and Trans-
2016;18;387:2496. fusion Annual Summary for FY2015. 11/2/2016. Available from:
33. Chen J, Scerbo M, Kramer G. A review of blood substitutes: http://www.fda.gov/BiologicsBloodVaccines/SafetyAvailability/
examining the history, clinical trial results, and ethics of ReportaProblem/TransfusionDonationFatalities/ UCM518148.pdf
6888_Ch01_001-023 29/10/18 4:39 PM Page 23

Chapter 1 Red Blood Cell and Platelet Preservation: Historical Perspectives and Current Trends 23

51. Schlenke P. Pathogen inactivation technologies for cellular 58. Cerus Corporation, INTERCEPT Blood System for Platelets –
blood components: an update. Transfus Med Hemother. 2014 Large Volume (LV) Processing Set. Package Insert, March 15,
Jul;41(4):309-25. doi: 10.1159/000365646. Epub 2014 Jul 21. 2016.
Review. 59. Jutzi M. AABB symposium on implementation of pathogen-
52. Blood Products Advisory Committee Meeting. FDA White reduced blood components. North Bethesda, MD, April 28, 2015.
Oak Campus, Silver Spring, MD November 30–December 1, 60. Blajchman MA. Substitutes and alternatives to platelet trans-
2017. Available from: https://www.fda.gov/downloads/Advisory fusions in thrombocytopenic patients. J Thromb Haemost.
Committees/CommitteesMeetingMaterials/BloodVaccinesand 2003;1:1637-41.
OtherBiologics/BloodProductsAdvisoryCommittee/UCM587085 61. Modery-Pawlowski CL, Tian LL, Pan V, McCrae KR, Mitragotri S,
.pdf Gupta AS. Approaches to synthetic platelet analogs. Biomaterials.
53. McDonald CP. Bacterial risk reduction by improved donor arm 2013;34:526-41.
disinfection, diversion and bacterial screening. Transfus Med. 62. Fischer TH, et al. Intracellular function in rehydrated
16:381-96, 2006. lyophilized platelets. Brit J Haematol. 2000;111:167-74.
54. AcrodoseTM PL Systems. A new platform for pre-storage pool- 63. Crowe JH, et al. Stabilization of membranes in human platelets
ing and testing of Whole Blood-Derived Platelets (WBDP). freeze-dried with trehalose. Chem Phys Lipids. 2003;122:
Haemonetics. Accessed from: https://www.haemonetics.com/ 41-52.
~/media/Sharepoint/Consumables/Whole_Blood/Acrodose/ 64. Anselmo AC, Modery-Pawlowski CL, Menegatti S, Kumar S,
COL-COPY-000539_US_Acrodose-PL-Systems.pdf.pdf, 2018. Vogus DR, Tian LL, et al. Platelet-like nanoparticles: mimicking
55. Tormey CA, et al. Analysis of transfusion reactions associated shape, flexibility, and surface biology of platelets to target
with prestorage-pooled platelets. Transfusion. 2009;49:1242-47. vascular injuries. Acs Nano. 2014;8:11243-53.
56. Verax PGD test FDA clearance accessible at https://www 65. Waters L, Cameron M, Padula MP, Marks DC, Johnson L.
.fda.gov/BiologicsBloodVaccines/BloodBloodProducts/Approved Refrigeration, cryopreservation and pathogen inactivation: an
Products/SubstantiallyEquivalent510kDeviceInformation/ucm updated perspective on platelet storage conditions. Vox Sang.
551518.htm. 2018 Feb 14. doi: 10.1111/vox.12640. [Epub ahead of print]
57. Food and Drug Administration Center for Biologics Evaluation Review.
and Research [Internet]: Washington (DC): FDA [cited 2017 66. Handigund M, Bae TW, Lee J, Cho YG. Evaluation of in vitro stor-
June 1]. New Drug Application. Available from:https://www age characteristics of cold stored platelet concentrates with N
.fda.gov/BiologicsBloodVaccines/BloodBloodProducts/ acetylcysteine (NAC). Transfus Apher Sci. 2016 Feb;54(1):
ApprovedProducts/NewDrugApplicationsNDAs/ucm342639.htm. 127-38. doi: 10.1016/j.transci.2016.01.006. Epub 2016 Jan 13.
6888_Ch02_024-044 29/10/18 4:40 PM Page 24

Chapter 2
Basic Genetics
Lorraine Caruccio, PhD, MT(ASCP)SBB and Helene DePalma, MS,
MT(ASCP)SBB, CQA(ASQ)

Introduction Meiosis Review Questions


Classic Genetics Cell Division Cycle References
Population Genetics Molecular Genetics Bibliography
Early Genetics and Mendel’s Laws Deoxyribonucleic Acid
of Inheritance Ribonucleic Acid
Hardy-Weinberg Principle Common Approaches in Modern
Inheritance Patterns Genetics Techniques
Cellular Genetics The Role of Genetics in the Future
Terminology Case Study 2-1
Mitosis Summary Chart

OBJECTIVES
1. Explain Mendel’s laws of independent segregation and random assortment, and describe how he developed them.
2. Correlate the concepts of dominant and recessive traits with examples of the inheritance of blood group antigens.
3. Explain the Hardy-Weinberg principle and how it applies to genetic traits.
4. Given the necessary information, solve Hardy-Weinberg problems for any blood group antigen.
5. Determine the inheritance pattern of a given trait by examining the pedigree analysis.
6. Describe the processes of mitosis and meiosis, and outline the differences between them.
7. Distinguish between X-linked and autosomal traits, and describe how each is inherited.
8. Describe in detail the processes of replication, transcription, and translation, including the basic mechanism of each.
9. List the various types of genetic mutations and describe how they can change the function of living cells and organisms.
10. Describe the cell’s different mechanisms for correcting mutations.
11. Identify some of the ways in which genetics can be used in the transfusion laboratory, including the necessary background
information for describing genetic testing techniques.
12. Describe in general the modern techniques used in the study of genetics.

Introduction groups. Knowledge of current methods of analysis is also


required to appreciate how problems in genetics are solved
One of the most important areas of modern biology is the and explained. Information about the inherited differences
science of genetics, the study of heredity. The understanding affecting red cell antigens among individuals forms the foun-
of the inheritance of blood group antigens and the testing dation of safe blood transfusion.
for disease markers at the molecular level, both of which are A solid understanding of classic genetics, including
vitally important in transfusion medicine, are based on the Mendel’s laws of inheritance and Hardy-Weinberg formulas,
science of genetics. This chapter covers the basic concepts cellular concepts that control chromosomes, cellular division
of genetics necessary to understand its role in blood banking. such as mitosis and meiosis, and the molecular structures
The fundamental principles of classic genetics will facilitate of nucleic acids, is required to fully understand modern
understanding of the molecular bases of individual blood genetics. How these theories, concepts, and principles apply

24
6888_Ch02_024-044 29/10/18 4:40 PM Page 25

Chapter 2 Basic Genetics 25

to transfusion medicine should be clearly understood, as polymorphism (SNP). For example, genotyping the donor
genetics is a very dynamic science that has its greatest poten- or recipient DNA, extracted from leukocytes, can predict
tial in direct applications. Many areas of transfusion medicine which antigens may be expressed on the red blood cells.
rely on an understanding of blood group genetics and on The predicted phenotype for a recipient obtained by geno-
accurate and sensitive methods of pathogen testing to main- typing can provide valuable insight on potential antibodies
tain the safety of the blood supply. Most of the antigens in that can be produced, especially if the recipient red
the various blood group systems (i.e., ABO, Rh, Kell, Kidd, blood cells can’t be typed by serologic methods due to recent
etc.) generally follow straightforward inheritance patterns, transfusion. Using this simple example, we see that in
usually of a codominant nature. modern blood banking genetics plays an important role. See
Historically, the major focus of genetics in blood banking Chapter 4 for more examples of clinical applications of red
has been in population genetics and inheritance patterns, but cell genotyping.
now cellular and molecular genetics are equally important.
Increasingly, modern genetic techniques are playing a role in Population Genetics
analyzing the antigen profile of blood donors and recipients,
which was previously performed only by serologic testing. The major areas of population genetics of concern to blood
Transfusion medicine professionals should understand banking include Mendel’s laws of inheritance, the Hardy-
classic genetics, such as interpretation of familial inheritance Weinberg principle, and inheritance patterns.
patterns as well as the concepts behind molecular methods,
such as in restriction mapping, sequencing, polymerase Early Genetics and Mendel’s Laws of Inheritance
chain reaction (PCR), and gene array technology. (See
Chapter 4, “Concepts in Molecular Biology.”) The Swedish biologist Carolus Linnaeus started the first clas-
In this chapter, a general overview of genetics at three sification system of living things in the 17th century and
different levels is provided. Population genetics, concern- used the unit of “species” as its principal definition. Deter-
ing genetic traits in large numbers of individuals; cellular mining factors that affected which classification group a
genetics, which pertains to the cellular organization of species would be put into was based on physical traits and
genetic material; and molecular genetics, based on the bio- observations. There was no attempt made to understand the
chemistry of genes and the structures that support them. underlying reasons for one trait versus another trait occur-
The chapter also provides a brief overview of molecular ring in one species or another. The amazing diversity of
techniques. Chapter 4 explains molecular biology testing species and the processes that might contribute to it were
methods in greater detail, including recombinant DNA not investigated further. In 1859, Charles Darwin published
technology, Southern and Northern blotting, restriction his epic book On the Origin of Species after many years of
fragment length polymorphism analysis, PCR techniques, intense study of various and diverse life-forms. Darwin’s
and cloning and sequencing. ambition was to understand the diversity of life and how
one organism could gain an advantage over another and
Classic Genetics better survive in a given environment, which is referred to
as “natural selection.” It created a revolution in the thinking
Modern genetics is based upon the understanding of the of modern biology and is still controversial today.
biochemical and biophysical nature of nucleic acids, The science of genetics found its modern development in
including deoxyribonucleic acid (DNA), ribonucleic acid the work of Gregor Mendel, published in 1865. Mendel was
(RNA), and the various proteins that are part of the chro- an Austrian monk and mathematician who used sweet pea
mosomal architecture. In addition, genetics is concerned plants growing in a monastery garden to study physical traits
with population studies and epidemiology. The under- in organisms and how they are inherited. He determined that
standing of inheritance patterns in which genetic traits are physical traits are due to factors he called elementen within
followed and analyzed, as well as the biochemical reactions the cell. In modern genetics, we know the physical basis of
that result in gene mutations that can give rise to new these so-called elementen are genes within the nucleus of the
alleles, are highly important in the study of genetics. Dis- cell. Mendel studied the inheritance of several readily observ-
covery of novel alleles can result in new blood group anti- able pea plant characteristics, notably flower color, seed color,
gens and provide insight on antigen-typing discrepancies and seed shape. He based his first law of inheritance, the law
due to weakened or variant alleles. of independent or random segregation, on these results.
All areas of transfusion medicine are influenced by The first generation in the study, called the parental, pure,
genetics, including HLA typing, relationship testing, and the or P1 generation, consisted of all red or all white flowers that
prediction of the phenotype of red cell, platelet, and neu- bred true for many generations. The plants were either
trophil antigens. The antigens present on all blood cells homozygous for red flowers (RR, a dominant trait; dominant
are expressed as a phenotype, but it is the genotype of the traits are usually written with uppercase letters) or homozy-
organism that controls what antigens may be expressed on gous for white flowers (rr, a recessive trait; recessive traits
the cell. Although many genetic mechanisms can generate are usually written with lowercase letters). When these
various blood group antigens, the majority are the result of plants were crossbred, the second generation, called first-
a change of one nucleotide referred to as a single nucleotide filial, or F1, had flowers that were all red. Thus the dominant
6888_Ch02_024-044 29/10/18 4:40 PM Page 26

26 PART I Fundamental Concepts

trait was the only trait observed. When plants from the F1 gene is not dominant over its allele, and the protein products
generation were crossbred to each other, the second-filial, of both genes are seen at the phenotypic level. An example
or F2, generation, of plants had flowers that were red and of this is seen in Figure 2–2 concerning the MNSs blood
white in the ratio of 3:1 (Fig. 2–1). All the plants from the group system, in which the red blood cells of a heterozygous
F1 generation are heterozygous (or hybrid) for flower color MN individual would type as both M and N antigen positive.
(Rr). The F2 generation has a ratio of three red-flowered (See Chapter 8, “Blood Group Terminology and the Other
plants to one white-flowered plant. This is because the plants Blood Groups.”)
that have the R gene, either RR homozygous or Rr heterozy- Mendel’s second law is the law of independent assortment
gous, will have red flowers because the red gene is dominant. and states that genes for different traits are inherited sepa-
Only when the red gene is absent and the white gene occurs rately from each other. This allows for all possible combina-
in duplicate, as in the rr homozygous white-flowered plant, tions of genes to occur in the offspring. Specifically, if a
will the recessive white gene expression be visible as a phe- homozygote that is dominant for two different characteristics
notype. This illustrates Mendel’s first law, the law of inde- is crossed with a homozygote that is recessive for both char-
pendent segregation. Specifically, Mendel’s first law shows acteristics, the F1 generation consists of plants whose phe-
that alleles of genes have no permanent effect on one another notype is the same as that of the dominant parent. However,
when present in the same plant but segregate unchanged by when the F1 generation is crossed in the F2 generation, two
passing into different gametes. Each gene is passed on to the general classes of offspring are found. One is the parental
next generation on its own. type; the other is a new phenotype called a reciprocal type
An intermediate situation can also occur when alleles and represents plants with the dominant feature of one plant
exhibit partial dominance. This is observed when the pheno- and the recessive feature of another plant. Recombinant
type of a heterozygous organism is a mixture of both homozy- types occur in both possible combinations. Mendel formu-
gous phenotypes seen in the P1 generation. An example of lated this law by performing studies with different types of
this is plants with red and white flowers that have offspring seeds produced by peas and noted that they can be colored
with pink flowers or flowers that have red and white sections. green or yellow and textured smooth or wrinkled in any
It is important to remember that although the phenotype does combination. An illustration of independent assortment of
not show dominance or recessive traits, the F1 generation has Mendel’s second law is given in Figure 2–3; his system of pea
the heterozygous genotype of Rr. It is essential to understand plant seed types are used as the example.
how a genotype can influence a phenotype, and using flower Mendel’s laws apply to all sexually reproducing diploid
color is a good basic model system to study this. organisms whether they are microorganisms, insects, plants,
Unlike the flower color of many types of plants, most or animals, or people. However, there are exceptions to the
blood group genes are inherited in a codominant manner. In Mendelian laws of inheritance. If the genes for separate traits
codominance, both alleles are expressed, and their gene are closely linked on a chromosome, they can be inherited as
products are seen at the phenotypic level. In this case, one a single unit. The expected ratios of progeny in F1 matings
may not be seen if the various traits being studied are linked.
There can also be differences in the gene ratios of progeny of
Parental RR rr F1 matings, if recombination has occurred during the process
of meiosis. An example of this in blood banking is the
MNSs system, in which the MN alleles and the Ss alleles are

Gametes R r

MN NN NN MM

First-Filial Rr

MN MN NN MN MN

Second-Filial R r

R RR Rr
MM MN NN MN NN

r Rr rr

Where R = red and r = White


MN NM MN NN
Figure 2–1. A schematic illustration of Mendel’s law of separation using
flower color. Figure 2–2. Independent segregation of the codominant genes of M and N.
6888_Ch02_024-044 29/10/18 4:40 PM Page 27

Chapter 2 Basic Genetics 27

Parental RRYY rryy


p q

Gametes RY ry
p p2 pq p2 + 2pq + q2 = 1.0

First-Filial RY Ry rY ry

q pq q2

Second-Filial RY Ry rY ry
Figure 2–4. Common inheritance patterns.

RY RRYY RRYy RrYY RrYy


and various conditions must be met to use the equations
Ry RRYy RRyy RrYy Rryy appropriately. These criteria are outlined in Box 2–1.
In any normal human population, it is almost impossible to
rY RrYY RrYy rrYY rrYy meet these demanding criteria. Although large populations
ry RrYy Rryy rrYy rryy exist, collecting sample data from a large enough representative
segment of a population is not always feasible. Also, mating is
Where R = round r = wrinkled not always random, and there is mixing of populations on a
global scale now that leads to “gene flow” on a constant basis.
Y = yellow y = green
Recently, sequencing of the human genome has revealed that
Figure 2–3. A schematic illustration of Mendel’s law of independent assortment gene mutations occur much more commonly than originally
using seed types. thought. Some of these mutations affect the phenotype of an
individual, such as loss of enzyme function, and some do not.
Despite these drawbacks, Hardy-Weinberg is still one of the
physically close on the same chromosome and are therefore best tools for studying gene frequencies in human populations
linked. Closely linked genes are inherited as a haplotype, and and is a cornerstone of population genetics.
the antigens encoded by the haplotypes have a different The allele frequency for blood group genes can be studied
prevalence than if there was random assortment. Recombi- using the Hardy-Weinberg equations. A relevant example
nation happens when DNA strands are broken and there is that demonstrates the use of the Hardy-Weinberg formula is
exchange of chromosomal material followed by activation of the frequency of the Rh antigen, D, in a given population.
DNA repair mechanisms. The exchange of chromosomal For simplicity, the absence of an RHD gene will be denoted
material results in new hybrid genotypes that may or may not as dd, using the same convention of upper- and lowercase
be visible at the phenotypic level. letters as in Figure 2–1. Refer to Chapter 7 for a detailed
Mendel’s laws of inheritance give us an appreciation of explanation of the genetics of the Rh system. To determine the
how diverse an organism can be through the variations in its frequency of each allele, we count the number of individuals
genetic material. The more complex the genetic material of who have the corresponding phenotype (remembering that
an organism, including the number of chromosomes and both Dd and DD will appear as D-positive) and divide this
the number of genes on the chromosomes, the greater the number by the total number of alleles. This value is repre-
potential for uniqueness among organisms of the same sented by p in the Hardy-Weinberg equation. Again, count-
species. Also, the more complex the genetic material, the ing the alleles lets us determine the value of q by counting
more complex and varied its responses to conditions in the alleles, bearing in mind that when p and q are added, they
environment. Therefore, as long as control is maintained must equal 1. The ratio of homozygotes and heterozygotes
during cell division and differentiation, organisms with is determined by solving the equation p2 + 2pq + q2 = 1. If in
greater genetic diversity and number can have an advantage our example we tested 1,000 random blood donors for
over other organisms in a given setting. the D antigen and found that DD and Dd (both genotypes
resulting in a D-positive phenotype) occurred in 84% of the
Hardy-Weinberg Principle

G.H. Hardy, a British mathematician, and W. Weinberg, a


BOX 2–1
German physician, developed a mathematical formula that
allowed the study of Mendelian inheritance in great detail. The Criteria for Use of the Hardy-Weinberg Formula
Hardy-Weinberg formula—p + q = 1, in which p equals the • The population studied must be large.
gene frequency of the dominant allele and q is the frequency • Mating among all individuals must be random.
of the recessive allele—can also be stated p2 + 2pq + q2 = 1 and • Mutations must not occur in parents or offspring.
specifically addresses questions about recessive traits and how • There must be no migration, differential fertility, or mortality of
they can be persistent in populations (Fig. 2–4). Like many genotypes studied.
mathematical formulations, however, certain ideal situations
6888_Ch02_024-044 29/10/18 4:40 PM Page 28

28 PART I Fundamental Concepts

population, and dd (Rh-negative) occurred in 16%, the gene A. Autosomal recessive


frequency calculations would be performed as follows:
p = gene frequency of D
q = gene frequency of d
p2 = DD, 2pq = Dd, which combined are 0.84
q2 = dd, which is 0.16
q = square root of 0.16, which is 0.4
p+q=1
p=1–q
p = 1 – 0.4 Propositus
B. X-linked dominant
p = 0.6
This example is for a two-allele system only. A three-allele
system would require use of the expanded binomial equation
p = q + r = 1 or p2 + 2pq + 2pr + q2 + 2qr + r2 = 1. More
complex examples using this formula can be found in
advanced genetics textbooks.

Inheritance Patterns C. X-linked recessive

The interpretation of pedigree analysis requires the under-


standing of various standard conventions in the representa-
tion of data figures. Males are always represented by squares
and females by circles, with open symbols indicating an
unaffected individual and a closed, filled-in symbol indicat-
ing an affected individual. A line joining a male and a female
indicates a mating between the two, and offspring are indi-
cated by a vertical line. A double line between a male and a
= affected = affected, heterozygous = not affected,
female indicates a consanguineous mating. A stillbirth or carrier
abortion is indicated by a small black circle. Deceased family
members have a line crossed through the symbol represent- Figure 2–5. Schematic illustration of common inheritance patterns.
ing them. The propositus or proband in the pedigree is
indicated by an arrow pointing to it and indicates the most
interesting or important member of the pedigree. Something his daughters will have the trait. This is because a father
unique about the propositus is often the reason the pedigree always passes his Y chromosome to his sons and his X chro-
analysis is undertaken and is referred to as the index case. mosome to his daughters. Women can be either homozygous
Figure 2–5 shows examples of different types of inheri- or heterozygous for an X-linked trait; therefore, when moth-
tance patterns seen in pedigree analysis. Almost all pedigrees ers have an X-linked trait, their daughters inherit the trait in
will follow one of these patterns or, rarely, a combination a manner identical to autosomal inheritance. The sons have
of them. The first example is a pedigree demonstrating a 50% chance of inheriting the trait. Because the trait is domi-
autosomal-recessive inheritance. Autosomal refers to traits nant, the sons who inherit it will express it. The Xga blood
that are not carried on the sex chromosomes. Sex-linked group system is one of the few blood group systems that
traits are encoded by a gene generally located on the X chro- follow an X-linked inheritance pattern (refer to Chapter 8).
mosome, as few functional genes are present on the Y chro- The third example illustrated is X-linked recessive inheri-
mosome. A recessive trait is carried by either parent or both tance. In this case, the father always expresses the trait but
parents but is not generally seen at the phenotypic level never passes it on to his sons. The affected father always
unless both parents carry the trait. In some cases, a heterozy- passes the trait to all his daughters, who are then carriers of
gous individual inherits the gene for a recessive trait but the the trait. The female carriers will pass the trait on to half of
trait may not be seen at the phenotypic level. With autoso- their sons, who also will be carriers. In the homozygous
mal recessive inheritance, the trait is expressed only when state, X’Y, the males will express the trait, whereas only the
an individual is homozygous for the allele and inherited the rare homozygous females, X’X’, will express it. In this situa-
recessive allele from both parents. For example, the offspring tion, with an X-linked recessive trait, a disease-carrying gene
of two parents with the Lub/Lu genotype will have a one can be passed from generation to generation, with many
in four chance of inheriting two recessive Lu genes, which individuals not affected. A classic example of this is the in-
result in an Lu(a–b–) red cell phenotype. heritance of hemophilia A, which affected many of the royal
In the second example, the pedigree demonstrates houses of Europe. Another example of X-linked recessive
X-linked dominant inheritance. If the father carries the trait inheritance from blood group systems is the XK gene.
on his X chromosome, he has no sons with the trait, but all Mutations in this gene result in red blood cells with McLeod
6888_Ch02_024-044 29/10/18 4:40 PM Page 29

Chapter 2 Basic Genetics 29

phenotype and reduced expression of Kell system antigens transcriptionally active. Euchromatin is the swollen form of
(refer to Chapter 8). chromatin in cells, which is considered to be more active in
In addition, there is autosomal dominant inheritance, in the synthesis of RNA for transcription.
which all the members of a family who carry the allele show Most cellular nuclei contain these different types of chro-
the physical characteristic. Generally, each individual with matin. The chromatin material itself, which chiefly com-
the trait has at least one parent with the trait, and the gene prises long polymers of DNA and various basic proteins
is expressed whether the individual is homozygous or called histones, is compressed and coiled to form chromo-
heterozygous for the allele. Unlike X-linked traits, autosomal somes during cell division. Each organism has a specific
traits usually do not show a difference in the distribution number of chromosomes, some as few as 4 and some as
between males and females, and this can be useful in their many as 50. In humans, there are 46 chromosomes in diploid
evaluation. Also, in autosomal and X-linked traits, if an cells. These 46 chromosomes are arranged into pairs, with
individual does not have the trait, he or she can be a carrier one of each being inherited from each parent. Humans have
and can pass it on to offspring. This is why recessive traits 22 autosomes and one set of sex chromosomes, XX in the
seem to skip generations, which is another helpful clue in female and XY in the male. This comprises the 2N state of
determining inheritance patterns. the cell, which is normal for all human cells except the
Autosomal dominant traits are routinely encountered in gametes (sex cells). N refers to the number of pairs of
the blood bank, as most blood group genes are codominant chromosomes in a cell.
and are on autosomal chromosomes. They are passed on
from one generation to the next and usually do not skip gen- Terminology
erations. Finally, unusually rare traits that occur in every
generation and in much greater frequency than the general Two gametes are required to make a fertilized egg with the
population are often the result of consanguineous matings. correct (2N) number of chromosomes in the nucleus of a
Table 2–1 provides examples of inheritance patterns in trans- cell. Therefore, each parent contributes only half (1N) of the
fusion medicine. inherited genetic information, or genes, to each child. In
order to be completely healthy, each child must have the cor-
Cellular Genetics rect number of genes and chromosomes (2N), without major
mutations affecting necessary biochemical systems.
Organisms may be divided into two major categories: The genetic material has a complex pattern of organiza-
prokaryotic, without a defined nucleus, and eukaryotic, tion that has been evolving for millions of years to an amaz-
with a defined nucleus. Human beings and all other mam- ing level of coordination and control. At the smallest level,
mals are included in the eukaryotic group, as are birds, genes are composed of discrete units of DNA arranged in a
reptiles, amphibians, fish, and some fungus species. The linear fashion, similar to a strand of pearls, with structural
nucleus of a cell contains most of the genetic material proteins wrapped around the DNA at specific intervals to
important for replication and is a highly organized complex pack it into tightly wound bundles. A genome is the full
structure. The nuclear material is organized into chromatin, complement of genes in an organism. Chromosomes are
consisting of nucleic acids and structural proteins, and the structures in the cell nucleus that contain the duplex
is defined by staining patterns. Heterochromatin stains (double-stranded) DNA. A gene is a section, often very large,
as dark bands, and achromatin stains as light bands and of DNA along the chromosome that encodes for a particular
consists of highly condensed regions that are usually not protein. The specific sequence of nucleotides and the loca-
tion on the chromosome determines a gene. In addition,
each gene has specific and general sequences that occur
upstream (before the start site) and downstream (after the
Table 2–1 Examples of Inheritance termination signals) that contribute to how the gene func-
Patterns in Transfusion tions. The specific location of a gene on a chromosome is
Medicine called a locus (plural = loci), and at each locus there may be
Type of Pattern only one or several alternative forms of the gene, which are
Examples
called alleles. As an example, in the Kidd blood group system
Autosomal dominant ABO system the alleles JK*A and JK*B encode the Jka and Jkb antigens,
In (Lu) phenotype (mutation in respectively. Jka and Jkb are considered antithetical antigens.
KLF1 gene) When alleles are carried on the same chromosome they are
in cis position and alleles found on opposite chromosomes
Autosomal recessive Group O phenotype (OO genotype)
of a homologous pair are in trans position.
Lu(a–b–) phenotype (LuLu genotype) It is important to note the distinction between phenotype
Autosomal codominant S+s+ phenotype (GYPB*S/s genotype) and genotype. Genotype is the complement of DNA that is
inherited. The phenotype is the observable expression of the
X-linked dominant Xga blood group system genotype, including an enzyme to control a blood group anti-
X-linked recessive Hemophilia A, XK gene gen; the length of long bones of the skeleton; the ratio of mus-
cle fibers; the level of hormones produced; and obvious traits
6888_Ch02_024-044 29/10/18 4:40 PM Page 30

30 PART I Fundamental Concepts

such as eye, skin, and hair color. A gene may be inherited but cells. The process of mitosis is illustrated in Figure 2–6 and
the gene product not expressed due to a mutation that outlined in Table 2–2.
silences the gene, resulting in a phenotype that
differs from the genotype. More than one gene can affect a Meiosis
particular trait (part of a phenotype), such as the height of
an individual; all relevant genes can be considered as part of A different process is used to produce the gametes or sex
the genotype for that trait. Depending on the alleles inherited, cells. The process is called meiosis and results in four
an organism can be either homozygous or heterozygous for unique, rather than two identical, daughter cells. The
a specific trait. The presence of two identical alleles at a par- uniqueness of the daughter cells generated with meiosis
ticular locus results in a homozygous genotype (i.e., AA), and allows for great genetic diversity in organisms and controls
the phenotype is group A blood. The inheritance of the number of chromosomes within dividing cells. If cells
different alleles from each parent results on a heterozygous with 2N chromosomes were paired, the resulting daughter
genotype. Hemizygous refers to the condition when one cells would have 4N chromosomes, which would not be
chromosome has a copy of the gene and the other chromo- viable. Therefore, gametes carry a haploid number of chro-
some has that gene deleted or absent. In blood groups, the mosomes, 1N, so that when they combine, the resulting cell
amount of antigen expressed is impacted by whether an has a 2N configuration. Meiosis occurs only in the germinal
individual is homozygous or heterozygous for an allele. tissues and is important for reproduction. Without the com-
Generally, higher antigen density is expected when there plicated process of meiosis, there would be no variation from
is homozygosity for an allele. Antigen expression can also generation to generation, and evolution would not occur
be affected by gene or protein interactions. or would happen too slowly for organisms to adapt to envi-
Another important concept is that of the “silent” gene, ronmental changes.
or amorph. An amorph is a gene that does not produce The first stages of meiosis are nearly identical to those in
any obvious, easily detectable traits and is seen only at mitosis, in which chromatin is condensed, homologous
the phenotypic level when the individual is homozygous chromosomes are paired in prophase, and chromosomes are
for the trait. aligned along the center of the cell. However, there is no
centromere division, and at anaphase and telophase, the cell
Mitosis divides and enters once again into interphase, in which there

During cell division, the chromosomes are reproduced in


Mitosis
such a way that all daughter cells are genetically identical to
the parent cell. Without maintaining the same number and
type of chromosomes, the daughter cells would not be viable. 2N
The process by which somatic cells divide to create identical
daughter cells is called mitosis. The chromosomes are
duplicated, and one of each pair is passed to the daughter
cells. During the process of mitosis, quantitatively and qual-
itatively identical DNA is delivered to daughter cells formed
Prophase 4N
by cell division.
The complex process of mitosis is usually divided into a
series of stages, characterized by the appearance and move-
ment of the chromosomes: interphase, prophase, metaphase,
anaphase, and telophase. Interphase is the resting stage
when the cells are not actively dividing. The DNA is in the Metaphase 4N
form of chromatin and is dispersed throughout the nucleus.
New DNA is synthesized by a process called replication. In
the next stage, prophase, the chromatin condenses to form
visible chromosomes and the nuclear envelope starts to
break down. During metaphase, the chromosomes are lined Anaphase 4N
up along the middle of the nucleus and paired with the cor-
responding chromosome. Preparations from chromosomes
in metaphase are made for chromosome analysis in cytoge-
netics, visualized in a photomicrograph called a karyotype.
In anaphase, the cellular spindle apparatus is formed and Telophase
the chromosomes are pulled to opposite ends of the cell. The and cell
division
cell becomes pinched in the middle, and cell division starts
to take place. In the last stage, telophase, the cell is pulled 2N 2N
apart, division is complete, and the chromosomes and Figure 2–6. Mitosis leads to two daughter cells having the same number of
cytoplasm are separated into two new identical daughter chromosomes as the parent cell.
6888_Ch02_024-044 29/10/18 4:40 PM Page 31

Chapter 2 Basic Genetics 31

Table 2–2 Stages of Mitosis and Meiosis between maternal- and paternal-derived chromosomes. This
allows for the creation of new DNA sequences that are
Mitosis different from the parent strains. Combined with random
Stage Description segregation, it is possible to have very large numbers of new
DNA sequences. In humans with 23 pairs of chromosomes,
1. Interphase (2N) Resting stage between cell divisions; during the total possible number is several million. Meiosis is
this period, cells are synthesizing RNA and illustrated in Figure 2–7 and outlined in Table 2–2.
proteins, and chromatin is uncondensed.
2. Prophase (4N) First stage of mitotic cell division. Chromo- Cell Division Cycle
somes become visible and condense. Each
chromosome has two chromatids from Cell division is a complicated process that also occurs with
duplication of DNA, and chromatids are various specific stages. In eukaryotic cells such as human
linked via the centromere.
cells, the cell cycle is divided into four distinct stages and is
3. Metaphase (4N) Chromosomes move toward the equator represented by a clock or circular scheme indicating that it
of the cell and are held in place by micro- can repeat itself or can be stopped at any one point in the
tubules attached at the mitotic spindle cycle. The first step or stage is the resting stage, or G0, and
apparatus.
is the state of cells not actively dividing. The prereplication
4. Anaphase (4N) The two sister chromatids separate. Each one
migrates to opposite poles of the cell, and the
diameter of the cell decreases at the equator.
Meiosis
5. Telophase (2N) Chromosomes are at the poles of the cell,
and the cell membrane divides between the
two nuclei. The cell divides, and each cell 2N
contains a pair of chromosomes
identical to the parent cell.

Meiosis
Stage Description
Prophase I 4N
1. Interphase (2N) Resting stage between cell divisions; during
this period, cells are synthesizing RNA and
proteins, and chromatin is uncondensed.
2. Prophase I (4N) First stage of meiotic division. Chromosomes
condense. Homologous chromosomes pair
to become bivalent. Chromosome crossing Metaphase I 4N
over occurs at this stage.
3. Metaphase I (4N) Bivalent chromosomes align at cell equator.
Bivalent chromosomes contain all four of
the cell’s copies of each chromosome.
4. Anaphase I (4N) Anaphase I 4N
Homologous pairs move to opposite poles of
the cell. The two sister chromatids separate.
5. Telophase I (2N) The cell separates to become two daughter
cells. The new cells are now 2N.
Telophase I
6. Metaphase II (2N) Homologues line up at the equator. 2N and cell 2N
division
7. Anaphase II (N) Homologues move to opposite poles of the
cell equator.
8. Telophase II (N) Each cell separates into two new cells.
There are now four (N) cells with a unique
genetic constitution. 2N Metaphase II 2N

is no replication of DNA. This is followed by the second Telophase II


Gametes and cell
prophase, in which chromosomes are condensed, and division
then the second metaphase, with the centromeres dividing.
Finally, in the second anaphase and telophase stages, the two N N N N
cells divide, giving rise to four 1N daughter cells. In addition, Figure 2–7. Meiosis produces four gametes having half the number of chromo-
during meiosis, crossing over and recombination occurs somes present in the parent cell.
6888_Ch02_024-044 29/10/18 4:40 PM Page 32

32 PART I Fundamental Concepts

stage is next and is called G1. The step at which DNA is DNA
DNA HISTONES
synthesized is the next stage, called the S stage. It is followed
by the G2 stage, or postreplication stage. Finally, there is the
M phase, in which mitosis occurs.
Chromosomes are in the interphase stage of mitosis in
the span from G0 to the end of the G2 phase. Cells that are
completely mature and no longer need to divide to increase
their numbers, such as nerve cells, can remain in the G0 stage
for a very long time. Normal cells divide in response to var-
ious intracellular and extracellular signals. It is a hallmark
of cancer cells, such as the transformed cells seen in the var-
ious leukemias and solid tumors, that they can go through
the stages of cell division much faster than nontransformed, Figure 2–8. Nucleosomes consist of stretches of DNA wound around histone
normal cells due to a disruption in the process that regulates proteins.
the balance between cell proliferation and cell death. In this
way, cancerous cells consume the bulk of nutrients needed
by the nontransformed cells and crowd them out of exis- that interact with it have an overall basic pH. This helps to
tence, potentially overgrowing the adult organism in which stabilize the overall complex structure. The complex of DNA
they occur (Table 2–3). and histone protein is referred to as a nucleosome. The DNA
and protein complex is bound together so tightly and
Molecular Genetics efficiently that extremely long stretches of DNA, several
inches in length, can be packaged inside the nucleus of a
The study of genetics at the molecular level requires an under- cell on a microscopic level. The DNA and histones are held
standing of the biochemistry of the molecules involved. This together by various proteins that keep the DNA in a very spe-
includes knowledge of the physical conformation of chromo- cific helical conformation. This conformation also protects
somes as well as the biological and chemical nature of the the DNA from degradation when it is not being replicated
polymers of the different nucleic acids and nuclear proteins. or transcribed.
All DNA in human cells is in the form of a two-stranded
Deoxyribonucleic Acid duplex, with one strand in one direction and its complemen-
tary strand in the opposite direction (the strands are said to
Deoxyribonucleic acid (DNA) is a masterpiece of architec-
be antiparallel). The formation of hydrogen bonds between
tural evolution and the “backbone” of heredity. Chromatin
two complementary strands of DNA is called hybridization.
is actually a type of polymer structure. Chromosomes are
DNA is composed of four nitrogenous bases, a 5-carbon
composed of long, linear strands of DNA tightly coiled
sugar molecule called deoxyribose and a phosphate group.
around highly basic proteins called histones (Fig. 2–8). Each
The sugar and phosphate moieties comprise the backbone
chromosome is a single, extremely long strand of duplex
of the DNA molecule, while the nitrogenous bases face each
DNA. DNA is a nucleic acid, therefore most of the proteins
other and are stabilized by hydrogen bonding and Van der
Waals forces. The backbone of a DNA molecule is joined by
Table 2–3 The Generative Cell Cycle phosphodiester linkages. Unlike what is observed in proteins
with an α-helical structure, there is little bonding forces
Stage Description Configuration between bases on the same strand, which allows DNA to be
G0 (Gap 0) Temporary resting 2N strong but flexible.
period, no cell The four different nitrogenous bases are adenine (A),
division cytosine (C), guanine (G), and thymine (T). Adenine and
G1 (Gap 1) Cells produce RNA 2N guanine are purines, consisting of double-ring structures.
and synthesize Cytosine and thymine are pyrimidines, which are single-ring
protein structures (Fig. 2–9). The hydrogen bonding in DNA is spe-
S (Synthesis)
cific, in which A bonds only to T with two hydrogen bonds,
DNA replication 4N
occurs thus forming the weaker pairing, and C bonds only with G
with three hydrogen bonds, forming the stronger pairing.
G2 (Gap 2) During the gap 4N This is the classic Watson-Crick base pairing that occurs
between DNA syn-
in the B-form, right-handed helical structure of DNA. It was
thesis and mitosis,
the cell continues first postulated by James Watson and Francis Crick at
to synthesize RNA Cambridge University in the early 1950s (Fig. 2–10).1 Since
and produce new then, it has been discovered that DNA can also occur in modi-
proteins fied forms such as Z-DNA, which is a type of left-handed
M (Mitosis) Cell division occurs 2N helix with a different three-dimensional conformation but
that contains the same four nitrogenous bases.2 In addition,
6888_Ch02_024-044 29/10/18 4:40 PM Page 33

Chapter 2 Basic Genetics 33

Thymine—Adenine which is the complementary strand that gives the correct


5′ to 3′ sequence of messenger RNA (mRNA) that corresponds
to the template, or coding strand, of the DNA molecule.
As there are only four different bases used to make up
DNA templates, and there are 20 different amino acids that
are used to construct proteins, it is evident that any single
deoxyribose
nucleotide cannot code for any specific amino acid. What
has been discovered is that triplets of nucleotides called a
deoxyribose
codon, such as ATG, code for one specific amino acid. The
genetic code is the term used to describe this relationship
Cytosine—Guanine between triplet nucleotides and amino acids. There is a
redundancy to the genetic code, however, in that most of the
20 amino acids have more than one triplet that codes
for their addition to the peptide chain formed during
translation. Generally, the more common the amino acid is
deoxyribose in proteins, the greater the number of codons it has. There
are four special codons, including the only codon specific
deoxyribose for the initiation of transcription and translation, called the
start codon, and three different codons that are used to stop
Dotted lines represent interatomic hydrogen bonds, which hold the addition of amino acids in the process of peptide synthe-
the base pairs together.
sis. Because these three codons cannot be charged with an
Figure 2–9. The pyrimidine and purine bases found in the DNA molecule. amino acid, they are called stop codons and result in termi-
nation of the peptide being translated from mRNA. The
genetic code is illustrated in Figure 2–11.
there are some unusual forms of nitrogenous bases resulting
from methylations or deaminations that can be incorporated
into DNA templates, giving rise to nucleotides with different
properties. The genetic code is triplet
The phosphates in the DNA backbone attach to the sugar
at the third and fifth carbon atoms. Note that all atoms in a First base Second base
molecular structure are numbered. The linkage of the purine
or pyrimidine nitrogenous base to the sugar is at carbon 1.
Therefore, the two DNA strands are antiparallel—that is, one U C A G
strand is 5′ to 3′ (pronounced “5 prime to 3 prime”) in one
} } }
}
UUU UCU UAU UGU
direction and the complementary strand is 5′ to 3′ in the other Phe Tyr Cys
U UUC UCC UAC UGC
direction. During transcription, only one strand is copied, Ser
UUA
UUG } Leu
UCA
UCG
UAA
UAG } STOP
UGA STOP
UGG Trp
The double helix has constant width

} } }
}
CUU CCU CAU CGU
His
CUC CCC CAC CGC
Leu Pro Arg
C CUA
CUG
CCA
CCG
CAA
CAG } Gln
CGA
CGG

A
AUU
AUC
} } Ile
ACU
ACC
Thr
AAU
AAC } Asn
AGU
AGC } Ser

AUA
AUG Met
ACA
ACG
AAA
AAG } Lys
AGA
AGG
} Arg

}
} } }
GUU GCU GAU GGU
Asp
G GUC GCC GAC GGC
Val Ala Gly
GUA
GUG
GCA
GCG
GAA
GAG } Glu
GGA
GGG

Figure 2–10 Base pairing in DNA and DNA structure. (From Lewin B: Genes VIII. Figure 2–11. The genetic code. (From Lewin B: Genes VIII. Prentice Hall/
Prentice Hall/Pearson Education, New York, 2004, p 6. Reprinted with permission Pearson Education, New York, 2004, p 168. Reprinted with permission of the
of the author.) author.)
6888_Ch02_024-044 29/10/18 4:40 PM Page 34

34 PART I Fundamental Concepts

DNA Replication replicated. This “primes” the replication process; therefore,


The replication, or copying, of DNA is a complex process these short oligonucleotide sequences are called primers.
involving numerous enzymes, nucleic acid primers, various Both DNA parent strands are replicated simultaneously in a
small molecules, and the DNA helix molecule that serves as 5′ to 3′ manner; however, the mechanism is different. Repli-
its own template for the replication process. DNA must be cation of the other parent strand, the lagging strand, is more
copied before mitosis can occur and must be copied in such complicated because the strands are antiparallel. As the
a way that each daughter cell will have the same amount of helix is opened, RNA primer sequences are added to the area
DNA and in the same sequence. Nearly all DNA replication of the opening fork, and the RNA primers are extended in a
is done in a bidirectional manner and is semiconservative 5′ to 3′ manner until the polymerase reaches the previously
in nature.3 Specifically, as enzymes involved in the replica- synthesized end. Rather than being replicated in a continu-
tion process open the double-stranded DNA helix, one ous manner, these replication forks open up all along the
strand of DNA is copied in a 5′ to 3′ manner, while the other lagging strand and are extended in this way. These small
strand is opened partially in sections and is copied 5′ to 3′ regions of newly replicated DNA are known as Okazaki
in sections as the double-stranded template continuously fragments.4 The fragments must be joined together to make
opens. Each strand of the parent DNA serves as a template a complete and continuous strand. This is accomplished by
for one of the newly synthesized strands, thereby maintain- the two enzymes DNA polymerase I and DNA ligase. The
ing the order of nucleotides. The replication process is RNA primers are synthesized and added to the DNA strands
shown in Figure 2–12. by an enzyme called primase, which anneals to the parent
In order for DNA to be replicated with the exact copying strands. After replication of the leading and lagging strands
of the template and its sequence into a new double-stranded is complete, DNA ligase joins the phosphodiester bonds
helix, many enzymes and proteins are involved. DNA repli- of the DNA backbone to complete the intact molecule.
cation occurs in specific steps, facilitated by certain enzymes Isomerase enzymes recoil the DNA; once this is completed
and other molecules at each step. First, sections of DNA must and the DNA is proofread by proofreading enzymes, the cell
be uncoiled from its supercoiled (or double-twisted) nature, can continue with mitosis and cell division.
and the two strands must be separated and kept apart; this is
done by the enzymes DNA gyrase (opens the supercoils) and DNA Repair
DNA helicase (separates the two strands of duplex DNA). DNA must be copied exactly or the information it contains
These enzymes, using energy derived from ATP hydrolysis, will be altered, possibly resulting in a decrease in the organ-
open the DNA molecules and keep the strands separate. In ism’s vitality. However, mistakes in the complex process of
the next step, DNA polymerase III can synthesize a new replication do occasionally occur, and a number of efficient
strand in the 5′ to 3′ direction on the leading strand. Proteins mechanisms have evolved to correct these mistakes. The
called single-stranded binding proteins interact with the open mechanisms can detect the mistakes, or changes, and correct
strands of DNA to prevent hydrogen bonding when it is not the actual DNA sequence. One of the most important mech-
needed during replication. DNA polymerase III also proof- anisms for correcting DNA replication errors is the proof-
reads the addition of new bases to the growing DNA strands reading ability of DNA polymerases. The proofreading
and can remove an incorrectly incorporated base, such as occurs in both the 5′ to 3′ and 3′ to 5′ directions and allows
G paired to T. the polymerase to backtrack on a recently copied DNA
In order for replication to take place on any piece of strand and remove an incorrect nucleotide, inserting the
DNA, there must first be a short oligonucleotide (composed correct one in its place. In addition to the proofreading abil-
of RNA) that binds to the beginning of the region to be ity of DNA polymerase enzymes, there is a second type of

The two new DNA strands have different features


Leading strand synthesis

Nucleotides added continuously to 3' end

3'
5'
5'
Leading strand
3'
Lagging strand 3'

5'
Lagging strand synthesis

Parental DNA Single strand Last fragment Previous fragment

Figure 2–12. DNA replication.


6888_Ch02_024-044 29/10/18 4:40 PM Page 35

Chapter 2 Basic Genetics 35

editing called mismatch repair, where an incorrect, noncom- be handled by recombination repair. Mismatch repair is
plementary nucleotide is removed and the correct one is activated when base pairing is incorrect and a bulge occurs
inserted in its place. in the duplex DNA. Mismatch repair enzymes are able to
In addition to errors in the primary nucleotide sequence remove the incorrect nucleotides and insert the correct ones.
of DNA molecules, there are other possible alterations that Methyl groups on adenines are used by the mismatch
can affect the way the sequence information is processed. enzyme systems to determine which nucleotide is correct and
Many different chemicals and environmental factors can alter which is a mistake. When DNA and cell damage occurs, SOS
DNA by modifying it chemically or physically. These include repair is induced. Damage can be caused by UV radiation,
alkylating agents, which react with guanine and result in chemical mutagens, and excessive heat, and by such treat-
depurination. Some cancer treatments are often based on the ments as exposure to cross-linking agents. There are certain
principle that the faster-replicating DNA in cancer cells does sections of highly conserved DNA that are activated when
greater damage and puts cancer cells at greater risk for cell DNA is damaged. The genes that are part of the SOS response
death than normal cells. Ionizing radiation and strong system must work in a coordinated manner to repair the
oxidants such as peroxides can cause single-strand breaks. damaged DNA through recombination events that remove
Ultraviolet (UV) radiation can alter thymine bases, resulting the damaged sections and replace them with the correct
in thymine dimers. Certain drugs such as the antibiotic sequences.
mitomycin C can form covalent linkages between bases on Repair systems have been studied closely in both
opposite strands, resulting in incorrect separation of the prokaryotes and eukaryotes. These systems are essential to
strands at that site during replication and subsequent muta- maintain the integrity of the DNA and minimize damage.
tions in daughter strands. Nearly all defects in DNA replica- The fact that so many repair systems exist in the cell proves
tion can be corrected by the various mechanisms used by the how important the correct structure and sequence of DNA
cell to maintain DNA integrity. However, if too many mis- is to cell viability. It also highlights how complex the DNA
takes occur, the repair systems can be overwhelmed, and molecule is in that many types of mutations can occur,
mistakes will not be corrected. The cell in which these mu- requiring a host of correction processes to repair them.
tations occur will have an altered DNA nucleotide sequence
and may or may not be viable. DNA Mutations
There are several major DNA repair systems. These in- Although many effective DNA proofreading and repair
clude photoreactivation, excision repair (also referred to as systems help to keep newly synthesized DNA from having
cut and patch repair), recombination repair, mismatch repair, mutations, none of the systems are foolproof and occa-
and SOS repair (Box 2–2). DNA repair systems can recog- sional mutations occur. Once a mutation is introduced into
nize mismatched base pairs, missing nucleotides, and a DNA coding strand, the information in that strand is now
altered nucleotides in DNA sequences. For example, when altered. It may be altered at the protein level if the mutation
thymine dimers are formed after exposure to UV light, the encodes for a different amino acid or a change in reading
photoreactivation enzyme becomes active and enzymatically frame. In general, a mutation is any change in the structure
cleaves the thymine dimers. Additionally, thymine dimers or sequence of DNA, whether it is physical or biochemical.
can be removed by the rather complex process of excision An organism is referred to as a mutant if its DNA sequence
repair, where the disrupted section of the DNA is removed. is different from that of the parent organism. The original
A cut is made on one side of the thymine dimer that bulges form of the DNA sequence, and the organism in which it
out from the rest of the duplex DNA. DNA polymerase I occurs, is called the wild type. The various chemicals and
synthesizes a short replacement strand for the damaged conditions that can cause mutations are referred to as
DNA section. The old strand is removed by DNA poly- mutagens. Many mutagens are also carcinogens, such as
merase I as it moves along the DNA and the newly formed the chemical benzene.
DNA segment is ligated into place. There are different types of mutations, and they may
Recombination repair uses the correct strand of DNA to have very different consequences for the organisms in
fill in the strand where the error was deleted. Polymerase I which they occur. Mutations can also be spontaneous. If
and DNA ligase then fill in the other strand. When large sec- they occur in the germinal tissue, they are passed on from
tions of DNA have been lost, the double-strand breaks can one generation to the next. The simplest type of mutation
is the point mutation, in which only one nucleotide in the
DNA sequence is changed. Point mutations include substi-
BOX 2–2
tutions, insertions, and deletions. Certain substitutions,
although they change the DNA sequence and the triplet
DNA Repair Systems codon(s), may not change the amino acid sequence in the
• Photoreactivation corresponding protein because of redundancy in the
• Excision repair genetic code. Recall that some of the amino acids have more
• Recombination repair than one codon that represents them. Therefore, if the new
• Mismatch repair codon is for the same amino acid, the mutation will be
• SOS repair silent, and the protein sequence will be the same. An
example of this is the amino acid threonine, which has ACA,
6888_Ch02_024-044 29/10/18 4:40 PM Page 36

36 PART I Fundamental Concepts

ACC, ACG, and ACU as possible codons. A substitution of the transferase protein of the A blood group.7 The frameshift
C, G, or U for A at the third position of the codon would mutation results in a nonfunctional transferase protein that
still have a peptide with threonine at that position. A type is seen phenotypically as the O blood group.
of “silent mutation” also occurs when a mutation happens Unusual genetic changes can also occur that cause gross
that causes a change in the peptide sequence, but that part mutations in the DNA sequence. Duplications, recombina-
of the peptide isn’t critical for its function; therefore, no tions, and large deletions have a lower frequency rate than
mutation is seen at the phenotypic level, such as enzyme the mutations in the replication of DNA noted above.
function or cell surface marker. A transition is a type of mu- Duplications can occur quite frequently and give rise to
tation in which one purine is substituted for another pseudogenes and other so-called junk DNA that does not
purine, or one pyrimidine is substituted for another pyrimi- code for proteins. A large part of DNA consists of such junk
dine. When a purine is substituted for a pyrimidine or a DNA without apparent ill effects and may even be necessary
pyrimidine for a purine, it is called a transversion. Exam- to some extent for the structural integrity of DNA and chro-
ples of DNA mutations are outlined in Table 2–4.5 mosomes. Also, there are highly conserved sequences from
Another type of mutation that can have a deleterious earlier evolution that are still present in human DNA, such
effect on the peptide sequence is called a missense point as Alu sequences. In addition, there may be evolutionary
mutation. A missense mutation results in a change in a pressures forcing duplications to occur. DNA contains short
codon, which alters the amino acid in the corresponding tandem repeats of various lengths throughout the entire
peptide. These changes cannot be accommodated by the DNA molecule, and these are useful in human identification
peptide while still maintaining its function. Typical examples and parentage testing.
of missense mutations include the alterations in the hemo- In blood banking, there are two good examples of dupli-
globin molecule at a single base pair, resulting in different cations. The first involves the glycophorin A and B genes; the
types of inherited anemias.6 A very specific type of serious second involves the genes RHD and RHCE. The Chido and
mutation, called a nonsense mutation, results when a point Rodgers blood group antigens, carried on the complement
change in one of the nucleotides of a DNA sequence causes components of the C4A and C4B genes, arose from duplica-
one of the three possible stop codons to be formed. The three tions and mutations of the C4 genes (refer to Chapter 8).
stop codons are amber (UAG), opal (UGA), and ochre In addition to these mutations, exchanges occasionally occur
(UAA), and these terminate the reading of the DNA between chromosomes, such as the Philadelphia chromo-
sequence, so the resulting peptide is truncated at its 3′ end. some present in chronic myelogenous leukemia, with a
A more severe type of mutation happens when there is an reciprocal translocation of genetic material between chromo-
insertion or deletion of one or more (but never multiplicities somes 9 and 22. Mutations involving recombination or cross-
of three) nucleotides in the DNA sequence. The result of this ing over take place during the process of meiosis in the
type of mutation is a change in the triplet codon sequence formation of gametes to generate sex cells that are different
and a subsequent alteration in the frameshift reading so that from the parental cells. Recombination involves breaking
a large change in the amino acid sequence occurs. Remember both double-stranded DNA homologues, exchanging both
that the DNA sequence is read three nucleotides at a time, strands of DNA, and then resolving the new DNA duplexes
called codons, and each codon can be charged with a specific by reconnecting phosphodiester bonds. Crossing over can be
amino acid. In transfusion medicine, it has been shown that single, double, or triple events (Fig. 2–13). An example of
there is a single base pair deletion in the gene encoding for such an event resulting in a hybrid formation is seen in the
MNSs blood group system. Crossover events have formed the
genes for the Dantu positive and GP.Hil phenotypes. One of
Table 2–4 Examples of DNA Mutations the most important events in the immune system is the
in Blood Groups recombination of the D, V, and J genes, which gives rise to the
vast array of immunoglobulin genes that are necessary to
Transition form the antibodies of the humoral system.
S antigen s antigen Deletion of large segments of DNA sequences covering
hundreds and possibly even thousands of nucleotides is also
Amino acid Methionine Threonine possible. It is a type of mutation that is not capable of being
mRNA code AUG ACG corrected by any of the DNA repair systems due to the size
and complexity of the mutation. Such mutations can result
DNA code TAC TGC in complete loss of a peptide, a severely truncated peptide,
Transversion or the formation of a nonfunctional peptide. An example of
this type of mutation in transfusion medicine is the Rodgers
N antigen He antigen negative phenotype, which results from a 30-kD deletion of
Amino acid Leucine Tryptophan both the complete CD4 (Rodgers) and 21-hydroxylase genes.8

mRNA code UUG UGG DNA Isolation


DNA code AAC ACC Genomic DNA is present in all nucleated cells. The ability
to successfully isolate DNA from the cell nucleus of cell
6888_Ch02_024-044 29/10/18 4:40 PM Page 37

Chapter 2 Basic Genetics 37

molecules within the cell. RNA requires greater care than


Crossing-over occurs at the 4-strand stage
DNA, as it is a single-stranded molecule and much more
labile. Chemicals that will inactivate RNase enzymes must
be used to obtain RNA that is not readily degraded and has
its sequence intact. As mRNA is only a small part of the total
RNA isolated, it can be purified by nature of its poly-A tail
using a column that contains poly-T attached to resin bead
molecules.
Methods such as complementary DNA (cDNA) synthesis,
in which RNA is converted into a DNA copy in vitro, allows
greater study of RNA sequences by making a more stable ver-
sion of it. RNA isolation also requires greater disruption of
the cellular material, usually with chaotropic agents such as
guanidine thiocyanate, lithium salts, and mechanical means
to disrupt cell membranes, such as douncing or shearing
with a needle and syringe.
Once DNA or RNA has been isolated, they can be stored
in low-salt buffers at the respective correct pH at –20°C or
lower for DNA and RNA at –80°C for many months to
many years. Tris-EDTA buffer (TE) solubilizes DNA and
Figure 2–13. Crossing over of DNA strands and chiasma formation at prophase I
RNA and protects against degradation. Because nucleic
of meiosis to create new sequences. (From Lewin B: Genes VIII. Prentice Hall/ acids are polymers, storage in water is not recommended
Pearson Education, New York, 2004, p 20. Reprinted with permission of the author.) as hydrolysis reactions will degrade them. Additionally,
water can contain substances such as DNase and RNase
that can degrade nucleic acids over time, even at low
cultures, blood, and other clinical specimens is fundamen- temperatures. The pH of the buffer used in nucleic acid
tal to optimal performance of other molecular procedures isolation and storage is critical, as acidic pH can result in
described in Chapter 4. The nucleic acid isolation method degradation of DNA and alkaline pH can degrade RNA.
should avoid damage of the nucleic acid and render it
free of other cellular contaminants, such as proteins or Ribonucleic Acid
lipids. The initial step involves lysing the cell to release the
cellular material. Common methods used for the organic Ribonucleic acid (RNA) is similar to DNA in structure but
isolation of nucleic acids use alkaline denaturation, phenol has certain key differences. It has a very different role to
extraction, and precipitation with alcohol. Inorganic isola- play in the cell. One of the key structural differences is that
tion utilizes low pH and high salt concentrations to precipi- unlike DNA, which is usually a double-stranded helix, RNA
tate proteins and leave the nucleic acid in solution. Nucleic occurs most often as a single-stranded structure, although
acids can be further purified from other unwanted cellular internal hydrogen bonding occurs frequently, probably to
components, such as the cytoplasmic membrane, by use of stabilize the RNA molecules. Both DNA and RNA are com-
specific columns or beads that will bind nucleic acids based prised of nucleotides, but in place of thymine in DNA there
on charge–charge interactions. Magnetic bead technology is uracil in RNA. Uracil is very similar to thymine except
is a commonly used, efficient method to isolate DNA that it lacks a methyl group. Another major difference is
and other molecules, and the procedure can be automated. the substitution of the sugar ribose for deoxyribose in the
Column chromatography has been in use for many years, backbone structure.
and different materials are used to prepare columns, In eukaryotes, RNA used to transmit genetic information
depending on the nature of the materials to be separated. (stored as DNA) from the nucleus to the cytoplasm is trans-
Silica particles can also be used to bind DNA under high- lated into peptides and proteins. DNA is copied into RNA by
salt conditions.9 Commercially prepared solid phase transcription, modified, and transported out of the nucleus
columns, containing columns or beads, are available as kits to the ribosomes, where it is translated into protein, which
that include the necessary lysing, buffering, and eluting is then modified if necessary for its proper function. There-
reagents. fore, RNA is the “go-between” from DNA, where the genetic
In earlier methods, DNA was isolated after treatment of information is stored to protein, the final product of the
cells with harsh mechanical or chemical agents often neces- expression of that genetic information. Proteins are the
sary to break open cell walls and disrupt proteins and fatty products of transcription and translation. This flow of
acids. Sonication, phenol, and chloroform, or various genetic information, from DNA to RNA to protein, is referred
chaotropic agents were used to disrupt cells, then followed to as the central dogma of molecular biology, described in
by high-speed centrifugation on cesium chloride or sucrose more detail in Chapter 4.
gradients to isolate the nucleic acids. This was necessary to In eukaryotes there are four major types of RNA; each
separate the high and low molecular weight DNA and RNA has a specific function as well as its own corresponding
6888_Ch02_024-044 29/10/18 4:40 PM Page 38

38 PART I Fundamental Concepts

polymerase. The first type of RNA molecule is ribosomal


tRNA is an adaptor
RNA (rRNA), which makes up a large part of the ribosomal
structure on the endoplasmic reticulum in the cytoplasm.
Amino acid linked
It is here that RNA is translated into peptide. RNA poly- to 3' end of tRNA
merase I transcribes rRNA. It is the most abundant and con-
sistent form of RNA in the cell. The second type of RNA is
messenger RNA (mRNA), the initial link between the infor-
mation stored in DNA and the translation of that informa-
tion into amino acids. It is this form that is transcribed from
DNA that encodes specific genes, such as those determining
the various blood groups. RNA polymerase II transcribes
mRNA. Unlike rRNA, mRNA molecules are very different
from each other, depending on the gene from which they Arm consists of:
were transcribed. In addition, eukaryotic mRNA undergoes Base-paired
stem
postsynthesis processing to remove introns before it can be
transferred out of the nucleus and translated. (See the Tran-
scription section for additional details on mRNA processing.)
The third major type of RNA is transfer RNA (tRNA); it is Single-
stranded loop
involved in delivering amino acids to the mRNA bound on Anticodon-
the ribosome. Each tRNA molecule can be charged with codon pairing
only one species of amino acid. However, as mentioned, the
genetic code is redundant, and many amino acids have more Figure 2–14. Schematic illustration of tRNA molecule and base pairing.
than one type of tRNA that codes for them. In addition, (Lewin B: Genes VIII. Prentice Hall/Pearson Education, New York, 2004, p 115.
tRNA has considerable internal hydrogen bonding to Reprinted with permission of the author.)
acquire its formal structure and keep it stabilized. All tRNA
molecules are long, single-stranded RNAs with a similar
secondary structure with intramolecular hydrogen bonding. Messenger RNA allows for highly efficient processing of the
They are often described as having a cloverleaf-shaped genetic code into the proteins that play nearly all the func-
conformation no matter which amino acid (aa) they are tional roles within a cell. Transcription begins when the
carrying to the translation machinery. There are two major enzyme RNA polymerase II binds to the region upstream (to
functional areas of the tRNA molecule. The first is the anti- the left of the 5′ start site) of a gene. Certain DNA short se-
codon: It consists of three nucleotides that hydrogen-bond quences, called consensus sequences, such as the CAAT box
to the correct site on the corresponding codon on the and TATA box, are located at specific sections upstream from
mRNA. It is this hydrogen bonding that assures the correct the gene to be transcribed. These are used to position RNA
aa is joined in the peptide chain by allowing only the correct polymerase properly so transcription of a gene starts in the
tRNA molecule with the right anticodon to bond to the correct position. This region is referred to as the promoter
correct mRNA codon. The second part of the tRNA mole- and is important in how and when a gene is expressed.
cule is at the 3′ hydroxyl end and binds an amino acid. Only Promoter regions also contain specific sequences that
one aa can bind to one tRNA molecule, with the specificity allow certain proteins, transcription factors, to bind to them
determined by the 3′ hydroxyl end. According to the recog- preferentially. This allows for certain genes to be more or less
nition region, an aminoacyl synthetase enzyme adds the active than other genes. Transcription factors are very critical
specific aa to the correct tRNA molecule. Only when the to proper cell growth and differentiation. Upstream
tRNA is charged with an amino acid can it transport it to sequences are not part of the mRNA itself and are never tran-
the ribosome for translation. See Figure 2–14. scribed; rather, they function to control transcription of
The fourth major type of RNA includes small and micro- mRNA. In addition to the promoter regions that can posi-
RNA molecules, which have various functions within the tively or negatively regulate various transcription factors and
cell. Some of these other functions include regulatory mech- other transcription-specific protein effectors, there are
anisms, cell development, and defense. These RNA mole- regions of DNA sequence called enhancers that can affect
cules, such as silencing RNA molecules, are necessary for transcription rates. Unlike promoters, these enhancer
proper gene expression and are altered in amount and type regions can have such effects without being close to the cod-
during cellular growth and differentiation. ing regions of the genes they influence. RNA is synthesized
in a 5′ to 3′ direction; transcription starts at the 3′ end of the
Transcription coding (or template) strand of the DNA duplex after it is
Transcription is the cellular process by which one strand of opened to two single strands and proceeds to the 5′ end. In
duplex DNA is copied into RNA. Although mRNA accounts this way, a 5′ to 3′ coding strand is generated with U in place
for only a small percentage of the total RNA inside a of T. The RNA transcript is complementary to the template
eukaryotic cell, it has the extremely important role of being strand and is equivalent to the coding strand of the DNA
a “transportable” and disposable form of the genetic code. molecule. Transcription is illustrated in Figure 2–15.
6888_Ch02_024-044 29/10/18 4:40 PM Page 39

Chapter 2 Basic Genetics 39

One strand of DNA is transcribed into RNA Eukaryotic mRNA is modified


and exported
5' TACGCGGTACGGTCAATGCATCTACCT
3' ATGCGCCATGCCAGTTACGTAGATGGA

5' UACGCGGUACGGUCAAUGCAUCUACCU

Figure 2–15. Transcription of RNA from DNA. (From Lewin B: Genes VIII.
Prentice Hall/Pearson Education, New York, 2004, p 241. Reprinted with permission
of the author.)

After RNA is transcribed, it is further processed before


it is transported to the ribosome and translated into pro-
tein. One major modification to eukaryotic mRNA is the
5′ 7-methyl guanosyl cap that is added to protect the
mRNA from degradation by nucleases. The cap also serves
as a recognition signal during translation. The 3′ end of
mRNA is modified by the addition of a string of adenines
that form a polyadenylation signal (poly-A tail), which
can vary in length from about 20 to 200 nucleotides and
is believed to increase mRNA stability. In addition to these
steps, eukaryotic mRNA has intervening sequences called
introns that do not code for protein or peptide and must
be removed before translation can begin. The process by Figure 2–16. Modification of mRNA and translation into protein. (Lewin B:
Genes VIII. Prentice Hall/Pearson Education, New York, 2004, p 122. Reprinted
which introns are removed from mRNA is called RNA splic- with permission of the author.)
ing. Specific nucleotide sequences at the beginning and
end of each intron signal the necessary enzymes to remove
the introns from the mRNA and degrade them. The tRNA-binding sites on the ribosome: the A site, P site, and
remaining sequences, which contain only exons that code E site. During translation, the charged tRNAmet must
for peptides and proteins, are joined together by ligation occupy the P site, and the A site next to the P site must
to form a mature mRNA molecule. The mature mRNA is have a charged tRNA with its correct matching amino acid
then transported out of the nucleus and to the ribosomes, in position.
where translation takes place. The process of translation involves three major steps:
initiation, elongation, and termination. The first event of the
Translation initiation sequence is the attachment of a free methionine to
Translation is the cellular process by which RNA tran- a transfer RNA molecule called tRNAmet, which requires the
scripts are turned into proteins and peptides, the functional presence of a high-energy molecule called guanine triphosphate
molecules of the cell (Fig. 2–16). Proteins can have many (GTP) and a special protein called initiation factor IF2. When
functions, such as structural, enzymatic, transport, or reg- GTP, IF2, and factors IF1 and IF3 are present at the ribosome,
ulatory. Through the genetic code, specific nucleic acid tRNAmet is able to bind the small 40S subunit of the ribosome
sequence is translated to a sequence of amino acids. Like to form an initiation complex. The larger subunit of the ribo-
DNA and RNA, proteins have a direction, reading their some, the 60S unit in eukaryotes, also binds to the complex
sequences left to right from the amino (NH2-) terminal to and hydrolyzes the GTP; then, the IFs are released.
the carboxy (-COOH) terminal. Peptides are composed of During the elongation step of translation, the incoming
amino acids joined to make a linear chain. Peptides consist tRNA binds to the A site in the presence of the elongation
of one strand of amino acids, and polypeptides (proteins) factor called E2F. Again, GTP is hydrolyzed as the energy
consist of more than one strand. Many peptide strands have source to move the tRNA in the A site to the P site. Peptidyl-
only primary (linear) and secondary (pleated or coiled) tRNA binds in the P site. The growing polypeptide is trans-
structure. Proteins can also have more complicated tertiary ferred from the P site to the next amino acid in the A site
(folded further) and quaternary (interaction between pro- carried by the aminoacyl-tRNA. Deacetylated tRNA, lacking
teins) structure. Translation takes place on the ribosomes any amino acid, exits the ribosome from the E site. The route
in the cytoplasm. Eukaryotic ribosomes are composed of a through the ribosome of an incoming tRNA charged with an
40S small subunit and a 60S large subunit. There are three amino acid is entry at the A site, then moves to the P site and
6888_Ch02_024-044 29/10/18 4:40 PM Page 40

40 PART I Fundamental Concepts

exits via the E site (Fig. 2-17). The ribosomes move down Protein synthesis has 3 stages
the codons on the mRNA one at a time, adding a correct Initiation 40s subunit on mRNA binding site
amino acid to the growing peptide chain. As the ribosome is joined by 60s subunit, and aminoacyl
moves down the mRNA, it eventually comes to one of the tRNA binds next
three stop codons—UAA, UGA, or UAG—and translation of
that mRNA is finished.
In the termination step, the completed polypeptide chain
is released and the ribosome dissociates from the mRNA.
Factors help the ribosome units to separate, and the peptide
chain is further processed. The three stages of translation are
illustrated in Figure 2-18.
Post-translational processing can consist of glycosylation
(addition of a sugar), phosphorylation (addition of a phos-
phate) or the removal of leader peptide sequences that are
used to traffic proteins to the cell membrane. Processing can
also consist of complicated folding schemes that ensure the
protein is in its correct tertiary form, such as the formation
of disulfide bonds via cysteine aa residues. A large family of
proteins called heat-shock proteins assists new proteins to
be folded correctly and stabilized by binding to hydrophobic
(water-avoiding) sections of the nascent protein chain.
Hydrogen bonding, van der Waals forces, and hydrophilic
(water-attracting) regions of proteins also help to give the
new protein its correct three-dimensional conformation.
After translation is complete, the mRNA is rapidly degraded
by enzymes (a process that helps to control gene expression)
or attaches to another ribosome, and the entire translation
process starts again.

Common Approaches in Modern


Genetics Techniques Figure 2–18. Stages of protein synthesis in translation. (Lewin B: Genes VIII.
Prentice Hall/Pearson Education, New York, 2004, p 137. Reprinted with permission
of the author.)
Over the past four decades, the knowledge of genetics has
advanced exponentially. Understanding the biochemical and
biophysical nature of DNA, RNA, and peptides and proteins
has allowed techniques to be developed to study these based on the biochemical and biophysical properties of genes
important molecules in great detail and to define the ways and chromosomes.
in which they interact in cells, groups of cells, complex mul- The following approaches are common to most tech-
ticellular organisms, and even in an entire human being. niques used in molecular biology:
Most of the techniques used in molecular biology (the 1. Any material that is to be studied must first be isolated
science of studying and manipulating genetic material) are intact without structural damage. This is often done with
chemicals that interact with one part of the cellular
material and not another. The methods to perform this
Peptide bond synthesis involves transfer of can be based on pH changes, salt gradients, detergent
polypeptide to aa-tRNA lysis, and enzyme activation.
2. There must be a way to visualize and locate the molecular
species to be studied. This is often done with probes that
act as signals or beacons that will bind specifically to the
molecule being analyzed; for example, DNA, by interac-
tion with parts of the molecule, such as hybridization
with the hydrogen bonds of DNA. The probes can be syn-
thesized with radiolabels, chemiluminescence molecules,
fluorochromes, or nanoparticles.
Figure 2–17. Peptide bond synthesis in translation. (Lewin B: Genes VIII. 3. Methods to amplify a specific DNA sequence, such as
Prentice Hall/Pearson Education, New York, 2004, p 137. Reprinted with permission PCR, can generate millions of copies of a particular
of the author.) sequence for analysis.
6888_Ch02_024-044 29/10/18 4:40 PM Page 41

Chapter 2 Basic Genetics 41

4. A method to separate out different species and subspecies disease treatment and prevention that takes into account
being studied, such as different sequences of DNA, must individual variability in genes, environment, and lifestyle
be available. An example of this involves separating for each person.”13 Pharmacogenomics is a segment of
nucleic acids by gel electrophoresis, binding them to precision medicine that investigates how an individual’s
columns, hybridizing them to membranes, or binding genes affect response to particular drugs and evaluates
them to other molecules. safe dosage of medications. With precision medicine,
5. A method to quantify the isolated and studied species information obtained from an individual’s genes, proteins,
must be used to obtain exact differences in amount for and environment can provide insight for disease preven-
the process under evaluation. Changes in optical density tion, targeted therapies, and diagnosis. Without question,
when exposed to UV radiation, radioisotope emission the science of genetics and the application of genetic
levels, fluorescence energy detection, and chemilumines- information will play an increasing role in treatment
cence emission detected by optical imaging instruments decisions.
can all be used to obtain data.
Several techniques used in molecular biology are reviewed
in detail in Chapter 4. CASE STUDY
Case 2-1
The Role of Genetics in the Future
History
The foundation for the science of genetics was developed
by the early research of various investigators working During the overnight shift in the blood bank of a small
with plants and bacteria. The hereditary basis of all living community hospital, a nurse from the newborn nursery
organisms is found in the sequence of its DNA. There are calls the lab to report a problem. A mother delivered an
many characteristics of nucleic acids that make them infant, her second child, earlier that day. She is dis-
useful for analyses for clinical purposes. The information tressed after inquiring about the blood type of the baby.
contained in the sequence of nucleotides that comprise From her prenatal testing, the mother knows that her
DNA provides the basis for genetic variation and patho- blood type is A Rh positive. She doesn’t know her hus-
logical traits in individuals. DNA-based tests are particu- band’s blood type, but the blood type of her first child is
larly applicable to transfusion medicine because the genes also A Rh positive. The blood type of the newborn was
encoding blood group antigens are known and the single reported to the nursery as O Rh negative on the cord
nucleotide and the genetic polymorphisms associated with blood specimen submitted to the lab. The mother insists
those antigens have been discovered. The remarkable pace the blood type is incorrect and requests the testing
of growth in molecular techniques combined with the be repeated. She is concerned the baby may have been
knowledge of the molecular bases associated with most switched in the nursery. What would explain the new-
blood groups allow application to many transfusion- born’s blood type?
related issues.10 1. Based on the information available, what are the pos-
Advances in molecular biology have facilitated the pro- sible ABO type(s) of the baby’s father that could result
gression into the age of molecular genetics. The technology in a group O infant?
is available to sequence genes more rapidly, allowing insight a. The father can only be group O
to disease at the nucleic acid level. DNA-based testing con- b. The father can only be group A or O
tinues to revolutionize clinical laboratory testing, with appli- c. The father can be group A, B, or AB
cations in cancer diagnosis, coagulation therapy, molecular d. The father can be group A, B, or O
pathology, prenatal practice, and the detection of infectious 2. If the blood types of the mother (A Rh positive) and
organisms.11 baby (O Rh negative), are both correct, what is the
On a broader scale, molecular testing is revolutionizing most likely explanation of the difference in the Rh
not only the testing in clinical laboratories but also the way type?
treatment decisions are made for patients. Genetic variation a. The mother must be Rh negative for the baby to be
can impact response to many types of treatment. An example Rh negative. The testing by the prenatal clinic must
of genetic variation influencing response to treatment can be be incorrect.
found with chronic hepatitis C (HCV) infection. Infected b. The baby can be Rh negative only if the father is Rh
patients of European ancestry have a significantly higher negative.
cure rate than patients of African ancestry when treated with c. It is possible for both parents to be Rh positive and
various peginterferon (PegIFN) drugs due to a genetic poly- produce a child who is Rh negative if each parent
morphism near the IL28B gene.12 inherited only one RHD gene.
The term precision medicine, often used interchangeably d. There was a switch in the nursery; this could not be
with personalized medicine, is defined by the National her baby.
Institutes of Health (NIH) as “an emerging approach for
6888_Ch02_024-044 29/10/18 4:40 PM Page 42

42 PART I Fundamental Concepts

SUMMARY CHART
 Genetics is defined as the study of inheritance or produces a complementary duplicate strand of nucleic
the transmission of characteristics from parents to acid. Each strand of DNA can act as a template to
offspring. It is based on the biochemical structure of be copied to make the opposite strand. DNA has a
chromatin, which includes nucleic acids and the struc- direction and is always read and written in the 5′ (left)
tural proteins that constitute the genetic material as to 3′ (right) direction unless specified differently for
well as various enzymes that assist in genetic processes certain applications.
such as replication.  Mutation refers to any structural alteration of DNA in
 All living organisms have specific numbers of chromo- an organism (mutant) that is caused by a physical or
somes. Humans have 22 pairs of autosomes and one chemical agent (mutagen) and can occur sponta-
set of sex chromosomes, females (XX) and males (XY), neously. Mutations can be beneficial or deleterious.
giving a total of 46 chromosomes in diploid cells. Some mutations are lethal and cannot be passed on to
 Mendel’s law of independent assortment states that another generation. Some mutations are silent and
factors for different characteristics are inherited have no consequence on the organism in which they
independent of each other if they reside on different occur and therefore have no selective pressure against
chromosomes. them in the population.
 Human chromosomes are composed of the genetic  Transcription is an enzymatic process whereby genetic
material chromatin, a complex of the nucleic acid poly- information in a DNA strand is copied into an mRNA
mer DNA wrapped around highly basic proteins called complementary strand. Eukaryotic mRNA is modified
histones. The helical structure of DNA allows a huge after it is made by various processing steps, such as the
amount of information to be packaged in a very small removal of introns and addition of a poly-A tail to
amount of space. the 3′ end. These processing steps take place in the
 Mitosis is the process by which somatic cells divide, nucleus of the cell before the mRNA is exported to the
with an end result of two identical daughter cells. cytoplasmic ribosomes for translation.
 Meiosis is the cell division process that takes place in  Translation is the complex process by which mRNA,
germ cells that concludes in the production of haploid which contains a mobile version of the DNA template
gametes, with an end result of four unique daughter encoding the genes for an organism, is turned into pro-
cells. teins, which are the functional units of an organism and
the cells of which it consists. Translation occurs on the
 Nucleic acids, DNA and RNA, are comprised of three
ribosomes, where there are three binding sites for tRNA.
components: a nitrogenous base, a sugar, and one or
Additional steps may be necessary to fold a specific pro-
more phosphates. The primary function of DNA is the
tein into its final mature form, such as proteins that re-
storage of genetic information. For the information to
quire disulfide linkages and proteins that are positioned
be utilized, it must be transcribed and then translated
within the cell membrane. Proteins are made of strings
to a protein in a process called gene expression.
of amino acids and are always read in an amino terminal
 Replication of DNA is semiconservative and is accom- (left) to carboxyl terminal (right) direction.
plished via the enzyme DNA polymerase, which

Review Questions 2. When a recessive trait is expressed:


a. One gene carrying the trait was present.
1. Which statement best describes the process of mitosis? b. Two genes carrying the trait were present.
a. Cell division by which only one-half of the daughter c. No gene carrying the trait was present.
cells produced are identical to the parent cell d. The gene is hemizygous.
b. Cell division of germ cells by which two successive 3. In a pedigree analysis, which statement about the symbols
divisions of the nucleus produce cells that contain half used is true?
the number of chromosomes of somatic cells a. Deceased family members have a line crossed through
c. Cell division that produces four daughter cells having the symbol.
the same number of chromosomes as the parent b. A consanguineous mating is indicated by a single line
d. Cell division that produces two daughter cells with the between a male and female.
same number of chromosomes as the parent cell c. The propositus is always found in the last generation
on the pedigree.
d. A stillbirth is indicated by a triangle.
6888_Ch02_024-044 29/10/18 4:40 PM Page 43

Chapter 2 Basic Genetics 43

4. Which of the following nitrogenous bases make up DNA? 12. Which statement correctly describes DNA replication in
a. Adenine, leucine, guanine, thymine eukaryotes?
b. Alanine, cytosine, guanine, thymine a. Semiconservative replication from RNA; requires
c. Adenine, lysine, uracil, guanine DNA polymerase
d. Adenine, cytosine, guanine, thymine b. Bidirectional replication; requires RNA primer
c. Conservative replication from DNA; requires RNA
5. Mutations can occur following DNA replication that
polymerase
escapes the proofreading and repair systems. Which state-
d. Occurs at the replication fork; both strands replicated
ment about DNA mutations is true?
in the same direction
a. All mutations result in a phenotypic change.
b. A frameshift mutation at the beginning of the coding 13. How is tRNA different from other types of RNA?
sequence is most likely to result in a phenotypic a. It has a 3′ poly-T tail.
change. b. The immature form contains introns.
c. A transversion always encodes for a stop codon. c. It has a 3′ methylated cap.
d. A missense point mutation never encodes for a stop d. It recognizes amino acids and nucleic acids.
codon.
14. Which statement about translation of proteins is false?
6. Which phenotype would be expected from the mating of a. It occurs on the ribosomes in the cytoplasm of
a Jk(a+b–) female and a Jk(a–b+) male? the cell.
a. Jk(a+b–) b. Post-translation processing can include glycosylation.
b. Jk(a+b+) c. mRNA delivers the amino acids to the growing
c. Jk(a–b+) peptide chain during elongation.
d. All of the above d. The codon UAA would terminate translation.

7. Which statement describes an intron? 15. The purpose of meiosis is to:


a. The part of a gene that contains nonsense mutations a. Generate two identical daughter cells after division.
b. The coding region of a gene b. Generate four identical daughter cells after division
c. The noncoding region of a gene with the same number of chromosomes as the parent
d. The resting stage between cell divisions cells.
c. Generate gametes with a diploid number of
8. Which statement about isolation of nucleic acids is true?
chromosomes.
a. All isolation methods involve the use of organic d. Generate daughter cells that contain half the number
solvents. of chromosomes of somatic cells that contain new
b. High protein concentration increases the DNA yield. DNA sequences.
c. mRNA can be effectively isolated with the use of
poly-A–coated beads. 16. The pattern of inheritance most commonly expressed by
d. Silica particles bind DNA under high salt con- blood group genes is:
centrations. a. X-linked recessive.
b. Autosomal recessive.
9. The purpose of transcription is to:
c. Autosomal codominant.
a. Produce a protein. d. X-linked codominant.
b. Read the mRNA by the ribosome.
c. Synthesize RNA using DNA as a template. References
d. Replicate DNA.
1. Watson JD, Crick FHC. Molecular structure of nucleic acids: a
10. When a male possesses a phenotypic trait that he passes structure for deoxyribose nucleic acid. Nature. 1953;171:737.
to all his daughters and none of his sons, the trait is said 2. Garrett RH, Grisham CM. Structure of nucleic acids. In Garret
to be: RH, Grisham CM. Biochemistry. 4th ed. Boston: Brooks/Cole,
Cengage Learning; 2010. p. 316-53.
a. X-linked dominant. 3. Knippers R, Ruff J. The initiation of eukaryotic DNA replication.
b. X-linked recessive. In: Fanning E, Knippers R, Winnacker E-L, editors. DNA replica-
c. Autosomal dominant. tion and the cell cycle. New York: Springer-Verlag; 1992. p. 2-10.
d. Autosomal recessive. 4. Okazaki R, Okazaki T, Sakabe K, Sugimoto K, Kainuma R,
Sugino A et al. In vivo mechanism of DNA chain growth. Cold
11. When a female possesses a phenotypic trait that she Spring Harb Symp Quant Biol. 1968;33:129-44.
passes to all of her sons and none of her daughters, the 5. Antonarakis SE, Cooper DN. Human gene mutation: mecha-
nisms and consequences. In: Speicher MR, Antonarakis SE,
trait is said to be: Motulsky AG, editors. Vogel and Motulsky’s human genetics:
a. X-linked dominant. problems and approaches. 4th ed. Berlin: Springer-Verlag; 2010.
b. X-linked recessive. p. 319-51.
c. Autosomal dominant. 6. Caskey CT, Pizzuti A, Fu YH, Fenwick Jr RG, Nelson DL.
Triplet repeat mutations in human disease. Science. 1992;
d. Autosomal recessive. 256:784-89.
6888_Ch02_024-044 29/10/18 4:40 PM Page 44

44 PART I Fundamental Concepts

7. Yamamoto F, White T, Marken J, Hakomori S. Molecular ge- Buckingham L. Molecular diagnostics: fundamentals, methods and
netic basis of the histo-blood group ABO system. Nature. 1990; clinical applications. 2 ed. Philadelphia: FA Davis; 2012.
345:229. Calladine CR, Drew HR, Luise BF, Travers AA. Understanding DNA,
8. Carroll MC, Palsdottir A, Belt KT, Porter RR. Deletion of the molecule and how it works. 3rd ed. San Diego: Elsevier;
complement C4 and steroid 21-hydroxylase genes in the HLA 2004.
class III region. EMBO J. 1985;4(10):2547-52. Clark DP, Russell LD. Molecular biology made simple and fun.
9. Carter MJ, Milton ID. An inexpensive and simple method for 4th ed. St. Louis (MO): Cache River Press; 2010.
DNA purification on silica particles. Nucleic Acids Res. Fung M, Grossman B, Hillyer C, Westhoff C. Blood group genetics:
1993;21(4):1044. technical manual. 18th ed. Bethesda (MD): AABB; 2014.
10. Westhoff CM. Molecular testing for transfusion medicine. Curr Fung M, Grossman B, Hillyer C, Westhoff C. Molecular biology
Opin Hematol. 2006;13:471-75. and immunology in transfusion medicine: technical manual.
11. Westhoff CM. The potential of blood group genotyping for 18th ed. Bethesda (MD): AABB; 2014.
transfusion medicine practice. Immunohematology. 2008; Glick BR, Pasternak JJ, Patten CL. Molecular biotechnology:
24(4):190-95. principles and applications of recombinant DNA. 4th ed.
12. Ge D, Fellay J, Thompson AJ, Simon JS, Shianna KV, Urban TJ, Washington, DC: ASM Press; 2009.
et al. Genetic variation in IL28B predicts hepatitis C treatment- Harmening DM. Modern blood banking and transfusion practices.
induced viral clearance. Nature. 2009;461(7262):399-401. 6th ed. Philadelphia: FA Davis; 2012.
13. Genetics Home Reference [Internet]. Bethesda (MD): National Hillyer CD, Silberstein LE, Ness PM, Anderson KC, Roback J. Blood
Library of Medicine (US) [updated 2015 Apr; cited 2017 banking and transfusion medicine, basic principles and practice.
Jul 18]. Available from: https://ghr.nlm.nih.gov/. Philadelphia: Churchill Livingstone; 2007.
Krebs J, Goldstein E, Kilpatrick S. Lewin’s genes XII. Burlington
Bibliography (MA): Jones and Bartlett Learning; 2018.
Lewin B. Genes VIII. New York: Prentice Hall; 2003.
Alberts B, et al. Molecular biology of the cell. 4th ed. New York: Quinley ED. Immunohematology: principles and practice. 3rd ed.
Garland Science; 2002. Philadelphia: Lippincott Williams & Wilkins; 2010.
Brown TA. Introduction to genetics: a molecular approach. London: Trent RJ. Molecular medicine. 3rd ed. Burlington (MA): Elsevier
Garland Science; 2011. Academic Press; 2005.
6888_Ch03_045-076 29/10/18 4:44 PM Page 45

Chapter 3
Fundamentals of Immunology
Lorraine Caruccio, PhD, MT(ASCP)SBB and Nadine M. Lerret, PhD, MLS(ASCP)

Introduction Membrane Attack Complex Enhancement Media


Overview of the Immune System Binding of Complement by RBC Protein Media
Innate and Acquired Immunity Antibodies Low Ionic Strength Solution Media
Innate Immunity Characteristics of Antigens Polyethylene Glycol and Polybrene
Acquired Immunity Characteristics of Blood Group Antibodies Proteolytic Enzymes
Cellular and Humoral Immunity Polyclonal and Monoclonal Antibodies Antihuman Globulin Reagents
Cells and Organs of the Immune System Naturally Occurring and Immune Chemical Reduction of IgG and IgM
B Cells Antibodies Molecules
T Cells Unexpected Antibodies Monoclonal Versus Polyclonal Reagents
Antigen-Presenting Cells Alloantibodies and Autoantibodies Nontraditional Laboratory Methods
Immune System Organs Characteristics of Antigen-Antibody Flow Cytometry
Reactions Diseases Important in Blood Bank
Immune Maturation
Intermolecular Binding Forces Serologic Testing
Cell Lineages and Markers
Antibody Properties Immunodeficiency
Cytokines and Immunoregulatory
Molecules Host Factors Hypersensitivity
Immune System Genetics Tolerance Monoclonal and Polyclonal
Characteristics of Immunoglobulins Detection of RBC Antigen-Antibody Gammopathies
Reactions Autoimmune Disease
Immunoglobulin Structure
Traditional Laboratory Testing Methods Hemolytic Disease of the Newborn
Immunoglobulins Significant for
Blood Banking Factors That Influence Agglutination Blood Product Transfusions and the
Reactions Immune System
Immunoglobulin Variations
Centrifugation Summary Chart
Immunoglobulin FC Receptors
Antigen-Antibody Ratio Review Questions
Complement System
Effect of pH References
Classical Complement Pathway
Temperature
Alternative Complement Pathway
Immunoglobulin Type
Lectin Complement Pathway

OBJECTIVES
1. Outline the different components of the immune system and identify their functions.
2. Describe the characteristics of the major cells of the immune system and their functions.
3. List the major effector molecules and the roles they play in the immune response.
4. Outline the basic steps of hematopoiesis in the immune system.
5. Explain the function of the major histocompatibility complex (MHC) class I and II molecules.
6. Describe the physical characteristics of immunoglobulins in relation to structure and list the different subtypes.
7. Explain the activation sequences of the three major complement pathways and describe how they come to a common
starting point.
8. List the methods used in the blood bank to detect antibodies and complement bound to red blood cells.
9. Describe the immune response, including antigen-antibody reactions, lymphocyte functions, and host factors that can activate
and suppress the immune system.
Continued

45
6888_Ch03_045-076 29/10/18 4:44 PM Page 46

46 PART I Fundamental Concepts

OBJECTIVES—cont’d
10. List the traditional laboratory techniques used in blood bank testing.
11. Identify the various factors that affect agglutination reactions.
12. Describe some of the common diseases that can affect blood bank testing.

Introduction reactions are influenced by a number of factors, including


distance, antigen-antibody ratio, pH, temperature, and
The immune system (IS) is complicated, tightly controlled, immunoglobulin type (Box 3–1).
and includes tissues, organs, cells, and biological mediators Both traditional and nontraditional laboratory testing
that coordinate to defend a host organism against intrusion methods are currently utilized in the transfusion service.
by a foreign substance or abnormal cells of self-origin. These methods focus on either the detection of antigens or
Immunity refers to the process by which a host organism on the detection of antibodies. This chapter provides an
protects itself from attacks by external and internal agents. introduction to the many areas of immunology and how they
Immunity also confers protection from nonself and abnormal impact transfusion medicine. It will include a brief introduc-
self-elements, which are controlled at different levels. The tion to the biology and biochemistry of the immune system
number of different types of nonself organisms includes and the application of testing procedures used in evaluating
unicellular and multicellular organisms, such as viruses, the immune response as related to blood transfusion. A brief
bacteria, mycoplasma, fungi, and parasites. Additionally, description of immune-mediated diseases important to trans-
tumor cells and cells destined for termination within the host fusion medicine is included at the end of the chapter.
must be recognized and eliminated.
The response and elimination of organisms and unwanted Overview of the Immune System
cells is accomplished through cellular and/or humoral
mechanisms. The cellular defense mechanism is mediated by All organisms at all levels of life are continually challenged
various cells of the IS, such as macrophages, T cells, and by potentially harmful organisms and molecules from their
dendritic cells, which function to eliminate viruses, bacteria, environment and must be protected against them. One of
cancer cells, and other cellular pathogens. The humoral the most fundamental concepts in immunology is the idea
mechanism is comprised of B cells and the specific antibodies of self versus nonself and how the IS distinguishes between
they produce, along with complement components that are the two. The two terms have now been broadened in mean-
produced in plasma, saliva, and other bodily secretions. These ing. Self refers to anything that is derived from the host
antibodies (immunoglobulins) bind to specific receptor sites genome and the rearrangement of host genes. It includes
on cells. Complement may also bind to immunoglobulin cells, fluids, molecules, and more complex structures of a
molecules that have specific complement receptor sites. The host organism. Anything put into the host body, even a close
extent of activation and the amount of damage that occurs genetic match, can be regarded as nonself and therefore can
to target cells is dependent upon the complement pathway be rejected by a response of the IS. Nonself refers to anything
involved and on several other host factors. physically outside the host, whether a living organism
Antibodies, or immunoglobulins, have special signifi- (parasites, fungus) or nonliving toxin (poison ivy fluid,
cance for transfusion medicine, because antigens present insect venom). When foreign objects or damaged host cells
on transfused cells may cause reactions in the recipient and are detected by the IS, an immune response occurs. Immune
complicate therapy. The majority of blood bank testing is responses occur at different levels and are either primary
focused on the prevention, detection, and identification of (natural, innate) or secondary (adaptive, acquired). The
blood group antibodies and on the typing of red blood cell overall mechanisms and components of each are outlined
(RBC) antigens. Antigen characteristics, as well as host fac- in Table 3–1 and Table 3–2.
tors, have an impact on the immune response. Knowledge The innate immune response consists of physical barri-
of these factors and characteristics enables testing problems ers, biochemical effectors, and immune cells. The first step
to be resolved or prevented from occurring in the first
place. Understanding the IS and its responses is essential
to understanding the factors that can affect agglutination BOX 3–1
reactions between RBCs and antibodies and various testing
modifications. Factors Affecting Antigen-Antibody Reactions
Detection of alloantibodies or autoantibodies in routine • Distance
blood bank testing procedures is of the utmost importance • Antigen-antibody ratio
in providing compatible blood to patients. This detection is • pH
dependent upon several factors, including binding forces • Temperature
between antigens and antibodies, properties of the antibody • Immunoglobulin type
itself, and individual host characteristics. Antigen-antibody
6888_Ch03_045-076 29/10/18 4:44 PM Page 47

Chapter 3 Fundamentals of Immunology 47

Table 3–1 Comparison of the Major Mechanisms of the Immune System


Innate or Natural Immunity Acquired or Adaptive Immunity
• Primary lines of defense • Supplements protection provided by innate immunity

• Early evolutionary development • Later evolutionary development—seen only in vertebrates

• Nonspecific • Specific

• Natural—present at birth • Specialized

• Immediately available • Acquired by contact with a specific foreign substance

• May be physical, biochemical, mechanical, or a • Initial contact with foreign substance triggers synthesis of specialized antibody proteins
combination of defense mechanisms resulting in reactivity to that particular foreign substance

• Mechanism does not alter on repeated exposure to • Memory


any specific antigen
• Response improves with each successive encounter with the same pathogen

• Remembers the infectious agent and can prevent it from causing disease later

• Immunity to withstand and resist subsequent exposure to the same foreign substance
is acquired

Table 3–2 Cellular and Humoral Components of the Immune System


Innate or Natural Immunity Acquired or Adaptive Immunity
First Line of Defense Second Line of Defense Third Line of Defense

Internal Components Internal Components Internal Components

Physical Cellular Cellular

• Intact skin • Phagocytic cells • Lymphocytes

• Mucous membranes • Macrophages-dendritic cells • T cells

• Cilia • Monocytes • TH

• Cough reflex • PMNs: Large granular leukocytes • TC

Biochemical • NK cells • T memory cells

• Secretions Humoral (fluid)/Biochemical • B cells

• Sweat • Complement-alternate pathway • B memory cells

• Tears • Cytokines • Plasma cells

• Saliva • Interferons Humoral

• Mucus • Interleukins • Antibodies

• Very low pH of vagina and stomach • Acute inflammatory reaction • Complement-classic pathway

• Cytokines

of innate defense is external, including skin and enzymes Importantly, it is the acquired immune response that
present on the skin’s surface. The second line of innate de- responds more rapidly and efficiently against a repeat attack
fense is internal and can recognize common invaders with a from a pathogen by establishing the memory response. The
nonspecific response, such as phagocytosis that does not IS’s specificity also prevents the host from becoming attacked
have to be primed. The last line of defense is the acquired and damaged during an immune response. The localized
immune response, which needs time (days to weeks) and nature of an immune reaction also prevents systemic damage
reorganization to mount an effective and specific reaction. throughout the host organism. The wide variety of potential
6888_Ch03_045-076 29/10/18 4:44 PM Page 48

48 PART I Fundamental Concepts

organisms and substances that can invade the host requires the macrophages in tissues). Phagocytic cells collaborate
a vast array of means to recognize and remove them. The with molecules of the IS. Opsonins are factors that include
acquired immune response must be capable of generating antibodies and complement components in plasma that coat
a near-infinite level of specific responses to all the different pathogens and facilitate phagocytosis. When phagocytes
complex organisms and substances that the host can ingest foreign cells and destroy them, they can become
encounter over its lifetime. activated to release soluble polypeptide substances called
cytokines that have numerous effects on other cells of the
Innate and Acquired Immunity immune and vascular systems (Table 3–3 is a partial list of
cytokines). There are a large number of different cytokines;
The immune system has two major arms, innate and some with unique functions and others with overlapping
acquired immunity, that work to prevent infection and dam- functions. Some work together, and some oppose the func-
aged cells from destroying the host. The innate part of the tions of other cytokines. Many are secreted, and some are
system is more primitive and does not function in a specific membrane receptors. Cytokines help to regulate the immune
way; rather, it recognizes certain complex repeating patterns response in terms of specificity, intensity, and duration.
present on common invading organisms. The innate arm can Another important component of the innate immune sys-
function immediately to stop host organisms from being tem is the complement system. Complement has three
infected. Therefore, innate immunity is the immediate (first) major roles in immunity: (1) the final lysis of abnormal and
line of defense from invading pathogens. pathogenic cells via the binding of antibody, (2) opsoniza-
The acquired immune response is more advanced and was tion and phagocytosis, and (3) mediation of inflammation.
developed after vertebrates had evolved. It relies on the for- The proteins of the complement system are enzymes that are
mation of specific antigen-antibody complexes and specific normally found in the plasma in a proenzyme inactive state.
cellular responses. Acquired immunity allows for a specific Three ways the complement proteins can be activated are the
response, and IS memory allows resistance to a pathogen that classic, alternative, and lectin pathways; all have essentially
was previously encountered. the final result of cell lysis and inflammation. The classic
pathway uses antigen-antibody binding and therefore is a
Innate Immunity specific activator of complement. The alternative pathway
activates complement by recognizing polysaccharides and
There are two important features of innate immunity. First, liposaccharides found on the surfaces of bacteria and tumor
the innate immune system is nonspecific. The same response cells; therefore, it uses nonspecific methods of activation.
is used against invading organisms, no matter what the The lectin pathway is activated by mannose-binding proteins
source is, as long as the innate IS can recognize them as non- bound to macrophages.
self. Innate immunity is present at birth and does not have Inflammation is also a critical component of the innate
to be learned or acquired. Second, it does not need modifi- IS and is familiar to most people when they have a minor
cations to function and is not altered with repeated exposure wound that has redness and warmth at the abrasion site.
to the same antigen. Because innate immunity functions so Inflammation is initiated by any type of tissue damage,
well as a first line of defense, it was maintained in the IS of whether it be to the skin or to an internal organ. Burns,
vertebrates during evolution. infections, fractures, necrosis, and superficial wounds all
The innate IS is comprised of physical and biochemical elicit an inflammatory response that is characterized by an
barriers as well as numerous cells. Physical barriers in- increase in blood flow to the wounded area, increased blood
clude intact skin, mucous membranes, cilia lining the mu- vessel permeability at the site to allow for greater flow of
cous membranes, and cough reflexes. Biochemical barriers cells, a mobilization of phagocytic cells into the site, and
of the innate system include bactericidal enzymes such as a possible activation of acute phase and stress response pro-
lysozyme and RNases, fatty acids, sweat, digestive enzymes teins at the site of tissue damage. Eventually the wound is
in saliva, stomach acid, and low pH. Innate immune cells repaired, new tissue grows in place of the damaged tissue,
include phagocytic leukocytes and natural killer (NK) cells. and inflammation is stopped. Uncontrolled inflammation
Phagocytic cells of different types are found in most tissues can result in unwanted damage to healthy tissues; therefore,
and organs of individuals, including the brain, liver, intes- the regulation of inflammation is tightly controlled and
tines, lungs, and kidneys. Phagocytes such as circulating requires signals to effectively turn it on and off.
monocytes in the blood and peripheral macrophages can
move between vessel walls. Phagocytes recognize complex Acquired Immunity
molecular structures on the surface of invading cells or in
the secretions and fluids of the host body. They remove the Acquired immunity is the other major arm of the host’s IS
invading organisms by engulfing and digesting them with and is the most highly evolved. It is also the most specific
vesicle enzymes. and allows the IS to have memory of pathogens it has
Two major cells that can use phagocytosis to remove encountered previously. The acquired system is present only
pathogens are the polymorphonuclear cells (which include in vertebrates. The term acquired refers to the fact that the
neutrophils, basophils, and eosinophils) and the mono- immunity is acquired via specific contact with a pathogen or
nuclear cells (which include the monocytes in plasma and aberrant cell. The term adaptive refers to the ability to adapt
6888_Ch03_045-076 29/10/18 4:44 PM Page 49

Chapter 3 Fundamentals of Immunology 49

Table 3–3 Various Cytokines and Their Functions


Cytokine Source Stimulatory Function
IL-1 (IL-1α, IL-1β) Mf, monocytes Induction of proinflammatory proteins, enhances production of other
cytokines

IL-2 T Proliferation of activated T and B cells, activates NK cells

IL-3 T, NK, MC Hematopoietic growth factor

IL-4 T, MC, NK Proliferation of activated B, T, MC, induces B to produce IgE

IL-5 T, MC Differentiation/proliferation of eosinophils, activated B, induces B to


produce IgA

IL-6 T, Monocytes, BM Stroma Differentiation of myeloid stem cells, convert B to plasma cells

IL-7 BM and thymic stroma Production/differentiation of B, T

IL-8 MF, Monocytes Chemotaxis/activation of neutrophils

IL-9 T Proliferation of T and MC

IL-10 T, B, Mf, monocytes Downregulates MHC class II and cytokine production by monocytes, MF

IL-11 BM stroma Differentiation of pro-B and megakaryocytes

IL-12 B, Mf, monocytes Enhances NK and CD8+T cytotoxicity

IL-13 T, MC Upregulates MHC class II, induces B switch to IgG and IgE

IL-14 T, B, and T lineage lymphomas Proliferation of activated B

IL-15 T, B, NK, Mf, monocytes Proliferation of T, NK, activated B

IL-16 T Induces MHC class II, chemoattractant for CD4+T

IL-17 T Proinflammatory-stimulates cytokine production

IL-18 Mf Induces T to produce IFN, enhances NK cytotoxicity

IL-19 Monocytes Modulates T helper activity

IL-20 Monocytes Regulation of inflammatory responses

IL-21 T Regulation of hematopoiesis, activates B, differentiation of NK

IL-22 T Inhibits IL-4 production

IL-23 Dendritic cells Induces IFNγ production by T

IL-24 T, Mf, monocytes Induction of TNF, IL-1, IL-6

IL-25 T, Mf, MC Induction of IL-4, IL-5, IL-13

Colony Stimulating Factors

GM-CSF T, Mf, fibroblasts, MC, endothelium Growth of granulocyte and Mf colonies

G-CSF Fibroblasts, endothelium Growth of neutrophil progenitors

M-CSF Fibroblasts, endothelium, epithelium Growth of monocyte progenitors

Steel Factor BM stroma Enhances cell division

Tumor Necrosis Factors

TNFα Mf, T Tumor cytotoxicity, promotes cytokine secretion, activates Mf, antiviral

TNFβ T Tumor cytotoxicity, promotes phagocytosis by neutrophils and Mf, lymphoid


organ development, antiviral
Continued
6888_Ch03_045-076 29/10/18 4:44 PM Page 50

50 PART I Fundamental Concepts

Table 3–3 Cytokines and Their Functions—cont’d


Cytokine Source Stimulatory Function
Interferons

IFNα Leukocytes Enhances MHC class II, antiviral

IFNβ Fibroblasts Antiviral, enhances MHC class II

IFNγ T, NK Inhibits T helper, induces B to IgG2, antagonizes IL-4

Other

TGFβ T, B, Mf, MC Proinflammatory, induces B to produce IgA, promotes tissue repair


LIF Thymic epithelium, BM stroma Induces acute phase proteins

Adapted from Delves, PJ, Martin, SJ, Burton, DR, Roitt, IM: Roitt’s Essential Immunology, 13th ed. Wiley-Blackwell, 2017
APC = antigen-presenting cells; BM = bone marrow; CSIF = cytokine synthesis inhibitory factor, FC = immunoglobulin Fc receptor for IgE; G-CSF = granulocyte colony-stimulating
factor; GM-CSF = granulocyte monocyte/macrophage colony-stimulating factor; IFN = interferon; IL = interleukin; LIF = leukocyte inhibitory factor; MC = mast cell; M-CSF =
monocyte colony-stimulating factor; MHC = major histocompatibility complex; Mf = macrophage; NK = natural killer cells; PGE2 = prostaglandin E2; T = T lymphocyte; TGF =
transforming growth factor; TNF = tumor necrosis factor

to and destroy new complex pathogens, although it must globulin because they are a type of globular soluble protein.
first react to them through complex recognition processes. They are found in the gamma globulin portion of plasma
Acquired immunity is specific in recognition of new or serum when it is separated by fractionation or elec-
pathogens and has specific responses, depending on the type trophoresis. The function of the antibody is to bind to for-
of pathogen it encounters. eign molecules called antigens. Most antigens are found on
The acquired IS uses antibodies as specific immune the surface of foreign cells or on damaged self-cells.
effectors. Antigen specificity and uniqueness determine A key feature of antigen-antibody reactions is their speci-
the particular antibody that will bind to it. The antigen- ficity. Only one antibody reacts with one antigen, or one part
antibody complex is a three-dimensional interaction, which (an epitope or antigenic determinant) of a complex anti-
helps to prevent the binding and induction of an immune gen. An immune reaction against an antigen stimulates the
response by nonspecific antigens. For example, antibodies production of antibodies that will match the epitope of
against one blood group antigen do not react against an- the antigen. The binding reaction of antigen and antibody
other blood group antigen. An antigen that an antibody is has often been called a lock and key mechanism, referring
made against is sometimes referred to as its antithetical to its specific conformation. Antigen-antibody complex
antigen. Antibodies do not always remain in plasma at lev- formation inactivates the antigen and elicits a number of
els observable with serologic testing, and if antigen-positive complicated effector mechanisms that will ultimately result
RBC units are transfused in a sensitized patient, the second in the destruction of the antigen and the cell to which it is
antibody response against the transfused cell antigens can bound. The laboratory study of antigen-antibody reactions
be more vigorous, resulting in intravascular RBC hemoly- is called serology and has been the basis of blood bank tech-
sis. Due to this second (memory) response, obtaining the nology for many years. Antibody screening methods such
medical history of a patient who requires a transfusion is as the indirect antibody test and crossmatching techniques
absolutely critical. rely on the detection of antigen-antibody complexes by
screening for antibodies in the plasma or sera. The direct
Cellular and Humoral Immunity antiglobulin (Coombs) test relies on the detection of anti-
bodies (and complement) bound to the surface of RBCs.
The two major components of the vertebrate adaptive IS are
cellular and humoral immunity. Cellular immunity is medi- Cells and Organs of the
ated by various IS cells, such as macrophages, T cells, and Immune System
dendritic cells. Lymphokines are powerful molecules that in-
clude cytokines and chemokines. These effector molecules The different types of cells within the immune system can
play a critical role in cellular immunity by activating and de- be distinguished by the membrane markers they possess.
activating different cells, and allowing cells to communicate These are referred to as clusters of differentiation (CD)
throughout the host body. markers and are detected by immunotyping methods to
Humoral immunity consists of the fluid parts of the identify cells of the immune system. These cell surface mole-
IS, such as antibodies and complement components found cules specify cellular definitions and functions, including
in plasma, saliva, and other secretions. One of the most maturation levels and lineage specificity.
important parts of humoral immunity is the antibody, Lymphocytes, which are critical for acquired immunity,
which is produced by B cells. Antibodies are also called are divided into two major types, the T lymphocyte (T cell)
immunoglobulins—immune because of their function, and and the B lymphocyte (B cell). The T lymphocyte matures
6888_Ch03_045-076 29/10/18 4:44 PM Page 51

Chapter 3 Fundamentals of Immunology 51

in the thymus gland and is responsible for making cytokines that remains in circulation in the plasma, body secretions,
and destroying virally infected host cells. B lymphocytes and lymphatic system. Antibodies can neutralize toxic sub-
mature in the bone marrow and, when stimulated by an anti- stances and antigens that are encountered by binding to
gen, evolve into plasma cells that secrete antibody. Natural them and thus preventing them from interacting with the
killer cells are another type of lymphocyte that plays a host. When the antigenic site is nonreactive because of anti-
role in immune protection against viruses. Dendritic cells body binding, it cannot interact with host cells to infect them
are present throughout many systems of the body and are or damage them. Also, binding of antigen by antibody brings
responsible for antigen processing and presenting. Addition- about opsonization, which aids in the direct killing of
ally, macrophages can also process antigens. pathogens by cell lysis. When complement is activated by an
Lymphocytes recognize only one specific antigen, which antigen-antibody complex, the pathogen can be destroyed.
is determined by the genetic programming of that lympho- In blood bank testing procedures, antibody testing is one of
cyte. B cells undergo gene rearrangement in order to have the most important components.
the correct antibody made that can react with the correct
antigen. Because there are so many different antigens that T Cells
pathogens can carry, the IS has adapted to recognize mil-
lions of different antigens, with a specific response that will T cells are another very important component of the cellular
match only one particular antigen. Therefore, if a foreign part of acquired immunity. One of the major differences of
antigen is present, only a small percentage of the host’s cellular immunity compared to humoral immunity is that
antibodies and T-cell receptors will recognize it. In cases T cells recognize antigens that are internalized within a host
where an antigen is recognized by more than one antibody, cell. The antigens are then processed and presented on the
a process of clonal selection happens, in which the different host cell surface in small peptide fragments. T cell–mediated
cells that recognize the different epitopes of the antigen are immunity is involved in the response against fungal and viral
expanded. In transfusion medicine, this plays a role in infections, intracellular parasites, tissue grafts, and tumors.
selecting the best antibody reagents for testing, as not all T-cell receptors do not recognize foreign antigen on their
epitopes of a complex antigen, including blood group anti- own, as B cells do; they require help in the form of cell mem-
gens, will be equally reactive. brane proteins known as major histocompatibility complex
The maturation of B and T cells occurs after an antigen is (MHC) molecules. Therefore, there is a certain level of
encountered. Both B and T cells mature into what are called restriction placed on this acquired cellular branch of the IS,
effector cells, which are the functional units of the IS. The as determined by the inherited MHC molecules on host cells.
final effect that B and T cells can induce is the elimination The MHC genes determine the human leukocyte antigens
of pathogens and foreign cells. Immune memory is also (HLA) present on leukocytes and other cells. HLA molecules
acquired when lymphocytes mature into memory cells after have been known for many years to be a major determinant
antigenic stimulation; thus allowing the IS to recognize anti- of tissue graft rejection.
gens from previous encounters. Once an infection is cleared,
some of the cells that can recognize the antigen remain in
circulation. The lymphocytes are permanently set to respond Advanced Concepts
to the same antigen again by the process of memory. The There are two major classes of MHC genes and antigens:
second time this antigen is encountered, memory allows for MHC class I and MHC class II. MHC class I antigens are
a more effective and rapid immune response. Memory cells found on most nucleated cells in the body, and MHC class II
can persist for the lifetime of the host. It is one of the reasons antigens are found on most antigen-presenting cells. In hu-
that some immunizations with vaccines do not have to be mans, MHC class I genes code for the HLA-A, HLA-B, and
repeated, except on a cautionary basis to keep the immu- HLA-C antigens, whereas MHC class II genes code for
nization active. HLA-DR, HLA-DQ, and HLA-DC antigens. MHC classes I
and II are important in the recognition of foreign sub-
B Cells stances and the immune reactions against them.
There are two major functions of T cells. The first major
Antibodies can be secreted or membrane-bound but are function is to produce immune mediating substances such
produced exclusively by mature B cells called plasma cells. as cytokines, which influence many immune functions
The receptor on the B cell that recognizes the antigen is a throughout the body. The second major function is to
membrane-bound antibody. When the receptor antibody on kill cells that contain foreign antigen. When T cells are
the B cell reacts with a specific antigen and recognizes it, the activated by antigen, they start to secrete cytokines and
B cell is activated to divide. The cells produced from this change their cellular interactions (see Table 3–3). T cells
rapid division mature into plasma cells and memory B cells. are also grouped into two major categories with two major
Memory B cells have antibody on their surfaces that is of the functions: T helper (TH) cells and T cytotoxic (TC) cells.
same conformation as that of the B cell from which they were TH cells are also distinguished by the cell receptor CD4;
derived. TC cells are distinguished by the cell receptor CD8. TH cells
Plasma cells are antibody factories that make large are further grouped into TH1 and TH2 and respond to
amounts of one specific type of antibody in a soluble form
6888_Ch03_045-076 29/10/18 4:44 PM Page 52

52 PART I Fundamental Concepts

different cytokines. TH cells have the ability to recognize an antigen is first encountered. The lag phase, until an
antigen, along with MHC class II molecules, and provide appropriate immune response occurs, is called the latency,
help to B cells to evolve into plasma cells and make anti- pre-seroconversion, or window period. It is during this time
bodies. TH cells therefore determine which antigens that antibody cannot be detected with serologic testing. Dur-
become IS targets, as well as which immune mechanisms ing the latency period, however, T and B cells are very active
will be used against them. TH cells aid in the proliferation in processing antigen and initiating the primary response
of immune cells after they encounter antigen. to the antigen. The first antibodies made against the new
antigen are different from the antibodies of the secondary
response. The primary antibodies are the immunoglobulin
Antigen-Presenting Cells M (IgM) subclass, whereas the antibodies of the secondary
response are the immunoglobulin G (IgG) subclass and have
There are several types of leukocytes that function as antigen- a different structure. Figure 3–1 depicts the primary and
presenting cells (APCs), including macrophages, neu- secondary antibody responses.
trophils, and some B cells. In addition to leukocytes, there After the antigen is cleared, memory cells are stored in
are specialized immune cells capable of antigen presentation. immune organs of the host. When the same antigen is en-
These include the different types of dendritic cells present countered again, the memory cells are activated and produce
in the skin (Langerhans cells), nervous tissue (glial cells), a stronger and more rapid response. IgG antibodies are
lymph nodes, spleen, intestines, liver (Kupffer cells), bone formed during the secondary response and are made in great
(osteoclasts), and thymus. These APCs first phagocytize the quantities, and although IgM is made during the primary
foreign antigen, process it internally, and then with the help response, there is a period when it overlaps with the produc-
of MHC molecules, present short peptide sequences of the tion of IgG antibodies at the beginning of the secondary
antigen on their cell membranes. TH cells can then recognize response. IgG secondary antibodies have a higher avidity for
the antigen in the context of MHC presentation and respond antigen and can be produced by much lower concentrations
to it by the appropriate immune reaction. of antigen. Secondary antibodies can usually be measured
within 1 to 2 days. For example, a primary antibody may re-
Immune System Organs quire more than 100-fold excess of antigen to initiate the first
response. Many of the antigens that stimulate the primary
The organs of the IS are divided into primary and secondary
response have multiple repeating epitopes such as polysac-
systems. The primary lymphoid organs are the thymus and
charides and therefore are good immune stimulators at lower
bone marrow, where immune cells differentiate and mature.
concentrations.
The secondary lymphoid organs include the lymph nodes
and spleen, in which immune cells interact with each other
Cell Lineages and Markers
and process antigens (Table 3–4).

Immune Maturation Advanced Concepts


Because of the complexity and diversity of immune responses, Nearly all cells of the host’s body, especially the various
a specific immediate immune response is not always pos- leukocytes, have specific cell receptors, CD markers, that
sible. It usually takes days to months for all the aspects of interact with other cells and receive signals from cellular
an efficient immune response to happen from the moment messenger systems. Macrophages, NK cells, T and B cells,
and APCs can interact directly with cell-to-cell communi-
cation or indirectly with soluble mediators through a com-
Table 3–4 Lymphoid Organs Associated plex system. These markers on cells can change during the
lifetime of the cell or in response to infection or activation.
With the Acquired Immune
Currently, there are more than 200 different CD markers
System known. CD markers are critical to identifying hematopoietic
Primary Lymphoid Secondary Lymphoid cell maturation stages and lineages.
Organs Organs All immune cells originate from pluripotent hematopoi-
etic progenitors (or CD34-positive cells) through one of two
• Thymus • Lymph nodes
pathways of lineage, the myeloid or the lymphoid. Growth
• Bone marrow • Spleen factors are responsible for differentiating and maturing stem
cells into the many different cells of the IS. Growth factors
• Mucosa-associated tissues
also allow progenitor stem cells to reproduce and differen-
Site of maturation for T and Site of cell function for mature T tiate. The cells of the myeloid lineage consist of phagocytic
B cells and B cells cells such as the monocytes and macrophages, often referred
Lymphocytes differentiate from Cells interact with each other and to as the mononuclear phagocytic system (MPS); the granu-
stem cells, then migrate to with accessory cells and antigens locytes or polymorphonuclear cells (PMNs), the neu-
secondary lymphoid organs trophils, eosinophils, and the basophils; and the APCs such
6888_Ch03_045-076 29/10/18 4:44 PM Page 53

Chapter 3 Fundamentals of Immunology 53

Antibody Concentration in Serum


PRIMARY SECONDARY
Total Ab
Steady state
IgG
Total Ab
Logarithmic Decline
increase IgG

IgM
IgM
Negative
Inductive period phase

Primary IgM Secondary


immunogenic immunogenic
stimulation stimulation

IgM
IgG

IgG
Figure 3–1. Schematic representation of "Memory" cells
primary and secondary antibody responses. Note
the enhanced antibody production and expanded
antibody-producing cell population during the
secondary antibody response. (From Herscowitz,
HB: Immunophysiology. In Bellanti, JA [ed]:
Immunology III. WB Saunders, Philadelphia, "Memory" cells
1985, p 117, with permission.)

as the dendritic cells in the skin and liver. Erythrocytes and inflammatory responses and have enzymes that allow them
platelets originate from this system. The lymphoid lineage to destroy engulfed pathogens. All three types of granulo-
consists of the various subpopulations of lymphoid cells, cytes possess receptors for the FC portion of IgG (CD16) and
the T cells, B cells, and NK cells. complement receptors C5a, CR1(CD35), and CR3(CD11b).
The myeloid-monocyte precursors originate in the bone Additionally, eosinophils possess low-affinity FC receptors
marrow and then differentiate into circulating blood mono- for IgE and therefore play a critical role in allergic reactions
cytes. When monocytes encounter antigen, they can differ- and inflammation in parasitic infections. Basophils and mast
entiate into tissue macrophages. Phagocytic cells process cells (a type of tissue basophil) possess high-affinity FC
antigens for acquired immunity and can directly kill many immunoglobulin E (IgE) receptors, are powerful effectors of
pathogens such as bacteria and fungi as part of innate im- inflammation and allergic reactions, and can cause the
munity. MPS cells present antigen to lymphocytes and inter- release of localized histamine.
act with other immune cells via cell membrane receptors. Lymphocytic cells are generated in the thymus or bone
The FC part of the antibody receptor and the complement marrow and travel through the circulatory system to the
receptor CR1 are used by phagocytes during opsonization.1 lymph nodes and spleen, where they mature and differen-
When these cells are functioning as APCs, they express the tiate. In a mature adult, the lymphocytes account for about
MHC class II molecules on their membranes and lack the 20% to 30% of the circulating leukocytes. In primary
FC receptor. Granulocytes originate in the bone marrow. organs, these cells acquire receptors that enable them to
They are the predominant leukocyte in the circulation of the interact with antigens and to differentiate between self
mature adult (60% to 70%). and nonself antigens; a very critical part of lymphocyte
Granulocytes all have granules in their cytoplasm and maturation and the adaptive IS in general.
are of three types, distinguished by the hematologic stain- In the secondary organs, immune cells are provided with
ing of their granules. The neutrophils stain a faint purple a highly interactive environment in which immune
or neutral color (neutral granules), the eosinophils a red- responses are exchanged and made specific. Remember,
dish orange color (acidic granules), and the basophils a there are two primary classifications of lymphocytes, T and
bluish black color (basic granules). The main role of these B cells, and these similar-looking cells can be distinguished
cells is phagocytosis; they function primarily in acute by the presence of specific cell markers by means of using
6888_Ch03_045-076 29/10/18 4:44 PM Page 54

54 PART I Fundamental Concepts

sophisticated immunologic methods of testing, such as produced by monocytes and macrophages. Cytokines
flow cytometry (discussed later in this chapter). Specific to function in a complex manner by regulating growth, mobil-
the T cell is the T-cell receptor (TCR), which is in proxim- ity, and differentiation of leukocytes. One cytokine may act
ity to and usually identified with the CD3 complex on the by itself or together with other cytokines. Other cytokines
T-cell membrane. It associates in cell-to-cell contacts and oppose the actions of one or more cytokines and function
interacts with both antigenic determinants and MHC pro- to quantitatively increase or decrease a particular immune
teins. In addition to CD3 on T cells, there is the separate reaction. Some cytokines are synergistic and need each
CD2 marker that is involved in cell adhesion. It has the other to have their full effect. The effects of cytokines
unique ability to bind with sheep erythrocytes in vitro.2 Re- can be in the immediate area of their release, or they
member that TH cells have the CD4 marker and recognize can travel through the plasma to affect distant cells and
antigen together with the MHC class II molecules, and TC tissues. There is often significant overlap in how cytokines
cells possess CD8 markers and interact with MHC class I function.
molecules.3 The ratios of T cells expressing CD4 versus Major classes of cytokines include the following: inter-
CD8-expressing cells can be a marker for particular dis- leukin (IL), interferon (IFN), tumor necrosis factor (TNF),
eases. One of the defining features of AIDS is a reversal of and colony-stimulating factor (CSF). Each class has several
the typical CD4 to CD8 ratio, which helped to explain members (see Table 3–3). Cytokines act by binding to spe-
some of the pathology of this illness. cific target cell receptors. When cytokines bind to their re-
There are also compounds referred to as superantigens, ceptors on cells, the number of receptors is often increased
which can stimulate multiple T cells, causing them to re- as the cell is stimulated. Internal cellular signaling pathways
lease large amounts of cytokines. Certain bacteria toxins become activated. The cell is no longer in a resting state and
are superantigens and can lead to lethal reactions in the can have a new function. After cytokine binding, both the
host if the immune system is overstimulated. receptor and the cytokine become internalized, which in-
B cells are defined by the presence of immunoglobulin duces the target cell to grow and differentiate. Differentiated
on their surface; however, they also possess MHC class II cells have specialized functions such as secreting antibodies
antigens (antigen presentation); the complement receptors or producing enzymes. Immune cells and other host cells
CD35 and CD21; FC receptors for IgG; and CD19, CD20, respond to cytokines and can react with chemoattraction, as
and CD22 markers, which are the CD markers used to well as antiviral, antiproliferation, and immunomodulation
identify B cells. Membrane-bound immunoglobulin may processes. Cytokines fine-tune the IS and also function as
act as an antigen receptor for binding simple structural critical cell activators.
antigens or antigens with multiple repeating determinants In addition to the cytokines, other cellular products of
(referred to as T cell–independent antigens, meaning they the IS, including chemokines, immunoglobulins, comple-
do not require the intervention of T-cell help). When T ment proteins, kinins, clotting factors, acute phase pro-
cell–dependent antigens (structurally complex and unique teins, stress-associated proteins, and the fibrinolytic
substances) are encountered, B cells require the interven- system, can have powerful effects. Cytokines typically com-
tion of T cells to assist in the production of antibody. When municate between cells through the plasma. Chemokines
B cells become activated, they mature and develop into are attractant molecules that interact between cells,
plasma cells, which produce and secrete large quantities of immunoglobulins, and complement proteins and are im-
soluble Ig into tissue or plasma. portant in destroying pathogens. Acute phase proteins and
The third major class of lymphocytes are the NK cells, fibrinolytic proteins play a role in inflammation, a process
also referred to as large granular lymphocytes. Unlike B cells, that recruits appropriate cells to an immune site and
NK cells do not have surface Ig or secrete Ig, nor do they modifies the vascular system. In addition to inflammation
have antigen receptors like the TCR of T cells. NK cells and cell-to-cell communication, some cytokines are
have the CD56 and CD16 markers and do not require the immunosuppressive. One of the important chemokine
presence of an MHC marker to respond to an antigen. They receptors in blood banking is the Duffy antigen group pres-
are thymus-independent and are able to lyse virally infected ent on RBCs. The lack of these antigens can prevent certain
cells and tumor cells directly in a process known as types of malaria parasites from infecting the host. The
antibody-dependent cell-mediated cytotoxicity (ADCC) Duffy antigen may also modulate immune response by
by anchoring immunoglobulin to the cell surface mem- acting as a sink for extra cytokines of the IL-8 family.
brane through an FC receptor.
Immune System Genetics

Cytokines and Immunoregulatory Individuals have a unique immune response based on their
Molecules genetic inheritance, and there are familial patterns of sus-
ceptibility and resistance. In blood banking this is impor-
Cytokines are soluble protein or peptide molecules that tant because not all transfusion recipients make antibodies
function as powerful mediators of the immune response. to alloantigens on transfused RBCs. Responders are people
There are two main cytokine types: lymphokines, which who have a tendency based on their inheritance to make
are produced by lymphocytes, and monokines, which are antibodies. The terms high responders and low responders
6888_Ch03_045-076 29/10/18 4:44 PM Page 55

Chapter 3 Fundamentals of Immunology 55

describe individual responses to antigen challenges. When Characteristics of Immunoglobulins


antigenic stimulation occurs, one B cell forms an antibody
with a single specificity. During the lifetime of the cell, the Immunoglobulin (Ig), also called antibody, is a complex self-
cell can switch to make a different isotypic class of anti- protein produced by plasma cells, with specificity to antigens
body that has the same antigen specificity. Isotype switch- (or immunogens), that stimulate their production. Igs act in
ing requires further DNA rearrangement in mature B cells. response to specific foreign, nonself proteins or other com-
Class switching is dependent on antigenic stimulation and plex molecules not tolerated by the host. Immunoglobulins
on the presence of cytokines released by T cells; this reflects make up approximately 20% of the proteins in disseminated
the further adaptation of the immune response. Isotype body fluids. Immunoglobulins bind antigen, fix comple-
switching is seen in blood banking when antibodies react ment, facilitate phagocytosis, and neutralize toxic substances
at different temperatures and phases. in the circulation. Thus, antibodies have multiple functions,
some more highly specialized and specific than others.
Immunoglobulins are classified according to the molecular
Advanced Concepts structure of their heavy chains. The five classifications are
IgA (α [alpha] heavy chain), IgD (δ [delta] heavy chain),
The major histocompatibility complex (MHC) is the region
IgE (ε [epsilon] heavy chain), IgG (γ [gamma] heavy chain),
of the genome that encodes the human leukocyte antigen
and IgM (µ [mu] heavy chain).
(HLA) proteins. (Refer to Chapter 23, “The HLA System.”)
Table 3–5 illustrates some of the various differences in the
The MHC is critical in immune recognition and regulation
classes of immunoglobulins, such as molecular weight, per-
of antigen presentation in cell-to-cell interactions, trans-
centage in serum, valency (number of antigen-binding sites),
plantation, paternity testing, and specific HLA patterns. It
carbohydrate content, half-life in the blood, and whether
also correlates with susceptibility to certain diseases. HLA
they exist as monomers or multimers. IgG is the most con-
molecules are categorized into two classes: MHC classes I
centrated in serum, comprising approximately 80% of the
and II. Class I molecules are found on all nucleated cells
total serum Ig; next is IgA, at about 13% (although it is the
except trophoblasts and sperm, and they play a key role in
major Ig found in body secretions); IgM is 6%; IgD is 1%;
cytotoxic T-cell function. Class II molecules are found on
and IgE is the least common, present at less than 1%.4
antigen-presenting cells such as B lymphocytes, activated
T cells, and the various dendritic cells. Class II molecules
on APCs are essential for presenting processed antigen Immunoglobulin Structure
to CD4 T cells and are necessary for T-cell functions and
B-cell help. In addition, there are class III molecules that All classes and subclasses of immunoglobulins have a com-
encode complement components such as C2, C4, and fac- mon biochemical structural configuration with similarities
tor B. The genes for MHC classes I through III molecules and are probably derived from a common evolutionary mo-
are located on the short arm of chromosome 6 and are lecular structure with well-defined function. In Figure 3–2,
highly polymorphic in nature with multiple alleles. the basic Ig structural unit is composed of four polypeptide
chains: two identical light chains (molecular weights of

Table 3–5 Characteristics of Serum Immunoglobulins


Characteristic IgA IgD IgE IgG IgM
Heavy chain type Alpha Delta Epsilon Gamma Mu

Sedimentation coefficient(s) 7–15* 7 8 6.7 19

Molecular weight (kD) 160–500 180 196 150 900

Biologic half-life (d) 5.8 2.8 2.3 21 5.1

Carbohydrate content (%) 7.5–9 10–13 11–12 2.2–3.5 7–14

Placental transfer No No No Yes No

Complement fixation (classical pathway) No No No + +++

Agglutination in saline + 0 0 ± ++++

Heavy chain allotypes Am None None Gm None

Proportion of total immunoglobulin (%) 13 1 0.002 80 6

*May occur in monomeric or polymeric structural forms.


d = days; kD = kilodaltons; 0 = negative reactivity; ± = weak reactivity; + = slight reactivity; +++ = strong reactivity; ++++ = very strong reactivity
6888_Ch03_045-076 29/10/18 4:44 PM Page 56

56 PART I Fundamental Concepts

A
Bi ntig
nd e
Si in n
te g

2
H2

NH
N

2
H2

NH
N

Heavy Chain
Hinge Region Variable region
Light Chain
Variable region
S S
Heavy Chain
S Constant region
S S
S S S Light Chain
Constant region

Fa
Fa

b
COOH
COOH

SS
S
SS

Figure 3–2. Schematic representation of


basic immunoglobulin structure. The inset shows
Fc formation of Fab and FC fragments after enzy-
matic cleavage of the IgG molecule by papain.

approximately 22,500 daltons [D]) and two identical heavy antibody specificity. Structurally and functionally, the Fab
chains (molecular weights from approximately 50,000 to fragments encompass the portions of the Ig from the hinge
75,000 D). In Figure 3–3, covalent disulfide bonding holds region to the amino terminal end and are the regions respon-
the light and heavy chains together, and the covalent disul- sible for binding antigen (see Fig. 3–2).
fide linkages in Ig molecules provide greater structural The domains of immunoglobulins are the regions of the
strength than do hydrogen bonding and van der Waals light and heavy chains that are folded into compact globular
forces. However, they limit the flexibility of the Ig molecule. loop structures (see Fig. 3–3). The domains are held together
The heavy chains are also interconnected by disulfide link- by intrachain covalent disulfide bonds; the V region of the
ages in the hinge region of the molecule. Although there are domain specifies the variable region, and the C is the con-
five types of heavy chains, there are only two types of light stant region. The domains are also specified according to
chains: κ (kappa) and λ (lambda). Both types are found in light and heavy chains. Looking at one half of an Ig mole-
all classes of immunoglobulins, regardless of heavy chain cule, one domain (VL) is the variable region, one domain
classification. (CL) is in the constant region of each light chain, and one
Ig molecules are proteins and therefore have two terminal variable domain (VH) is on each heavy chain. The number
regions: the amino (-NH2) terminal and the carboxyl of domains is determined by the isotype. There are three
(-COOH) terminal. The carboxyl region of all heavy chains constant domains, CH1 to CH3, on the heavy chains of IgA,
has a relatively constant amino acid sequence and thus is IgD, and IgG, and four constant domains, CH1 to CH4, on
named the constant region. Like the heavy chain, the light the heavy chains of IgE and IgM. The antigen-binding, or
chain also has a constant region. Due to certain common Ig idiotypic, regions (which distinguish one V domain from all
structures, enzyme treatment yields specific cleavage prod- other V domains) are located within the three-dimensional
ucts of defined molecular weight and structure. The enzyme structures formed by the VL and VH domains together. Cer-
papain splits the antibody molecule at the hinge region to tain heavy chain domains are associated with particular bio-
give three fragments: one crystallizable FC fragment and two logical properties of immunoglobulins, especially those of
antigen-binding fragments, Fab. The FC fragment encom- IgG and IgM, and include complement fixation. They are
passes that portion of the Ig molecule from the carboxyl identified with the CH2 domain and the CH3 domains, which
region to the hinge region and is responsible for complement serve as attachment sites for the FC receptor of monocytes
fixation and monocyte binding by FC receptors on cells. The and macrophages.
FC fragment on IgG antibody only is also responsible for pla-
cental transfer. In contrast to the carboxyl terminal regions, Immunoglobulins Significant for Blood Banking
the amino terminal regions of both light and heavy chains
of immunoglobulins are known as the variable regions be- All immunoglobulins can be significant for transfusion medi-
cause they are structured according to the great variation in cine; however, IgG, IgM, and IgA have the most significance
6888_Ch03_045-076 29/10/18 4:44 PM Page 57

Chapter 3 Fundamentals of Immunology 57

Heavy Chain

Light Chain
Hinge Region

Complement (C1q)
Attachment Site

Attachment Site for


Macrophage Fc Receptor

Figure 3–3. Schematic illustration of the


domain structure within the IgG molecule.

for the blood bank. Most clinically significant antibodies blood products. IgG antibodies are important in
that react at body temperature (37°C) are IgG isotype and hemolytic disease of the newborn (HDN), because antibod-
are capable of destroying transfused antigen-positive RBCs, ies can be formed in response against alloantigens on fetal
causing anemia and transfusion reactions of various severi- RBCs that enter the mother’s circulation, usually during
ties. IgM antibodies are most commonly encountered as delivery. While HDN can be fatal, antibody screening, Rh
naturally occurring antibodies in the ABO system and are typing, and passive anti-D antibody have prevented HDN
believed to be produced in response to commonly occurring from developing in D-negative mothers who give birth to
antigens such as intestinal flora and pollen grains. (Refer to D-positive babies.
Chapter 6, “The ABO Blood Group System.”) Other blood IgG has the greatest number of subclasses: IgG1, IgG2,
groups such as Lewis, Ii, P, and MNS may also produce IgM IgG3, and IgG4, and all four are easily separated by
antibodies, which usually react best at ambient temperature
(22°C to 24°C). The primary testing problem encountered
with IgM antibodies is that they can interfere with the de-
tection of clinically significant IgG antibodies by masking J Chain
their reactivity. Unlike IgG, IgM exists in both monomeric
and polymeric forms (as pentamers) containing a J (joining)
chain (Fig. 3–4).

Advanced Concepts
The pentameric form of IgM can be dissociated through
cleavage of covalent bonds interconnecting the monomeric
subunits and the J chain by chemical treatment with
sulfhydryl reducing reagents such as β-2-mercaptoethanol
(2-ME) or dithiothreitol (DTT). These reagents can distin-
guish a mixture of IgM and IgG antibodies, because only
IgM is removed by the use of these compounds; therefore, IgM
the removal allows unexpected IgG antibodies to be Monomer
detected. IgG antibodies are significant in transfusion medi-
cine because they are the class of immunoglobulins that are Figure 3–4. Schematic representation of the pentameric configuration of the
made in response to transfusion with nonself antigens on IgM immunoglobulin.
6888_Ch03_045-076 29/10/18 4:44 PM Page 58

58 PART I Fundamental Concepts

electrophoresis. The small differences in the chemical struc- acquires a glycoprotein secretory component as it passes
ture within the constant regions of the gamma heavy chains through epithelial cell walls of mucosal tissues and appears
designate the various subclasses, and the number of disul- in nearly all body fluids. IgA is important in immunohe-
fide bonds between the two heavy chains in the hinge re- matology because about 30% of anti-A and anti-B anti-
gion of the molecule constitutes one of the main differences bodies are of the IgA class (the remaining percentages are
between subclasses. Functional differences between the IgM and IgG).6 Also, anti-IgA antibodies can cause severe
subclasses include the ability to fix complement and cross anaphylaxis if IgA are transfused in plasma products to
the placenta (Table 3–6). IgG blood group antibodies of a patients who are deficient in IgA. Additionally, IgA can
single specificity are not necessarily one specific subclass; increase the effect of IgG-induced RBC hemolysis.7
all four subclasses may be present, or one may predomi- IgE is normally found only in monomeric form in trace
nate. For example, the antibodies to the Rh system antigens concentrations in serum, about 0.004% of total im-
are mostly of the IgG1 and IgG3 subclasses, whereas anti-K munoglobulins, and is important in allergic reactions. The
(Kell), and anti-Fy (Duffy) antibodies are usually of the FC portion of the IgE molecule attaches to basophils and
IgG1 subclass. Anti-Jk (Kidd) antibodies are mainly IgG3 mast cells and facilitates histamine release when an allergen
and may account for the unusual nature of these different binds to the Fab portion of the molecule and cross-links
antibodies with regard to testing and clinical significance. with a second molecule on the cell surface. Although
The purpose for the existence of biological differences in hemolytic transfusion reactions are not caused by IgE,
subclass expression is still not completely understood. urticaria may occur because of the presence of IgE antibod-
Especially important in blood banking, severe HDN has ies. Histamine is thought to be a primary mediator of aller-
been most often associated with IgG1 antibodies.5 gic reactions and hypersensitivity. Because IgE causes
Like IgM, IgA exists in two main forms—a monomer transfusion reactions by release of histamines, patients who
and polymer form—as dimers or trimers composed of two have several allergic reactions to blood products can be pre-
or three identical monomers, respectively, joined by a treated with antihistamines to counteract the response
J chain. IgA is located in different parts of the IS, depending when receiving blood products.2 IgD is scarce in serum,
on subclass. Serum IgA is found in both monomeric and present at less than 1% of total serum immunoglobulins,
polymeric forms; however, secretory IgA is usually found and has a short half-life of only 1 to 3 days. IgD is usually
in the mucosal tissues of the body. Its polymer form bound to the membrane of immature B cells and appears
to have functions that deal primarily with maturation of
B cells into plasma cells; however, its function is not fully
understood.8
Table 3–6 Biological Properties of IgG
Subclasses
Immunoglobulin Variations
Characteristic IgG1 IgG2 IgG3 IgG4
There are three main types of antibody-inherited variation:
Proportion of total serum 65–70 23–28 4–7 3–4 isotype, allotype, and idiotype. Isotype variation (or class
IgG (%)
variation) refers to variants present in all members of a
Complement fixation ++ + +++ 0 species, including the different heavy and light chains and
(classic pathway) the different subclasses. All humans have the same Ig classes
Binding to macrophage +++ ++ +++ ± and subclasses. Allotypic variation is present primarily in
Fc receptors the constant region; not all variants occur in all members of
a species. Idiotypic variation, which determines the antigen-
Ability to cross placenta + ± + + binding specificity (or complementary determining regions,
Dominant antibody [CDRs]) of antibodies and T-cell receptors, is found only in
activities: the variable (and hypervariable) regions and is specific for
each antibody molecule.
• Anti-Rh ++ 0 + ±
The allotypes of a growing fetus may cause the maternal
• Anti-factor VII 0 0 0 + IS to become immunized to paternal allotypic determinants
on fetal immunoglobulins.4 Just as alloimmunization can
• Anti-dextran 0 + 0 0
occur during pregnancy, it can occur from transfusions,
• Anti-Kell + 0 0 0 especially in patients who have received multiple transfusions
of blood, plasma, or gamma globulin. Idiotypes can be seen
• Anti-Duffy + 0 0 0
as nonself because they are often present at concentrations
• Anti-platelet 0 0 + 0 too low to induce self-tolerance. Idiotypes are determined
by the antigens that react with them and exist in a type of
Biological half-life (days) 21 21 7–8 21
equilibrium with anti-idiotypic antibodies. The presence of
0 = negative reactivity; ± = weak (or unusual) reactivity; + = slight (or usual) reactivity; antigen disrupts this equilibrium and may be important in
++ = moderate (or more common) reactivity; +++ = strong reactivity controlling an immune response.
6888_Ch03_045-076 29/10/18 4:44 PM Page 59

Chapter 3 Fundamentals of Immunology 59

Immunoglobulin FC Receptors medicine testing procedures is useful in the investigation


of hemolytic transfusion reactions and autoimmune
Macrophages and monocytes have receptors for the attach-
hemolytic anemias.
ment of IgG and can bind the CH3 domain of the FC portion.
Only the IgG1 and IgG3 subclasses are capable of attachment
to phagocytic receptors. This is one way that incompatible
Classical Complement Pathway
RBCs coated with IgG antibody are removed by phagocyto-
sis. The other phagocytic cells with FC receptors include The activation of the classical complement pathway is
neutrophils, NK cells, and mature B cells.4,9 initiated when antibody binds to antigen. This allows the
binding of the complement protein C1 to the FC fragment
Complement System of an IgM, IgG1, or IgG3 subclass antibody. Complement
activation by IgG antibody depends on concentration of
The complement system, or complement, is a complex group
cell surface antigen and antigen clustering, in addition to
of over 20 circulating and cell membrane proteins that have
antibody avidity and concentration. IgM is large and has
a multitude of functions within the immune response.
FC monomers close to each other on one immunoglobulin
Primary roles include direct lysis of cells, bacteria, and
molecule; therefore, only one IgM molecule is necessary to
enveloped viruses as well as assisting with opsonization to
activate complement. The C1 component is actually a com-
facilitate phagocytosis. Another role is the production of
plex composed of three C1 subunits, C1q, C1r, and C1s,
peptide fragment split products, which play roles in inflam-
which are stabilized by calcium ions. Without calcium pres-
matory responses such as increased vascular permeability,
ent, there is no stabilization of the C1q, r, s complex, and
smooth muscle contraction, chemotaxis, migration, and ad-
complement is not activated.
herence. Complement components circulate in inactive form
In the C1 complex bound to antigen antibody, C1q is
as proenzymes, with the exception of factor D of the alter-
responsible for catalyzing the C1r to generate activated C1s.
nate pathway. The complement proteins are activated in a
Activated C1s is a serine-type protease. The C1q, r, s complex
cascade of events through three main pathways: the classical,
(actually as a C1q[r, s]2 unit) acts on C2 and C4 to form
alternative, and lectin pathways. The three pathways con-
C4b2a. C4b2a uses component C3 as a natural substrate.
verge at the activation of the component C3. The classical
By-products that result from the activation of the classical
pathway is activated by the binding of an antigen with an
sequence include C3a; C4b, which binds to the cell surface;
IgM, IgG1, or IgG3 antibody. The alternative pathway is ac-
C4a, which stays in the medium and has modest anaphyla-
tivated by high molecular weight molecules with repeating
toxin activity; and C3b, which attaches to the microbial sur-
units found on the surfaces of target cells. The lectin path-
face. Note that complement fragments that have the b type
way is activated by attachment of plasma mannose-binding
are usually bound to membranes, whereas the a fragments
lectin (MBL) to microbes. MBL in turn activates proteins of
often have anaphylatoxic activity.
the classical pathway.
Human RBCs have CR1 receptors for C4d and C3b, and
some of the cleavage products will attach to the RBC mem-
brane. Eventually these fragments are further degraded to
Advanced Concepts C4d and C3d through the action of C4BP, factor H (which
Complement components are sequentially numbered C1 binds C3b), and factor I (which degrades both C4b and
through C9, but this refers to their discovery date, not to C3b). Some of C3b binds to the C4b2a complex, and the re-
their activation sequence. The four unique serum proteins sulting C4b2a3b complex functions as a C5 convertase. The
of the alternative pathway are designated by letters: factor B, C5 convertase then acts on C5 to produce C5a (a strong
factor D, factor P (properdin), and IF (initiating factor). stimulator of anaphylatoxins) and C5b, which binds to the
Some activation pathways require Ca2+ and Mg2+ cations as cell membrane and recruits C6, C7, C8, and C9 to the cell
cofactors for certain components. In certain nomenclature, membrane. When C5b and C6, C7, C8, and C9 are bound,
complement components that are active are designated by the membrane attack complex forms; this causes lysis of
a short bar placed over the appropriate number or letter. foreign cells. Figure 3–5 shows the sequential activation of
The cleavage products of complement proteins are distin- the complement system via the classical and alternative path-
guished from parent molecules by suffixes such as C3a and ways. Figure 3–6 illustrates the mechanism of antibody-
C3b. The complement system is able to modulate its own dependent cell-mediated cytotoxicity (ADCC).
reactions by inhibitory proteins such as C1 inhibitor
(C1INH), factor H, factor I, C4-binding protein (C4BP), Alternative Complement Pathway
anaphylatoxin inactivator, anaphylatoxin inhibitor, mem-
brane attack complex (MAC) inhibitor, and C3 nephritic The alternative pathway is older in evolution and allows
factor (NF). This regulation of complement is important to complement to be activated without acquired immunity. The
ensure that complement proteins do not destroy healthy alternative pathway is activated by surface contacts with
host cells and inflammation is controlled. The evaluation complex molecules and artificial surfaces such as dialysis
of C3b and C3d complement components in transfusion membranes and dextran polymers. There are four important
proteins in this pathway: factor D, factor B, properdin, and
6888_Ch03_045-076 29/10/18 4:44 PM Page 60

60 PART I Fundamental Concepts

CLASSIC PATHWAY ALTERNATIVE PATHWAY

C4b, 2C

MEMBRANE
ATTACK
MECHANISM

C5b67

C5b6789
Figure 3–5. Schematic diagram illustrating
the sequential activation of the complement
CELL LYSIS system via the classical and alternative pathways.

C3. In this pathway, complement component factor D is C3b encounters normal cells, it is rapidly eliminated through
analogous to C1s in the classical pathway; factor B is analo- the combined interactions of factors H and I. The accumu-
gous to C2; and the cleavage product C3b is analogous lation of C3b on microbial cell surfaces is associated with
to C4. Also, factor C3bBbP is analogous to C4b2a, and attachment of C3b to factor B. The complex of C3b and fac-
C3b2BbP is analogous to C4b2a3b in the classical pathway. tor B is then acted on by factor D. As a result of this action,
Activation of the alternative pathway requires that a C3b factor B is cleaved, yielding a cleavage product known as Bb.
molecule be bound to the surface of a target cell. Small The C3bBb complex is stabilized by the presence of prop-
amounts of C3b are generated continuously, owing to the erdin (P), yielding C3bBbP. This complex cleaves C3 into
spontaneous hydrolytic cleavage of the C3 molecule. When additional C3a and C3b. C3b, therefore, acts as a positive
feedback mechanism for driving the alternative pathway. The
C3b2BbP complex acts as a C5 convertase and initiates the
IgG Antibody
later steps of complement activation (see Fig. 3–5).

Lectin Complement Pathway


Lysed Target Cell
The third pathway of activation, the lectin pathway, is acti-
vated by the attachment of MBL to microbes. The subsequent
Red Cell Antigenic Determinant Site reactions are the same as those of the classical pathway.
Remember that all three methods of activation lead to a final
common pathway for complement activation and membrane
Effector Cell
(Monocyte) attack complex (MAC) formation.
C3b Receptor
Membrane Attack Complex

The final step of complement activation is the formation


Fc Receptor of the MAC, which is composed of the terminal components
of the complement sequence. There are two ways in which
the MAC is initiated. In either classical or alternative path-
Figure 3–6. Schematic representation of the mechanism of antibody-dependent
cell-mediated cytotoxicity (ADCC). Note the role of effector cell surface receptors ways, the formation of C5 convertase is necessary. In the
for the FC fragment of IgG. activation of the classical pathway, the MAC is initiated by
6888_Ch03_045-076 29/10/18 4:44 PM Page 61

Chapter 3 Fundamentals of Immunology 61

the enzymatic activity of component C4b2a3b on C5. In the antigen is more commonly used in blood banking because
alternative pathway, it is C3b2Bb that has the ability to cleave the primary testing is for the detection of antibodies to blood
C5. The next step in either pathway is the same. Component group antigens. The immune reaction to any immunogen,
C5 is split into the fragments C5a (a potent anaphylatoxin) including antigens, is determined by the host response and
and C5b; the C5b fragment attaches to cell membranes and by several biochemical and physical characteristics of the im-
can continue the complement cascade by initiating the mem- munogen. Properties such as size, complexity, conformation,
brane attachment of C6, C7, and C8. After the attachment charge, accessibility, solubility, digestibility, and biochemical
of C9 to the C5b678 complex, a small transmembrane chan- composition influence the amount and type of immune
nel or pore is formed in the lipid bilayer of the cell mem- response (Box 3–2).
brane, and this allows for osmotic lysis and subsequent death Molecules that are too small cannot stimulate antibody pro-
of the cell. duction. Immunogens having a molecular weight (MW) less
than 10,000 D, for example, are called haptens and usually do
Binding of Complement by RBC Antibodies not elicit an immune response on their own; however, coupled
with a carrier protein having a MW greater than 10,000 D, they
Disruption in the activation of either complement pathway can produce a reaction. Antibodies and cellular responses are
can result in damage to the host’s cells. The formation of an- very specific for an antigen’s physical conformation as opposed
tibodies with complement capacity that can bring about the to its linear sequence. Overall charge is important, as antibody
destruction of RBCs are of particular concern to transfusion response is also formed to the net charge of a molecule,
medicine specialists. In order to initiate activation, C1 mole- whether it is positive, negative, or neutral. As an antigen must
cules bind with two adjacent Ig FC regions. A pentameric be seen by the IS, the accessibility of epitopes influences the
IgM molecule provides two FC regions side by side, thereby immune response. Also, antigenic substances that are less
binding complement. However, a monomeric IgG molecule soluble are less likely to elicit an immune response. The bio-
binds C1q less efficiently because two IgG molecules are chemical composition of the stimulus plays a role in immune
needed in close proximity to bind complement. Therefore, stimulation. Remember that RBC antigens are very diverse in
many molecules of IgG are required to be bound to a single structure and composition and may be proteins (such as the
target in order for complement activation to occur. For Rh, M, and N blood group substances) or glycolipids (such as
example, as many as 800 IgG anti-A molecules may need to the ABH, Lewis, Ii, and P blood group substances). Further-
attach to one adult group A RBC in order to bind a single C1 more, human leukocyte antigens are glycoproteins. Because of
molecule, so there is little complement activation by IgG these differences in structure, conformation, and molecular
anti-A immunoglobulins.4 nature, not all blood group substances are equally immuno-
Antibodies against the Rh antigens usually do not bind genic in vivo (Table 3–7).
complement due to the low level of Rh antigens on RBC sur-
faces. Antibodies to the Lewis blood group system are gener- Characteristics of Blood Group
ally IgM, and as such can activate complement but rarely cause Antibodies
hemolytic transfusion reactions due to their low optimal
reactivity temperature. Therefore, in blood banking, with the There are many different and important characteristics of
exception of the ABO system, only a few antibodies activate blood group antibodies, such as whether they are polyclonal
the complement sequence to induce complement-mediated or monoclonal, naturally occurring or immune, and allo-
intravascular hemolysis. However, extravascular hemolysis antibodies or autoantibodies.
usually occurs as a result of antibody coating of RBCs, and the
split products of complement activation can stimulate the Polyclonal and Monoclonal Antibodies
reticuloendothelial system and cause anaphylatoxic effects.
Antibody-coated RBCs, either self or nonself, are removed In laboratory testing there are two types of antibody (reagent
by cells of the mononuclear phagocyte system and by the antibodies are called antisera) that are available for use; they
cells lining the hepatic and splenic sinusoids. These phago-
cytic cells are able to clear antibody-coated RBCs because
they have cell surface receptors for complement CR1 (C3b) BOX 3–2
and Ig FC receptors (see Fig. 3–6). IgM-coated RBCs are not
Characteristics of Antigens: Properties That Influence
eliminated through FC receptor–mediated phagocytosis, but
if erythrocytes are coated with IgG and complement, they
Immune Response
will be cleared rapidly from circulation by monocytes and • Size
macrophages. If RBCs are coated with only C3b, they may • Complexity
not be cleared but only sequestered temporarily. • Conformation
• Charge
Characteristics of Antigens • Accessibility
• Solubility
The immune response is initiated by the presentation of an • Digestibility
antigen (initiates formation of and reacts with an antibody) • Chemical composition
or immunogen (initiates an immune response). The term
6888_Ch03_045-076 29/10/18 4:44 PM Page 62

62 PART I Fundamental Concepts

Table 3–7 Relative Immunogenicity of from the cell culture contains antibody from a single type of
B cell, clonally expanded, and therefore has the same variable
Different Blood Group Antigens
region and a single epitope specificity. This results in a mono-
Blood Blood clonal antibody suspension. Monoclonal antibodies are
Group Group Immunogenicity preferred in testing because they are highly specific, well
Antigen System (%)* characterized, and uniformly reactive. Most reagents used
D (Rho) Rh 50 today are monoclonal in nature or are a blend of monoclonal
antisera.
K Kell 5

c (hr’) Rh 2.05 Naturally Occurring and Immune Antibodies


E (rh’’) Rh 1.69 There are two types of antibodies that concern blood bank-
ing: one is naturally occurring and the other is immune.
k Kell 1.50
Both are produced in reaction to encountered antigens. RBC
e (hr’’) Rh 0.56 antibodies are considered naturally occurring when they are
found in the serum of individuals who have never been pre-
Fya Duffy 0.23
viously exposed to RBC antigens by transfusion, injection,
C (rh’) Rh 0.11 or pregnancy. These antibodies are probably produced in
response to substances in the environment that resemble
Jka Kidd 0.07
RBC antigens such as pollen grains and bacteria membranes.
S MNSs 0.04 The common occurrence of naturally occurring antibodies
suggests that their antigens are widely found in nature and
Jkb Kidd 0.03
have a repetitive complex pattern. Most naturally occurring
s MNSs 0.03 antibodies are IgM cold agglutinins, which react best at room
temperature or lower, activate complement, and may be he-
Adapted from Kaushansky, K, et al: Williams Hematology, 8th ed. McGraw-Hill molytic when active at 37°C. In blood banking, the common
Professional, New York, 2010.
*Percentage of transfusion recipients lacking the blood group antigen (in the first naturally occurring antibodies react with antigens of the ABH,
column) who are likely to be sensitized to a single transfusion of red blood cells Hh, Ii, Lewis, MN, and P blood group systems. Some naturally
containing that antigen. occurring antibodies found in normal serum are manufactured
without a known environmental stimulus.10 In contrast to nat-
ural antibodies, RBC antibodies are considered immune when
are manufactured differently and have different properties. found in the serum of individuals who have been transfused
An antigen usually consists of numerous epitopes, and it is or who are pregnant. These antigens have a molecular makeup
the epitopes, not the entire antigen, that a B cell is stimulated that is unique to human RBCs. Most immune RBC antibodies
to produce antibody against. Therefore, these different epi- are IgG antibodies that react best at 37°C and require the use
topes on a single antigen induce the proliferation of a variety of antihuman globulin sera (Coombs’ sera) for detection. The
of B-cell clones, resulting in a heterogeneous population of most common immune antibodies encountered in testing
serum antibodies. These antibodies are referred to as poly- include those that react with the Rh, Kell, Duffy, Kidd, and Ss
clonal or serum antibodies and are produced in response to blood group systems.10
a single antigen with more than one epitope.
In vivo, the polyclonal nature of antibodies improves the Unexpected Antibodies
immune response with respect to quality and quantity. Anti-
bodies against more than one epitope are needed to give Naturally occurring anti-A and anti-B antibodies are routinely
immunity against an entire antigen, such as a pathogen. detected in human serum and depend on the blood type of
However, this diversity is not optimal in the laboratory, and the individual. Blood group A has anti-B; blood group B has
in vitro reagents produced by animals or humans can give anti-A; blood group O has both and anti-A, B. Blood group
confusing test results. Consistency and reliability are needed AB has neither antibody present in their serum. These natu-
in laboratory testing, and polyclonal sera can vary in anti- rally occurring antibodies, or isoagglutinins, are significant
body concentration from person to person and animal to and useful in blood typing. They are easily detected by use of
animal. Sera from the same animal can also vary somewhat, A and B reagent RBCs with a direct agglutination technique.
depending on the animal’s overall condition. In normal, healthy individuals, anti-A and anti-B are generally
Individual sera also differ in the serologic properties of the only RBC antibodies expected to be found in a serum
the antibody molecules they contain, the epitopes they rec- sample. People with an IS that does not function normally
ognize, and the presence of additional nonspecific or cross- may not have the expected naturally occurring antibodies.
reacting antibodies. One way to avoid this problem is to use All other antibodies directed against RBC antigens are con-
monoclonal antibodies produced by isolating individual B sidered unexpected and must be detected and identified
cells from a polyclonal population and propagating them in before blood can be safely transfused, no matter their reaction
cell culture with hybridoma technology. The supernatant strength or profile.
6888_Ch03_045-076 29/10/18 4:44 PM Page 63

Chapter 3 Fundamentals of Immunology 63

The reactivity of unexpected antibodies is highly varied and antigen; this antibody amino acid sequence cannot be changed
unpredictable, as they may be either isotype IgM or IgG; rarely, without altering its specificity. The extent of the reciprocal
both may be present in the same sample. These antibodies relationship, also called the fit between the antigen and its
may be able to hemolyze, agglutinate, or sensitize RBCs. Some binding site on the antibody, is often referred to as a lock and
antibodies require special reagents to enhance their reactivity key mechanism. Factors influencing antigen-antibody reac-
and detection, especially if more than one antibody occurs in tions include intermolecular binding forces, antibody proper-
the sample. Due to the enormous polymorphism of the human ties, host factors, and tolerance.
population, a diversity of RBC alleles and antigens exist, re-
quiring a variety of standardized immunologic techniques and Intermolecular Binding Forces
reagents for their detection and identification.
The in vitro analysis of unexpected antibodies involves Intermolecular binding forces such as hydrogen bonding,
the use of antibody screening procedures to optimize anti- electrostatic forces, van der Waals forces, and hydrophobic
gen-antibody reactions. The majority of these procedures bonds are all involved in antigen-antibody binding reactions.
include reacting unknown serum from a donor or patient Stronger covalent bonds are not involved in this reaction,
sample with known reagent cells at various amounts of time, although they are important for the intramolecular confor-
temperature, and media. All routine blood bank testing re- mation of the antibody molecule. Hydrogen bonds result
quires the use of samples for both expected and unexpected from weak dipole forces between hydrogen atoms bonded
antibodies. to oxygen or nitrogen atoms in which there is incomplete
transfer of the electronic energy to the more electronegative
Alloantibodies and Autoantibodies oxygen or nitrogen. When two atoms with dipoles come
close to each other, there is a weak attraction between them.
Antibodies can be either alloreactive or autoreactive. Allo- Although hydrogen bonds are singularly weak, many of
antibodies are produced after exposure to genetically differ- them together can be strong. They are found in complex
ent, or nonself, antigens, such as different RBC antigens after molecules throughout the human body.
transfusion. Transfused components may elicit the formation Electrostatic forces result from weak charges on atoms in
of alloantibodies against antigens (red blood cell, white cell, molecules that have either a positive or negative overall
and platelets) not present in the recipient. A potentially charge (like charges repel and unlike charges attract). This
serious problem for blood bankers is transfused patients who is seen in the formation of salt bridges. Van der Waals forces
have alloantibodies that are no longer detectable in the are a type of weak interaction between atoms in larger mole-
patients’ plasma or serum. If these individuals are transfused cules. Hydrophobic (water-avoiding) bonds result from the
with the immunizing antigen again, they will make a overlap of hydrophobic amino acids in proteins. The hydro-
stronger immune response against those RBC antigens, phobic amino acids bury themselves together to avoid water
which can cause severe and possibly fatal transfusion reac- and salts in solution. The repulsion of these amino acids to
tions. These recipients will then have a positive autocontrol prevent contact with water and aqueous solutions can be
or direct antiglobulin test (DAT), also referred to as the very strong collectively.
direct Coombs’ test. In addition, there are hydrophilic (water-loving) bonds
Autoantibodies on the other hand, are produced in that allow for the overlap of amino acids that are attracted to
response to self-antigens. They can cause reactions in the water. Hydrophobic and hydrophilic bonds repel each other,
recipient if they have a specificity that is common to the and these repulsive forces also play a role in the formation of
transfused blood. Some autoantibodies do not have a the antigen-antibody bond. All of these bonds affect the total
detectable specificity. Autoantibodies can react at different conformation and strength of antigen-antibody reactions and
temperatures, and both cold and warm autoantibodies may IS molecules.
be present. Patients with autoantibodies frequently have
autoimmune diseases and may require considerable num- Antibody Properties
bers of blood products and special techniques to find com-
patible units. Autoantibodies can be removed from RBCs Just as antigen properties influence the amount and type of
by special adsorption and elution techniques and then immune response, antibody properties influence the strength
tested against reagent RBCs. Therefore, in order to trans- and characteristics of the immune response. Antibody affinity
fuse blood safely, the identity of antibodies should be is often defined as the strength of a single antigen-antibody
determined and recorded. bond produced by the summation of attractive and repulsive
forces. In other words, affinity is the strength of the interac-
Characteristics of Antigen-Antibody tion between the antigen and the antibody’s binding site at
Reactions one individual site. Avidity is used to express the binding
strength of a multivalent antigen with antisera produced in
There are many complex properties of antigen and antibody an immunized individual. Avidity, therefore, is a measure of
reactions that influence serologic and other testing methods the functional affinity of an antiserum for the whole antigen
involving antibodies. The antigen-binding site of the antibody and is sometimes referred to as a combination of affinities.
molecule is uniquely structured to recognize a corresponding Avidity can be important in blood banking because high-titer,
6888_Ch03_045-076 29/10/18 4:44 PM Page 64

64 PART I Fundamental Concepts

Specific Reaction Cross-Reaction No Reaction BOX 3–3

Host Factors: Properties of the Host That Influence


Immune Response
• Nutritional status
• Hormones
• Genetics
• Age
• Race
Immunizing Antigen Determinant Shared No Shared Determinant
• Exercise level
Figure 3–7. Types of antigen-antibody reactions: specific reaction, cross-reaction, • Disease
and no reaction. • Injury

low-avidity antibodies exhibit low antigen-binding capacity


but still show reactivity at high serum dilutions.11 is seen in malaria infection. The majority of African Ameri-
The specificity of an antiserum (or antibody) is one of its cans who do not inherit the Duffy blood system antigens Fya
most important characteristics and is related to its relative or Fyb are resistant to malarial invasion with Plasmodium
avidity for antigen. Antibody specificity can be further clas- knowlesi and Plasmodium vivax. The absence of these anti-
sified as a specific reaction, cross-reaction, or no reaction gens may make these individuals ideal donors for those
(Fig. 3–7). A specific reaction implies reaction between who have developed Duffy system antibodies. (Refer to
similar epitopes. A cross-reaction results when certain epi- Chapter 8, “Blood Group Terminology and the Other Blood
topes of one antigen are shared by another antigen and the Groups.”) This illustrates the importance of the historical
same antibody can react with both antigens. No reaction record of the patient or donor.
occurs when there are no shared epitopes.
Another term frequently used when describing antibod- Tolerance
ies is valency. The valency of an antibody is the number of
antigen-binding sites on an antibody molecule or the num-
ber of antibody-binding sites on an antigen. Antigens are Advanced Concepts
usually multivalent, but antibodies are usually bivalent. Tolerance is defined as the lack of an immune response or
IgM and IgA antibodies can have higher valences due to the an active immunosuppressive response. It has been
multimeric nature of some of their structures. Because the observed post-transfusion for many years, but the precise
biochemistry controlling the forces of antibody binding is mechanism is still unclear. Tolerance can be either natu-
well understood, the influence of these forces can be ma- rally occurring or experimentally induced. Exposure to
nipulated to enhance the reactivity of certain RBC antigens an antigen during fetal life usually produces tolerance to
with antibodies. When more than one antibody is present that antigen. An example of this type of tolerance is found
in a blood bank sample, it is often necessary to use special in the chimera, an individual who receives an in utero
techniques to identify all the antibodies. These techniques cross-transfusion of ABO-incompatible blood from a dizy-
and reagents were developed with an understanding of gotic (fraternal) twin.2 The chimera does not produce an-
antibody reactivity. tibodies against the A and B antigen of the twin, and the
ABO group of such an individual may appear as a testing
Host Factors discrepancy.
More frequently, the induction of tolerance is used to pre-
Host factors play a key role in an individual’s immune vent D-negative mothers from developing anti-D antibodies
response (Box 3–3). These factors are important in the host’s after delivering Rh-positive infants. When a D-negative
overall immune response and in various specific immune woman gives birth to a D-positive infant, she is exposed to
reactions. Each individual’s immune system is unique and D-positive RBCs, most of which occurs during delivery.
determines how that individual is able to resist disease. Host Approximately 50% to 70% of Rh-negative mothers develop
factors include nutritional status, hormones, genetic inher- anti-D antibodies on first exposure to D-positive cells. These
itance, age, race, sex, physical activity level, environmental antibodies can result in hemolytic disease of the newborn
exposure, and the occurrence of disease or injury. It is also (HDN) upon later pregnancies with a D-positive fetus. Use
believed that immune function decreases as age increases; of passive immunization to prevent the formation of these
this is demonstrated by increased vulnerability to cancer antibodies can prevent HDN. This is accomplished by
and viral illness in the aged population.12 A decrease in an- administering IgG Rh-immune globulin (RHIG) within 48
tibody levels in older individuals may result in false-negative to 72 hours after the birth of the infant. About 25% to 30%
reactions, especially in reverse ABO blood typing. of D-negative individuals are nonresponders and do not
In some instances, an individual’s race may be a factor in produce anti-D antibodies, even when subjected to repeated
susceptibility to certain diseases. A dramatic example of this
6888_Ch03_045-076 29/10/18 4:44 PM Page 65

Chapter 3 Fundamentals of Immunology 65

exposure to D-positive cells. Currently, testing to determine The routine methods used today have to be highly efficient
who will respond is not routine; therefore, it is recom- to meet clinical laboratory requirements.
mended that all D-negative mothers who deliver D-positive
newborns receive RHIG to prevent immunization.
Advanced Concepts
Many different types of tests are available, and it is impor-
Detection of RBC Antigen-Antibody tant to be familiar with the different purposes of each. In
Reactions vitro testing for the detection of antigens or antibodies may
be accomplished by commonly used techniques, including
The most important factors influencing the detection of RBC
hemagglutination (a special type of agglutination), precipi-
antigen-antibody reactions include: having the correct sam-
tation, agglutination inhibition, and hemolysis. Other
ple and the proper reagents; performing the correct test; and
techniques that quantify antigen or antibody with the use
understanding how the test should be interpreted.
of an enzyme or fluorescent label—such as enzyme-linked
Different tests in the blood bank may require different
immunosorbent assay (ELISA) or enzyme immunoassay
specimens. Some tests require the use of serum to ensure
(EIA), Western blotting (WB), and immunofluorescence
that adequate amounts of viable complement are available
(IF)—may be used in automated or semiautomated blood
for fixation by blood group antibodies. An anticoagulated
banking instrumentation.2 These techniques are also im-
sample would not be conducive to complement activation
portant in testing for viral pathogens in donor units and
studies because anticoagulants bind divalent Ca2+ and Mg2+
patient samples. Recently, blood bank automated methods
ions and inhibit complement activity. The commonly used
for the typing of whole blood units and patient samples
anticoagulant ethylenediaminetetraacetic acid (EDTA) at a
have become more popular and may become routine in the
ratio of 2 mg to 1 mL of serum will totally inhibit comple-
near future.
ment activation by binding calcium and, to a lesser extent,
In most transfusion laboratory testing, hemagglutination
magnesium. Another anticoagulant, sodium heparin, inhibits
reactions are the major technique used. Hemagglutination
the cleavage of C4. These problems can be avoided by using
methods for the analysis of blood group antigen-antibody
serum instead of plasma for blood bank procedures that
responses and typing for ABO, Rh, and other blood group
require fresh complement.
antigens is accomplished by red blood cell agglutination
Serum is obtained when no anticoagulant is used in the
reactions. Agglutination is a straightforward process and can
sample collection tube. The sample clots and is centrifuged
be shown to develop in two stages. In the first stage, sensi-
to separate the clotted cells and the liquid serum fraction.
tization, antigen binding to the antibody occurs. Epitopes
Currently, however, plasma is used routinely in place of
on the surfaces of RBC membranes combine with the antigen-
serum. Years of testing with both samples have shown that
combining sites (Fab region) on the variable regions of the
for most tests, plasma is comparable to serum. Plasma sam-
immunoglobulin heavy and light chains. Antigen and anti-
ples are preferred for DAT and elution studies because they
body are held together by various noncovalent bonds, and
lack fibrin strands, which can cause false-positives. Serum
no visible agglutination is seen at this stage.
should be removed from a clotted blood sample as soon as
In the second stage, a lattice-type structure composed
possible. If testing cannot begin immediately, then serum
of multiple antigen-antibody bridges between RBC antigens
should be removed and stored at 4°C for no longer than
and antibodies is formed. A network of these bridges forms,
48 hours. After this time, serum should be frozen at –50°C
and visible agglutination is present during this stage. The
or lower to retain complement activity. The complement sys-
development of an insoluble antigen-antibody complex, re-
tem may become activated during storage of preserved RBC
sulting from the mixing of equivalent amounts of soluble
products. For example, in citrate phosphate dextrose ade-
antigen and antibody, is known as a precipitation reaction.
nine (CPDA-1)–preserved RBCs, activation of the alternate
Agglutination inhibition is a method in which a positive
pathway can be caused by contact of plasma C3 with plastic
reaction is the opposite of what is normally observed in ag-
surfaces of blood bags. This may cause hemolysis that will
glutination. Agglutination is inhibited when an antigen-
become visible as the RBCs settle upon storage.13
antibody reaction has previously occurred in a test system.
The antigen and antibody cannot bind because another sub-
Traditional Laboratory Testing strate has been added to the reaction mixture and blocks the
Methods formation of the agglutinates. Inhibition reactions are used
in secretory studies to determine whether soluble ABO sub-
The various complex immunologic responses that are im- stances are present in body fluids and secretions. The se-
portant to blood banking have been studied by a number of creted substance can combine with the antibody and block
immunologic methods. These in vitro testing methods were RBC antigen-antibody reactions. People who have blood
developed with an understanding of the biochemistry and group antibodies in their body fluids are called secretors.
biophysics of the IS. Many of the methods used today are Hemolysis represents a strong positive result and indi-
modifications of previous blood bank techniques or more cates that an antigen-antibody reaction has occurred in
generalized immunologic testing methods that have been which complement has been fixed and RBC lysis occurs.
adapted to the needs of immunohematology laboratories.
6888_Ch03_045-076 29/10/18 4:44 PM Page 66

66 PART I Fundamental Concepts

The Lewis blood group antibodies anti-Lea and anti-Leb gravitational forces on the reactants and bringing reactants
may be regarded as clinically significant if hemolysis occurs closer together. High-speed centrifugation is one of the most
in their in vitro testing reactions. efficient methods used in blood banking. Under the right
ELISA (or EIA), IF, and WB techniques are immunologic centrifugation conditions, sensitized RBCs overcome their
methods based on detection and quantification of antigen natural repulsive effect (zeta potential) on each other and
or antibody by the use of enzyme or fluorescent labels. agglutinate more efficiently. Having RBCs in closer physical
Either the antigen or the antibody can be labeled in these proximity allows for an increase in antigen-antibody lattice
tests. These techniques measure the interaction of the bind- formation, which results in enhanced agglutination.
ing of antigen with antibody and are extremely sensitive.
Most of these techniques employ reagents that use either Antigen-Antibody Ratio
antigen or antibody, which is bound in a solid or liquid
Antigen and antibody have optimal concentrations; in ideal
phase in a variety of reaction systems ranging from plastic
reactive conditions, an equivalent amount of antigen and
tubes or plates to microscopic particles or beads. These
antibody binds. Any deviation from this decreases the effi-
methods use a separation system and washing steps to iso-
ciency of the reaction and a loss of the zone of equivalence
late bound and free fractions and a detection system to
between antigen and antibody ratio, which is necessary for
measure the amount of antigen-antibody interaction. The
agglutination reactions to occur. An excess of unbound im-
values of unknown samples are then calculated from the
munoglobulin leads to a prozone effect, and a surplus of
values of standards of known concentration.
antigen leads to a postzone effect (Fig. 3–8). In either situ-
ation, the lattice formation and subsequent agglutination
may not occur, which can give false-negative results. If this
Factors That Influence Agglutination
is suspected, steps need to be taken to correct it. If the prob-
Reactions
lem is excessive antibody, the plasma or serum may be
Typical of most biochemical reaction systems, agglutination diluted with the appropriate buffer. The problem of excessive
reactions are influenced by the concentration of the reactants antigen can be solved by increasing the serum-to-cell ratio,
(antigen and antibody) and by factors such as pH, tempera- which tends to increase the number of antibodies available
ture, and ionic strength. The surface charge, antibody isotype, to bind with each RBC. Therefore, antigen-antibody test sys-
RBC antigen dosage, and the use of enhancement media, anti- tems can be manipulated to overcome the effects of excessive
human globulin reagents, and enzymes are all important in antigen or antibody.
antigen-antibody reactions. The most important factors are Another reason antigen amount may be altered is due to
discussed in the following sections. weak expression of antigen on RBCs (dosage effect). Weak
expression occurs as a result of the inheritance of genotypes
Centrifugation that give rise to heterozygous expression of RBC antigens
and resultant weaker phenotypes. For example, M-positive
Centrifugation is an effective way to enhance agglutination RBCs from an individual having the genotype MM (homozy-
reactions because it decreases reaction time by increasing the gous) have more M antigen sites than M-positive cells from

Figure 3–8. Schematic representation of the


effects of varying concentrations of antigen and
antibody on lattice formation.

Equivalence
Antibody Excess (Optimum Proportions of Antigen Excess
(Prozone) Antigen and Antibody) (Postzone)
6888_Ch03_045-076 29/10/18 4:44 PM Page 67

Chapter 3 Fundamentals of Immunology 67

an individual having the MN (heterozygous) genotype. In IgG antibodies are those directed against Ss, Kell (Kk, Jsa,
the Kidd system, Jka homozygous inheritance has greater Jsb, Kpa, Kpb), Rh (DCEce), Lutheran (Lub), Duffy (Fya, Fyb),
RBC antigen expression than Jka heterozygous. The same is and Kidd (Jka, Jkb) antigens (see Fig. 3–8). IgM antibodies
true for Jkb homozygous and heterozygous expression. In are generally capable of agglutinating RBCs suspended in a
the Rh system, the C, c, E, and e antigens also show dosage 0.85% to 0.90% saline medium. The IgM antibody is 160 Å
effects. Dosage effect is sometimes used when preparing larger than an IgG molecule and approximately 750 times as
reagent RBCs for testing. efficient as IgG in agglutination reactions. This allows it to
easily bridge the distance between two RBCs.14
Effect of pH Another factor that contributes to the difference in re-
activity between the IgM and IgG molecules is the number
The ideal pH of a test system for antigen-antibody reactions of antigen-combining sites on each type of immunoglobu-
ranges between 6.5 and 7.5, which is similar to the pH of lin. The IgM molecule has the potential to bind 10 separate
normal plasma or serum. Exceptions include some anti-M antigen sites, while an IgG molecule has only two binding
and some Pr(Sp1) group antibodies that show stronger reac- sites per molecule, which implies that an IgG molecule
tivity below pH 6.5.14 Acidification of the test serum may aid would have to bind two RBCs with only one binding site
in distinguishing anti-M and anti-Pr(Sp1) antibodies from on each cell.14 However, IgM molecules rarely bind 10 sites,
other antibodies. owing to the size and spacing of antigens in relation to the
size and configuration of the IgM molecule. More typically,
Temperature when the IgM molecule attaches to two RBCs, two or three
antigen-combining sites attach to each RBC. Agglutination
Different isotypes of antibodies may exhibit optimal reactiv-
reactions involve more than one immunoglobulin mole-
ity at different temperatures. IgM antibodies usually react
cule, and these conditions are multiplied many times in
optimally at ambient temperatures or below 22°C at the
order to represent the agglutination reaction. Because of
immediate spin phase of testing, whereas IgG antibodies
the basic differences in the nature of reactivity between IgM
usually require 37°C incubation and react optimally at the
and IgG antibodies, different serologic systems must be
antihuman globulin (AHG) phase of testing. Because clini-
used to optimally detect both classes of clinically significant
cally significant antibodies may be in both temperature
antibodies. An overview of the serologic methods tradition-
ranges, it is important to do testing with a range of temper-
ally used for antibody detection in the blood bank labora-
atures (Fig. 3–9).
tory is outlined in Table 3–8.
Immunoglobulin Type
Enhancement Media
Examples of IgM antibodies that have importance in blood
Agglutination reactions for IgM antibodies and their corre-
banking include those against the ABH, Ii, MN, Lewis (Lea, Leb),
sponding RBC antigens are easily accomplished in saline
Lutheran (Lua), and P blood group antigens. Important
medium, as these antibodies usually do not need enhance-
ment or modifications to react strongly with antigens. How-
ever, detection of IgM antibodies may not have the same
clinical significance as the detection of most IgG antibodies
because IgG antibodies react best at 37°C and are generally
Types of Antibodies responsible for hemolytic transfusion reactions and HDN.
Reaction Phases To discover the presence of IgG antibodies, enhancement
techniques or potentiators are available (Table 3–9).
A, B, H One of the key ways to enhance the detection of IgG anti-
I bodies is to increase their reactivity. The net negative charge
Immediate M, N IgM surrounding RBCs (and most other human cells) in a cationic
Spin Phase Lea, Leb media is part of the force that repels RBCs from each other
P1 and is due to sialic acid molecules on the surface of RBCs.
D, C, E Most acids have a negative charge, and the large concentra-
Antiglobulin c,e IgG tion of these molecules on RBCs creates a “zone” of negative
Phase (37ºC) K, Fy, Jk charge around the RBC. This zone is protective and keeps
S, s RBCs from adhering to each other in the peripheral blood. A
Lea, Leb potential is created because of the ionic cloud of cations (pos-
itively charged ions) that are attracted to the zone of negative
charges on the RBC membrane (Fig. 3–10).14 This potential
around the RBC is called the zeta potential and is an expres-
Figure 3–9. Types of antibodies and reaction phases. (From Kutt, SM, Larison, sion of the difference in electrostatic charges at the RBC
PJ, and Kessler, LA: Solving Antibody Problems, Special Techniques. Ortho Diagnostic surface and the surrounding cations. IgM and IgG antibodies
Systems, Raritan, NJ, 1992, p 10, with permission.) have differences in how they react to the same zeta potential.
6888_Ch03_045-076 29/10/18 4:44 PM Page 68

68 PART I Fundamental Concepts

Table 3–8 Serologic Systems Used in Traditional Laboratory Methods for Red Blood Cell
Antibody Detection
Ig Class Purpose and Tests That Use
Reaction Commonly Mechanism of Serologic Type of Antibodies
Phase Detected Reaction System Commonly Detected
Immediate spin IgM IgM antibodies react best at ABO reverse testing Expected ABO alloantibodies
cold temperatures

IgM is an agglutinating anti- Cross-match Unexpected cold-reacting


body that has the ability to alloantibodies or autoantibodies
easily bridge the distance
between red blood cells.

37°C incubation IgG IgG antibodies react best at Antibody screening/


warm temperatures. identification

Autocontrol

Antibody screening/
identification

No visible agglutination Cross-match (if needed)


commonly seen.

IgG is sensitizing antibody Autocontrol


with fewer antigen-binding
sites than IgM and cannot
undergo the second stage
of agglutination, lattice
formation.

Complement may be bound


during reactivity, which may
or may not result in visible
hemolysis.

Antiglobulin test IgG Antihuman globulin (AHG) Antibody screening/ Unexpected warm-reacting
has specificity for the identification alloantibodies or autoantibodies
FC portion of the heavy
chain of the human IgG
molecule or complement
components.
Cross-match (if needed)

Autocontrol

AHG acts as a bridge cross- Direct antiglobulin test


linking red blood cells sensi- (DAT)
tized with IgG antibody or
complement.

Commercially available enhancement media reduces the zeta neutral and go into solution because of their microscopic
potential and allows the more positively charged antibodies size. There are several colloidal solutions currently utilized
to get closer to the negatively charged RBCs and therefore in blood bank testing to enhance agglutination reactions.
increases RBC agglutination by IgG molecules. Colloids include albumin, polyethylene glycol (PEG), poly-
brene, polyvinylpyrrolidone (PVP), and protamine. These
Protein Media substances work by increasing the dielectric constant
(a measure of electrical conductivity), which then reduces
Colloidal substances, or colloids, are a clear solution that the zeta potential of the RBC.
contains particles permanently suspended in solution. Col-
loidal particles are usually large moieties like proteins as Low Ionic Strength Solution Media
compared with the more familiar crystalloids, which usually
have small, highly soluble molecules, such as glucose, that Low ionic strength solutions (LISS), or low salt media,
are easily dialyzed. The colloidal solutes can be charged or generally contain 0.2% sodium chloride. They decrease
6888_Ch03_045-076 29/10/18 4:44 PM Page 69

Chapter 3 Fundamentals of Immunology 69

Table 3–9 Potentiators


Reagent Action Procedure Type of Antibody ID
Primarily IgM; IgG if incubated at 37°C

AHG Cross-links sensitized cells, resulting 1. DAT: AHG added directly to 1. Polyspecific; anti-IgG + anticomplement
in visible agglutination washed RBCs 2. IgG monospecific: anti-IgG only
2. IAT: serum + screen cells;
incubation at 37°C for time
determined by additive used;
cell washing before addition
of AHG

22% albumin* Causes agglutination by adjusting Incubation at 37°C for 15–60 min; IgG
zeta potential between RBCs cell washing prior to indirect
antiglobulin test (IAT)

LISS* Low ionic strength environment Incubation at 37°C for 5–15 min; IgG
causes RBCs to take up antibody cell washing before IAT
more rapidly

PEG* Increases test sensitivity; Incubation at 37°C for 10–30 min; IgG
cell washing before IAT
aggregates RBCs causing closer NOTE: The test mixture cannot
proximity of RBCs to one another, be centrifuged and examined
assisting in antibody cross-linking reliably for direct agglutination
after 37°C incubation.
Enzymes Reduces RBC surface charge; 1. One step: enzymes added Destroys Fya, Fyb, MNS; enhances reactiv-
destroys or depresses some RBC directly to serum/RBC mixture ity to Rh, Kidd, P1, Lewis, and I antibodies
antigens; enhances other RBC 2. Two step: RBC pretreated with
antigens enzymes before addition of
serum

*All additives should be added after the immediate spin phase immediately before 37°C incubation.
LISS = low ionic strength solutions; PEG = polyethylene glycol

Figure 3–10. Schematic representation of No Agglutination Erythrocyte


the ionic cloud concept and its relevance to Ionic Cloud
hemagglutination induced by IgM and IgG anti-
bodies. Compare the size of the IgG antibody
with that of the IgM molecule. The size of the IgG IgG
molecule is not large enough to span the distance
between two adjacent RBCs. 14 nm

25 nm

35 nm

IgM

Agglutination
6888_Ch03_045-076 29/10/18 4:44 PM Page 70

70 PART I Fundamental Concepts

the ionic strength of a reaction medium, which reduces the Antihuman Globulin Reagents
zeta potential and therefore allows antibodies to react more
efficiently with RBC membrane antigens. LISS media are When RBCs become coated with antibody, complement, or
often used because they result in an increased rate of anti- both, but do not agglutinate in regular testing, special
body uptake during sensitization and a decreased reaction reagents are needed to produce agglutination. The direct
incubation time (from 30 to 60 minutes to 5 to 15 minutes antihuman globulin (AHG) test is designed to determine if
as compared with protein potentiators such as albu- RBCs are coated with antibody or complement or both. Poly-
min).14,15 However, this media can result in false-positive specific AHG can determine if RBCs have been sensitized
reactions and may require testing to be repeated with with IgG antibody or complement (components C3b or
albumin. C3d) or both. Monospecific AHG reagents react only
with RBCs sensitized with IgG or complement.15 Refer to
Polyethylene Glycol and Polybrene Chapter 5, “The Antiglobulin Test.”
Blood bank reagents can be either polyclonal or monoclonal
in source. In the indirect antiglobulin test, the same AHG
Advanced Concepts reagents are used to detect antibodies or complement that
PEG and polybrene are macromolecule additives used with have attached to RBC membranes but with a prior incubation
LISS to bring sensitized RBCs closer to each other in order step with serum (or plasma). If the antibodies present in
to facilitate antibody cross-linking and agglutination reac- serum do not cause RBC agglutination but only sensitize the
tions. They are often used in place of albumin. Polybrene RBCs, then the AHG reagents will allow for agglutination to
can detect ABO incompatibility and clinically significant occur by cross-linking the antibodies on the RBCs. The use
IgG alloantibodies, whereas PEG produces very specific of AHG reagents allows blood bank testing to be more sensi-
reactions with reduction in false-positive or nonspecific tive, and the development of AHG is one of the milestones in
reactions. PEG is considered to be more effective than blood bank testing (see Tables 3–8 and 3–9).
albumin, LISS, or polybrene for detection of weak anti-
bodies. These reagents have been used in automated and Chemical Reduction of IgG and IgM Molecules
manual testing systems.
Advanced Concepts
Proteolytic Enzymes There are special reagents available that can be used to help
identify the different antibodies present in a mixture of
Enzymes are protein molecules that function by altering alloantibodies or alloantibodies occurring with autoantibod-
reaction conditions and bringing about changes in other ies. The reagents generally act on covalent sulfhydryl bonds
molecules without being changed themselves. Only a small and facilitate antibody identification by removal of either
part of a large enzyme molecule reacts with another mole- IgG or IgM antibodies. Dithiothreitol (DTT) and β-2-
cule, called its substrate. Enzymes are specific for the mole- mercaptoethanol (2-ME) are thiol-reducing agents that break
cules they target and can modify proteins, nucleic acids, the disulfide bonds of the J (joining) chain of the IgM mol-
carbohydrates, or lipids. Proteolytic enzymes target protein ecule but leave the IgG molecule intact.10 Another reagent,
molecules. Certain enzymes have been found to modify ZZAP, which consists of a thiol reagent plus a proteolytic
various blood group antigens and are useful in testing, enzyme, causes the dissociation of IgG molecules from the
especially in cases in which there are multiple antibodies surface of sensitized RBCs and alters the surface antigens of
present in a sample. the RBC.2 Chemical reduction of the disulfide bond of the
Enzymes used in the detection and identification of blood IgG molecule is also used to produce chemically modified
group antibodies include ficin (isolated from fig plants), reagents that react with RBCs in saline. Sulfhydryl com-
papain (from papaya), trypsin (from pig stomach), and pounds reduce the strong but less flexible covalent disulfide
bromelin (from pineapple). It is thought that treating RBCs bonds in the hinge region of the IgG molecule, allowing
with enzymes results in the release of sialic acid from the the Fab portions more flexibility in facilitating agglutination
membrane, with a subsequent decrease in the negative reactions.14
charges and zeta potential of the RBCs. It has also been
suggested that enzyme treatment removes hydrophilic gly-
coproteins from the membrane of RBCs, causing the mem- Monoclonal Versus Polyclonal
brane to become more hydrophobic, which would allow Reagents
RBCs to come closer together. Also, because of the removal
of glycoproteins from the membrane, antibody molecules Traditionally, polyclonal antisera reagents have been pro-
may no longer be subject to steric hindrance from reacting duced by immunizing donors with small amounts of RBCs
with RBC antigens. The use of enzymes provides enhanced positive for an antigen that they lack and then collecting
antibody reactivity to Rh, Kidd, P1, Lewis, and I antigens the serum and isolating antibodies against that antigen.
and destroys or decreases reactivity to Fya, Fyb, M, N, and AHG reagents were originally made by injecting animals
S antigens.15 (usually rabbits) with human globulin components and then
6888_Ch03_045-076 29/10/18 4:44 PM Page 71

Chapter 3 Fundamentals of Immunology 71

collecting the antihuman antibodies. One antigen can have a Cells that are coated with fluorescent-labeled antibody emit
number of different epitopes, and polyclonal reagents are di- a brightly fluorescent color of a specific wavelength. Fluo-
rected against multiple epitopes found on the original antigen rescent dyes are used as markers in immunologic reactions,
used to stimulate antibody production. (Refer to Chapter 5.) and a fluorochrome-labeled antibody can be visualized by an
Monoclonal reagents are directed against specific epi- instrument that detects emitted light. There are many differ-
topes. They are made by hybridoma technology with spleen ent fluorochromes available that have various colors, and
lymphocytes from immunized mice that are fused with rap- they can be coupled to nearly any antibody with minimal
idly proliferating myeloma cells. The spleen lymphocytes problems with spectral overlap.
have single epitope specificity, while the myeloma cells (a Flow cytometry can be used to obtain quantitative and
type of immortalized, cultured cell) make vast amounts of qualitative data on cell populations and to sort different cell
antibody. After extensive screening and testing, these very populations. There are direct and indirect procedures for
efficient hybrid cells are selected and cultured to produce labeling with fluorochromes. In a direct procedure, the specific
lines of immortal cell clones that make a large amount of one antibody, called the primary antibody, is directly conjugated
type of antibody that reacts with one specific epitope. Mono- with a fluorescent dye and reacts with a specific antigen.
clonal reagents do not use human donors and therefore do Indirect procedures need a secondary antibody that is con-
not use a human source for reagent purposes. jugated to a fluorochrome, which reacts with an unlabeled
Monoclonal reagents have several important advantages specific primary antibody that has reacted with antigen. In-
over polyclonal reagents. Because monoclonal reagents are direct methods often require more complex analysis because
produced from immortal clones maintained in vitro, no batch these reactions have higher amounts of nonspecific binding.
variation exists, and nearly unlimited high titers of antibodies The secondary antibody is usually made against the species
can be produced. Also, the immortal clones can be kept grow- of the primary antibody.
ing in in vitro culture for years without loss of production and The principle of flow cytometry is based on the scattering
are therefore cost-efficient. Monoclonal antibodies react very of light as cells are bathed in a fluid stream through which a
specifically and often have higher affinities. For these reasons, laser beam enters. The cells move into a chamber one at a
monoclonal reagents are not subject to cross-reactivity and time and absorb light from the laser. If labeled antibody is
interference from nonspecific reactions and can react strongly bound to the cell, then the light emitted is different from light
with the small quantities of antigen in some antigen sub- emitted by cells without antibody. The flow cytometer instru-
groups; therefore, AHG phase testing may not be needed. ment makes the distinction between different wavelengths of
However, there are some disadvantages of monoclonal an- light. The light signal is amplified and analyzed. There are a
tisera use, such as overspecificity. Not all monoclonal antisera number of components to the flow cytometry system, includ-
have the ability to fix complement and thus may cause false- ing one or more lasers for cell illumination, photodetectors
negative results. In addition, problems with oversensitivity for signal detection, a possible cell-sorting device, and a com-
may cause false-positive results. Some of the disadvantages of puter to manage the data.16 Fluorescence is used in place of
monoclonal reagents can be overcome by using blends of dif- hemagglutination as the endpoint of the reaction. Immuno-
ferent monoclonal reagents or by using polyclonal reagents fluorescent antibodies and flow cytometry have been used to
and monoclonal reagents together.2 Regardless, monoclonal quantify feto-maternal hemorrhage, identify transfused cells
antisera has generally replaced polyclonal antisera and is and follow their survival in recipients, measure low levels of
used for HLA typing, AHG testing, and RBC and lymphocyte cell-bound IgG, and distinguish homozygous from heterozy-
phenotyping. gous expression of blood group antigens.2

Nontraditional Laboratory Methods Diseases Important in Blood Bank


Serologic Testing

Advanced Concepts
Sophisticated testing methods have become more common- Advanced Concepts
place in transfusion laboratories. These technologies are A number of immune-mediated diseases are important in
discussed in more detail in Chapter 12, “Blood Bank Test- blood bank testing. For further detail, please refer to the
ing Technologies and Automation.” appropriate chapter in this textbook. However, some of
these diseases will be discussed within the context of this
chapter to emphasize their importance in blood testing.
Flow Cytometry

Some of the most vital techniques used to study immuno- Immunodeficiency


logic reactions and systems are fluorescence-assisted cell
sorting (FACS) and flow cytometry. Flow cytometry makes Immunodeficiency diseases can result from defects at many
use of antibodies that are tagged with a fluorescent dye. Flu- different levels of immune function and may be congenital
orescence occurs when the compound absorbs light energy or acquired. They can result from defects in either innate or
of one wavelength and emits light of a different wavelength. adaptive immunity or both, and often the cause is not
6888_Ch03_045-076 29/10/18 4:44 PM Page 72

72 PART I Fundamental Concepts

known. Some of the more well-known immunodeficiencies Type III reactions, like type II, involve phagocytes and IgG
are listed in Box 3–4.17 Immunodeficiencies can influence and IgM and complement. Type III reactions result in tissue
blood bank test results and transfusion decisions, such as damage from the formation of immune complexes of
when there is a false-negative reverse grouping for ABO due antigen-antibody aggregates, complement, and phagocytes
to low immunoglobulin levels. and are therefore very serious. Penicillin and other drug-
induced antibodies can lead to hemolytic reactions through
Hypersensitivity type III hypersensitivity. The type IV reaction involves only
T cell–mediated responses and their cytokines and can be
Hypersensitivity is an inflammatory response to a foreign fatal if untreated. The most important type IV reaction is
antigen and can be cell- or antibody-mediated or both. There graft-versus-host, of which there is also more than one type.
are four different types of hypersensitive reactions, and the Immunocompromised and immunosuppressed patients
symptoms and treatment required for each are different. All must receive irradiated blood products so that T lympho-
four types can be caused by blood product transfusions and cytes do not engraft and attack the host tissues. Type IV
may be the first sign of a transfusion reaction. Type I reac- reactions are a problem for bone marrow transplant and stem
tion, also called anaphylaxis or immediate hypersensitivity, cell transfusion recipients as well. (Refer to Chapter 17,
involves histamine release by mast cells or basophils with “Adverse Effects of Blood Transfusion.”)
surface IgE antibody. It can occur in IgA-deficient individuals
who receive plasma products containing IgA. Urticarial re- Monoclonal and Polyclonal Gammopathies
actions (skin rashes) may also result from transfusion of cer-
tain food allergens or drugs in plasma products. A type II Plasma cell neoplasms result in proliferation of abnormal im-
reaction can involve IgG or IgM antibody with complement, munoglobulin from either a single B-cell clone (monoclonal
phagocytes, and proteolytic enzymes. HDN or transfusion gammopathies) or multiple clones (polyclonal gammo-
reactions caused by blood group antibodies and autoimmune pathies) and may be a specific isotype or involve just light
hemolytic reactions are all type II reactions. or heavy chain molecules. Increased serum viscosity is a

BOX 3–4

Classification of Immunodeficiency Disorders


Antibody (B Cell) Immunodeficiency Disorders • Cellular immunodeficiency with abnormal immunoglobulin synthesis
(Nezelof syndrome)
• X-linked hypogammaglobulinemia (congenital
hypogammaglobulinemia) • Immunodeficiency with ataxia-telangiectasia
• Transient hypogammaglobulinemia of infancy • Immunodeficiency with eczema and thrombocytopenia
(Wiskott-Aldrich syndrome)
• Common, variable, unclassifiable immunodeficiency (acquired by
hypogammaglobulinemia) • Immunodeficiency with thymoma
• Immunodeficiency with hyper-IgM • Immunodeficiency with short-limbed dwarfism
• Selective IgA deficiency • Immunodeficiency with adenosine deaminase deficiency
• Selective IgM deficiency • Immunodeficiency with nucleoside phosphorylase deficiency
• Selective deficiency of IgG subclasses • Biotin-dependent multiple carboxylase deficiency
• Secondary B-cell immunodeficiency associated with drugs, • Graft-versus-host disease
protein-losing states • AIDS
• X-linked lymphoproliferative disease Phagocytic Dysfunction
Cellular (T Cell) Immunodeficiency Disorders • Chronic granulomatous disease
• Congenital thymic aplasia (DiGeorge syndrome) • Glucose-6-phosphate dehydrogenase deficiency
• Chronic mucocutaneous candidiasis (with or without endocrinopathy) • Myeloperoxidase deficiency
• T-cell deficiency associated with purine nucleoside phosphorylase • Chediak-Higashi syndrome
deficiency • Job’s syndrome
• T-cell deficiency associated with absent membrane glycoprotein • Tuftsin deficiency
• T-cell deficiency associated with absent class I or II MHC antigens or • Lazy leukocyte syndrome
both (base lymphocyte syndrome) • Elevated IgE, defective chemotaxis, and recurrent infections
Combined Antibody-Mediated (B Cell) and Cell-
Mediated (T Cell) Immunodeficiency Disorders
• Severe combined immunodeficiency disease (autosomal recessive,
X-linked, sporadic)

Ammann, AJ: Mechanisms of immunodeficiency. In Stites, DP, Terr, AI, and Parslow, TG (eds): Basic and Clinical Immunology, 8th ed. Appleton & Lange, Norwalk, 1994, with
permission.
6888_Ch03_045-076 29/10/18 4:44 PM Page 73

Chapter 3 Fundamentals of Immunology 73

result of these diseases and can interfere with testing. The antibodies and may require exchange transfusion. Antigens
increased concentrations of serum proteins can cause of the ABO, Rh, and other blood group systems such as Kell
nonspecific aggregation (as opposed to agglutination) of have been shown to cause HDFN. (Refer to Chapter 20,
erythrocytes called rouleaux, which is seen as a stacking of “Hemolytic Disease of the Fetus and Newborn [HDFN].”)
RBCs. It often occurs in multiple-myeloma patients. If
rouleaux is suspected, saline replacement technique may Blood Product Transfusions and the
be needed to distinguish true cell agglutination from non- Immune System
specific aggregation. Rouleaux primarily causes problems in
the ABO reverse grouping, antibody screening, and compat- The immune system may undergo transient depression fol-
ibility testing procedures. However, excess immunoglobulin lowing the administration of blood and blood products.
coating red blood cells can cause spontaneous aggregation This is referred to as transfusion-related immunomodula-
to occur as well. tion (TRIM). With a weakened immune system, there is an
increased risk of an individual developing infections or
Autoimmune Disease certain cancers. The exact mechanism(s) causing TRIM are
not known and have not been fully elucidated, but three
Autoantibodies are produced against the host’s own cells and major mechanisms seem to have emerged: clonal deletion,
tissues. It is unknown why this loss of tolerance to self- induction of anergy, and immune suppression. In clonal
antigens occurs, but there are many possible explanations deletion, alloreactive lymphocytes are inactivated and
such as aberrant antigen presentation, failure to obtain clonal removed, thus preventing a potential graft rejection (as in
deletion, anti-idiotypic network breakdown, and cross- kidney organ). With induction of anergy, there is an unre-
reactivity between self and nonself antigens. Autoimmune sponsiveness of the immune system, most likely due to
hemolytic anemias are an important problem in testing and a failure of T cells to respond. In immune suppression,
transfusion. They may produce antibodies that cause responding cells appear to be inhibited by some sort of
RBC destruction and anemia and result in antibody- or cellular mechanism or cytokine.
complement-sensitized RBCs. The direct antiglobulin test Immune suppression can be due to a number of factors.
should be done to detect sensitized RBCs and determine if Certain cytokines, including interleukins and some growth
the cells are coated with antibody or complement. Special factors, can decrease immune responsiveness. Medically in-
procedures such as elution or chemical treatment to remove duced suppression of some IS components is critical at times
antibody from cells may be required to prepare RBCs so the IS does not become overactivated and attack host cells.
for antigen typing. Serum autoantibodies may interfere Immature T and B cells that recognize host cells, and poten-
with testing for clinically significant alloantibodies. Special tially destroy them, must be removed before they can deve-
reagents and procedures to denature immunoglobulins lop into mature lymphocytes. Specific suppressor cells
may be required to remove autoantibodies from serum so have recently been isolated that have unique CD profiles
they do not interfere with the testing. (Refer to Chapter 21, and play a role in modulating lymphocyte function. Immune
“Autoimmune Hemolytic Anemias.”) suppression can also occur when there is too little im-
munoglobulin made or when too many T and B cells are lost
Hemolytic Disease of the Newborn through severe infection, extreme immune stimulation, or
IS organ failure.
Hemolytic disease of the fetus and newborn (HDFN) can re- It is believed that some constituents of cellular blood
sult when the maternal IS produces an antibody directed at products may be responsible for TRIM. These constituents
an antigen present on fetal cells but absent from maternal include allogeneic mononuclear cells, chemicals released by
cells. The mother is exposed to fetal RBCs as a result of allogenic mononuclear cells as they age, and soluble HLA
fetomaternal transfer of cells during pregnancy or childbirth. peptides that circulate in plasma. The decision to transfuse
Maternal memory cells can cause a stronger response during blood products is approached with caution, and if transfu-
a second pregnancy if the fetus is positive for the sensitizing sion is required, the patient’s immune status must be taken
antigens. IgG1, IgG3, and IgG4 are capable of crossing the into consideration. The use of leukoreduction filters may
placenta and attaching to fetal RBCs, whereas IgG2 and IgM help to reduce the risk for TRIM. (Refer to Chapter 16,
are not. Severe HDFN is most often associated with IgG1 “Transfusion Therapy.”)
6888_Ch03_045-076 29/10/18 4:44 PM Page 74

74 PART I Fundamental Concepts

SUMMARY CHART
 The immune system interacts with other host systems and  Membrane-bound antibodies are manufactured by
maintains the host equilibrium. The two major roles for B cells and inserted into their cell membranes, where
the IS are: they act as antigen receptors.
• Protection from pathogens and foreign substances  Stimulated B cells differentiate into plasma cells to
• Removal of abnormal and damaged host cells secrete humoral immunoglobulin; B cells receive T-cell
 The two major branches of the IS are: help for antibody production and for immunologic
• Innate, or natural—first to respond, primitive IS memory; a single B-cell clone manufactures Ig of a sin-
• Acquired, or adaptive—the specific, evolved IS gle specificity for a specific antigen for its entire cell
 The two major arms of the acquired IS are: lifetime.
• Humoral, mediated by B cells and antibody  The primary, or original, immune response occurs after
production the first exposure to an antigen. The secondary, or
• Cellular, mediated by T cells and lymphokines anamnestic, immune response happens after a second
 The basic mechanisms used by the IS are: exposure with the same specific antigen.
• Recognition of self and nonself organisms, cells, and  Complement consists of a large group of different en-
tissues zymatic proteins (convertases/esterases) that circulate
• Removal of unwanted organisms, cells, and tissues in an inactive proenzyme form. Once the cascade is
(self or nonself) started, complement proteins activate each other in a
• Repair of damaged host tissues sequence to form products that are involved in opti-
 The acquired IS demonstrates diversity and uniqueness: mizing phagocytosis and cell lysis.
• Individual B and T cells have unique membrane  Complement can be activated through three pathways:
molecules that have limitless configurations to • The classical pathway is initiated by antigen-antibody
match nearly any antigen in the environment. complexes and requires C1q for activation to proceed.
• Each individual lymphocyte has one unique recep- • The alternative pathway is activated by certain
tor per cell that recognizes one epitope. macromolecules on the cell walls of bacteria, fungi,
• Antibodies and T-cell receptors recognize and react parasites, and tumor cells and requires C3b; serum
only with the antigen that matches and fits their factors B, D, properdin; and initiating factor.
specific configuration. • The lectin pathway is activated by binding of MBL
• Selected T and B cells can remain dormant and later to microbes.
respond more rigorously upon secondary exposure  All three complement pathways meet at a common point
of a previously recognized antigen. in the cascade and result in the formation of the mem-
 The acquired IS demonstrates tolerance: This indicates brane attack complex to remove unwanted cells.
that immune responses against the host are either  There are five classes (or isotypes) of immunoglobu-
removed or downregulated. lins, all of which have a basic four-chain protein struc-
 There are three types of lymphocytes: T cells, B cells, ture consisting of two identical light chains and two
and NK cells. identical heavy chains. Disulfide (covalent) bonds link
 T cells (or lymphocytes) have a TCR, which is usually each light chain to a heavy chain and link the two
associated with the CD3 complex, and T cells require heavy chains to each other.
APCs to respond to antigens.  Antibody molecules are isotypic (based on heavy chain
 There are two well-characterized subpopulations of subtype), allotypic (based on one heavy chain muta-
T cells distinguished by CD markers—T helper tion), or idiotypic (based on hypervariable and variable
(TH, CD4-positive) and T cytotoxic (TC, CD8-positive) regions of light and heavy chains) and are reflected in
lymphocytes. the Ig sequences.
 TH lymphocytes have CD4 markers on their cell mem-  Blood group antibodies may be characterized by such
branes, provide B-cell help to stimulate the immune factors as epitope and variable region diversity (mon-
response, release lymphokines when stimulated, and oclonal or polyclonal), mode of sensitization (naturally
recognize antigens in association with MHC class II occurring or immune), expected or unexpected pres-
molecules. ence in routine serum samples, isotype class (IgM, IgG,
 TC lymphocytes have the CD8 marker on their mem- or, rarely, IgA), activity (warm or cold reactive or both,
branes and can eliminate specific target cells without agglutinating or sensitizing), clinical significance,
the help of antibody (cytotoxicity). alloantibody or autoantibody specificity, serum titer,
and chemical reactivity (influence of enzymes, sensitivity
 B cells (or lymphocytes) make up about 5% to 15% of
to pH, DTT, or 2-ME reagents).
circulating lymphocytes and are characterized by their
membrane-bound antibodies (or immunoglobulins).
6888_Ch03_045-076 29/10/18 4:44 PM Page 75

Chapter 3 Fundamentals of Immunology 75

9. Which portion of the immunoglobulin molecules con-


Review Questions
tains complement binding sites?
1. Which of the following is not involved in the acquired a. Heavy chain variable region
(adaptive) immune response? b. Light chain variable region
c. Heavy chain constant region
a. Phagocytosis
d. Light chain constant region
b. Production of antibody or complement
c. Induction of immunologic memory 10. Which complement pathway is activated by the forma-
d. Accelerated immune response upon subsequent tion of antigen-antibody complexes?
exposure to antigen a. Classical
2. Which cells are involved in the production of antibodies? b. Alternative
c. Lectin
a. Dendritic cells
d. Retro
b. T lymphocytes
c. B lymphocytes 11. Which of the following is known as the “recognition
d. Macrophages unit” in the classical complement pathway?
3. Which of the following cells is involved in antigen recog- a. C1q
nition following phagocytosis? b. C3a
c. C4
a. B lymphocytes
d. C5
b. T lymphocytes
c. Macrophages 12. Which of the following is known as the “membrane
d. Granulocytes attack complex” in the classical complement pathway?
4. The role of the macrophage during an antibody response a. C1
is to: b. C3
c. C4, C2, C3
a. Make antibody.
d. C5b, C6, C7, C8, C9
b. Lyse virus-infected target cells.
c. Activate cytotoxic T cells. 13. Which of the following immunoglobulin classes is
d. Process antigen and present it. capable of crossing the placenta and causing hemolytic
disease of the newborn?
5. Which of the following immunoglobulins is produced in
the primary immune response? a. IgA
b. IgE
a. IgA
c. IgG
b. IgE
d. IgM
c. IgG
d. IgM 14. Which of the following refers to the effect of an excess
amount of antigen present in a test system?
6. Which of the following immunoglobulins is produced in
the secondary immune response? a. Postzone
b. Prozone
a. IgA
c. Zone of equivalence
b. IgE
d. Endzone
c. IgG
d. IgM 15. Which of the following refers to the presence of an
excess amount of antibody present in a test system?
7. Which of the following MHC classes encodes comple-
ment components? a. Postzone
b. Prozone
a.Class I
c. Zone of equivalence
b.Class II
d. Endzone
c.Class III
d.Class IV 16. Which one of the following properties of antibodies is
NOT dependent on the structure of the heavy chain
8. Which of the following immunoglobulins is most efficient
constant region?
at binding complement?
a. Ability to cross the placenta
a. IgA
b. Isotype (class)
b. IgE
c. Ability to fix complement
c. IgG
d. Affinity for antigen
d. IgM
6888_Ch03_045-076 29/10/18 4:44 PM Page 76

76 PART I Fundamental Concepts

17. Molecules that promote the update of bacteria for 6. Rieben R, Buchs JP, Flückiger E, Nydeggar UE. Antibodies
phagocytosis are: to histo-blood group substances A and B: agglutination titers,
Ig class, and IgG subclasses in healthy persons of different age
a. Opsonins. categories. Transfusion. 1991;31:607-15.
b. Cytokines. 7. Sokol RJ, Hewitt S, Booker DJ, Bailey A. Red cell autoantibodies,
c. Haptens. multiple immunoglobulin classes, and autoimmune hemolysis.
d. Isotypes. Transfusion. 1990;30:714-17.
8. Chen J, Cerutti A. The function and regulation of immuno-
18. Select the term that describes the unique confirmation globin D. Curr Opin Immunol. 2011;23(3):345-52.
of the antigen that allows recognition by a correspond- 9. Klein, HG. Red cell antibodies against self-antigens, bound
antigens and induced antigens. In: Anstee DJ, editor. Mollison’s
ing antibody. blood transfusion in clinical medicine. 12th ed. Oxford, UK:
a. Immunogen Wiley-Blackwell; 2014. p. 259-302.
b. Epitope 10. Klein, HG. ABO, Lewis and P groups and Ii antigens. In: Anstee
c. Avidity DJ, editor. Mollison’s blood transfusion in clinical medicine.
12th ed. Oxford, UK: Wiley-Blackwell; 2014. p. 118-66.
d. Clone
11. Turgeon ML. Fundamentals of immunohematology. 2nd ed.
19. Which of the following terms refers to the net negative Philadelphia: Lippincott Williams & Wilkins; 2003.
12. Thompson WW, Shay DK, Weintraub E, Brammer L, Cox N,
charge surrounding red blood cells? Anderson LJ, et al. Mortality associated with influenza and
a. Dielectric constant respiratory syncytial virus in the United States. JAMA.
b. Van der Waals forces 2003;289(2):179-86.
c. Hydrogen bonding 13. Schleuning M, Schmid-Haslbeck M, Utz H, Jochum M, Heim M,
Mempel W, et al. Complement activation during storage of blood
d. Zeta potential
under normal blood bank conditions: Effects of proteinase
inhibitors and leukocyte depletion. Blood. 1992;79:3071-75.
References 14. Issitt PD, Anstee DJ. Applied blood group serology. 4th ed.
Durham (NC): Montgomery Scientific; 1998. p. 34-35.
1. Waytes AT, Malbran A, Bobak DA, Fries LF. Pre-ligation of CR1
15. Kutt SM, Larison PJ, Kessler LA. Rh blood group system anti-
enhances IgG-dependent phagocytosis by cultured human
gens, antibodies, nomenclature, and testing. Raritan (NJ):
monocytes. J Immunol. 1991;146:2694.
Ortho Diagnostic Systems;1990. p. 13-14.
2. Fung MK, Eder AF, Spitalnik SL, Westhoff CM. Technical
16. Stevens, CD, Miller LE. Clinical immunology and serology:
manual. 19th ed. AABB, 2017.
a laboratory perspective. 4th ed. Philadelphia: FA Davis
3. Sunshine G, Coico R. Immunology: a short course. 7th ed.
Company; 2017.
Hoboken (NJ): John Wiley & Sons; 2015.
17. Ammann, AJ. Mechanisms of immunodeficiency. In: Stites DP,
4. Parslow TG, Stites DP, Imboden JB. Medical immunology.
Terr AI., Parslow, TG, editors. Basic and clinical immunology.
10th ed. Terr AI, editor. New York: McGraw-Hill; 2001.
8th ed. Norwalk: Appleton & Lange; 1994.
5. Nance SJ, Arndt PA, Garratty G, Nelson JM. Correlation of IgG
subclass with the severity of hemolytic disease of the newborn.
Transfusion. 1990;30:381-82.
6888_Ch04_077-102 29/10/18 4:45 PM Page 77

Chapter 4
Concepts in Molecular Biology
Maria P. Bettinotti, PhD, dip. ABHI, Barbara Kraj, PhD, MLS(ASCP)CMMBCM,
and Jules G. Zinni, MLS(ASCP)CMMBCM

Introduction Detection of Nucleic Acids and Red Blood Cell Genotyping


DNA Is the Genetic Material Proteins Genetic Basis of Blood Groups
Features Relevant to Molecular Nucleic Acid Hybridization Molecular Basis of Blood Group
Techniques PCR-Based and Other Amplification Polymorphism
Expression of Genetic Information Techniques Clinical Applications of Red Blood
The Central Dogma of Molecular Biology, Antibodies as Probes for Proteins Cell Genotyping
Expanded Techniques for Studying Gene Case Studies
Recombinant DNA Polymorphism Case 4-1
The Coding Sequence of a Gene Restriction Fragment Length Case 4-2
Tools for DNA Cloning Polymorphism Case 4-3
Expression of Cloned Genes: PCR and Allele-Specific Probes Summary Chart
Recombinant Proteins in Clinical Use DNA Sequencing Review Questions
The Polymerase Chain Reaction DNA Profiling or “Fingerprinting” References
DNA Sequencing Systems Biology

OBJECTIVES
1. Refer to the contributions by Griffith, Avery, Hershey, and Chase, and explain how DNA was proven to be the carrier of genetic
information.
2. Explain the significance of Chargaff’s rules and x-ray diffraction studies by Wilkins and Franklin in Watson and Crick’s discovery
of the DNA double helix structure.
3. Describe DNA features fundamental in the design of molecular assays and define the applicable terms: complementarity,
hydrogen bonding, melting point, denaturation, annealing, and polarity.
4. Explain what the central dogma of molecular biology is and how it expanded after the discovery of reverse transcriptase
enzyme.
5. Explain what the recombinant DNA technology is and describe the tools used in molecular cloning: restriction endonucleases,
vectors, and host cells.
6. Define gene expression and explain how it may be studied and used in manufacturing of recombinant proteins.
7. Summarize the procedures of plasmid DNA isolation and gel electrophoresis.
8. Explain the principles of the polymerase chain reaction as an in vitro molecular procedure mimicking the process of semi-
conservative DNA replication occurring in vivo.
9. Discuss the differences between classic PCR, reverse-transcriptase, and real-time PCR.
10. Describe the principle of transcription-mediated amplification.
11. Describe Sanger’s sequencing dideoxy chain termination method.
12. Recognize the differences between immunoblotting and hybridization methods: Southern and Northern analysis, microarrays,
and fluorescent in situ hybridization.
Continued

77
6888_Ch04_077-102 29/10/18 4:45 PM Page 78

78 PART I Fundamental Concepts

OBJECTIVES—cont’d
13. Explain major principles of methods used in studying gene polymorphisms: restriction fragment length polymorphism (RFLP),
variable number of tandem repeats (VNTR), sequence-specific PCR (SSP), and sequence-specific oligonucleotide probe (SSOP).
14. Explain how nucleic acid testing (NAT) in donor blood improves the process of screening for infectious disease.
15. Describe the applications of red blood cell (RBC) molecular antigen typing.

Introduction some of the experiments and concepts that led to the discov-
ery and understanding of the mechanisms underlying these
Molecular biology is the study of macromolecule interactions phenomena are reviewed. These experiments are direct pred-
that take place in the living cell. For the molecular biologist, ecessors of modern benchtop molecular techniques relevant
the master molecules are the nucleic acids: the deoxyribo- to immunohematology. This chapter provides a brief intro-
nucleic acid (DNA) and the ribonucleic acid (RNA). Accord- duction to molecular biology and supplements the descrip-
ing to the original central dogma of molecular biology, the tion of molecular applications presented in other sections of
basic information of life flows from DNA through RNA to pro- this book. Blood group genotyping, a relatively new and
teins.1 By means of the genetic code, a sequence of nucleotides evolving application of molecular technology in transfusion
within a gene is transcribed and translated into a sequence of medicine, is introduced at the end of this chapter.
amino acids that form a peptide or protein. The processes of
transcription and translation (reviewed in Chapter 2, “Basic DNA Is the Genetic Material
Genetics”) are referred to as gene expression. The key to the
chemistry of life is the variety of gene expression products that Living organisms can replicate and pass hereditary traits
are unique structural or functional protein molecules. (genes) to successive generations; this is their most distinctive
A protein may be shaped into a building component of a characteristic. Refer to Chapter 2 for an in-depth review of
cell. For example, spectrin is a main structural protein of the Mendel’s laws and other concepts fundamental to heredity.
red blood cell (RBC) skeleton. Defects in the spectrin gene lead In 1928, the British microbiologist Frederick Griffith pre-
to hereditary spherocytosis (HS), a condition characterized sented a model system that was key to demonstrating that
by fragile, spherical RBCs, which the spleen traps and destroys. DNA was the material that carried the genetic information
A protein may temporarily bind particular substrate mole- responsible for the features of living organisms (Fig. 4–1).
cules, enabling these molecules to chemically react with each He used two naturally occurring strains of Streptococcus
other. Proteins that act as catalysts of these chemical reac-
tions are called enzymes. A good example is the ABO blood
1 2
group system. The H gene codes for protein fucosyl trans-
ferase, an enzyme that adds sugar L-fucose to a series of pre- R Strain
S Strain
cursor structures. Once L-fucose has been added, the A and
B gene–specified enzymes can add sugars to the chains that
now carry the H residue. In other words, these genes deter-
mine a carbohydrate structure through the action of their
encoded proteins acting as enzymes. (Refer to Chapter 6, Mouse Dies Mouse Lives
“The ABO Blood Group System.”) Some proteins may be
shaped to bind to DNA at specific nucleotide sequences in Heat-killed
3 4 S Strain
the chromatin to influence gene expression. These proteins Heat-killed +
act as transcription factors. Through all these mechanisms, S Strain Live R Strain
the genotype of a cell is translated into its phenotype.
Transfusion medicine is tied to three branches of molecu-
lar biology: (1) molecular genetics, because donor-recipient
compatibility depends on the genetic transmission of poly- Mouse Lives Mouse Dies
morphic tissue markers, such as blood groups and human
leukocyte antigens (HLAs); (2) biotechnology, because of Live S Strain
isolated from
the production of recombinant proteins relevant to blood mouse
banking, such as growth factors, erythropoietin, and clotting
factors; and (3) molecular diagnostics, because of the Figure 4–1. Griffith’s transformation experiment. Bacterial cells can be trans-
applications of molecular-based methods in the detection of formed from nonpathogenic to virulent by heat-resistant component of the virulent
strain: (1) Mice injected with encapsulated Streptococcus pneumoniae strain S die.
transfusion-transmitted pathogens. Therefore, blood bankers
The strain is virulent. (2) Mice injected with nonencapsulated Streptococcus pneu-
must learn the basic principles of molecular biology. moniae strain R live. The strain is nonvirulent. (3) Mice injected with heat-killed
Chapter 2 describes in detail the biochemistry of gene S strain live. (4) Mice injected with a mixture of R live strain and heat-killed S strain
replication, transcription, and translation. In this chapter, die, and live S strain can be isolated from them.
6888_Ch04_077-102 29/10/18 4:45 PM Page 79

Chapter 4 Concepts in Molecular Biology 79

pneumoniae bacterium that differed in their infectivity of


mice.2 The virulent smooth (S) strain, which has a smooth 1 1
polysaccharide capsule, kills mice through pneumonia. The
nonvirulent rough (R) strain lacks the immunoprotective 35
S Protein 32
P DNA
outer capsule and is nonlethal to mice because it is easily
recognized and destroyed by the murine immune system.
Injection of S-strain bacteria killed by heat does not cause
any disease. However, simultaneous coinjection of the 2 2
mixture of nonvirulent R strain with heat-killed S strain is
lethal, and virulent S-strain bacteria can be recovered from
35 32
mice infected with this mixture. Some heat-resistant com- S Protein P DNA
ponent (a “transforming principle”) from the heat-killed
S strain is able to transform the living R strain from innocu-
ous to virulent (S).
This experiment has been known as Griffith’s transfor-
mation. It is still the basis for transfer of genetic material
from one organism to another in the laboratory setting. 3 3
Griffith was lucky choosing this particular species to demon-
strate the transformation, because many cell types are very
resistant to this process, which is now facilitated by harsh
conditions such as use of chemicals (detergents), electricity
(electroporation), or liposomes.
But how do we know that this genetic transformation
occurred due to transfer of DNA? Oswald T. Avery and his 4 4
group at the Rockefeller Institute were able to reproduce Grif- 35
S Protein
fith’s transformation in vitro. In 1944, they reported that they
had purified the “transforming principle.”3 By molecular com-
position and weight, it was mainly DNA. Moreover, treatment
with RNA or protein hydrolytic enzymes did not degrade it,
32
but treatment with DNase caused loss of activity. Avery’s group’s P DNA
conclusion that DNA was responsible for the transformation
was not accepted immediately. Some in the scientific commu-
nity suspected that inadequately destroyed proteins that con-
taminated the isolated DNA caused the transformation.
In 1952, Alfred Hershey and Martha Chase reported an
experiment that convinced the scientific community that
genes are constituted by DNA (Fig. 4–2). They infected in Figure 4–2. The Hershey-Chase phage experiment. DNA is the molecule that
carries genetic information: (1) Bacteriophages were labeled with 35S or with 32P;
parallel two bacterial cultures with phages (bacterial viruses) 35S was incorporated into proteins and 32P into DNA. (2) Two Escherichia coli
in which either the protein capsule was labeled with radioac- (E. coli) cultures were infected in parallel with the two different phages. (3) Phages
tive sulfur (35S) or the DNA core was labeled with radioactive were dislodged from bacteria by treatment with a blender. (4) After centrifugation,
phosphorus (32P). After dislodging the phage particles from 35S (proteins) were detected only in the supernatant of the first culture and 32P

the bacteria with a blender, they pelleted the bacteria by cen- (DNA) only in the pellet of the second culture. The phages remained in the super-
natant and the bacteria in the pellet. From the bacterial pellet, a new generation of
trifugation. The phage particles were left in the supernatant.
viruses could be raised.
Following the centrifugation, 35S (protein) was only in the
supernatant and 32P (DNA) was only in the pellet. From this
pellet, a new generation of phages arose. The investigators that suggested a helical structure. Based on these clues, in
concluded that the phage protein removed from the cells 1953, James Watson and Francis Crick published a seminal
by stirring was the empty phage coat, whose mission was finding in which they proposed that the DNA molecule was
to transport the DNA from cell to cell. The DNA was the a double α helix.6 DNA was a helical ladder, the rails of
material of life, containing the phage genetic information which were built from alternating units of deoxyribose and
necessary for the viral reproduction inside the bacteria.4 phosphate. Each rung of the ladder was composed of a pair
of nitrogen-containing nucleotides (a base pair) held to-
Features Relevant to Molecular Techniques gether by hydrogen bonds.
The double helix consisted of two strands of nucleotides
In 1950, Erwin Chargaff of Columbia University reported that ran in opposite directions (they were antiparallel),
results of DNA quantitative analysis that became known as designated by polarity (directionality) labels 3′ and 5′, which
Chargaff’s rules.5 Later, Maurice Wilkins and Rosalind refer to the number assigned by convention to the deoxyri-
Franklin produced x-ray diffraction photographs of DNA bose carbon atom linked to either the hydroxyl or phosphate
6888_Ch04_077-102 29/10/18 4:45 PM Page 80

80 PART I Fundamental Concepts

group (see Chapter 2 and Fig. 2–10). The 3′ and 5′ ends de- and T). Proteins are polymers of 20 different amino acids,
termine the direction of DNA replication and transcription. the sequence of which determines their structure and func-
Consistent with x-ray diffraction, about 10 base pairs were tion. But how could the sequence of nucleotides determine
stacked on top of each other at each turn of a helix. Adenine the sequence of amino acids?
always paired by two hydrogen bonds with thymine; guanine The discovery of the molecular basis of sickle cell anemia
always paired by three hydrogen bonds with cytosine. Due provided the first experimental evidence that the nucleotide
to this complementarity resulting from formation of non- sequence of a DNA molecule indeed determined the amino
covalent hydrogen bonds, the nucleotide alphabet of one-half acid sequence of a protein. Sickle cell anemia is a genetic dis-
of the DNA helix determined the alphabet of the other half. ease inherited according to an autosomal-recessive pattern.
Chemistry and dynamics of hydrogen bonding is pivotal Patients with this disease have two copies of the mutated
in understanding the fundamentals of molecular testing beta globin chain gene located on chromosome 11. Individ-
methods, as many procedures are based on the processes uals with only one mutated copy of the gene show mild or
of DNA strands’ separation (denaturation) and annealing no clinical manifestation and are called carriers. In 1949,
(renaturation). These processes occur at different tempera- Linus Pauling and Vernon Ingram showed that the hemoglo-
tures, depending on the total number of hydrogen bonds in bin of individuals with and without sickle cell anemia had
the molecule, which determines the DNA melting point different mobility in an electric field (electrophoretic mobil-
(Tm). The value of Tm is necessary in calculations used to ity).9 This indicated that hemoglobin from the patients had
choose temperatures for polymerase chain reaction or other a different electric charge; therefore, amino acid composition
hybridization-based assays.7 The DNA melting point has was different from normal hemoglobin. Carriers of the
nothing to do with the conversion of solid state into liquid, disease had both types of hemoglobin.
as the term could imply. Rather, it is the temperature at In 1957, Ingram identified that substituting valine for glu-
which half of all hydrogen bonds in the molecule are tamic acid in the sixth amino acid position from the NH2 end
broken, while the other half are still intact. In other words, of the β-globin chain was the cause of the defective hemo-
the Tm determines how much energy is needed to keep globin.10 Thus, a direct link was established between a gene
the strands of the helix apart. Temperatures above the Tm mutation and the structure of a protein. Concurrent research
promote the separation of the strands, while temperatures showed that proteins were synthesized in the cytoplasm in
below Tm keep the strands together. eukaryotic cells. These studies established that the micro-
The Watson-Crick model and the chemistry of hydrogen somal fraction, which we now call ribosomes, was the major
bonding reveal how DNA could replicate. During replication, cellular component required for protein synthesis.11
the hydrogen bonds break, the strands separate, and each one Because DNA was in the nucleus, separated from the
functions as a template for the synthesis of another comple- cytoplasm by the nuclear membrane, an intermediary molecule
mentary half molecule. In this enzymatic process, two identical had to be responsible for conveying the genetic information
DNA molecules are generated, each containing the original from the nucleus to the cytoplasm. Ribonucleic acid (RNA)
strand and a new complementary strand. Because each was a good candidate for this role. Its structure suggested
daughter double helix contains an “old” strand and a newly that RNA could be produced based on a DNA template, and
synthesized strand, this model of replication is called semicon- RNA was located mainly in the cytoplasm where protein
servative. The mechanism of DNA replication was studied synthesis occurred.
using cellular components to reconstruct, in vitro, the bio- Experimental evidence supporting the role of RNA as the
chemical reactions that occur within the cell. In this way, the intermediary molecule came from the studies on bacterial
whole process was dissected so that molecular testing proce- viruses (bacteriophages). When bacteriophage T4 infects
dures could be developed that mimicked the natural process E. coli, the synthesis of bacterial RNA stops, and the only new
that occurred in vivo. In eukaryotes, DNA polymerase III is RNA synthesized is transcribed from T4 DNA. Using radioac-
able to form a new strand using deoxyribonucleoside triphos- tive labeling, Sidney Brenner, François Jacob, and Matthew
phates (dNTPs) as substrates, double-stranded DNA as tem- Meselson showed that the newly synthesized T4 RNA associ-
plate, and magnesium as an enzyme cofactor. This enzyme can ated with bacterial ribosomes, unlike ribosomal RNA.12 These
add nucleotides onto the hydroxyl group (OH) at the 3′ end RNA molecules, which served as templates for protein synthe-
of an existing nucleic acid fragment. Both strands are synthe- sis, were given the name messenger RNAs (mRNAs).
sized at the same time8 (see Fig. 2–12). But how could nucleotides direct the incorporation of an
The examples of techniques mimicking DNA replication amino acid into a protein? The experimental answer to this
are DNA sequencing and polymerase chain reaction (PCR), question came again from studying cell-free extracts. In these
described later in this chapter. experiments, a third type of RNA molecule, which we now
call transfer RNA (tRNA), was isolated. These were short
Expression of Genetic Information molecules, 70 to 80 nucleotides long, and some displayed
the remarkable characteristic of having amino acids cova-
Genes act by determining the sequence of amino acids and lently attached to their 3′ end. The enzymes responsible for
therefore the structure of proteins. Knowledge of DNA struc- attaching a specific amino acid to a specific tRNA were also
ture revealed that the genetic information must be specified isolated from cell-free extracts and were given the name
by the order of the four nitrogen-containing bases (A, C, G, aminoacyl tRNA synthetases. Sequencing of several tRNAs
6888_Ch04_077-102 29/10/18 4:45 PM Page 81

Chapter 4 Concepts in Molecular Biology 81

showed that they all had a common three-dimensional struc- can be transcribed to its complementary DNA (cDNA) and
ture, but a unique three-nucleotide-long sequence was studied by recombinant DNA techniques. For example,
always present in a loop region. This sequence, which we infectious disease screening assays designed to detect HIV
now call anticodon, provided the specificity for each tRNA, in donors’ blood and other specimens start with the process
carrying its specific amino acid, to align with the correspond- of reverse transcription because HIV viruses are retro-
ing codon (triplet) in the mRNA.8 (Refer to Chapter 2 and viruses and their genetic material consists of RNA. (Refer
Figs. 2–15 through 2–18 for more information on the to Chapter 14, “Transfusion-Transmitted Diseases.”) The
process of translation.) cDNA synthesized during reverse transcription is subse-
The next question was which triplet of nucleotides of the quently amplified in polymerase chain reaction, which
mRNA corresponded to which amino acid; in other words, allows for creation of enough copies of genetic material to
it was time to decipher the genetic code. By 1966, Marshall analyze (refer to “Reverse Transcriptase PCR” section in
Nirenberg and Har Gobind Khorana had cracked the code. this chapter).
Nirenberg used cell-free extracts containing ribosomes, Scientists have added further disclaimers to the central
amino acids, tRNAs, and aminoacyl tRNA synthetases, and dogma:
added synthetic mRNA polymer containing only uracil
• Genes are not “fixed.” Some of them do not have a desig-
(-UUUUUU-). He was able to produce in vitro a polymer
nated chromosomal locus and function as transposable
peptide containing only the amino acid phenylalanine. In
genetic elements in the eukaryotic genome. Another mech-
this way, he showed that UUU was the codon for phenylala-
anism of physical rearrangement is in vivo DNA recombi-
nine.13 During the next few years, all possible mRNA com-
nation. The examples are the immunoglobulin chain and
binations were tried. The 61 different triplets were assigned
T-cell receptor (TCR) gene rearrangements.
to their corresponding amino acids, and three triplets were
• DNA sequences and protein amino acid sequences do not
found to code for a stop signal (refer to Fig. 2–11). The
exactly correspond to one another. In many organisms, in-
genetic code is called degenerate because one amino acid
cluding humans, coding sequences (exons) are interrupted
may be encoded by more than one nucleotide triplet. This
by noncoding sequences (introns) that are excised from the
serves as a mechanism that protects the code from devastat-
immature RNA shortly after transcription. This introduces
ing effects of mutations occurring in the third or second base
the possibility of alternative splicing to create different pro-
of the triplet. In most cases, the mutation results in amino
teins from the same gene. Also, different reading frames can
acid change only when it affects the first base of the triplet.
generate different mRNAs and therefore different proteins.
Viruses frequently use this last mechanism. In vitro reverse
The Central Dogma of Molecular transcription procedures will result in cDNA that corre-
Biology, Expanded sponds only to sequences originally present within exons.
Hence, cDNA derived from very long genes is a much
Deciphering the genetic code confirmed the central dogma
shorter representation of the gene (Fig. 4–3). Scientists have
of molecular biology. The genetic material is DNA. DNA is
discovered further roles for RNA. Some RNA molecules
self-replicating and is transcribed into mRNA, which in turn
display catalytic activity; by RNA interference, small RNA
serves as a template for the synthesis of proteins. Although
molecules help to regulate gene expression.16
the central dogma remains true, the knowledge acquired in
the following years has refined and enlarged it, a process that Recombinant DNA
continues now and into the future.
A different method of information flow in biological sys- The classic experiments in molecular biology, which we have
tems was discovered. A particular group of animal RNA described, used simple and rapidly replicating organisms
viruses called tumor viruses can cause cancer in infected (bacteria and viruses) as study models. However, the
animals. In the early 1960s, Howard Temin discovered that genomes of eukaryotes are much more complicated, and
replication of these viruses required DNA synthesis in the scientists had to explore new ways to isolate and study indi-
host cells. He formulated the hypothesis that RNA tumor vidual genes of these organisms. The big breakthrough came
viruses, subsequently called retroviruses, replicated via the with the development of recombinant DNA technology. A
synthesis of a DNA intermediate or provirus. In 1970, Temin fragment of DNA from one organism can be cut and pasted
and David Baltimore showed independently that these into a carrier DNA molecule or vector (e.g., a plasmid or bac-
viruses contain an enzyme that catalyzes the synthesis of teriophage). The new DNA molecule, which is a “recombi-
DNA using RNA as a template.14,15 nant” of the original DNA with the vector DNA, can be
The presence of viral DNA in replicating host cells was introduced using various techniques into another, usually
also demonstrated. The synthesis of DNA from RNA, now simpler, host organism. Because the genetic code is almost
called reverse transcription, was thus definitely established universal, the host organism treats the gene as its own and
as a mode of biological information transfer. In vitro reverse replicates this “transgene” along with its own DNA. This
transcription is an important tool in molecular biology technique is called molecular cloning. The ABO gene was
techniques. Natural or recombinant enzymes with reverse cloned by Yamamoto’s group in 199017 (Table 4–1).
transcriptase activity can be used to generate DNA copies Cloning is the reproduction of daughter cells from one
of any RNA molecule. Using reverse transcriptase, mRNA single cell by fission or mitotic division, giving rise to a
6888_Ch04_077-102 29/10/18 4:45 PM Page 82

82 PART I Fundamental Concepts

Promoter Exon 1 Intron 1 Exon 2 Intron 2 Exon 3


5′ 3′
3′ 5′
DNA

Transcription

Pre-mRNA 5′ 3′ Figure 4–3. In Eukaryotes, the coding sequences (exons) are


interrupted by noncoding sequences (introns). Newly transcribed RNA
Processing (pre-mRNA) is considered heterogeneous RNA (hnRNA) and loses the
introns in the process of splicing. The mature mRNA is modified
mRNA 7 Me G AAAAA by polyadenylation of the 3’ end and addition of 7-methylguanine
5′ 3′ “cap” at the 5’ end, and it contains only the exons. In vitro reverse
transcription procedures will result in cDNA that corresponds only to
Reverse transcription
sequences originally present within exons. Hence, cDNA is a shorter
representation of the gene. (Modified from Buckingham, L,: Molecular
cDNA Diagnostics. Fundamentals, Methods and Clinical Applications.
F.A. Davis Company, Philadelphia, PA, 2012.)

Table 4–1 Selected Major Events Leading to Establishment of Molecular Immunohematology


Year Major Event
1924 Bernstein, applying the Hardy-Weinberg principle, proposes that the ABO antigens are coded by one gene.

1944 Avery, MacLeod, and McCarty prove DNA as the carrier of inheritance (following Griffith’s transformation in 1928).

1953 Watson and Crick decipher DNA structure based on Chargaff’s rules (A + G = T + C and T = A and C = G) and x-ray studies by R. Franklin
(1962 Nobel Prize with Wilkins).

1970 Smith and Wilcox isolate the first restriction enzyme from H. influenzae (RFLP method developed in 1978).
Temin and Baltimore discover enzyme reverse transcriptase.

1975 Maxam and Gilbert invent the first DNA sequencing method. Sanger improves it in 1977.

1977–1978 First demonstration of a molecular basis for blood group polymorphisms: two aa differences in glycophorin A identified as the basis for
M and N blood groups (several researchers).

1981 First demonstration of a molecular basis for rare blood group polymorphisms: basis for Mg and Mc rare blood group variants identified
(H. Furthmayr and O. O. Blumenfeld groups).

1986 Kary Mullis publishes polymerase chain reaction (PCR) to amplify DNA fragments using thermostable Taq enzyme.

1986 Glycophorin A and glycophorin C are cloned.

1987 CD55 (DAF), responsible for the Cromer system, and glycophorin B are cloned.

1990 F. Yamamoto and colleagues clone the ABO gene.


R. D. Larsen and colleagues clone the FUT1 gene (H/h system).

1991 RHD and RHAG genes of the Rh system and KEL gene are cloned.

1992 Higuchi and others at Roche Molecular Systems and Chiron demonstrate real-time PCR (simultaneous amplification and detection).

1994 Genes responsible for the Lutheran system, Colton blood group antigens, and Kidd (JK) system are cloned.

1996 S. Iwamoto and colleagues identify promoter mutations as responsible for the Duffy a-b phenotype.

1999 The Blood Group Antigen Gene Mutation Database (BGMUT) is set up by O. Bloomenfeld and S. K. Patnaik. Moved in 2006 to National
Center for Biotechnology Information

2000 Complete sequence of the human genome is published.

2004 Consortium for Blood Group Antigens established with the purpose to “provide education for laboratories involved in DNA/RNA testing”;
initiative by NYBC (M. Reid).
First International Workshop of Molecular Blood Group Genotyping (ISBT initiative).

2005 Microarray-based blood group typing is described by multiple researchers (Transfusion, 45).
6888_Ch04_077-102 29/10/18 4:45 PM Page 83

Chapter 4 Concepts in Molecular Biology 83

Table 4–1 Selected Major Events Leading to Establishment of Molecular


Immunohematology—cont’d
Year Major Event
2006 Ortho sponsors Rh molecular workshop in Heidelberg.
FDA workshop Molecular Methods in Immunohematology (Transfusion, July 2007 Suppl). AABB Annual Meeting—BioArray
Solutions Technology
2007 AABB News publishes “The Omics Revolution.”
2014 First FDA-approved molecular platform for predicted erythrocyte antigens.56

Modified from: dbRBC, Blood Group Antigen Gene Mutation Database.55

population of genetically identical clones. Successive divi- controlled and predictable fashion and “paste” it into another
sions of the host cell create a population of clones containing fragment using other enzymes (ligases). Restriction endonu-
the DNA fragment of interest. The gene or genes of interest cleases are isolated from bacteria. From the early 1950s, it
can be studied using the techniques available for the host was observed that certain strains of E. coli were resistant to
organism. These techniques have allowed detailed molecular infection by various bacteriophages. This form of primitive
studies of the structure and function of eukaryotic genes and bacterial immunity was called restriction because the host was
genomes.18 able to restrict the growth and replication of the virus. The
resistant bacteria had enzymes that selectively recognized and
The Coding Sequence of a Gene destroyed foreign DNA molecules by cutting it into pieces
(endonuclease activity). At the same time, these enzymes
In humans and most other eukaryotes, the coding part of a modified (by methylation) the host’s chromosomal DNA,
gene consists of exons, which in the genomic DNA alternate protecting it from self-destruction. Numerous restriction en-
with noncoding introns. Newly transcribed RNA (pre-mRNA) donucleases have been isolated from different bacteria. These
is considered heterogeneous RNA (hnRNA) and loses the in- enzymes have been classified as type I, II, and III according
trons in the process of splicing. The mature mRNA contains to their mechanism of action. Only the type II enzymes are
only the exons, the coding sequence of nucleotides that directs relevant for our description of molecular cloning.
the process of translation into the amino acid sequence of the In 1970, Hamilton Smith and Kent Wilcox at Johns
corresponding protein.19 Hopkins University isolated the first type II restriction en-
Both the genomic DNA and the coding sequence of a gene donuclease. Type II restriction enzymes are extremely useful
can be cloned. First, the mRNA is reverse transcribed in vitro for the molecular biologist; they cut DNA at a precise posi-
to its cDNA, using either synthetic or retroviral enzyme re- tion within its recognition sequence and have no modifying
verse transcriptase. Because eukaryotic mRNAs end with a activity.20 More than 200 restriction endonucleases, covering
poly-A tail, a short synthetic oligo-dT nucleotide can be used more than 100 different recognition sites, are available com-
as a universal primer starting the cDNA synthesis (this mercially. The name given to each enzyme reflects its origin.
process may also be achieved using the random hexamers For example, Smith and Wilcox’s endonuclease was called
or sequence-specific primers, depending on the applica- HindII because it was obtained from Haemophilus influenzae
tion). The final product is a hybrid double strand of mRNA strain Rd. The number II indicates that it was the second
and cDNA. The mRNA is degraded, and a DNA strand com- endonuclease to be identified in these bacteria. Restriction
plementary to the cDNA is synthesized using DNA poly- endonucleases usually recognize sequences 4 to 8 nucle-
merase. The cDNA, which has the information for the coding otides in length. The recognition sequences are palindromic;
gene sequence, can be subjected to the same techniques as that is, they read the same in the 5′ to 3′ direction on both
genomic DNA.18 strands of the double helix. Some enzymes cut both strands
in the middle of the target sequence, generating blunt ends.
Tools for DNA Cloning Some enzymes cut both strands off the center, generating
staggered ends. For example enzyme EcoRI, isolated from
The following sections describe the essential tools for molec-
E. coli, recognizes this sequence:
ular cloning: restriction endonucleases, gel electrophoresis,
vectors, and host cells. An example of gene cloning follows. 5′GAATTC3′
3′CTTAAG5′
Restriction Endonucleases
EcoRI cuts after the first G in both strands, generating
The first breakthrough in the production of recombinant
fragments with the following ends:
DNA came with the discovery and isolation of restriction
endonucleases. These enzymes cleave DNA at specific 5′G AATTC3′
sequences and allow scientists to “cut” a DNA fragment in a 3′CTTAA G5′
6888_Ch04_077-102 29/10/18 4:45 PM Page 84

84 PART I Fundamental Concepts

The single-strand overhanging ends are complementary Table 4–2 Examples of Type II Restriction
to any end cut by the same enzyme. These sticky, or cohe-
Endonucleases
sive, ends are extremely useful for “cutting and pasting”
DNA from different origins, thus creating recombinant DNA. Enzyme Recognition Sequence
Examples of other restriction endonucleases and correspond- AluI AG↓CT
ing recognition sequences are listed in Table 4–2.
Finding out the sequences in a given DNA recognized by AosI TGC↓GCA
these enzymes, and establishing the distance between such ApyI CC↓(A,T)GG
sites, is called restriction enzyme mapping. In short
sequences (about a few thousand base pairs [bp] long), such AsuI G↓GNCC
as in plasmids, which are bacterial extrachromosomal DNA, AsuII TT↓CGAA
the mapping is simple because there are only a few sites rec-
ognized by each enzyme. However, restriction maps of long AvrII C↓CTAGG
eukaryotic genomes are quite complex. Restriction fragment BalI TGG↓CCA
sizes may be altered by changes in or between enzyme
recognition sites. The restriction site may disappear upon BamHI G↓GATCC
various mutations, and a new enzyme recognition site BclI T↓GATCA
may appear.
The discovery of these naturally and pathologically BglII A↓GATCT
occurring changes contributed to the establishment of the BstEII G↓GTNACC
restriction fragment length polymorphism (RFLP) method.
The analysis of unique RFLP patterns of various loci proved BstNI CC↓(A,T)GG
useful in crime-victim identification and in kinship studies. BstXI CCANNNNN↓NTGG
Blood bank applications of this initially laborious method
were expanded greatly after the discovery of polymerase ClaI AT↓CGAT
chain reaction. For example, the PCR-RFLP method was DdeI C↓TNAG
used to discriminate between RHD+ and RHD genotypes,
based on the different number of PstI enzyme restriction EcoRI G↓AATTC
sites in the region flanking the RHD gene (so-called EcoRII ↓CC(A,T)GG
Rhesus box).21
Fnu4HI GC↓NGC

Gel Electrophoresis FnuDII CG↓CG


The restriction patterns mentioned in the previous section HaeI (A,T)GG↓CC(T,A)
may be examined by gel electrophoresis (Fig. 4–4), another
technique fundamental for DNA cloning. It allows not only HaeII PuGCGC↓Py
for visual analysis of DNA fragment length but also for its HaeIII GG↓CC
isolation and purification for further applications.18
The word electrophoresis means “to carry with electric- HhaI GCG↓C
ity.” Agarose gel electrophoresis is a simple and rapid method HincII GTPy↓PuAC
of separating DNA fragments. The DNA sample is intro-
duced into a well in a gel immersed in buffer and is exposed HindII GTPy↓PuAC
to electric current. The DNA molecules run at different HindIII A↓AGCTT
speeds according to their size. After staining with a fluores-
cent dye, bands of DNA of specific length can be seen under HinfI G↓ANTC
ultraviolet (UV) light and isolated from the gel. In solution, HpaI GTT↓AAC
at a neutral pH, DNA is negatively charged because of its
phosphate backbone. When placed in an electric field, DNA HpaII C↓CGG
molecules are attracted toward the positive pole (anode) and MboI ↓GATC
repelled from the negative pole (cathode).
To make a gel, molten agarose (a seaweed extract) is MstI TGC↓GCA
poured into a mold where a plastic comb is suspended. As NotI GC↓GGCCGC
it cools, the agarose hardens to form a porous gel slab with
preformed wells. The gel has a consistency similar to gelatin PstI CTGCA↓G
and is placed in a tank full of buffer and with electrodes at RsaI GT↓AC
opposing ends. The samples containing the DNA fragments
are pipetted into the wells after mixing them with a loading SacI GAGCT↓C
solution of high density, such as sucrose or glycerol. The SacII CCGC↓GG
dense solution helps the DNA sink when loaded into the
6888_Ch04_077-102 29/10/18 4:45 PM Page 85

Chapter 4 Concepts in Molecular Biology 85

Cathode Anode
(–) (+)

D
N
EB
UV A
GEL
Figure 4–4. Agarose gel electrophoresis. L
Negatively charged DNA molecules migrate to- E
ward the positive pole. The agarose gel acts as a
N
molecular sieve: The shorter the DNA molecule,
Direction of DNA Migration G
the longer the distance of migration. DNA bands
are detected by exposure to ultraviolet light (UV) T
after staining with the fluorescent dye ethidium H
bromide (EB) or SYBR Green. DNA Length

wells and allows for monitoring of the progress of elec- from the chromosome. The plasmid vectors used in molecular
trophoresis due to presence of bromophenol blue dye cloning are “designer plasmids,” modified to make perfect
(a tracer). When electric current (80 to 120 V) is applied to recombinant DNA carriers. A vast selection of plasmids are
electrodes on the tank, the DNA fragments migrate toward commercially available that are useful for different purposes,
the anode. The porous gel matrix acts as a sieve through such as DNA sequencing, protein expression in bacteria, and
which larger molecules move with more difficulty than protein expression in mammalian cells.
smaller ones. The distance run by a DNA fragment is All plasmids contain an origin of replication. Plasmid
inversely proportional to its molecular weight and therefore vectors can be under “stringent” control of replication, in
to its length or number of nucleotides. which case they replicate only once per cell division. By con-
DNA bands can be detected in the gel upon staining with trast, the “relaxed” plasmids replicate autonomously and can
a fluorescent dye, such as ethidium bromide (EB) or SYBR grow as hundreds of copies per cell. Relaxed plasmids are
Green. The gel may be soaked in a diluted fluorescent dye used to amplify large amounts of cloned DNA. Naturally
solution, or alternatively, the electrophoresis is run with a occurring plasmids contribute significantly to bacterial
gel and buffer containing the dye. EB is a planar molecule, genetic diversity by encoding functions, such as resistance
which intercalates between the stacked nucleotides of the to certain antibiotics, which provides the bacterium with
DNA helix. This feature makes it a mutagenic factor, which a competitive advantage over antibiotic-sensitive strains.
calls for cautious handling of gels, equipment exposed to this Plasmids used as vectors also contain genes that encode for
agent, and waste. antibiotic resistance. When bacteria grow in a culture
SYBR Green dye isn’t mutagenic, and many facilities have medium with the given antibiotic, only host cells containing
adopted this dye in their staining procedures to assure labo- the plasmid will survive. Thus, a pure population of bacteria
ratory safety.22 When the gel is exposed to ultraviolet light can be obtained. Examples of antibiotics used for bacterial
of 300 nm, the stained DNA fragments are seen as fluores- cloning are listed in Box 4–1.
cent orange (EB) or green (SYBR) bands and can be docu- Cloning a DNA fragment requires inserting it into the
mented by photography. Each band of DNA is formed by plasmid vector. Plasmids are designed to contain one or
millions of DNA molecules of equal length, which is estab- more cloning sites, also called polylinkers, which are a
lished by comparison with standard bands of size marker series of recognition sequences for different restriction
reagents. Gel electrophoresis, in low-melting-point agarose, endonucleases. Most commonly, these are recognition sites
is used in molecular cloning to isolate and purify the vector for enzymes that cut both strands off the center, which gen-
and insert, which will be used to form a recombinant erates cohesive (“sticky”) ends. Otherwise, the procedure is
DNA vector. called “blunt end cloning.” The circular DNA of the plasmid

Vectors
A vector is a DNA molecule of known nucleotide sequence BOX 4–1
that is used to carry a foreign DNA fragment into a host
organism. Vectors can be used to produce large quantities Antibiotics Used in Bacterial Cloning
of a target DNA fragment. They can also be used to get a • Ampicillin
foreign gene expressed in a host cell to study the function of • Carbenicillin
the gene or to manufacture large quantities of the encoded • Chloramphenicol
protein product.18 • Hydromycin B
• Kanamycin
Plasmids. Plasmids are the simplest kind of vectors. They are • Tetracycline
bacterial, circular genetic elements that replicate independently
6888_Ch04_077-102 29/10/18 4:45 PM Page 86

86 PART I Fundamental Concepts

is cut (or linearized) with a chosen restriction enzyme. The libraries. Cosmids have a small region of bacteriophage λ
DNA fragment to be inserted is cut with the same enzyme. necessary for packaging viral DNA into λ particles. The
The sticky ends of the linearized plasmid can form hydrogen linear genomic DNA fragment is inserted into the vector and
bonds with the complementary nucleotides in the overhang packaged into bacteriophage particles. After infecting the
of the DNA fragment to be inserted (hence called an insert). E. coli cells, the vector circularizes into a large plasmid con-
Both fragments are run in a preparative, low-melting-point taining the DNA fragment of interest.
agarose gel. The bands corresponding to the expected size Bacterial artificial chromosomes (BACs) are circular
of the linearized plasmid and insert are cut out, and the pu- DNA molecules that contain a unit of replication, or replicon,
rified DNA fragments are isolated. Both fragments are pasted capable of carrying fragments as large as a quarter of the
together with DNA ligase, which forms phosphodiester E. coli chromosome. BACs can maintain DNA inserts of up
bonds between adjacent nucleotides (Fig. 4–5). to 350 kb. A typical BAC library contains inserts of 120 kb.
Typical plasmid vectors consist of DNA 2 to 4 kb (kilo-
bases) long and can accommodate inserts up to 15 kb long. Host Cells
Several other vectors are available for cloning fragments of The recombinant DNA molecule, which was produced in
DNA of different sizes. vitro, must then be introduced into a living cell in order to
reveal if it is functional. The typical host organisms are
Other Vectors. Lambda (λ) vectors contain the part of the bac- various strains of E. coli. As we have previously described
teriophage’s genome necessary for lytic replication in E. coli using the Griffith’s experiment, the uptake and expression
and one or more restriction endonuclease sites for insertion of foreign DNA by a cell is called transformation.2 However,
of the DNA fragment of interest. They can accommodate for- natural transformation that was observed due to DNA uptake
eign DNA 5 to 14 kb long. The recombinant DNA is “pack- by the rough (nonencapsulated) strain of Streptococcus
aged” into viral particles and used to infect E. coli. pneumoniae is a rare event. The efficiency of the bacterial
Cosmids are vectors that can accept DNA 28 to 45 kb transformation can be increased by chemical or electrical
long. They are useful for producing large-insert genomic (electroporation) methods that modify the cellular mem-
brane.18 The bacteria treated by these methods are called
competent.
Because E. coli is a normal inhabitant of the human colon,
1 EcoRI Digestion ATTC
GAT AAG
it grows best in vitro at 37°C in a culture medium containing
CT nutrients similar to those available in the human digestive
GAATTC Target GAATTC
Circular tract, including carbohydrates, amino acids, nucleotide phos-
CTTAAG Gene CTTAAG
Amp

Plasmid
phates, salts, and vitamins. To isolate individual colonies,
r

Or
i cells are spread on the surface of agar plates containing an
antibiotic used as a selectable marker. A suspension of the
5'AATT 3' 5'AATT Linearized 3' cells is diluted enough so that each cell will give rise to an
Insert individual colony. Plates are incubated at 37°C. After 12 to
3' TTAA5' 3' Plasmid TTAA5'
24 hours (at the maximum rate, cells divide every 22 min-
2 Agarose Gel Electrophoresis utes), visible colonies of identical daughter cells appear. In-
(–) dividual colonies are picked and further propagated in liquid
culture medium with antibiotic. The incubation proceeds
Targe
t with continuous shaking to maintain the cells in suspension
T
Gene AATAA and to promote aeration and production of cell mass that
TT
may be used for efficient isolation of vector DNA containing
TT ATT
AA

(+)
the insert.
A

Recombinant Plasmid Isolation


3 Elution of insert and Plasmid
Linearized Plasmid Plasmids can be easily separated from the host’s chromoso-
mal DNA because of their small size and circular structure.
Am

A rapid method for making a small preparation of purified


p

ri

4 Ligation
r

plasmid from 1 to 5 mL of culture is called miniprep. The


Insert + Linearized
Plasmid
overnight culture is centrifuged, and the pellet containing
the bacteria is treated with a solution that includes sodium
dodecyl sulfate (SDS) and sodium hydroxide. SDS is an ionic
detergent that lyses the cell membrane, releasing the cell
Figure 4–5. Gene cloning: part 1. (1) The target DNA and the vector plasmid contents. Sodium hydroxide denatures DNA into single
are digested with EcoRI. (2) The insert and linearized plasmid are submitted to
strands. Treating the solution with potassium acetate and
agarose gel electrophoresis. (3) The bands containing the insert and linearized
plasmid are cut from the gel and the DNA fragments eluted. (4) Insert and linearized acetic acid (“salting out”) causes an insoluble precipitate of
plasmid are ligated together and a recombinant plasmid containing the target gene detergent, phospholipids, and proteins and neutralizes the
is obtained. sodium hydroxide.
6888_Ch04_077-102 29/10/18 4:45 PM Page 87

Chapter 4 Concepts in Molecular Biology 87

At neutral pH, the long strands of chromosomal DNA 1 Transform E. coli with recombinant plasmid
renature only partially and become trapped in the precipi- Bacterial
Chromosome
tate. The plasmid DNA renatures completely and remains in
solution. After centrifugation, the pellet is discarded. Ethanol
or isopropanol is added to the supernatant to precipitate the Recombinant
plasmid DNA out of the solution. After centrifugation, the Plasmid

Am
DNA pellet is washed with 70% ethanol and dissolved in

ri
p

O
r
water or buffer. RNase is used to destroy the RNA that
coprecipitates with the plasmid. A miniprep provides enough
material for some downstream applications such as sequenc- 2 Plate bacteria on LB agar
medium with ampicillin
ing or subcloning into a different plasmid vector. However,
it can be easily scaled up to a “midiprep” or “maxiprep”
when more plasmid DNA is needed.
3 Isolate and grow bacterial colony
DNA Cloning: An Example in liquid medium with ampicillin
To summarize the process of DNA cloning, we will describe
a hypothetical example in which a gene of interest, or target
gene, is obtained from genomic DNA for sequencing pur-
poses. In Figure 4–5, the target gene, flanked by two EcoRI
restriction sites, is cut from genomic DNA and inserted
into a plasmid vector. The plasmid contains an origin of
replication (Ori) so it can be copied by the bacteria’s DNA
replication machinery. It also contains a gene that provides
4 Lyse bacteria and
resistance to the antibiotic ampicillin (Ampr); therefore, the isolate recombinant
bacteria that have the plasmid will be able to grow in culture plasmids
medium with the antibiotic, whereas the bacteria that did
not get the plasmid will die.
In Figure 4–6, competent E. coli are transformed with the Miniprep
plasmid and plated at a very low dilution in a Petri dish in
solid LB medium with ampicillin. Only the bacteria contain- Figure 4–6. Gene cloning: part 2. (1) The recombinant plasmid is used to
ing the plasmid are able to grow and form colonies, which transform competent E. coli. (2) E. coli bacteria are plated onto solid agar medium
are visible after overnight incubation at 37°C. A colony containing ampicillin. Only bacteria containing the plasmid survive. Isolated colonies,
is isolated and grown in liquid LB medium with ampicillin. each from a single cell, grow after overnight incubation at 37°C. (3) A colony
is picked and cultured overnight in 1 to 5 mL of liquid LB medium containing
The plasmid replicates independently from the bacterial
ampicillin. (4) The recombinant plasmid is isolated from the bacterial culture
chromosome, and several plasmids are formed inside each (“miniprep”).
bacterium. From this cell suspension, the recombinant plas-
mid is isolated. The resulting miniprep contains abundant
copies of the gene of interest (target gene) that can be used DNA was isolated from cells and partially digested with re-
as a template for obtaining the sequence of the target gene. striction enzymes, obtaining large fragments of around 100
Currently, all genes coding for the 30 recognized human to 200 kb. Inserts of these cloned BACs were subcloned as
blood group systems have been cloned.23 smaller fragments into phage or plasmid vectors, which
were used for sequencing.24 Complementary DNA libraries
Recombinant DNA Libraries can be used to determine gene-coding sequences from
A way of isolating single genes is the production of recom- which the amino acid sequence of the encoded protein can
binant DNA libraries. Instead of trying to fish one gene out be deduced.
of a mass of genomic DNA or cDNA, each gene is physically Oligonucleotides can be synthesized on the basis of par-
separated and introduced into a vector. The target gene is tial amino acid sequence of the protein of interest and used
selected by screening each of the individual pieces with a to screen a recombinant cDNA library. The clones that con-
specific probe. Recombinant DNA libraries are collections tain the gene encoding the target protein are isolated and
of clones that contain all genomic or mRNA sequences of a sequenced. The complete protein sequence can thus be
particular cell type. Clones containing a specific gene are obtained by translating the mRNA codons into amino acids.
identified by hybridization with a labeled probe, such as a This approach was used in 1990 by Fumi-Chiro Yamamoto
cDNA or genomic clone or a PCR product (see the “Detec- and his colleagues to isolate the gene that codes for the group
tion of Nucleic Acids and Proteins” section). A transferase.17 Another approach for isolating a gene on the
As seen in the section on vectors, different vectors are basis of the protein that it encodes is using antibodies spe-
useful for isolating DNA fragments of different sizes. For cific against the target protein to screen expression libraries.
example, the Human Genome Project used BAC vectors to In this case, the cDNA library is generated in an expression
produce recombinant human genomic libraries. Genomic vector that drives protein synthesis in E. coli.
6888_Ch04_077-102 29/10/18 4:45 PM Page 88

88 PART I Fundamental Concepts

Expression of Cloned Genes: Recombinant treatments seek to enhance the host antitumor responses by
Proteins in Clinical Use overexpression of cytokines or by genetic alteration of the
tumor cell.
The cost and effort necessary to isolate proteins with ther- One approach is the use of viral vectors, such as retro-
apeutic functions from their original source make their viruses, adenoviruses, herpesviruses, and lentiviruses. Most
use impractical. Also, the risk of viral contamination is a human clinical trials to date have used retroviral vectors.
fundamental consideration in any therapeutic product that However, the use of these vectors carries serious potential
is purified from mammalian cells, in particular from human risks that must be weighed against the severity of the under-
tissues. For these reasons, the use of engineered in vitro, lying illness. The exogenous gene may cause disease if over-
recombinant proteins is a major improvement in the treat- expressed or expressed in certain cell types. Contaminants
ment of several diseases. Large amounts of recombinant may be introduced during vector manufacture. Also of
proteins can be obtained for biochemical studies or for ther- concern is the potential for recombination of gene therapy
apeutic use by molecular cloning of genes that code for vectors with human endogenous sequences. The search for
them. The cloned gene is inserted into an expression vector, better vectors is underway in animal models and holds
usually a plasmid, which directs the production of large promise for a safer and more effective gene therapy.
amounts of the protein in either bacteria or eukaryotic cells.
A great variety of expression vectors are available for recom- The Polymerase Chain Reaction
binant protein expression in E. coli, yeast, insect cells, and
mammalian cells. An alternative to cloning for isolating large amounts of a sin-
The list of recombinant proteins used as therapeutic gle DNA fragment or gene is the polymerase chain reaction
agents increases constantly. Interferon-α used to treat hairy (PCR), which was developed by Kary Mullis at Cetus
cell leukemia and hepatitis C and B, recombinant hepatitis Corporation in 1985.26 As opposed to cloning, which is
B vaccine, recombinant antihemophiliac factor, and recom- performed in a living cell, PCR is carried out completely in
binant coagulation factor IX are some examples of recombi- vitro. Provided that some sequence of the DNA molecule is
nant proteins useful for transfusion medicine. Granulocyte known, PCR can be used to amplify a defined segment of
colony-stimulating factor (GCSF) is used to increase the DNA several million times.
production of hematopoietic stem cells (HSC) for HSC trans- In PCR, DNA (or cDNA) is replicated in the test tube or
plantation (HSCT). Epoetin alfa is used to treat anemia on a microtiter-like plate, replacing the steps that normally
caused by chronic renal failure (Box 4–2). occur within a cell by adding synthetic or recombinant
reagents and by cyclic changes in the reaction temperature.
Gene Therapy The role of the enzyme primase, which in vivo generates the
Gene therapy is the introduction of new genetic material primers to which DNA polymerase attaches the successive
into the cells of an organism for therapeutic purposes. In nucleotides, is replaced by synthetic oligonucleotides called
the United States, only somatic gene therapy is allowed in forward and reverse primers. These primers are usually 15
humans—that is, the treated cells are somatic and not to 20 nucleotides long and flank the target region to be
germline cells. The obvious clinical use of gene therapy is amplified. In other words, the primers are designed to anneal
the treatment of inherited diseases. In the first clinical trial to complementary DNA sequences at the 3′ end of each
of that kind, the gene coding for adenosine deaminase was strand of the DNA fragment (Fig. 4–7).
introduced into lymphocytes of children suffering from The product obtained (the amplicon) will have the
severe combined immunodeficiency disease.25 However, sequence of the template bracketed by the primers. The two
several current clinical trials are focused on oncology. These primers are mixed with a DNA sample containing the target
sequence; thermostable DNA polymerase; the four deoxyri-
bonucleoside triphosphates (dNTPs); and magnesium,
which acts as enzyme cofactor. Thermus aquaticus, a bac-
BOX 4–2 terium that grows in hot springs, was the original source of
Recombinant Proteins Used in Transfusion Medicine heat-resistant polymerase, consequently named Taq.
Currently, numerous recombinant enzymes are on the
• Factor VIII market. The thermostability of the enzyme is of utmost im-
• Factor IX portance because the first step of the PCR is heat denatura-
• Factor VII tion. Applying high temperatures breaks the hydrogen bonds
• Factor XIII between the complementary nucleotides and plays the role
• Thrombopoietin of the helicases and gyrases that unwind the DNA so that the
• G-CSF DNA polymerase complex can reach the template. At 94°C,
• GM-CSF the double helix is denatured into single strands. In the sec-
• Epoetin α ond step, annealing of the primers to the target sequences
• Interferon-α flanking the fragment of interest is achieved by cooling to a
temperature of 50° to 60°C, which promotes renaturation
G-CSF = granulocyte colony stimulating factor; GM-CSF = granulocyte
macrophage colony-stimulating factor (reestablishment of hydrogen bonds). The exact optimum
temperature of annealing is determined by the number of
6888_Ch04_077-102 29/10/18 4:45 PM Page 89

Chapter 4 Concepts in Molecular Biology 89

Region under
Sense (plus) strand investigation
Template DNA
5′ 3′
G A A T CG T CG A GC T GC T A GC T T T G T T CG A
Forward GA A A C A A
3′ 5′
Figure 4–7. The FORWARD primer always anneals to the minus Primer Primer
strand (also called antisense strand). It is complementary to the 3’ end 5′ 3′ Reverse
of the minus strand (and identical to the 5’ end of the plus strand). A T CG T C
The REVERSE primer always hybridizes with the plus strand (also called C T T A GC A GC T CG A CG A T CG A A A C A A GC T
3′ 5′
the sense strand). It is complementary to the 3’ end of the plus strand
and is identical to the 5’ end of the minus strand. (Modified from Template DNA
Buckingham, L: Molecular Diagnostics. Fundamentals, Methods and Antisense (minus) strand PCR
Clinical Applications. F.A. Davis Company. Philadelphia, PA, 2012.)

G and C residues in the primers: The more GC, the more end of the primer and ends wherever the Taq or other ther-
hydrogen bonds to break and the higher the melting point mostable polymerase ceases to function. After the second
Tm of the molecule. Typically, the optimum annealing cycle, DNA is synthesized, using the newly copied strands as
temperature is 5°C below the Tm. templates. In this case, synthesis stops when it reaches the end
The third step, primer extension by DNA polymerase, is of the molecule defined by the primer. By the end of the third
done at 72°C. This is the optimal temperature for the poly- cycle, a new blunt-ended double-stranded product is formed,
merase, which incorporates nucleotides to the 3′ OH end of with its 5′ and 3′ ends precisely coinciding with the primers.
the primers using the target DNA as template. The cycles of These blunt-ended fragments accumulate exponentially
DNA synthesis, which consist of denaturation, annealing, and during subsequent rounds of amplification, while the orig-
extension, are repeated simply by repeating the cycle of inal DNA that was outside the region flanked by the
temperatures. The reaction is carried out in a programmable primers isn’t amplified. Thus, the majority of fragments in
heating and cooling machine called a thermocycler. The the later PCR cycles have both ends defined. A single DNA
instrument contains a heating block built from materials char- molecule amplified through 30 cycles of replication would
acterized by high thermal conductivity (e.g., silver) that allow theoretically yield 230 (approximately 1 billion) progeny
for rapid changes of temperatures and shortens time between molecules. In reality, the exponential phase of product
the steps of each cycle. Figure 4–8 shows a schematic PCR accumulation during PCR is not indefinite; therefore, the
reaction. In each cycle, an original template strand is copied theoretical number of product molecules is not reached.
to generate a complementary strand, which begins at the 5′ After a certain number of cycles, the accumulation of

Figure 4–8. Polymerase chain reaction DNA Template + Forward Primer + Reverse Primer + Taq + dNTP's
(PCR). Each cycle of PCR consists of denatu- 5' 3' 5' 3' 3' 5'
ration of the double-stranded DNA by heating 3' 5'
at 94°C, annealing of the forward and reverse
primers to the template by cooling to 60°C, 1st CYCLE
and extension at 72°C of the growing com- TARGET
94°C Denature
plementary strand catalyzed by the enzyme DNA
Taq polymerase. The target DNA sequence, 3rd CYCLE
5' 3'
bracketed by the forward and reverse 5' 3'
primers, is indicated by a black box in the 3' 5'
3' 5'
template DNA. In the third cycle, blunt-ended 5' 3'
double-stranded products (also indicated by 3' 5'
a black box) are formed with its 5’ and 60°C Anneal
5' 3'
3’ ends coinciding with the primers. These
target DNA fragments continue to grow 3' 5'
5' 3'
exponentially with each cycle. 5' 3' 3' 5' 5' 3'
3' 5' 3' 5'
2nd CYCLE
5' 3' 5' 3'
72°C Extend
3' 5' 3' 5'
5’ 3'
5' 3'
5' 3' 3' 5'
3' 5' 3' 5'
5' 3'
5' 3'
3' 5'
3' 5'
5' 3' 5' 3' 5' 3'
3' 5' 3' 5' 3' 5'
6888_Ch04_077-102 29/10/18 4:45 PM Page 90

90 PART I Fundamental Concepts

product reaches a plateau, the level of which depends on


Template Taq Polymerase
the initial number of target sequences, the quantity of dNTP's
5' ACTGCGGCTACGCCCGTATGCG 3'
reagents, and the efficiency of extension. Primer (5' ACTGCGG 3')
3' TGACGCCGATGCGGGCATACGC 5'
Aside from cloning, the PCR has numerous applications + ddTTP
in immunohematology—for example, various modifications 94OC Denature ddATP
of this method have been used in the detection of infectious ddCTP
agents in donors’ blood.27,28 (Refer to Chapter 14.) 5' ACTGCGGCTACGCCCGTATGCG 3'
3' TGACGCCGATGCGGGCATACGC 5' ddGTP

DNA Sequencing 65OC Anneal and Extend

Individual fragments of DNA can be obtained in sufficient 5' ACTGCGG 3'


amount for determining their nucleotide sequence either by 3' TGACGCCGATGCGGGCATACGC 5'
molecular cloning or by PCR. Current methods of DNA
5' ACTGCGG 3'
sequencing are rapid and efficient. The easiest way of deter- 5' 3'
mining an amino acid sequence within a protein is the 5' 3'
sequencing of a cloned gene, as the coding sequence of a 5' 3'
gene can be unequivocally translated into the amino acid 5' 3'
sequence of its encoded protein. 5' 3'
Modern sequencing is based on the chain termination 5' 3'
method developed in 1977 by Fred Sanger at the Medical 5' 3'
Research Council’s Laboratory of Molecular Biology in Cam- 5' 3'
5' 3'
bridge, England.29 Just like the PCR, the Sanger sequencing
5' 3'
method is a controlled in vitro process mimicking DNA syn-
5' 3'
thesis. When a short synthetic primer (complementary 5' 3'
to the beginning of the fragment to be sequenced) is 5' 3'
hybridized to a single-stranded template in the presence of 5' 3'
the four dNTPs, DNA polymerase can synthesize a new
strand of DNA complementary to the template. The addi- 25 Cycles
tion of an incoming nucleotide occurs via the 3′ hydroxyl C T A C G C C C G T A T G C G
group in the deoxyribose necessary to form the phosphodi-
ester linkage. The principle of the Sanger method is based
on including small amounts of modified dNTPs that are
lacking that hydroxyl group into the reaction mix. These
modified dNTPs are 2′, 3′-dideoxyribonucleoside triphos- Figure 4–9. DNA sequencing: automated fluorescent cycle method. The
phates. They are designated by a double d (ddNTPs). Upon double-stranded DNA template is denatured by heating at 94°C. Cooling to 65°C
incorporation into the newly synthesized DNA, they cause allows the annealing of the oligonucleotide primer. In the presence of the four dNTPs
an immediate chain termination (hence the name of the and the four fluorescently labeled ddNTPs, Taq polymerase incorporates nucleotides
method), as no new phosphodiester linkage with the fol- into the growing chain, following the template sequence in a 5’ to 3’ direction. Once
in approximately 100 nucleotides, a ddNTP will be incorporated and synthesis will
lowing nucleotide is possible. Originally, Sanger sequencing stop. A fragment of DNA is generated that contains a fluorescent tag in its last
required using radioisotopes and laborious casting of poly- nucleotide. After capillary gel electrophoresis, the fluorescent labels are detected as
acrylamide electrophoretic gels in which the DNA frag- the terminated fragments pass a laser aimed at the capillary. The final output is an
ments were separated by size in denaturing conditions electropherogram showing colored peaks corresponding to each nucleotide position.
created by the presence of urea.
Until the development of the so called “next generation
sequencing” platforms, most laboratories have been using the discrete size, labeled fluorescently according to its last
automated fluorescent cycle sequencing method (Fig. 4–9). nucleotide, is produced. At the end of 20 to 30 cycles of the
The reaction, which is carried out in a thermocycler, contains reaction, millions of copies of the template DNA sequence
a DNA template, a primer, DNA polymerase, the four dNTPs, are terminated at each nucleotide so a mixture of fragments
and four ddNTPs, each labeled with a different fluorescent is obtained, each longer by one nucleotide than the previ-
dye. The reaction is heated to 94°C, which causes DNA de- ous one. These fragments are separated by size by capillary
naturation to a single strand, followed by cooling to 65°C to gel electrophoresis. The fluorescent labels are detected as
allow annealing of the primer and its extension by enzymatic the terminated fragments pass a laser aimed at the capillary.
addition of nucleotides. Several cycles are repeated. When the fluorescent terminators are struck by the laser
Working from the primer, the polymerase adds dNTPs beam, each emits a colored light of a characteristic wave-
or ddNTPs that are complementary to the DNA template. length, which is collected by the detector and interpreted
The ratio of dNTPs to ddNTPs is adjusted so that a ddNTP by the computer software as A (green), T (red), C (blue),
is incorporated into the elongating DNA chain, approxi- or G (yellow). The final output is an electropherogram
mately once in every 100 nucleotides. Each time a ddNTP showing colored peaks corresponding to each nucleotide
is incorporated, synthesis stops and a DNA fragment of a position.
6888_Ch04_077-102 29/10/18 4:45 PM Page 91

Chapter 4 Concepts in Molecular Biology 91

Detection of Nucleic Acids electrophoresis.30 Due to an enormous number of some


and Proteins restriction enzyme recognition sites in genomic DNA,
there may be hundreds of various-length fragments run-
The concepts of molecular cloning, creating expression DNA ning on the gel within a short distance from one another,
libraries and gene therapy, are fundamental in the contem- which upon staining is visible as a “smear” rather than
porary molecular research and development setting. Yet well-distinguished bands. To capture only the fragments
cloning may seem esoteric to a clinical laboratorian pre- of interest, the gel is placed over a nitrocellulose or nylon
sented with the task of identifying a specific mutation or membrane and overlaid with transfer buffer. The DNA
confirming the presence of a pathogen. In the following para- fragments are transferred or “blotted,” when a vacuum is
graphs, some classical and newer methods for detecting applied by the flow of a transfer buffer. The membrane-
specific nucleic acids and proteins will be described. bound fragments have the same relative positions as the
fragments separated by size on the gel. The filter is then
Nucleic Acid Hybridization incubated with a labeled probe under very specific tem-
perature conditions. Upon hybridization, the fragments
Base pairing between complementary strands of DNA or containing the sequence of interest are detected as labeled
RNA allows the specific detection of nucleic acid sequences. bands. The classical, laborious, and time-consuming
Double-stranded nucleic acids denature at high temperature Southern blotting is being replaced by PCR-based detec-
(90°C to 100°C) and renature when cooled to form double- tion methods (Fig. 4–10).
stranded molecules as dictated by complementary base pairing.
This process of renaturation is called nucleic acid hybridiza- Northern Blotting
tion. Nucleic acid hybrids can be formed between two strands This technique is a variation of Southern blotting and is used
of DNA, two strands of RNA, or one strand of RNA and one of to detect RNA instead of DNA. Total cellular RNAs are
DNA. DNA or RNA sequences complementary to any purified extracted and fractioned by size through gel electrophoresis
DNA fragment can be detected by nucleic acid hybridization. in the presence of substances, preventing RNA degradation
The DNA fragment—for example, a cloned gene or a PCR by the ubiquitous RNases. The RNAs are then blotted onto
product—is labeled using radioactive, fluorescent, or chemi- a filter and detected by hybridization with a labeled probe.
luminescent tags. The labeled DNA is used as a probe for This technique is used for studying gene expression and
hybridization with any DNA or RNA complementary se- is being replaced by quantitative reverse-transcriptase poly-
quence. The resulting hybrid double strand will be labeled and merase chain reaction (RT-qPCR).
can easily be detected by autoradiography or digital imaging.
DNA Microarrays
Southern Blotting DNA microarrays, also called gene chips, allow tens of thou-
In this technique developed by E. M. Southern, the DNA sands of genes to be analyzed simultaneously.31 A gene chip
analyzed is digested with one or more restriction endonu- consists of a glass slide or a membrane filter onto which
cleases, and the fragments are separated by agarose gel oligonucleotides or fragments of cDNA, which serve as

Figure 4–10. Southern blotting. (1) The DNA restric- 1 Agarose gel
tion digest is submitted to agarose gel electrophoresis. electrophoresis
(2) The DNA fragments are transferred from the gel to Gel
a filter membrane by flow of buffer with negative pres- Cathode 2 Blotting with transfer buffer Filter
sure under vacuum. (3) The filter is hybridized with a (–)
labeled probe, and the labeled DNA bands are detected Sealing Mask
by digital imaging.
Sealing Frame
Porous Support

Vacuum

Gel 3 Hybridization with


probe and detection
Anode
(+)

Filter
6888_Ch04_077-102 29/10/18 4:45 PM Page 92

92 PART I Fundamental Concepts

multiple probes, are printed by a robot in small spots at high beacons or FRET probes) labeled with fluorophores that emit
density. Because a chip can have more than 10,000 unique fluorescent light when binding to the newly synthesized
DNA sequences, scientists can produce DNA microarrays con- amplicon. If the assay is used to detect polymorphism, a
taining sequences representing all the genes in a cellular post-PCR monitoring of amplicon/probe separation upon
genome. A common application of this technique is the study slow increase of temperature is performed. This process is
of differential gene expression; for example, the comparison called melting curve analysis and allows for determination
of the genes expressed by a normal cell as opposed to those of of homo- or heterozygosity. A classical example of melting
a tumor cell. Messenger RNA is extracted from both types of curve analysis is the detection of factor V Leiden mutation in
cells, and cDNA is obtained via reverse transcription. The patients with this inherited coagulopathy.35
two sets of cDNAs are labeled with different fluorescent dyes Real-time PCR allows the quantification of specific
(typically red and green), and a mixture of the cDNAs is mRNAs in complex mixtures and is extremely useful in
hybridized to the gene chip that contains many of the probes the study of gene expression when combined with reverse
that represent genes already known in the human genome. transcription.
The array is analyzed using a laser scanner. The ratio of
red-to-green fluorescence at each specific spot on the array Reverse Transcriptase PCR
indicates the relative extent of transcription of the given gene Single copies of RNA can be detected by adding a step of cDNA
in the tumor cells compared with the normal cells. synthesis by reverse transcription (RT), prior to the PCR
Another application of microarrays, especially desirable in amplification (RT-PCR). By RT-PCR, cDNA molecules can
contemporary immunohematology, is the analysis of the be specifically amplified from total RNA obtained from cell
genome content rather than its expression. This represents a extracts, tissue sections, or plasma. The high sensitivity of
sophisticated form of genotyping (a process during which PCR-based DNA and RNA detection techniques makes them
polymorphisms within multiple genes may be identified valuable for early detection of transfusion-transmitted viruses,
simultaneously). In 2007, a prototype BLOOD-1 Illumina such as HIV and hepatitis B and C. The first RT-PCR assays de-
BeadChip microarray-based system was developed in the veloped by National Genetics Institute (UltraQual HIV-1) and
United States and was used to screen for 18 single nucleotide by Roche (COBAS AmpliScreen HIV-1) were designed to detect
polymorphisms (SNPs) within genes coding for RBC antigens these pathogens in donors’ blood and were approved by the
in the Duffy, Colton, Lewis, Diego, and other blood groups.32 FDA in 2001 and 2003.28,36 Real-time RT-PCR is employed in
GenomeLab SNPStream by Beckman Coulter is another the multiplex assay for concurrent detection of HIV, HCV, and
example of microarray technology that was customized for HBV (Roche Cobas TaqScreen MPX).37
RBC antigen typing at the Genome Quebec Innovation Center
in Canada.33 Transcription Mediated Amplification
Other FDA approved tests for HIV, hepatitis C virus (HCV),
Fluorescent In Situ Hybridization hepatitis B virus (HBV), and West Nile virus in donors’ blood
In this technique, scientists use fluorescent probes to detect are performed by means of another method developed by
homologous DNA or RNA sequences in chromosomes or Gen-Probe: transcription-mediated amplification (TMA).
intact cells. In this case, the hybridization of the probe to The starting material in this assay is RNA. The RNA serves
specific cells or subcellular structures is determined by direct as a template for a reverse transcriptase to synthesize a
microscopic examination. In 1995, this method was used to complementary DNA, which is bound to the RNA by hydro-
localize the ABO gene to chromosome 9.34 gen bonds to form an RNA/DNA hybrid. The DNA is never
amplified in the assay. After enzymatic removal of the RNA
PCR-Based and Other Amplification Techniques from the hybrid, the DNA is transcribed to hundreds of
RNA molecules. These multiple RNA molecules go through
Amplification of DNA by PCR allows the detection of even sin- another reverse transcription process to produce more
gle copies of DNA molecules. By contrast, Southern blotting RNA/DNA hybrids, and the whole process is repeated many
has a detection limit of around 100,000 copies. The specificity times. The detection of the pathogen (if its RNA is present
of the PCR amplification depends on the primers that hybridize in the sample) occurs via hybridization protection assay
to complementary sequences of the template molecule span- (HPA) technology. In this technology, ssDNA probes labeled
ning the target DNA fragment. With carefully chosen primers, with chemiluminescent molecules are added to form hybrids
PCR can be used to selectively amplify DNA molecules from with the amplified RNA molecules produced during TMA.
complex mixtures, such as genomic DNA from cell extracts. Light is emitted upon hybridization of the probes to the RNA
and captured by a luminometer.
Real-Time PCR Nucleic acid testing (NAT) is rapidly becoming a stan-
In real-time PCR, conducted in a special type of thermocycler dard method in blood banking. It allows the detection of
(LightCycler), the product formed during each cycle of am- pathogens before the appearance of a testable immune
plification is detected by fluorescence at the same time that response, such as screening of antibodies. Narrowing the
it is produced. In addition to primers, the real-time reaction preseroconversion window (during which donors can be
mixture contains DNA probes that are complementary to the infected but do not yet test positive by serologic methods)
region between the primers (called TaqMan or molecular helps to enhance the safety of blood products.38,39 Since NAT
6888_Ch04_077-102 29/10/18 4:45 PM Page 93

Chapter 4 Concepts in Molecular Biology 93

implementation, the average seroconversion window for HIV


has been shortened to 11 days (as compared to 22 days when
using serologic testing and 16 days when performing the
standard confirmatory p24 antigen testing). The average
seroconversion window for HCV has been shortened to
10 days (as compared to the original 82 days when serologic
testing is used). Consequently, the residual risk of HIV trans-
mission decreased from 9.7 to 1.2 incidents per million do-
nations, and the residual risk of HCV transmission decreased
from 2 to 1 incident per million donations. A significant
progress in testing for the transfusion-transmitted pathogens
in donor units was the ability of the NAT assays to detect
these pathogens concurrently. In 2008, the Gen-Probe assay,
initially approved for detection of HIV-1 and HCV only, was
also approved for the detection of HBV.36
Figure 4–11. Restriction fragment length polymorphisms (RFLPs). Southern blot
analysis of genomic DNA to determine HLA-DR genotype.
Antibodies as Probes for Proteins

Gene expression at the protein level can be studied by using typing for transplantation (Fig. 4–11), in paternity testing,
labeled antibodies as probes. In particular, monoclonal anti- and in forensic science (refer to Chapter 24, “Relationship
bodies are widely used in the technique called immunoblot- Testing”).
ting.40 This technique is also known as Western blotting by
analogy to Southern and Northern blotting. Proteins in cell PCR and Allele-Specific Probes
extracts are separated by polyacrylamide gel electrophoresis
in the presence of the ionic detergent SDS. In SDS-PAGE PCR can be used to amplify polymorphic genes from ge-
electrophoresis, each protein binds many detergent mole- nomic DNA. The PCR products are blotted on a nylon filter
cules, which causes its denaturation and provides a negative and hybridized with specific probes that allow the distinction
charge. Proteins will migrate to the anode; the rate of migra- of the different known alleles. This method, usually called
tion depends on their size. The proteins are then transferred dot blotting or sequence-specific oligonucleotide probe
from the gel onto a filter membrane. The protein/antigen of (SSOP), is commonly used for HLA typing (Fig. 4–12).
interest is detected by incubation of the membrane with a A variation of this method is sequence-specific PCR
specific labeled antibody. An example of Western blot is the (SSP), in which the alleles are distinguished by PCR ampli-
HIV p24 confirmatory testing. fication with primer pairs specific for one allele or a group
of alleles. Genomic DNA is submitted to PCR amplification
Techniques for Studying Gene with a battery of primer pairs. The products of the different
Polymorphism PCR reactions are run in an agarose gel and stained with EB
or SYBR Green. The presence of fluorescent bands of a
By recombinant DNA techniques, experimental analysis pro- defined size in the gel indicates the presence of the allele for
ceeds either from DNA into protein or from protein into which the primer pair is specific (Fig. 4–13).
DNA. An important consequence for transfusion medicine
has been the introduction of techniques for typing molecular DNA Sequencing
polymorphism, not only at the protein level but also at the
genetic level. These techniques are usually grouped under Alleles of polymorphic genes can also be specifically am-
the term DNA typing. plified by PCR and sequenced after cloning into plasmid

Restriction Fragment Length Polymorphism

Restriction fragment length polymorphism (RFLP) can be


used to detect the glycosyl transferase gene in a person who DRw52 LRS PROBE
is type AO or BO. The nucleotide deletion of the O allele re-
sults in the loss of a BstEII site (GGTGACC) and creation
of a new KpnI site (GGTACC). RFLP can also be used to
determine if an individual carries the mutation for sickle
cell anemia. The gene that codes for hemoglobin S (HbS)
has one nucleotide difference with the normal β-globin
coding allele. The sixth codon of the defective gene is GTG
(Val). The sixth codon of the normal gene is GAG (Glu).
The restriction site for MstII (CCTGAGG) is lost in the Figure 4–12. Sequence-specific oligonucleotide probe (SSOP). Dot blot of PCR
mutated gene (CCTGTGG). RFLP is widely used in HLA products hybridized to an allelic-specific HLA probe.
6888_Ch04_077-102 29/10/18 4:45 PM Page 94

94 PART I Fundamental Concepts

DRB1*13 DRB1*08 Systems Biology


The molecular techniques developed in recent years and
older techniques, still valuable in the study of nucleic acids
and the cellular processes in which they are involved, keep
improving and expanding. Many are being automated and
miniaturized. While it is necessary to have the exact
sequence of DNA that is of interest (e.g., if it is associated
with an inherited disease), it is equally important to under-
stand how that DNA is expressed inside a living cell. As
technology is developed, it will be interesting to see what
new methods become available and occur in common use.
Of particular interest to biotechnology is the development
of nanotechnology and nanostring technology applications,
high throughput DNA systems, and computational bio-
informatics in which complex models of expression and
regulation (such as proteasomes) are being assembled and
analyzed.
DQB1*03 DQB1*04 DRB3*00
Some of the most exciting recent advances in molecular
Figure 4–13. Sequence-specific PCR (SSP). Genomic DNA was PCR amplified biology have been the result of analyzing the complete nu-
with primer pairs specific for different HLA class II alleles or group of alleles. cleotide sequence of the human genome. A draft sequence
Note the control band indicating successful PCR. (Courtesy of HLA Laboratory,
Department of Transfusion Medicine, NIH.)
of the human genome was published in 2001 by two inde-
pendent teams of researchers.24,41 The public effort was led
by the International Human Genome Project, which used
vectors. Alternatively, direct sequencing of PCR products BAC libraries (briefly described in the “Tools for DNA
can be done without previous cloning, if the product ob- Cloning” section). The other group, led by Craig Venter of
tained is pure enough. This procedure is currently used in Celera Genomics, used a “shotgun” approach in which small
HLA typing for allogeneic hematopoietic stem cell trans- fragments of genomic DNA were cloned and sequenced.
plant (HSCT). Overlaps between the fragment sequences were used to as-
semble the whole genome. This draft encompassed about
DNA Profiling or “Fingerprinting” 90% of the euchromatin portion of the human genome. Con-
tinuing work by researchers around the world rendered a
Minisatellites and microsatellites are regions of DNA inter- high-quality human genome sequence in 2003—50 years
spersed in the human genome formed by a variable number after the Nature paper in which Watson and Crick described
of tandem repeats (VNTRs) of short nucleotide sequences. the structure of DNA. This wealth of information, which is
The variability depends on the number of repeat units con- constantly updated, can be accessed through the National
tained within each “satellite.” Minisatellites or variable tan- Center for Biotechnology Information (NCBI) website
dem repeats are composed of repeated units ranging in size (www.ncbi.nlm.nih.gov).42
from 9 to 80 bp. Microsatellites or short tandem repeats In its first 50 years, molecular biology has been primarily
(STRs) contain repeat units of 2 to 5 nucleotides. an analytical science, concerned mainly with reducing
The polymorphism of these loci in the human population biological problems to the level of individual genes. This
is so high that there is a very low probability that two indi- approach has been extraordinarily successful in finding the
viduals will have the same number of repeats. When several key genes involved in cellular replication and development
loci are analyzed simultaneously, the probability of finding and in identifying the genes involved in genetic disorders.
two individuals in the human population with the same The challenge for the next few decades is going to be
polymorphic pattern is extremely low. The independence of understanding the synchronized activity of numerous genes
inheritance of the different loci tested is ensured by choosing during key biological events, such as development and
loci on different chromosomes. The pattern of polymor- memory, and understanding the multiple genes presumably
phism (the differences in number of repeats or length) can involved in complex diseases such as cancer and diabetes.
be determined by RFLP or PCR analysis. In 1997, the Federal Another complication is the fact that several different
Bureau of Investigation recommended a panel of 13 STRs, proteins can be produced from a single gene by alternative
plus an XY marker, for criminal investigations. With this RNA splicing and that additional forms are created by post-
number of independently inherited polymorphisms, the translational modifications.
probability of even the most common combinations is less To solve these complex issues, biologists are turning to
than 1 in 10 billion. Thus, modern DNA testing can uniquely more synthetic approaches that allow them to examine the
identify each person, hence the name DNA fingerprinting. coordinated expression of multiple genes. Experiments
DNA profiling is used in forensic applications, paternity test- performed using DNA and protein chips are starting to
ing, and to follow chimerism after HSCT. show how hundreds or thousands of genes are expressed
6888_Ch04_077-102 29/10/18 4:45 PM Page 95

Chapter 4 Concepts in Molecular Biology 95

coordinately in response to different developmental and Since then, several updates have been published.46,47 The full
environmental stimuli. Analysis of these data is one of current classification can be found at the ISBT website
the applications of bioinformatics, a discipline that uses (http://www.isbtweb.org ).48
computer algorithms to manage and analyze large-scale Most of the blood group systems are coded by variants of
experiments. a single gene. There are four exceptions: Rh; Xg; Chido/
The functions of genes implicated in a specific pathway Rodgers, which have two genes each; and MNS, which has
can be further explored by reverse genetics. Genes can be three. As an example, the Rh system consists of two genes:
“knocked out” or “knocked in” as wild type or mutated into D and CE. These genes have 97% homology, which can add
the germline of a mouse or other animal model.43,44 The difficulty in molecular methods for genotyping this system.
resulting knockout transgenic animal can be analyzed for Table 4–3 enumerates the blood group systems, their genes
metabolic or behavioral changes, which can give clues to the with the chromosomal location, and the corresponding
mechanism of function of the gene under study. A novel way antigens.
of studying specific gene function is RNA interference, by
which small synthetic RNA molecules can be used to silence Molecular Basis of Blood Group Polymorphism
a specific target gene.16 This new biology that is emerging is
called systems biology, and it requires integration of knowl- According to the Blood Group Antigen Gene Mutation Data
edge from experiments done in vitro, in vivo, and in silico Base (BGMUT) in March 2014, there were 34 blood group
(in a computer). systems (as compared to 30 reported in 2010), 44 genes, and
1,545 alleles known in the human population.49 By 2018, the
Red Blood Cell Genotyping number of identified blood group systems increased to 36.
The systems ABO, P, Lewis, H, I, and MNS comprise
Molecular RBC antigen typing could be of tremendous antigens that are carbohydrates on glycoproteins or glyco-
advantage in situations where serologic testing is impossi- lipids. The genes that constitute these systems code for
ble or inconclusive. Currently considered as supplemental glycosyltransferases that catalyze oligosaccharide chain
rather than routine practice, it has the potential to revolu- synthesis. For the rest of the blood group systems, the anti-
tionize the way blood cell inventory may be searched for gens are direct consequences of amino acid variation in the
multiple antigen-negative units. The knowledge of theoreti- protein sequence. Most blood group variants are the result
cal principles and practical skills to perform red blood cell of one or more single nucleotide polymorphisms (SNPs)
genotyping is becoming indispensable in contemporary encoding amino acid substitutions in a glycosyltransferase
blood banking. or the extracellular domain of a membrane protein. Other
mechanisms of polymorphism generation are gene deletion,
Genetic Basis of Blood Groups single nucleotide deletion, insertion, and intergenic recom-
bination. Clinically significant blood group systems are
Early important events in the molecular biology of transfu- listed in Table 4–3.
sion medicine includes the cloning and sequencing of the
gene that codes for the MN polymorphism, GYPA, in 1986, Clinical Applications of Red Blood
followed by the ABO gene in 1990 (review Table 4–1). Now Cell Genotyping
the genes for all the blood group systems have been identi-
fied and localized in specific chromosomal regions.23 Also There are several important applications for red blood cell
known are the alleles that code for the blood group polymor- genotyping in use today.50,51 These methods help to confirm
phisms, the structure of the gene products, and in many serologic testing. In cases where serology is not possible or
cases their function. This knowledge has allowed the deve- not sensitive enough or where discrepancies occur, genotyp-
lopment of red blood cell group typing at the DNA level. ing provides results that can be used to obtain a blood
Molecular genotyping is being increasingly used in clinical type on a donor or patient. Applications of genotyping
blood banking, where it does not replace but rather comple- include fetal DNA typing, blood group typing of donors for
ments traditional phenotyping. Molecular biology provides alloimmunized patients, screening blood donors to locate
information to help make decisions in transfusion medicine, rare blood group phenotypes, screening blood inventory for
such as obtaining compatible blood in cases of massive trans- antigen-negative units, determining the frequency of blood
fusion or transplantation. The field of molecular biology is group polymorphisms in a given population, determining
constantly changing and improving. Methods become more zygosity for the fathers of fetuses at risk for hemolytic disease
efficient and less expensive. As greater numbers of popula- of the fetus and newborn (HDFN), and blood group typing
tions are analyzed, different alleles are discovered and added of patients with autoimmune hemolytic anemia and other
to databases that are updated frequently. diseases.
A committee of the International Society of Blood Trans-
fusion (ISBT) is responsible for the nomenclature of red Fetal DNA Typing
blood cell surface antigens. A monograph published in 2004 Initially, ABO and Rh typing should be done on a pregnant
described the ISBT terminology for red blood cell surface mother and should include an antibody screening. If the
antigens and genes and tabulated a complete classification.45 woman is D negative and has a negative antibody screen
6888_Ch04_077-102 29/10/18 4:45 PM Page 96

96 PART I Fundamental Concepts

Table 4–3 Blood Group Systems With Number of Antigens, Coding Gene(s),
and Chromosomal Location48
ISBT System ISBT System Chromosomal No. of
No. Name Symbol Gene Name(S) Location Alleles
001 ABO ABO ABO 9q34.2 4

002 MNS MNS GYPA, GYPB, GYPE 4q31.21 46

003 P P1 P1 22q11.2–qter 1

004 Rh RH RHD, RHCE 1p36.11 > 50

005 Lutheran LU LU 19q13.32 20

006 Kell KEL KEL 7q34 31

007 Lewis LE FUT3 19p13.3 6

008 Duffy FY DARC 1q23.2 6

009 Kidd JK SLC14A1 18q12.3 3

010 Diego DI SLC4A1 17q21.31 21

011 Yt YT ACHE 7q22.1 2

012 Xg XG XG, MIC2 Xp22.33 2

013 Scianna SC ERMAP 1p34.2 7

014 Dombrock DO ART4 12p12.3 6

015 Colton CO AQP1 7p14.3 3

016 Landsteiner-Wiener LW ICAM4 19p13.2 3

017 Chido/Rodgers CH/RG C4A, C4B 6p21.3 9

018 H H FUT1 19q13.33 1

019 Kx XK XK Xp21.1 1

020 Gerbich GE GYPC 2q14.3 8

021 Cromer CROM CD55 1q32.2 15

022 Knops KN CR1 1q32.2 9

023 Indian IN CD44 11p13 4

024 Ok OK BSG 19p13.3 1

025 Raph RAPH CD151 11p15.5 1

026 John Milton Hagen JMH SEMA7A 15q24.1 5

027 I I GCNT2 6p24.2 1

028 Globoside GLOB B3GALT3 3q26.1 1

029 Gill GIL AQP3 9p13.3 1

030 Rh-associated glycoprotein RHAG RHAG 6p21-qter 3

031 FORS FORS GBGT1 9q34.2 5

032 JR JR ABCG2 4q22.1 29

033 LAN LAN ABCB6 2q35 42

034 VEL VEL SMIM1 1p36.32 3

035 CD59 CD59 CD59 11p13 3

036 Augustine AUG SLC29A1 6p21.1 4


6888_Ch04_077-102 29/10/18 4:45 PM Page 97

Chapter 4 Concepts in Molecular Biology 97

using IgG AHG techniques, she is a candidate for RhIG. prohibitively expensive to use on a large scale. Genotyping
If the antibody screening is positive, then the antibody speci- with PCR techniques allows for the probes (or primers) to
ficity must be determined. If the father has the correspond- be synthesized at low cost and used in DNA-based methods,
ing antigen, then his zygosity, either heterozygous or as long as information about the sequence of the gene is
homozygous, should be established. If the father is heterozy- known. Different alleles can be tested at the same time in the
gous, then the genotype of the fetus can be determined using multiplex PCR assays or on microarrays. Once known, the
PCR testing of amniocentesis samples, chorionic villus sam- genotype information can be put into a database and does
pling, or cordocentesis samples. not require repeated testing, which saves considerable time
PCR has a very high sensitivity, approaching 100%, and a and money. A rare donor profile can be generated, linking
low false-negative rate, so it is an excellent method. DNA potential donors to potential recipients for better recruitment
probes are known for most if not all genes playing a role in in cases of emergency. For patients requiring donor units
HDFN. As an alternate approach, the typing for the D anti- with rare phenotypes, the costs and time involved in geno-
gen at the molecular level can be done on maternal samples typing such donor units are acceptable.
as early as the second trimester. Fetal DNA in maternal
samples is derived from apoptotic syncytiotrophoblasts; it Determining the Frequency of Blood Group
increases in concentration throughout pregnancy and is Polymorphisms in a Population
cleared soon after delivery. Genotyping databases can be used to predict the possibility
Maternal-only sample DNA testing will likely become of finding a specific allele in a given population once a
more routine in the near future, as it has many advantages. similar population has been analyzed at the genotype level.
All DNA-based information allows earlier intervention Because most blood group alleles are the result of a single
in cases where HDFN is suspected.52 (Refer to Chapter 19, nucleotide polymorphism, screening of blood samples for
“Hemolytic Disease of the Fetus and Newborn [HDFN].”) allele frequency can be relatively easy and done by simple
In addition to HDFN, there is risk to the developing fetus in DNA-based methods. Several different methods employed
cases of platelet and neutrophil antigen incompatibility, re- for this include RFLP and SSP-PCR (single or multiplex),
sulting in fetal and neonatal alloimmune thrombocytopenia real-time quantitative PCR, sequencing, and microarray tech-
(FNAIT) and neonatal alloimmune neutropenia (NAN). nology using DNA probes on chips.
Genotyping can help resolve these cases and determine
if there is an associated risk due to maternal platelet and Blood Group Typing of Patients With Autoimmune
neutrophil antigens and HLA type. Hemolytic Anemia and Other Diseases
Patients with autoimmune hemolytic anemia and other con-
Extensive Blood Group Typing of Donors ditions such as sickle cell anemia can have trouble obtaining
for Alloimmunized Patients compatible donor units. They are often sensitized to many anti-
Patients with multiple antibodies may require extensive test- gens and have a rapid turnover of red blood cells, requiring
ing to find compatible blood for transfusion. It is now possi- frequent transfusion. Because they can have a complex popu-
ble to genotype potential donors and the patient to prevent lation of red blood cells, with autologous cells mixed with
further immunization and to characterize the donor at a more transfused cells from more than one donor, genotyping meth-
specific level.53,54 DNA testing can be done in place of exten- ods are an excellent way to type these donors. A small amount
sive family studies to determine the genotype. Sequencing of of patient white cells or cheek cells can be used to determine
the entire gene of interest, often required to obtain sufficient the genotype using standard DNA-based methods, and the sub-
information, may be labor-intensive. The amount of work sequent patient phenotype can be determined from the geno-
may be lessened if exon sequencing is done first and if RFLPs type. Buccal swabs are also beneficial in cases of hematopoietic
can be done on certain areas of the gene at the beginning of stem cell transplant patients with a mixed chimerism. The
the study. Focused sequencing with comparisons to antigenic DNA obtained from the buccal swab can be used to predict the
profiles can also be used. Having extensive information on recipient’s native phenotype, while DNA from the donor can
the patients’ and the potential donors’ genotypes allows trans- be used to predict the donor’s phenotype.
fusion to take place in a safer environment. Knowing the anti- Although crossmatching may not provide totally compati-
gens that the patient does not have based on genotyping ble results, especially with autoimmune diseases, at least
prevents further alloantibodies from being made from the alloantigens can be ruled out when transfusion is required,
transfusion of blood that is positive for these antigens. Be- thereby preventing further alloimmunization in an already-
cause red blood cells have lost their nuclei, white cells can be difficult-to-transfuse patient. In addition to saving the time
used for genotyping and other normal adult diploid cells. required for repeated adsorptions, elutions, and extensive
phenotyping in these patients and in donor units, the infor-
Screening of Blood Donors to Find Rare Blood mation obtained by DNA-based methods is highly accurate
Group Phenotypes and easy to obtain. It is a permanent record of the patient’s
Using DNA-based methods to screen blood donors allows genetic profile, and it is necessary information in cases where
for a high-throughput system screening in comparison with transplant may be considered. It is also a permanent record
serology-based methods. Blood group antisera often are not of the donor’s profile when performed, unless the patient
readily available for rare blood group phenotypes or are undergoes a hematopoietic stem cell transplant.
6888_Ch04_077-102 29/10/18 4:45 PM Page 98

98 PART I Fundamental Concepts

CASE STUDIES 5.7

Case 4-1
48
A 56-year-old female presented for cardiac valve surgery 60.5
68.8
with no prior blood bank history and no transfusions
84.5 85.8
within the last 3 months. Her blood tested group A
RhD positive and the antibody screen reacted with all 94.3
cells tested. An anti-E, warm autoantibody was identi- O+ O+mf
fied, as well as anti-C like antibody in the autoadsorbed 62 52 140
plasma. The patient serologically typed C positive, and 39.5
the anti-C was thought to be autoimmune. Predicted 31.2
human erythrocyte antigen (pHEA) testing was per- 15.5 14.2
formed on the sample and indicated the patient may
25 39 53 70 74 80 95 108 136 143
express the C variant, r′S. The r′S presents a partial C,
Anti-E
normal c, weakened e, normal E, and VS positive phe-
1st HEA 2nd HEA Anti-D O–
notype. These patients type C positive when typed with Sample Sample 144
monoclonal MS24 antisera. Days Post-Transplant Buccal
HEA
1. Was the anti-C an autoantibody or alloantibody, and
= Recipient (%) = Donor (%)
why?
2. What antigen-negative units should be crossmatched
Figure 4–14. Graphical representation of hematopoietic stem cell engraft-
to this patient?
ment in a patient who received a transplant. Quantitative PCR was used to mon-
itor the engraftment by analyzing both the donor and recipient. Monitoring
Case 4-2 typically uses, but is not limited to, CD3+ or CD33+ cells.

A 64-year-old group O RhD-negative (D–) male with


acute myeloid leukemia (AML) in his second clinical buccal swab of the patient, obtain some DNA from the
remission received an HSCT from a group O RhD-positive MUD, and perform the pHEA testing to supply antigen-
(D+) matched unrelated donor (MUD). The HLA match negative units corresponding with both the donor and
was a 10/10, and the patient had no history of anti-D. recipient. Figure 4-14 illustrates the patient’s peripheral
The patient received a reduced intensity conditioning of blood chimerisms with defining serological dates.
busulfan and fludarabine in the days preceding the trans- Platelet, neutrophil, and red blood cell counts confirmed
plant. The patient’s RhD typed 4+ on day 49 post HSCT engraftment. The graph shows when engraftment oc-
and was a confirmed D+ conversion on day 62. He was curred, when the donor progenitor cells began produc-
discharged on day 17 and maintained an average hemo- ing red blood cells, and when his leukemia relapses.
globin level of 8.9 g/dL. The first positive antibody Table 4–4 compares the pHEA tests of DNA from the
screen was a sample drawn on day 95. Anti-E, possible whole-blood samples collected on days 70 and 95, the
anti-Jka, warm and cold autoantibodies were identified. buccal swab, and the MUD DNA.
The possible anti-Jka reactivity was not confirmed in
1. Were the antibodies being identified produced by the
any subsequent specimens. pHEA testing was performed
recipient or donor B cells?
on the day 95 sample. On day 80, the patient was
2. Assuming there was no crossover contamination, what
48% donor, so pHEA testing was also performed on
was the probable cause for the indeterminate calls on
a day 70 sample when the patient’s chimera study
the day 70 sample?
demonstrated the donor at just about 68.8%. The two
samples gave discrepant results on the C antigen predic-
tions. The day 70 sample produced indeterminate calls
(IC) on the E, and N predictions. The IC is usually made Table 4–4 Discrepant Predicted
when there is crossover contamination in a sample. On Antigens
day 136, the patient was admitted due to his hemoglobin C E Jka N
decreasing to 5.5 g/dL. Following three transfusions of
E- and Jka-negative D+ units, the hemoglobin remained Day 70 + IC + IC
at 5.0 g/dL through day 137. On day 136, the patient
Day 95 0 0 + +
had a strongly reacting direct antiglobulin test and anti-
body identification of anti-D found in the alloadsorbed Buccal 0 0 + +
plasma. On day 140, the patient typed RhD mixed field
MUD + + + 0
after receiving only 1 D– and 17 D+ red blood cell trans-
fusions in the past 3 months. It was decided to collect a
6888_Ch04_077-102 29/10/18 4:45 PM Page 99

Chapter 4 Concepts in Molecular Biology 99

Case 4-3 for sequencing to identify the cause for the discrepancy.
The SLC14A1 gene codes for the Kidd antigens, with
A 64-year-old woman with coronary artery disease, no one SNP of 838G>A in exon 9 dictating the difference
history of red blood cell transfusions, and a current between aspartic acid or asparagine translation and
negative antibody screen had a warfarin-related Jka or Jkb expression. Exons 4–11 of the SLC14A1 are
retroperitoneal hemorrhage. She received seven units of expressed. The sequencing revealed a single-nucleotide
red blood cells on the first 2 days of admission. The deletion at c.1038G in exon 11 coding for a premature
antibody screen collected on day 12 was positive, and stop codon five amino acids downstream.
anti-Jkb was identified. pHEA by allelic-specific testing
was performed on the day 12 sample, and predicted the 1. What could have been the cause for the discrepancy
patient to be Jk(a+b+). The patient was then serologically between the serological and molecularly predicted
typed and found to be Jk(b–). A sample was then sent Jkb phenotype?

SUMMARY CHART
 The central dogma of molecular biology: DNA →  By reverse transcription, mRNA is transcribed into
RNA → protein. complementary DNA (cDNA).
 Proteins have structural, enzymatic, and gene-  Automated fluorescent DNA sequencing based on the
regulatory function. Through these mechanisms, the Sanger dideoxy, chain termination method is a stan-
genotype of a cell is translated into its phenotype. dard laboratory procedure.
 DNA is the genetic material.  DNA sequencing of a cloned cDNA corresponding to
 DNA is a double helix, consisting of two antiparallel a given gene is the easiest way to determine the amino
strands of stacked nucleotides paired through hydro- acid sequence of a protein.
gen bonds. Adenine (A) always pairs with thymine (T),  Genomic and cDNA libraries are collections of clones
and cytosine (C) always pairs with guanine (G). containing the genetic material of a cell.
 The structure of DNA determines its function. The  Base pairing between complementary strands of DNA or
sequence of nucleotides of one strand determines RNA (hybridization with a labeled probe) is used to de-
the sequence of its complementary strand, the basis tect specific nucleic acid sequences in complex mixtures.
for the semiconservative way of replication.  Southern blotting, Northern blotting, and dot blotting
 A gene is transcribed into precursor RNA, and the are hybridization-based techniques for nucleic acid
spliced mRNA is translated into the amino acid sequence-specific recognition.
sequence of the coded protein. The sequence of mRNA  The polymerase chain reaction (PCR) is an in vitro
unequivocally determines the sequence of the protein. method for DNA amplification.
 Recombinant DNA is the DNA of one organism “cut  Molecular polymorphism is studied at the genetic level
and pasted” into a carrier vector. The foreign gene by DNA typing. Methods for DNA typing relevant for
introduced in a host organism is functional because the transfusion medicine are restriction fragment length
genetic code is universal. polymorphism, allele-specific oligonucleotide probe
 By DNA cloning, recombinant genes of complex hybridization, allele-specific PCR amplification, DNA
animals, such as humans, are introduced into simple sequencing, and DNA profiling (DNA fingerprinting).
organisms, such as bacteria, and other model organ-  PCR and TMA are used for the early detection of trans-
isms, such as mice, allowing structural and functional fusion-transmitted pathogens.
studies.  Other therapeutic uses of molecular biology are gene
 Restriction endonucleases, bacterial enzymes that recog- therapy and the clinical use of recombinant proteins,
nize and cut specific DNA sequences, are fundamental such as interferons, coagulation factors, and growth
tools for DNA cloning. factors.
 Gel electrophoresis separates nucleic acids by size.  Molecular RBC antigen typing is used to either confirm
 The most common host cell is the bacterium E. coli. serologic testing or in cases where serology is not pos-
 Plasmids, the most commonly used vectors, are inde- sible or is not sensitive enough or where discrepancies
pendently replicating circular DNA molecules modified occur.
to provide the host cell with resistance to antibiotics  Molecular RBC antigen typing is a prediction from
(selectable marker) and one or more restriction enzyme DNA of the antigens that may be expressed on the RBC
sites for inserting the recombinant gene. membrane.
6888_Ch04_077-102 29/10/18 4:45 PM Page 100

100 PART I Fundamental Concepts

Review Questions 10. Recombinant DNA techniques:


a. Are not used in a clinical setting.
1. The central dogma of molecular biology states that: b. Are useful research tools.
a. DNA is the genetic material. c. Are not used in blood banking.
b. RNA is the genetic material. d. Are useful only for research.
c. DNA is translated to mRNA. 11. Transcription-mediated amplification:
d. Proteins are transcribed from mRNA.
a. Requires thermostable DNA polymerase.
2. Recombinant-DNA technology is possible because: b. Is an isothermal procedure.
a. Restriction endonucleases cut RNA. c. Is an obsolete method currently replaced by SSOP.
b. Restriction endonucleases cut proteins. d. Utilizes probes labeled with fluorescent tags.
c. The genetic code is universal. 12. Preseroconversion window:
d. Bacteria are difficult to culture.
a. Is the time when donors can be infected but do not
3. Agarose gel electrophoresis is a technique used for: yet test positive by serologic methods.
a. DNA synthesis. b. May be narrowed by using molecular methods.
b. RNA synthesis. c. Refers mainly to viral pathogens.
c. Separation of DNA molecules by size. d. All of the above
d. Oligonucleotide synthesis. 13. Red blood cell molecular antigen typing is useful in all
4. Restriction fragment length polymorphism (RFLP) is listed situations except:
based on the use of the enzymes: a. In screening RBC inventory for antigen-negative
a.Reverse transcriptases. units.
b.Bacterial endonucleases. b. When reagent antibodies are weak or unavailable.
c.DNA polymerases. c. In quantitative gene expression analysis.
d.RNA polymerases. d. When resolving ABO discrepancies.

5. The polymerase chain reaction (PCR): References


a. Is carried out in vivo.
1. Crick F. Central dogma of molecular biology. Nature.
b. Is used for peptide synthesis. 1970;227(5258):561-63.
c. Requires RNA polymerase. 2. Griffith F. The significance of pneumococcal types. J Hyg
d. Is used for the amplification of DNA. (Lond). 1928;27(2):113-59.
3. Avery OT, MacLeod CM, McCarty M. Studies on the chemical
6. Plasmids are: nature of the substance inducing transformation of pneumo-
a. Vectors used for molecular cloning. coccal types. Induction of transformation by a deoxyribonu-
b. Antibiotics. cleic acid fraction isolated from Pneumococcus type III. J Exp
Med. 1944;79(2):137-58.
c. Enzymes. 4. Hershey AD, Chase M. Independent functions of viral protein
d. Part of chromosomes. and nucleic acid in growth of bacteriophage. J Gen Physiol.
1952;36(1):39-56.
7. Some model organisms: 5. Chargaff E, Magasanik B, Visher E, Green C, Doniger R, Elson
a. Simplify the study of human disease. D. Nucleotide composition of pentose nucleic acids from yeast
b. Are used to produce recombinant proteins. and mammalian tissues. J Biol Chem. 1950;186(1):51-67.
c. Are prokaryotes and some are eukaryotes. 6. Watson JD, Crick FHC. Molecular structure of nucleic acids: a
structure for deoxyribose nucleic acid. Nature. 1953;171:
d. All of the above 737-38.
8. DNA sequencing: 7. Breslauer KJ, et al. Predicting DNA duplex stability from the
base sequence. Proc Natl Acad Sci USA. 1986;83(11):3746-50.
a. Is more difficult than peptide sequencing. 8. Lewin B. Genes VIII. New York: Prentice Hall; 2004. p. 392.
b. Requires the use of RNA polymerase. 9. Pauling L, Itano, HA, et al. Sickle cell anemia a molecular
c. Can never be automated. disease. Science. 1949;110(2865):543-48.
d. Is an enzymatic in vitro reaction. 10. Ingram VM. Gene mutations in human hemoglobin: the chemi-
cal difference between normal and sickle cell hemoglobin.
9. RFLP and SSP are techniques used for: Nature. 1957;180:326-28.
11. Keller EB, Zamecnik PC, Loftfield RB. The role of microsomes
a. Protein isolation. in the incorporation of amino acids into proteins. J Histochem
b. RNA isolation. Cytochem. 1954;2(5):378-86.
c. DNA typing. 12. Brenner S, Jacob F, Meselson M. An unstable intermediate
d. Protein typing. carrying information from genes to ribosomes for protein
synthesis. Nature. 1961;190:576-81.
13. Nirenberg M, Leder, P. RNA codewords and protein synthesis.
Science. 1964;145:1399-407.
14. Temin HM, Mizutani S. RNA-dependent DNA polymerase in
virions of Rous sarcoma virus. Nature. 1970;226:1211-13.
6888_Ch04_077-102 29/10/18 4:45 PM Page 101

Chapter 4 Concepts in Molecular Biology 101

15. Baltimore D. RNA-dependent DNA polymerase in virions of infectious agents and HIV diagnostic assays. Available from:
RNA tumour viruses. Nature. 1970;226:1209-11. www.fda.gov/cber/products/testkits.htm.
16. Sharp PA. RNA interference—2001. Genes Dev. 2001;15: 37. Wiedmann M, Kluwick S, Walter M, Fauchald G, Howe J,
485-90. Bronold M, et al. HIV-1, HCV and HBV seronegative window
17. Yamamoto F, et al. Cloning and characterization of DNA com- reduction by the new Roche cobas TaqScreen MPX test in
plementary to human Histo-blood group A transferase mRNA. seroconverting donors. J Clin Virol. 2007;39(4):282-87.
J Biol Chem. 1990;265:1146-151. 38. Glynn SA, Kleinman SH, Wright DJ, Busch MP. International
18. Sambrook J, Russell DW. Molecular cloning: a laboratory application of the incidence rate/window period model. Trans-
manual. 3rd ed. Cold Spring Harbor (NY): Cold Spring Harbor fusion. 2002;42:966-72.
Laboratory Press; 2001. 39. Dodd RY, Notari EP, Stramer S. Current prevalence and
19. Alberts B, et al. Molecular biology of the cell. 4th ed. New York: incidence of infectious disease markers and estimated window
Garland Publishing; 2002. period risk in the American Red Cross blood donor population.
20. Nathans D, Smith HO. Restriction endonucleases in the analy- Transfusion. 2002;42:975-79.
sis and restructuring of DNA molecules. Ann Rev Biochem. 40. Kohler G, Milstein C. Continuous cultures of fused cells
1975;44:273-293. secreting antibody of predefined specificity. Nature. 1975;256:
21. Wagner FF, Flegel WA. RHD gene deletion occurred in the 495-97.
Rhesus box. Blood. 2000;95:3662-68. 41. Venter JC, et al. The sequence of the human genome. Science.
22. Kirsanov KI, Lesovaya EA, Yakubovskaya MG, Belitsky GA. 2001;291:1304-51.
SYBR Gold and SYBR Green II are not mutagenic in the Ames 42. National Center for Biotechnology Information [Internet].
test. Mutat Res. 2010;699(1-2):1-4. Bethesda (MD): NCBI. Available from: www.ncbi.nlm.nih.gov.
23. Logdberg L, Reid M, Zelinski T. Human blood group genes 43. Capecchi MR. Altering the genome by homologous recombi-
2010: chromosomal locations and cloning strategies revisited. nation. Science. 1989;244:1288-92.
Transfus Med Rev. 2011;25(1):36-46. 44. Bronson SK, Smithies O. Altering mice by homologous recom-
24. International Human Genome Sequencing Consortium. Initial bination using embryonic stem cells. J Biol Chem. 1995;
sequencing and analysis of the human genome. Nature. 269:27155-58.
2001;409:860-921. 45. Daniels GL, et al. Blood group terminology 2004: from the
25. Gaspar HB. Bone marrow transplantation and alternatives for International Society of Blood Transfusion committee on
adenosine deaminase deficiency. Immunol Allergy Clin North terminology for red cell surface antigens. Vox Sanguinis.
Am. 2010;30(2):221-36. 2004;87:304-16.
26. Saiki RK, Gelfand DH, Stoffel S, Scharf R, Higuchi R, Horn GT, 46. Daniels GL, et al. International Society of Blood Transfusion
et al. Primer-directed enzymatic amplification of DNA with a Committee on terminology for red cell surface antigens:
thermostable DNA polymerase. Science. 1988;239:487-91. Cape Town report. Vox Sanguinis. 2007;92:250-53.
27. Busch MP, Kleinman SH. Nucleic acid amplification testing 47. Daniels GL, et al. International Society of Blood Transfusion
of blood donors for transfusion-transmitted diseases. Report Committee on terminology for red blood cell surface antigens:
of the Interorganizational Task Force on nucleic acid ampli- Macao report. Vox Sanguinis. 2009;96:153-56.
fication testing of blood donors. Transfusion. 2000;40: 48. International Society of Blood Transfusion Committee: http://
143-59. www.isbtweb.org/ accessed February 28, 2018.
28. Kraj B, Nadder T. Blood donor screening process and infectious 49. Patnaik, SK., Helmberg, W, Blumenfeld, OO. BGMUT database
disease testing using molecular methods. Adv Med Lab Profes- of allelic variants of genes encoding human blood group anti-
sionals. 2007;Oct. 8:17-20. gens. Transfus Med Hemother, 2014: 41(5):346-51.
29. Sanger F, Nicklen S, Coulson AR. DNA sequencing with chain- 50. Reid ME. Transfusion in the age of molecular diagnostics.
terminating inhibitors. Proc Natl Acad Sci USA. 1977;74: Hematology. 2009;171-77.
5463-67. 51. Casas J, Friedman DF, Jackson T, Vege S, Westhoff CM, Chou ST.
30. Southern EM. Detection of specific sequences among DNA Changing practice: red blood cell typing by molecular methods
fragments separated by gel electrophoresis. J Mol Biol. for patients with sickle cell disease. Transfusion, 2015:55
1975;98:503-17. (6 Pt 2):1388-93.
31. Brown PO, Botstein D. Exploring the new world of the genome 52. van der Schoot CE, de Haas M, Clausen FB. Genotyping to
with DNA microarrays. Nat Genet. 1999;21:33-37. prevent Rh disease: has the time come? Curr Opin Hematol.
32. Hashmi G, et al. A flexible array format for large-scale, rapid 2017:24(6):544-550.
blood group DNA typing. Transfusion. 2005;45(5):680-88. 53. Anstee DJ. Red cell genotyping and the future of pretransfusion
33. Montpetit A, et al. High-throughput molecular profiling of testing. Blood. 2009;114:(2):248-56.
blood donors for minor red blood cell and platelet antigens. 54. Tormey CA, Hendrickson JE. Routine non-ABO blood group
Transfusion. 2006;46(5):841-48. antigen genotyping in sickle cell disease: the new frontier
34. Bennett EP, et al. Genomic cloning of the human histo-blood in pretransfusion testing? Transfusion 2015; 55(6 Pt 2):
group ABO locus. Biochem Biophys Res Commun. 1374-1377.
1995;206(1):318-25. 55. Patnaik, SK, Helmberg W, Blumenfeld OO: BGMUT: NCBI
35. Lyondagger E, Millsondagger A, Phan T, Wittwer, CT. Detec- dbRBC database of allelic variations of genes encoding anti-
tion and identification of base alterations within the region of gens of blood group systems. Nucleic Acids Res. 2012; 40:
factor V Leiden by fluorescent melting curves. Mol Diagn. D1023-1029.
1998;3(4):203-9. 56. U.S. Food and Drug Administration. May 21, 2014 Approval
36. U.S. Food and Drug Administration. Washington DC: U.S. Letter - Immucor PreciseType Human Erythrocyte Antigen
FDA. 2018. Complete list of donor screening assays for (HEA) Molecular BeadChip Test. Available at www.fda.gov
6888_Ch04_077-102 29/10/18 4:45 PM Page 102
Blood Groups and
Serologic Testing

Chapter 5
The Antiglobulin Test
Ralph E. B. Green, BAppSc, FAIMLS, MACE, Jayanna Slayten, MS,
MT(ASCP)SBBCM and Justin R. Rhees, MS, MLS(ASCP)CMSBBcM

Introduction Direct Antiglobulin Test Sources of Error


History of the Antiglobulin Test Principle and Application Modified and Automated Antiglobulin
Antihuman Globulin Reagents DAT Panel Test Techniques
Polyspecific AHG Evaluation of a Positive DAT Solid-Phase Technology
Monospecific AHG Indirect Antiglobulin Test Gel Test
Preparation of AHG Factors Affecting ttie Antiglobulin Test Comparison of AHG Methodologies
Polyspecific AHG Ratio of Serum to Cells Case Studies
Monospecific AHG Reaction Medium Case 5-1
Antibodies Required in AHG Temperature Case 5-2
Anti-lgG lncubatio Time Summary Chart
Anticomplement Washing of RBCs Review Questions
Use of Polyspecific Versus Monospecific Saline for Washing References
AHG in the IAT Addition of AHG
Principles of the Antiglobulin Test Centrifugation for Reading

OBJECTIVES
1. State the principle of the antiglobulin test.
2. Differentiate monoclonal from polyclonal and monospecific from polyspecific antihuman globulin (AHG) reagents.
3. Describe the preparation of monoclonal and polyclonal AHG reagents.
4. List the antibody requirements for AHG reagents.
5. Explain the use of polyspecific versus monospecific AHG in the indirect antiglobulin test (IAT).
6. List the advantages and disadvantages of anticomplement activity in polyspecific AHG.
7. Compare and contrast the IAT and the direct antiglobulin test (DAT).
8. Explain the principle and applications of red blood cell sensitization.
9. Explain the reasons for the procedural steps in the DAT and IAT.
10. Interpret the results of a DAT and IAT panel.
Continued

103
6888_Ch05_103-118 29/10/18 4:46 PM Page 104

104 PART II Blood Groups and Serologic Testing

OBJECTIVES—cont’d
11. Identify the factors that affect the antiglobulin test.
12. Formulate the steps necessary to resolve errors associated with the performance of the antiglobulin test.
13. Describe the multiple methodologies utilized in AHG testing.

Introduction to avoid prozone interference, the AHG serum still retained


sufficient antibody activity to permit cross-linking of adja-
The antihuman globulin (AHG) test, which is also referred cent RBCs sensitized with IgG antibodies. The cross-linking
to as the Coombs’ test,1,2 is based on the principle that of sensitized RBCs by AHG produced hemagglutination,
antihuman globulins obtained from immunized nonhuman indicating that the RBCs had been sensitized by an antibody
species bind to human globulins such as IgG or comple- that had reacted with an antigen present on the cell surface.
ment, either free in serum or attached to antigens on red Early AHG reagents were prepared using a crude globulin
blood cells (RBCs). The AHG test is an essential testing fraction as the immunogen. In 1947, Coombs and Mourant
methodology for transfusion medicine. demonstrated that the antibody activity that detected Rh
There are two major types of blood group antibodies: antibodies was associated with the anti-gamma globulin
IgM and IgG. Because of their large pentameric structure, IgM fraction in the reagent.5 In 1951, Dacie6 presented the first
antibodies bind to corresponding antigen and directly agglu- evidence that there might be another antibody activity
tinate RBCs suspended in saline. Some IgG antibodies are present that influenced the final reaction. He observed that
termed nonagglutinating, or incomplete antibodies, because different reaction patterns were obtained when dilutions of
their single monomer structure is too small to directly agglu- AHG were used to test cells sensitized with warm (IgG,
tinate sensitized RBCs (refer to Chapter 3, “Fundamentals of 37°C) as compared with cold antibodies (IgM, <22°C). In
Immunology”). Adding AHG reagent containing anti-IgG to 1957, Dacie and coworkers7 published data showing that the
RBCs sensitized with IgG antibodies allows for hemaggluti- reactivity of AHG to cells sensitized with warm antibodies
nation of these sensitized cells. An anti-antibody is used resulted from anti-gamma globulin activity, whereas anti-
to observe the formation of an Ag/Ab complex that would nongamma globulin activity was responsible for the activity
otherwise go undetected.3 Some blood group antibodies have of cells sensitized by cold antibodies. The nongamma globu-
the ability to bind complement to the RBC membrane. In lin component was shown to be beta globulin and had
such cases, an anticomplement component can be added to specificity for complement. Later studies8,9 revealed that the
the AHG reagent, rendering it polyspecific (anti-IgG and anti- complement activity was a result of C3 and C4.
complement). Antiglobulin tests detect IgG or complement- The antiglobulin test can be used to detect RBCs sensi-
sensitized RBCs. tized with IgG alloantibodies, IgG autoantibodies, and com-
plement components. Sensitization can occur either in vivo
History of the Antiglobulin Test or in vitro. The use of AHG to detect in vitro sensitization
of RBCs is a two-stage technique referred to as the indirect
Before the antiglobulin test was developed, only IgM
antiglobulin test (IAT). In vivo sensitization is detected by
antibodies were detected by direct testing methods. The
a one-stage procedure called the direct antiglobulin test
introduction of the AHG test permitted the detection of
(DAT). The IAT and DAT still remain the most common pro-
nonagglutinating IgG antibodies and led to the discovery and
cedures performed in blood group serology.10
characterization of many new blood group systems.
In 1945, Coombs, Mourant, and Race1 described the use
Antihuman Globulin Reagents
of the antiglobulin test for the detection of weak and non-
agglutinating Rh antibodies in serum. In 1946, Coombs and Several AHG reagents have been defined by the U.S. Food
coworkers2 described the use of AHG to detect in vivo and Drug Administration (FDA) Center for Biologics Evalu-
sensitization of the RBCs of neonates suffering from ation and Research (CBER). These are listed in Table 5–1
hemolytic disease of the fetus and newborn (HDFN). The and are described in the following paragraphs. Antihuman
test was originally called the Coombs’ test but later was globulin reagents can be polyspecific or monospecific.
called the direct antiglobulin test (DAT).3 Although Coombs
and associates1 were instrumental in introducing the Polyspecific AHG
antiglobulin test to blood group serology, the principle of the
test had in fact been described by Moreschi4 in 1908. Polyspecific AHG reagents contain antibody to human IgG
Moreschi’s studies used rabbit anti-goat serum to agglutinate and to the C3d component of human complement. Other
rabbit RBCs that were sensitized with low nonagglutinating anticomplement antibodies, such as anti-C3b, may also be
doses of goat anti-rabbit RBC serum. present. Therefore, its use can facilitate agglutination when
The Coombs’ test involved the injection of human serum RBCs have been sensitized with IgG or C3d or both. Com-
into rabbits to produce antihuman serum. After absorption mercially prepared polyspecific AHG contains little, if any,
to remove heterospecific antibodies and after dilution studies activity against IgA and IgM heavy chains. However, the
6888_Ch05_103-118 29/10/18 4:46 PM Page 105

Chapter 5 The Antiglobulin Test 105

Table 5–1 FDA Licensed Antihuman Globulin Reagents


Reagent Definition
Polyspecific

1. Rabbit polyclonal Contains anti-lgG and anti-C3d (may contain other anticomplement and other anti-immunoglobulin antibodies)

2. Rabbit/murine monoclonal blend Contains a blend of rabbit polyclonal antihuman IgG and Anti-C3d is a murine monoclonal IgM antibody.

Monospecific

1. Anti-IgG Contains anti-IgG with no anticomplement activity


(Rabbit polyclonal)

2. Anti-IgG Murine monoclonal IgM antibody secreted by a hybridoma cell line


(Gamma-clone AHG)
3. Anticomplement

Anti-C3d The main component of this reagent is a murine monoclonal antibody to C3d. Anti-C3d will cause the aggluti-
nation of red blood cells coated with human C3d and/or C3b complement components.

Accessed From: https://www.fda.gov/BiologicsBloodVaccines/BloodBloodProducts/ApprovedProducts/LicensedProductsBLAs/BloodDonorScreening/BloodGroupingReagent/


default.htm

polyspecific mixture may contain antibody activity to kappa human complement components injected into a rabbit result
and lambda light chains common to all immunoglobulin in anticomplement. This type of response produces a poly-
classes, thus reacting with IgA or IgM molecules.10 clonal antiglobulin serum. Polyclonal antibodies are a
mixture of antibodies from different plasma cell clones. The
Monospecific AHG resulting polyclonal antibodies recognize different antigenic
determinants (epitopes), or the same portion of the antigen
Monospecific AHG reagents contain only one antibody but with different affinities. Hybridoma technology can be
specificity: either anti-IgG or antibody to specific components used to produce monoclonal antiglobulin serum. Mono-
of complement, such as C3b or C3d (i.e., anticomplement). clonal antibodies are derived from one clone of plasma cells
Licensed monospecific AHG reagents in common use are and recognize a single epitope.
anti-IgG and anti-C3b,-C3d.10
Polyspecific AHG
Anti-IgG
Reagents labeled anti-IgG contain no anticomplement activ- Polyspecific AHG can be made using polyclonal or mono-
ity. Anti-IgG reagents contain antibodies specific for the Fc clonal antibodies. The two types of antibody production
fragment of the gamma heavy chain of the IgG molecule. If processes are very different from one another, yielding two
not labeled “gamma heavy chain–specific,” anti-IgG may very different advantages and disadvantages in their usage.
contain anti–light chain specificity and may therefore react
with cells sensitized with IgM, IgA, and IgG.11 Polyclonal AHG Production
Polyclonal AHG is usually prepared in rabbits. In contrast
Anticomplement with the early production methods, in which a crude globu-
Anticomplement reagents, such as anti-C3b,-C3d reagents, lin fraction of serum was used as the immunogen, modern
are reactive against only the designated complement compo- production commences with the purification of the immuno-
nents and contain no activity against human immunoglobu- gen from a large pool of normal sera.
lins.11 Monospecific anticomplement reagents are often Conventional polyspecific antiglobulin reagents are pro-
a blend of monoclonal anti-C3b and monoclonal anti-C3d duced by immunizing one colony of rabbits with human
(see the following sections for descriptions of monoclonal immunoglobulin (IgG) antigen and another colony with
and polyclonal antibody production). human C3 antigen. Because of the heterogeneity of IgG mol-
ecules, using serum from many donors to prepare the pooled
Preparation of AHG IgG antigen to immunize the rabbits and the pooling of anti-
IgG from many immunized rabbits is essential in producing
The classic method of AHG production involves injecting polyclonal reagents for routine use that are capable of de-
human serum or purified globulin into laboratory animals tecting the many different IgG antibodies. This is an advan-
such as rabbits. The human globulin behaves as foreign anti- tage of using anti-IgG of polyclonal origin for antiglobulin
gen, the rabbit’s immune response is triggered, and an anti- serum.12
body to human globulin is produced. For example, human Both colonies of animals are hyperimmunized to produce
IgG injected into a rabbit results in anti-IgG production; high-titer, high-avidity IgG antibodies. Blood specimens are
6888_Ch05_103-118 29/10/18 4:46 PM Page 106

106 PART II Blood Groups and Serologic Testing

drawn from the immunized animals, and if the antibody potency of anti-C3b and anti-C3d individually is one of the
potency and specificity meet predetermined specifications, difficulties with polyclonal reagents that can be avoided with
the animals are bled for a production batch of reagent. monoclonal products.12
Separate blends of the anti-IgG and anticomplement anti-
bodies are made, and each pool is then absorbed with A1, B, Monoclonal AHG Production
and O cells to remove heterospecific antibodies. The prepa- The monoclonal antibody technique devised by Kohler and
ration of polyclonal AHG is diagrammed in Figure 5–1. The Milstein13 has been used to produce AHG and has proved par-
total antibody content of each pool is determined, and the ticularly useful in producing high-titer antibodies with well-
potency of the pools is analyzed to calculate the optimum defined specificities to IgG and to the fragments of C3.14–16
antibody dilution for use. Block titrations for anti-IgG pools Monoclonal antibody production begins with the immu-
are performed by reacting dilutions of each antibody against nization of laboratory animals, usually mice, with purified
cells sensitized with different amounts of IgG. This is a human globulin. After a suitable immune response, mouse
critical step in the manufacturing process because excess spleen cells containing antibody-secreting lymphocytes
antibody (especially anti-IgG) may lead to prozone, which are fused with myeloma cells. The resulting hybridomas
leads to false-negative test results. (lymphocyte–myeloma hybrid cells) are screened for anti-
Because it is not possible to coat cells with measured bodies with the required specificity and affinity. The antibody-
amounts of components of complement, the potency of anti- secreting clones may then be propagated in tissue culture
C3 pools is measured using at least two examples each of a or by inoculation into mice, in which case the antibody is
C3b- and C3d-coated cell. Both anti-C3b and anti-C3d are collected as ascites (fluid accumulation in the abdomen of the
present in the polyclonal anti-C3 pool. The level of anti-C3d mouse). Because the clonal line produces a single antibody,
is critical in keeping false-positive tests to a minimum yet there is no need for absorption to remove heterospecific anti-
also detecting clinically significant amounts of RBC-bound bodies. A hybridoma is a hybrid cell that is used as the basis
C3d. Additionally, if the dilution of the anti-C3 pool is for the production of large amounts of antibodies for thera-
determined on the basis of the amount of anti-C3d present, peutic, research, or diagnostic applications.
the level of anti-C3b varies. The inability to determine the All antibody molecules produced by a clone of hybridoma
cells are identical in terms of antibody structure and antigen
specificity. This has advantages and disadvantages in AHG
Conventional Method production. Once an antibody-secreting clone of cells has
Polyclonal Antihuman Globulin been established, antibody with the same specificity and re-
action characteristics will be available indefinitely. This allows
Complement IgG
the production of a consistently pure and uncontaminated
Rabbit is injected with AHG reagent. The disadvantage is that all antibodies pro-
pooled donor antigen duced by a clone of cells recognize a single epitope present
on an antigen. For antigens composed of multiple epitopes
such as IgG, several different monoclonal antibodies reacting
with different epitopes may need to be blended, or a mono-
clonal antibody specificity for an epitope on all variants of a
particular antigen may need to be selected to ensure that all
different expressions of the antigen are detected.
Monoclonal antibodies to human complement compo-
nents anti-C3b and anti-C3d may be blended with polyclonal
anti-IgG from rabbits to achieve potent reagents that give
fewer false-positive reactions as a result of anticomplement.
Antibody collected
from multiple rabbits Immucor, Inc., manufactures AHG reagents from an entirely
and purified monoclonal source. The anti-IgG component is produced by
Monospecific Monospecific exposing mice to RBCs coated with IgG. The resulting anti-
Polyclonal Polyclonal IgG component is a murine monoclonal IgM antibody that
Anti-C3b,d Anti-IgG reacts with the CH3 domain of the Fc region of human IgG
subclasses 1, 2, and 3. The antibody does not react with
human antibodies of the IgG4 subclass, but these are not con-
sidered to be clinically significant. Blending the monoclonal
AHG Polyspecific
Polyclonal Blend anti-IgG with a monoclonal anti-C3b and monoclonal anti-
C3d results in a polyspecific AHG reagent. The preparation
Figure 5–1. Preparation of polyclonal AHG reagents. Pooled donor antigen
of monoclonal AHG is diagrammed in Figure 5–2.
allows for a broader spectrum of reactivity, but the source of antibody is limited to
the life span of the inoculated animal. Polyspecific antihuman globulin may be Before the AHG is available for purchase, manufacturers
manufactured by combining polyclonal anti-IgG with either polyclonal or monoclonal must subject their reagents to an evaluation procedure, and
anticomplement components. the results must be submitted to the FDA for approval.
6888_Ch05_103-118 29/10/18 4:46 PM Page 107

Chapter 5 The Antiglobulin Test 107

Hybridoma Technology
Monoclonal Antihuman Globulin

Complement IgG

Mouse is injected with


purified human antigen

Antibody-producing
lymphocytes collected
from spleen

Myeloma Myeloma
cell cell
Fusion with
myeloma cell to
form hybridoma

Clones grown in
Monoclonal tissue culture to Monoclonal
Anti-C3b,d,g yield monoclonal Anti-IgG
Figure 5–2. Preparation of monoclonal AHG reagents. The production
of antibody is longer lasting than the polyclonal source, as the hybridoma antibodies
can live indefinitely. A monoclonal blend may be manufactured by blending
monoclonal anti-C3b, monoclonal anti-C3d, and monoclonal anti-IgG. Mono-
specific antihuman globulin reagents can be manufactured by conventional AHG Polyspecific
or hybridoma technology. Monoclonal Blend

Whether produced by the polyclonal or monoclonal tech- a mixture of IgG1 and IgG3 subclasses. Rarely, nonaggluti-
nique, the final polyspecific product is one that contains nating IgM antibodies may be found; however, they have
both anti-IgG and anticomplement activity at the correct been shown to fix complement and may be detected by anti-
potency for immediate use. The reagent also contains buffers, complement.17 While some antibodies to blood group anti-
stabilizers, and bacteriostatic agents and may be dyed green gens can have IgA components, the only RBC alloantibodies
for visual distinction in the laboratory reagent rack. that have been reported as being solely IgA have been exam-
ples of anti-Pr,18 and those antibodies were agglutinating.
Monospecific AHG IgA autoantibodies have been reported, although very
rarely.19 Therefore, anti-IgG activity must be present in the
Monospecific AHG production process is similar to that AHG reagent. Anti-IgM and anti-IgA activity may be present,
described above for polyspecific AHG; however, it contains but neither is essential. The presence of anti–light chain
only one antibody specificity. Monospecific anti-IgG is activity allows detection of all immunoglobulin classes.
produced as a monoclonal, polyclonal, or blended formula. IgG1, IgG2, IgG3, and IgG4 are the human IgG subclasses;
however, changes in the constant region has defined 29 isoal-
Antibodies Required in AHG lotypes to date.20 Monospecific IgG AHG has the advantage
of specificity, but there have been limitations/disadvantages
The need to use both polyspecific and monospecific AHG
noted based on the detection of IgG1–IgG4 and the reactivity
reagents has already been discussed. However, the reader
of isoallotypes. Gamma-clone anti-IgG (Immucor, Inc.,
should be aware of the specific requirements for the sub-
Norcross, GA), is the only FDA-licensed monoclonal IgG
classes and components of IgG and complement required
AHG in the United States.21 Gamma-clone detects IgG1,
in the reagent.
IgG2, and IgG3, but as described in the package insert, it
Anti-IgG fails to react with IgG4. This is not regarded as a significant
limitation since IgG4-only antibody specificities are uncom-
AHG must contain antibody activity to nonagglutinating mon, have not been associated with acute hemolytic reac-
blood group antibodies. The majority of these antibodies are tions, and often have high-titer low-avidity-type features
6888_Ch05_103-118 29/10/18 4:46 PM Page 108

108 PART II Blood Groups and Serologic Testing

associated with clinically insignificant reactivity.21 Howie Table 5–2 Antibodies Capable of Binding
and workers22 also reported in 2016 that the Gamma-clone
Complement
anti-IgG AHG failed to detect IgG4 and this presents a rare
risk of not detecting reported IgG4 only, anti-JMH23 and Most Some Rare
anti-Ch,24 reported to rarely cause transfusion reactions. The ABO Xga D
work of Howie et al.22 demonstrates further the limitation
of monoclonal anti-IgG, and further studies will continue to Lea LKE P1
improve the specificity of the anti-IgG produced. Leb Lan Lua
Jka Lub
Anticomplement
Jkb Kell
Some antibodies “fix” complement components to the RBC
membrane after complexing of the antibody with its corre- Sc1 Fya
sponding antigen. These antibodies are listed in Table 5–2 Co3 Fyb
and are described in more detail in Chapters 6 through 9.
The terms most, some, and rare in the table refer to antibodies Ge2 Coa
that bind complement the vast majority of the time, that Ge3 Cob
show variability in their ability to bind complement, and that
rarely bind complement, respectively. These membrane- Ii Dia
bound complement components can be detected by the P S
anticomplement activity in AHG.
As a result of studies published during the 1960s25–28 P1PK s
indicating the need for anticomplement activity in AHG to Vel Yta
allow the IAT to detect antibodies, a polyspecific reagent was
introduced. Evidence was also presented showing that the
presence of anticomplement activity would enhance the Use of Polyspecific Versus
reactions of clinically significant antibodies (e.g., anti-Fya Monospecific AHG in the IAT
and anti-K).25 For a review of complement activation, refer
to Chapter 3. Polyspecific AHG contains both anti-IgG activity and anti-
C3d activity. There is considerable debate among blood
bankers over the use of monospecific anti-IgG versus poly-
Advanced Concepts specific AHG for routine antibody detection and pretrans-
fusion testing. Because most clinically significant antibodies
During complement activation, C3 is split into two com-
detected during antibody screening are IgG, the most
ponents. C3b binds to the RBC membrane and is opsonic,
important function of polyspecific AHG is the detection of
whereas C3a passes into the fluid phase.29-30 Additional
IgG antibodies.
cofactors further cleave C3b, releasing C3c into the fluid
There have been numerous reports of clinically significant
phase, and leaving C3dg bound to the membrane. Tissue
RBC alloantibodies that were undetectable with monospe-
proteases then rapidly transform C3dg into C3d.31 The
cific anti-IgG but were detected with the anticomplement
ISBT/ICSH Joint Working Party32 considered anti-C3c to
component of AHG.36
be the most important anticomplement component because
of its limited capacity to cause nonspecific reactions. How-
ever, when RBCs are incubated with serum for longer than
15 minutes, the number of C3c determinants falls rapidly Advanced Concepts
because C3c is split off the C3bi molecule. This finding fur- Unfortunately, polyspecific AHG has also been associated
ther supports the use of anti-C3d in international reference with unwanted positive reactions that are not caused
reagents by the Joint Working Party. by clinically significant antibodies. To investigate these
The final degradation step has been shown to occur in variables, Petz and coworkers37 examined 39,436 sera
vivo33 and is a common occurrence in both warm and cold comparing monospecific anti-IgG with polyspecific AHG.
autoimmune hemolytic anemias. Engelfriet and others34 They also compared the albumin technique with low
have also shown that degradation of C3b to C3d can occur ionic strength solutions (LISS)–suspended RBCs. Four
in vitro, providing that the incubation period is greater than Jka antibodies were detected only with polyspecific
1 hour. In 1976, Garratty and Petz35 confirmed the need anti-IgG using albumin or LISS-suspended RBCs. An
for anti-C3d activity in AHG for use in the DAT. They also additional anti-Jka was detected only with polyspecific
confirmed Engelfriet’s observation that, given sufficient AHG when using LISS but not with albumin. Also, five
time, cell-bound C3b could be degraded to C3d in vitro. antibodies of anti-K, anti-Jka, and Fya specificities were
Refer back to Table 5-1 for a detailed listing of all AHG detected when using LISS, but not albumin, with both
reagents’ components. polyspecific AHG and anti-IgG. Their results concluded
6888_Ch05_103-118 29/10/18 4:46 PM Page 109

Chapter 5 The Antiglobulin Test 109

that some clinically significant antibodies are detected antibody to the C3d component of human complement,
with the anticomplement component of AHG but not with and polyspecific reagents that contain both anti-IgG and
anti-IgG. This is especially true for anti-Jka, a complement- anti-C3d activity.
binding IgG antibody often associated with delayed hemolytic AHG reacts with human globulin molecules, either bound
transfusion reactions. to RBCs or free in serum. Washed RBCs coated with human
Petz and others37 also determined the number of false- globulin are agglutinated by AHG.
positive reactions obtained when using polyspecific AHG
versus anti-IgG with LISS and albumin. False-positive The complete procedures for the direct and indirect
reactions were defined as those caused by antibodies with antihuman globulin tests can be found in Procedures
no definable specificity or by antibodies considered to be 5–1 and –2 on the textbook’s companion website.
clinically insignificant because of optimum reactivity at Figure 5–3 illustrates in vitro sensitization detected in the
cold temperatures (anti-I, anti-H, anti-P1, anti-M). Of the IAT and in vivo sensitization detected by the DAT.
unwanted positive reactions, 93% were caused by C3 on
the cells. The authors emphasize that, if the first step in Direct Antiglobulin Test
evaluating a weakly positive AHG reaction is to repeat
using the prewarmed technique, about 60% of the false- Described above are the overall principles of the antiglobulin
positive weak reactions become negative. test. Figure 5–3 depicts two variations of the AHG tests: the
In a 3-year study, Howard and associates38 found eight DAT and the IAT. The following section details the DAT’s
patients whose antibodies were detected primarily or solely principle, application, construct, and evaluation.
by AHG containing anticomplement activity. Seven of these
antibodies had anti-Jka or anti-Jkb specificity. Some of them Principle and Application
could be detected using homozygous Jka or Jkb cells
The direct antiglobulin test (DAT) detects in vivo sensitiza-
and an AHG containing only anti-IgG activity. Two of the
tion of RBCs with IgG or complement components. Clinical
anti-Jka antibodies were associated with delayed hemolytic
conditions that can result in in vivo coating of RBCs with
transfusion reactions. The complement-only Kidd antibod-
antibody or complement are the following:
ies represented 23% of all Kidd antibodies detected during
the study. The authors concluded that they would continue • Hemolytic disease of the fetus and newborn (HDFN)
to use polyspecific AHG reagent for routine compatibility • Hemolytic transfusion reaction (HTR)
testing. • Autoimmune and drug-induced hemolytic anemia (AIHA).
Table 5–3 lists the clinical application and in vivo sensi-
One must balance the advantage of detecting clinically tization detected for each situation.
significant complement-only antibodies with the disadvan- The DAT is not a required test in routine pretransfusion
tages resulting from using antiglobulin serum containing protocols. Eder40 tested the clinical utility of the DAT at a
anticomplement activity.36 Deciding to use the AHG large tertiary care hospital in Philadelphia. A retrospective
reagent for indirect tests is the prerogative of the individ- study was performed from 1999 to 2002. DATs with anti-
ual blood bank. Many blood banks have begun using IgG were performed on 15,662 pretransfusion patient sam-
monospecific anti-IgG for routine pretransfusion testing, ples; 15% were positive. Subsequent eluate testing revealed
citing cost containment measures necessitated by the high 76% were nonreactive, 9% were pan reactive, and 12% pas-
number of repeats versus the rarity of complement-only sively acquired ABO or D antibodies. Only one case
detected antibodies such as anti-Jka. Milam39 states that demonstrated an RBC antibody in the eluate that was not
rare clinical transfusion intolerance, when using monospe- detected in the serum, concluding that even in a tertiary
cific anti-IgG over polyspecific AHG reagents to screen care setting, the routine DAT is inefficient, yielding a
for unexpected antibodies and to test for blood group positive predictive value of 0.16%. Judd and coworkers
compatibility, offers reliability without interference from revealed similar findings on 65,049 blood samples in a
common and clinically insignificant IgM-complement 29-month period, where only 5.5% of samples resulted in
fixing antibodies. a positive DAT.41

DAT Panel
Principles of the Antiglobulin Test
Initial DATs include testing one drop of a 3% to 5% suspen-
The antiglobulin test is based on the following simple
sion of washed RBCs with polyspecific (anti-IgG, anti-C3d)
principles:10
reagent (please refer to the DAT procedure on the DavisPlus
• Antibody molecules and complement components are website). Positive results are monitored by a DAT panel using
globulins. monospecific anti-IgG and anti-C3d to determine the spe-
• Injecting an animal with human globulin stimulates the cific type of protein sensitizing the cell. In an effort to save
animal to produce antibody to the foreign protein (i.e., valuable tech time, some institutions run polyspecific and
AHG). Serologic tests employ a variety of AHG reagents monospecific reagents at one time as well as a saline control.
reactive with various human globulins, including anti-IgG The saline control serves to detect spontaneous agglutination
6888_Ch05_103-118 29/10/18 4:46 PM Page 110

110 PART II Blood Groups and Serologic Testing

IgG

Figure 5–3. The antihuman globulin (AHG) test. (A) Indirect AHG (IAT), with sensitization occurring in vitro, looking for unknown antibody in the patient’s serum/plasma.
(Note: If the IAT was identifying RBC antigens, the RBCs would be from the patient and the antibody would be the known reagent). (B) Direct AHG (DAT) with sensitization
already occurring in vivo. (Note: The polyspecific AHG reagent is a mixture of anti-IgG and anti-C3d.)

Table 5–3 Direct Antiglobulin Test Table 5–4 DAT Panel: Patterns of
Reactivity in Autoimmune
Clinical Application In Vivo Sensitization
Hemolytic Anemia*
HDFN Maternal antibody coating fetal RBCs
Anti-IgG Anti-C3d Type of AIHA
HTR Recipient antibody coating donor RBCs
+ + WAIHA
AIHA Autoantibody coating individual’s RBCs
+ _ WAIHA

_ + CAS; PCH, WAIHA


of cells or reactions occurring without the addition of AHG + + Mixed-type AIHA (cold and warm)
reagents. In warm AIHA, including drug-induced hemolytic
anemia, the RBCs may be coated with IgG or C3d, or both. Modified from Fung MK, et al. Technical Manual, 19th ed. AABB, Bethesda, MD, 2017.
Patterns of reactivity and the type of protein sensitization in *The DAT with monospecific antiglobulin reagents is helpful in classifying AIHAs. Other
AIHA are summarized in Table 5–4. procedures and studies are necessary to diagnose and characterize which form of
autoimmune disease is present.
In a transfusion reaction workup, the DAT may detect CAS = cold agglutinin syndrome; PCH = paroxysmal cold hemoglobinuria;
the presence of IgG or C3d, or both, depending on the WAIHA = warm autoimmune hemolytic anemia
6888_Ch05_103-118 29/10/18 4:46 PM Page 111

Chapter 5 The Antiglobulin Test 111

nature and specificity of the recipient’s antibody. In the drug therapy, and recent transfusion history. A positive DAT
investigation of HDFN, testing for complement proteins is may occur without clinical manifestations of immune-
not necessary inasmuch as the protein sensitizing the new- mediated hemolysis. Table 5–5 outlines the in vivo phenom-
born RBCs is presumed to be maternal IgG. Problems can ena that may be associated with a positive DAT.
arise in accurate D typing in the case of a newborn with a The AABB Technical Manual states that “ a positive DAT
positive DAT. If the DAT is positive due to IgG and the im- result alone is not diagnostic of hemolytic anemia. Under-
mediate spin for D typing is negative, a test for weak D standing the significance of this positive result requires
cannot be performed. The same is true for a patient with knowledge of the patient’s diagnosis; recent drug, pregnancy,
AIHA due to a warm IgG antibody coating the patient transfusion, and hematopoietic transplantation history;
cells. The antibody must be removed from the RBCs for and the presence of acquired or unexplained hemolytic
accurate phenotyping. Other techniques can be used to anemia.”11 Answering the following questions before inves-
remove antibody from the patient’s RBCs. These include tigating a positive DAT for patients other than neonates will
chloroquine diphosphate, EDTA-glycine, and murine help determine what further testing is appropriate:
monoclonal antibodies.
• Is there evidence of in vivo hemolysis?
Evaluation of a Positive DAT • Has the patient been transfused recently? If so, did the
patient receive blood products or components containing
Clinical consideration should dictate the extent to which a ABO-incompatible plasma?
positive DAT is evaluated. Interpreting the significance of a • Does the patient’s serum contain unexpected antibodies?
positive DAT requires knowledge of the patient’s diagnosis, • Is the patient receiving any drugs?

Table 5–5 In Vivo Phenomena Associated With a Positive DAT


Condition Cause
Transfusion 1. Recipient alloantibody and Alloantibodies in the recipient of a recent transfusion that
donor antigen react with antigen on donor RBCs

2. Donor antibody and recipient Antibodies present in donor plasma that react with antigen
antigen on a transfusion recipient’s RBCs

Drug Induced 1. Type I (hapten-dependent Ab) Drug binds covalently to membrane proteins and stimu-
lates hapten-dependent Ab
2. Type II (autoantibody) Drug induces autoantibody specific for RBC membrane
proteins through unknown mechanism; Ab reacts with
normal RBCs in the absence of drug.

3. Type III (drug-dependent Ab) Drug induces Ab that binds to RBC only when drug is
present in soluble form, unknown mechanism; Ab reacts
with normal RBCs when soluble drug is present.

Autoimmune Hemolytic Anemia 1. WAIHA (IgG and/or C3) Autoantibody reacts with patient’s RBCs in vivo.

2. CAS (C3) Cold-reactive IgM autoagglutinin binds to RBCs in periph-


eral circulation (32°C). IgM binds complement as RBCs
return to warmer parts of circulation; IgM dissociates,
leaving RBCs coated only with complement.

3. PCH (IgG) The IgG autoantibody reacts with RBCs in colder parts of
body, causes complement to be bound irreversibly to
RBCs, and then elutes at warmer temperature.

Hemolytic disease of fetus and newborn Maternal alloantibody crosses Maternal (IgG) alloantibody, specific for fetal antigen,
placenta (IgG) coats fetal RBCs. DAT is reactive with anti-IgG.

Miscellaneous 1. Absorbed proteins; administra- Heterophile antibodies that are present in ALG or ATG coat
tion of equine preparations of recipient’s RBCs. High levels of protein causing RBCs to
antilymphocyte globulin (ALG) spontaneously agglutinate.
and antithymocyte globulin (ATG)

2. Administration of high-dose Nonantibody-mediated binding of immunoglobulin to


IV gamma globulin and RBCs in patients with hypergammaglobulinemia
hypergammaglobulinemia

Modified from Fung MK, Grossman BJ, Hillyer CD and Westhoff CM (eds): Technical Manual, 18th ed. AABB, Bethesda, MD, 2014.
AIHA = autoimmune hemolytic anemia; CAS = cold agglutinin syndrome; PCH = paroxysmal cold hemoglobinuria
6888_Ch05_103-118 29/10/18 4:46 PM Page 112

112 PART II Blood Groups and Serologic Testing

• Is the patient receiving antilymphocyte globulin or Table 5–7 Tasks and Purposes of the
antithymocyte globulin?
Indirect Antiglobulin Test
• Is the patient receiving intravenous immune globulin
(IVIG) or intravenous Rh immune globulin (IV RhIG)? Task Purpose
• Has the patient received a marrow or other organ transplant? Incubate RBCs with antisera Allows time for antibody molecule
attachment to RBC antigen
Indirect Antiglobulin Test Perform a minimum of three Removes free globulin molecules
saline washes
The IAT is performed to determine in vitro sensitization of
RBCs and is used in the following situations: Add antiglobulin reagent Forms RBC agglutinates (RBC Ag +
Ab + anti-IgG)
• Detection of incomplete (nonagglutinating) antibodies to
potential donor RBCs (compatibility testing) or to screen- Centrifuge Accelerates agglutination by bring-
ing cells (antibody screen) in serum ing cells closer together
• Determination of RBC phenotype using known antisera Examine for agglutination Interprets test as positive or
(e.g., weak D, any other antigen testing that requires IAT) negative
• Titration of incomplete antibodies
Grade agglutination reactions Determines the strength of reaction
Table 5–6 lists the IATs and the in vitro sensitization
Add antibody-coated RBCs to Checks for neutralization of anti-
detected for each application. For in vitro antigen-antibody negative reactions sera by free globulin molecules
reactions, the IAT tasks are listed and explained in Table 5–7. (Coombs’ control cells are
The DAT does not require the incubation phase because D-positive RBCs coated with
of the antigen-antibody complexes formed in vivo. anti-D)

Factors Affecting the Antiglobulin Test


The DAT can detect a level of 100 to 500 IgG molecules per • Addition of AHG
RBC and 400 to 1,100 molecules of C3d per RBC.34 • Centrifugation for reading
For the IAT, there must be between 100 and 200 IgG or
C3 molecules on the cell to obtain a positive reaction. The Ratio of Serum to Cells
number of IgG molecules that sensitize an RBC and the rate
at which sensitization occurs can be influenced by several Increasing the ratio of serum to cells increases the sensitiv-
factors, including the following: ity of the test system. Generally, a minimum ratio can be
achieved by using 2 drops of serum and 1 drop of a 5% vol-
• Ratio of serum to cells ume of solute per volume of solution (v/v) suspension of
• Reaction medium cells.39 When using cells suspended in saline, it is often
• Temperature advantageous to increase the ratio of serum to cells in an
• Incubation time effort to detect weak antibodies (e.g., 4 drops of serum with
• Washing of RBCs 1 drop of a 3% [v/v] cell suspension will give a ratio of
• Saline for washing 133:1).

Reaction Medium
Table 5–6 Indirect Antiglobulin Test
Reaction mediums include albumin, LISS, and polyethylene
In Vitro glycol.
Application Tests Sensitization
Albumin
Antibody detection Compatibility Recipient antibody react-
testing ing with donor cells The macromolecules of albumin allow antibody-coated
cells to come into closer contact with each other so that
Antibody Antibody reacting with
agglutination occurs. In 1965, Stroup and MacIlroy42 re-
screening screening cells
ported on the increased sensitivity of the IAT if albumin
Antibody Antibody panel Antibody reacting with was incorporated into the reaction medium. Their reaction
identification panel cells mixture, consisting of 2 drops of serum, 2 drops of 22%
Antibody titration Rh antibody Antibody and selected (w/v) bovine albumin, and 1 drop of 3% to 5% (v/v) cells,
titer Rh cells was shown to provide the same sensitivity at 30 minutes of
incubation as a 60-minute saline-only test. The use of
RBC phenotype RBC antigen Specific antisera + RBCs
albumin does not seem to provide any advantage over LISS
detection(ex: to detect antigen
weak D, K, Fy) techniques and adds to the cost of the test.42 Petz and
coworkers36 also showed that an albumin technique may
6888_Ch05_103-118 29/10/18 4:46 PM Page 113

Chapter 5 The Antiglobulin Test 113

miss several clinically significant antibodies. Therefore, it antibody to elute from the RBCs, decreasing the sensitivity
is rarely, if ever, used as an IAT media for routine tests. of the test.47 However, this could not be confirmed by Voak
and coworkers.48
Low Ionic Strength Solutions
Low ionic strength solutions (LISS) enhance antibody Washing of RBCs
uptake and allow incubation times to be decreased—from
30 to 60 minutes to 10 to 15 minutes—by reducing the When both the DAT and IAT are performed, RBCs must be
zeta potential surrounding the RBC. Some LISS also saline-washed a minimum of three times before adding AHG
contain macromolecular substances. The LISS technique, reagent. Washing the RBCs removes free unbound serum
introduced by Low and Messeter,43 has critical require- globulins. Inadequate washing may result in a false-negative
ments with respect to the serum-to-cell ratio. Moore and reaction because of neutralization of the AHG reagent by
Mollison44 showed that optimum reaction conditions were residual unbound serum globulins. The washing phase of a
obtained using 2 drops of serum and 2 drops of a 3% (v/v) DAT and IAT is one of the most important steps in testing.
suspension of cells in LISS. Increasing the serum-to-cell The wash phase can be controlled using check cells, or group
ratio increased the ionic strength of the reaction mixture, O cells sensitized with IgG.
leading to a decrease in sensitivity and counteracting the Washing should be performed immediately after being re-
shortened incubation time of the test. A LISS medium moved from the incubator and in as short a time as possible
may be achieved by either suspending RBCs in LISS or to minimize the elution of low-affinity antibodies. The cell
using a LISS additive reagent, with the latter being the button should be completely resuspended before adding the
more common practice. next saline wash. All saline should be discarded after the
final wash because residual saline dilutes the AHG reagent
Polyethylene Glycol and therefore decreases the sensitivity of the test. Centrifu-
Polyethylene glycol (PEG) is a water-soluble linear gation at each wash should be sufficient to provide a firm
polymer and is used as an additive to increase antibody cell button and therefore minimize the possible loss of cells
uptake. Its action is to remove water molecules surround- with each discard of saline.
ing the RBC (the water of hydration theory), thereby
effectively concentrating antibody. Anti-IgG is the AHG Saline for Washing
reagent of choice with PEG testing to avoid false-positive
reactions.11 Because PEG may cause aggregation of RBCs, Ideally, the saline used for washing should be fresh (sug-
reading for agglutination following 37°C incubation in the gested open expiration of 30 days) and buffered to a pH of
IAT is omitted. Several investigators45 compared the per- 7.2 to 7.4. Saline stored for long periods in plastic contain-
formance of PEG as an enhancement media with that of ers has been shown to decrease in pH, which may increase
LISS. Findings indicated that PEG increases the detection the rate of antibody elution during the washing process,
of clinically significant antibodies while decreasing detec- yielding a false-negative result.49 Changes in pH may have
tion of clinically insignificant antibodies. Barrett and important implications when monoclonal AHG is used,
associates46 reported that since PEG has been used for pre- inasmuch as monoclonal antibodies have been shown to
transfusion antibody screening, 6,353 RBC components have narrow pH ranges for optimum reactivity. Significant
have been transfused without any reported acute or levels of bacterial contamination in saline have been
delayed HTRs. reported;50 this situation can contribute to false-positive
results.
Temperature
Addition of AHG
The rate of reaction for the majority of IgG antibodies is
optimal at 37°C; therefore, this is the usual incubation tem- AHG should be added to the cells immediately after wash-
perature for the IAT. This is also the optimum temperature ing to minimize the chance of antibody eluting from the
for complement activation. cell and subsequently neutralizing the AHG reagent. The
volume of AHG added should be as indicated by the man-
Incubation Time ufacturers. However, Voak and associates51 have shown
that adding two volumes of AHG may overcome washing
For cells suspended in saline, incubation times may vary problems when low levels of serum contamination remain.
between 30 and 120 minutes. The majority of clinically These authors indicated that the neutralization of AHG
significant antibodies can be detected after 30 minutes of is a problem only with free IgG left in serum following
incubation, and extended incubation times are usually not inadequate saline washings and not with residual serum
necessary. If a LISS or PEG technique is being used,43,44 in- complement components. The complement fragments free
cubation times may be shortened to 10 to 15 minutes. With in serum are not the same as the complement fragments
these shortened times, it is essential that tubes be incubated bound to RBCs. Therefore, residual serum does not con-
at a temperature of 37°C. Extended incubation (i.e., up to tain C3b and C3d to neutralize the anti-C3b and anti-C3d
40 minutes) in the LISS technique has been shown to cause in AHG reagent.
6888_Ch05_103-118 29/10/18 4:46 PM Page 114

114 PART II Blood Groups and Serologic Testing

Centrifugation for Reading hemagglutination of the RBCs with the anti-IgG in the
AHG reagent. If no hemagglutination follows the addition
Centrifugation of the cell button for reading of hemagglu- of IgG-coated check cells, the test result is invalid and the
tination along with the method used for resuspending test must be repeated. The most common technical errors
the cells is a crucial step in the technique. The CBER- that result in failure to demonstrate hemagglutination after
recommended method for the evaluation of AHG uses the addition of IgG-coated RBCs are inadequate washing,
1,000 relative centrifugal forces (RCFs) for 20 seconds, nonreactive AHG reagent, and failure to add AHG reagent.
although the technique described in this chapter suggests Monospecific anti-C3d reactivity may be verified with the
500 RCFs for 15 to 20 seconds. The use of higher RCFs addition of C3d-coated RBCs to negative reactions.52
yields more sensitive results; however, depending on how
the button is resuspended, it may give weak false-positive Modified and Automated Antiglobulin
results because of inadequate resuspension, or may give a Test Techniques
negative result if resuspension is too vigorous. The opti-
mum centrifugation conditions should be determined for Modifications to the antiglobulin test technique (LISS, PEG,
each centrifuge. and albumin) have just been described; however, additional
modifications have historically been used, including the low-
Sources of Error ionic polybrene technique53-56 and the enzyme-linked
antiglobulin test (ELAT). Solid-phase and gel technologies
Some of the most common sources of error associated with are fully automated modifications now widely used in
the performance of the AHG test have been outlined in the immunohematology laboratories.
previous section. Box 5–1 lists reasons for false-positive and
false-negative AHG reactions. An anticoagulant such as Solid-Phase Technology
EDTA should be used to collect blood samples for the DAT
to avoid the in vitro complement attachment associated with Solid-phase technology may be used for performing
refrigerated clotted specimens.52 antiglobulin tests. Several different techniques have been re-
All negative antiglobulin test reactions must be checked ported using either test tubes57 or microplates.58,59 With the
by the addition of IgG-sensitized cells. Adding IgG-coated availability of microplate readers, this technology can be
RBCs to negative test reactions should demonstrate fully automated. Direct and indirect tests can be performed

BOX 5–1

Sources of Error in Antihuman Globulin Testing


False-Positive Results False-Negative Results
• Improper specimen (refrigerated, clotted) may cause in vitro • Inadequate or improper washing of cells
complement attachment • Failure to wash additional times when increased serum volumes are
• Overcentrifugation and overreading used
• Centrifugation after the incubation phase when PEG or other • Contamination of AHG by extraneous protein (i.e., glove, wrong
positively charged polymers are used as an enhancement medium dropper)
• Bacterial contamination of cells or saline used in washing • High concentration of IgG paraproteins in test serum
• Dirty glassware • Early dissociation of bound IgG from RBCs due to interruption in testing
• Presence of fibrin in the test tube may mimic agglutination. • Early dissociation of bound IgG from RBCs due to improper testing
• Cells with a positive DAT will yield a positive IAT. temperature (i.e., saline or AHG too cold or hot)
• Polyagglutinable cells • AHG reagent nonreactive because of deterioration or neutralization
• Saline contaminated by heavy metals or colloidal silica (improper reagent storage)
• Using a serum sample for a DAT (use EDTA, ACD, or CPD anticoagu- • Excessive heat or repeated freezing and thawing of test serum
lated blood) • Serum nonreactive because of deterioration of complement
• Samples collected in gel separator tubes may have unauthentic com- • AHG reagent, test serum, or enhancement medium not added
plement attachment. • Undercentrifuged or overcentrifuged
• Complement attachment when specimens are collected from infusion • Cell suspension either too weak or too heavy
lines infusing dextrose solutions • Serum-to-cell ratios are not ideal.
• Preservative-dependent antibody directed against reagents • Rare antibodies are present that are only detectable with polyspecific
AHG and when active complement is present.
• Low pH of saline
• Inadequate incubation conditions in the IAT
• Poor reading technique

Modified from Fung MK, et al. Technical Manual, 19th ed. AABB, Bethesda, MD, 2017.
6888_Ch05_103-118 29/10/18 4:46 PM Page 115

Chapter 5 The Antiglobulin Test 115

using solid-phase methodology. In the former, antibody Comparison of AHG Methodologies


is attached to a microplate well, and RBCs are added. If
antibody is specific for antigen on RBCs, the bottom of the Transfusion service departments typically work to detect all
well will be covered with suspension; if no such specificity clinically significant antibodies, both DAT and IAT types, and
occurs, RBCs will settle to the bottom of the well. In the as few clinically insignificant antibodies such as warm and
latter, known RBCs are bound to a well that has been treated cold-reacting autoantibodies. A common question that arises
with glutaraldehyde or poly L-lysine. Test serum is added to in these departments is which detection method should be
RBC-coated wells, and if antibody in serum is specific for employed to reach such goals. Chapter 12 describes various
antigen on fixed RBCs, a positive reaction occurs as previ- AHG methods for DAT and IAT testing. Table 5–8 outlines
ously described. For a detailed description of this technology, some of the advantages and disadvantages in various AHG
see Chapter 12, “Blood Bank Testing Technologies and testing methodologies.
Automation.”

Gel Test
CASE STUDIES
The gel test is a process that detects RBC antigen-antibody
Case 5-1
reactions by means of a chamber filled with polyacrylamide
gel. The gel acts as a trap; free unagglutinated RBCs form You are working second shift at a suburban hospital. Your
buttons in the bottom of the tube, whereas agglutinated laboratory utilizes the conventional tube testing method
RBCs are trapped in the tube for hours. Therefore, negative using LISS as an enhancement medium. About an hour
reactions appear as buttons in the bottom of the microtube, before the end of your shift, the phlebotomist brings you
and positive reactions are fixed in the gel. For a detailed a routine type and screen for a patient just admitted to
description of this technology, see Chapter 12.

Table 5–8 Comparison of AHG Methodologies


Testing Methodology Advantages Disadvantages
Saline-tube testing • No additives • Long incubation
• Reduced cost • Least sensitive
• Avoids reactivity with auto Abs • Requires highly trained staff
• Ability to assess multiple phases of reactivity • Most procedural steps
• Fewer method-dependent Abs detected

LISS-tube testing • Reduced cost • Inability to be automated


• Avoids reactivity with auto Abs • Requires highly trained staff
• Shortest incubation time • Many procedural steps
• Increased Ab uptake • Fewer method-dependent Abs detected
• Most common tube method
• Ability to assess multiple phases of reactivity

PEG-tube testing • Reduced cost • Requires highly trained staff


• Decreased incubation time • Many procedural steps
• Increased Ab uptake • Detects more unwanted Abs
• Enhances most Abs • Inability to be automated
• Ability to assess multiple phases of reactivity • Fewer method-dependent Abs detected
(not 37°C)

Gel • More sensitive DAT method • Warm auto Abs enhanced


• No washing steps • Mixed-cell agglutination with cold Abs
• No need for check cells • Increased costs
• Stable endpoints • Increased need for additional instrumentation
• Small test volume • Increased chances of detected unwanted Abs
• Enhanced anti-D detection
• Ability to be automated

Solid phase • No need for check cells • Increased sensitivity for all Abs
• Stable endpoints • Detects unwanted Abs
• Small test volume • Warm auto Abs enhanced
• Enhanced anti-D • Increased costs
• Increased sensitivity for all Abs • Increased need for additional instrumentation
• Ability to be automated

Ab = antibody; LISS = low ionic strength saline; PEG = polyethylene glycol


6888_Ch05_103-118 29/10/18 4:46 PM Page 116

116 PART II Blood Groups and Serologic Testing

the medical surgical floor. Because you are confident you is O-positive, and the blood bank technologist performed
can finish before your shift ends, you decide to proceed an antibody screen using the patient serum and a two-cell
with testing. The patient types as an A-positive. The anti- screen using the solid-phase automated platform. One of
body screen results were negative in all phases of testing. the cells in the antibody screen is weakly reactive (1+),
Therefore, check cells were added to all tubes and the so you proceed with an antibody identification using a
reactions after centrifuging were also negative. panel. However, only two cells are weakly reactive (1+)
and show no pattern of reactivity. A second panel is run
1. Can the antibody screen be interpreted as negative?
and yields similar results. You decide to run an antibody
2. What steps must be taken to resolve the problem?
identification panel using the conventional tube testing
3. What is the most likely cause for the discrepant
method with LISS as your enhancement medium. All
results?
reactions are negative using LISS.
4. What are other causes for the results?
1. Can the antibody screen be interpreted as negative?
Case 5-2 2. Provide an explanation for the observed results.
3. Are there additional tests that should be conducted to
A 44-year-old white female is admitted for an exploratory
complete testing on this patient? If so, which tests
laparotomy. A type and screen is ordered prior to her
should be run?
scheduled surgery. ABO and Rh typing reveal the patient

SUMMARY CHART
 The antiglobulin test is used to detect RBCs sensitized  The IAT detects in vitro sensitization of RBCs and can
by IgG alloantibodies, IgG autoantibodies, and com- be applied to compatibility testing, antibody screen,
plement components. antibody identification, RBC phenotyping, and titra-
 AHG reagents containing anti-IgG are needed for the tion studies.
detection of IgG antibodies because the IgG monomeric  A positive DAT is followed by a DAT panel using
structure is too small to directly agglutinate sensitized monospecific anti-IgG and anti-C3d to determine the
RBCs. specific type of protein sensitizing the RBC.
 Polyspecific AHG sera contain antibodies to human  EDTA should be used to collect blood samples for the
IgG and the C3d component of human complement. DAT to avoid in vitro complement attachment associ-
 Monospecific AHG sera contain only one antibody speci- ated with refrigerated, clotted specimens.
ficity: either anti-IgG or antibody to anti–C3b–C3d.  There are multiple sources of error that can be intro-
 Classic AHG sera (polyclonal) are prepared by inject- duced into the AHG procedure.
ing human globulins into rabbits, and an immune  LISS, PEG, polybrene, and albumin can all be used as
stimulus triggers production of antibody to human enhancement media for AHG testing, with each having
serum. its own advantages and disadvantages.
 Hybridoma technology is used to produce monoclonal  Conventional tube testing, gel technology, enzyme-
antiglobulin serum. linked technology, and solid-phase testing are available
 The DAT detects in vivo sensitization of RBCs with methods to use in AHG testing.
IgG or complement components. Clinical conditions  Method-dependent antibodies do exist and should be
that can result in a positive DAT include HDFN, HTR, evaluated on a case-by-case basis.
and AIHA.

2. Polyspecific AHG reagent contains:


Review Questions
a. Anti-IgG and anti-IgA.
1. A description of the antiglobulin test is: b. Anti-IgG and anti-IgM.
c. Anti-IgG and anti-C3d.
a. IgG and C3d are required for RBC sensitization.
d. Anti-IgA and Anti-C3d.
b. Human globulin is completely eluted from RBCs
during saline washings. 3. Monoclonal anti-C3d is:
c. Human globulin is injected into an animal. a. Derived from one clone of plasma cells.
d. AHG reacts with human globulin molecules bound to b. Derived from multiple clones of plasma cells.
RBCs. c. Derived from immunization of rabbits.
d. Reactive with C3b and C3d.
6888_Ch05_103-118 29/10/18 4:46 PM Page 117

Chapter 5 The Antiglobulin Test 117

4. Which of the following is a clinically significant 11. A positive DAT may be found in which of the following
antibody whose detection has been reported in some situations?
instances to be dependent on anticomplement activ- a. A weak D-positive patient
ity in polyspecific AHG? b. A patient with anti-M
a. Anti-Jka c. HDFN
b. Anti-Lea d. An incompatible crossmatch
c. Anti-P1
12. What do Coombs’ check cells consist of?
d. Anti-H
a. Type A-positive cells coated with anti-IgG
5. After the addition of IgG-coated RBCs (check cells) b. Type A-negative cells coated with anti-IgG
to a negative AHG reaction during an antibody c. Type O-positive cells coated with anti-D
screen, a negative result is observed. Which of the d. Type B-negative cells coated with anti-D
following is a correct interpretation based on these
findings? 13. Which of the following IAT methods requires the use of
check cells?
a. The antibody screen is negative.
b. The antibody screen cannot be interpreted. a. Manual tube method with albumin
c. The saline washings were adequate. b. Gel
d. AHG reagent was added. c. Automated solid-phase analyzer
d. Enzyme-linked
6. RBCs must be washed in saline at least three times
before the addition of AHG reagent to: 14. Which uncontrollable factor can affect AHG testing?
a. Wash away any hemolyzed cells. a. Temperature
b. Remove traces of free serum globulins. b. Antibody affinity
c. Neutralize any excess AHG reagent. c. Gravitational force in the centrifuge
d. Increase the antibody binding to antigen. d. Incubation time

7. An in vivo phenomenon associated with a positive 15. Which would be the most efficient method for a labora-
DAT is: tory staffed by medical laboratory technicians?
a. Passive anti-D detected in the maternal sample. a. LISS
b. Positive antibody screen tested by LISS. b. Polybrene
c. Identification of alloantibody specificity using a c. Solid-phase or gel
panel of reagent RBCs. d. Enzyme-linked
d. Maternal antibody coating fetal RBCs. 16. A 27-year-old group O mother has just given birth to a
8. False-positive DAT results are most often associated group A baby. Since the mother has IgG anti-A, anti-B
with: and anti-A, B in her plasma, which of the following
methods and tests would be most effective at detecting
a. Use of refrigerated, clotted blood samples in which
the anti-A on the baby’s RBCs?
complement components coat RBCs in vitro.
b. A recipient of a recent transfusion manifesting an a. DAT using common tube technique
immune response to recently transfused RBCs. b. DAT using gel
c. Presence of antispecies antibodies from adminis- c. IAT using common tube technique
tration of immune globulin (IVIG). d. IAT using gel
d. A positive autocontrol caused by polyagglutination.
References
9. Polyethylene glycol (PEG) enhances antigen-
1. Coombs RRA, et al. A new test for the detection of weak and
antibody reactions by: “incomplete” Rh agglutinins. Br J Exp Pathol. 1945;26:255.
a. Decreasing zeta potential. 2. Coombs RRA, et al. In-vivo isosensitisation of red cells in babies
b. Concentrating antibody by removing water. with haemolytic disease. Lancet. 1946;i:264.
c. Increasing antibody affinity for antigen. 3. McCullough J. Transfusion medicine., 4th ed. Wiley-Blackwell
2016.
d. Increasing antibody specificity for antigen. 4. Moreschi C. Neue Tatsachen über die Blutkorperchen Aggluti-
10. Solid-phase antibody screening is based on: nationen. Zentralbl Bakteriol. 1908;46:49.
5. Coombs RRA, Mourant RA. On certain properties of antisera
a. Adherence. prepared against human serum and its various protein frac-
b. Agglutination. tions; their use in the detection of sensitisation of human red
c. Hemolysis. cells with incomplete Rh antibody, and on the nature of this
d. Precipitation. antibody. J Pathol Bacteriol. 1947;59(1-2):105-11.
6. Dacie JF. Differences in the behaviour of sensitized red cells to
agglutination by antiglobulin sera. Lancet. 1951;ii:954.
7. Dacie JV, et al. “Incomplete” cold antibodies: role of complement
in sensitization to antiglobulin serum by potentially haemolytic
antibodies. Br J Haematol. 1957;3:77.
6888_Ch05_103-118 29/10/18 4:46 PM Page 118

118 PART II Blood Groups and Serologic Testing

8. Harboe M, et al. Identification of the component of comple- 33. Brown DL, et al. The in vivo behaviour of complement-coated
ment participating in the antiglobulin reaction. Immunology. red cells: studies in C6-deficient, Ce-depleted and normal
1963;6:412. rabbits. Clin Exp Immunol. 1970;7:401.
9. Jenkins GC, et al. Role of C4 in the antiglobulin reaction. 34. Engelfriet CP, et al. Autoimmune haemolytic anemias: 111
Nature. 1960;186:482. preparation and examination of specific antisera against com-
10. U.S. Food & Drug Administration, Blood Grouping and plement components and products, and their use in serological
Phenotyping Reagents, Licensed Product Approval Information. studies. Clin Exp Immunol. 1970;6:721.
Available from: https://www.fda.gov/BiologicsBloodVaccines/ 35. Garratty G, Petz LD. The significance of red cell bound com-
BloodBlood Products/ApprovedProducts/LicensedProductsBLAs/ plement components in development of standards and quality
BloodDonorScreening/BloodGrouping/reagent/default.htm assurance for the anticomplement components of antiglobulin
11. Fung MK, editor. Technical manual. 19th ed. Bethesda (MD): sera. Transfusion. 1976;16:297.
AABB, 2017. 36. Petz LD, et al. Clinical practice of transfusion medicine.
12. Issitt C. Monoclonal antiglobulin reagents. Dade International 3rd ed. New York: Churchill Livingstone; 1996. p. 207.
Online, 1997. Available from www.dadeinternational.com/hemo/ 37. Petz LD, et al. Compatibility testing. Transfusion. 1981;21:633.
papers/monoanti.htm. 38. Howard JE, et al. Clinical significance of the anticomplement
13. Kohler G, Milstein C. Continuous cultures of fused cells component of antiglobulin antisera. Transfusion. 1982;22:269.
secreting antibody of predefined specificity. Nature. 1975; 39. Milam JD. Laboratory medicine parameter: utilizing monospe-
256:495. cific antihuman globulin to test blood group compatibility.
14. Lachman PJ, et al. Use of monoclonal antibodies to characterize Am J Clin Pathol. 1995;104:122.
the fragments of C3 that are found on erythrocytes. Vox Sang. 40. Eder AF. Evaluation of routine pre-transfusion direct antiglobulin
1983;45:367. test in a pediatric setting. Lab Med. 2003;24:680.
15. Holt PDJ, et al. NBTS/BRIC 8: a monoclonal anti-C3d antibody. 41. Judd WJ, et al. The evaluation of a positive direct antiglobulin
Transfusion. 1985;25:267. test in pre-transfusion testing revisited. Transfusion. 1986;26:220.
16. Voak D, et al. Monoclonal antibodies—C3 serology. Biotest 42. Stroup M, MacIlroy M. Evaluation of the albumin antiglobulin
Bull. 1983;1:339. technique in antibody detection. Transfusion. 1965;5:184.
17. Simon TL , McCullough J, Snyder EL, Solheim BG, Strauss RG 43. Low B, Messeter L. Antiglobulin test in low-ionic strength salt
(Editors). Rossi’s principles of transfusion medicine. 5th ed. Wiley- solution for rapid antibody screening and cross-matching. Vox
Blackwell 2016. Sang. 1974;26:53.
18. Garratty G, et al. An IgA high titre cold agglutinin with an 44. Moore HC, Mollison PL. Use of a low-ionic strength medium
unusual blood group specificity within the Pr complex. Vox in manual tests for antibody detection. Transfusion. 1976;
Sang. 1973;25:32. 16:291.
19. Petz LD, Garratty G. Acquired immune hemolytic anemias. 45. Shirley R, et al. Polyethylene glycol versus low-ionic strength
New York: Churchill Livingstone; 1980. p. 193. solution in pre-transfusion testing: a blinded comparison study.
20. Vidarsson G, et al. IgG subclasses and allotypes: from structure Transfusion. 1994;34:5.
to effector functions. Front Immunol. 2014;5:520. 46. Barrett V, et al. Analysis of the routine use of polyethylene glycol
21. Branch DR, Westhoff CM. Shining a light on AHG “blind (PEG) as an enhancement medium. Immunohematology.
spot(s). Transfusion. 2016;56:2913-15. 1995;11:1.
22. Howie HL, et al. Serological blind spots for variants of human 47. Jorgensen J, et al. The influence of ionic strength, albumin and
IgG3 and IgG4 by a commonly used anti-immunoglobulin incubation time on the sensitivity of indirect Coombs’ test. Vox
reagent. Transfusion. 2016;56:2953-62. Sang. 1980;36:186.
23. Geisland J, et al. An example of anti-JMH with characteristics 48. Voak D, et al. Low-ionic strength media for rapid antibody
of a clinically significant antibody. Immunohematology. 1990; detection: optimum conditions and quality control. Med Lab
6:9-11. Sci. 1980;37:107.
24. Westhoff CM, et al. Severe anaphylactic reactions following 49. Bruce M, et al. A serious source of error in antiglobulin testing.
transfusions of platelets to a patient with anti-Ch. Transfusion. Transfusion. 1986;26:177.
1985;32:34-38. 50. Green C, et al. Quality assurance of physiological saline used
25. Polley MJ, Mollison PL. The role of complement in the detec- for blood grouping. Med Lab Sci. 1968;43:364.
tion of blood group antibodies: special reference to the 51. Voak D, et al. Antihuman globulin reagent specification: the
antiglobulin test. Transfusion. 1961;1:9. European and ISBT/ICSH view. Biotest Bull. 1986;3:7.
26. Polley MJ, et al. The role of 19S gamma-globulin blood group 52. Mallory D, et al. Immunohematology methods and procedures.
antibodies in the antiglobulin reaction. Br J Haematol. Washington (D.C.): American National Red Cross; 1993.
1962;8:149. pp. 40–41.
27. Stratton F, et al. The preparation and uses of antiglobulin 53. Lalezari P, Jiang RF. The manual polybrene test: a simple and
reagents with special reference to complement fixing blood rapid procedure for detection of red cell antibodies. Transfusion.
group antibodies. Transfusion. 1962;2:135. 1980;20:206.
28. Stratton F, et al. Value of gel fixation on Sephadex G-200 in the 54. Crawford MN, et al. Microplate system for routine use in blood
analysis of blood group antibodies. J Clin Pathol. 1968;21:708. bank laboratories. Transfusion. 1970;10:258.
29. Lachman PJ, Muller-Eberhard HJ. The demonstration in 55. Redman M, et al. Typing of red cells on microplates by low-
human serum of “conglutinogen-activating-factor” and its ionic polybrene technique. Med Lab Sci. 1986;43:393.
effect on the third component of complement. J Immunol. 56. Liu JC, Wang Y, Liu FP, He YS. The manual polybrene test has
1968;100:691. limited sensitivities for detecting the Kidd blood group system.
30. Muller-Eberhard HJ. Chemistry and reaction mechanisms of Scand J Clin Lab Invest. 2009;69(7): 797–800.
complement. Adv Immunol. 1968;8:1. 57. Moore HH. Automated reading of red cell antibody identifica-
31. Merle NS, et al. Complement system part I—molecular mech- tion tests by a solid phase antiglobulin technique. Transfusion.
anisms of activation and regulation. Front Immunol. 2015; 1984;24:218.
6:262. 58. Plapp FV, et al. A solid phase antibody screen. Am J Clin
32. Case J, et al. International reference reagents: antihuman Pathol. 1984;82:719.
globulin, an ISBT/ICSH Joint Working Party Report. Vox Sang. 59. Lapierre Y, et al. The gel test: a new way to detect red cell
1999;77:121. antigen-antibody reactions. Transfusion. 1990;30:2.
6888_Ch06_119-148 31/10/18 10:55 AM Page 119

Chapter 6
The ABO Blood Group System
Denise M. Harmening, PhD, MT(ASCP) and Beth L. Manning,
BS, MT(ASCP)SBBCM

Introduction Comparison of A, B, and H Antigens ABO Discrepancies


Historical Perspective and Routine ABO on RBCs with A, B, and H Soluble Technical Errors
Testing Substances Resolution
ABO Antibodies ABO Subgroups Categories of ABO Discrepancies
Inheritance of the ABO Blood Groups A Subgroups Case Study
Formation of A, B, and H Red Blood Weak A Subgroups Case 6-1
Cell Antigens Weak B Subgroups Summary Chart
Interaction of Hh and ABO Genes The Bombay Phenotypes (Oh) Review Questions
Molecular Genetics of ABO The Para-Bombay Phenotypes References
Formation of A, B, and H Soluble ABH Antigens and Antibodies in Disease
Antigens

OBJECTIVES
1. Describe the reciprocal relationships between ABO antigens and antibodies for blood types O, A, B, and AB.
2. Identify the frequencies of the four major blood types in the white, black, Hispanic, and Asian populations.
3. Explain the effect of age on the production of ABO isoagglutinins.
4. Describe the immunoglobulin classes of ABO antibodies in group O, A, and B individuals.
5. Predict the ABO phenotypes and genotypes of offspring from various ABO matings.
6. Explain the formation of H, A, and B antigens on the red blood cells (RBCs) from precursor substance to immunodominant
sugars.
7. Describe the formation of H, A, and B soluble substances.
8. Explain the principle of the hemagglutination inhibition assay for the determination of secretor status.
9. Describe the qualitative and quantitative differences between the A1 and A2 phenotypes.
10. Describe the reactivity of Ulex europaeus with the various ABO groups.
11. Describe the characteristics of the weak subgroups of A (A3, Ax, Aend, Am, Ay, Ael).
12. Describe the characteristics of the Bombay phenotypes.
13. Explain the effects of disease on the expression of ABH antigens and antibodies.
14. Interpret the results from an ABO typing and resolve any discrepancies, if present.

Introduction through transfusion or pregnancy. Due to the presence of


these antibodies, transfusion of an incompatible ABO type
The ABO system is the most important of all blood groups may result in immediate lysis of donor RBCs. This produces
in both transfusion and transplant medicine.. It is the only a very severe, if not fatal, transfusion reaction in the patient.
blood group system in which individuals already have anti- Testing to detect ABO incompatibility between a donor and
bodies in their serum to antigens that are absent from their potential transfusion recipient is the foundation on which
red blood cells (RBCs) without any prior exposure to RBCs all other pretransfusion testing is based.
119
6888_Ch06_119-148 31/10/18 10:55 AM Page 120

120 PART II Blood Groups and Serologic Testing

Even today, transfusion of the wrong ABO group remains


a cause of death in hemolytic transfusion reaction fatalities BOX 6–1
reported to the FDA; however, transfusion-related acute lung Causes of Fatal Hemolytic Transfusion Reactions Due
injury (TRALI) was the most frequent cause of death in fiscal to ABO-Incompatible Blood Transfusions in FY 2015
year (FY) 20151 (Table 6–1). In FY 2015, there were two re-
• Case 1: Sample collected from improperly identified patient
ports of fatal hemolytic transfusion reactions due to ABO- (phlebotomist error)
incompatible blood product transfusions (Box 6–1 lists the • Case 2: A group B patient received a group A apheresis platelet.
causes in each of these two cases).1 This chapter presents the The platelet was later identified with a high anti-B titer of 1:2048.
ABO blood group system and discusses the biochemistry,
properties, and characteristics of ABO antigens and antibod- Data from 2015, F. R. (n.d.): Vaccines, Blood & Biologics; U.S. Food and Drug
ies. In addition, weak subgroups and common discrepancies Administration. Fatalities Reported to FDA Following Blood Collection and
Transfusion: Annual Summary for Fiscal Year 2015. Retrieved March 10, 2017,
will be introduced to provide a working knowledge for from www.fda.gov/BiologicsBloodVaccines/SafetyAvailability/ReportaProblem/
routine ABO testing. TransfusionDonationFatalities/ucm518148.pdf.

Historical Perspective and Routine


ABO Testing and B cells. Figure 6–2 outlines the steps of performing the
reverse ABO grouping (see color insert following page 128),
Karl Landsteiner truly opened the doors of blood banking and Table 6–3 summarizes the results of the procedures.
with his discovery of the first human blood group system, Table 6–4 lists the characteristics of the routine reagents used
ABO. This marked the beginning of the concept of individual for ABO testing in the blood bank laboratory.
uniqueness defined by the RBC antigens present on the RBC ABO grouping is the most frequently performed test in
membrane. In 1901, Landsteiner drew blood from himself the blood bank. Both ABO forward and reverse grouping
and five associates, separated the cells and serum, and then tests must be performed on all donors and patients.3 There
mixed each cell sample with each serum.2 He was inadver- is always an inverse reciprocal relationship between the for-
tently the first individual to perform forward and reverse ward and reverse type; thus, one serves as a check on the
grouping. Forward grouping (front type) is defined as using other. For example, if the individual has A antigens only on
known sources of commercial antisera (anti-A, anti-B) to de- their red blood cells, there will be an “expected” naturally
tect antigens on an individual’s RBCs. Figure 6–1 outlines occurring anti-B antibody in their serum since they lack the
the steps of performing the forward grouping for ABO (see B antigen.
color insert following page 128), and Table 6–2 lists the re- It has been postulated that bacteria, pollen particles, and
sults of the forward grouping procedure. Reverse grouping other substances present in nature are chemically similar to
(back type) is defined as detecting ABO antibodies in the A and B antigens. Bacteria are widespread in the environ-
patient’s serum by using known reagent RBCs, namely A1 ment, which constantly exposes individuals to A-like and

Table 6–1 Fatality Complication Breakdown by Imputability FY 2015


Doubtful/ Undetermined
Definite/ Probable/ Unlikely/ Ruled Out/ Not Assessable
Category Certain Likely Possible Improbable Excluded or Evaluable Total
TRALI** 5 N/A* 7 1 1 – 14

HTR** (non-ABO) 2 1 1 1 – – 5

HTR (ABO) 2 – – – – – 2

Contamination 3 – 2 – – 5
(Bacterial)

TACO** 3 6 2 – – 1 12

Allergy or 2 – – – – – 2
Anaphylaxis

Hypotensive – 1 – 1 – – 2
Reaction

*Definitions based on the Canadian Consensus Panel on TRALI.


** HTR = hemolytic transfusion reaction; TACO = transfusion-associated circulatory overload; TRALI = transfusion-related acute lung injury
Data from 2015, F. R. (n.d.): Vaccines, Blood & Biologics; U.S. Food and Drug Administration. Fatalities Reported to FDA Following Blood Collection and Transfusion: Annual Summary
for Fiscal Year 2015. Retrieved March 10, 2017, from www.fda.gov/BiologicsBloodVaccines/SafetyAvailability/ReportaProblem/TransfusionDonationFatalities/ucm204763.htm
6888_Ch06_119-148 31/10/18 10:55 AM Page 121

Chapter 6 The ABO Blood Group System 121

Table 6–2 ABO Forward Grouping: B-like antigens. This exposure serves as a source of stimula-
tion of anti-A and anti-B. All other defined blood group sys-
Principle—Detection of Antigens
tems do not regularly have “naturally occurring” antibodies
on Patient’s RBCs With Known
expected in their serum to antigens they lack on their RBCs.
Commercial Antisera Antibody production in most other blood group systems
Patient RBCs Patient RBCs Interpretation requires the introduction of foreign RBCs by either transfu-
with Anti-A with Anti-B of Blood Group sion or pregnancy, although some individuals can occasion-
ally have antibodies present that are not related to the
0 0 O
introduction of foreign RBCs. (These antibodies are usually
4+ 0 A of the IgM type and are not consistently present or expected
in everyone’s serum.) Performance of serum grouping is,
0 4+ B
therefore, unique to the ABO blood group system. The reg-
4+ 4+ AB ular occurrence of anti-A and/or anti-B in persons lacking
the corresponding antigen(s) serves as a confirmation of re-
+ = visual agglutination; 0 = negative sults in ABO grouping. Table 6–5 summarizes the forward
Note: Reaction gradings vary from patient to patient.
and reverse grouping for the common ABO blood groups.
The frequency of the ABO blood groups differs among
Table 6–3 ABO Reverse Grouping: selected populations and ethnic groups (Table 6–6).4 For
example, group B is found twice as frequently in blacks and
Principle—Detection of ABO
Asians as in whites. There is also a significant decrease in
Antibodies (Isoagglutinins) in
group A distribution in these two ethnic populations com-
Serum of Patient With Known pared to whites. It has been reported that subgroup A2 is
Commercial RBCs rarely found in Asians.5
Patient Serum Patient Serum
with Reagent with Reagent Interpretation ABO Antibodies
A1 Cells B Cells of Blood Group
Individuals normally produce antibodies directed against the
4+ 4+ O A and/or B antigen(s) absent from their RBCs. These anti-
0 3+ A bodies have been described as naturally occurring because
they are produced without any exposure to RBCs. The ABO
3+ 0 B antibodies are predominantly IgM, activate complement, and
0 0 AB react at room temperature or colder.5 ABO antibodies
produce strong direct agglutination reactions during ABO
+ = visual agglutination; 0 = negative testing. ABO antibody production is initiated at birth,
Note: Reaction gradings vary from patient to patient. but titers are generally too low for detection until infants are

Table 6–4 Characteristics of Routine Reagents Used for ABO Testing


Forward Grouping Anti-A Reagent Anti-B Reagent
• Monoclonal antibody* • Monoclonal antibody*

• Highly specific • Highly specific

• IgM • IgM

• Clear blue-colored reagent • Clear yellow-colored reagent (contains an acriflavine dye)

• Expected 3+ to 4+ reaction • Expected 3+ to 4+ reaction

• Usually use 1–2 drops • Usually use 1–2 drops

Reverse Grouping Reagent A1 and B Cells


• Human source
• 4%–5% RBC suspension
• Expected 2+ to 4+ reaction usually use 1 drop

*General rule: Always drop clear solutions first and RBCs second to make sure you have added both a source of antibody and antigen.
6888_Ch06_119-148 31/10/18 10:55 AM Page 122

122 PART II Blood Groups and Serologic Testing

Table 6–5 Summary of Forward and Reverse Groupings


Forward Group Patient’s Cells With Reagents Reverse Group Patient’s Serum With Reagents

Blood Antigen(S) Antibody(ies)


Group Anti-A Anti-B on RBCS A1 Cells B Cells in Serum
O 0 0 No A or B antigen 4+ 4+ A and B

A 4+ 0 A 0 2+ B

B 0 4+ B 3+ 0 A

AB 3+ 3+ A and B 0 0 No A or B antibodies

0 = negative (no agglutination); + = visual agglutination


Note: Reaction gradings vary from patient to patient.
*Percentages rounded to the nearest whole number

Table 6–6 ABO Phenotype Frequencies if group O serum is adsorbed with A or B cells, the antibody
eluted will react with both A and B cells.6,7 Anti-A,B antibody
of Ethnic Groups in the United
is not a combination of anti-A and anti-B but is a separate
States
“cross-reacting” antibody that is usually IgG in nature.5
U.S. Frequencies (%) (Rounded to the Nearest Knowing the amount of IgG anti-A, anti-B, or anti-A,B in
Whole Number) a woman’s serum sometimes allows prediction or diagnosis
Phenotype Whites Blacks Hispanic Asian** of hemolytic disease of the fetus and newborn (HDFN)
caused by ABO incompatibility.8 (See Chapter 20,
O 45 50 56 40 “Hemolytic Disease of the Fetus and Newborn [HDFN]”).
A 40 26 31 28 Both immunoglobulin classes of ABO antibodies react
preferentially at room temperature (20°C to 24°C) or below
B 11 20 10 25 and efficiently activate complement at 37°C.9
AB 4 4 3 7 Anti-A,B reagent is routinely used for performing ABO
confirmation of group O donor units simply because it is
*Hispanic includes Mexican, Puerto Rican, Cuban, and other Hispanics. more economical to use one reagent instead of both anti-A
**Asian includes Chinese, Filipino, Indian, Japanese, Korean, and Vietnamese. and anti-B.3 Use of anti-A,B reagent is not required for
Data from Garratty G, Glynn SA, McEntire R. ABO and Rh (D) phenotype frequencies of
different racial/ethnic groups in the United States. Transfusion. 2004;44:703–706. routine patient ABO testing.10 Some believe anti-A,B is more
effective at detecting weakly expressed A and B antigens than
reagent anti-A or anti-B. The production and use of mono-
clonal antisera, however, have made anti-A and anti-B
3 to 6 months old.5 Therefore, most antibodies found in cord reagents much more sensitive, to the point where weak A
blood serum are of maternal origin. Results of serum ABO and B antigens are detected routinely. Reagent anti-A,B can
testing before 3 to 6 months of age cannot be considered be prepared using blended monoclonal anti-A and anti-B;
valid because some or all of the antibodies present may polyclonal human anti-A,B; or a blend of monoclonal
be IgG maternal antibodies that crossed the placenta. As a anti-A, anti-B, and anti-A,B.3 Always consult the manufac-
result, it is logical to perform only forward grouping on cord turer’s product insert to determine if a reagent anti-A,B reacts
blood from newborn infants. with a specific weak A phenotype.
Antibody production peaks between 5 and 10 years of age
and declines later in life.5 Elderly people usually have lower Inheritance of the ABO Blood Groups
levels of anti-A and anti-B; therefore, antibodies may be
undetectable in the reverse grouping (see the “ABO Discrep- The theory for the inheritance of the ABO blood groups
ancies” section later in this chapter). ABO antibodies can was first described in 1924 by indicating that an individual
cause rapid intravascular hemolysis if the wrong ABO group inherits one ABO gene from each parent and that these two
is transfused, potentially resulting in patient death.1 genes determine which ABO antigens are present on the
Although anti-A (from a group B individual) and anti-B RBC membrane. The inheritance of ABO genes, therefore,
(from a group A individual) contain predominantly IgM follows simple Mendelian genetics. ABO, like most other
antibody, small quantities of IgG may also be present.5 Serum blood group systems, is codominant in expression.9 (For a
from group O individuals contains anti-A, anti-B, and review of genetics, see Chapter 2, “Basic Genetics.”) One
anti-A,B. Anti-A,B reacts with both A and B cells. Anti-A,B position, or locus, on each chromosome 9 is occupied by
antibody activity, originally thought to be just a mixture an A, B, or O gene.11,12 The O gene is considered an
of anti-A and anti-B, cannot be separated into a pure speci- amorph, as no detectable antigen is produced in response
ficity when adsorbed with either A or B cells.6,7 For example, to the inheritance of this gene. The group O phenotype is
6888_Ch06_119-148 31/10/18 10:55 AM Page 123

Chapter 6 The ABO Blood Group System 123

an autosomal recessive trait with the inheritance of two Table 6–7 ABO Groups of Offspring from
nonfunctional O genes.
Various Possible ABO Matings
The designations group A and B refer to phenotypes,
whereas AA, BO, and OO denote genotypes. In the case of Offspring Mating
an O individual, both phenotype and genotype are the same Possible Mating Phenotypes
because that individual would have to be homozygous for Phenotypes Genotypes (and Genotypes)
the O gene. An individual who has the phenotype A (or B) A×A AA × AA A (AA)
can have the genotype AA or AO (or BB or BO). Box 6–2
lists the ABO genotypes and phenotypes. Serologically, it is AA × AO A (AA or AO)
not possible to determine the genotype from the phenotype AO × AO A (AA or AO) or O(OO)
of an A or B individual. Family studies or molecular assays
would need to be performed to determine the exact geno- B×B BB × BB B (BB)
type. The phenotype and genotype are the same in an AB BB × BO B (BB or BO)
individual because of the inheritance of both the A and
B gene. Table 6–7 lists possible ABO phenotypes and geno- BO × BO B (BB or BO) or O (OO)
types from various matings. AB × AB AB × AB AB (AB) or A (AA) or B(BB)

Formation of A, B, and H Red Blood Cell Antigens O×O OO × OO O (OO)

A×B AA × BB AB (AB)
The formation of ABH antigens results from the interaction
of genes at three separate loci (ABO, Hh, and Se). These AO × BB AB (AB) or B (BO)
genes do not actually code for the production of antigens but
AA × BO AB (AB) or A (AO)
rather produce specific glycosyltransferases that add sugars
to a basic precursor substance (Table 6–8). A paragloboside AO × BO AB (AB) or A (AO) or B
or glycan is the same basic precursor material from which (BO) or O (OO)
A, B, and H antigens all originate. Specific enzyme trans- A×O AA × OO A (AO)
ferases elicited by an inherited gene attach sugars to the para-
globoside/glycan.11–13 AO × OO A (AO) or O (OO)
When the terminal galactose on the precursor substance A × AB AA × AB AB (AB) or A (AA)
is attached to the N-acetylglucosamine in a beta 1 → 4 link-
age (Fig. 6–3), the precursor substance on erythrocytes is re- AO × AB AB (AB) or A (AA or AO)
ferred to as type 2. ABH antigens on the RBC are constructed or B (BO)
on oligosaccharide chains of a type 2 precursor substance.14 B×O BB × OO B (BO)
A type 1 precursor substance refers to a beta 1 → 3 linkage
between galactose and N-acetylglucosamine (see “Formation BO × OO B (BO) or O (OO)
of A, B, and H Soluble Antigens” section). B × AB BB × AB AB (AB) or B (BB)
The H antigen is the precursor structure on which A and
B antigens are made. Inheritance of the H gene results in BO × AB AB (AB) or B (BB or BO)
or A (AO)
formation of the H antigen. The FUT 1 (H) and FUT 2 (Se)
genes are closely linked and located on chromosome 19, AB × O AB × OO A (AO) or B (BO)
in contrast to the ABO genes located on chromosome 9. The

H and Se genes are not part of the ABO system; yet, their in-
heritance influences A and B antigen expression. The H gene
BOX 6–2
must be inherited to form ABO antigens on the RBCs, and
ABO Genotypes and Phenotypes the Se gene must be inherited to form ABO antigens in
Genotype Phenotype secretions.
A1A1 A1 The ABH antigens develop early in fetal life and do not
A1A2 A1 increase much in strength during the gestational period. The
A1O A1 RBCs of the newborn have been estimated to carry anywhere
A2A2 A2 from 25% to 50% of the number of antigenic sites found on
A2O A2 the adult RBC.9 Consequently, reactions of newborn RBCs
A1B A1B with ABO reagent antisera are frequently weaker than reac-
A2B A2B tions with adult cells. The expression of A and B antigens on
OO O the RBCs is fully developed by 2 to 4 years of age and
BB B remains constant throughout life.9 In addition to age, the
BO B phenotypic expression of ABH antigens may vary with race,
genetic interaction, and disease states.15
6888_Ch06_119-148 31/10/18 10:55 AM Page 124

124 PART II Blood Groups and Serologic Testing

Table 6–8 Glycosyltransferases and Immunodominant Sugars Responsible for H, A, and


B Antigen Specificities
Gene Glycosyltransferase Immunodominant Sugar Antigen
H (FUT1) α-2-L-fucosyltransferase L-fucose H

A α-3-N-acetylgalactosaminyltransferase N-acetyl-D-galactosamine A

B α-3-D-galactosyltransferase D-galactose B

(6) CH2OH Fucose:


5 0 GAL FUC
Immunodominant
4 D-galactose 1
GLNAC sugar responsible
2 "H" antigen for "H" specificity
3
0 (6) CH2OH GAL
"1 4 Linkage" 5 0
4 N-acetylglucosamine 1 GL Protein
3 2
Type-2 precursor chain NHCOCH3
Ceramide
D-galactose

Glucose Spectrin

HH L-fucosyl
( Precursor
Structure )
P.S.
Hh transferase
"H" antigen

(genotype inherited)

Figure 6–3. Type 2 precursor chain. Figure 6–4. Formation of the H antigen.

Interaction of Hh and ABO Genes hh is extremely rare. The term Bombay has been used to
refer to the phenotype that lacks normal expression of the
Individuals who are blood group O inherit at least one ABH antigens because of the inheritance of the hh geno-
FUT 1(H) gene (genotype HH or Hh) and two O genes. The type. The hh genotype does not elicit production of α-2-
H gene elicits the production of an enzyme called α-2-L- L-fucosyltransferase. Accordingly, L-fucose is not added to
fucosyltransferase that transfers the sugar L-fucose to an the type 2 chain and H substance is not expressed on the
oligosaccharide chain on the terminal galactose of type 2 RBC. Even though Bombay (hh) individuals may inherit
chains.15 Sugars occupying the terminal positions of this pre- ABO genes, normal expression, as reflected in the formation
cursor chain and conferring blood group specificity are called of A, B, or H antigens, does not occur. (See “The Bombay
the immunodominant sugars. Therefore, L-fucose is the Phenotypes (Oh)” section.)
sugar responsible for H specificity (blood group O; Fig. 6–4). In the formation of blood group A, the A gene (AA or AO)
The O gene at the ABO locus is sometimes referred to as an codes for production of α-3-N-acetylgalactosaminyltrans-
amorph and does not elicit the production of a catalytically ferase, which transfers an N-acetyl-D-galactosamine(GalNAc)
active polypeptide transferase; therefore, the H substance re- sugar to the H substance. This sugar confers A specificity
mains unmodified.13 Consequently, the O blood group has (blood group A; Fig. 6–5). The A-specific immunodominant
the highest concentration of H antigen. The H substance sugar is linked to a type 2 precursor substance that now con-
(L-fucose) must be formed for the other sugars to be attached tains H substance through the action of the H gene.
in response to an inherited A and/or B gene. The A gene tends to elicit higher concentrations of trans-
The H gene is present in more than 99.99% of the ran- ferase than the B gene, leading to conversion of nearly all of
dom population. Its allele, h, is quite rare, and the genotype the H antigen on the RBCs to A antigen sites. As many as
6888_Ch06_119-148 31/10/18 10:55 AM Page 125

Chapter 6 The ABO Blood Group System 125

D-Galactose: Immunodominant sugar responsible


GALNAC N-Acetylgalactosamine:
for "B" specificity
Immunodominant sugar
FUC GAL responsible for "A"
specificity
"A" antigen GLNAC GAL

GAL FUC GAL

GL Protein GLNAC "B" antigen

GAL
Ceramide
GL Protein

Ceramide
Spectrin

Spectrin

AA N-Acetylgalactosaminyl "A"
"H" structure
AO transferase antigen
BB Galactosyl
"H" structure "B" antigen
BO transferase
( genotype
)
inherited

Figure 6–5. Formation of the A antigen.


( genotype
inherited )

Figure 6–6. Formation of the B antigen.

810,000 to 1,170,000 antigen sites exist on an A1 adult RBC


in response to inherited genes.5 located on chromosome 9, consists of seven exons.11 The
Individuals with blood group B inherit a B gene (BB or BO) last two exons (6 and 7) encode for the catalytic domain
that codes for the production of α-3-D-galactosyltransferase of the ABO glycosyltransferases.15 Exons 6 and 7 constitute
and attaches D-galactose (Gal) sugar to the H substance pre- 77% of the gene, with most of the coding sequence lying
viously placed on the type 2 precursor substance through in exon 7.17,18 Amino acid substitutions, resulting primarily
the action of the H gene.14 This sugar is responsible for from deletions, mutations, or gene recombinations within
B specificity (blood group B; Fig. 6–6). Anywhere from these two exons of the coding DNA of variant ABO glyco-
610,000 to 830,000 B antigen sites exist on a B adult RBC syltransferases, are responsible for the less efficient transfer
in response to the conversion of the H antigen by the α-3- of the immunodominant sugar to H substance, resulting in
D-galactosyltransferase produced by the B gene.5 weak serologic reactions observed in ABO subgroups.15 The
When both A and B genes are inherited, the B enzyme seven common ABO alleles include A1, A1variant (A1v), A2,
(α-3-D-galactosyltransferase) seems to compete more effi- B1, O1, O1variant(O1v), and O2.19,20
ciently for the H substance than the A enzyme (α-3- All ABO antigens arise from mutations in the single ABO
N-acetylgalactosaminyltransferase). As a result of this en- gene. Only three specific mutations showing a high fre-
zyme competition, the average number of A antigens on an quency in the population indirectly lead to changes in the
AB adult cell is approximately 600,000 sites, compared epitope structures resulting in A, B, or O specificities. Two
with an average of 720,000 B antigen sites.5 Interaction of of the mutations (substitutions) change the specificity of
the Hh and ABO genes is reviewed in Figure 6–7. the enzyme from an α-N-acetylgalactosaminyltransferase
(the A enzyme) to an α-3-D-galactosyltransferase (the B en-
Molecular Genetics of ABO zyme).20 These glycosyltransferases, in turn, introduce an
α 1,3 N-acetylgalactosamine (A) or an α 1,3 galactose (B)
carbohydrate at the ends of type H oligosaccharide
Advanced Concepts (glycan) chains. The third mutation, a deletion within the
Since the cloning in 1990 of the complementary DNA cor- 5' region of the catalytic domain, creates a premature stop
responding to messenger RNA transcribed at the blood codon that inactivates the enzyme altogether, leaving
group ABO locus, more than 250 ABO alleles have been the H glycan unmodified.21 These different glycans define
identified by molecular investigation.13,16 The ABO gene, the A, B, or O antigens or epitopes.
6888_Ch06_119-148 31/10/18 10:55 AM Page 126

126 PART II Blood Groups and Serologic Testing

ar
s ug
nt )
en i na m ine
g
nti om sa
"A
a od cto
mun gala
(im l
ety
ac
N-
e
feras
ns
l tra
ny
mi
sa
cto
/A
O ala
AA t y lg
e
ac
"H structure" N- "B antigen"
Type-2-precursor chain HH / Hh
BB/BO
(immunodominant (immunodominant
(Precursor structure) L-fucosyl transferase D-galactosyl transferase
sugar L-fucose) sugar galactose)
A
an
dB
N- ge
ga ace ne
lac tyl s
tos ga
yl lac
hh tra tos
ns am
fer in
as yl
es an
d "A
B
su an
ga tig
rs en
an N- "(
dg ac im
Precursor structure unchanged ala ety mu
cto lg n od
se ala o
(Bombay phenotype) ) cto mina
sa nt
mi
ne

Figure 6–7. Interaction of the Hh and ABO genes.

Formation of A, B, and H Soluble Antigens

ABH antigens are integral parts of the membranes of RBCs,


endothelial cells, platelets, lymphocytes, and epithelial cells.8 Genotype: Se se water-soluble secretions
ABH-soluble antigens can also be found in all body secretions. AB produced by tissue cells
Their presence is dependent on the ABO genes inherited and HH
on the inheritance of another set of genes called Sese (secretor
genes) that regulate their formation. Eighty percent of the ran-
dom U.S. population are known as secretors because they have
inherited a secretor gene (SeSe or Sese). The inheritance of a
FUT 2 (Se) gene codes for the production of the transferase
α-2-L-fucosyltransferase that modifies the type 1 precursor
substance in secretions to form H substance.22 If the corre- H A B
sponding gene is present, this H substance can then be further
modified to express A and B substance in secretions such as
saliva. For example, a group A individual who is a secretor
(SeSe or Sese) will secrete glycoproteins carrying A and H anti-
gens. However, the Se gene does not affect the formation of A, = A-Acetylgalactosamine
B, or H antigens on the RBC. It is the presence of the Se gene– = D-Galactose
specified α-2-L-fucosyltransferase that determines whether = N-Acetylglucosamine
ABH-soluble substances will be secreted (Fig. 6–8).22 People = L-fucose
who inherit the sese genotype are termed nonsecretors. = Protein backbone

Comparison of A, B, and H Antigens on RBCs


with A, B, and H Soluble Substances Se AB A, B, H
P.S. H antigen soluble
H antigens
The formation of soluble A, B, and H substances is the same
as that described for the formation of A, B, and H antigens Figure 6–8. Secretor ABH glycoprotein substances.
6888_Ch06_119-148 31/10/18 10:55 AM Page 127

Chapter 6 The ABO Blood Group System 127

on the RBCs, except for a few minor distinctions that are linkage position of galactose (Gal) to N-acetylglucosamine
compared in Table 6–9. (GlcNAc); specifically type 1 has a beta 1→3 linkage while
In the past, tests for ABH secretion were used to establish type 2 has a beta 1→4 linkage.22 Addition of specific immu-
the true ABO group of an individual with poorly developed nodominant sugars to the type 2 and 4 chains leads to for-
RBC antigens. The term secretor refers only to secretion mation of A, B, and H antigens on the RBC membrane, with
of A, B, and H soluble antigens in body fluids. The demon- the majority being present in the form of type 2 chains.
stration of A, B, and H substances in saliva is evidence of the Adding the same immunodominant sugars to type 1 and
inheritance of an A gene, B gene, H gene, and Se gene. The 3 chains in the body secretions allow for A, B, and H soluble
glycoprotein-soluble substances (or antigens) normally substances to be made in body secretions. Box 6–3 summa-
found in the saliva of secretors are listed in Table 6–10. Both rizes the body fluids in which ABH-soluble substances can
ABH red blood cell antigens and ABH-soluble substances be found.
are formed due to the attachment of an immunodominant
The procedure for determining the secretor status
sugar to an oligosaccharide chain. Although several types
(saliva studies) can be found as Procedure 6-1 on
of oligosaccharide chains exist, types 1 and 3 are primarily
the textbook’s companion website.
associated with body secretions, while types 2 and 4 are
associated with the red blood cell membrane.22 Type 1 and
ABO Subgroups
2 chains are more abundant, and they differ only in the
The original reports of most ABO subgroups were made
Table 6–9 Comparison of ABH Antigens before the availability of the monoclonal typing reagents
currently used in routine ABO grouping. ABO subgroups
on RBCs with A, B, and
represent phenotypes showing weaker and variable serologic
H Soluble Substances
reactivity with the commonly used human polyclonal
ABH Antigens A, B, and H Soluble anti-A, anti-B, and anti-A,B reagents.
on RBCs Substances
RBC antigens can be Secreted substances are
A Subgroups
glycolipids, glycoproteins, glycoproteins.
or glycosphingolipids. In 1911, von Dungern described two different A antigens
based on reactions between group A RBCs and anti-A and anti-
RBC antigens are synthesized Secreted substances are A1.23 Group A RBCs that react with both anti-A and anti-A1
only on type 2 precursor chains. primarily synthesized on are classified as A1, whereas those that react with anti-A and
type 1 precursor chains.
not anti-A1 are classified as A2 (Table 6–11 and Figs. 6–9 and
Type 2 chain refers to a Type 1 chain refers to a 6–10). RBCs from A1 and A2 individuals react equally strong
beta 1→4 linkage in which beta-1→3 linkage in which
the number one carbon the number one carbon of the
of the galactose is attached to galactose is attached to the
the number four carbon of number three carbon of BOX 6–3
the N-acetylglucosamine sugar the N-acetylglucosamine sugar
of the precursor substance. of the precursor substance. Fluids in Which A, B, and H Substances Can Be
Detected in Secretors
The enzyme produced by The enzyme produced by
the H (FUT 1) gene (α-2-L- the Se (FUT 2) gene (α-2-L- • Saliva
fucosyltransferase) acts fucosyltransferase) preferen- • Tears
primarily on type 2 chains, tially acts on type 1 chains in • Urine
which are prevalent on the secretory tissues. • Digestive juices
RBC membrane.
• Bile
• Milk
• Amniotic fluid
Table 6–10 ABH Substance in the Saliva • Pathological fluids: pleural, peritoneal, pericardial, ovarian cyst
of Secretors (SeSe or Sese)*
Substances in Saliva
ABO Group A B H Table 6–11 A1 Versus A2 Phenotypes
O None None ↑↑ Reactions of Patient’s RBCs With
A ↑↑ None ↑ Blood Anti-A Reagent Anti-A1 Lectin
Group (anti-A plus anti-A1) Reagent
B None ↑↑ ↑
A1 + +
AB ↑↑ ↑↑ ↑
A2 + 0
* Nonsecretors (sese) have no ABH substances in saliva.
↑↑ and ↑, respectively, represent the concentration of ABH substances in saliva. + = positive (agglutination); 0 = negative (no agglutination)
6888_Ch06_119-148 31/10/18 10:55 AM Page 128

128 PART II Blood Groups and Serologic Testing

A1 A1 A A1 A A
A
A1 A A A1 A A A
A1 A1 A

A1 A2

Reactions of Patients’ Red Cells with


Anti-A Anti-A1
Blood Group Antigen Present (Anti-A plus Anti-A1) lectin

A1 A1 A + +
A2 A + 0
Figure 6–9. A1 versus A2 phenotypes.

A1 A1 A A A
A1 A1
A1 A1 A A
A1

A1 A2

Reactions of Patients’ Red Cells with


Anti-A Anti-A1
Blood Group Antigen Present (Anti-A plus Anti-A1) lectin

A1 A1 + +
A + 0 Figure 6–10. A1 versus A2 phenotypes (alternative con-
A2
ceptual presentation).

with current reagent monoclonal anti-A in ABO forward The production of both types of antigens is a result of an
typing tests.2 inherited gene at the ABO locus. Inheritance of an A1 gene
The A subgroups are more common than B subgroups. elicits production of high concentrations of the enzyme
The weaker serologic reactivity of ABO subgroups is attrib- α-3-N-acetylgalactosaminyltransferase, which converts
uted to the decreased number of A and B antigen sites on almost all of the H precursor structure to A1 antigens on the
their red blood cells. Classification into A1 and A2 pheno- RBCs. The very potent gene A1 creates between 810,000 and
types accounts for 99% of all group A individuals. The cells 1,170,000 antigen sites on the adult A1 RBC, whereas inher-
of approximately 80% of all group A (or AB) individuals are iting an A2 gene results in production of only 240,000 to
A1 (or A1B), and the remaining 20% are A2 (or A2B) or 290,000 antigen sites on the adult A2 RBC.9 The A2 allele is
weaker subgroups. The differences between A1 and A2 are characterized by a single base substitution at nucleotide 467
both quantitative and qualitative (Table 6–12). and a single base deletion at nucleotide 1060 (1060delC)
in exon 7.19 These substitutions alter the active site of the
coding region and subsequently change the specificity of the
A glycosyltransferase. The immunodominant sugar on both
Table 6–12 Quantitative and Qualitative A1 and A2 RBCs is N-acetyl-D-galactosamine.
Differences of Subgroups A1 Qualitative differences also exist, since 1% to 8% of A2 in-
and A2 dividuals produce anti-A1 in their serum and 22% to 35% of
Quantitative Qualitative A2B individuals produce anti-A1.9 This antibody can cause
discrepancies between forward and reverse ABO testing and
• ↓ Number of antigen sites • Differences in the precursor incompatibilities in crossmatches with A1 or A1B cells. Anti-
oligosaccharide chains
A1 is a naturally occurring IgM cold-reacting antibody and
• ↓ Amount of transferase • Subtle differences in trans- is unlikely to cause a transfusion reaction because it usually
enzyme ferase enzymes reacts only at temperatures well below 37°C. It is considered
• ↓ Amount of branching • Formation of anti-A1, in a per-
clinically significant if it is reactive at 37°C.
centage of some subgroups The antigens present on the RBCs of A1 and A2 individu-
als can be depicted in two ways. Most often, A1 RBCS are
6888_Ch06_119-148 31/10/18 10:55 AM Page 129

Chapter 6 The ABO Blood Group System 129

illustrated as having both A and A1 antigens on the cell sur- naturally occurring IgM cold agglutinin that reacts best below
face, in contrast to A2 cells which only contain A (Fig. 6–9). room temperature. As can be expected, this antibody is
Alternatively, A1 RBCs can also be conceptualized as having formed in response to a natural substance and reacts most
only A1 antigen sites and A2 as only having A antigen sites strongly with cells of group O individuals (which have the
(Fig. 6–10). In routine forward grouping, reagent anti-A greatest amount of H substance on their RBCs) and weakly
strongly agglutinates both A1 and A2 phenotypes. Differen- with the RBCs of A1B individuals (which contain small
tiation of A1 and A2 phenotypes is determined serologically amounts of H substance). It is an insignificant antibody in
using anti-A1 lectin, a reagent made from the seeds of terms of transfusion purposes because it has no reactivity at
the plant Dolichos biflorus. Lectins are seed extracts that body temperature (37°C). However, high-titered anti-H may
agglutinate human cells with some degree of specificity.24 react at room temperature and present a problem in antibody
Anti-A1 lectin agglutinates A1 (or A1B) cells but does not screening procedures because reagent screening cells are
agglutinate A2 (or A2B cells). Box 6–4 lists some of the group O (see Chapter 10, “Detection and Identification of
lectins used in blood banking. Characteristics of the A1 and Antibodies”). This high-titered anti-H may also present a
A2 phenotypes are presented in Table 6–13. Due to the problem with compatibility testing (see Chapter 11, “Pre-
A2 glycosyltransferase being less efficient at adding the transfusion Testing”). Anti-H lectin, Ulex europaeus, closely
immunodominant sugar to the H antigen precursor, A2 parallels the reactions of human anti-H. Both antisera agglu-
RBCs show increased reactivity with anti-H lectin, Ulex tinate RBCs of group O and A2 and react very weakly or not
europaeus, compared to A1 RBCs. at all with groups A1 and A1B.24 Group B cells give reactions
H antigen is found in greatest concentration on the RBCs of variable strength (Fig. 6–11).
of group O individuals. H antigen may not be detectable in
group A1 individuals, because in the presence of the A1 gene,
almost all of the H antigen is converted to A1 antigen Advanced Concepts
by placing the large N-acetyl-D-galactosamine sugar on the The discussion thus far has presented a basic overview of
H substance. Due to the presence of so many A1 antigens, the two major ABO subgroups, A1 and A2. A more plausi-
the H antigen on A1 and A1B RBCs may be hidden and there- ble, yet more detailed, theory of ABO subgroups has been
fore may not be available to react with anti-H antisera. In proposed by the identification of four different forms of
the presence of an A2 gene, only some of the H antigen is H antigens, two of which are unbranched straight chains
converted to A antigens, and the remaining H antigen is (H1, H2) and two of which are complex branched chains
detectable on the cell. (H3, H4; Fig. 6–12).24 The antigens H1 through H4 corre-
Weak subgroups of the A antigen will often have an spond to the precursor structures on which the A enzyme
inverse reciprocal relationship between the amount of H anti- can act to convert H antigen to blood group A active
gen on the RBC and the amount of A antigens formed (i.e., glycolipids. Although the chains differ in length and com-
the more A antigen formed, the less H antigen expressed on plexity of branching, the terminal sugars giving rise
the RBC). The H antigen on the RBCs of A1 and A1B individ- to their antigenic specificity are identical. Studies on the
uals is so well hidden by N-acetyl-D-galactosamine that chemical and physical characteristics of the A1 and A2
anti-H is occasionally found in the serum. This anti-H is a enzyme transferases have demonstrated that these two en-
zymes are different qualitatively.17,25 Straight chain H1 and
H2 glycolipids can be converted to Aa and Ab antigens, re-
BOX 6–4 spectively, by both A1 and A2 enzymes, with the A2 enzyme
being less efficient. The more complex branched H3 and
Lectins Used in Blood Banking H4 structures can be converted to Ac and Ad antigens by
• Dolichos biflorus—agglutinates A1 or A1B A1 enzyme and only very poorly by A2 enzyme.24 As a re-
• Bandeiraea simplicifolia—agglutinates B cells sult, more unconverted H antigens (specifically H3 and H4)
• Ulex europaeus—agglutinates O cells (H specificity) and other ABO are available on group A2 RBCs, and only Aa and Ab deter-
blood groups depending on the amount of H antigen available. minants are formed from H1 and H2 structures.25

Table 6–13 Characteristics of A1 and A2 Phenotypes


Reagents Antibodies in Serum Other
Substances Number of
Present in Saliva Antigen Sites
Phenotypes Anti-A Anti-B Anti-A,B Anti-A1 Common Unexpected of Secretors RBC x 103

A1 4+ 0 4+ 4+ Anti-B None A, H 810–1,170

A2 4+ 0 4+ 0 Anti-B Anti-A1 (1%–


8% of cases) A, H 240–290
6888_Ch06_119-148 31/10/18 10:55 AM Page 130

130 PART II Blood Groups and Serologic Testing

O > A2 > B > A2B > A1 > A1B On the RBCs of some A2 individuals, Ac is extremely low
and Ad is completely lacking (Box 6–5). These are the
greatest least individuals in whom one would likely find anti-A1 in the
amount of amount of
H H serum. This anti-A1 antibody could really be an antibody
to Ac and Ad determinants, which the A2 individuals lack.
Figure 6-11. Reactivity of anti-H antisera or anti-H lectin with ABO blood
Furthermore, anti-A1 can be found in the serum in 22% to
groups.

GALNAC GALNAC

Fuc GAL Fuc GAL

GlcNac GlcNac
Aa
H1
GAL GAL

Glc GlcNac Ab
H2

GAL
Ceramide

RBC Glc

Ceramide
H1
RBC

GALNAC GALNAC
H2

Fuc GAL GAL Fuc


Fuc GALNAC
GlcNac GlcNac
GAL
GAL

GlcNac Fuc GALNAC


GlcNac
Ac
H3
Fuc GAL GAL
GAL
GlcNac GlcNac
Glc
GAL

GlcNac Ad
Ceramide H4

RBC GAL

Glc
H3

Ceramide
Denotes immunodominant
sugar for H antigen RBC

Denotes immunodominant
sugar for A antigen
H4

Figure 6–12. H-active antigenic structures.


6888_Ch06_119-148 31/10/18 10:55 AM Page 131

Chapter 6 The ABO Blood Group System 131

• Increased variability in the detectability of H antigen,


BOX 6–5 resulting in strong reactions with anti-H
Structural Characteristics of A1 and A2 RBCs • Presence or absence of anti-A1 in the serum
• A2 RBCs: Predominantly Aa and Ab and unconverted H3 and H4 Secretor studies, adsorption-elution tests, and molecular
antigen sites testing can be utilized to subdivide A individuals into A3, Ax,
• A1 RBCs: Aa, Ab, Ac, and Ad determinants and no unconverted H3 Aend, etc.27 (Table 6–14).
and H4 antigen sites Occasionally, weak subgroups of A may present practical
problems, for example, if an Ax donor was mistyped as
group O and was transfused to a group O patient. This is
35% of A2B individuals. Since the B enzyme transferase is potentially dangerous because the group O patient possesses
usually more efficient than the A enzyme in converting H anti-A,B, which agglutinates and lyses Ax RBCs, causing
structures to the appropriate antigen, A2 enzymes would rapid intravascular hemolysis.
probably fail completely when paired with a B enzyme. As
a result, A2B individuals would be far more likely to lack
Ac and Ad components with subsequent production of anti- Advanced Concepts
Ac and anti-Ad (anti-A1). Weak A phenotypes can be serologically differentiated
As stated previously, most group A infants appear to be using the following techniques:
A2 at birth, with subsequent development to A1 a few
months later. Newborns have a deficiency of the branched • Forward grouping of A and H antigens with anti-A,
H3 and H4 antigens and therefore the Ac and Ad antigens as anti-A,B, and anti-H
well, possibly accounting for the A2 phenotype. Adult • Reverse grouping of ABO isoagglutinins and the presence
cells contain a higher concentration of branched H3 and of anti-A1
H4 structures and therefore Ac and Ad determinants of the • Adsorption-elution tests with anti-A
A antigen in A1 individuals.25 • Saliva studies to detect the presence of A and H substances
Additional special procedures such as molecular testing
for mutations or serum glycosyltransferase studies for de-
Weak A Subgroups
tecting the A enzyme can be performed for differentiation
Subgroups weaker than A2 occur infrequently and are most of weak subgroups.26,27 Absence of a disease process should
often recognized through an ABO discrepancy (unex- be confirmed before subgroup investigation because ABH
pected reactions in the forward and reverse grouping). antigens are altered in various malignancies and other
These subgroups of A make up 1% of those encountered hematologic disorders. (See “ABH Antigens and Antibodies
in the laboratory and therefore are mainly of academic in Disease” and “ABO Discrepancies” sections).
interest.26 Characteristics of weak A subgroups include the Weak A subgroups can be distinguished as A3, Ax,
following: Aend, Am, Ay, and Ael using the serologic techniques men-
tioned previously (see Table 6–14). The characteristics
• Decreased number of A antigen sites per RBC (resulting in of each weak A subgroup are presented in the following
weak or no agglutination with human polyclonal anti-A) paragraphs.
• Varying degrees of agglutination by human anti-A,B8

Table 6–14 Characteristics of Weak ABO Phenotypes


Reagents Antibodies in Serum Other
Substances
present Presence of Number of
in saliva A transferase antigen sites
Phenotypes Anti-A Anti-B Anti-A,B Anti-H Anti-A Anti-B Anti-A1 of secretors in serum RBC x 103

A3 ++mf 0 ++mf 3+ No Yes Sometimes A, H Sometimes 35

Ax wk/0 0 2+ 4+ 0/wk Yes Almost always H Rarely 5

Aend wk mf 0 wk mf 4+ No Yes Sometimes H No 3.5

Am* 0/wk 0 0/+ 4+ No Yes No A, H Yes 1

Ay* 0 0 0 4+ No Yes No A, H Trace 1

Ael* 0 0 0 4+ Some Yes Yes H No 0.7

*A specificity demonstrated only by absorption/elution procedures.


0 = negative; mf = mixed-field agglutination; wk = weak
6888_Ch06_119-148 31/10/18 10:56 AM Page 132

132 PART II Blood Groups and Serologic Testing

A3 RBCs characteristically demonstrate a mixed-field Am RBCs. Trace amounts of A glycosyltransferase is detectable


pattern of agglutination with anti-A and most anti-A,B in the serum of Ay individuals, and saliva secretor studies
reagents.28 Mixed-field can be defined as small agglutinates demonstrate H and A substance, with A substance present in
within predominantly unagglutinated RBCs. The estimated below-normal quantities.18 Ay individuals usually do not pro-
number of A antigen sites is approximately 35,000 per duce anti-A1. The Ay phenotype can be observed in siblings,
RBC.28 Weak α-3-N-acetylgalactosaminyltransferase activ- implicating a recessive mode of inheritance. This phenotype
ity is detectable in the serum. However, there appears to be does not represent expression of an alternate allele at the ABO
molecular heterogeneity in the A3 glycosyltransferases iso- locus but rather as a germline mutation of an A gene within
lated from various A3 phenotypes. A3 enzyme is a product a family.23
of an allele at the ABO locus inherited in a dominant man- Ael RBCs typically are unagglutinated by anti-A or
ner; however, the A3 blood group has been reported to be anti-A,B reagents; however, adsorption and elution can be
very heterogeneous at the molecular level.29–31 Anti-A1 may used to demonstrate the presence of the A antigen. No
be present in serum of A3 individuals, and A substance is detectable A enzyme activity can be demonstrated in
detected in the saliva of A3 secretors. the serum or in the RBC membranes of Ael individuals by
Ax RBCs characteristically are not agglutinated by glycosyltransferase studies.18 The Ael phenotype is inher-
anti-A reagent but do agglutinate with most examples of ited as a rare gene at the ABO locus.36 Ael individuals
anti-A,B.28 The estimated number of A antigen sites is usually produce an anti-A1 that is reactive with A1 cells and
approximately 4,000 per RBC.28 Anti-A can be adsorbed sometimes produce anti-A, which agglutinates A2 RBCs.28
and then eluted from Ax cells without difficulty. A trans- Secretor studies demonstrate the presence of only H sub-
ferase is not usually detectable in the serum or in the RBC stance in the saliva of Ael secretors.
membranes of Ax individuals. The molecular genetics of Ax It should be noted that there are still some reported A
reflect the considerable heterogeneity of the serologic phe- variants that do not fit into any of the weak subgroups de-
notypes.32 Ax individuals almost always produce anti-A1 in scribed, alluding to the existence of new alternate alleles
their serum. Routine secretor studies detect the presence or regulation by modifier genes.37,38
of only H substance in Ax secretors. However, Ax secretors A general flowchart for the process of elimination
contain A substance detectable only by agglutination/ and identification of various subgroups is presented in
inhibition studies using Ax RBCs as indicators.33 Caution Figure 6–13. It is assumed that the patient’s medical
should be used in interpreting results of secretor studies history, such as recent transfusion, pregnancy history,
using Ax indicator cells and anti-A, because not all Ax cells disease states, and medications, has been investigated
are agglutinated by anti-A. and excluded as a source of the discrepancy.
Aend RBCs characteristically demonstrate very weak
mixed-field agglutination with some anti-A and anti-A,B
reagents.28 The estimated number of A antigen sites on the Weak B Subgroups
few agglutinable RBCs is approximately 3,500 per RBC,
whereas no detectable A antigens are demonstrated on Subgroups of B are very rare and much less frequent than A
RBCs that do not agglutinate.33 No A glycosyltransferase is subgroups. Subgroups of B are usually recognized by varia-
detectable in the serum or in the RBC membranes of Aend tions in reaction strength using anti-B and anti-A,B. Inheri-
individuals. Aend is inherited as an allele at the ABO locus.23 tance of B subgroups, similar to that of the majority of A
Secretor studies detect the presence of only H substance subgroups, is considered to be a result of alternate alleles at
in the saliva of Aend secretors. Anti-A1 is found in some Aend the B locus. Criteria used for differentiation of weak B phe-
sera.28 The phenotypes of Afinn and Abantu are also variants notypes include the following techniques:
of the Aend subgroup.28 • Strength and type of agglutination with anti-B, anti-A,B,
Am RBCs are characteristically not agglutinated, or are and anti-H
agglutinated only weakly, by anti-A or anti-A,B reagents.34 • Presence or absence of ABO isoagglutinins in the serum
A strongly positive adsorption or elution of anti-A confirms • Adsorption-elution studies with anti-B
the presence of A antigen sites. The estimated number • Presence of B substance in saliva
of A antigen sites varies from 200 to 1,900 per RBC in Am • Molecular testing
individuals.33 An A enzyme of either the A1 or A2 type
previously described is detectable in the serum of Am sub-
groups.34 Am is inherited as a rare allele at the ABO locus.35
These individuals usually do not produce anti-A1 in their Advanced Concepts
sera. Normal quantities of A and H substance are found in RBCs demonstrating serologic activity that is weaker than
the saliva of Am secretors.28 normal are designated weak B phenotypes or B subgroups
Ay RBCs are not agglutinated by anti-A or anti-A,B and include B3, Bx, Bm, and Bel phenotypes39 (Table 6–15).
reagents. Adsorption and elution of anti-A is the method used There are no B subgroups reported that are equivalent to
to confirm the presence of A antigens. Activity of eluates from Aend or Ay. A classification system similar to A subgroups
Ay RBCs is characteristically weaker than that of eluates from has been used because of common serologic characteristics.
6888_Ch06_119-148 31/10/18 10:56 AM Page 133

Chapter 6 The ABO Blood Group System 133

Red Cell Reactions


with anti-A and/or anti-A,B

weakly agglutinated no agglutination


(possibly A3, Ax, Aend)

adsorbs and elutes anti-A


(possibly Am, Ay, Ael)

mixed-field (mf) ⭐10% weak agglutination


agglutination red cells with anti-A,B only
show very
1) anti-A easily 1) anti-A adsorbed 1) anti-A
weak mf
adsorbed and and eluted adsorbed
agglutination
eluted and eluted
2) secretors contain
2) secretors small amount 2) secretors
demonstrate of A substance contain
quantities of A in saliva only H
substance in saliva substance
and no A
substance
in saliva

A3 Aend Ax Am Ay Ael

Figure 6–13. Investigation of weak A subgroups.

Table 6–15 Characteristics of B Phenotypes


Reagents Antibodies in Serum Other
Substances Presence of a
Present in Saliva B Transferase
Phenotypes Anti-A Anti-B Anti-A,B Anti-H Common Unexpected of Secretors in Serum

B 0 4+ 4+ 2+ Anti-A None B,H Yes


B3 0 ++mf ++mf 3+ Anti-A None B,H Yes (wk pos)

Bx 0 wk wk 3+ Anti-A Weak anti-B H No

Bm* 0 0/wk 0/wk 3+ Anti-A None B,H Yes (wk pos)

Bel* 0 0 0 3+ Anti-A Sometimes a H No


weak anti-B

*B specificity demonstrated only by adsorption/elution procedures.


0 = negative; mf = mixed-field; wk = weak

Serologic techniques can be used to characterize B sub- of secretors. The B3 subgroup is the most frequent weak
groups in the following categories: B3, Bx, Bm, and Bel. The B phenotype.28,40
characteristics of each weak B subgroup are presented in Bx RBCs typically demonstrate weak agglutination with
the following paragraphs. anti-B and anti-A,B antisera. B glycosyltransferase has not
The B3 phenotype generally results from the inheri- been detected in the serum or in the RBC membranes of
tance of a rare gene at the ABO locus and is characterized Bx phenotypes, but a weakly reactive anti-B usually is pro-
by a mixed-field pattern of agglutination with anti-B and duced.41 Bx RBCs readily adsorb and elute anti-B. Secretor
anti-A,B antisera.28 B glycosyltransferase is present in the studies demonstrate large amounts of H substance and some
serum but not in the RBC membranes of these individu- B substance that often can only be detected by inhibiting
als. Anti-B is absent in the serum of B3 phenotypes, yet agglutination of Bx cells with anti-B.23 Family studies
B substance is present in normal amounts in the saliva suggest that Bx is a rare allele at the ABO locus.42
6888_Ch06_119-148 31/10/18 10:56 AM Page 134

134 PART II Blood Groups and Serologic Testing

Bm RBCs are characteristically unagglutinated by anti-B Bombay phenotype is 0.048%, 0.004%, and 0.005%, respec-
or anti-A,B antisera. The Bm RBCs easily adsorb and elute tively.50 In comparison, prevalence of the Bombay pheno-
anti-B. B glycosyltransferase is present in the serum of Bm type in Europe is 0.0003%.51
phenotypes but is usually lower in activity and varies from The (Oh) Bombay phenotype is inherited as an autoso-
individual to individual.43 Only very small amounts of mal recessive trait. The underlying molecular defect is
B transferase activity are demonstrated in Bm RBC mem- most often a mutation in the gene FUT1 (H gene), produc-
branes. Reduced activity of B enzyme in hematopoietic ing a silenced gene incapable of coding for the enzyme,
tissue is clearly the defect causing the formation of the Bm α-2-L-fucosyltransferase (H transferase).49 This enzyme
subgroup, since normal B plasma incubated with Bm RBCs normally catalyzes the transfer of L-fucose in an α-1,2
and uracil diphosphate (UDP)-galactose transforms this linkage to the terminal galactose of the precursor molecule
subgroup into a normal group B phenotype. Anti-B is not on RBCs forming the H antigen. This mutation underlying
characteristically present in the serum of Bm individuals. the Bombay phenotype is also associated with a silenced
Normal quantities of H and B substance are found in the FUT2 gene (Se gene), which usually encodes a very similar
saliva of Bm secretors. α-2-L-fucosyltransferase that when active, transfers a
The Bm phenotype is usually the result of inheritance of fucose to form H antigens in secretions.49 Box 6–7 sum-
a rare allele at the ABO locus, although the subgroup Bm marizes the general characteristics of the Bombay pheno-
may be the product of an interacting modifying gene linked type. When family studies demonstrate which ABO genes
closely to the ABO locus.44 This modifier gene may depress are inherited in the Bombay phenotype, the genes are
expression of the B gene, resulting in decreased B enzyme written as superscripts (OhA, OhB, OhAB).52
activity.45 The Bm subgroup is reported to be more frequent Since these RBCs lack normal ABH antigens, they fail to
in Japan.45 react with anti-A, anti-B, and anti-H antisera. In RBC testing
Bel RBCs are unagglutinated by anti-B or anti-A,B anti- using anti-A and anti-B, the Bombay would phenotype as an
sera. This extremely rare phenotype must be determined O blood group. However, RBCs of the Bombay phenotype
by adsorption and elution of anti-B. No B glycosyltrans- (Oh) do not react with the anti-H lectin (Ulex europaeus),
ferase has been identified in the serum or RBC membrane unlike those of the normal group O individual, which react
of Bel individuals. Bel is inherited as a unique mutation in strongly with anti-H lectin.49 Bombay serum contains
exon 7 of the B gene at the ABO locus.46 A weak anti-B may anti-A, anti-B, anti-A,B, and anti-H. Unlike the anti-H found
be present in the serum of this subgroup. Only H substance occasionally in the serum of A1 and A1B individuals, the
is demonstrated in saliva of Bel secretors. Bombay anti-H can often be potent and reacts strongly at
Other weak B phenotypes have been reported that do 37°C. It is an IgM antibody capable of binding complement
not possess the appropriate characteristics for classification and causing RBC lysis. Transfusing normal group O blood
into one of the groups previously discussed.47 These may (with the highest concentration of H antigen) to a Bombay
represent new classifications and new representations of recipient (anti-H in the serum) would cause immediate cell
ABO polymorphism. lysis. Only blood from another Bombay individual will be
compatible and can be transfused to a Bombay recipient.
ABH substance is also absent in the saliva of individuals with
The Bombay Phenotypes (Oh) the Bombay phenotype.49

The Bombay phenotype was first reported by Bhende in


1952 in Bombay, India.48 The phenotype results from the
BOX 6–7
inheritance of a double dose of the h gene, producing the
very rare genotype hh. As a result, the ABO genes cannot be General Characteristics of Bombay Oh (Hnull)
expressed and ABH antigens cannot be formed, since there Phenotypes
is no H antigen made in the Bombay phenotype (Box 6–6).
• Absence of H, A, and B antigens; no agglutination with anti-A,
More than 130 Bombay phenotypes have been reported in anti-B, or anti-H lectin
various parts of the world.49 In the Indian states of Andhra • Presence of anti-A, anti-B, anti-A,B, and a potent wide thermal
Pradesh, Tamil Nadu, and Karnataka, prevalence of the range of anti-H in the serum
• A, B, H nonsecretor (no A, B, or H substances present in saliva)
• Absence of α-2-L-fucosyltransferase (H enzyme) in serum and
H antigen on red blood cells
BOX 6–6
• Presence of A or B enzymes in serum (depending on ABO
The Bombay Phenotype (Oh) genotype)
• A recessive mode of inheritance (identical phenotypes in children
• hh genotype but not in parents)
• No H antigens formed; therefore, no A or B antigens formed • RBCs of the Bombay phenotype (Oh) will not react with the anti-H
• Phenotypes as blood group O lectin (Ulex europaeus)
• Anti-A, anti-B, anti-A,B, and anti-H present in the serum • RBCs of the Bombay phenotype (Oh) are compatible only with the
• Can only be transfused with blood from another Bombay (Oh) serum from another Bombay individual
6888_Ch06_119-148 31/10/18 10:56 AM Page 135

Chapter 6 The ABO Blood Group System 135

The Para-Bombay Phenotypes with cord cells and is not inhibited by secretor saliva. Due
to their secretor status, normal levels of H substances are
present in the saliva.54 A and B substances are present in
Advanced Concepts the secretions when A and B genes are present.54
The para-Bombay phenotypes are those rare phenotypes in
which the RBCs either completely lack H antigens or have
small amounts of H antigen present.52 The genetic basis for ABH Antigens and Antibodies
the para-Bombay phenotype is either a mutated FUT1 in Disease
(H gene) with or without an active FUT2 gene (Se gene) or
a silenced FUT1 gene with an active FUT2 gene.53 With Associations between ABH antigens and practically any dis-
the mutated FUT1 gene para-Bombay, the encoded α-2-L- order known to man can be found throughout medical liter-
fucosyltransferase enzyme activity is greatly reduced, and ature. Even more profound are the associations of blood
very small amounts of H, A, and B antigens are produced. group specificity with such things as a more pronounced
RBCs from these individuals expressing weak forms of “hangover” in A blood groups, “criminality” in group B
A and B antigens are serologically undetectable using rou- blood groups, and “good teeth” in group O individuals.
tine ABO testing methods and are detected only using There are also several papers correlating blood groups with
adsorption and elution techniques. If a person is genetically personality traits. It is no surprise that many scientists refer
A or B, the respective enzymes can be detected, but no to these associations as a part of blood group mythology.
H enzyme is detectable, even though it has been shown However, more relevant associations between blood groups
that there is limited production of H antigen on the RBC.54 and disease are important to the blood banker in terms of
The notations Ah and Bh, respectively, have been used to blood group serology.
describe these individuals. ABh individuals have also been Various disease states seem to alter RBC antigens, result-
reported. Ah, Bh, and ABh have been reported mainly in ing in either progressively weaker reactions or additional
individuals of European origin.52 No H, A, or B antigen is acquired pseudoantigens, which can be seen during forward
present in the saliva, yet anti-H is present in the serum. The grouping.55 Leukemia, chromosome 9 translocations, and
serum of Ah individuals contains anti-B and no anti-A, any hemolytic disease that induces stress hematopoiesis
although anti-A1 is usually present.52 (e.g., thalassemia) have been shown to depress antigen
In Bh serum, anti-A is always present and anti-B may strength.55 Often the cells will appear to show a mixed-field
be detected.52 It is postulated that homozygous inheri- agglutination (tiny agglutinates in a sea of unagglutinated
tance of a mutant H (FUT1) gene codes for the production cells). Hodgkin’s disease also has been reported to weaken or
of low levels of H transferase activity. The small amount depress ABH RBC antigens, resulting in variable reactions dur-
of H substance on the RBC is completely consumed by ing forward grouping similar to those found in leukemia.55
the A or B transferase present, resulting in small quantities The weakening of the antigen tends to follow the course
of A or B antigen being present on the RBC with no of the disease.55 The antigen strength increases again as the
detectable H antigen. The anti-H present in the serum is patient enters remission.
weaker in reactivity than the anti-H found in the Bombay The isoagglutinins (anti-A, anti-B, or anti-A,B) also may
phenotype, although it may be reactive at 37°C.54 be weak or absent in leukemias demonstrating hypogamma-
In the silenced FUT1 gene with the active FUT2 gene globulinemia, such as chronic lymphocytic leukemia (CLL).
para-Bombays, the α-2-L-fucosyltransferase enzyme asso- Various lymphomas, such as the malignant (non-Hodgkin’s)
ciated with the FUT2 gene (α2FucT2) produces H, A, and variety, may yield weak isoagglutinins, owing to moderate
B type 1 antigens in secretions, including plasma. These decreases in the gamma globulin fraction. Immunodefi-
type 1 antigens in the plasma may adsorb onto the RBC ciency diseases, such as congenital agammaglobulinemia,
membrane. H-deficient secretors have been found in a will also yield weak or absent isoagglutinins. If this problem
variety of ethnic groups and nationalities.53 RBCs have is suspected, a simple serum protein electrophoresis will
little or no A, B, and H antigens. RBCs of Oh secretors are confirm or rule out this condition. Individuals with intestinal
not agglutinated by most examples of anti-H but may be obstruction, carcinoma of the colon or rectum, or other
agglutinated by strong anti-H reagents. Adsorption and disorders of the lower intestinal tract may have increased
elution of anti-H may reveal the presence of some H anti- permeability of the intestinal wall, which allows passage of
gen on the RBC.53 Cells are not usually agglutinated by the bacterial polysaccharides from Escherichia coli serotype
anti-A and anti-B; however, some OhA RBCs can mimic the O86 into the patient’s circulation and results in the acquired B
behavior of Ax cells and can be agglutinated by anti-A,B phenomenon in group A1 individuals.55 The patient’s group A
and potent examples of anti-A. The same reactions with RBCs absorb the B-like polysaccharide, which reacts with
anti-A,B and potent examples of anti-B can be seen with human source anti-B.
OhB RBCs.23 A weak H-like antibody, called anti-IH, that A lack of detectable ABO antigens can occur in patients
is reactive at low temperature is almost always present in with carcinoma of the stomach or pancreas.55 The patient’s
the serum (see Chapter 8, “Blood Group Terminology and red blood cell antigens have not been changed, but the serum
Common Blood Groups”). This antibody is nonreactive contains excessive amounts of blood group–specific soluble
substances (BGSS) that may neutralize the antisera utilized
6888_Ch06_119-148 31/10/18 10:56 AM Page 136

136 PART II Blood Groups and Serologic Testing

in the forward grouping. All these disease states previously saline suspension of RBCs can usually resolve the ABO dis-
mentioned may result in discrepancies between the forward crepancy. It is important to make sure any and all technical
and reverse groupings, indicating that the patient’s red blood factors that may have given rise to the ABO discrepancy are
cell group is not what it seems. All ABO discrepancies must reviewed and corrected. It is also essential to obtain adequate
be resolved before blood for transfusion is released for that information regarding the patient’s age, diagnosis, transfu-
patient. In some cases, secretor or molecular studies may sion history, medications, and history of pregnancy. If
help confirm the patient’s true ABO group. the discrepancy persists and appears to be due to an error
in specimen collection or identification, a new sample must
ABO Discrepancies be drawn from the patient and all RBC and serum testing
repeated.
ABO discrepancies occur when unexpected reactions are ob- When a discrepancy is encountered, all results must be
tained in the forward and/or reverse grouping. These can be recorded, but interpretation of the ABO type must be delayed
due to problems with the patient’s serum (reverse grouping), until the discrepancy is resolved. If the blood is from a
problems with the patient’s RBCs (forward grouping), or potential transfusion recipient, it may be necessary to admin-
problems with both the serum and cells. The unexpected ister group O, Rh-compatible RBCs before the discrepancy is
reaction(s) may be due to an extra positive reaction or a resolved. In general, when investigating ABO discrepancies,
weak or missing reaction in the forward and reverse group- always remember that RBC and serum grouping reactions
ing. All ABO discrepancies must be resolved prior to report- are very strong (3+ to 4+) and the weaker reactions typically
ing a patient or donor ABO group. represent the discrepancy. Figure 6–14 shows an algorithm
for resolving ABO discrepancies.
Technical Errors
Categories of ABO Discrepancies
Technical errors can also cause ABO discrepancies. Examples
include: blood sample and test tube labeling errors, failure to ABO discrepancies may be arbitrarily divided into four
add reagents, or the addition of incorrect reagents or sample. major categories: group I, group II, group III, and group IV
Serum and antiserum should always be added first, followed discrepancies.
by the patient or reagent RBCs to avoid both reagent contam-
ination and potential omission of either patient sample or Group I Discrepancies
reagent. Results must be recorded immediately after obtaining Group I discrepancies are associated with unexpected reac-
them to avoid transcription errors. Always examine reagent tions in the reverse grouping due to weakly reacting or miss-
vials concurrently while performing ABO testing and quality ing antibodies. These discrepancies are more common than
control testing for possible contamination. Some of the com- those in the other groups listed. When a reaction in the
mon causes of technical errors leading to ABO discrepancies serum grouping is weak or missing, a group I discrepancy
in the forward and reverse groupings are listed in Box 6–8. should be suspected because, normally, RBC and serum
grouping reactions are very strong (4+). One of the reasons
Resolution for the missing or weak isoagglutinins is that the patient has
depressed antibody production or cannot produce the ABO
If initial testing was performed using RBCs suspended in
antibodies. Common populations with discrepancies in this
serum or plasma, repeat testing of the same sample using a
group are:
• Newborns (ABO antibody production is not detectable
until 3 to 6 months of age)
BOX 6–8
• Elderly patients (production of ABO antibodies is
Common Sources of Technical Errors Resulting depressed)
in ABO Discrepancies • Patients with a leukemia (e.g., chronic lymphocytic
leukemia) or lymphoma (e.g., malignant lymphoma)
• Incorrect or inadequate identification of blood specimens, test
tubes, or slides demonstrating hypogammaglobulinemia
• Cell suspension either too heavy or too light • Patients using immunosuppressive drugs that yield
• Clerical errors or incorrect recording of results hypogammaglobulinemia
• A mix-up in samples • Patients with congenital or acquired agammaglobulinemia
• Missed observation of hemolysis or immunodeficiency diseases
• Failure to add reagents • Patients with bone marrow or hematopoietic progenitor
• Failure to add sample stem cell (HPC) transplants (patients develop hypogam-
• Failure to follow manufacturer’s instructions maglobulinemia from therapy and start producing a
• Uncalibrated centrifuge different RBC population from that of the transplanted
• Overcentrifugation or undercentrifugation bone marrow)
• Contaminated reagents • Patients whose existing ABO antibodies may have been
• Warming during centrifugation diluted by plasma transfusion or exchange transfusion
• ABO subgroups
6888_Ch06_119-148 31/10/18 10:56 AM Page 137

Chapter 6 The ABO Blood Group System 137

ABO DISCREPANCY
Forward and Reverse testing
do not match as expected.
Note: The initial testing was
performed using patient’s RBCs If an error in specimen collection
suspended in serum or plasma. and identification is suspected.
Wash patient’s RBCs with
saline and repeat Testing.
Request a new sample to
be drawn from the patient.

No Discrepancy Same unexpected Rx between


Forward and Reverse testing
Repeat Testing

Report out ABO group Look up information


on Patient: No Discrepancy
Age, Diagnosis,
Medications,Transfusions,
and Pregnancy History Report out ABO group

Determine whether the discrepancy is in the Red Cell


or Serum results by observing weakest reactivity.

Example: Patient Serum Problem Example: Red Cell Problem Example: Both Serum and RBC Problem
Weak Reactions in the Weak Reactions in the Weak Reaction in both
Reverse grouping Forward grouping Forward and Reverse grouping

Anti-A 4⫹ Anti-A 2⫹mf Anti-A 1⫹


Anti-B 4⫹ Anti-B 0 Anti-B 1⫹
A1 Cell 2⫹ A1 Cell 0 A1 Cell 1⫹
B Cell 2⫹ B Cell 4⫹ B Cell 2⫹

Probable Group AB with Probable Group A with Probable Group O with


the following possibilities: the following possibilities: the following possibilities:
1. Cold Reacting Alloantibody 1. Out of Group Transfusion 1. Cold Autoantibody
(i.e. Anti-M, Anti-P1 most common) (i.e. Group O units 2. Cold Autoantibody and
2. Cold Reacting Autoantibody transfused to an A patient.) Cold Alloantibody
(i.e. Anti-I, Anti-H, Anti-IH) 2. Out of Group Bone Marrow/ 3. Out of Group Bone Marrow/
3. Passively Acquired Antibody Stem Cell Transplantation Stem Cell Transplantation
(i.e. plasma exchange, 3. Leukemia/Lymphoma 4. Passively Acquired Antibody
mismatched platelets) 4. Fetal-Maternal Bleed
4. Rouleaux 5. A3 Subgroup

Resolution:
Resolution: 1. Wash patient cells with
(Refer to chapter 11) warm saline and retest
1. Run Antibody Screen Resolution: 2. Run DAT and Auto Control
2. Run Auto Control (Refer to chapter 5) (Refer to chapter 5)
3. Run saline replacement 1. Run DAT 3. Run Antibody Screen
for rouleaux 2. Run Auto Control (Refer to chapter 11)

Figure 6–14. Algorithm for resolving ABO discrepancies.

Resolution of Common Group I Discrepancies serum by incubating the patient serum with reagent A1 and
Obtaining patient clinical history may immediately resolve B cells at room temperature for approximately 15 to 30 min-
this type of discrepancy. If the history indicates the patient utes. If there is still no reaction after centrifugation, the
is an elderly individual or has a diagnosis indicative of serum-cell mixtures can be incubated at 4°C for 15 to
hypogammaglobulinemia, the best way to resolve this dis- 30 minutes. An auto control and O cell control must always
crepancy is to enhance the weak or missing reaction in the be tested concurrently with the reverse typing when trying
6888_Ch06_119-148 31/10/18 10:56 AM Page 138

138 PART II Blood Groups and Serologic Testing

to solve the discrepancy since the lower temperature of lives of the individuals. In utero exchange of blood oc-
testing will most likely enhance the reactivity of other com- curs because of vascular anastomosis. As a result, two
monly occurring cold agglutinins, such as anti-I, that react cell populations emerge, both of which are recognized as
with all adult RBCs (see Chapter 8, “Blood Group Terminol- self, and the individuals do not make anti-A or anti-B.
ogy and Common Blood Groups”). Expected isoagglutinins are not present in the reverse
Table 6–16 shows a type of discrepancy that may be seen grouping, depending on the percentage of the population
with weak or missing antibodies. of RBCs that exist in each twin. If the patient or donor
The RBC results present a group O individual and the has no history of a twin, then the chimera may be due
serum results indicate an AB individual. Since serum prob- to dispermy (two sperm fertilizing one egg) and indi-
lems are more common, it is more likely that the serum cates mosaicism. More commonly, artificial chimeras
immunoglobulins are decreased. occur, which yield mixed cell populations as a result of
Mixed-field agglutination may look like small to large the following:
agglutinates with unagglutinated cells. Mixed-field may
also appear as a “halo” or “puff of smoke” of unagglutinated • Blood transfusions (e.g., group O cells given to an A or
RBCs as the RBC button is dislodged from the test tube B patient)
bottom. Common causes of mixed-field reactions include • Transplanted bone marrow or hematopoietic progenitor
receiving non-ABO-type specific RBCs, ABO subgroups (A3), stem cells (HPC) of a different ABO type
and bone marrow or hematopoietic stem cell transplantation. • Exchange transfusions
Before selecting RBCs for transfusion, it is imperative to • Fetal-maternal bleeding
investigate the patient’s transfusion records and clinical
history to verify the cause of the mixed-field agglutination. In contrast to transfusion medicine and solid organ
transplantation, ABO incompatibility is not a critical com-
Group I Discrepancies ponent of HPC donor selection.56 HPC engraftment is un-
restrained in the company of circulating ABO antibodies
Advanced Concepts since pluripotent and early-committed HPCs lack ABO
Chimerism is defined as the presence of two cell popula- antigens.3 A major ABO incompatibility occurs when the
tions in a single individual (Table 6–17). It was discovered donor’s red RBCs are incompatible with the recipient’s plasma
in twins born to a group O mother and group B father with (e.g., the donor is group B and the recipient is group O
a mixture of both B and O cells instead of the expected with naturally occurring anti-B).56 Complications of major
group of either B or O. Detecting a separate cell population ABO incompatibility include hemolysis of RBCs at time of
may be easy or difficult, depending on what percentage of infusion and continued antibody production directed
cells of the minor population are present. Reactions from against erythroid progenitors and donor RBCs by the en-
chimerism are typically mixed-field. grafted HPCs.3 A minor ABO incompatibility occurs when
True chimerism occurs only in twins and is rarely the donor’s plasma is incompatible with the recipient’s
found. The two cell populations will exist throughout the RBCs (e.g., the donor is group O with naturally occurring
anti-B and the recipient is group B). Bidirectional ABO

Table 6–16 Example of ABO Discrepancy Seen With Weak or Missing Antibodies
Forward Grouping Reaction of Patient’s Reverse Grouping Reaction of Patient’s
Cells With Serum With
Anti-A Anti-B A1 Cells B Cells
Patient 0 0 0 0
Patient’s probable group: O (elderly patient or newborn)

Note: The absence of agglutination with reagent cells in the reverse type is because the production of ABO antibodies can be weak or absent in the elderly.
Resolution: (1) Check age of the patient. (2) Increase incubation time to 30 minutes (not appropriate for newborn sample). (3) Lower the temperature to 4°C for 15 minutes
(include O cells and an autocontrol).

Table 6–17 ABO Grouping in Chimera Twins


Patient Anti-A Anti-B Anti-A,B A1 Cells B Cells RBC %
Twin 1 0 2+mf 2+mf 4+ 0 70% B; 30% O

Twin 2 0 +wk +wk 4+ 0 30% B; 70% O

0 = negative; mf = mixed-field; wk = weak


6888_Ch06_119-148 31/10/18 10:56 AM Page 139

Chapter 6 The ABO Blood Group System 139

incompatibility occurs when both a major and minor Table 6–19 shows the ABO testing results of an acquired
incompatibility are present (e.g., the donor is group A and B phenomenon.
the recipient is group B).56
Since ABO antigens are histo-blood group antigens Resolution of Common Group II Discrepancies
present on many tissues, including the lung, pancreas,
The agglutination of weakly reactive antigens with the
bowel, endothelium, heart, and kidney, recipients of ABO-
reagent antisera can be enhanced by incubating the test mix-
incompatible bone marrow or hematopoietic progenitor
ture at room temperature for up to 30 minutes to increase
stem cell transplantation pose distinctive challenges to
the association of the antibody with the RBC antigen. If the
transfusion service staff when selecting blood components
reaction is still negative, incubate the text mixture at 4°C for
for transfusion. Following an ABO-incompatible HPC
15 to 30 minutes. Include group O and autologous cells as
transplant, the pretransplant ABO type will remain in these
controls. RBCs can also be pretreated with enzymes and
tissues for the rest of the patient’s life. Accordingly, the
retested with reagent antisera.
patient will never make antibody against the ABO type the
The acquired B antigen arises when bacterial enzymes
body still sees as self (e.g., a group B recipient converted
modify the immunodominant blood group A sugar (N-acetyl-
to group A will never make anti-B). RBCs, platelets, and
D-galactosamine) into D-galactosamine, which is sufficiently
plasma products must be compatible with both the donor
similar to the group B sugar (D-galactose) to cross-react with
and recipient blood types (see Chapter 19, “Cellular Ther-
anti-B antisera. This pseudo-B antigen is formed at the ex-
apy in the Transplant Setting”).
pense of the A1 antigen and disappears following the patient’s
Group II Discrepancies recovery.55 The reaction of the appropriate antiserum with
these acquired antigens demonstrates a weak reaction, often
Group II discrepancies are associated with unexpected reac-
yielding a mixed-field appearance.
tions in the forward grouping due to weakly reacting or miss-
Blood group reagents of a monoclonal anti-B clone (ES4)
ing antigens. This group of discrepancies is probably the least
strongly agglutinate cells with the acquired B antigen. The
frequently encountered. The following are some of the
pH of reagents containing ES4 has been lowered, and conse-
causes of discrepancies in this group:
quently, only those cells with the strongest examples of ac-
• Subgroups of A or B may be present (see the “ABO Sub- quired B antigen react with the antisera. Testing the patient’s
groups” section). serum or plasma against autologous RBCs gives a negative re-
• Leukemias may yield weakened A or B antigens (Table 6–18), action, because the anti-B in the serum does not agglutinate
and Hodgkin’s disease has been reported in some cases to the patient’s RBCs with the acquired B antigen. The acquired
mimic the depression of antigens found in leukemia. B antigen is also not agglutinated when reacted with anti-B
• The “acquired B” phenomenon will show weak reactions that has a pH greater than 8.5 or less than 6.28 Secretor studies
with anti-B antisera and is most often associated with dis- can be performed when trying to characterize the acquired
eases of the digestive tract (e.g., cancer of the colon). B phenomenon. If the patient is in fact a secretor, only the

Table 6–18 Serologic Reactions Typical of Leukemia


Forward Grouping Reaction of Patient Reverse Grouping Reaction of Patient
Cells With Serum With
Patient Phenotype Anti-A Anti-B A1 Cells B Cells

A +mf 0 0 3+

B 0 ±/+ 4+ 0

Note: Weak reactivity with anti-A and anti-B is because the disease, leukemia, has resulted in the weakened expression of the corresponding antigen.

Table 6–19 Example of ABO Discrepancy Caused by an Acquired B Antigen


Forward Grouping Reaction of Patient’s Reverse Grouping Reaction of Patient’s
Cells With Serum With
Anti-A Anti-B A1 Cells B Cells

Patient 4+ 2+ 0 4+

Patient’s probable group: A

Note: Patient RBCs have acquired a B-like antigen that reacts with reagent anti-B and is associated with cancer of the colon or other diseases of the digestive tract.
Resolution: (1) Acidify Anti-B reagent to a pH of 6. (2) Run DAT (refer to Chapter 5, “The Antiglobulin Test”). (3) Run autocontrol.
6888_Ch06_119-148 31/10/18 10:56 AM Page 140

140 PART II Blood Groups and Serologic Testing

A substance is secreted in the acquired B phenomenon. Treat- this discrepancy is by repeating the forward type, using
ing RBCs with acetic anhydride reacetylates the surface antisera with a different lot number. If the cause of the
molecules, then markedly decreases the reactivity of the cells discrepancy is indeed a low-prevalence antibody in the
tested with anti-B. The reactivity of normal B cells is not reagent antisera reacting with a low-prevalence antigen
affected by treatment with acetic anhydride.28 on the patient’s cells, the antibody will most likely not be
present in a different lot number of reagent. This phenom-
Rare Group II Discrepancies
enon is only seen when human source antiserum is used.
Most ABO reagents in use today are monoclonal antibodies
Advanced Concepts and these reagents are free of contaminating antibodies to
Weakly reactive or missing reactions in RBC grouping may low-prevalence antigens.
be due to excess amounts of blood group–specific soluble
(BGSS) substances present in the plasma, which sometimes
occurs with certain diseases, such as carcinoma of the Group III Discrepancies
stomach and pancreas. Excess amounts of BGSS substances These discrepancies between forward and reverse groupings
will neutralize the reagent anti-A or anti-B, leaving no are caused by protein or plasma abnormalities and result in
unbound antibody to react with the patient cells and yields rouleaux formation or pseudoagglutination, attributable to
a false-negative or weak reaction in the forward grouping. the following:
Washing the patient cells free of the BGSS substances with
saline should alleviate the problem, resulting in correlating • Elevated levels of globulin from certain disease states, such
forward and reverse groupings. as multiple myeloma, Waldenström’s macroglobulinemia,
Antibodies to low-prevalence antigens in reagent anti-A other plasma cell dyscrasias, and certain moderately
or anti-B can also cause weakly reactive or missing reac- advanced cases of Hodgkin’s lymphomas
tions in RBC grouping (Table 6–20). It is impossible for • Elevated levels of fibrinogen
manufacturers to screen reagent antisera against all known • Plasma expanders, such as dextran and polyvinylpyrrolidone
RBC antigens. Even though rarely, it has been reported that • Wharton’s jelly in cord blood samples
this additional antibody in the reagent antisera has reacted • Table 6–21 shows an example of ABO discrepancy caused
with the corresponding low-prevalence antigen present on by rouleaux formation.
the patient’s RBCs. This produces an unexpected reaction
Resolution of Common Group III Discrepancies
of the patient’s cells with anti-A or anti-B or both, mimick-
ing the presence of a weak antigen. The best way to resolve Rouleaux is a stacking of erythrocytes that adhere in a coin-
like fashion, giving the appearance of agglutination. It can

Table 6–20 Example of ABO Discrepancy Caused by Low-Prevalence Antibodies


in the Reagent Antisera
Forward Grouping Reaction of Patient Reverse Grouping Reaction of Patient
Cells With Serum With
Anti-A Anti-B A1 Cells B Cells

Patient 0 1+ 4+ 4+

Patient’s probable group: O

Note: Reaction with anti-B in the forward type is due to agglutination between a low-prevalence antibody in reagent anti-B and the corresponding antigen on the patient’s cells.
Resolution: Use a different lot number for reagent anti-B.

Table 6–21 Example of ABO Discrepancy Caused by Rouleaux Formation


Forward Grouping Reaction of Patient Reverse Grouping Reaction of Patient
Cells With Serum With
Anti-A Anti-B A1 Cells B Cells

Patient 4+ 4+ 2+ 2+

Patient’s probable group: AB

Note: Agglutination with A1 and B cells in reverse type is due to rouleaux formation as a result of increased serum protein or plasma abnormalities.
Resolution: (1) Microscopic examination. (2) Saline replacement technique. (3) Wash cells with saline three times. (4) Run antibody screen (refer to Chapter 10).
6888_Ch06_119-148 31/10/18 10:56 AM Page 141

Chapter 6 The ABO Blood Group System 141

be observed on microscopic examination (Fig. 6–15). Cell


grouping can usually be accomplished by washing the
patient’s RBCs several times with saline. Performing a saline
replacement technique will free the cells in the case of
rouleaux formation in the reverse type. In this procedure,
serum is removed and replaced by an equal volume of saline.
In true agglutination, RBC clumping will still remain after
the addition of saline. Rouleaux can be a nuisance in the
laboratory, since it is an in vitro problem observed during
laboratory testing. It is not an in vivo problem for the patient.
The complete procedure for saline replacement can
be found in Procedure 11-3 on the textbook’s com-
panion website.
Cord blood samples can pose a problem in ABO testing,
since cord cells may be contaminated with a substance called
Wharton’s jelly. This substance is a viscous mucopolysaccha-
ride material present on cord blood cells that may cause the Figure 6–16. Autoagglutination in a patient with cold agglutinin disease.
red blood cells’ aggregation. Thoroughly washing cord cells
six to eight times with saline should alleviate spontaneous
rouleaux due to Wharton’s jelly and result in an accurate Resolution of Common Group IV Discrepancies
ABO grouping. However, since testing is usually not per- Potent cold autoantibodies can cause spontaneous agglutina-
formed on cord serum because the antibodies detected are tion of the patient’s cells. These cells often yield a positive direct
usually of maternal origin, reverse grouping may still not Coombs’ or antiglobulin test (see Chapter 21, “Autoimmune
correlate with the RBC forward grouping. Hemolytic Anemias”). If the antibody in the serum reacts with
Group IV Discrepancies all adult cells, for example, anti-I, the reagent A1 and B cells
These discrepancies between forward and reverse groupings used in the reverse grouping also agglutinate.
are due to miscellaneous problems that have the following To resolve this discrepancy, the patient’s RBCs could be
causes: incubated at 37°C for a short period, then washed with saline
at 37°C three times and retyped. If this is not successful in
• Cold reactive autoantibodies in which RBCs are so heavily resolving the forward type, the patient’s RBCs can be treated
coated with antibody that they spontaneously agglutinate, with 0.01 M dithiothreitol (DTT) to disperse IgM-related
independent of the specificity of the reagent antibody agglutination. As for the serum, the reagent RBCs and serum
(Fig. 6–16 and Table 6–22). can be warmed to 37°C for 10 to 15 minutes, mixed, tested,
• Circulating RBCs of more than one ABO group due to RBC and read at 37°C. The test can be converted to the antihuman
transfusion or marrow/stem cell transplant globulin phase if necessary. Weakly reactive anti-A or anti-B
• Unexpected ABO isoagglutinins may not react at 37°C, which is outside their optimum
• Unexpected non-ABO alloantibodies thermal range. If the reverse typing is still negative (and a
positive result was expected), a cold autoabsorption (patient
cells with patient serum) could be performed to remove the
cold autoantibody from the serum. The absorbed serum can
then be used to repeat the serum typing at room tempera-
ture. (Refer to Chapter 10 for cold autoadsorption and
alloadsorption with rabbit erythrocyte stroma [RESt] for the
removal of cold autoantibodies.)
Unexpected ABO isoagglutinins in the patient’s serum
react at room temperature with the corresponding antigen
present on the reagent cells (Table 6–23). Examples of this
type of ABO discrepancy include A2 and A2B individuals,
who can produce naturally occurring anti-A1, or A1 and A1B,
individuals who may produce naturally occurring anti-H.
(Refer to the previous sections on ABO subgroups.) Serum
grouping can be repeated using at least three examples of A1,
A2, B cells; O cells; and an autologous control (patient’s
serum mixed with patient’s RBCs).3 The specificity of the
antibody can be determined by examining the pattern of
Figure 6–15. Rouleaux. reactivity (e.g., if the antibody agglutinates only A1 cells, it
6888_Ch06_119-148 31/10/18 10:56 AM Page 142

142 PART II Blood Groups and Serologic Testing

Table 6–22 Example of ABO Discrepancy Caused by Cold Autoantibodies


Forward Grouping Reaction of Patient Reverse Grouping Reaction of Patient
Cells With Serum With
Anti-A Anti-B A1 Cells B Cells

Patient 2+ 4+ 4+ 2+

Patient’s probable group: B

Note: Reaction with anti-A in forward type is due to spontaneous agglutination of antibody-coated cells; reaction with B cells in reverse type is due to cold autoantibody (e.g., anti-I)
reacting with I antigen on B cells.
Resolution: (1) Wash patient cells with warm saline and retest. (2) Run DAT and autocontrol. (3) Run antibody screen (refer to Chapter 10).

Table 6–23 Example of ABO Discrepancy Caused by an Unexpected ABO Isoagglutinin


Forward Grouping Reaction of Patient Reverse Grouping Reaction of Patient
Cells With Serum With
Anti-A Anti-B A1 Cells B Cells

Patient 4+ 4+ 1+ 0

Patient’s probable group: A2B

Note: Reactions with patient serum are due to anti-A1 agglutinating A1 reagent red blood cells.
Resolution: (1) Test cells with anti-A1 lectin. (2) Test serum with A1, A2, and O cells. (3) Run an autocontrol.

can most likely be identified as anti-A1). The patient’s RBCs onto patient’s RBCs. One example is an individual with
can be tested with Dolichos biflorus to confirm the presence an antibody against acriflavine, the yellow dye used
of the ABO subgroup. Dolichos biflorus will agglutinate cells in some commercial anti-B reagents. The acriflavine-
of the A1 but not the A2 phenotype. antiacriflavine complex attaches to the patient’s RBCs,
Unexpected alloantibodies in the patient’s serum other causing agglutination in the forward type. Washing the
than ABO isoagglutinins (e.g., anti-M) may cause a discrep- patient’s cells three times with saline and then retyping
ancy in the reverse grouping (Table 6–24). Reverse grouping them should resolve this discrepancy.
cells possess other antigens in addition to A1 and B, and it Cis-AB refers to the inheritance of both AB genes from
is possible that other unexpected antibodies present in the one parent carried on one chromosome and an O gene in-
patient’s serum will react with these cells. In this situation, herited from the other parent.57 This results in the offspring
an antibody identification panel should be performed with inheriting three ABO genes instead of two (Fig. 6–17). The
the patient’s serum. Once the unexpected alloantibodies are designation cis-AB is used to distinguish this mode of in-
identified, reagent A1 and B cells negative for the correspond- heritance from the more usual AB phenotype in which the
ing antigen should be used in the reverse grouping. alleles are located on different chromosomes. The cis-AB
phenotype was first discovered in 1964, when a Polish fam-
Rare Group IV Discrepancies
ily was described in which the father was group O and the
mother was group AB and gave birth to children who were
Advanced Concepts all group AB.23 It was resolved by the fact that the A and
Antibodies other than anti-A and anti-B may react to B genes were inherited together and were both on the same,
form antigen-antibody complexes that may then adsorb or cis, chromosome; thus the term cis-AB.

Table 6–24 Example of ABO Discrepancy Caused by an Unexpected Non-ABO Alloantibody


Forward Grouping Reaction of Patient Reverse Grouping Reaction of Patient
Cells With Serum With
Anti-A Anti-B A1 Cells B Cells

Patient 4+ 4+ 1+ 1+

Patient’s probable group: AB

Note: Reactions with patient serum are due to non-ABO alloantibody agglutinating an antigen other than A1 and B on reagent red blood cells.
Resolution: (1) Run an antibody screen and panel (refer to Chapter 10).
6888_Ch06_119-148 31/10/18 10:56 AM Page 143

Chapter 6 The ABO Blood Group System 143

RBCs with the cis-AB phenotype (a rare occurrence)


express a weakly reactive A antigen (analogous to A2 cells)
Group: AB Group: O and a weak B antigen.23 The B antigen usually yields a
Genotype: AB Genotype: OO
Chromosomes:
weaker reaction with the anti-B from random donors, with
Chromosomes:
A A O O mixed-field agglutination typical of subgroup B3 reported
B B in several cases. Weak anti-B (present in the serum of most
unequal crossing-over
cis-AB individuals) leads to an ABO discrepancy in the
(rare occurrence) reverse grouping. The serum of most cis-AB individuals
contains a weak anti-B, which reacts with all ordinary B
RBCs, yet not with cis-AB RBCs. A and B transferase levels
are lower than those found in ordinary group AB sera.57
Various hypotheses have been offered to explain the
cis-AB phenotype. Many favor an unequal crossing over be-
tween the A and B gene with gene fusion and the formation
Group: AB Group: O
Genotype: ABO (cis-AB) Genotype: OO of a new gene. However, the banding pattern of the distal
Chromosomes: Chromosomes: end of the long arm of chromosome 9 representing the ABO
A O
locus is normal. There have been other examples of cis-ABs
O O
B that do not fit the above scenario. In these examples, there
was a point mutation at the ABO locus, and an enzyme was
produced that was capable of transferring both A-specific
Group: AB and B-specific sugars to the precursor molecule.57 Many
Genotype: ABO (cis-AB) families have been reported in other parts of the world,
Chromosomes: with a high incidence of cis-AB being found in Japan.
A O
Table 6–25 provides some examples of serologic reac-
B
tions involving ABO discrepancies, with possible causes
Figure 6–17. Example of cis-AB inheritance to unequal crossing over. 䊊 = male; and resolution steps. Figure 6–18 provides a simplified
䊐 = female summary of ABO discrepancies.

Table 6–25 ABO Discrepancies Between Forward and Reverse Grouping


Forward
Grouping Reverse Grouping
Patient Anti-A Anti-B A1 Cells B Cells O Cells Autocontrol Possible Cause Resolution Steps

1 0 0 0 0 0 0 Group O newborn or elderly Check age and diagnosis


patient may have hypogam- of patient and im-
maglobulinemia or agamma- munoglobulin levels; if
globulinemia or may be taking possible incubate at RT
immunosuppressive drugs for 30 min or at 4°C for
15 min; include group
O and autologous cells
at 4°C
2 4+ 0 1+ 4+ 0 0 Subgroup of A: probable A2 React patient cells
with anti-A1 with anti-A1 lectin, test
serum against addi-
tional A1, A2, and O
cells; run autocontrol.
3 4+ 4+ 2+ 2+ 2+ 2+ (1) Rouleaux (multiple (1) Wash RBCs; use
myeloma patient; any patient saline dilution or saline
with reversed albumin-to- replacement technique
globulin ratio or patients
given plasma expanders)

(2) Cold autoantibody (2) Perform cold panel


(probable group AB with and autoabsorb or
an auto anti-I) rabbit erythrocyte
stroma (RESt) absorb
(see Chapter 10) or
reverse type at 37°C
Continued
6888_Ch06_119-148 31/10/18 10:56 AM Page 144

144 PART II Blood Groups and Serologic Testing

Table 6–25 ABO Discrepancies Between Forward and Reverse Grouping—cont’d


Forward
Grouping Reverse Grouping
Patient Anti-A Anti-B A1 Cells B Cells O Cells Autocontrol Possible Cause Resolution Steps

(3) Cold autoantibody with (3) Perform cold panel


underlying cold or RT reacting autoabsorb or RESt,
alloantibody (probable group and run panel on ab-
AB with an auto anti-I and a sorbed serum; select
high-frequency cold antibody reverse cells lacking
[e.g., anti-P1, anti-M, anti-Leb]) antigen for identified
alloantibody; repeat
reverse group on
absorbed serum to
determine true
ABO group or at 37°C

4 4+ 4+ 1+ 0 0 0 Subgroup of AB; probable A2B Use anti-A1 lectin, test


with anti-A1 serum against addi-
tional A1, A2, and
O cells

5 4+ 0 0 4+ 3+ 0 A1 with potent anti-H Confirm A1 group with


anti-A1 lectin; test
additional A2, O, and
A1 cells and an Oh if
available

6 0 0 4+ 4+ 4+ 0 Oh Bombay Test with anti-H lectin;


test Oh cells if available;
send to reference labo-
ratory for confirmation

7 0 0 2+ 4+ 0 0 Subgroup of A; probable Ax Perform saliva studies or


with anti-A1 absorption/elution

8 4+ 2+ 0 4+ 0 0 Group A with an acquired B Check history of patient


antigen for lower gastrointesti-
nal problem or sep-
ticemia; acidify anti-B
typing reagent to pH
6.0 by adding 1 or
2 drops of 1 N HCl to
1 mL of anti-B antisera,
and measure with a
pH meter (this acidified
anti-B antisera would
agglutinate only true
B antigens not acquired
B antigens), test serum
against autologous cells

9 4+ 4+ 2+ 0 2+ 0 Group AB with cold antibody Perform antibody screen


and panel, identify
room temperature
antibody, repeat serum
type with antigen
negative reagent cells
or perform serum type
at 37°C

10 0 4+ 4+ 1+ 1+ 1+ Group B with cold autoantibody Enzyme-treat RBCs and


perform autoabsorption
at 4°C or perform
prewarmed testing

*Absorption should not be performed on patient’s cells that have been transfused within the last 3 months
RT = room temperature
6888_Ch06_119-148 31/10/18 10:56 AM Page 145

Chapter 6 The ABO Blood Group System 145

Causes of ABO Discrepancies

Forward Reverse
(RBCs) (plasma)

Missing/weak Extra Mixed Missing/weak Extra


Antigens Antigens Field Antibodies Antibodies

A or B Acquired B Group O Anti-A1


Newborns,
Subgroups (intestinal transfusions
Elderly,
disease) Immunocom-
promised

Disease Rouleaux Stem Cell Cold


(leukemia) (hyperpro- Transplant Alloanti-
teinemia) bodies

Cold Auto- A3 or B3 Rouleaux


antibodies Phenotype (hyperpro-
teinemia)

Wharton’s Cold Auto-


jelly antibodies

Passively
Acquired Antibody
(i.e. plasma exchange,
Figure 6–18. Simplified summary of ABO mismatched
discrepancies. platelets)

CASE STUDY 1. Where is the discrepancy?


2. What testing would you perform next to resolve the
Case 6-1 discrepancy?
A 45-year-old woman, who has given birth to three Part 2
children and has a history of five cases of dilation and The patient’s serum was then tested with A2 cells and
curettage, is scheduled for a partial hysterectomy at a O cells and the patient’s red blood cells tested with
community hospital. Preoperative laboratory tests include Anti-A1 lectin.
a type and screen. There is no history of transfusions. A2 Cells O Cells Anti-A1 Lectin
Part 1 Patient serum 0 0 Patient RBCs 0
ABO and Rh Typing
3. How would you interpret these results?
Anti-A Anti-B Anti-A,B A1Cells B Cells Anti-D 4. Why were O POS RBCs chosen for transfusion?
3+ 0 3+ 2+ 4+ 3+
Antibody Screen
37°C AHG CC
SCI 0 0 √
SCII 0 0 √
SCIII 0 0 √
6888_Ch06_119-148 31/10/18 10:56 AM Page 146

146 PART II Blood Groups and Serologic Testing

SUMMARY CHART
 ABO frequencies: group O, 45%; group A, 40%; group  Group O persons have the greatest amount of H sub-
B, 11%; group AB, 4%. stance; group A1B persons contain the least amount of
 ABO blood group system has naturally occurring anti- H substance.
bodies that are primarily IgM.  Approximately 80% of individuals inherit the A gene
 ABO genes, like those of most other blood groups, are phenotype as A1; the remaining 20% phenotype as A2
inherited in a codominant manner. or weaker subgroups.
 ABH-soluble antigens are secreted by tissue cells and  Approximately 1% to 8% of A2 persons produce anti-
are found in all body secretions. The antigens secreted A1 in their serum.
depend on the person’s ABO group.  Glycoproteins in secretions are formed on type 1 pre-
 ABO reverse grouping is omitted from cord blood test- cursor chains.
ing on newborns because their antibody titer levels are  The ABH antigens on RBCs are formed on type 2 pre-
generally too low for detection. cursor chains.
 ABO RBC antigens can be glycolipids, glycoproteins,  Forward and reverse grouping normally yield strong
or glycosphingolipids; ABO-secreted substances are (3+ to 4+) reactions.
glycoproteins.  Group A persons have anti-B in their serum; group
 L-fucose is the immunodominant sugar responsible for B persons have anti-A in their serum; group AB per-
H specificity. sons have neither anti-A nor anti-B in their serum;
 N-acetylgalactosamine is the immunodominant sugar group O persons contain both anti-A and anti-B in
responsible for A specificity. their serum.
 D-galactose is the immunodominant sugar responsible  Approximately 78% of the random population inherit
for B specificity. the Se gene and are termed secretors; the remaining
 The hh genotype is known as the Bombay phenotype, 22% inherit the se gene and are termed nonsecretors.
or Oh, and lacks normal expression of the ABH  The Se gene codes for the production of
antigens. L-fucosyltransferase.

Review Questions 4. The immunodominant sugar responsible for blood group


A specificity is:
1. An ABO type on a patient gives the following reactions: a. L-fucose.
b. N-acetyl-D-galactosamine.
Patient Cells With Patient Serum With c. D-galactose.
Anti-A Anti-B A1 cells B cells d. Uridine diphosphate-N-acetyl-D-galactose.

4+ 4+ Neg Neg 5. What ABH substance(s) would be found in the saliva of


a group B secretor?
What is the patient’s blood type? a. H
a. O b. H and A
b. A c. H and B
c. B d. H, A, and B
d. AB
6. An ABO type on a patient gives the following reactions:
2. The major immunoglobulin class(es) of anti-B in a group
A individual is (are): Patient Cells With Patient Serum With
a. IgM. Anti-A Anti-B Anti-A1 A1 cells B cells
b. IgG.
c. IgM and IgG. 4+ 4+ Neg 2+ Neg
d. IgM and IgA.
The reactions above may be seen in a patient who is:
3. What are the possible ABO phenotypes of the offspring a. A1 with acquired B.
from the mating of a group A to a group B individual? b. A2B with anti-A1.
a. O, A, B c. AB with increased concentrations of protein in the
b. A, B serum.
c. A, B, AB d. AB with an autoantibody.
d. O, A, B, AB
6888_Ch06_119-148 31/10/18 10:56 AM Page 147

Chapter 6 The ABO Blood Group System 147

7. Which of the following ABO blood groups contains the 9. Klein HG, Anstee DJ. Mollison’s Blood Transfusion in Clinical
least amount of H substance? Medicine. 12th ed. Somerset (NJ): Wiley-Blackwell Publishing;
2014.
a. A1B 10. American Association of Blood Bank Standards. 31th ed.
b. A2 Bethesda (MD): American Association of Blood Banks; 2018.
c. B 11. Olsson ML, Chester MA. Polymorphism and recombination
d. O events at the ABO locus: a major challenge for genomic ABO
blood grouping strategies. Transfus Med. 2001;11:295-313.
8. You are working on a specimen in the laboratory that 12. Chester MA, Olsson ML. The ABO blood group gene: a locus
you believe to be a Bombay phenotype. Which of the fol- of considerable genetic diversity. Transfus Med Rev. 2001;15:
177-200.
lowing reactions would you expect to see? 13. Yamamoto F, Cid E, Yamamoto M, Saitou N, Bertranpetit J,
a. Patient’s cells + Ulex europaeus = no agglutination Blancher A. An integrative evolution theory of histo-blood
b. Patient’s cells + Ulex europaeus = agglutination group ABO and related genes. Scientific Reports. 2014;4:6601.
c. Patient’s serum + group O donor RBCs = no doi:10.1038/srep06601.
14. Yamamoto F. Molecular genetics of ABO. Vox Sang. 2000;78
agglutination (2 Suppl 2):91-103. Review.
d. Patient’s serum + A1 and B cells = no agglutination 15. Lee AH, Reid ME. ABO blood group system: a review of mo-
lecular aspects. Immunohematology. 2000;16:1-6.
9. An example of a technical error that can result in an 16. Yamamoto F, Cid E, Yamamoto M, Blancher A. ABO
ABO discrepancy is: research in the modern era of genomics. Transfus Med Rev. 2012;
a. Acquired B phenomenon. 26(2):103-18.
b. Missing isoagglutinins. 17. Yamamoto F. Cloning and regulation of the ABO genes. Transfus
Med. 2001; 11:281-94.
c. Cell suspension that is too heavy.
18. Seltsam A, Hallensleben M, Kollmann A, Burkhart J, Blasczyk, R.
d. Acriflavine antibodies. Systematic analysis of ABO gene diversity within exons 6 and
7 by PCR screening reveals new ABO alleles. Transfusion. 2003;
10. An ABO type on a patient gives the following reactions: 43:428-39.
19. Seltsam A, Hallensleben M, Kollmann A, Blasczyk, R. The nature
Patient Cells of diversity and diversification at the ABO locus. Blood. 2003;
102:3035-42.
With Patient Serum With 20. Hosseini-Maaf B, Hellberg A, Chester MA, Olsson ML. An ex-
tensive polymerase chain reaction-allele-specific polymor-
Anti-A Anti-B A1 cells B cells O cells Autocontrol
phism strategy for clinical ABO blood group genotyping that
4+ Neg 2+ 4+ 2+ Neg avoids potential errors caused by null, subgroup, and hybrid
alleles. Transfusion. 2007;47(11):2110-25.
These results are most likely due to: 21. Blumenfeld OO. The ABO gene—more variation! Blood.
2003;102:2715.
a. ABO alloantibody. 22. Sano R, Nakajima T, Takahashi Y, Kubo R, Kobayashi M,
b. Non-ABO alloantibody. Takahashi M, et al. Epithelial expression of human ABO group
c. Rouleaux. genes is dependent upon a downstream regulatory element
d. Cold autoantibody. functioning through an epithelial cell-specific transcription
factor, Elf5. J Biol Chem. 2016;291(43):22594-606. doi:10.1074/
jbc.M116.730655.
References 23. Watkins WM. The ABO blood group system: historical back-
ground. Transfus Med. 2001;11:243-65.
1. U.S. Food and Drug Administration [Internet]. Silver Spring 24. Gorakshakar AC, Ghosh K. Use of lectins in immunohematol-
(MD): Fatalities reported to FDA following blood collection and ogy. Asian Journal of Transfusion Science. 2016;10(1):12-21.
transfusion: annual summary for fiscal year 2015. [Cited 2017 doi:10.4103/0973-6247.172180.
March 10]. Available from: www.fda.gov/BiologicsBloodVaccines/ 25. Fujitani N, Liu Y, Toda S, Shirouzu K, Kimura H. Expression
SafetyAvailability/ReportaProblem/TransfusionDonationFatalities/ of H type 1 antigen of ABO histo–blood group in normal colon
ucm518148.pdf. and aberrant expressions of H type 2 and H type 3/4 antigens
2. Landsteiner K. Uber agglutinationserscheinungen normalen in colon cancer. Glycoconj J. 2000;17:331-8.
menschlichen blutes. Wien Klin Wschr. 1901;14:1132-4. 26. Mizuno N, Ohmori T, Sekiguchi K, Kato T, Fujii T, Fujii K, et al.
3. Fung MK, Eder AF, Spitalnik SL, et al. Technical Manual Alleles responsible for ABO phenotype-genotype discrepancy
19th Edition. AABB; 2017. and alleles in individuals with a weak expression of A or B anti-
4. Garratty G, Glynn SA, McEntire R. ABO and Rh (D) phenotype gens. J Forensic Sci. 2004;49(1):21-8.
frequencies of different racial/ethnic groups in the United States. 27. Seltsam A, Blasczyk R. Missense mutations outside the catalytic
Transfusion. 2004;44:703-6. domain of the ABO glycosyltransferase can cause weak blood
5. Daniels G, Bromilow I. Essential Guide to Blood Groups. group A and B phenotypes. Transfusion. 2005;45(10):1663-9.
Somerset (NJ): Wiley-Blackwell Publishing; 2013. 28. Daniels, G. Human blood groups. 3rd ed. Oxford, UK: Wiley-
6. Landsteiner K, Witt DH. Observations on the human blood Blackwell; 2013.
groups: Irregular reactions. Isoagglutinin in sera of group 4. The 29. Novaretti MCZ, Domingues AE, Pares MMNS, Dorlhiac-Llacer
fact of A1. J Immunol. 1967;2:221. PE, Chamone DAF. Rapid detection of 871G>A mutation by
7. Dodd BE, Lincoln PJ, Boorman KE. The cross-reacting antibod- sequence specific PCR in ABO*A301 blood donors. Blood. ASH
ies of group O sera: immunological studies and a possible Annual Meeting Abstracts. 2004;104:4098.
explanation of the facts. Immunology. 1967;12:39. 30. Barjas-Castro ML, Carvalho MH, Locatelli MF, Bordin S, Saad ST.
8. Murphy MF, Pamphilon DH. Practical transfusion medicine. Molecular heterogeneity of the A3 group. Clin Lab Haemato.
3rd ed. Hoboken (NJ): Wiley-Blackwell; 2009. 2001;22:73-8.
6888_Ch06_119-148 31/10/18 10:56 AM Page 148

148 PART II Blood Groups and Serologic Testing

31. Olsson ML, Irshaid NM, Hosseini-Maaf, B, Hellberg A, Moulds specificity of human blood group B-synthesizing galactosyl-
MK, Sareneva H, et al. Genomic analysis of clinical samples transferase. J Biol Chem. 2003;278:12403-5.
with serologic ABO blood grouping discrepancies: identifica- 44. Seltsam A, Gruger D, Just B, Fiqueiredo C, Gupta CD,
tion of 15 novel A and B subgroup alleles. Blood. 2001; Deluca DS, et al. Aberrant intracellular trafficking of a variant
98:1585-93. B glycosyltransferase. Transfusion. 2008;48(9):1898-1905.
32. Deng ZH, Yu Q, Wu GG, Lian YL, Su YQ, Li DC, et al. Molec- 45. Koscielak J, Pacuszka T, Dzierkowa-Borodej W. Activity of
ular genetic analysis for Ax phenotype of the ABO blood group B-gene-specified galactosyltransferase in individuals with Bm
system in Chinese. Vox Sang. 2005;89(4):251-6. phenotypes. Vox Sang. 1976;30:58-67.
33. Hosseini-Maaf B, Hellberg Å, Rodrigues MJ, Chester MA, 46. Lin PH, Li L, Lin-Tsai SJ, Lin KT, Chen JM, Chu DC. A unique
Olsson ML. ABO exon and intron analysis in individuals with 502C>T mutation in exon 7 of ABO gene associated with the
the A weak B phenotype reveals a novel O1v-A2 hybrid allele Bel phenotype in Taiwan. Transfusion. 2003;43:1254-9.
that causes four missense mutations in the A transferase. BMC 47. Boose GM, Issitt C, Issitt PD. Weak B antigen in a family. Trans-
Genetics. 2003;4:17. doi:10.1186/1471-2156-4-17. fusion. 1978; 18:570-1.
34. Catron JP, Gerbal A, Badet J, Ropars C, Salmon C. Assay of 48. Bhende YM, Deshpande CK, Bhatia HM, Sanger R, Race RR,
alpha-N-acetylgalactosaminyltransferase in human sera: fur- Morgan WT, et al. A “new” blood group characteristic related
ther evidence for several types of Am individuals. Vox Sang. to the ABO system. Lancet. 1952;1:9034.
1975;28:347-65. 49. Storry JR, Johannesson JS, Poole J, Strindberg J, Rodrigues MJ,
35. Asamura, H, Ota M, Takayanagi K, Saito S, Tsukada K, Yahalam V, et al. Identification of six new alleles at the FUT1
Fukushima H. Molecular genetic analysis of the Am phenotype and FUT2 loci in ethnically diverse individuals with Bombay
of the ABO blood group system. Vox Sang. 2002;83:263-7. and para-Bombay phenotypes. Transfusion. 2006;46(12):
36. Sun CF, Chen DP, Tseng CP, Wang WT, Liu JP. Identification of 2149-55.
a novel A1v-O1v hybrid allele with G829A mutation in a 50. Verma A, Vani KG, Chaitanya Kumar IS, Jothi Bai DS. Preva-
chimeric individual of AelBel phenotype. Transfusion. 2006; lence of Bombay blood group in a tertiary care hospital, Andhra
46(5):780-9. Pradesh, India. Asian J Transfusion Sci. 2011;5:57-58.
37. Yazer MH, Hosseini-Maaf B, Olsson ML. Blood grouping dis- 51. Wagner FF, Flegel WA. Polymorphism of the h allele and the
crepancies between ABO genotype and phenotype caused by population frequency of sporadic nonfunctional alleles. Trans-
O alleles. Curr Opin Hematol. 2008;15(6):618-24. fusion. 1997;37:284-90.
38. Zhu F, Tao S, Xu X, Ying Y, Hong X, Zhu H, et al. Distribution 52. Salmon C, Cartron JP, Rouger P, Liberge G, Juszczak G,
of ABO blood group allele and identification of three novel Mulet C, et al. H-deficient phenotypes: a proposed practical
alleles in the Chinese Han population. Vox Sang. 2010; classification of Bombay Ah, H2, Hm. Blood Transfus Immuno-
98(4):554-9. haematol. 1980;23:233-48.
39. Cai XH, Jin S, Liu X, Shen W, Lu Q, Wang JL, et al. Molecular 53. Storry JR, Olsson ML. The ABO blood group system revis-
genetic analysis for the B subgroup revealing two novel alleles ited: a review and update. Immunohematology. 2009;25(2):
in the ABO gene. Transfusion. 2008;48(11):2442-47. 48-59.
40. Cho D, Yazer MH, Shin M, et al. Dispermic chimerism in a 54. Yan L, ZhuF, Xu X, Hong X, Lv Q. Molecular basis for para-
blood donor with apparent B3 blood group and mosaic 47, XYY Bombay phenotypes in Chinese persons, including a novel
syndrome. Blood. ASH Annual Meeting Abstracts. 2005; nonfunctional FUT1 allele. Transfusion. 2005;45:725-730.
106(11):1896. 55. Liumbruno GM, Franchini M. Beyond immunohaematology:
41. Hosseini-Maaf B, Letts JA, Persson M, Smart E, LePennec PY, the role of the ABO blood group in human diseases.
Hustinx H, et al. Structural basis for red cell phenotypic Blood Transfusion. 2013;11(4):491-499. doi:10.2450/2013
changes in newly identified, naturally occurring subgroup .0152-13.
mutants of the human blood group B glycosyltransferase. 56. Booth GS, Gehrie EA, Bolan CD, Savani, BN. Clinical guide to
Transfusion. 2007;47(5):864-75. ABO-incompatible allogeneic stem cell transplantation. Biol
42. Seltsam A, Wagner FF, Gruger D, Gupta CD, Bade-Doeding C, Blood Marrow Transplant. 2013;19:1152-8.
Blasczyk, R. Weak blood group B phenotypes may be caused 57. Chun S, Ryu MR, Cha S-Y, Seo J-Y, Cho D. ABO mistyping of
by variations in the CCAAT-binding factor/NF-Y enhancer cis-AB blood group by the automated microplate technique.
region of the ABO gene. Transfusion. 2007; 47(12):2330-5. Transfusion Medicine and Hemotherapy. 2018;45(1):5-10.
43. Marcus SL, Polakowski R, Seto NO, Leinala E, Borisova S, doi:10.1159/000475506.
Blancher A, et al. A single point mutation reverses the donor
6888_Ch07_149-172 31/10/18 9:46 AM Page 149

Chapter 7
The Rh Blood Group System
Gale Suwe, MT(ASCP)SBB and Merilyn Wiler, MAEd, MT(ASCP)SBB

Introduction Weak D: Variations of D Antigen Rh Deficiency Syndrome: RhNULL and RhMOD


History Expression Unusual Phenotypes and Rare Alleles
Terminology Position Effect: C in Trans to D Cw
Fisher-Race: DCE Terminology Weak D: Quantitative Changes Due to f (ce)
Wiener: Rh-Hr Terminology Fewer D Antigen Sites rhi (Ce)
Rosenfield and Coworkers: Del G
Alphanumeric Terminology Partial D or D Mosaic Rh17 (Hr0)
International Society of Blood D Epitopes on RhCE Protein Rh23, Rh30, Rh40 and Rh52
Transfusion Committee: Updated RH Antibodies Rh33 (Har)
Numeric Terminology Characteristics of Rh Antibodies Rh32
Rh Nomenclatures Overview Rh Antibodies in Pregnancy Rh43 (Crawford)
Genetics Testing for RH Antigens and Antibodies e Variants
Rh Genes Rh Typing Reagents V and VS
RH-Associated Glycoprotein Rh Testing Requirements for
(RHAG) Deletions
Pretransfusion, Obstetric Patients,
Rh-Positive Phenotypes and Blood Donors Landsteiner Wiener Blood Group System
Rh-Negative Phenotypes Detecting and Identifying Rh Case Studies
Most Probable Genotypes Antibodies Case 7-1
RH Antigens Selecting Compatible Blood for Case 7-2
Biochemistry Transfusion Case 7-3
Antigen Characteristics Clinical Considerations Summary Chart
D Antigen Transfusion Reactions Review Questions
Other Common Rh Antigens (C, E, Hemolytic Disease of the Fetus References
c, e) and Newborn

OBJECTIVES
1. Explain the derivation of the term Rh.
2. Compare and contrast the Fisher-Race and Wiener theories of Rh inheritance.
3. Translate the five major Rh antigens, haplotypes, and predicted haplotypes, from one nomenclature to another, including
Fisher-Race, Wiener, and Rosenfield.
4. Describe the interaction of the RHD and RHCE genes with the RHAG gene.
5. Define the basic biochemical structure of Rh antigens.
6. Compare and contrast the genetic pathways for the regulator type of Rhnull and the amorphic Rhnull.
7. Describe the role of RHAG in antigen expression.
8. Describe and differentiate mechanisms that result in weakened expression of D on red blood cells.
9. Describe the requirements for Rh testing for pretransfusion and obstetric patients and compare with the requirements
for blood donors.
10. Discuss Rh typing reagents for both routine and special testing.
11. Interpret Rh typing results and resolve any discrepancies if present.
Continued

149
6888_Ch07_149-172 31/10/18 9:46 AM Page 150

150 PART II Blood Groups and Serologic Testing

OBJECTIVES—cont’d
12. Define the characteristics of Rh antibodies.
13. Describe the risk of hemolytic disease of the fetus and newborn as it relates to anti-D.
14. Describe unusual Rh phenotypes and alleles in terms of prevalence and the challenges in transfusion.
15. Determine the most probable genotype of an individual when given the red blood cell typing results and ethnicity.

Introduction antibody agglutinated 85% of human RBCs and was named


anti-Rh after the rhesus monkey.
This chapter describes in detail the Rh blood group system. A subsequent investigation by Levine and coworkers3
The Rh blood group is highly complex, and the alloimmu- demonstrated that the agglutinin (antibody causing direct
nization to Rh blood group antigens can complicate trans- agglutination of RBCs) causing the hemolytic transfusion re-
fusion and pregnancy. It is imperative to have a basic action, and the antibody described by Landsteiner and
understanding of Rh, as RhD typing is a critical component Wiener2 appeared to define the same blood group. Many
of pretransfusion and prenatal testing. years later it was recognized that the two antibodies were
The term Rh refers to a specific red blood cell (RBC) anti- different. However, the name Rh was retained for the human-
gen (D) and to a complex blood group system currently com- produced antibody, and anti-rhesus formed by the animals
posed of 61 different antigenic specificities. Although Rh was renamed anti-LW in honor of those first reporting it
antibodies were among the first to be described, researchers (Landsteiner and Wiener).
continue to explore the complexities of the Rh blood group By the mid-1940s, five antigens were defined in the Rh
system from its serology to its mode of inheritance, genetic system. In the 1980s, molecular testing further defined the
control, and the biochemical structure of the Rh antigens. structure of the RH genes. The Rh blood group system con-
Rh is the second in importance only to ABO blood group tinues to be explored, and at present, over 60 different Rh
system in terms of transfusion, as the Rh system antigens are antigen specificities have been identified.
very immunogenic. Unlike ABO antibodies that are routinely
found in individuals who lack the corresponding antigen, Terminology
Rh antibodies are produced only after exposure to foreign
red blood cells. Once present, they can produce significant There are four terminologies used to describe the Rh system.
hemolytic disease of the fetus and newborn (HDFN) as well Two are based on postulated genetic theories of Rh inheri-
as hemolytic transfusion reactions. tance. The third common terminology used describes only the
The terms Rh-positive and Rh-negative are used to de- presence or absence of a given antigen. The fourth was estab-
scribe the presence or absence of the D antigen. Rh-positive lished by the International Society of Blood Transfusion (ISBT)
indicates that an individual’s red blood cells possess one par- Committee on Terminology for Red Cell Surface Antigens.
ticular Rh antigen, the D antigen. Rh-negative indicates that It is important for the student to distinguish between phe-
the red blood cells lack the D antigen. notype and genotype before exploring the Rh nomenclature.
The phenotype of a given RBC is defined by the serologic
History detection of antigens using specific antisera. Therefore an Rh
phenotype represents the results for serologic testing of RBC
In 1939 Levine and Stetson1 described a hemolytic transfusion for D, C, c, E, and e antigens. A genotype is an individual’s
reaction in an obstetrical patient. After delivering a stillborn actual genetic makeup. An RH genotype refers to the actual
infant, the woman required transfusions. Her husband, who RH genes inherited by the individual from his parents. Sero-
had the same ABO type, was selected as her donor. After trans- logic results may not exactly correspond with the genetic ex-
fusion, the recipient demonstrated classic symptoms of an pression. For a more detailed explanation of this concept,
acute hemolytic transfusion reaction (AHTR). Subsequently, refer to Chapter 2, “Basic Genetics.”
an antibody was isolated from the mother’s serum that reacted
at both 37oC and 20°C with the father’s RBCs. It was postu- Fisher-Race: DCE Terminology
lated that the fetus and the father possessed a common factor
that the mother lacked. While the mother carried the fetus, In the mid-1940s Fisher and Race defined the five common
she was exposed to this factor and subsequently produced an Rh antigens and postulated that the antigens of the system
antibody that showed positive reactivity when tested against were produced by three closely linked genes (D/d, C/c, and
the transfused RBCs from the father.1 The antibody specificity E/e)4 (Fig. 7–1). Each gene was responsible for producing
was not identified, but it was found to react with 80% of cells one antigen on the RBC surface. Each antigen and correspond-
tested and was suspected to be a maternal antibody to a factor ing gene were given the same letter designation (when refer-
on the fetal cells. A year later Landsteiner and Wiener2 de- ring to the gene, the letter is italicized). Fisher and Race
scribed an antibody made by guinea pigs and rabbits when named the antigens of the system D, d, C, c, E, and e. Now
they were transfused with rhesus macaque monkey RBCs. The it is known that there is no d antigen, but some references
6888_Ch07_149-172 31/10/18 9:46 AM Page 151

Chapter 7 The Rh Blood Group System 151

Antigens Agglutinogen Rh Factor


D Gene D
Close Production Factor Rh0 Rh0
Linkage C / c Gene C/c
Rh° gene Factor hr' hr'
Pathway
E / e Gene E/e Factor hr'' hr''

RBC Surface RBC Surface


Figure 7–1. Fisher-Race concept of Rh (simplified). Each gene produces one
Figure 7–2. Wiener’s agglutinogen theory. Antibody will recognize each factor
product.
within the agglutinogen.

continue to use the letter d with Fisher-Race terminology as of the haplotype. Each factor is an antigen recognized by an
a placeholder for D-negative individuals. antibody. The original Wiener nomenclature named the five
According to the Fisher-Race theory, an individual inherits common Rh antigens as Rho, rh⬘, rh⬙, hr⬘, and hr⬙, but these
a set of RH genes from each parent (i.e., one D or d, one C or terms are no longer used in favor of a modified form of
c, and one E or e). The combination of genes inherited from Wiener notation.
one parent is called a haplotype. For example, if one parent In the modified Wiener nomenclature, an agglutinogen is
has the genes D, C, and e, then the haplotype is written as described by a letter and symbol assigned based on the fac-
DCe. The pairing of maternal and paternal haplotypes deter- tors present. The uppercase R denotes the presence of the
mines the offspring’s genotype (the RH genes inherited from D antigen. The lowercase r indicates the absence of D antigen.
each parent). The genotype is written as two haplotypes sep- The presence of the C antigen is indicated by a 1 or a single
arated by a / (i.e., DCe/Dce). There are rare phenotypes that prime (⬘). The c antigen is implied when there is no 1 or ⬘
involve deletions of specific genes, and in those cases the indicated. The E antigen is indicated by a 2 or double prime
deletion is represented with a dash. For example an individ- (⬙) and the e antigen is implied when no 2 or ⬙ is indicated.
ual having only D and no C/c and E/e, the Fisher-Race haplo- That is, R1 is the same as DCe; r′ denotes Ce; and Ro is equiv-
type is written as D–. Placing parenthesis around (D), (C), alent to Dce. The presence of E is indicated by the Arabic
and (e) indicates weakened antigen expression. Table 7-1 number 2 or double prime (⬙). Lowercase e is implied when
compares serologic reactions to Fisher-Race nomenclature for there is no 2 or ⬙ indicated—that is, R2 is the same as DcE; r⬙
haplotypes. denotes cE, and r is equivalent to ce (again, it is assumed that
a c antigen is present). When both C and E are present, the
Wiener: Rh-Hr Terminology letter z or y is used. Rz denotes DCE, whereas ry represents
CE. Phenotypes of less common Rh types such as Rhnull
In his early work defining the Rh antigens, Wiener believed
and Rhmod are written as stated. The genotype for the Rhnull
there was one gene responsible for defining Rh that produced
that arises from an amorphic gene at both Rh loci is written
an agglutinogen containing three Rh factors5 (Fig. 7–2). The ⫽ and pronounced “little r double bar.” These will be
as rr
agglutinogen may be considered the phenotypic expression
discussed later in this chapter. See Table 7–2 for a sum-
mary of this shorthand nomenclature for the common
agglutinogens.
Table 7–1 Fisher-Race Haplotype Modified Wiener terminology allows one to convey Rh
Terminology antigens inherited on one chromosome or haplotype and
Designation makes it easier to discuss a genotype. Fisher-Race nomen-
clature may be converted to Wiener nomenclature and vice
Phenotype Reaction With Antisera versa. It is important to remember that an agglutinogen in
D C E c e Wiener nomenclature actually represents the presence of a
single haplotype expressing three different antigens. In the
+ + 0 0 + DCe
Wiener nomenclature, there is no designation for the ab-
0 + 0 0 + Ce sence of D antigen. By using these designations, the labora-
torian should be able to recognize immediately which
+ 0 + + 0 DcE
antigens are present on the RBCs.
0 0 + + 0 cE
Rosenfield and Coworkers: Alphanumeric
+ + + 0 0 DCE
Terminology
0 + + 0 0 CE
In the early 1960s, Rosenfield and associates proposed a sys-
+ 0 0 + + Dce tem that assigned a number to each antigen of the Rh system
0 0 0 + + ce in order of its discovery or recognized relationship to the Rh
system.6 This system has no genetic basis, nor was it proposed
6888_Ch07_149-172 31/10/18 9:46 AM Page 152

152 PART II Blood Groups and Serologic Testing

Table 7–2 Wiener Haplotype Terminology


Shorthand Symbol D C E c e Designation
R 1 + + 0 0 + R1

r ⬘ 0 + 0 0 + r⬘

R 2 + 0 + + 0 R2

r ⬙ 0 0 + + 0 r⬙

R Z + + + 0 0 Rz

r y 0 + + 0 0 ry

R 0 + 0 0 + + R0
r 0 0 0 + + r

based on a theory of Rh inheritance, but it simply demon- Table 7–3 Rh Genotypes by Three
strates the presence or absence of the antigen on the RBC.
Nomenclatures
Each antigen is assigned a number. A minus sign preceding a
number designates the absence of the antigen. If an antigen Wiener Fisher-Race Rosenfield
has not been phenotyped, its number will not appear in the R1r DCe/ce Rh:1, 2, –3, 4, 5
sequence. An advantage of this nomenclature is that the RBC
phenotype is thus succinctly described. R1R1 DCe/DCe Rh:1, 2, –3, –4, 5
For the five major antigens, D is assigned Rh1, C is Rh2, R1R2 DCe/DcE Rh:1, 2, 3, 4, 5
E is Rh3, c is Rh4, and e is Rh5. For RBCs that type D-
positive, C-positive, E-positive, c-negative, and e-negative, R2r DcE/ce Rh:1, –2, 3, 4, 5
the Rosenfield designation is Rh: 1, 2, 3, –4, –5. If the sample R2R2 DcE/DcE Rh:1, –2, 3, 4, –5
was not tested for e, the designation would be Rh: 1, 2, 3, –4.
The numeric system is well suited to electronic data process- Ror Dce/ce Rh: 1,-2, -3, 4, 5
ing. Its use expedites data entry and retrieval. Its primary RoRo Dce/Dce Rh: 1,-2, -3, 4, 5
limiting factor is that there is a similar nomenclature for nu-
merous other blood groups, such as Kell, Duffy, Kidd, and Rzr DCE/ce Rh:1, 2, 3, 4, 5
more. Therefore, when using the Rosenfield nomenclature rr ce/ce Rh: –1, –2, –3, 4, 5
on the computer, one must use both the alpha (Rh:) and the
numeric (1, 2, –3, etc.) to denote a phenotype. Table 7-3 lists r⬘r Ce/ce Rh: –1, 2, –3, 4, 5
common Rh genotypes comparing the nomenclatures of r⬘r⬘ Ce/Ce Rh: –1, 2, –3, –4, 5
Wiener, Fisher-Race, and Rosenfield.
r⬙r cE/ce Rh: –1, –2, 3, 4, 5
International Society of Blood Transfusion r⬙r⬙ cE/cE Rh: –1, –2, 3, 4, –5
Committee: Updated Numeric Terminology
r⬘r⬙ Ce/cE Rh: –1, 2, 3, 4, 5
As the world of blood transfusion began to cooperate and
share data, it became apparent there was a need for a univer- (ryr) CE/ce Rh: -1, 2, 3, 4, 5
sal language. The International Society of Blood Transfusion
(ISBT) formed the Committee on Terminology for Red Cell
Surface Antigens. Its mandate was to establish a uniform unchanged but were converted to all uppercase letters (e.g.,
nomenclature that is both eye- and machine-readable and is Rh, Kell became RH, KELL). Therefore, D is RH1, C is RH2,
in keeping with the genetic basis of blood groups.7 The ISBT and so forth. (Note: There is no space between the RH and
adopted a six-digit number for each authenticated antigen the assigned number.) The phenotype designation includes
belonging to a blood group system. The first three numbers the alphabetical symbol that denotes the blood group,
represent the system, and the remaining three the antigenic followed by a colon and then the specificity numbers of the
specificity. Number 004 was assigned to the Rh blood group antigens defined. A minus sign preceding the number in-
system, and then each antigen assigned to the Rh system was dicates that the antigen was tested for but was not present.
given a unique number to complete the six-digit computer The phenotype D + C – E + c + e + or DcE/ce or R2r would
number. Table 7–4 provides a listing of these numbers. be written RH:1, –2, 3, 4, 5.
When referring to individual antigens, an alphanumeric When referring to a gene, an allele, or a haplotype, the
designation similar to the Rosenfield nomenclature may symbols are italicized. A haplotype is followed by a space or
be used. The alphabetic names formerly used were left an asterisk, and the numbers of the specificities are separated
6888_Ch07_149-172 31/10/18 9:46 AM Page 153

Chapter 7 The Rh Blood Group System 153

Table 7–4 Antigens of the Rh Blood Group System in Four Nomenclatures


Rosenfield Fisher-Race Wiener ISBT Number Other Names or Comment
Rh1 D Rh0 004001

Rh2 C rh⬘ 004002

Rh3 E rh⬙ 004003

Rh4 c hr⬘ 004004

Rh5 e hr⬙ 004005

Rh6 ce hr 004006 f

Rh7 Ce rhi 004007

Rh8 Cw rhw1 004008

Rh9 Cx rhx 004009

Rh10 V hrv 004010 ceS

Rh11 Ew rhw2 004011

Rh12 G rhG 004012


Rh17† Hr0 004017

Rh18 Hr 004018 HrS (high prevalence)

Rh19 hrs 004019

Rh20 VS es 004020

Rh21 CG 004021

Rh22 CE Rh 004022 Jarvis

Rh23 Dw 004023 Wiel

Rh24† ET 004024

Rh26 c-like 004026 Deal

Rh27 cE rhii 004027

Rh28 hrH 004028 Hernandez

Rh29 004029 Total Rh

Rh30 Dcor 004030 Goa (low prevalence) DIVa

Rh31 hrB 004031

Rh32 RN 004032 Troll (low prevalence)

Rh33 R0Har 004033 DHAR (low prevalence)

Rh34 HrB 004034 Bastiaan

Rh35 004035 (low prevalence)

Rh36 004036 Bea (Berrens; low prevalence)

Rh37 004037 Evans (low prevalence)

Rh38† Formerly Duclos

Rh39 C-like 004039

Rh40 Tar 004040 Targett (low prevalence)


Continued
6888_Ch07_149-172 31/10/18 9:46 AM Page 154

154 PART II Blood Groups and Serologic Testing

Table 7–4 Antigens of the Rh Blood Group System in Four Nomenclatures—cont’d


Rosenfield Fisher-Race Wiener ISBT Number Other Names or Comment
Rh41 Ce-like 004041

Rh42 CeS , CceS rhis 004042 Thornton

Rh43 004043 Crawford (low prevalence)

Rh44 004044 Nou (high prevalence)

Rh45 004045 Riv

Rh46 004046 Sec (high prevalence)

Rh47 “Allelic” to RN 004047 Dav (high prevalence)

Rh48 004048 JAL (low prevalence)

Rh49 004049 Stem

Rh50 004050 FPTT (low prevalence)

Rh51 004051 MAR (high prevalence)

Rh52 004052 BARC (low prevalence)

Rh53 004053 JAHK (low prevalence)

Rh54 004054 DAK (low prevalence)

Rh55 004055 LOCR (low prevalence)

Rh56 004056 CENR (low prevalence)

Rh57 004057 CEST (high prevalence)

RH58 004058 CELO (high prevalence)

RH59 004059 CEAG (high prevalence)

RH60 004060 PARG (low prevalence)

RH61 004061 CEVf

by commas. The R1 haplotype or DCe would be RH 1,2,5 or Rhce, or RhCE proteins (Fig. 7–3). RHD and RHCE are
RH*1,2,5. codominant, which means that all products inherited typi-
cally produce antigens detectable on RBCs. RHD and RHCE
Rh Nomenclatures Overview genes each have 10 exons and are 97% identical. Each gene
has a number of alleles, most of which have been identified
Tables 7–3, 7–4, and 7–5 summarize the data presented in through molecular testing techniques.
this section. As the genetics and biochemistry of the Rh Numerous mutations have been described in the RH
blood group system continue to be investigated, so too does genes. Greater than 250 alleles have been determined in the
the terminology continue to change. For consistency of use, RHD gene, and 50 alleles have been found in the RHCE allele
RHD and RHCE, all uppercase and in italics, will be used and the number continues to grow. A complete list of the
from this point forward in the text to indicate genes. RhD, alleles found on the RHD and RHCE genes can be found on
RhCe, RhcE, Rhce, and RhCE will be used to designate pro- the Rhesus-data base and the website for the National Center
teins on which the Rh antigens reside. for Biotechnology Information (NCBI), which maintains a
database for human blood group mutation.10,11 Fortunately
Genetics for the medical laboratory scientist, many of these mutations
are rare and do not change the serology observed in day-to-
Rh Genes day testing.
RHD and RHCE are two closely linked genes located on chro- Rh-Associated Glycoprotein (RHAG)
mosome 1 that control expression of Rh proteins.8,9 The gene
RHD codes for the presence or absence of the RhD protein, Another gene important to Rh antigen expression is RHAG,
and the second gene RHCE codes for either RhCe, RhcE, and it resides on chromosome 6.12 The product of this gene
6888_Ch07_149-172 31/10/18 9:46 AM Page 155

Chapter 7 The Rh Blood Group System 155

Table 7–5 Prevalence of the Principal Rh Haplotypes


Fisher-Race Nomenclature Modified Wiener Nomenclature Prevalence (%)
White African American Asian

DCe R1 42 17 70

DcE R2 14 11 21

Dce Ro 4 44 3

DCE Rz <0.01 <0.01 1

ce r 37 26 3

Ce r⬘ 2 2 2

cE r⬙ 1 <0.01 <0.01

CE ry <0.01 <0.01 <0.01

(Modified From: AABB Technical Manual 18th ed. Philadelphia: American Association of Blood Banks, Bethesda, MD, 2016, p. 320, with permission.)

RH Genes — Rh Positive RH Genes in Rh-Negative Caucasians

Locus 1 Locus 2 Locus 1 Locus 2


RHD RHCE RHCE

1 2 3 4 5 6 7 8 9 10 1 2 3 4 5 6 7 8 9 10 1 2 3 4 5 6 7 8 9 10

Exons Exons Exons


A D antigen C/c and E/e antigens B No D antigens ce antigens
Figure 7–3. Locus 1 presence of RHD codes for the presence of D (A) or no D (B). Locus 2 presence of RHCE codes for Ce, CE, cE, ce. Each gene consists of 10 coding
exons.

is Rh-associated glycoprotein (RHAG). This polypeptide is


very similar in structure to the Rh proteins, with the differ-
ence being that it is glycosylated (carbohydrates attached).
Within the RBC membrane, it forms complexes with the Rh DCe/dce DcE/dce
proteins. RhAG is termed a coexpressor and must be present
for successful expression of the Rh antigens. However, by it-
self, this glycoprotein does not express any Rh antigens.
When mutations in the RHAG gene occur, it can result in
missing or significantly altered RhD and RhCE proteins,
DCe/DcE DCe/dce dce/DcE dce/dce
affecting antigen expression. In rare instances, individuals
express no Rh antigens on their RBCs. These individuals are Figure 7–4. Example of a normal pattern of Rh inheritance.
said to have the Rhnull phenotype and are discussed in more
detail later in this chapter. RHAG has been assigned as a sep-
arate blood group system and is discussed in further detail Numerous mutations in the RHD gene have been discov-
in Chapter 8, “Blood Group Terminology and the Other ered that cause weakened expression of the RhD antigen
Blood Groups.” detected in routine testing. This is generally termed weak D
or weak expression of D and will be discussed in more detail
Rh-Positive Phenotypes later in the chapter.

RH genes are inherited as codominant alleles. Rh-positive Rh-Negative Phenotypes


individuals inherit one or two RHD genes, which result in
expression of RhD antigen and are typed Rh-positive. In Rh-negative phenotypes are so called because the RBCs lack
addition to the RHD gene(s), two RHCE genes are inherited, detectable D antigen. Rh-negative individuals can arise from
one from each parent. Figure 7–4 is an example of a normal several pathways. The most common Rh-negative phenotype
Rh inheritance pattern. results from the complete deletion of the RHD gene—that is,
6888_Ch07_149-172 31/10/18 9:46 AM Page 156

156 PART II Blood Groups and Serologic Testing

the individuals possess no RHD gene but have inherited two Most Probable Genotypes
RHCE genes.13 Refer back to Figure 7–3 for a depiction of
Rh genes. Serologic testing for Rh antigens only defines phenotype. One
may predict the genotype of the individual based on the
prevalence of RH genes. Determining most probable geno-
Advanced Concepts types was used in the past for parentage studies, also known
African Ethnicity as relationship testing, and for population studies. Other mo-
lecular methods are proving more powerful today but it re-
An unusual form of the RHD gene, termed RHD pseudo-
mains important for the student to be able to predict the most
gene or RHDψ, occurs in 66% of individuals of African eth-
probable genotype based on gene frequency and ethnicity.
nicity. This gene is an RHD-negative allele whose sequence
There are substantial differences in the frequency of hap-
is identical to the RHD gene except for missense mutations
lotypes in the white, black, and Asian populations. Refer back
in exon 5 and exon 6, a nonsense mutation in exon 6, and
to Table 7–5 to recall the prevalence of RH haplotypes in both
a 37-bp insertion at the intron 3/exon 4 boundary.14 Indi-
Fisher-Race and Wiener nomenclatures. Table 7-6 shows the
viduals with this gene do not produce RhD protein.
serologic reactions with the most probable genotypes for
Asian Ethnicity Caucasian and African American populations. Knowing the
prevalence of RH haplotypes is important to predict the cor-
Another mutation has been described in Asians that
rect most probable genotype. Consider, for example, an Rh
alters the RHD gene, causing an individual to type as
phenotype of D+C-E-c+e+. In the Caucasian population, the
D-negative.15 This is termed Del and will be discussed in
genotype Ror would be the most likely because the Ro haplo-
more detail later in this chapter in the section on weak
type has a prevalence of only 4% of those individuals while
D phenotypes.
the r haplotype has a prevalence of 37%. If the individual

Table 7–6 Reaction Patterns with Five Antisera and Most Probable Genotypes (MPG)*
African Other
Caucasian MPG American Possibilities
D C E c e and % MPG and % (Both Groups)
+ + 0 + + R1r R1Ro Ror⬘
34.9% 15%

+ + 0 0 + R1R1 R1R1 R1r⬘


18.5% 3%

+ + + + + R1R2 R1R2 R1r⬙, R2r⬘, Rzr, RzRo, Rory


13.3% 4%

+ 0 + + + R2r R2Ro r⬙
11.8% 10%

+ 0 + + 0 R2R2 R2R2 R2r⬙


2.3 % 1%

+ 0 0 + + Ror Ror RoRo


2.1% 23% 19%

0 0 0 + + rr rr -
15.1% 7%

0 + 0 + + r⬘r r⬘r -
0.8% 1%

0 + + + + r⬘r⬙ r⬘r⬙ ryr


0.05%

0 + 0 0 + r⬘r⬘ r⬘r⬘ -
rare rare

0 0 + + 0 r⬙r⬙ -
rare rare

+ + + 0 + RzR1 RzR1 Rzr⬘, R1ry


0.2% rare

+ + + + 0 RzR2 RzR2 Rzr⬙, R2ry


0.1% rare
6888_Ch07_149-172 31/10/18 9:46 AM Page 157

Chapter 7 The Rh Blood Group System 157

Table 7–6 Reaction Patterns with Five Antisera and Most Probable Genotypes (MPG)*—cont’d
African Other
Caucasian MPG American Possibilities
D C E c e and % MPG and % (Both Groups)
+ + + 0 0 RzRz RzRz Rzry
0.1% rare

0 + + 0 + ryr⬘ ryr⬘
rare rare

0 + + + 0 ryr⬙ ryr⬙
rare rare

0 + + 0 0 ryry ryry
rare rare

* Percentages are rounded off.

were African American, the most likely genotype would be number of D antigen sites on a variety of Rh phenotypes.18
RoRo as the Ro haplotype is far more prevalent at 44%. This The results are summarized in Table 7-7. The greatest num-
explains why the phenotype D+C-E-c+e+ is most commonly ber of D antigen sites are on cells of the rare Rh phenotype
seen in the African American population but is considered D– (refer to the “Deletions” section). However, of the com-
relatively rare in Caucasians. These differences must be re- monly encountered Rh genotypes, R2R2 cells possess the
membered when trying to locate compatible blood for recip- largest number of D antigen sites.
ients with unusual or multiple Rh antibodies.

RH Antigens
C/c (Ser/Pro) E/e (Pro/Ala)
Biochemistry 103 226
Cx
Cw
The product of RH genes are nonglycosylated proteins. This
means no carbohydrates are attached to the protein. Rh anti- RhCE
gens reside on transmembrane proteins and are an integral
part of the RBC membrane.16 The gene products of RHD and
RHCE are remarkably similar in that both encode for proteins NH2 Leu245Val 417
composed of 416 amino acids that traverse the cell membrane V+VS+
12 times. Their proteins differ by only 32 to 35 amino acids.
Amino acid position 103 is important in determining C or c
expression, and position 226 differentiates E from e (Fig. 7–5).
RhD
Only small loops of Rh proteins are exposed on the surface of
the RBC and provide the conformational requirements for
many serologic differences between the Rh blood types.
RhD and RhCE proteins and RhAG are exclusively on red NH2 417
blood cells. As they are transmembrane, it is not surprising
they play a role in maintaining the structural integrity of red
blood cells. Based on their structure, it appears they may also
be transporters. Westhoff and colleagues showed they may RhAG
have a role in transporting ammonia.17 An alternative hy- Rh-associated
pothesis is that they may be CO2 transporters. glycoprotein

NH2 409
Advanced Concepts
In researching the biochemistry of Rh antigens, investiga- Figure 7–5. Predicted model of the RhCE, RhD, and RhAG proteins in the RBC
membrane. The amino acid differences are shown as circles, and those that are
tions have been performed to determine the quantity of
extracellular on RhD are shown as black dots. C/c differ by several amino acids,
antigen sites on RBCs of various Rh phenotypes. In com- but only Ser103Pro on loop 2 is predicted to be extracellular. E/e differs by
parison with ABO and Kell (K) blood groups, A1 cells pos- Pro226Ala on loop 4, and CW and CX antigens are located on the first extracellular
sess approximately 1 × 106 A antigens, whereas RBCs loop of RhCE. The antigens V and VS result from a Leu245Val change located in
possessing a double-dose expression of the K antigen have the predicted eighth transmembrane region of RhCE. RhAG does not carry Rh anti-
gens. (From: Westhoff, CM: The Rh blood group system in review: A new face for
6,000 K sites. Hughes-Jones and coworkers measured the
the next decade. Transfusion 44 (11): 1663-73, 2004, with permission.)
6888_Ch07_149-172 31/10/18 9:46 AM Page 158

158 PART II Blood Groups and Serologic Testing

Table 7–7 Number of D Antigen Sites of D Antigen


Cells With Various Phenotypes Rh antigens are highly immunogenic; the D antigen is the
Rh Genotype Number of D Antigen Sites most potent.19 This is not surprising given that most Rh-
negative individuals lack the entire RhD protein. Exposure
R1r 9,900–14,600
to less than 0.1 mL of Rh-positive RBCs can stimulate
R0r 12,000–20,000 antibody production in an Rh-negative person. Eighty-five
percent of the general population is Rh-positive, with 15%
R2r 14,000–16,600
typing as Rh-negative; however, the percentages vary
R1R1 14,500–19,300 among ethnicities. The Hispanic and African American
populations in the United States have significantly higher
R1R2 23,000–31,000
percentages of Rh-positive individuals (92.7% and 92.9%,
R2R2 15,800–33,300 respectively).20 Anti-D was the most common cause of
HDFN until Rh-immune globulin was introduced. (See
D— 110,000–202,000
Chapter 20, “Hemolytic Disease of the Newborn and
Fetus.”) There are rare variations of the D antigen, which
will be discussed later in this chapter.
Antigen Characteristics

As mentioned above, Rh antigens are proteins integral to Other Common Rh Antigens (C, E, c, e)
the RBC membrane, passing through the RBC wall 12 times
(Fig. 7–5).The antigens are found only on RBCs, and are not While the D antigen is most immunogenic, c antigen is
soluble or expressed on other cells. The Rh antigens are well the next most likely Rh antigen to elicit an immune re-
developed at birth, and as such can cause hemolytic disease sponse, followed by E, C, and e. Figure 7–6 depicts the
of the fetus and newborn (HDFN). Rh antigens are also im- relative immunogenicity of the common Rh antigens. It is
munogenic, and exposure to foreign antigens through trans- not uncommon to see several Rh antibodies in a patient;
fusion or pregnancy can cause an immune response with for example, anti-D and anti-C or anti-c and anti-E (see
the production of corresponding antibodies. Case Study 7-2). C, c, E, and e antigens are inherited
The five common Rh antigens are D, C, E, c, and e, and through the RHCE gene. The alleles are codominant,
their specific corresponding antibodies account for the major- therefore specificities from both haplotypes are expressed
ity of problems due to Rh antibodies. The D antigen does not as RBC antigens. The antigen frequencies vary in the Cau-
have an allele, but C and c are alleles and E is allelic to e. The casian and African American populations. (Refer back to
frequency of some of the Rh antigens is listed in Table 7–8. Table 7–8.) There are unusual phenotypes and rare anti-
(Antigens other than the common five are discussed later in gens that are variations of these antigens, which will be
this chapter). discussed later in this chapter.

Table 7–8 Frequency of Rh Antigens


Antigen Name Frequency (%) Caucasian Frequency (%) African American
D 85 92

C 70 34
E 30 21

c 80 97

e 98 99

ce/f 64 75

Ce 70 52

Cw 1 rare

G 86 86

V 1 30

VS 1 32
6888_Ch07_149-172 31/10/18 9:46 AM Page 159

Chapter 7 The Rh Blood Group System 159

D > c > E > C > e The D antigens expressed appear to be complete but fewer
Figure 7–6. Immunogenicity of common Rh antigens. in number. On a molecular level, mutations in the RHD gene
occur, causing changes or deletions in amino acids present
in the transmembrane or intracellular region of the RhD
Weak D: Variations of D Antigen protein, thus causing conformational changes in the protein
Expression (Fig. 7–7). Wagner, Gassner, and others described the
classification of weak D RBCS based on single nucleotide
When Rh-positive RBC samples are typed for the D antigen, polymorphisms (SNP).23 Mutations for these weak D types
they are expected to show strong positive reactivity with anti- occur because SNP affects amino acid insertion of the protein
D reagents. However, some individuals have RBCs that possess on the RBC membrane. When these changes occur, normal
variations in the quantity of D antigen or the specificity RhD antigen expression is altered because there are fewer
of D antigen epitopes, resulting in weakened expression of the antigen sites overall.
D antigen when tested with serologic methods. Serologic weak Individuals with weak D phenotype rarely make anti-D,
D is noted when initial anti-D testing is negative, or less than since the RhD antigens are present as expected, but protein
or equal to 2+ strong, but detectable at the indirect antiglobulin changes occur “inside” the red blood cell. On a molecular
testing (IAT) phase. Advances in Rh testing methodology and level changes in the RHD gene occur, causing changes in
reagents have resulted in detecting more weak D types without amino acids present in the transmembrane region of the RhD
the need for IAT, but there are still circumstances in which protein. Mutations in the RHD gene causing this altered ex-
weak D testing is necessary. Individuals with RBCs carrying pression have been categorized with types 1 and 3 being the
weaker D antigen (historically called Du type) can produce most common.
anti-D if they are missing epitopes of the D antigen.
For many years, the generic term weak D was used to iden- Del
tify individuals whose D was not detectable at immediate spin,
without defining the nature of the weakened expression. Now Del is a phenotype occurring in individuals whose red blood
it is known that weak D can arise from several mechanisms. cells possess an extremely low number of D antigen sites that
Weak D types can be separated into three categories: position most reagent anti-D are unable to detect. Adsorbing and elut-
effect, quantitative, and partial-D antigen (missing one or ing anti-D from the individual’s red blood cells is often the
more alleles). It has been shown that up to 2% of individuals only way to detect the D antigen. This procedure requires
with European ancestry possess some altered form of D anti- incubating anti-D with the RBCs in question at 37°C fol-
gen.21 Altered D antigen occurs more often in individuals of lowed by eluting the anti-D off the adsorbed red blood cells.
African descent, but the exact prevalence is not known. Fi- The 37°C incubation provides time for the anti-D to bind to
nally, to further complicate matters, there are rare individuals the few D antigen sites present on these red blood cells. Mo-
who possess D epitopes on their RhCE protein. lecular studies can detect a mutant RHD gene that alters ex-
pression of the RhD protein. This phenotype occurs most
Position Effect: C in Trans to D often in individuals of Southeast Asian descent, occurring in
up to 30% of that population.15 It is rare in Caucasians.
The first mechanism that may result in weakened expression Del individuals of Southeast Asian descent have not been
of D antigen was originally described as a position effect or reported to make anti-D in response to transfusion or preg-
gene interaction effect.22 The allele carrying RHD is trans nancy, but there have been a few instances of anti-D produc-
(or in the opposite haplotype) to the allele carrying C; for tion in Caucasian individuals.24
example, Dce/dCe. The Rh antigen on the RBC is normal, but
the steric arrangement of the C antigen in relationship to the
D antigen appears to interfere with the expression of D anti-
Weak D Partial D
gen. This interference with D expression does not occur
when the C gene is inherited in the cis position to RHD, such
as DCe/dce. It is not possible to serologically distinguish ge- Exterior
netic weak D from the position effect weak D. Molecular
studies would differentiate the two types. Practically speak-
ing, this is unnecessary because the D antigen is structurally Plasma
complete. These individuals can receive D-positive RBCs membrane
with no adverse effects.
Interior
Weak D: Quantitative Changes Due to Fewer D
Antigen Sites Type 1 Type 2
Figure 7–7. Three-dimensional illustrations of weak and partial D. (From: Chou
The second mechanism results from inheritance of RHD ST, Westhoff CM. The Rh System. In: Roback JD, Grossman BJ, Harris T, Hillyer CD,
genes that code for a weakened expression of the D antigen. eds. Technical Manual. 17th edition. AABB, Bethesda, MD, pp. 389-410, 2011.)
6888_Ch07_149-172 31/10/18 9:46 AM Page 160

160 PART II Blood Groups and Serologic Testing

Normal RHD Normal RHCE


Advanced Concepts
1 2 3 4 5 6 7 8 9 10 1 2 3 4 5 6 7 8 9 10
There is currently no national consensus on the handling
and testing of weak D phenotypes. In the transfusion set-
ting, patients are generally typed for D with immediate Partial DVI
spin or other routine methods, and a negative result is
interpreted as Rh negative without any further testing. 1 2 3 4 5 6 7 8 9 10 1 2 3 4 5 6 7 8 9 10
Donors who initially type as D-negative must be tested
further to determine if they are really weak D. Sandler and Figure 7–8. One type of partial-DVI gene where three exons of RHCE gene are
others have suggested that patients who initially type as inserted into RHD gene.
D-negative should have RHD genotyping performed to as-
sess their true D status.25 The goal of this further testing
or transfusion reactions, or both. Once anti-D is identified,
would be to improve accuracy in Rh typing, conserve
Rh-negative blood should be used for transfusion. Note that
Rh-negative units for those who truly need them, and pre-
the anti-D produced is an alloantibody because it is directed
vent unnecessary administration of Rh immune globulin
against an antigenic epitope that the patient lacks. So while
in women with weak D serotypes.
the patient may type as D-positive, the serum antibody will
not react with the patient’s own cells.
Partial D or D Mosaic The identification of a person with a partial D routinely
occurs after the person begins producing anti-D unless
The third mechanism in which D antigen expression can be there are discrepant Rh (D) typings. This discovery should
weakened is when one or more D epitopes within the entire prompt collection of additional samples to be sent to an
D protein is either missing or altered, termed partial D immunohematology reference laboratory for further RhD
(sometimes called “D Mosaic”). The D antigen is not com- classification.
plete because one or more epitopes are missing. The sero-
logic detection of the D antigen varies greatly for partial-D
individuals. Some individuals have red blood cells with Advanced Concepts
partial-D antigens that may type weaker than expected or
With the advent of monoclonal antibodies and the deple-
that may not react at all when routine procedures are used
tion and deterioration of the available anti-D made by per-
with most commercial anti-D reagents. Others have partial-
sons with partial-D phenotypes, Tippett and coworkers
D types that may show normal typing with reagent anti-D.
pursued the classification of partial-D antigens using mon-
Wiener and Unger postulated that the D antigen is made
oclonal anti-D (MAb-D).29 While partial D phenotypes are
of antigenic subparts, genetically determined, that could be
not common, category VI is the most often encountered
absent in rare instances.26 If an individual lacks one (or
partial D phenotype. Today, a commercial monoclonal anti-D
more) pieces, or epitopes, of the total D antigen, alloantibody
panel is available. Although helpful, it does not define all
can be made to the missing epitope(s) if exposed to RBCs
partial D and weak D types clearly. A combination of sero-
that possess the complete D antigen. This theory has become
logic typing and molecular analysis is often required to
well accepted.
accurately categorize partial-D types.
Tippett and Sanger worked with RBCs and sera of partial-D
individuals to classify these antigens.27 Seven categories were
recognized, designated by Roman numerals I through VII. D Epitopes on RhCE Protein
Category I is now obsolete, and a few of the categories have
been further subdivided. In some cases, the loss of D epi- Like RhD proteins that can express some RhCE protein, re-
topes can result in a new antigen forming. For example, in- sulting in the partial-D phenotypes, RhCE protein can ex-
dividuals in category VI have RBCs that carry the BARC press RhD epitopes detected by some monoclonal anti-D.
antigen. This can cause discrepant RhD type results with historical
Partial-D antigens can be classified on a molecular level information on a patient or donor. There are rare individuals
and are attributed to hybrid genes resulting from portions who possess these unusual proteins and when typed with
of the RHD gene being replaced by portions of the RHCE anti-D will show positive reactivity even though the D epi-
gene, as shown in Figure 7–8. The resulting protein con- tope is on the RhCE protein. Examples of these unusual
tains a portion of RHD and RHCE in various combinations, phenotypes include DHAR and ceCF (Crawford).
depending on the hybrid gene’s makeup. These protein R0Har (RH33) results from a hybrid gene RHCE-RHD-
changes occur external to the red blood cell membrane. If RHCE, in which only a small portion of RHD is inserted into
an individual with a hybrid RhD-RhCe-RhD protein is ex- the RHCE gene (Fig. 7–9).30 If this hybrid gene is paired with
posed to red blood cells possessing normal RhD protein, a normal RHD gene, the individual will type Rh-positive.
they will make antibody to the portion of the RhD protein However, if paired with a D-deletion, the individual may type
they are missing.28 D-positive, depending on the anti-D reagent being used.
Anti-D made by individuals expressing partial D can These individuals should be classified as RhD-negative since
cause hemolytic disease of the fetus and newborn (HDFN) they essentially lack the RhD protein.
6888_Ch07_149-172 31/10/18 9:46 AM Page 161

Chapter 7 The Rh Blood Group System 161

D Epitope on RHCE Gene — D HAR Rh antibodies do not bind complement. For complement
to be fixed (or the complement cascade activated), two IgG
Locus 1 Locus 2
immunoglobulins must attach to an RBC antigen in close
RHCE proximity to each other. Rh antigens (to which the antibody
would attach) are not situated on the RBC surface this
closely. Therefore, when an Rh antibody coats the RBCs, in-
1 2 3 4 5 6 7 8 9 10 travascular, complement-mediated hemolysis does not occur.
RBC destruction resulting from Rh antibodies is primarily
Exons extravascular. However, a few rare examples of complement
No D antigens ce antigens binding Rh antibodies have been reported.
Figure 7–9. DHAR results from one RHD exon inserted into the RHCE gene.
Rh Antibodies in Pregnancy

The Crawford (ceCF) phenotype, RH34, is found in in- Because Rh antibodies are primarily IgG and can traverse the
dividuals of African descent. It results from a specific amino placenta and because Rh antigens are well developed early
acid change in the RHce gene, resulting in an RhD epitope in fetal life, Rh antibodies formed by pregnant women cross
on the Rhce protein.31 the placenta and may coat fetal RBCs that carry the corre-
sponding antigen. This results in the fetal cells having a pos-
RH Antibodies itive direct antiglobulin test; in HDFN, the coated fetal cells
are removed prematurely from the fetal circulation which
Rh antibodies are fairly straightforward to detect and identify can result in anemia. HDFN due to anti-D can be prevented
as compared to understanding the molecular genetics and with the use of Rh-Immune Globulin, but complications due
biochemistry of the Rh blood group system. Further discus- to other Rh antibodies cannot be prevented. Until the dis-
sion on detecting antibodies made by individuals to Rh anti- covery of Rh-immune globulin, anti-D was the most frequent
gens follows. cause of HDFN. (See Chapter 20, “Hemolytic Disease of the
Newborn and Fetus.”)
Characteristics of Rh Antibodies
Testing for RH Antigens
Although the Rh system was first recognized by saline tests and Antibodies
used to detect IgM antibodies, most Rh antibodies are IgG im-
munoglobulins and react optimally at 37°C or after antiglob- Rh Typing Reagents
ulin testing in any method used for antibody detection. Rh
antibodies are usually produced following exposure of the Reagents used to type for D and for the other Rh antigens
individual’s immune system to foreign RBCs, through either may be derived from a variety of sources. The reagents may
transfusion or pregnancy. Rh antibodies may show dosage, be high-protein based or low-protein based, saline based,
reacting preferentially with RBCs possessing double-dose Rh chemically modified, monoclonal, or blends of monoclon-
antigen. For example, anti-E may show 3+ positive reactivity als. The goal is to use a reagent anti-D that will allow for
with E+e– RBCs versus 2+ positive reactivity with E+e+ RBCs. typing individuals’ RBCs as quickly and accurately as typing
In addition, Rh antibodies are enhanced when testing with for ABO.
enzyme-treated RBCs. Saline reactive reagents, which contain IgM immunoglob-
IgG1, IgG2, IgG3, and IgG4 subclasses of Rh antibodies ulin, were the first typing reagents available to test for the
have been reported. IgG1 and IgG3 are of the greatest clinical D antigen. Saline anti-D has the advantage of being low-protein
significance because the reticuloendothelial system rapidly based and can be used to test cells that are already coated
clears RBCs coated with IgG1 and IgG3 from the circulation. with IgG antibody, as in patients who have autoantibodies
IgA Rh antibodies have also been reported but are not rou- binding to their RBCs. If a high-protein–based reagent was
tinely tested for in the blood bank. used when typing these antibody-coated RBCs, a false-
As with most blood group antibodies, IgM Rh antibodies positive reaction would be obtained because the RBCs would
are formed initially, followed by a transition to IgG. Rh anti- agglutinate on their own without the addition of anti-D. The
bodies often persist in the circulation for years. An individual primary disadvantages of saline typing reagents are their lim-
with low-titer Rh antibody may experience an anamnestic ited availability, cost of production, and lengthy incubation
(secondary) antibody response if exposed to the same sensi- time. Because saline anti-D is an IgM immunoglobulin, it
tizing antigen. Therefore, in the clinical setting, accuracy of cannot be used for weak D typing.
D typing is essential, as is the careful checking of patient his- In the 1940s, high-protein anti-D reagents were developed
tory to determine whether an Rh antibody has been identi- that consisted primarily of IgG anti-D. Human plasma con-
fied previously. Most commonly found Rh antibodies are taining high-titer D-specific antibody was used as the raw
considered clinically significant. Therefore, antigen-negative material. Potentiators of bovine albumin and macromolecu-
blood must be provided to any patient with a history of Rh- lar additives, such as dextran, were added to the source ma-
antibody sensitization, whether the antibody is currently terial to optimize reactivity in the standard slide and rapid
demonstrable or not. tube tests to allow for direct agglutination of red blood cells
6888_Ch07_149-172 31/10/18 9:46 AM Page 162

162 PART II Blood Groups and Serologic Testing

using an IgG anti-D. In essence, this media causes RBCs to controls, and accurate interpretation of test and control re-
be in closer proximity to each other and allows IgG anti-D sults. Table 7–9 summarizes several common causes of false
to crosslink and cause direct agglutination. These reagents Rh typing results and suggests corrective actions that may
are commonly referred to as high-protein reagents. be taken to obtain an accurate Rh type.
However, the presence of potentiators and the higher pro-
tein concentration increase the likelihood of false-positive Rh Testing Requirements for Pretransfusion,
reactions. To assess the validity of the high-protein Rh typing Obstetric Patients, and Blood Donors
results, a control reagent was manufactured and had to be
Rh typing requirements vary depending on the reason for
tested in parallel with each Rh test. If the control reacted,
performing the testing. For blood donors, it is critical to
the test result was invalid and had to be repeated using a dif-
make every effort to identify the presence of the D antigen,
ferent technique or reagent anti-D. The major advantages of
because it is so highly immunogenic. AABB Standards
high-protein anti-D reagents are reduced incubation time
require that patients must be tested for the D antigen, but
and the ability to perform weak D testing and slide typing
additional testing for weak D is optional.32
with the same reagent. This type of anti-D reagent is also
Blood centers perform Rh typing on all donors each time
polyspecific. More than one clone of anti-D is produced by
a unit of blood is collected. Current Rh type is compared to
immunized human donors and is therefore able to recognize
previous results as a method to check for sampling or testing
multiple epitopes on the RhD protein.
error. All discrepancies must be fully resolved before the unit
Chemically modified Rh typing reagents alter the IgG
can be labeled and released for transfusion. Rh typing is typ-
anti-D molecule by breaking the disulfide bonds that main-
ically performed with a method designed to screen for the
tain the antibody’s rigid shape. This allows the antibody to
D antigen at the immediate spin phase (if tube testing) or
relax and to span the distance between RBCs in a low-protein
other screening method. Donors who test D-negative on the
medium. The chemically modified reagents can be used for
initial test must have additional testing performed to explore
both slide and tube testing and do not require a separate,
the possibility of a weak D phenotype.32 Even the smallest
manufactured Rh control as long as the samples type as A,
amount of D antigen may trigger an immune response in a
B, or O. When samples test AB Rh-positive or when the Rh
patient exposed to the donated RBCs. The unit of blood is
test is performed by itself, a separate saline control or 6% to
labeled as Rh-positive or Rh-negative. It is not necessary to
8% albumin control must be used to ensure the observed re-
note any weak D phenotypes on the final labeling.
actions are true agglutination and not a result of spontaneous
In the transfusion service, patients are tested for the
agglutination. Fewer false-positive test reactions are obtained
D antigen as part of routine testing, usually in combination
because of the lower-protein suspending medium. Because
with performing the ABO typing. Patients who type as neg-
of its lower-protein base and ready availability, the chemically
ative on the initial test for the D antigen are often interpreted
modified anti-D replaced the need for saline (IgM) anti-D
as Rh-negative and given Rh-negative units for transfusion.
reagents.
It is not required that a weak D test be performed because
As monoclonal antibody production became available, Rh
even if the patient truly was a weak D phenotype, giving
monoclonal antibodies were produced. These reagents are
Rh-negative RBCs to an Rh-positive individual is not a safety
derived from single clones of antibody-producing cells. The
risk.33 Additionally, transfusion services are required to
antibody-producing cells are hybridized with myeloma cells
repeat the Rh typing on all red blood cell units labeled as
to increase their reproduction rate, thereby maximizing their
Rh-negative. This testing is a safety measure to verify that
antibody-producing capabilities. Because the D antigen is
the unit of blood is truly Rh-negative and prevent transfusion
composed of many epitopes and the monoclonal Rh antibod-
errors due to testing or labeling errors.
ies have a narrow specificity, monoclonal anti-D reagents are
Pregnant women are tested for the D antigen to see if they
usually a combination of monoclonal anti-D reagents from
are candidates for Rh-immune globulin. Testing for weak
several different clones to ensure reactivity with a broad
D phenotype is optional. Women who test Rh-negative on
spectrum of Rh-positive RBCs. Reagents that are a blend of
initial screening may be interpreted as Rh-negative, and
IgM and IgG anti-D are routinely used to maximize visuali-
treated as such for the entire pregnancy. (See Chapter 20,
zation of reactions at immediate spin testing and to allow in-
“Hemolytic Disease of the Newborn and Fetus.”)
direct antiglobulin testing for weak D antigen with the same
reagent. Monoclonal blends can be used for slide, tube, mi- Detecting and Identifying Rh Antibodies
crowell, and most automated Rh testing, but the U.S. Food
and Drug Administration has also approved monoclonal IgM Patients who are pregnant or anticipating the need for trans-
anti-D reagents for column agglutination techniques. Be- fusion must be tested for the presence of all clinically signif-
cause these reagents are not human-derived, they lack all icant antibodies, including the Rh antibodies.32 The method
potential for transmitting infectious disease. must include incubation at 37°C, which culminates in an
Reagents are also available to test for C, E, c, and e anti- antiglobulin test. Recall that Rh antibodies are IgG class and
gens to aid in antibody identification and screening for optimally react at 37°C. If Rh antibodies are suspected, then
antigen-negative units of blood. As with all commercial typ- a complete antibody identification workup should be per-
ing reagents, Rh antigen typing must be performed with formed to identify the antibody specificity (or specificities)
strict adherence to manufacturer’s directions, use of proper present in the serum.
6888_Ch07_149-172 31/10/18 9:46 AM Page 163

Chapter 7 The Rh Blood Group System 163

Table 7–9 False Reactions With Rh Typing Reagents


False-Positives False-Negatives
Likely Cause Corrective Action Likely Cause Corrective Action

1. Cell suspension too 1. Adjust suspension, retype 1. Immunoglobulin-coated 1. Use saline-active typing reagent
heavy cells (in vivo)

2. Cold agglutinins 2. Wash with warm saline, retype 2. Saline-suspended cells 2. Use unwashed cells
(slide)

3. Test incubated too long 3. Follow manufacturer’s 3. Failure to follow manufac- 3. Review directions; repeat test
or drying (slide) instructions precisely turer’s directions precisely

4. Rouleaux 4. Use saline-washed cells, retype 4. Omission of reagent 4. Always add reagent first and
manufacturer’s directions check before adding cells

5. Fibrin interference 5. Use saline-washed cells, retype 5. Resuspension too vigorous 5. Resuspend all tube tests gently

6. Contaminating low- 6. Try another manufacturer’s 6. Incorrect reagent selected 6. Read vial label carefully; repeat
incidence antibody in reagent or use a known serum
reagent antibody

7. Polyagglutination 7. See chapter on polyagglutination 7. Variant antigen 7. Refer sample for further
investigation

8. Bacterial contamination 8. Open new vial of reagent, retype 8. Reagent deterioration 8. Open new vial
of reagent vial

9. Incorrect reagent 9. Repeat test; read vial label 9. Incorrect reagent selected 9. Repeat test, read vial label
selected carefully carefully

10. Centrifugation too long 10. Repeat test using shorter 10. Centrifugation too short 10. Repeat test using longer
centrifugation time centrifugation time

11. rpm too high 11. Repeat test using lower rpm 11. rpm too low 11. Repeat testing using higher rpm

Rh antibodies react stronger with cells that have a double need for blood negative for high-prevalence antigens can be
dose of the corresponding antigen. An example of this is an facilitated by contacting the blood supplier or the American
anti-C that reacts stronger with a cell that is C+c– and Red Cross American Rare Donor Program (ARDP).34
weaker with a cell that is C+c+. Identification of Rh antibod-
ies can be aided by the use of special techniques and Clinical Considerations
reagents. Rh antibody reactivity can be enhanced by adding
reagents such as albumin, low ionic strength solution (LISS), Determining an individual’s RhD type and testing to detect
and polyethylene glycol (PEG) to the test system. Another and identify RH antibodies has been reviewed. It is important
method to enhance the reactions is to treat the reagent red to understand the clinical implications when Rh incompati-
blood cells with proteolytic enzymes such as ficin. For bility exists.
more discussion of antibody identification techniques, see
Chapter 10, “Detection and Identification of Antibodies.” Transfusion Reactions

Selecting Compatible Blood for Transfusion When Rh antibodies are detected, a careful medical history
will reveal RBC exposure through pregnancy or transfusion
Rh antibodies are clinically significant, so units that are neg- of products containing RBCs. Circulating antibody can ap-
ative for the corresponding antigens must be selected for pear as soon as 10 to 14 days of a primary exposure and
transfusion. Commercial antisera is available to test for the within 1 to 7 days after a secondary exposure.
five common Rh antigens, as well as some of the other less Rh-mediated hemolytic transfusion reactions, whether
common types. The search for compatible units can be aided caused by primary sensitization or secondary immunization,
by recalling the frequencies of the Rh antigens. For example, usually result in extravascular destruction of immunoglobulin-
the C antigen is present in 70% of the Caucasian population coated RBCs. The transfusion recipient may have an unex-
and 34% of the African American population. Using com- plained fever, a mild bilirubin elevation, and a decrease in
mercial antisera to screen the available ABO/Rh-compatible hemoglobin and haptoglobin. The direct antiglobulin test
units in house can be accomplished easily. The e antigen is (DAT) is usually positive, and the antibody screen may or
present in up to 99% of the general population, so screening may not demonstrate circulating antibody. When the DAT in-
available units in house may not yield a compatible unit. The dicates that the donor cells within the recipient’s circulation
6888_Ch07_149-172 31/10/18 9:46 AM Page 164

164 PART II Blood Groups and Serologic Testing

are coated with IgG, elution studies may be helpful in defin- In the second type of Rhnull syndrome (the amorphic
ing the offending antibody specificity. If antibody is detected type), there is a mutation in each of the RHCE genes inher-
in either the serum or eluate, subsequent RBC transfusions ited from each parent as well as the deletion of the RHD gene
should lack the implicated antigen. It is not unusual for a per- found in most D-negative individuals. The RHAG gene is
son with a single Rh antibody to produce additional Rh anti- normal.
bodies if further stimulated.35 Even after the Rh antibodies It should be noted that Rhnull individuals of either regu-
are no longer detectable in the patient’s serum, RBCs must be lator or amorphic type are negative for the high-prevalence
selected that lack the corresponding antigens to prevent antigen LW and for FY5, an antigen in the Duffy blood group
transfusion reaction. system. S, s, and U antigens found on glycophorin B may
also be depressed.38 Individuals with Rhnull syndrome
Hemolytic Disease of the Fetus and Newborn demonstrate a mild compensated hemolytic anemia, reticu-
locytosis, stomatocytosis, a slight-to-moderate decrease in
Hemolytic disease of the fetus and newborn (HDFN) is hemoglobin and hematocrit levels, an increase in hemoglo-
briefly described here because of the historic significance of bin F, a decrease in serum haptoglobin, and possibly an ele-
the discovery of the Rh system in elucidating its cause. Anti-D vated bilirubin level.38 The severity of the syndrome is highly
was a frequent cause of fetal death before the implementation variable from individual to individual, even within one fam-
of Rh-immune globulin therapy. This was to be expected ily. Rhnull individuals, if exposed to normal Rh cells through
because while 15% of the population is Rh-negative, 85% transfusion or pregnancy, can produce a potent antibody,
of the population is Rh-positive; therefore, the likelihood anti-Rh29, which reacts with all cells except for those that
an Rh-negative woman is carrying an Rh-positive fetus is are Rhnull. When transfusion of individuals with Rhnull syn-
significant. HDFN caused by Rh antibodies is often severe drome is necessary, only Rhnull blood can be given. The Rhnull
because the Rh antigens are well developed on fetal cells, phenotype is usually written in Fisher-Race as —/—, in
and Rh antibodies are primarily IgG, which readily cross the Wiener as Rhnull, and in Rosenfield nomenclature as (RH: –1,
placenta. –2, –3, –4, –5).
After years of research, a method was developed to pre- Individuals of the Rhmod phenotype have a partial sup-
vent susceptible (D-negative) mothers from forming anti-D, pression of RH gene expression caused by mutations in the
thus preventing RhD HDFN. Rh-immune globulin, a purified RHAG gene. When the resultant RhAG protein is altered,
preparation of IgG anti-D, is given to D-negative woman dur- normal Rh antigens are also altered, often causing weak-
ing pregnancy and following delivery of a D-positive fetus.36 ened expression of the normal Rh and LW antigens.38 (See
Rh-immune globulin is effective only in preventing RhD the Landsteiner Wiener Blood Group System section later
HDFN. No effort has been made to develop immune globulin in this chapter.) Rhmod RBCs exhibit other blood group
products for other Rh antigens (e.g., C, c, E, e). Passively ac- antigens; however, like Rhnull, S, s, and U antigen expres-
quired anti-D in the serum may be noted in mothers who re- sion may be depressed. Rhmod individuals exhibit RBC ab-
ceive antepartum Rh-immune globulin, so it is critical to normalities similar to those with the Rhnull syndrome;
obtain an accurate medical history to establish the reason for however, clinical symptoms are usually less severe and
the presence of the anti-D. When present, RhD HDFN may rarely clinically remarkable.38
be severe and may require aggressive treatment. Refer to
Chapter 20, “Hemolytic Disease in the Newborn and Fetus,” Unusual Phenotypes and Rare Alleles
for a more detailed discussion of HDFN—its etiology, serol-
Several of the less frequently encountered Rh antigens are
ogy, and treatment.
described briefly in the following paragraphs. While the an-
tibodies directed against these antigens are less commonly
Rh Deficiency Syndrome: Rhnull encountered, they can be clinically significant and, in some
and Rhmod cases, cause transfusion reactions, HDFN, or both.
Rare individuals have Rh deficiency or Rhnull syndrome and Cw
fail to express any Rh antigens on the RBC surface. Rhnull
syndrome is inherited in one of two ways—amorphic and Cw was originally considered an allele at the C/c locus. Later
regulator. Other rare individuals exhibit a severely reduced studies showed that it can be expressed in combination with
expression of all Rh antigens, a phenotype called Rhmod. both C and c and in the absence of either allele. It is now
Individuals who lack all Rh antigens on their RBCs are known that the relationship between C/c and Cw is only phe-
said to have Rhnull syndrome, which can be produced by two notypic and that Cw is antithetical to the high-prevalence
different genetic mechanisms.37 In the regulator-type Rhnull antigen MAR.39 Cw results from a single amino acid change
syndrome, a mutation occurs in the RHAG gene. This results most often found on the RhCe protein. Cw is found in about
in no RhAG protein expression and subsequently no RhD or 2% of Caucasians and is very rare in African Americans.
RhCE protein expression on the RBCs, even though these Anti-Cw has been identified in individuals without
individuals usually have a normal complement of RHD and known exposure to foreign RBCs and after transfusion or
RHCE genes. These individuals can pass normal RHD and pregnancy. Anti-Cw may show dosage (i.e., reacting more
RHCE genes to their children. strongly with cells from individuals who are homozygous
6888_Ch07_149-172 31/10/18 9:46 AM Page 165

Chapter 7 The Rh Blood Group System 165

for Cw). Commercial anti-Cw reagent is not readily avail- identification testing, anti-G reacts as though it were a com-
able, but because of the low prevalence of the Cw antigen bination of anti-C plus anti-D because most C-positive and
RBCs compatible at the crossmatch in the antiglobulin D-positive cells are G+.42 G was originally described in an
phase may be selected for transfusion. Anti-Cw may not rr person who received D+C–E–c+e+ RBCs. Subsequently,
always be detected on routine antibody screens because the the recipient produced an antibody that appeared to be anti-
low frequency of the antigen means that some screening cell D plus anti-C, which should be impossible because the
sets may not have a Cw-positive cell included. C antigen was not on the transfused RBCs. Further investi-
gation showed the antibody was directed toward D + G anti-
f (ce) gens only and did not include anti-C. This also explains
situations in which an Rh-negative individual who has re-
The f antigen is expressed on the RBC when both c and e are ceived Rh-negative blood will look like they made anti-D.
present on the same haplotype. It has been called a com- The Rh-negative blood the patient received was C-positive,
pound antigen; however, f is likely a single entity resulting thus G-positive and the antibody produced is actually anti-G,
from conformational changes in the RHCE protein.40 The not anti-D and anti-C.
antigen f was included in a series of these compound anti- Anti-G versus anti-D and anti-C is important when eval-
gens, which were previously referred to as cis products to in- uating obstetric patients. If the patient has produced anti-G
dicate that the antigens were on the same haplotype. It is now and not anti-D, then she is considered a candidate for Rh-
known they are expressed on the RHCEe protein. Phenotyp- immune globulin.36 Elaborate adsorption and elution stud-
ically, DCE/dce and DcE/DCe cells will react the same when ies, usually performed in an immunohematology reference
tested with the five major Rh antisera: D+C+E+c+e. However, laboratory, may be valuable in these situations to discrimi-
when tested with anti-f, only the DCE/dce shows positive nate anti-D from anti-C from anti-G.
reactivity, confirming the former genotype (Table 7–10). For transfusion purposes, it is not necessary to discrimi-
Anti-f is generally a weakly reactive antibody often found nate anti-D and anti-C from anti-G, as the patient would
with other antibodies. It has been reported to cause HDFN receive D-negative and C-negative blood regardless if the
and transfusion reactions. In case of transfusion, f-negative antibody is anti-D, anti-C, or anti-G.
blood should be provided. Anti-f is not available as a reagent.
It is not necessary to give blood that is negative for both c Rh17 (Hr0)
and e antigens. It is adequate to provide either c-negative or
e-negative blood since all c-negative or e-negative individu- Rh17, also known as Hr0, is an antigen present on all RBCs
als are f-negative. with the “common” Rh phenotypes (e.g., R1R1, R2R2, rr).43
In essence, this antibody is directed to the entire protein
rhi (Ce) resulting from the RHCE genes. When RBCs phenotype as
D– (i.e. D+C-E-c-e-) the most potent antibody they make is
Similar to f, rhi was considered a compound antigen present often one directed against Rh17 (Hr0), which would react
when C and e are on the RhCe protein.41 Therefore anti-rhi with all cells except D–.
would only react with cells from an individual with a haplo-
type of DCe or Ce (Table 7–10). Rh23, Rh30, Rh40, and Rh52
Antigens cE (RH27) and CE (Rh22) also exist, but anti-
bodies produced to these antigens are not commonly seen Rh23, Rh30, and Rh40 are all low-prevalence antigens
(Table 7–10). associated with a specific category of partial D. These low-
prevalence antigens result from the formation of the hybrid
G proteins seen in individuals with partial-D phenotypes.
Rh23 (also known as Wiel and Dw) is an antigenic marker
G is an antigen present on most D-positive and all C-positive for category Va partial-D.44 Rh30 (also known as Goa or
RBCs. The antigen results from the amino acid serine at po- Dcor) is a marker for partial DIVa.45 Rh40 (also known as
sition 103 on the RhD, RhCe, and RhCE proteins. In antibody Tar or Targett) is a marker for partial DVII.46 Rh52 or BARC
is associated with some partial-DVI types.29
Table 7–10 Differentiation of Unusual
Rh33 (Har)
Rh Antibodies
R1R1 R1R2 rr R2R2 RoRo Rzr The low-prevalence antigen Rh33 is most often found in
cell cell cell cell cell cell whites and is associated with the rare variant haplotype
called R0Har.47 R0Har gene codes for normal amounts of c,
Anti-rhi (Ce) + + 0 0 0 0 reduced amounts of e, reduced f, reduced Hr0, and reduced
Anti-f 0 0 + 0 + + amounts of D antigen written as (D)c(e). The D reactions
are frequently so weak that the cells are often typed as Rh-
Anti-RH27 (cE) 0 + 0 + 0 0 negative. As previously discussed, R0Har or DHAR results
Anti-RH22 (CE) 0 0 0 0 0 + from a hybrid gene RHCE-RHD-RHCE in which only a small
portion of RHD is inserted into the RHCE gene.
6888_Ch07_149-172 31/10/18 9:46 AM Page 166

166 PART II Blood Groups and Serologic Testing

Rh32 The V antigen, also historically referred to as ceS, is found


in about 30% of randomly selected African Americans. (It is
Rh:32 is a low-prevalence antigen associated with a variant most often found in individuals with Gly263 amino acid
=
of the R1[D(C)(e)] haplotype called RN (pronounced “R dou- change in Rhce; several other mutations also occur in this
ble bar N”).48 The C antigen and e antigen are expressed gene.)
weakly. The D antigen expression is exaggerated or exalted. The VS antigen, also known as eS, occurs in 32% of
This gene has been found primarily in African Americans. African Americans. It results from a single amino acid change
that occurs at position Val245 in the Rhce protein. Most hrB-
Rh43 (Crawford) individuals with r’s genotype are VS+. The Val245 mutation
in this haplotype is found in the hybrid RHDIIIa-RHCE-
Rh43, also known as the Crawford antigen, is a low-prevalence RHDIIIa gene described previously.50 Most V+ individuals
antigen on a variant Rhce protein.31 The Crawford (ceCF) anti- are also VS+.
gen is of very low prevalence found in individuals of African
descent. Deletions

e Variants There are very uncommon phenotypes that demonstrate no


Cc and/or Ee reactivity. Many examples lacking all Cc or Ee
Like the variant D antigen seen in individuals possessing a often have an unusually strong D antigen expression, fre-
hybrid or mutated RHD gene, some individuals of African quently called exalted D. The deletion phenotype is indicated
or mixed ethnic backgrounds possess e antigen that by the use of a dash (–), as in the following example: D–.
exhibits similar qualities as those described for partial-D This phenotype results from individuals possessing normal
phenotypes—that is, an individual may have a phenotype RHD gene(s) and hybrid RHCE-RHD-RHCE in which the
of e-positive but produce antibodies behaving as anti-e.49 Rhce protein is replaced with RhD. This helps explain the
These variant types result from multiple mutations in the exalted D, as there are extra D antigens recognized on the
RHCE gene. Individuals who possess two altered RHCE resulting hybrid protein. The antibody made by D–/D– peo-
genes may have a phenotype of e-positive but produce ple is called anti-Rh17 or anti-Hr0.51
antibodies behaving as anti-e. A variation has been recognized within the deletion D–,
The Rh antigens hrB (Rh31) and hrS (Rh19 )are rarely called D••. The D antigen in the D•• is stronger than that
encountered in routine blood banking. They are normally in DC–, or Dc–, samples but weaker than that of D– samples.
present in individuals who possess normal RhCe or Rhce A low-prevalence antigen called Evans (Rh:37) accompanies
protein but are lacking in individuals with normal RhcE the Rh structure of D•• cells.52
or RhCE proteins (i.e., e-negative). Several antigens, most Some deletion phenotypes are missing E/e only and are
notably hrB and hrS, are lacking on the Rhce proteins because indicated as Dc– or DC–. To date, a deletion of only C/c has
of a variant RHCE gene. If individuals with these variant not been reported.
genes who are hrB-negative or hrS-negative are immunized, Transfusion of individuals with a D– or D•• phenotype is
they may produce anti-hrB or anti-hrS. In routine antibody difficult, as blood of a similar phenotype or even the rarer
identification, these antibodies are generally nonreactive Rhnull blood, all lacking Rh17, would be required.
with e-negative red blood cells (and therefore are also hrB-
negative and hrS-negative), appearing to have anti-e-like Landsteiner Wiener Blood Group
specificity. Individuals with an anti-hrB or anti-hrS require System
RBCs that are lacking in the specific antigens, which are
exceedingly rare. A discussion of the Landsteiner Wiener (LW) antigen begins
To further complicate this, the variant RHCE gene associ- with the time when Rh antigens were first recognized. The
ated with the hrB-negative phenotype is usually found with antibody produced by injecting rhesus monkey RBCs into
a variant RHD-CE-D hybrid gene. The RHCE is inserted in guinea pigs and rabbits was identified as having the same
the middle of the RHDIIIa gene. The resulting protein lacks specificity as the antibody Levine and Stetson described ear-
D antigen but possesses an unusual form of the C antigen. lier.1 The antibody was given the name anti-Rh, for anti-
The hybrid gene occurs in 22% of individuals of African rhesus, and the blood group system was established. Many
Americans.49 This haplotype that includes the RHDIIIa- years later, it was recognized that the two antibodies were
RHCE-DIIIa hybrid gene and variant RHCE genes is referred not identical; the anti-rhesus described by Landsteiner and
to as r’s[(C)ces]. Wiener was renamed anti-LW in their honor.2
Phenotypically, there is a similarity between the Rh and
V and VS LW systems.53 Anti-LW reacts strongly with most D-positive
RBCs, weakly with Rh-negative RBCs (and sometimes not at
V and VS are antigens of low prevalence in the Caucasian all), and never with Rhnull cells. A weak anti-LW may be pos-
population but are more prevalent among African Americans. itive only with D-positive RBCs, and enhancement tech-
These antigens can be used as predictors of an individual’s niques may be required to demonstrate its reactivity with
ethnic background because of this difference in prevalence. D-negative cells. Anti-LW shows equal reactivity with cord
6888_Ch07_149-172 31/10/18 9:46 AM Page 167

Chapter 7 The Rh Blood Group System 167

cells regardless of their D type. This is an important charac- techniques can help distinguish between Anti-D and LW
teristic to remember when trying to differentiate anti-LW antibodies. One method is to treat the reagent panel cells
from anti-D. Also, anti-LW more frequently appears as an with 0.2 m dithiothreitol (DTT) and test the patient serum
autoantibody, which does not present clinical problems. Fur- against the treated cells. LW antigens are denatured with
ther discussion can be found in Chapter 9, “Uncommon DTT treatment while D antigens are unaffected.54 Another
Blood Groups.” strategy is to test the patient serum against Rh-positive and
Rh-negative cord blood cells. Anti-D will react only with
the Rh-positive cord cells whereas the LW antibodies will
Advanced Concepts react with all cord cells tested, regardless of Rh type. See
Since LW antibodies can mimic the reactivity of anti-D, Chapter 10, “Detection and Identification of Antibodies,”
antibody identification can be challenging. Using special for more information on special techniques.

CASE STUDIES
Case 7-1

Two units of blood are ordered for an 89-year-old woman with myelodysplastic syndrome. She had been transfused 3 years
ago and has six children. Routine pretransfusion testing follows.
Type and Screen Results
Anti-A Anti-B Anti-D A1 Cells B Cells
O O O 4+ 4+
Rh MNS P Lewis Kell Duffy Kidd GEL

D C E c e f M N S s P1 Lea Leb K k Fya Fyb Jka Jkb IAT


R1R1 + + 0 0 + 0 0 + 0 + + 0 + 0 + 0 + 0 + 2+
R2R2 + 0 + + 0 0 + + + 0 0 + 0 0 + + 0 + 0 2+
rr 0 0 0 + + + + 0 + 0 + 0 + + + + 0 + + 0
Because the antibody screen was positive, the patient’s plasma was tested against an antibody identification cell panel
using the GEL technique:
GEL Antibody Identification Panel
Rh MNS P Lewis Kell Duffy Kidd GEL
D C E c e f M N S s P1 Lea Leb K k Fya Fyb Jka Jkb IAT
R1r + + 0 + + + 0 + 0 + + 0 + 0 + 0 + 0 + 2+
R2R2 + 0 + + 0 0 + + + 0 0 + 0 0 + + 0 + 0 2+
R1R2 + + + + + 0 + 0 + 0 + 0 + + + + 0 + + 2+
R1R1 + + 0 0 + 0 + + + + 0 0 + 0 + + + + + 2+
rr 0 0 0 + + + + 0 + 0 + + 0 + 0 + + 0 + 0
r⬘r 0 + 0 + + + 0 + 0 + 0 0 + 0 + 0 + + 0 0
r⬘r⬙ 0 + + + + 0 + 0 + + + 0 + + + + 0 + + 0
r⬘r⬘ 0 + 0 0 + 0 0 + 0 + 0 0 + 0 + 0 0 0 + 0
RoRo + 0 0 + + + 0 + 0 + + 0 + 0 + + + + + 2+
ryr⬙ 0 + + 0 0 0 + 0 + 0 0 0 + + 0 + 0 + 0 0
R1Ro + + 0 + + + + 0 + + + 0 + + + 0 + 0 + 2+
Patient 0
Cells
1. What is the ABO/Rh of the patient?
2. How would you interpret the results of the antibody detection test/screen?
3. Which antibody(antibodies) is(are) present in the patient’s plasma?
6888_Ch07_149-172 31/10/18 9:46 AM Page 168

168 PART II Blood Groups and Serologic Testing

4. What additional testing is needed (if any)?


5. What ABO/Rh type RBCs should be selected for transfusion?

Case 7-2

A 55-year-old Caucasian man is scheduled for knee replacement surgery and a type and screen was ordered. He has a
history of two prior transfusions. The first was 3 years ago during a prostatectomy, at which time he received two units of
RBCs. The second transfusion was 6 months ago during surgery to remove a bowel obstruction, when he received one
unit of RBCs.
Anti-A Anti-B Anti-D A1 cells B cells
O O 3+ 4+ 4+
Rh MNS P Lewis Kell Duffy Kidd GEL

D C E c e f M N S s P1 Lea Leb K k Fya Fyb Jka Jkb IAT


R1R1 + + 0 0 + 0 0 + 0 + + 0 + 0 + 0 + 0 + 0
R2R2 + 0 + + 0 0 + + + 0 0 + 0 0 + + 0 + 0 2+
rr 0 0 0 + + + + 0 + 0 + 0 + + + + 0 + + 2+
Because the antibody screen was positive, a full antibody identification panel was tested with the patient’s plasma and
the results are below.
GEL Antibody Identification Panel
Rh MNS P Lewis Kell Duffy Kidd GEL

D C E c e f M N S s P1 Lea Leb K k Fya Fyb Jka Jkb IAT


1-R1r + + 0 + + + 0 + 0 + + 0 + 0 + 0 + 0 + 2+
2-RoRo + 0 0 + + + + + + 0 0 + 0 + 0 + 0 + 0 2+
3-R1r + + 0 + + + + 0 + 0 + 0 + + + + 0 + + 2+
4-R1R1 + + 0 0 + 0 + 0 + 0 0 0 + 0 + 0 + 0 + 0
5-R1R1 + + 0 0 + 0 0 + 0 + + + 0 + + + 0 + 0 0
6-r⬘r⬘ 0 + 0 0 + 0 0 + 0 + 0 0 + 0 + 0 + + 0 0
7-R1r⬘ + + 0 0 + 0 + 0 + + + 0 + + + + 0 + + 0
8-ryr⬘ 0 + + 0 + 0 0 + 0 + 0 0 + 0 + 0 0 0 + 2+
9-R1Rz + + + 0 + 0 0 + 0 + + 0 + 0 + + + + + 2+
10-ryr⬘ 0 + + 0 + 0 + 0 + 0 0 0 + + 0 + 0 + 0 2+
11-R1Ro + + 0 + + + + 0 + + + 0 + + + 0 + 0 + 2+
Patient + + 0 0 + 0 + 0 + 0 + 0
cells*
* Patient cells were phenotyped for some antigens and the results are written above
1. What is the patient’s ABO/RH?
2. Which antibody specificity(specificities) is(are) present?
3. What antibodies (if any) cannot be ruled out based on the panel results? For this particular case study, please rule out
using ONLY cells that have a double dose of the antigen.
4. What additional testing should be performed?
5. Using Fisher-Race and Wiener nomenclature, what is the patient’s most probable genotype?
6. Does the patient’s antigen type confirm or conflict with your antibody identification?
7. What type of RBCs should be selected in the event of a transfusion? (That is, which ABO/Rh; list any special antigen
typing that must be performed.)

Case 7-3 (Advanced)

A 29-year-old female, pregnant for the second time, presented to her OB physician in her early first trimester for initial
prenatal care. She states that her first pregnancy was uneventful and the baby was born without incident.
6888_Ch07_149-172 31/10/18 9:46 AM Page 169

Chapter 7 The Rh Blood Group System 169

Pretransfusion Type and Screen


Anti-A Anti-B Anti-D A1 cells B cells
4+ O O O 4+
Antibody Screen
Rh MNS P Lewis Kell Duffy Kidd GEL

D C E c e f M N S s P1 Lea Leb K k Fya Fyb Jka Jkb IAT


R1R1 + + 0 0 + 0 0 + 0 + + 0 + 0 + 0 + 0 + 2+
R2R2 + 0 + + 0 0 + + + 0 0 + 0 0 + + 0 + 0 2+
rr 0 0 0 + + + + 0 + 0 + 0 + + + + 0 + + 0
GEL Antibody Identification Panel
Rh MNS P Lewis Kell Duffy Kidd GEL

D C E c e f M N S s P1 Lea Leb K k Fya Fyb Jka Jkb IAT


1-R1r + + 0 + + + 0 + 0 + + 0 + 0 + 0 + 0 + 2+
2-RoRo + 0 0 + + + + + + 0 0 + 0 0 + + 0 + 0 2+
3-R2r + 0 + + + + + 0 + 0 + 0 + + + + 0 + + 2+
4-R2R2 + 0 + + 0 0 + 0 + 0 0 0 + 0 + 0 + 0 + 2+
5-R1R1 + + 0 0 + 0 0 + 0 + + + 0 + 0 + 0 + 0 2+
6-rr 0 0 0 + + 0 0 + 0 + 0 + 0 0 + 0 + + 0 0
7-r⬘r⬘ 0 + 0 0 + 0 + 0 + + + 0 + + + + 0 + + 2+
8-ryr 0 + + + + 0 0 + 0 + 0 0 + 0 + 0 0 0 + 2+
9-rr 0 0 0 + + 0 0 + 0 + + 0 + 0 + + + + + 0
10-r⬙r⬙ 0 0 + + 0 0 + 0 0 + 0 0 + + 0 + 0 + 0 0
rr 0 0 0 + + + + 0 + 0 + 0 0 + + 0 + 0 + 0
Patient 0 0 0 + + 0 + + + + 0 + 0 + 0 + + + 0
cells *
* Patient cells were phenotyped by the laboratory to aid in antibody identification, and the results are recorded here.
1. What is the patient’s ABO, Rh type?
2. Which antibody(antibodies) is(are) present in the serum?
3. Which antibodies could not be ruled out using a double-dose cell?
4. What additional testing is recommended?
5. Discuss the likelihood of the risk of hemolytic disease of the fetus and newborn for this patient.

SUMMARY CHART
 The Rh antibody was so named on the basis of anti-  A person who expresses no Rh antigens on the RBC
body production by guinea pigs and rabbits when is said to be Rhnull, and the genotype may be written
transfused with rhesus monkey RBCs. as –––/–––.
 Historically, Rh antibodies were a primary cause of  In the Wiener Rh-Hr nomenclature, it is postulated
HDFN, erythro-blastosis fetalis, and a significant cause that the gene responsible for defining Rh actually pro-
of hemolytic transfusion reactions. duces an agglutinogen that contains a series of three
 Fisher-Race DCE terminology is based on the theory blood factors in which each factor is an antigen recog-
that antigens of the system are produced by three nized by an antibody.
closely linked sets of alleles and that each gene is re-  In the Rosenfield alpha/numeric terminology, a num-
sponsible for producing a product (or antigen) on the ber is assigned to each antigen of the Rh system
RBC surface. in order of its discovery (Rh1 = D, Rh2 = C, Rh3 = E,
Rh4 = c, Rh5 = e).
Continued
6888_Ch07_149-172 31/10/18 9:46 AM Page 170

170 PART II Blood Groups and Serologic Testing

SUMMARY CHART—cont’d
 It is currently accepted that two closely linked genes  The Rh antigens are inherited as codominant alleles.
on chromosome 1 control the expression of Rh;  A partial-D individual is characterized as lacking one
one gene (RHD) codes for the presence of RhD, and or more pieces or epitopes of the total D antigen and
a second gene (RHCE) codes for the expression of may produce an alloantibody to the missing fraction if
CcEe antigens. These genes interact with the RHAG exposed to RBCs with the complete D antigen.
gene on chromosome 6 to form Rh antigens.  Blood donor units for transfusion are considered
 Rh antigens are characterized as nonglycosylated pro- Rh-positive if either the D or weak D test is positive; if
teins in the RBC membrane, and do not appear on both the D and weak D tests are negative, blood for
other tissues or in body fluids. transfusion is considered Rh-negative.
 D antigen is highly immunogenic: exposure to less  Rh antibodies are IgG and can cross the placenta to
than 0.1 mL of Rh-positive RBCs can stimulate anti- coat fetal (Rh-positive) RBCs.
body production in an Rh-negative person.  Rh-mediated hemolytic transfusion reactions usually
 The most common phenotype in Caucasians is R1r result in extravascular hemolysis.
(31%); the most common phenotype in African
Americans is R0r (23%), followed by R0R0 at 19%.

6. Biochemically speaking, what type of molecules are Rh


Review Questions
antigens?
1. The Rh system genes are: a. Glycophorins
b. Simple sugars
a. RHD and RHCE.
c. Proteins
b. RHD and LW.
d. Lipids
c. RHD and RHAG.
d. RHCE and RHAG. 7. Rh antibodies react best at what temperature (°C)?
2. What Rh antigen is found in 85% of the Caucasian pop- a. 15
ulation and is always significant for transfusion purposes? b. 18
c. 22
a. d
d. 37
b. c
c. D 8. Rh antibodies are primarily of which immunoglobulin
d. E class?
3. How are weaker-than-expected reactions with anti-D a. IgA
typing reagents categorized? b. IgD
c. IgG
a. Rhmod
d. IgM
b. Weak D
c. DAT positive 9. Rh antibodies have been associated with which clinical
d. Dw condition?
4. Cells carrying a weak D antigen require the use of what a. Hemolytic disease of the fetus and newborn
test to demonstrate its presence? b. Thrombocytopenia
c. Hemophilia A
a. Indirect antiglobulin test
d. Stomatocytosis
b. Direct antiglobulin test
c. Microplate test 10. What do Rhnull cells lack?
d. Warm autoadsorption test a. Lewis antigens
5. How are Rh antigens inherited? b. Normal oxygen-carrying capacity
c. Rh antigens
a. Autosomal recessive alleles
d. Hemoglobin
b. Sex-linked genes
c. Codominant alleles
d. X-linked
6888_Ch07_149-172 31/10/18 9:46 AM Page 171

Chapter 7 The Rh Blood Group System 171

11. Convert the following genotypes from Wiener nomen- 11. Blood Group Antigen Gene Mutation Database [Internet].
clature to Fisher-Race and Rosenfield nomenclatures, Bethesda (MD): National Center for Biotechnology Informa-
tion (NCBI) [updated 2014 Aug 15; cited 2017 March 20].
and list the antigens present in each haplotype. Available from: https://www.ncbi.nlm.nih.gov/projects/gv/mhc/
a. R1r xslcgi.cgi?cmd=bgmut/systems_info&system=rh
b. R2R0 12. Ridgewell K, Spurr NK, Laguda B, MacGeoch C, Avent ND,
c. RzR1 Tanner MJ. Isolation of cDNA clones for a 50 kDa glycoprotein
of the human erythrocyte membrane associated with Rh (rhe-
d. r⬘r
sus) blood-group antigen expression. Biochem J. 1992;287:
12. Which Rh phenotype has the strongest expression of D? 223-28.
13. Colin Y, Cherif-Zahar B, Le Van K, Raynal V, Van Huffel V,
a. DCe/ce Cartron JP. Genetic basis of the RhD-positive and RhD-negative
b. DCe/DCe blood group polymorphism as determined by Southern analy-
c. DcE/DcE sis. Blood. 1991;78:2747-52.
d. D– 14. Singleton BK, Green CA, Avent ND, Martin PG, Smart E,
Daka A, et al. The presence of an RHD pseudogene contain-
13. An individual has the following serologic reactions: ing a 37 base pair duplication and nonsense mutation in
D+C+E+c+e+f–. What is the most probable genotype? Africans with the RhD-negative blood group phenotype.
Blood. 2000;95(1):12-18.
a. R1R2 15. Shao CP, Maas JH, Su YQ, Kohler M, Legler TJ. Molecular back-
b. Rory ground of Rh D-positive, D-negative, D (el) and weak D phe-
c. Rzr notypes in Chinese. Vox Sang. 2002;83(2):156-61.
d. R1r⬙ 16. Avent ND, Butcher SK, Liu W, Mawby WJ, Mallinson G,
Parsons SF, et al. Localization of the C termini of the Rh
14. Which of the following is the most common haplotype (rhesus) polypeptides to the cytoplasmic face of the human
erythrocyte membrane. J Biol Chem. 1992;267(21):15134-39.
in the African American population? 17. Westhoff CM, Wylie DE. Transport characteristics of mam-
a. DCe malian Rh and Rh glycoproteins expressed in heterologous sys-
b. DcE tems. Transfus Clin Biol. 2006;13(1-2):132-38.
c. Dce 18. Hughes-Jones NC, Gardner B, Lincoln PJ. Observations of the
number of available c, D, and E antigen sites on red cells. Vox
d. ce
Sang. 1971;21(3):210.
15. If a patient who is R1R1 is transfused with RBCs that are 19. Westhoff CM. The Rh blood group system in review: a new face
for the next decade. Transfusion. 2004;44(11):1663-73.
Ror, which antibody is he most likely to produce? 20. Garratty G, Glynn S, McEntire R. ABO and Rh(D) phenotype
a. Anti-D frequencies of different racial/ethnic groups in the United
b. Anti-c States. Transfusion. 2004:44(5):703-6.
c. Anti-e 21. Müller T, Wagner F, Trockenbacher A, Eicher N, Flegel W,
Schönitzer D, et al. PCR screening for common weak D types
d. Anti-G shows different distributions in three Central European popu-
lations. Transfusion. 2001;41(1):45-52.
References 22. Ceppellini R, Dunn LC, Turri M. An interaction between alleles
at the Rh locus in man which weakens the reactivity of the Rh0
1. Levine P, Stetson RE. An unusual case of intragroup agglutina- factor (D0). Proc Natl Acad Sci. 1955;15;41(5):283-88.
tion. JAMA. 1939;113(2):126-27. 23. Wagner F, Gassner C, Müller TH, Schönitzer D, Schunter F,
2. Landsteiner K, Wiener AS. An agglutinable factor in human Flegel WA. Molecular basis of weak D phenotypes. Blood.
blood recognized by immune sera for rhesus blood. Proc Soc 1999;93(1):385-93.
Exp Biol. 1940;43:223. 24. Daniels G. Variants of RhD-current testing and clinical conse-
3. Levine P, Burnham L, Katzen E, Vogel P. The role of isoim- quences. Br J Hematol. 2013;161(4):461-67.
munization in the pathogenesis of erythroblastosis fetalis. 25. Sandler SG, Flegel W, Westhoff CM, Denomme GA, Delaney
Am J Obstet Gynecol. 1941;42(6):925. M, Keller, MA et al. It’s time to phase in RHD genotyping for
4. Race RR. The Rh genotypes and Fisher’s theory. Blood. 1948; patients with a serologic weak D phenotype. Transfusion.
(3 Suppl 2):27-42. 2014;55(3):680-89.
5. Wiener AS. Genetic theory of the Rh blood types. Proc Soc Exp 26. Wiener AS, Unger LJ. Rh factors related to the Rh0 factor as a
Biol. 1943;54:316. source of clinical problems. JAMA. 1959;169(7):696-99.
6. Rosenfield RE, Allen F, Swisher S, Kochwa S. A review of Rh 27. Tippett P, Sanger R. Observations on subdivisions of the Rh
serology and presentation of a new terminology. Transfusion. antigen D. Vox Sang. 1962;7(1):9.
1962;2(5):287. 28. Huang CH, Liu P, Cheng JG. Molecular biology and genetics
7. Lewis M, Anstee DJ, Bird G, Brodheim E, Cartron JP, Contr- of the Rh blood group system. Semin Hematol. 2000;37(2):
eras M, et. al. Blood group terminology 1990. Vox Sang. 150-65.
1990;58(2):152-69. 29. Tippett P, Lomas-Francis C, Wallace M. The Rh antigen D:
8. Tippett P. A speculative model for the Rh groups. Ann Human partial D antigens and associated low incidence antigens.
Genet. 1986;50(3): 241-47. Vox Sang. 1996;70(3):123-31.
9. Le van Kim C, Mouro I, Cherif-Zahar B, Raynal V, Cherrier C, 30. Beckers EA, Faas BH, von dem Borne AE, Overbeeke MA, van
Cartron JP, et al. Molecular cloning and primary structure of Rhenen DJ, van der Schoot CE. The R0Har RH:33 phenotype re-
human blood group RhD polypeptide. Proc Natl Acad Sci USA. sults from substitution of exon 5 of the RHCE gene by the corre-
1992; 89(22):10925-29. sponding exon of the RHD gene. Br J Haem. 1996;92(3):751-57.
10. The Rhesus Site [Internet]. Ulm: Wagner FF, Flegal WA. 31. Flegel WA, Wagner FF, Chen Q, Schlanser G, Frame T, West-
[Updated 2017 Feb 9; cited 2017 Mar 20]. Available from: hoff CM, et al. The RHCE allele ceCF: the molecular basis of
http://www.uni-ulm.de/~wflegel/RH/ Crawford (RH43). Transfusion. 2006;46(8):1334-42.
6888_Ch07_149-172 31/10/18 9:46 AM Page 172

172 PART II Blood Groups and Serologic Testing

32. AABB. Standards for blood banks and transfusion services. 44. Chown B, Lewis M, Kaita H, Phillips S. The Rh antigen Dw
31st ed. Bethesda (MD): AABB; 2018. (Wiel). Transfusion. 1964;4(3):169.
33. Mintz P. Transfusion therapy clinical principals and practice. 45. Von Zabern I, Wagner FF, Moulds JM, Moulds JJ, Flegel WA. D
3rd ed. Bethesda (MD): AABB Press; c2011. p. 317. category IV: a group of clinically relevant and phylogenetically
34. Flickinger C, Petrone T, Church A. Review: American diverse partial D. Transfusion. 2013;53(11 Pt 2):2960-73.
Rare Donor Program. Immunohematology. 2004:20(4): 46. Lewis M, Kaita H, Allderdice PW, Bartlett S, Squires WG,
239-43. Huntsman RG. Assignment of the red cell antigen Targett
35. Mintz P. Transfusion therapy clinical principals and practice. (Rh 40) to the Rh blood group systems. Am J Hum Genet.
3rd ed. Bethesda: AABB Press; 2011. p. 634. 1979;31(5):630-33.
36. De Haas M, Thurik FF, Koelewijn, JM, van der Schoot, CE. 47. Giles CM, Skov F. An Rh gene complex which results in a
Haemolytic disease of the fetus and newborn. Vox Sang. 2015; “new” antigen detectable by a specific antibody, anti-Rh 33.
109(2):99-113. Vox Sang. 1971;21(4):289.
37. Chown B, Lewis, M, Kaita, H, Lowen, B. An unlinked modifier 48. Rosenfield RE, Haber GV, Schroeder R, Ballard R. Problems in
of Rh blood groups: effects when heterozygous and when Rh typing as revealed by a single Negro family. Am J Hum
homozygous. Am J Hum Genet. 1972;24(6 part 1):623. Genet. 1960;12(2):147.
38. Schmidt PJ, Vos GH. Multiple phenotypic abnormalities asso- 49. Chou ST, Westhoff CM. Molecular biology of the Rh system:
ciated with Rhnull (—/—). Vox Sang. 1967;13(1):18. clinical considerations for transfusion in sickle cell disease.
39. Sistonen P, Saraneva H, Pirkola A, Eklund J. MAR. A novel Hematology. 2009;178-84.
high-incidence Rh antigen revealing the existence of an allele 50. Daniels GL, Faas BH, Green CA, Smart E, Maaskant-van Wilk
sub-system including Cw (Rh8) and Cx (Rh9) with exceptional PA, Advent ND, et al. The VS and V blood group polymor-
distribution in the Finnish population. Vox Sang. 1994;66(4): phisms in Africans: a serologic and molecular analysis. Trans-
287-92. fusion. 1998;38(10):951-58.
40. Chen YX, Peng J, Novaretti M, Reid ME, Huang CH. Deletion 51. Reid M, Lomas-Francis C, Olsson M. The blood group antigen
of arginine codon 229 in the Rhce gene alters e and f but not fact book. 3rd ed. London (Eng). 2012.
c antigen expression. Transfusion. 2004;44(3):391-98. 52. Contreras M, Stebbing B, Blessing M, Gavi, J. The Rh antigen
41. Rosenfield RE, Haber GV. An Rh blood factor, Rh1 (Ce) and its Evans. Vox Sang. 1978;34(4):208-11.
relationship to hr (ce). Am J Hum Genet. 1958;10(4):474. 53. Daniels, G. Human blood groups. 3rd ed. Oxford: Wiley-
42. Allen FH, Tippett PA. A new Rh blood type which reveals the Blackwell; 2013. p. 391-92.
Rh antigen G. Vox Sang. 1958;3(5):321. 54. Miola MP, Cervo SVB, Fachini RM, Ricci Jr. O. Do not confuse
43. Issitt PD. Applied blood group serology. 4th ed. Miami (FL): anti-LW autoantibodies with anti-D. Revista Brasileira de
Montgomery Scientific Publication; 1998. Hematologia e Hemoterapia. 2013;35(3):198.
6888_Ch08_173-211 29/10/18 5:27 PM Page 173

Chapter 8
Blood Group Terminology and Common Blood
Groups: The Lewis, P, I, MNS, Kell, Duffy,
Kidd, and Lutheran
Regina M. Leger, MSQA, MT(ASCP)SBB, CMQ/OE(ASQ)
and Angela K. Novotny MT(ASCP)SBBCM

Introduction The MNS (002) System Fyx


Blood Groups and Terminology M and N Antigens Fy3 Antigen and Antibody
The Lewis (007) System S and s Antigens Fy5 Antigen and Antibody
Lewis Antibodies Anti-M The Kidd (009) System
Genetics and Biosynthesis Anti-N Jka and Jkb Antigens
Development of Lewis Antigens Anti-S and Anti-s Anti-Jka and Anti-Jkb
Other Lewis Antigens Biochemistry Biochemistry
The P Blood Group: P1PK (003) and Genetics Genetics
Globoside (028) Systems and Related GPA- and GPB-Deficient Phenotypes Jk(a–b–) Phenotype and the Recessive
(209) Antigens Other Antibodies in the MNS System Allele, Jk
The P1 Antigen Autoantibodies Jk(a–b–) Phenotype and the
Anti-P1 Disease Associations Dominant In(Jk) Allele
Biochemistry The Kell (006) and Kx (019) Systems Anti-Jk3
Genetics K and k Antigens Autoantibodies
Other Sources of P1 Antigen and Kpa, Kpb, and Kpc Antigens The Lutheran (005) System
Antibody Lua and Lub Antigens
Jsa and Jsb Antigens
Anti-PP1Pk Anti-Lua
Anti-K
Alloanti-P Anti-Lub
Antibodies to Kpa, Jsa, and Other Low-
Autoanti-P Associated With Prevalence Kell Antigens Biochemistry
Paroxysmal Cold Hemoglobinuria Genetics
Antibodies to k, Kpb, Jsb, and Other
Antibodies to Compound Antigens High-Prevalence Kell Antigens Lu(a–b–) Phenotypes
Luke (LKE) Antigen Biochemistry Anti-Lu3
PX2 Antigen Genetics Applications to Routine Blood Banking
Disease Associations The Kx Antigen Case Studies
The I (027) System and i Antigen The Ko Phenotype and Anti-Ku Summary Chart
The I and i Antigens The McLeod Phenotype and Syndrome Review Questions
Anti-I Altered Expressions of Kell Antigens References
Anti-i Autoantibodies
Biochemistry and Genetics The Duffy (008) System
Other Sources of I and i Antigen Fya and Fyb Antigens
The IT Antigen and Antibody Anti-Fya and Anti-Fyb
Antibodies to Compound Antigens Biochemistry
Disease Associations Genetics

173
6888_Ch08_173-211 29/10/18 5:27 PM Page 174

174 PART II Blood Groups and Serologic Testing

OBJECTIVES
Blood Groups and Terminology
1. Describe how antigens, antibodies, genes, and phenotypes are correctly written.
2. List the four categories for classification of RBC surface blood group antigens used by the ISBT.
3. For each of the blood group systems described in this chapter:
• List the major antigens and common phenotypes.
• Describe the serologic characteristics and clinical significance of the antibodies.
• Identify null phenotypes.

Antigen Characteristics
4. Describe the formation of Lewis antigens and their adsorption onto RBCs.
5. Define the interaction of Lewis genes with ABO, H, and secretor genes.
6. List substances present in secretions and the Lewis phenotypes based on a given genotype.
7. Describe the reciprocal relationship of I antigen to i antigen.
8. List the antigen frequencies for the common antigens K, M, S, s, Fya, Fyb, Jka, Jkb, and P1.
9. Define Kpa, Jsa, and Lua as low-prevalence antigens and Kpb, Jsb, Lub, and I as high-prevalence antigens.
10. Define the association of M and N with glycophorin A (GPA) and S and s with glycophorin B (GPB).
11. Describe the antigen phenotypes S–s–U–, Js(a+), and Fy(a–b–) associated with blacks.
12. Define the null phenotypes Mk, p, Ko, Fy(a–b–), Jk(a–b–), and Lu(a–b–), and describe their role in problem-solving.
13. Compare dominant and recessive forms of the Lu(a–b–) and Jk(a–b–) phenotypes.
14. Explain I and P1 antigens as being poorly expressed on cord RBCs.
15. Describe the phenotypic relationship between LW and Rh.
16. List the antigens that are denatured by routine blood bank enzymes (M, N, S, s, Fya, Fyb) and antigens whose reactivity with
antibody is enhanced with enzymes (I, i, P1, Jka, Jkb).
17. List which antigens are destroyed by treatment with DTT (dithiothreitol).

Antibody Characteristics
18. List the characteristics of the Lewis antibodies, including clinical significance.
19. Define antibodies to M, N, I, and P1 as being typically non-RBC–induced (“naturally occurring”), cold-reacting agglutinins
that are usually clinically insignificant.
20. Describe antibodies to K, k, S, s, Fya, Fyb, Jka, and Jkb as usually induced by exposure to foreign RBCs (“immune”),
antiglobulin-reactive antibodies that are clinically significant.
21. List the antibody specificities that commonly show dosage (anti-M, -N, -S, -s, -Fya, -Fyb, -Jka and -Jkb).

Clinical Significance and Disease Association


22. List and correlate the common 37°C antihuman globulin–reactive antibodies to K, k, S, s, Fya, Fyb, Jka, and Jkb with hemolytic
transfusion reactions (HTR) and hemolytic disease of the fetus and newborn (HDFN).
23. Describe why the Kidd antibodies are a common cause of delayed HTR.
24. Define the relationship of autoanti-I with Mycoplasma pneumoniae infections and autoanti-i with infectious mononucleosis.
25. Describe the common characteristics of the McLeod syndrome, including very weak Kell antigen expression, acanthocytosis,
and late onset of muscular and neurological abnormalities.
26. Describe the association of the Fy(a–b–) phenotype with Plasmodium vivax resistance.
27. Explain Jk(a–b–) RBCs with a resistance to lysis that normally occurs in 2M urea, a common diluting fluid used for automated
platelet counters.
6888_Ch08_173-211 29/10/18 5:27 PM Page 175

Chapter 8 Blood Group Terminology and Common Blood Groups 175

Introduction RBCs. Some genes code for complex structures that carry
more than one antigen (e.g., the glycophorin B structure,
The first part of this chapter covers blood group terminology which carries S or s antigen, also carries the U antigen).
and how antigens are organized into a classification scheme. Silent, or amorphic, alleles exist that make no antigen, but
Conventions for the correct use of terminology are included they are rare. When paired chromosomes carry the same
so that the student will be able to accurately communicate silent allele, a null phenotype results. Null phenotype RBCs
information about antigens and antibodies, phenotypes, and can be very helpful when evaluating antibodies to unknown
genotypes. high-prevalence antigens. For example, an antibody reacting
The ABO and Rh blood groups are the most significant in with all test cells except those with the phenotype Lu(a–b–)
transfusion practice. However, there are currently 346 RBC may be directed against an antigen in the Lutheran system or
antigens in 36 systems that are formally recognized inter- an antigen phenotypically related to the Lutheran system. In
nationally. With the advances in genotyping leading to iden- some blood group systems, the null phenotype results in RBC
tification of variants within the blood group systems, the abnormalities.
number of RBC antigens and blood group systems can be Some blood group systems have regulator or modifying
expected to grow. Refer to the International Society of Blood genes, which alter antigen expression. These are not neces-
Transfusion (ISBT) website (www.isbtweb.org) for the Red sarily located at the same locus as the blood group genes they
Cell Immunogenetics and Blood Group Terminology Work- affect and may segregate independently. For example, RBCs
ing Party page for current classifications. Blood group anti- with the dominant type of Lu(a–b–) have suppressed expres-
gens are defined by carbohydrates (sugars) attached to sion of all the antigens in the Lutheran blood group system
glycoprotein or glycolipid structures or by amino acids on as well as many other antigens, including P1 and i. This
a protein. The carbohydrate blood groups Lewis, P, and modifying gene is inherited independently of the genes
I will be presented first. The antigens of these carbohydrate coding for Lutheran, P1, and i antigens.
systems (like ABO and H) are not encoded by their genes Blood group antigens are detected by alloantibodies,
directly. Rather, the genes encode specific glycosyltrans- which occur naturally (i.e., without a known immune stim-
ferases that in turn synthesize the carbohydrate epitopes by ulus) or as a response by the immune system after exposure
sequential addition of sugars to a precursor. to nonself RBC antigens introduced by blood transfusion or
The next blood groups discussed are MNS, Duffy, Kell, pregnancy. Over the last century, blood group antigens have
Kidd, and Lutheran. These are significant in routine trans- been named using several styles of symbols: uppercase single
fusion medicine; antibodies to these antigens are more com- letters, uppercase and lowercase letters to represent alleles,
monly encountered. Other uncommon blood groups and letters derived from the name of the system (some with
selected antigens/antibodies will be covered in Chapter 9, superscripts to represent alleles), letter symbols followed by
“Uncommon Blood Groups.” numbers, and finally the more recent use of all uppercase
Antigen and antibody characteristics are discussed for letters.
each blood group presented. Included are the effects of en- Although gene and antigen names seem confusing at
zymes and chemical treatments on test RBCs. This infor- first, certain conventions are followed when writing alleles,
mation is a tool for antibody identification. For example, antigens, and phenotypes.1 Some examples are given in
if an antibody no longer reacts with a panel of RBCs after Table 8–1. Genes are written in italics or underlined when
the RBCs have been pretreated with an enzyme such as ficin italics are not available (e.g., when handwritten), and their
or papain, then only those antibodies that don’t react after allele number or letter is always superscript. Antigen names
enzyme treatment are considered for the specificity, and all are written in regular type without italics or underlining;
other specificities can be excluded. These antigen and an- some antigens have numbers or superscript letters. A phe-
tibody characteristics are summarized on the front and notype is a description of which antigens are present on an
back cover of the book so that students can easily access individual’s RBCs and simply indicates the results of sero-
pertinent information while performing routine antibody logic tests on those RBCs. It is important to use subscripts,
identification. superscripts, and italics appropriately. For example, A1,
A1, and A1 mean the antigen, the phenotype, and the allele,
Blood Groups and Terminology respectively.
How the phenotype is written depends on the antigen
A blood group system is one or more antigens produced nomenclature and whether letters or numbers are used. For
by alleles at a single gene locus or at loci so closely linked letter antigens, a plus sign or minus sign written on the same
that crossing over does not occur or is very rare.1 With a few line as the antigen is used to designate that the antigen is
notable exceptions, most blood group genes are located on present or absent, respectively. Examples are M+ and K–. For
the autosomal chromosomes and demonstrate straightfor- antigens that have superscripts, the letter of the superscript
ward Mendelian inheritance. is placed in parentheses on the same line as the letter defin-
Most blood group alleles are codominant and express a ing the antigen—for example, Fy(a+) and Jk(a–). When test-
corresponding antigen. For example, a person who inherits ing for both of the antithetical antigens, both results are
alleles K and k expresses both K and k antigens on his or her written within the parentheses—for instance, Fy(a–b+). For
6888_Ch08_173-211 29/10/18 5:27 PM Page 176

176 PART II Blood Groups and Serologic Testing

Table 8–1 Examples of Terminology for Genes, Antigens, Antibodies, and Phenotypes
Gene Antigen Antibody Phenotype
System Conventional ISBT Antigen Positive Antigen Negative
Lewis Le LE Lea Anti-Lea Le(a+) Le(a–)

Leb Anti-Leb Le(b+) Le(b–)


le

MNS M MNS*1 M Anti-M M+ M–


N MNS*2 N Anti-N N+ N–
S MNS*3 S Anti-S S+ S–
s MNS*4 s Anti-s s+ s–
Kell K KEL*1 K Anti-K K+ K–
k KEL*2 k Anti-k k+ k–
Kidd Jka JK*1 Jka Anti-Jka Jk(a+) Jk(a–)
Jkb JK*2 Jkb Anti-Jkb Jk(b+) Jk(b–)

antigens that have a numerical designation, the letter(s) Table 8–2 Examples of Correct and
defining the system is followed by a colon, followed by the
Incorrect Terminology
number representing the antigen. No plus sign is written if
the antigen is present, but a minus sign is placed before the Correct Incorrect
negative result, and multiple results are separated by a Fy(a+) Fya+, Fy(a+), Fya+, Fya(+), Duffy a-positive, Duffya+
comma (e.g., Sc:–1,2). Phenotypes of more than one blood
group system are separated by a semicolon—for example, Fy(a–b+) Fya–b+, Fya(–)Fyb(+)
S+s+; K–; Fy(a+b–). Anti-Fya Anti Fya, anti-Duffy, anti-Duffya
Remember that serologic tests determine only RBC phe-
notype, not genotype. A genotype is composed of the actual K Kell (name of system), K1 (obsolete)
genes that an individual has inherited and can be determined Anti-k Anti-Cellano, anti-K2 (obsolete)
only by family or DNA studies. Sometimes the genotype can
be predicted or inferred by the phenotype. When based on M+N– M(+), MM
results from RBC antigen typing, the genotype is a probable Ge:–2 Ge2–, Ge:2–, Ge2-negative
interpretation as to which genes the individual carries in
order to have the observed phenotype.
Antibodies are described by their antigen notation with
the prefix anti-, including a hyphen before the antigen sym- three digits represent the system, collection, or series, and
bol. Use of correct blood group terminology, especially for the second three digits identify the antigen. The antigens
antibodies identified in a patient’s serum, is very important within the system are numbered sequentially in order of dis-
so that correct information is conveyed for patient care. covery. Each system also has an alphabetical symbol. For ex-
Some examples of correct and incorrect terminology are ample, using the ISBT terminology, the K antigen is 006001
given in Table 8–2. with the first three numbers (006) representing the system
Numeric terminology was originally introduced for the and the second three numbers (001) representing the number
Kell and Rh systems and was subsequently applied to other assigned to K, or as 6.1(omitting the sinistral zeroes). Anti-
systems. To facilitate computer storage and retrieval of blood gens can also be written using the system symbol followed
group information and to help standardize blood group sys- by the antigen number; for instance, KEL1. This committee’s
tem and antigen names, the International Society of Blood work is ongoing, and the assignment of RBC antigens to
Transfusion (ISBT) formed the Working Party on Terminol- blood group systems is periodically updated. The first mono-
ogy for Red Cell Surface Antigens in 1980.2 The numeric graph was released in 1990, and updates are published in
system this group proposed was not intended to replace tra- Vox Sanguinis or the associated ISBT Science Series 3 and on
ditional terminology but rather to enable communication the Working Party’s page of the ISBT website (http://www
on computer systems where numbers are necessary. Each .isbtweb.org/working-parties/red-cell-immunogenetics-and-
antigen is given a six-digit identification number. The first blood-group-terminology/).4
6888_Ch08_173-211 29/10/18 5:27 PM Page 177

Chapter 8 Blood Group Terminology and Common Blood Groups 177

The ISBT assigns RBC antigens to a system, collection, and Collections are antigens that have a biochemical, sero-
low- or high-prevalence series. This chapter uses traditional logic, or genetic relationship but do not meet the criteria for
terminology, but the ISBT symbol and number are indicated a system.5 Antigens classified as a collection are assigned a
for the blood group systems and collections discussed. 200 number. Some of the previously established collections
As defined by the ISBT, a blood group system “consists of have been made obsolete as criteria have been met to estab-
one or more antigens controlled at a single gene locus, or by lish a system or incorporate antigens into an existing system.
two or more very closely linked homologous genes with little See Table 8–4 for a listing of the ISBT collections.4
or no observable recombination between them.”5 Each sys- All remaining RBC antigens that are not associated with a
tem is genetically distinct. To date, there are 36 blood group system or a collection are catalogued into the 700 series of low-
systems (Table 8–3).4 prevalence antigens or the 901 series of high-prevalence

Table 8–3 International Society of Blood Transfusion (ISBT) Blood Group Systems
Gene Name Chromosomal No. of
Number Name Symbol ISBT Location Antigens CD Numbers
001 ABO ABO ABO 9q 4
002 MNS MNS GYPA, GYPB, 4q 49 CD235a (GPA)
(GYPE)
CD235b (GPB)
003 P1PK P1PK A4GALT 22q 3 CD77
004 Rh RH RHD, RHCE 1p 55 CD240
005 Lutheran LU BCAM 19q 24 CD239
006 Kell KEL KEL 7q 36 CD238
007 Lewis LE FUT3 19p 6
008 Duffy FY ACKR1 (formerly 1q 5 CD234
known as DARC )
009 Kidd JK SLC14A1 18q 3
010 Diego DI SLC4A1 17q 22 CD233
011 Yt YT ACHE 7q 3
012 Xg XG XG, MIC2 Xp 2 CD99 (MIC2)
013 Scianna SC ERMAP 1p 7
014 Dombrock DO ART4 12p 10 CD297
015 Colton CO AQP1 7p 4
016 Landsteiner-Wiener LW ICAM4 19p 3 CD242
017 Chido/Rodgers CH/RG C4A, C4B 6p 9
018 H H FUT1 19q 1 CD173
019 Kx XK XK Xp 1
020 Gerbich GE GYPC 2q 11 CD236
021 Cromer CROM CD55 (formerly 1q 19 CD55
known as DAF)
022 Knops KN CR1 1q 9 CD35
023 Indian IN CD44 11p 5 CD44
024 Ok OK BSG 19p 3 CD147
025 Raph RAPH CD151 11p 1 CD151
Continued
6888_Ch08_173-211 29/10/18 5:27 PM Page 178

178 PART II Blood Groups and Serologic Testing

Table 8–3 International Society of Blood Transfusion (ISBT) Blood Group Systems—cont’d
Gene Name Chromosomal No. of
Number Name Symbol ISBT Location Antigens CD Numbers
026 John Milton JMH SEMA7A 15q 6 CD108
Hagen
027 I I GCNT2 6p 1
028 Globoside GLOB B3GALT1 3q 2
029 Gill GIL AQP3 9p 1
030 Rh-associated RHAG RHAG 6p 4 CD241
glycoprotein
031 FORS FORS GBGT1 9q 1
032 JR JR ABCG2 4q 1 CD338
033 LAN LAN ABCB6 2q
034 Vel VEL SMIM1 1q 1
035 CD59 CD59 CD59 11p 1 CD59
036 Augustine AUG SLC29A1 6p 2

Table 8–4 International Society of Blood Transfusion Blood Group Collections


Collection Antigen
Number Name Symbol Number Symbol Incidence %

205 Cost COST 205001 Csa 95


205002 Csb 34

207 Ii I 207002 i

208 Er ER 208001 Era >99


208002 Erb <1
208003 Er3 >99

209 GLOB 209003 LKE 98

210 210001 Lec 1


210002 Led 6

213 MN CHO 213001 Hu


213002 M1
213003 Tm
213004 Can
213005 Sext
213006 Sj

antigens. Refer to Table 8–5 for a listing of low-prevalence anti- by the terms high- and low-prevalence, reflecting the occurrence
gens and Table 8–6 for a listing of high-prevalence antigens of an inherited characteristic at the phenotypic level.6
recognized by the ISBT.4 Antigens in these series represent
those with a prevalence of less than 1% or more than 90% of The Lewis (007) System
most random populations, respectively. As terminology has
evolved, the terms high- and low-incidence, also previously The Lewis blood group system is unique because the Lewis
known as high- and low-frequency, are currently being replaced antigens are not intrinsic to RBCs but are on type 1
6888_Ch08_173-211 29/10/18 5:27 PM Page 179

Chapter 8 Blood Group Terminology and Common Blood Groups 179

Table 8–5 International Society of Blood Transfusion Antigens of Low (700 Series) Prevalence
Number Name Symbol Number Name Symbol
700002 Batty By 700039 Milne

700003 Christiansen Chra 700040 Rasmussen RASM

700005 Biles Bi 700044 JFV

700006 Box Bxa 700045 Katagiri Kg

700017 Torkildsen Toa 700047 Jones JONES

700018 Peters Pta 700049 HJK

700019 Reid Rea 700050 HOFM

700021 Jensen Jea 700054 REIT

700028 Livesay Lia

Table 8–6 International Society of Blood Transfusion Antigens of High (901 Series) Prevalence
Number Name Symbol Number Name Symbol
901008 Emm 901014 PEL

901009 Anton AnWj 901015 ABTI

901012 Sid Sda 901016 MAM

glycosphingolipids that are passively adsorbed onto the RBC Table 8–7 Phenotypes of the Lewis
membrane from the plasma. The Lewis system was named
System
after one of the first individuals to make the antibody,
reported by Mourant in 1946.7 This antibody, later called Adult Phenotype
anti-Lea, agglutinated RBCs from about 25% of English Phenotype Prevalence (%)
people.8 In 1948, an antibody, later called anti-Leb, was found Whites Blacks
that reacted with Le(a–) individuals. It was thought that Lea
and Leb were antithetical antigens, but we now know this is Le(a+b–) 22 23
not so because they do not result from alternative alleles of a Le(a–b+) 72 55
single gene. Rather, they result from the interaction of two
fucosyltransferases encoded by independent genes, Le and Se. Le(a–b–) 6 22
The Lewis blood group system has been assigned the ISBT Le(a+b+) Rare Rare
system number 007 and the system symbol LE.
There are several Lewis antigens, but the two of primary
concern are Lea and Leb. The Lewis (Le, FUT3) gene is located
on chromosome 19 at position 19p13.3, as is the secretor (Se, Le(a–b–) RBC phenotype are either secretors or nonsecretors.8
FUT2) gene at 19q13.3.4 There are two alleles at the Lewis The Le(a–b–) phenotype is found more frequently among
locus, Le and the amorph le, and there are two alleles at the Africans. The Le(a+b+) phenotype is rare among whites and
secretor locus, Se and the amorph se. The biochemistry and Africans but is more frequent among Asians, with a prevalence
interaction of the genes at these two loci will be explained of 10% to 40%.10 The antigens of the Lewis blood group sys-
in a later section, but in short, the Le gene must be present tem recognized by the ISBT are listed in Table 8–8.
for a precursor substance to be converted to Lea, but the Se Lewis antigens are not expressed on cord RBCs and are
gene must also be present for conversion to Leb. The four often diminished on the mother’s RBCs during pregnancy.
phenotypes resulting from the interaction of these two genes Lewis antigens are found on lymphocytes and platelets and on
are shown in Table 8–7. other tissues such as the pancreas, stomach, intestine, skeletal
In 1948, it was observed that individuals with Le(a+) muscle, renal cortex, and adrenal glands.10 In addition, soluble
RBCs were mostly nonsecretors of ABH.9 As a result, in gen- Lewis antigens are found in saliva as glycoproteins.
eral for adults, Le(a+b–) RBCs are from ABH nonsecretors and Lewis antigens are resistant to treatment with the en-
Le(a–b+) RBCs are from ABH secretors (refer to Chapter 6, zymes ficin and papain, dithiothreitol (DTT), and glycine-
“The ABO Blood Group System”). Individuals with the acid EDTA. Reactivity of Lewis antibodies can be greatly
6888_Ch08_173-211 29/10/18 5:27 PM Page 180

180 PART II Blood Groups and Serologic Testing

Table 8–8 Antigens of the Lewis Blood Group System


Antigen ISBT Number Comment
Lea LE1

Leb LE2

Leab LE3 Anti-Leab reacts with Le(a+b–) and Le(a–b+) RBCs from adults and with 90% cord RBCs.*

LebH LE4 Anti-LebH reacts with group O Le(b+) and A2 Le(b+) RBCs.

ALeb LE5 Anti-Aleb reacts with group A Le(b+) and AB Le(b+) RBCs.

BLeb LE6 Anti-BLeb reacts with group B Le(b+) and AB Le(b+) RBCs.

*Reactive cord RBCs [serologically Le(a–b–)] are from babies with Le gene.

enhanced by testing with enzyme-treated RBCs; hemolysis recognizes any Leb antigen regardless of the ABO type.
of enzyme-treated RBCs may be seen if serum is tested. Anti-LebH should be suspected when anti-Leb is identified
with a panel of RBCs (group O) but most or all group A donor
Lewis Antibodies units are crossmatch compatible (as only about 20% of the
group A RBC units will be from group A2 individuals).
Lewis antibodies are often naturally occurring and made by Lewis antigens are not intrinsic to the RBC membrane and
Le(a–b–) persons; that is, they occur without any known are readily shed from transfused RBCs within a few days
RBC stimulus. They are generally IgM and do not cross the of transfusion. Also, Lewis blood group substance present
placenta. Because of this and because the Lewis antigens are in transfused plasma neutralizes Lewis antibodies in the
not well developed on fetal RBCs, the antibodies do not recipient.6 This is why it is exceedingly rare for anti-Lea or
cause hemolytic disease of the fetus and newborn (HDFN). anti-Leb to cause hemolysis of transfused RBCs.
Anti-Lea and anti-Leb may occur together and can be neu-
tralized by the Lewis substances present in plasma or saliva Genetics and Biosynthesis
or with commercially prepared Lewis substance. Lewis anti-
bodies occur quite frequently in the sera of pregnant women
who transiently exhibit the Le(a–b–) phenotype. Advanced Concepts
Most Lewis antibodies agglutinate saline-suspended The Le (FUT3) gene is linked to Se (FUT2) and H (FUT1),
RBCs, but these agglutinates are often fragile and can be eas- all located on chromosome 19. The synthesis of Lewis
ily dispersed if the cell button is not gently resuspended after antigens depends on the interaction of the transferases
centrifugation. Lewis antibodies can bind complement, and produced by the Lewis and secretor genes. The Lewis and
when fresh serum is tested, anti-Lea may cause in vitro he- secretor transferases preferentially fucosylate type 1
molysis of incompatible RBCs, although this is more often chains, whereas the H gene (FUT1) preferentially fucosy-
seen with enzyme-treated RBCs than with untreated RBCs. lates type 2 chains. Type 1 chain refers to the beta linkage
Anti-Lea is the most commonly encountered of the Lewis of the number 1 carbon of galactose to the number 3
antibodies and is often detected in room temperature tests, carbon of N-acetylglucosamine (GlcNAc) residue of the
but it sometimes reacts at 37°C and in the indirect antiglobulin precursor structure (Fig. 8–1). As shown in Figure 8–2,
test. Rare hemolytic transfusion reactions (HTR) have been
reported in patients with anti-Lea who were transfused with
Le(a+) RBCs, so anti-Lea that are reactive at 37°C, particularly
those that cause in vitro hemolysis, should not be ignored.1 It
Type 1 GAL Predominant
is relatively easy to find Le(a–) units since 80% of the popu-
type in
lation are secretors. Obtaining donor units that are typed GlcNAc secretions,
Le(a–) with reagent antisera is generally not considered nec- plasma, some
GAL ␤1 3 tissues
essary for these patients; units that are crossmatch compatible
␤1 3
in tests performed at 37°C are acceptable. Persons whose RBCs
are Le(a–b+) do not make anti-Lea, because small amounts of
Type 2
unconverted Lea are present in their plasma and saliva.
Anti-Leb is not as common or generally as strong as anti- GAL Predominant
type on
Lea. It is usually an IgM agglutinin and can bind complement. GAL GlcNAc RBCs
Anti-Leb is infrequently made by Le(a+b–) individuals and can ␤1 3
be classified into two categories: anti-LebH and anti-LebL. Anti- ␤1 4
LebH reacts best when both the Leb and the H antigens are Figure 8–1. Precursor type 1 and type 2 chains. The type 1 chain has a beta 1→3
present on the RBC, such as group O and A2 cells. Anti-LebH linkage of the terminal galactose (Gal) to the N-acetylglucosamine (GlcNAc) of the pre-
represents an antibody to a compound antigen. Anti-LebL cursor structure. The type 2 chain has a beta 1→4 linkage of the terminal galactose.
6888_Ch08_173-211 29/10/18 5:27 PM Page 181

Chapter 8 Blood Group Terminology and Common Blood Groups 181

Type 1 precursor Gal(␤1 3)GlcNAc-R


Le ␣1,4
Gal(␤1 3)GlcNAc-R Fuc
Se Lea

Gal(␤1 3)GlcNAc-R Gal(␤1 3)GlcNAc-R


␣1,2 Le ␣1,2 ␣1,4
Fuc Type 1H Fuc Fuc
Leb
A B
GalNAc(␣1 3)Gal(␤1 3)GlcNAc-R Gal(␣1 3)Gal(␤1 3)GlcNAc-R
␣1,2 ␣1,2
Fuc Type 1A Fuc Type 1B
Le Le

GalNAc(␣1 3)Gal(␤1 3)GlcNAc-R Gal(␣1 3)Gal(␤1 3)GlcNAc-R


␣1,2 ␣1,4 ␣1,2 ␣1,4
Figure 8–2. Formation of Lewis antigens. Gal = galactose; Fuc Fuc Fuc Fuc
GlcNAc = N-acetylglucosamine; Fuc = fucose; GalNAc =
ALeb BLeb
N-acetylgalactosamine.

secretor α1,2-L-fucosyltransferase adds a terminal fucose Table 8–9 Antigens and Phenotypes
to the type 1 chain to form type 1H. The Le allele codes Resulting From Interaction
for α1,4-L-fucosyltransferase, which transfers L-fucose to of Lewis, Secretor, and
type 1H chain on glycoprotein or glycolipid structures to ABO Genes
form Leb. Small amounts of Lea are made before the secre-
tor enzyme is able to add the terminal fucose. If these in- Lewis, Secretor, and ABO Genes
dividuals also have A or B genes, type 1H structures will Genes Antigens in Secretions RBC Phenotype
be converted to A or B structures and the Le fucosyltrans-
ferase will then produce ALeb or BLeb (see Fig. 8–2). Le, Se, A/B/H Lea, Leb, A, B, H A, B, H, Le(a – b+)
Individuals who are nonsecretors do not have the en- lele, Se, A/B/H A, B, H A, B, H, Le(a – b –)
zyme to convert type 1 chains in secretions to type 1H.
Consequently, the type 1 precursor is available for action Le, sese, A/B/H Lea A, B, H, Le(a+b –)
by the Le fucosyltransferase, with the result of Lea antigen lele, sese, A/B/H None A, B, H, Le(a – b –)
in secretions and Le(a+b–) RBCs. The Lea antigen cannot
be converted to Leb because of steric hindrance from the Le, sese, hh, A/B Lea Oh, Le(a+b –)
fucose added to make Lea. Individuals with a weak Se gene, Le, Se, hh, A/B Lea, Leb, A, B, H A*, B*, Le(a – b+)
common in Asia, produce a fucosyltransferase that com-
petes less effectively with the Le fucosyltransferase, result- *para-Bombay
ing in RBCs with the Le(a+b+) phenotype.8 The Le(a+b+)
phenotype is rare in European populations, and it occurs
in 10% to 40% of some Asian populations.10 Mutations that to Le(a+b–) or Le(a–b+), respectively. With saliva as a
result in inactivation of the fucosyltransferase, represented source of Lewis substances, Le(a–b–) RBCs cannot be con-
by the amorphic or silent le allele, are responsible for the verted into Lewis-positive phenotypes because Lewis sub-
Le(a–b–) phenotype. stances in saliva, being glycoproteins, are not adsorbed
All Le(a–b+) individuals are ABH secretors and also se- onto the RBC membranes.
crete Lea and Leb; very little Lea will be detected in the
plasma or on the RBCs. All Le(a+b–) individuals are ABH
nonsecretors, yet all secrete Lea. Approximately 78% to Development of Lewis Antigens
80% of whites are secretors, and 20% are nonsecretors. In
terms of Le(a–b–) individuals, 80% are ABH secretors, and Depending on the genes inherited, Lea and Leb glycoproteins
20% are ABH nonsecretors. The interaction of Lewis, se- will be present in the saliva of newborns, but Lewis glycolipids
cretor, and ABO genes is summarized in Table 8–9. are not detectable in the plasma until about 10 days after
Lewis antigens produced in saliva and other secretions birth. As a result, cord blood and RBCs from newborn infants
are glycoproteins, but Lewis cell-bound antigens absorbed phenotype as Le(a–b–). Some can be shown to be weakly
from plasma onto the RBC membranes are glycolipids. The Le(a+) when tested with a potent anti-Lea or with methods
gastrointestinal tract is thought to be the primary source of more sensitive than direct agglutination. Lewis antigens will
Lewis glycolipid in plasma.6 Le(a–b–) RBCs incubated with start to appear shortly after birth, with Lea developing first
plasma from Le(a+) or Le(b+) individuals can be converted when the Le gene is present. The Lewis fucosyltransferase is
more active than the secretor fucosyltransferase in newborns,
6888_Ch08_173-211 29/10/18 5:27 PM Page 182

182 PART II Blood Groups and Serologic Testing

so more type 1 chains are available for conversion to Lea. As system (028, symbol GLOB), and LKE is assigned to the
the secretor transferase activity increases, converting type 1 Globoside collection (209, symbol GLOB). The reader will
to type 1H, Leb will be detected. In children who inherit both notice the confusing use of GLOB as the symbol for both a
Le and Se genes, the transformation can be followed from system and a collection. For simplicity in this chapter, these
the Le(a–b–) phenotype at birth to Le(a+b–) after 10 days to antigens will be referred to as the P blood group.
Le(a+b+) and finally to Le(a–b+), the true Lewis phenotype, The P blood group was introduced in 1927 by Landsteiner
after about 6 years. In contrast, children who inherit Le and and Levine. In their search for new antigens, they injected
sese genes phenotype as Le(a–b–) at birth and transform to rabbits with human RBCs and produced an antibody, initially
Le(a+b–) after 10 days; the Le(a+b–) phenotype persists called anti-P, that divided human RBCs into two groups: P+
throughout life. Individuals with lele genes phenotype as and P–.8
Le(a–b–) at birth and for the rest of their lives. In 1951, Levine and colleagues11 described anti-Tja (now
known as anti-PP1Pk), an antibody to a high-prevalence
Other Lewis Antigens antigen that Sanger12 later showed was related to the P
blood group. Because anti-Tja defined an antigen common
Leab is present on all Le(a+b–) and Le(a–b+) RBCs and on 90% to P+ and P– cells and was made by an apparent P null
of cord RBCs. The antigen was previously known as Lex, but individual, the original antigen and phenotypes were
in 1998 the ISBT renamed it Leab.10 Anti-Leab is fairly common renamed. Anti-P became anti-P1; the P+ phenotype became
and is frequently found with anti-Lea or anti-Leb. The antibody P1; the P– phenotype became P2; and the rare P null indi-
is heterogeneous and occurs mainly in Le(a–b–) secretors of vidual became p.
group A1, B, or A1B. The antigens now known as Lex and Ley The P blood group became more complex in 1959 when
are products of FUT3 on type 2 precursor chains and are not Matson and coworkers13 described a new antigen, Pk. This
associated with the RBC surface and are not part of the Lewis antigen is expressed on all RBCs except those of the very rare
blood group.11 p phenotype, but it is not readily detected unless P is absent
ALeb and BLeb result from the addition of the A or B (i.e., in the P1k and P2k phenotypes).
immunodominant sugar, respectively, to type 1H chain in The phenotypes, antigens, and antibodies associated with
individuals who have at least one Se and one Le allele. Se the P blood group are summarized in Table 8–10. There are
converts type 1 chains to type 1H, providing a suitable two common phenotypes: P1 and P2, and three rare pheno-
acceptor for the A and B carbohydrates (see Fig. 8–2). types: p, P1k, and P2k. The P1 phenotype describes RBCs that
react with anti-P1 and anti-P; the P2 phenotype describes
The P Blood Group: P1PK (003) RBCs that do not react with anti-P1 but do react with anti-P.
and Globoside (028) Systems When RBCs are tested only with anti-P1 and not with
and Related (209) Antigens anti-P, the phenotype should be written as P1+ (or P1) or P1–.
Only when P1– RBCs are tested and found to be reactive
Traditionally, the P blood group comprised the P, P1, and Pk with anti-P should they be designated as phenotype P2. RBCs
antigens and, later, Luke (LKE), PX2, and NOR. The bio- of the p phenotype do not react with anti-P1, anti-P, or
chemistry and molecular genetics, although not completely anti-Pk. RBCs of the P1k phenotype react with anti-P1 and
understood as yet, make it clear that at least two biosynthetic anti-Pk but not with anti-P. RBCs of the P2k phenotype react
pathways and genes at different loci are involved in the de- with anti-Pk but not with anti-P1 or anti-P.
velopment and expression of these antigens. Consequently, Individuals with the p phenotype (P null) are very rare:
these antigens cannot be considered a single blood group 5.8 in a million. P nulls are slightly more common in Japan,
system. Currently, in ISBT nomenclature, P1, Pk, and NOR North Sweden, and in an Amish group in Ohio.1
are assigned to the P1PK blood group system (003, symbol The antibodies generally fall into two categories: clinically
P1PK), P and PX2 are assigned to the Globoside blood group insignificant or potently hemolytic.

Table 8–10 P Blood Group: Phenotypes, Antigens, and Antibodies


Phenotype Antigens Present Possible Antibodies Prevalence
Whites Blacks

P1 P1, P, Pk* None 79% 94%

P2 P, Pk* Anti-P1 21% 6%

p None Anti-PP1Pk Rare Rare

P1k P1, Pk Anti-P Very rare Very rare

P2k Pk Anti-P, anti-P1 Very rare Very rare

*Trace amounts of Pk antigen, not detectable by agglutination test.


6888_Ch08_173-211 29/10/18 5:27 PM Page 183

Chapter 8 Blood Group Terminology and Common Blood Groups 183

The P blood group antigens, like the ABH antigens, are phase, without typing for P1, is an acceptable approach to
synthesized by sequential action of glycosyltransferases, transfusion.
which add sugars to precursor substances. The precursor of Rare examples of anti-P1 that react at 37°C can cause in
P1 can also be glycosylated to type 2H chains, which carry vivo RBC destruction; both immediate and delayed HTRs
ABH antigens. P1, P, or Pk may be found on RBCs, lympho- have been reported.14 Anti-P1 is usually IgM; IgG forms are
cytes, granulocytes, and monocytes; P can be found on rare. HDFN is not associated with anti-P1, presumably
platelets, epithelial cells, and fibroblasts. P and Pk have also because the antibody is usually IgM and the antigen is so
been found in plasma as glycosphingolipids and as glycopro- poorly developed on fetal RBCs.
teins in hydatid cyst fluid.8 The antigens have not been iden-
tified in secretions. RBCs carry approximately 14 × 106 Biochemistry
copies of globoside, the P structure, per adult RBC and about
5 × 105 copies of P1.10
The P blood group antigens are resistant to treatment with Advanced Concepts
ficin and papain, DTT, chloroquine, and glycine-acid EDTA. The RBC antigens of the P blood group exist as glyco-
Reactivity of the antibodies can be greatly enhanced by sphingolipids. As with ABH, the antigens result from the
testing with enzyme-treated RBCs. sugars added sequentially to precursor structures. Bio-
chemical analyses have shown that the precursor substance
The P1 Antigen for P1 is also a precursor for type 2H chains that carry ABH
antigens. However, the genes responsible for the formation
The P1 antigen is poorly expressed at birth and may take up of the P1 and ABH antigens are independent.
to 7 years to be fully expressed.8 Antigen strength in adults There are two distinct pathways for the synthesis of
varies from one individual to another: RBCs from some P1+ the P blood group antigens, as shown in Figure 8–3. The
individuals are P1 strong (P1+s) and others are P1 weak common precursor is lactosylceramide (or Gb2, also
(P1+w). These differences may be controlled genetically or known as ceramide dihexose or CDH). The pathway on the
may represent homozygous versus heterozygous inheritance figure’s left results in the formation of paragloboside,
of the gene coding for P1. The strength of P1 can also vary P1 and PX2. Paragloboside is also the type 2 precursor for
with race. Blacks have a stronger expression of P1 than ABH. The pathway shown on the right side leads to the
whites. The gene mutation responsible for the In(lu) type production of the globoside series: Pk, P, NOR, and Luke
Lu(a–b–) RBCs, discussed in the Lutheran section, also af- (LKE). The NOR antigen is a low-prevalence antigen
fects the expression of P1 so that P1 individuals who inherit resulting from a single nucleotide change in A4GALT.10
this gene mutaion may type serologically as P1–.
The P1 antigen deteriorates rapidly on storage. When
older RBCs are typed or used as controls for typing reagents Genetics
or when older RBCs are used to detect anti-P1 in serum,
The P1PK gene (A4GALT) encoding the enzyme responsible
false-negative reactions may result.
for the synthesis of Pk, a 4-α-galactosyltransferase (Gb3 or
Pk synthase), was cloned independently by three research
Anti-P1 groups in 2000.15 The Globoside gene (B3GALNT1) encod-
ing the 3-β-N-acetylgalactosaminyltransferase (Gb4 syn-
Anti-P1 is a common, naturally occurring IgM antibody in
thase) that is responsible for converting Pk to P was also
the sera of P1– individuals. Anti-P1 is typically a weak, cold-
reactive saline agglutinin optimally reactive at 4°C and not
seen in routine testing. Stronger examples react at room tem- Lactosylceramide (Gb2)
perature, and rare examples react at 37°C and bind comple- A4GALT
ment, which is detected in the antiglobulin test when
polyspecific (anti-IgG plus anti-C3) reagents are used. Anti- Pk antigen
Lactotriaosylceramide Globotriosylceramide (Gb3)
body activity can be neutralized or inhibited with soluble P1
substance. If room temperature incubation is not included, B3GALNT1
antibody activity can often be bypassed altogether. Examples
of anti-P1 that react only at temperatures below 37°C can be Paragloboside P antigen
considered clinically insignificant. (type 2 precursor) Globoside (Gb4)
Because P1 antigen expression on RBCs varies and dete- FUT1
A4GALT
riorates during storage, antibodies may react only with RBCs A4GALT B3GALNT1
that have the strongest expression and give inconclusive pat- H antigen
P1 antigen
terns of reactivity when antibody identification is performed. NOR
When anti-P1 is suspected, incubating tests at room temper- PX2
ature or lower or pretreating test cells with enzymes can en-
hance reactions to confirm specificity. Providing units that LKE
are crossmatch compatible at 37°C and the antiglobulin Figure 8–3. Biosynthetic pathways of the P blood group antigens.
6888_Ch08_173-211 29/10/18 5:27 PM Page 184

184 PART II Blood Groups and Serologic Testing

cloned in 2000.15 Several mutations in both genes have been alloantibody in the sera of Pk individuals. Its reactivity is
identified that result in the p and Pk phenotypes. A polymor- similar to that of anti-PP1Pk in that it is usually a potent
phism in the Pk synthase was recently identified that ties the hemolysin reacting with all cells except the autocontrol and
P1 and Pk antigens together at the genetic level; conse- those with the p phenotype. However, it differs from anti-
quently, the P system (003), to which the P1 antigen was PP1Pk in that it does not react with cells that have the
assigned, was renamed P1PK.4 extremely rare Pk phenotype, and the individual making the
The P1PK (A4GALT) gene is located on chromosome 22 antibody may type P1+. Alloanti-P is rarely seen, but because
at position 22q13.2 and the Globoside (B3GALT1) gene is it is hemolytic with a wide thermal range of reactivity, it is
located on chromosome 3 at position 3q25, and they are very significant in transfusion. IgG class alloanti-P may occur
inherited independently.4 The gene for the synthesis of LKE and has been associated with habitual early abortion.
has not yet been cloned.
Autoanti-P Associated With Paroxysmal Cold
Other Sources of P1 Antigen and Antibody Hemoglobinuria

The discovery of strong anti-P1 in two P1– individuals in- Anti-P specificity is also associated with the cold-reactive
fected with Echinococcus granulosus tapeworms led to the IgG autoantibody in patients with paroxysmal cold hemo-
identification of P1 and Pk substance in hydatid cyst fluid. globinuria (PCH). Historically, this rare autoimmune disor-
This fluid was subsequently used in many of the studies der was seen in patients with tertiary syphilis; it now more
that identified the biochemical structures of the P blood commonly presents as a transient, acute condition second-
group. Strong antibodies to P1 have also been found in ary to viral infection, especially in young children. The IgG
patients with fascioliasis (bovine liver fluke disease) and in autoantibody in PCH is described as a biphasic hemolysin:
bird handlers. in vitro, the antibody binds to RBCs in the cold, and, via
Soluble P1 substances have potential use in the blood bank complement activation, the coated RBCs lyse as they are
and are commercially available. When it is necessary to con- warmed to 37°C. The autoantibody typically does not react
firm antibody specificity or to identify underlying antibodies, in routine test systems but is demonstrable only by the
these substances can be used to neutralize anti-P1. Donath-Landsteiner test. The etiology and diagnosis of PCH
are more fully discussed in Chapter 21, “Autoimmune
Anti-PP1Pk Hemolytic Anemias.”

Originally called anti-Tja, anti-PP1Pk was first described in Antibodies to Compound Antigens
the serum of Mrs. Jay, a p individual with adenocarcinoma
of the stomach.11 Her tumor cells carried P system antigens, Considering the biochemical relationship of the P blood
and the antibody was credited as having cytotoxic properties group antigens to ABH and I, it is not surprising that anti-
that may have helped prevent metastatic growth postsurgery bodies requiring more than one antigenic determinant have
(the T in the Tja refers to tumor). been described, including anti-IP1, -iP1, -ITP1, and -IP. Most
Anti-PP1Pk is produced by p individuals early in life examples are cold-reactive agglutinins.
without RBC sensitization and reacts with all RBCs except
those of the p phenotype. Unlike antibodies made by other Luke (LKE) Antigen
blood group null phenotypes, the anti-P, anti-P1, and anti-
Pk components of anti-PP1Pk are separable through adsorp- In 1965, Tippett and colleagues17 described an antibody in
tion.8 Components of anti-PP1Pk have been shown to be the serum of a patient with Hodgkin’s lymphoma that
IgM and IgG.8 They react over a wide thermal range and divided the population into three phenotypes: 84% tested
efficiently bind complement, which makes them potent Luke+, 14% were weakly positive or Luke(w), and 2% were
hemolysins. Anti-PP1Pk has the potential to cause severe Luke–. LKE has also been associated with metastasis in renal
HTRs and HDFN. cell carcinoma.10 The LKE antigen has a frequency of 98%
The antibody is also associated with an increased inci- in all populations and is expressed on cord cells. The antigen
dence of spontaneous abortions in early pregnancy. Al- is resistant to enzyme and chemical treatment with enzyme
though the reason for this is not fully known, it has been treatment showing a markedly enhanced expression. De-
suggested that having an IgG anti-P component is an im- creased expression of the LKE is seen in individuals with the
portant factor. Women with anti-P and anti-PP1Pk and a Se gene with secretors having a 3- to 4-fold decreased risk
history of multiple abortions have successfully delivered of E.coli infections.10 Although this Mendelian-dominant
infants after multiple plasmaphereses to reduce their anti- character segregated independently of the P blood group, it
body level during pregnancy.16 was thought to be phenotypically related because the anti-
body reacted with all RBCs except 2% of P1 and P2 pheno-
Alloanti-P types and those having the rare p and Pk phenotypes. All
individuals with the p and Pk phenotype are Luke–. Anti-
In addition to being a component of the anti-PP1Pk in p in- bodies to the LKE antigen are rare. They are IgMs reacting
dividuals (see above), anti-P is found as a naturally occurring at room temperature or below with some antibodies having
6888_Ch08_173-211 29/10/18 5:27 PM Page 185

Chapter 8 Blood Group Terminology and Common Blood Groups 185

the ability to bind complement. Anti-LKE has no reported provide the distinction between the I (027001) and i
transfusion reactions and does not cause HDFN. (207002) antigens.

PX2 Antigen The I and i Antigens

The PX2 antigen was added to the GLOB collection in 2010 The antigens are best introduced by classic serologic facts.
after antibodies from Pk individuals were shown to react with Both I and i are high-prevalence antigens, but they are ex-
the p phenotype (PP1Pk–). PX2 was then reassigned to the pressed in a reciprocal relationship that is developmentally
GLOB blood group system (028) in 2016 as GLOB2 when it regulated. At birth, infant RBCs are rich in i; I is almost un-
was determined to be a product of B3GALNT1.18 The PX2 detectable. During the first 18 months of life, the quantity
antigen has a frequency of >99.9% in all population and is of i slowly decreases as I increases until adult proportions
expressed on cord cells.10 Enzyme treatment enhances the are reached; adult RBCs are rich in I and have only trace
expression of the PX2 antigen. Antibodies to the PX2 antigen amounts of i antigen.
appear to be naturally occurring and primarily IgG with a There is no true I– or i– phenotype. The strength of I and
possible mixture of IgM that react best at IAT on papain- i varies from individual to individual, and the relative amount
treated RBCs. There is no data concerning the clinical sig- detected will depend on the example of anti-I or anti-i used.
nificance of anti-PX2 with transfusion or HDFN. Data suggest that i reactivity on RBCs is inversely propor-
tional to marrow transit time and RBC age in circulation.
Disease Associations Some people appear not to change their i status after birth.
They become the rare i adult. RBCs of i adult generally express
Several pathological conditions associated with the P blood more i antigen than do cord RBCs. A spectrum of Ii phenotypes
group antigens have been described: parasitic infections are and their characteristic reactivity are shown in Table 8–11.
associated with anti-P1, early abortions with anti-PP1Pk or Treatment of RBCs with ficin and papain enhances reac-
anti-P, and PCH with autoanti-P. The P system antigens also tivity of the I and i antigens with their respective antibodies.
serve as receptors for P-fimbriated uropathogenic E. coli—a The I and i antigens are resistant to treatment with DTT and
cause of urinary tract infections. The Pk antigen is a receptor glycine-acid EDTA.
for Shiga toxins, which cause Shigella dysentery and E. coli–
associated hemolytic uremic syndrome. In addition, P is the Anti-I
receptor of human parvovirus B19. Recent studies demon-
strate that Pk provides some protection against HIV infection Anti-I is a common autoantibody that can be found in virtu-
of peripheral blood mononuclear cells.19 ally all sera, although testing at 4°C and/or against enzyme-
treated RBCs may be required to detect the reactivity.1
The I (027) System and i Antigen Consistently strong agglutination with adult RBCs and weak
or no agglutination with cord or adult i RBCs define its
The existence of cold agglutinins in the serum of normal in- classic activity (see Table 8–11).
dividuals and in patients with acquired hemolytic anemia has Autoanti-I, found in the serum of many normal healthy
long been recognized. In 1956, Wiener and coworkers2,8 gave individuals, is benign—that is, not associated with in vivo
a name to one such agglutinin, calling its antigen I for “indi- RBC destruction. It is usually a weak, naturally occurring,
viduality.” The antibody reacted with most blood specimens saline-reactive IgM agglutinin with a titer less than 64
tested. The few nonreactive I– specimens were thought to be at 4°C. Stronger examples agglutinate test cells at room
from homozygotes for a rare gene producing the “i” antigen; temperature and bind complement, which can be detected
the I– phenotype in adults was called adult i. In 1960, Marsh in the antiglobulin test if polyspecific reagents are used.
and Jenkins20 reported finding anti-i, and the unique relation- Some examples may react only with the strongest I+ RBCs
ship between I and i began to unfold. According to ISBT, the and give inconsistent reactions with panel RBCs.
correct designation is “I adult and i adult.” Incubating tests in the cold enhances anti-I reactivity and
I and i are not antithetical antigens. Rather, they are helps confirm its identity; albumin and enzyme methods also
branched and linear carbohydrate structures, respectively, enhance anti-I reactivity. Testing enzyme-treated RBCs with
that are formed by the action of glycosyl transferases. The
gene encoding the transferase that converts i active straight
chains into I active branched chains has been cloned, and Table 8–11 I and i Antigens
several mutations responsible for the rare adult i phenotype Phenotype Strength of Reactivity With:
have been identified.21 The synthesis of i antigen is not con-
trolled by this same gene. Consequently, I has been raised to Anti-I Anti-i Anti-IT
blood group system status (system number 027, symbol I) I adult Strong Weak Weak
and the i antigen remains in the Ii collection (collection
number 207, symbol I). The reader will notice the confusing Cord Weak Strong Strong
use of the same ISBT symbol, capital I, for both the I system i adult Weak Strong Weakest
and Ii collection; in ISBT terminology, the antigen numbers
6888_Ch08_173-211 29/10/18 5:27 PM Page 186

186 PART II Blood Groups and Serologic Testing

slightly acidified serum may even promote hemolysis. Occa- test cells (except cord RBCs) have poor i expression (see
sionally, benign cold autoanti-I can cause problems in pre- Table 8–11).
transfusion testing. Usually, avoiding room temperature Unlike anti-I, autoanti-i is not seen as a common antibody
testing and using anti-IgG instead of a polyspecific antihu- in healthy individuals. Potent examples are associated with
man globulin help to eliminate detection of cold-reactive an- infectious mononucleosis (Epstein-Barr virus infections) and
tibodies that may bind complement at lower temperatures. some lymphoproliferative disorders. High-titer autoantibod-
Cold autoadsorption to remove the autoantibody from the ies with a wide thermal range may contribute to hemolysis,
serum may be necessary for stronger examples; cold autoad- but because i expression is generally weak, they seldom
sorbed plasma or serum can also be used in ABO typing. cause significant hemolysis. IgG anti-i has also been de-
Pathogenic autoanti-I (e.g., the type associated with cold scribed and has been associated with HDFN.1
agglutinin disease) typically consists of strong IgM agglu-
tinins with higher titer and a broad thermal range of activity, Biochemistry and Genetics
reacting up to 30°C. When peripheral circulation cools in
response to low ambient temperatures, these antibodies
attach in vivo and cause autoagglutination and peripheral Advanced Concepts
vascular occlusion (acrocyanosis) or hemolytic anemia. An early association of I and i to ABH was demonstrated by
Refer to Chapter 21 for more information. complex antibodies involving both ABH and Ii specificity
Pathogenic anti-I typically reacts with adult and cord RBCs (see the “Antibodies to Compound Antigens” section). I and
equally well at room temperature and at 4°C, and antibody i antigens are precursors for the synthesis of ABO and Lewis
specificity may not be apparent unless the serum is diluted or antigens, and thus they are internal structures on these
warmed to 30°C or 37°C. Potent cold autoantibodies can also oligosaccharide chains.
mask clinically significant underlying alloantibodies and can ABH and Ii determinants on the RBC membrane are
complicate pretransfusion testing. Procedures to deal with carried on type 2 chains that attach either to proteins or to
these problems are discussed in Chapters 11 and 21. lipids. See Figure 8–4 for examples of glycolipid structures
The production of autoanti-I may be stimulated by mi- for i and I antigens. The i antigen activity is defined by at least
croorganisms carrying I-like antigen on their surface. Patients two repeating N-acetyllactosamine [Gal(β1-4)GlcNAc(β1-3)]
with M. pneumoniae often develop strong cold agglutinins with units in linear form. I antigen activity is associated with a
I specificity and can experience a transient episode of acute branched form of i antigen. The IGnT (also known as
abrupt hemolysis just as the infection begins to resolve. Al- GCNT2) gene on chromosome 6 at position 6p24.2 encodes
loanti-I exists as an IgM or IgG antibody in the serum of most the N-acetylglucosaminyltransferase, which adds GlcNAc to
individuals with the i adult phenotype. Although i adult RBCs form the branches.4,21,22
are not totally devoid of I, the anti-I in these cases does not In summary, fetal, cord, and i adult RBCs carry predomi-
react with autologous RBCs. It has been traditional to trans- nantly unbranched chains and have the i phenotype. Normal
fuse compatible i adult units to these people, although such adult cells have more branched structures and express I anti-
practice may be unnecessary, especially when the antibody is gen. The gene responsible for I antigen (IGnT) codes for the
not reactive at 37°C.1 Technologists must be aware that strong branching enzyme. Family studies show that the i adult phe-
autoanti-I can mimic alloanti-I: if enough autoantibody and notype is recessive. Heterozygotes (e.g., children inheriting
complement are bound to a patient’s RBCs, blocking the anti- I from one parent and i from the other parent) have inter-
genic sites, they may falsely type I-negative. mediate I antigen expression. Several gene mutations have
Anti-I is not associated with HDFN because the antibody been identified that result in the i adult phenotype.
is IgM, and the I antigen is poorly expressed on infant RBCs.

Anti-i Other Sources of I and i Antigen

Alloanti-i has never been described. Autoanti-i is a fairly rare I and i antigens are found on the membranes of leukocytes
antibody that gives strong reactions with cord RBCs and and platelets in addition to RBCs. It is quite likely that the
i adult RBCs and weaker reactions with I adult RBCs. Most antigens exist on other tissue cells, much like ABH, but this
examples of autoanti-i are IgM and react best with saline- has not been confirmed.
suspended cells at 4°C. Only very strong examples of I and i have also been found in the plasma and serum of
autoanti-i are detected in routine testing because standard adults and newborns and in saliva, human milk, amniotic

Antigen Structure Figure 8–4. The linear and branched structures carrying
i and I activity.
(none) Gal(␤1-4)Glc-Cer
i Gal(␤1-4)GlcNAc(␤1-3)Gal(␤1-4)GlcNAc(␤1-3)Gal(␤1-4)Glc-Cer
Gal(␤1-4)GlcNAc(␤1-3)
I Gal(␤1-4)GlcNAc(␤1-3)Gal(␤1-4)Glc-Cer
Gal(␤1-4)GlcNAc(␤1-6)
6888_Ch08_173-211 29/10/18 5:27 PM Page 187

Chapter 8 Blood Group Terminology and Common Blood Groups 187

fluid, urine, and ovarian cyst fluid. The antigens in secretions Disease Associations
do not correlate with RBC expression and are thought to de-
velop under separate genetic control. For example, the quan- Well-known associations between strong autoantibodies and
tity of I antigen in the saliva of i adult individuals and disease or microorganisms have already been discussed: anti-
newborns is quite high. I with cold agglutinin disease and M. pneumoniae, and anti-
i with infectious mononucleosis.
The IT Antigen and Antibody Diseases can also alter the expression of I and i antigens
on RBCs. Conditions associated with increased i antigen on
In 1965, Curtain and coworkers23 reported a cold agglutinin in RBCs include those with shortened marrow maturation time
Melanesians that did not demonstrate classical I or i specificity. or dyserythropoiesis: acute leukemia, hypoplastic anemia,
In 1966, Booth and colleagues24 confirmed these observations megaloblastic anemia, sideroblastic anemia, thalassemia,
and carefully described the agglutinin’s reactivity. This agglu- sickle cell disease, paroxysmal nocturnal hemoglobinuria
tinin reacted strongly with cord RBCs, weakly with normal (PNH), and chronic hemolytic anemia.1,8 Except in some
adult RBCs, and most weakly with i adult RBCs. They con- cases of leukemia, the increase in i on RBCs is not usually
cluded that the agglutinin recognizes a transition state of associated with a decrease in I antigen; the expression of I anti-
i into I and designated the specificity IT (T for “transition”). gen can appear normal or sometimes enhanced. Chronic
However, detection of IT on fetal RBCs ranging in age dyserythropoietic anemia type II or hereditary erythroblastic
from 11 to 16 weeks does not support this hypothesis.25 This multinuclearity with a positive acidified serum test (HEMPAS)
benign IgM anti-IT was frequently found in two populations: is associated with much greater i activity on RBCs than control
Melanesians and the Yanomami Indians in Venezuela. cord RBCs. HEMPAS RBCs are very susceptible to lysis with
Whether it is associated with an organism or parasite in these both anti-i and anti-I, and lysis by anti-I appears to be the
regions is unknown. Examples of IgM and IgG anti-IT react- result of increased antibody uptake and increased sensitivity
ing preferentially at 37°C have also been found in patients to complement.1 In Asians, the i adult phenotype has been
with warm autoimmune hemolytic anemia, with a special associated with congenital cataracts.22
association with Hodgkin’s disease.25
The MNS (002) System
Antibodies to Compound Antigens
Following the discovery of the ABO blood group system,
Many other I-related antibodies have been described: anti-IA, Landsteiner and Levine began immunizing rabbits with
-IB, -IAB, -IH, -iH, -IP1, -ITP1, -IHLeb, and -iHLeb. Bearing in human RBCs, hoping to find new antigen specificities.
mind the close relationship of I to the biochemical structures Among the antibodies recovered from these rabbit sera were
of ABH, Lewis, and P antigens, it is not surprising to find an- anti-M and anti-N, both of which were reported in 1927.8
tibodies that recognize compound antigens. These specifici- Data from family studies suggested that M and N were anti-
ties are not mixtures of separable antibodies; rather, both thetical antigens. In 1947, after the implementation of the
antigens must be present on the RBCs for the antibody to antiglobulin test, Walsh and Montgomery discovered S,
react. For example, anti-IA reacts with RBCs that carry both a distinct antigen that appeared to be genetically linked to
I and A but will not react with group O, I+, or group A i adult M and N. Its antithetical partner, s, was discovered in 1951.
RBCs. (Table 8–12 summarizes some common cold autoan- Family studies (and later, molecular genetics) demonstrated
tibodies.) Anti-IH is commonly encountered in the serum of the close linkage between the genes controlling M, N,
group A1 individuals. Anti-IH reacts stronger with group O and S, s antigens. There is a disequilibrium in the expression
and group A2 RBCs than with group A1 RBCs. Anti-IH should of S and s with M and N. In whites, the common haplotypes
be suspected when serum from a group A individual directly were calculated to appear in the following order of relative
agglutinates all group O RBCs but is compatible with most frequency: Ns > Ms > MS > NS.1,7 The prevalence of the com-
group A donor units. mon MN and Ss phenotypes are listed in Table 8–13.

Table 8–12 Typical Reactions of Some Cold Autoantibodies*


Antibody A1 Adult A2 Adult B Adult O Adult O Cord A Cord Oh Adult O i Adult
Anti-I ++++ ++++ ++++ ++++ 0/+ 0/+ ++++ (0)

Anti-i 0/+ 0/+ 0/+ 0/+ ++++ ++++ 0/+ ++++

Anti-H 0/+ ++ +++ ++++ +++ 0/+ (0) +++

Anti-IH 0/+ ++ +++ ++++ 0/+ 0/+ (0) (0)

Anti-IA ++++ +++ 0/+ 0/+ 0/+ 0/+ (0) (0)

*Reactions vary with antibody strength; very potent examples may need to be diluted before specificity can be determined.
0 = negative; + = positive
6888_Ch08_173-211 29/10/18 5:27 PM Page 188

188 PART II Blood Groups and Serologic Testing

Table 8–13 Prevalence of Common MN In 1953, an antibody to a high-prevalence antigen, U


(for almost universal distribution), was named by Weiner.
and Ss Phenotypes
The observation by Greenwalt and colleagues26 that all
Phenotype Whites (%) Blacks (%) U– RBCs were also S–s– resulted in the inclusion of U into
M+N– 28 26 the system.
Forty-nine antigens have been included in the MNS sys-
M+N+ 50 44 tem, making it almost equal to Rh in size and complexity
M–N+ 22 30 (Table 8–14). Most of these antigens are of low prevalence
and were discovered in cases of HDFN or incompatible
S+s– 11 3 crossmatch. Others are high-prevalence antigens. Antibodies
S+s+ 44 28 to these low- and high-prevalence antigens are not com-
monly encountered in the blood bank. The genes encoding
S–s+ 45 69 the MNS antigens are located on chromosome 4.
S–s–U– 0 <1 The MNS blood group system has been assigned the ISBT
number 002 (symbol MNS), second after ABO.

Table 8–14 Summary of MNS Antigens


Antigen Name ISBT Number Prevalence (%) Year Discovered
M MNS1 Polymorphic 1927

N MNS2 Polymorphic 1927

S MNS3 Polymorphic 1947

s MNS4 Polymorphic 1951

U MNS5 High 1953

He MNS6 Low 1951

Mia MNS7 Low 1951

Mc MNS8 Low 1953

Vw MNS9 Low 1954

Mur MNS10 Low 1961

Mg MNS11 Low 1958

Vr MNS12 Low 1958

Me MNS13 Low 1961

Mta MNS14 Low 1962

Sta MNS15 Low 1962

Ria MNS16 Low 1962

Cla MNS17 Low 1963

Nya MNS18 Low 1964

Hut MNS19 Low 1966

Hil MNS20 Low 1966

Mv MNS21 Low 1966

Far MNS22 Low 1968

sD MNS23 Low 1978

Mit MNS24 Low 1980

Dantu MNS25 Low 1982


6888_Ch08_173-211 29/10/18 5:27 PM Page 189

Chapter 8 Blood Group Terminology and Common Blood Groups 189

Table 8–14 Summary of MNS Antigens—cont’d


Antigen Name ISBT Number Prevalence (%) Year Discovered
Hop MNS26 Low 1977

Nob MNS27 Low 1977

Ena MNS28 High 1969

ENKT MNS29 High 1986

‘N’ MNS30 High 1977

Or MNS31 Low 1964

DANE MNS32 Low 1991

TSEN MNS33 Low 1992

MINY MNS34 Low 1992

MUT MNS35 Low 1992

SAT MNS36 Low 1991

ERIK MNS37 Low 1993

Osa MNS38 Low 1983

ENEP MNS39 High 1995

ENEH MNS40 High 1993

HAG MNS41 Low 1995

ENAV MNS42 High 1996

MARS MNS43 Low 1992

ENDA MNS44 High 2005

ENEV MNS45 High 2006

MNTD MNS46 Low 2006

SARA MNS47 Low 2016

KIPP MNS48 Low 2016

JENU MNS49 High 2016

M and N Antigens ZZAP, a combination of DTT and papain or ficin, but they
are not affected by DTT alone, 2-aminoethyliso-thiouronium
The M and N antigens are found on a well-characterized bromide (AET), α-chymotrypsin, chloroquine, or glycine-
glycoprotein called glycophorin A (GPA), the major RBC acid EDTA treatment. Treating RBCs with neuraminidase,
sialic acid–rich glycoprotein (sialoglycoprotein, SGP). The which cleaves sialic acid (also known as neuraminic acid or
M and N antigens are antithetical and differ in their amino NeuNAc), abolishes reactivity with only some examples of
acid residues at positions 1 and 5 (Fig. 8–5). M is defined by antibody. M and N antibodies are heterogeneous; some may
serine at position 1 and glycine at position 5; N has leucine recognize only specific amino acids, but others recognize
and glutamic acid at these positions, respectively. The anti- both amino acids and carbohydrate chains.
body reactivity may also be dependent on adjacent carbohy-
drate chains, which are rich in sialic acid. There are about S and s Antigens
106 copies of GPA per RBC.14 The antigens are well devel-
oped at birth. S and s antigens are located on a smaller glycoprotein called
Because M and N are located at the outer end of GPA, they glycophorin B (GPB) that is very similar to GPA (see the
are easily destroyed by the routine blood bank enzymes ficin, next “Biochemistry” section and Fig. 8–5). S and s are dif-
papain, and bromelin and by the less common enzymes ferentiated by the amino acid at position 29 on GPB. Methio-
trypsin and pronase. The antigens are also destroyed by nine defines S, whereas threonine defines s. The epitope may
6888_Ch08_173-211 29/10/18 5:27 PM Page 190

190 PART II Blood Groups and Serologic Testing

NH react with M+ reagent RBCs or donor RBCs stored in preser-


M N
1 Ser 1 Leu
1 vative solutions containing glucose but do not react with
2 Ser 2 Ser freshly collected M+ RBCs.
3 Thr 3 Thr NH2 As long as anti-M does not react at 37°C, it is not clinically
4 Thr 4 Thr
5 Gly 5 Glu ‘N’ 1 significant for transfusion and can be ignored. When anti-M
29 S/s ⫽ Met/Thr reacts at 37°C, it is sufficient to provide units that are cross-
U⫽[ 39
match compatible at 37°C and at the antiglobulin phase
72
without typing for M antigen. Anti-M rarely causes HTRs,
decreased red blood cell survival, or HDFN.
Glu ⫽ glutamic acid
Gly ⫽ glycine 96 72
Leu ⫽ leucine Anti-N
Met ⫽ methionine
Ser ⫽ serine 131 The serologic characteristics of the common anti-N (made
Thr ⫽ threonine GPA GPB by individuals whose RBCs type M+N – and S+ or s+) are
⫽ N-linked glycan similar to those of anti-M: a cold-reactive IgM or IgG saline
agglutinin that does not bind complement or react with
Figure 8–5. Comparison of glycophorin A (GPA) and glycophorin B (GPB).
enzyme-treated RBCs. Anti-N can demonstrate dosage, re-
acting better with M–N+ RBCs than with M+N+ RBCs. Rare
also include the amino acid residues at position 34 and examples are pH- or glucose-dependent.
35 and the carbohydrate chain attached to threonine at Also like anti-M, anti-N is not clinically significant unless
position 25.8 There are fewer copies of GPB (about 200,000) it reacts at 37°C. It has been implicated only with rare cases
than GPA per RBC.8 In addition, there are about 1.5 times of mild HDFN.1 Anti-N is less common than anti-M. A
more copies of GPB on S+s– RBCs than on S–s+ RBCs.1 S and potent anti-N can be made by the rare individual whose
s also are well developed at birth. RBCs type M+N–S–s– because they lack both N and GPB
S and s antigens are less easily degraded by enzymes that has ‘N’ activity (see the next “Biochemistry” section).
because the antigens are located farther down the glycopro- Anti-N was also seen in renal patients, regardless of their
tein, and enzyme-sensitive sites are less accessible. Ficin, MN type, who were dialyzed on equipment sterilized with
papain, bromelin, pronase, and α-chymotrypsin can destroy formaldehyde. Dialysis-associated anti-N reacts with any N+
S and s activity, but the amount of degradation may depend or N– RBCs treated with formaldehyde and is called anti-Nf.
on the strength of the enzyme solution, the length of treat- Formaldehyde may alter the M and N antigens so that they
ment, and the enzyme-to-cell ratio. Trypsin does not destroy are recognized as foreign. The antibody titer decreases when
the S and s antigens, and neither does DTT, AET, chloro- dialysis treatment and exposure to formaldehyde stops.
quine, or glycine-acid EDTA treatment. Because anti-Nf does not react at 37°C, it is clinically in-
significant for transfusion.
Anti-M
Anti-S and Anti-s
Many examples of anti-M are naturally occurring saline
agglutinins that react below 37°C. Although we may think Most examples of anti-S and anti-s are IgG, reactive at 37°C
of agglutinating anti-M as IgM, 50% to 80% are IgG or have and the antiglobulin phase of testing. A few express optimal
an IgG component.1 They do not bind complement, regard- reactivity between 10°C and 22°C by saline indirect antiglob-
less of their immunoglobulin class, and they do not react ulin test. If anti-S or anti-s specificity is suspected but the
with enzyme-treated RBCs. Anti-M appears to be more com- pattern of reactivity is not clear, incubating tests at room
mon in children than in adults and is particularly common temperature and immediately performing the antiglobulin
in patients with bacterial infections.8 test (without incubating at 37°C) may help in identification.
Because of antigen dosage, many examples of anti-M may Dosage effect can be exhibited by many examples of anti-S
react better with M+N– RBCs (genotype MM) than with and anti-s, although it may not be as dramatic as seen with
M+N+ RBCs (genotype MN). Very weak anti-M may not anti-M and anti-N.1
react with M+N+ RBCs at all, making antibody identification The antibodies may or may not react with enzyme-treated
difficult. Antibody reactivity can be enhanced by increasing RBCs, depending on the extent of treatment and the efficiency
the serum-to-cell ratio or incubation time, or both, by de- of the enzyme. Although seen less often than anti-M, anti-S
creasing incubation temperature or by adding a potentiating and anti-s are more likely to be clinically significant. They may
medium such as albumin, low-ionic strength saline solution bind complement, and they have been implicated in severe
(LISS), or polyethylene glycol (PEG). HTRs with hemoglobinuria. They have also caused HDFN.
Some examples of anti-M are pH-dependent, reacting best Units selected for transfusion must be antigen negative and
at pH 6.5. These antibodies may be detected in plasma crossmatch compatible. Because only 11% of whites and 3% of
(which is slightly acidic from the anticoagulant) but not in blacks are s–, it can be difficult to provide blood for a patient
unacidified serum.1 Other examples of anti-M react only with anti-s. S– units are much easier to find (45% of whites
with RBCs exposed to glucose solutions. Such antibodies and 69% of blacks are S–). Antibodies to low-prevalence
6888_Ch08_173-211 29/10/18 5:27 PM Page 191

Chapter 8 Blood Group Terminology and Common Blood Groups 191

antigens are commonly found in reagent anti-S; these can section). GPA is restriced to erythroid cells. GPA is expressed
cause discrepant antigen typing results. on renal endothelium and epithelium, but only as an incom-
pletely sialylated glycophorin .10
Biochemistry

Genetics
Advanced Concepts
GPA, the structure carrying the M and N antigens, is a The genes GYPA and GYPB, which code for GPA and GPB,
single-pass membrane SGP that consists of 131 amino acids. respectively, are located on chromosome 4 at position
The hydrophilic NH2 terminal end, which lies outside 4q31.21.4 The known alleles for GYPA (M/N) and GYPB (S/s)
the RBC membrane, has 72 amino acid residues, 15 O- are codominant. The genes are highly homologous (meaning
glycosidically linked oligosaccharide chains (GalNAc-serine/ they are very similar) and probably arose by gene duplica-
threonine), and 1 N-glycosidic chain (sugar-asparagine). The tion. GYPA is considered to be the ancestral gene.27
portion that traverses the membrane is hydrophobic and GYPA is organized into seven exons. GYPB has a size and
contains 23 amino acids. The hydrophilic COOH end, which arrangement similar to those of GYPA but has only five
contains 36 amino acids and no carbohydrates, lies inside coding exons plus one noncoding or pseudoexon. A third,
the membrane and interacts weakly with the membrane highly homologous gene, GYPE, does not appear to make a
cytoskeleton. glycoprotein that has been definitively recognized on the
GPB, the structure carrying the S, s, and U antigens, con- RBC surface, but it participates in gene rearrangements that
sists of 72 amino acids and 11 O-linked oligosaccharide result in variant alleles.
chains and no N-glycans. It has an outer glycosylated por- Misalignment of GYPA and GYPB during meiosis, fol-
tion of 44 amino acids, a hydrophobic portion of 20 amino lowed by an unequal crossing over, appears to explain some
acids that traverses the RBC membrane, and a short cyto- of the variant glycophorins observed in the MNS system. The
plasmic “tail” of 8 amino acids. resulting new reciprocal GYP(A-B) and GYP(B-A) genes
The first 26 amino acids on GPB are identical to the first encode GP(A-B) and GP(B-A) hybrid glycophorins, respec-
26 amino acids on the N form of GPA (GPAN). This N activity tively (Fig. 8–6).8,28 The point of fusion between the GPA
of GPB is denoted as ‘N’ (N-quotes) to distinguish it from the and GPB part in the hybrid glycophorin can give rise to novel
N activity of GPAN. Anti-N reagents are formulated to not rec- antigens (e.g., antigen of low prevalence).
ognize the weak reactivity of the ‘N’ structure. The U antigen, For more complex variant glycophorins, a gene conversion
expressed when normal GPB is present, is located very close event probably occurs. Gene conversion involves a nonrecip-
to the RBC membrane (see the “U– Phenotype” section). rocal exchange of genetic material from one gene to another
Most O-linked carbohydrate structures on GPA and GPB homologous gene (Fig. 8–7).8,28 As with hybrids resulting
are branched tetrasaccharides containing one GalNAc, one from crossing over, the new amino acid sequence at the junc-
Gal, and two NeuNAc (sialic acid). NeuNAc helps give the tion of the hybrid glycophorin can give rise to novel antigens
RBC its negative charge. About 70% of the RBC NeuNAc is of low prevalence. Also, expected antigens may be missing if
carried by GPA, and about 16% is carried by GPB. the coding exons are replaced by the inserted genetic material.
Both GPA and GPB appear to be associated with the
band 3/Rh macrocomplex that is linked to the spectrin ma- GPA- and GPB-Deficient Phenotypes
trix of the cytoskeleton via protein 4.2 and ankyrin. GPA
RBCs of three rare phenotypes lack GPA or GPB or both GPA
is required for the expression of the antigen Wrb of the
and GPB; consequently, they lack all MNS antigens that are
Diego blood group system (located on protein band 3). The
normally expressed on those structures.
association of GPB with the Rh protein and Rh-associated
glycoprotein complex is evidenced by the greatly reduced
S and s expression on Rhnull RBCs.8
GYPA GYPB
Other antigens within the MNS system have been eval-
uated biochemically and at the molecular level. Some are
associated with altered GPA because of amino acid substi- Misaligned
tutions or changes in carbohydrate chains. Others are
expressed on variants of GPA or GPB. Still others result GYPA GYPB
from a genetic event that encodes a hybrid glycophorin that
has parts of both GPA and GPB. The altered glycophorins
are associated with changes in glycosylation, changes in GYPA GYP(B-A) GYPB
molecular weight, loss of high-prevalence antigens or the
appearance of novel low-prevalence antigens, or alterations
in the expression of MNS antigens.1,8,10
GYP(A-B)
RBCs that lack GPA or GPB are not associated with disease
or decreased RBC survival (see the “Disease Associations” Figure 8–6. Depiction of how misalignment during meiosis can lead to a
crossover and reciprocal GYP(B-A) and GYP(A–B) hybrids.
6888_Ch08_173-211 29/10/18 5:27 PM Page 192

192 PART II Blood Groups and Serologic Testing

GYPA GYPB portion. Anti-EnaTS recognizes a trypsin-sensitive (TS)


area on GPA between amino acids 20 and 39, anti-EnaFS
Misaligned reacts with a ficin-sensitive (FS) area between amino acids
46 and 56, and anti-EnaFR reacts with a ficin-resistant (FR)
area around amino acids 62 to 72.1,8
GYPA GYPB Although the gene responsible for this phenotype has been
DNA repair termed En, it is now known that the En(a–) phenotype has
more than one origin. Most often, the En(a–) phenotype
GYPA GYP(B-A-B) results from homozygosity for a rare gene deletion at the
GYPA locus; consequently, no GPA is produced, but GPB
is not affected. This type of En(a–) inheritance is called
GYPA GYPB En(a–)Fin, representing the type described in the Finnish
report.31 However, the En(a–) phenotype in the English
Figure 8–7. In a gene conversion event, nucleotides from one strand of DNA
are transferred to the misaligned homologous gene. If this is the coding strand, a report30 probably represents heterozygosity for a hybrid gene
hybrid glycophorin will be formed; the partner chromosome will be repaired and along with the very rare Mk gene; this type is often called
carry the naïve (unaltered) GYPA and GYPB genes. En(a–)UK. Several other En(a–) phenotypes have been re-
ported that result from homozygosity for a variant allele or
inheritance of two dissimilar variant alleles.
U– Phenotype
Anti-Ena can be confused with two other specificities that
The U antigen is located on GPB very close to the RBC mem- react with all normal RBCs—anti-Pr and anti-Wrb. Anti-Pr
brane between amino acids 33 and 39 (see Fig. 8–5). This does not react with enzyme-treated RBCs and can be con-
high-prevalence antigen is found on RBCs of all individuals fused with anti-EnaFS; anti-Wrb does not react with En(a–)
except about 1% of African Americans (and 1% to 35% of RBCs but does react with enzyme-treated RBCs and can be
Africans) who lack GPB because of a partial or complete confused with anti-EnaFR. Anti-Ena has caused severe HTRs
deletion of GYPB. The RBCs usually type S–s–U–, and these and HDFN. It is extremely difficult and may be impossible
individuals can make anti-U in response to transfusion or to find units compatible for patients with anti-Ena; siblings
pregnancy. Anti-U is typically IgG and has been reported to are a potential source of compatible blood if they are also
cause severe and fatal HTRs and HDFN. ABO and Rh compatible.
The U antigen is resistant to enzyme treatment; thus,
most examples of anti-U react equally well with untreated Mk Phenotype
and enzyme-treated RBCs. However, there are rare examples The rare silent gene Mk was named by Metaxas and Metaxas-
of broadly reactive anti-U that do not react with papain- Buhler in 1964 when they found an allele that did not
treated RBCs.1 produce M or N.8 A second family showed that Mk was
Some examples of anti-U react with apparent U– RBCs, also silent at the Ss locus. Several other Mk heterozygotes
although weakly, by adsorption and elution.8 Such RBCs are have been found. In 1979, two related MkMk blood donors
said to be U variant (Uvar); these have an altered GPB that were found in Japan. The RBCs of these individuals typed
does not express S or s. There is a strong correlation between M–N–S–s–U–En(a–)Wr(a–b–), but they had a normal hema-
the low-prevalence antigen He, found in about 3% of African tologic picture. More individuals have since been identified,
Americans, and Uvar expression.29 and it is now known that the Mk gene represents a single,
Because examples of anti-U are heterogeneous, U– units near-complete deletion of both GYPA and GYPB8; thus,
selected for transfusion must be crossmatched to determine MkMk is the null phenotype in the MNS system. The MkMk
compatibility. Some patients may tolerate Uvar units; others genotype is associated with decreased RBC sialic acid content
may not. Many examples of anti-U are actually anti-U plus but increased glycosylation of RBC membrane band 3.
anti-GPB. If the patient is U– and N–, the antibody may
actually be a potent anti-N plus anti-U, making the search Other Antibodies in the MNS System
for compatible blood more difficult.
Antibodies to antigens other than M, N, S, and s are rarely
En(a–) Phenotype encountered and can usually be grouped into two categories:
In 1969, Darnborough and coworkers30 and Furuhjelm those directed against low-prevalence antigens and those
and colleagues31 described an antibody to the same high- directed against high-prevalence antigens.
prevalence antigen, called Ena (for envelope), which reacted Antibodies to high-prevalence antigens are easily
with all RBCs except those of the propositi. In both of these detected with antibody detection RBCs. Antibodies to low-
cases, the En(a–) individuals appeared to be M–N– with prevalence antigens are rarely detected by the antibody
reduced NeuNAc on their RBCs. The RBCs of the two indi- detection test but are seen as an unexpected incompatible
viduals were mutually compatible. crossmatch or an unexplained case of HDFN. Few hospital
Most En(a–) individuals produce anti-Ena, which is an blood banks have the test cells available to identify the
umbrella term for reactivity against various portions of GPA specificity, but enzyme reactivity and MNS antigen typing
unrelated to M or N, but not all antibodies detect the same may offer clues.
6888_Ch08_173-211 29/10/18 5:27 PM Page 193

Chapter 8 Blood Group Terminology and Common Blood Groups 193

When antibodies to low-prevalence antigens are encoun- The Kell (006) and Kx (019) Systems
tered, it is common practice to transfuse units that are cross-
match compatible at 37°C and in the antiglobulin phase of The Kell blood group system consists of 36 high-prevalence
testing. Typing sera for MNS antigens other than M and N and low-prevalence antigens; it was the first blood group sys-
and S and s are not generally available, so the antigen status tem discovered after the introduction of antiglobulin testing.
of compatible RBCs can seldom be confirmed. Also, when Anti-K was identified in 1946 in the serum of Mrs. Kelleher.
one antibody to a low-prevalence antigen is found, antibod- The antibody reacted with the RBCs of her newborn infant, her
ies to other low-prevalence antigens are also frequently pres- older daughter, her husband, and about 7% of the random
ent in the same serum. If the antibody is directed to a population.8 In 1949, anti-k, the high-prevalence antithetical
high-prevalence antigen, the assistance of an immunohema- partner to K, was described. Kell remained a two-antigen sys-
tology reference laboratory is generally needed to identify tem until the antithetical antigens Kpa and Kpb were described
the antibody and obtain appropriate antigen-negative units. in 1957 and 1958, respectively. Likewise, Jsa (described in
1958) and Jsb (described in 1963) were found to be antithetical
Autoantibodies and related to the Kell system. The discovery of the null phe-
notype in 1957, designated Ko, helped associate many other
Autoantibodies to M and N have been reported.1 Not all ex- antigens with the Kell system. Antibodies that reacted with all
amples of anti-M in M+ individuals or anti-N in N+ individ- RBCs except those with the Ko phenotype recognized high-
uals are autoantibodies. Many fail to react with the patient’s prevalence antigens that were phenotypically related.
own RBCs. It may be that these individuals have altered GPA The 36 antigens included in the Kell blood group sys-
and that their antibody is specific for a portion of the com- tem, designated by the symbol KEL or 006 by the ISBT, are
mon antigen they lack. Autoantibodies to U and Ena are listed in Table 8–15. In 1961, a numerical notation was
more common and may be associated with warm-type proposed for the Kell system. As new antigens were discov-
autoimmune hemolytic anemia. ered, some received only a number for the antigen name.
However, the traditional notation persisted and is more
Disease Associations useful in conveying antithetical relationships (e.g., K and
Jsa). Consequently, the commonly used nomenclature for
The MNS antigen can serve as receptors for complement, bac- Kell antigens is a mixture of symbols using letters and
teria, and viruses.10 GPAM may serve as the receptor by which numbers; obsolete terminology has been deleted from
certain pyelonephritogenic strains of E. coli gain entry to the Table 8-15. The associated antigen Kx is the only antigen
urinary tract. The malaria parasite Plasmodium falciparum in the Kx system, ISBT number 019 and symbol XK.
appears to use alternative receptors, including GPA and GPB, Kell blood group antigens are found only on RBCs.
for cell invasion; some of these receptors also involve NeuNAc. They have not been found on platelets or on lymphocytes,

Table 8–15 Kell System Antigens


Numeric Antithetical
Name Terminology ISBT Numeric Prevalence (%) Antigen(S) Year Discovered
K KEL1 9 k 1946
k KEL2 99.8 K 1949
Kpa KEL3 2 (whites) Kpb, Kpc 1957
Kpb 4 KEL4 >99.9 Kpa, Kpc 1958
Ku KEL5 >99.9 1957
Jsa KEL6 <0.1 whites Jsb 1958
20 blacks
Jsb KEL7 >99.9 whites Jsa 1963
99 blacks
Ula KEL10 <3 Finns 1968
K11 KEL11 >99.9 K17 1971
K12 KEL12 >99.9 1973
K13 KEL13 >99.9 1973
Continued
6888_Ch08_173-211 29/10/18 5:27 PM Page 194

194 PART II Blood Groups and Serologic Testing

Table 8–15 Kell System Antigens—cont’d


Numeric Antithetical
Name Terminology ISBT Numeric Prevalence (%) Antigen(S) Year Discovered
K14 KEL14 >99.9 K24 1973

K16 KEL16 99.8 1976

Wka K17 KEL17 0.3 K11 1974

K18 KEL18 >99.9 1975

K19 KEL19 >99.9 1979

Km KEL20 >99.9 1979

Kpc KEL21 <0.1 Kpa, Kpb 1945

K22 KEL22 >99.9 1982

K23 KEL23 <0.01 1987

K24 KEL24 <2 K14 1985

VLAN KEL25 <0.1 VONG 1996

TOU KEL26 >99.9 1995


RAZ KEL27 >99.9 1994
VONG KEL28 <0.1 VLAN 2003
KALT KEL29 >99.9 2006
KTIM KEL30 >99.9 2006
KYO KEL31 <0.1 KYOR 2006
KUCI KEL32 >99.9 2007
KANT KEL33 >99.9 2007
KASH KEL34 >99.9 2007
KELP KEL35 >99.9 2010
KETI KEL36 High 2011
KHUL KEL37 High KEAL 2011
KYOR KEL38 High KYO 2013
KEAL KEL39 Low KHUL 2017

Obsolete terminology deleted

granulocytes, or monocytes. The associated Xk protein is to enzyme), destroy Kell antigens but not Kx. Glycine-acid
found in erythroid tissues and in other tissues, such as EDTA (an IgG-removal agent) also destroys Kell antigens.
brain, lymphoid organs, heart, and skeletal muscle.32 The prevalence of common Kell phenotypes are listed in
The K antigen can be detected on fetal RBCs as early as Table 8–16. There are eight sets of antithetical antigens; an-
10 weeks and is well developed at birth. The k antigen has tithetical relationships have not been established for the
been detected at 7 weeks. The total number of K antigen sites other high- and low-prevalence antigens. Some of the Kell
per RBC is quite low: only 3,500 up to 18,000 sites per antigens (e.g., Jsb) are more prevalent in certain populations.
RBC.10 Despite its lower quantity, K is very immunogenic.
The antigens are not denatured by the routine blood bank K and k Antigens
enzymes ficin and papain but are destroyed by trypsin and
chymotrypsin when used in combination.8 Thiol-reducing Excluding ABO, K is rated second only to D in immuno-
agents, such as 100 to 200 mM DTT, 2-mercaptoethanol genicity. Most anti-K appear to be induced by pregnancy and
(2-ME), AET, and ZZAP (which contains DTT in addition transfusion.8 Fortunately, the prevalence of K antigen is low
6888_Ch08_173-211 29/10/18 5:27 PM Page 195

Chapter 8 Blood Group Terminology and Common Blood Groups 195

Table 8–16 Prevalence of Common Kell 20-day-old infant with an E. coli O125:B15 infection whose
mother did not make anti-K. The organism was shown to
System Phenotypes
have a somatic K-like antigen that reacted with the infant’s
Phenotype Whites (%) Blacks (%) antibody, so the bacterial antigen was thought to have been
K–k+ 91 96.5 the stimulus. The antibody disappeared after recovery.
Some examples of anti-K react poorly in methods incor-
K+k+ 8.8 3.5 porating low-ionic media, such as LISS, and in some auto-
K+k– 0.2 <0.1 mated systems. The most reliable method of detection is the
indirect antiglobulin test. The potentiating medium, PEG,
Kp(a+b–) <0.1 0 may increase reactivity.
Kp(a+b+) 2.3 Rare Anti-K has been implicated in severe HTRs. Although
some examples of anti-K bind complement, in vivo RBC
Kp(a–b+) 97.7 100 destruction is usually extravascular via the macrophages in
Js(a+b–) 0 1 the spleen. Anti-K is also associated with severe HDFN. The
antibody titer does not always accurately predict the severity
Js(a+b+) Rare 19 of disease; stillbirth has been seen with anti-K titers as low
Js(a–b+) 100 80 as 64. Fetal anemia in anti-K HDFN is associated with sup-
pression of erythropoiesis due to destruction of erythroid
precursor cells, which can be additional to destruction of cir-
culating antigen-positive RBCs, as seen in anti-D HDFN. Kell
(9% in whites), and the chance of receiving a K+ unit is glycoprotein is expressed on fetal RBCs at a much earlier
small. If anti-K develops, compatible units are easy to find. stage of erythropoiesis than Rh antigens.34 When a pregnant
Antibodies to k antigen are seldom encountered. Only woman is identified as making anti-K, it is prudent to type
2 in 1,000 individuals lack k and are capable of developing the father for the K antigen. If he is K+, the fetus should be
the antibody. The likelihood that these few individuals will monitored carefully for signs of HDFN.
receive transfusions and become immunized is even less.
Antibodies to Kpa, Jsa, and Other Low-Prevalence
Kpa, Kpb, and Kpc Antigens Kell Antigens

Alleles Kpa and Kpc are low-prevalence mutations of their Antibodies to the low-prevalence Kell antigens are rare be-
high-prevalence partner Kpb. The Kpa antigen is found in cause so few people are exposed to these antigens. Because
about 2% of whites. The Kpa gene is associated with suppres- routine antibody detection RBCs do not carry low-prevalence
sion of other Kell antigens on the same molecule, including antigens, the antibodies are most often detected through un-
k and Jsb.8 The effect appears to result from a reduced expected incompatible crossmatches or cases of HDFN.
amount of the Kell glycoprotein (produced by the Kpa allele) The serologic characteristics and clinical significance of
inserted in the RBC membrane. The Kpc antigen is even these antibodies parallel anti-K. The original anti-Kpa was
rarer. naturally occurring, but most antibodies result from trans-
fusion or pregnancy.
Jsa and Jsb Antigens
Antibodies to k, Kpb, Jsb, and Other
The Jsa antigen, antithetical to the high-prevalence antigen Jsb, High-Prevalence Kell Antigens
is found in about 20% of blacks but in fewer than 0.1% of
whites.10 The prevalence of Jsa in blacks is almost 10 times Antibodies to high-prevalence Kell system antigens are rare
greater than the prevalence of the K antigen in blacks. Jsa and because so few people lack these antigens. They also parallel
Jsb were linked to the Kell system when it was discovered that anti-K in serologic characteristics and clinical significance.
Ko RBCs were Js(a–b–). The high-prevalence antibodies are easy to detect but dif-
ficult to work with because most blood banks do not have
Anti-K the antigen-negative panel cells needed to exclude other
alloantibodies, nor do they have typing reagents to phenotype
Outside of the ABO and Rh antibodies, anti-K is the most the patient’s RBCs. Testing an unidentified high-prevalence
common antibody seen in the blood bank. Anti-K is usually antibody against DTT- or AET-treated RBCs is a helpful tech-
IgG and reactive in the antiglobulin phase, but some exam- nique: reactivity that is abolished with DTT or AET treatment
ples agglutinate saline-suspended RBCs. The antibody is suggests that the antibody may be related to the Kell system
usually made in response to antigen exposure through preg- and enables the technologist to exclude common alloantibod-
nancy and transfusion and can persist for many years. ies. Caution is needed before assigning Kell system specificity
Naturally occurring IgM examples of anti-K are rare and until antigen-negative RBCs are tested because DTT also
have been associated with bacterial infections. Marsh denatures JMH and high-prevalence antigens in the LW,
and colleagues33 studied an IgM anti-K in an untransfused Lutheran, Dombrock, Cromer, and Knops systems. Finding
6888_Ch08_173-211 29/10/18 5:27 PM Page 196

196 PART II Blood Groups and Serologic Testing

compatible units for transfusion can be difficult; siblings and found to carry the encoding alleles on opposite chromosomes.
rare-donor inventories are the most likely sources. Patients For example, someone who types Kp(a+) and Js(a+) is genet-
with antibodies to high-prevalence antigens should be en- ically kKpaJsb on one chromosome and kKpbJsa on the other.
couraged to donate autologous units and, if possible, to par- However, in a 2009 report, two unrelated individuals with
ticipate in a rare-donor program. very weak K antigens were found to be heterozygous for KKpa
and kKpb.
Biochemistry The gene XK, which encodes the Kx antigen, is indepen-
dent of KEL and is located on the short arm of the X chromo-
some at position Xp21.1.4,8
Advanced Concepts
The Kell antigens are located on a glycoprotein that con- The Kx Antigen
sists of 731 amino acids and spans the RBC membrane
once. The N-terminal domain is intracellular, and the large Kx is present on all RBCs except those of the rare McLeod
external C-terminal domain is highly folded by disulfide phenotype (see “The McLeod Phenotype and Syndrome”
linkages (Fig. 8–8). The Kell glycoprotein is covalently section below). Ko and Kmod phenotype RBCs have increased
linked with another protein, called Xk, by a single disulfide Kx antigen.8 When Kell antigens are denatured with AET or
bond. The Xk protein (440 amino acids) is predicted to DTT, the expression of Kx increases.
span the RBC membrane 10 times. Kell antigen expression
is dependent upon the presence of the Xk protein. The Ko Phenotype and Anti-Ku
The Kell glycoprotein is a member of the neprilysin
Ko RBCs lack expression of all Kell antigens. Ko RBCs have
(M13) family of zinc endopeptidases associated with the
no membrane abnormality and survive normally in circula-
cleavage of big endothelins, but how this relates to the phys-
tion. The phenotype is rare; data suggest a frequency of
iological role of the Kell glycoprotein remains unclear. The
1:25,000 in whites.8
structure of the Xk protein suggests a membrane transport
Immunized individuals with the Ko phenotype typically
protein. The absence of Xk results in McLeod syndrome (see
make an antibody called anti-Ku that recognizes the “universal”
“The McLeod Phenotype and Syndrome” section below).
Kell antigen (Ku) present on all RBCs except Ko. Anti-Ku ap-
pears to be a single specificity and cannot be separated into
Genetics components. Anti-Ku has caused both HDFN and HTRs.8
Because Ko RBCs are negative for k, Kpb, Jsb, and so forth,
The gene KEL, located on chromosome 7 at position 7q33, they are very useful in investigating complex antibody prob-
is organized into 19 exons of coding sequence.4 Single base lems. They can help confirm a Kell system specificity or rule
mutations encoding amino acid substitutions are responsible out other underlying specificities. When Ko RBCs are not
for the different Kell antigens. Several different mutations available, they can be made artificially by treating normal
(e.g., point, frameshift, or splice site mutations) have been RBCs with DTT, AET, or glycine-acid EDTA.
found that result in the rare null phenotype Ko.8,10
Historically, no Kell haplotype had been shown to code for The McLeod Phenotype and Syndrome
more than one low-prevalence antigen. People who tested pos-
itive for two low-prevalence Kell antigens had always been In 1961, Allen and coworkers35 described a young male
medical student who initially appeared to be Kell null but
who demonstrated the weak expression of k, Kpb, and Jsb
detectable by adsorption-elution methods. This unusual
Kell phenotype was called McLeod, after the student.
COOH The McLeod phenotype is very rare. All who have it are
male, and inheritance is X-linked through a carrier mother.
McLeod phenotype RBCs lack Kx and the Kell system high-
Kx prevalence antigen, Km, and have marked depression of all
other Kell antigens. The weakened expression of the Kell anti-
gens is designated by a superscript w for “weak”—for example,
S-S
K–k+w Kp(a–b+w). The McLeod phenotype has been associated
with several mutations and deletions at the XK locus.
A significant proportion of the RBCs in individuals with
NH2 the McLeod phenotype are acanthocytic (having irregular
NH2 shapes and protrusions) with decreased deformability
COOH and reduced in vivo survival. As a result, individuals with
the McLeod phenotype have a chronic but often well-
Figure 8–8. Proposed structures for Kell and Kx proteins. The two proteins are
linked through one disulfide bond. The conformation of the large external domain compensated hemolytic anemia characterized by reticulo-
of the Kell glycoprotein is unknown; 15 cysteine residues suggest the presence of cytosis, bilirubinemia, splenomegaly, and reduced serum
disulfide bonds and extensive folding. haptoglobin levels.
6888_Ch08_173-211 29/10/18 5:27 PM Page 197

Chapter 8 Blood Group Terminology and Common Blood Groups 197

Individuals with the McLeod phenotype have a variety of antigens are also seen on RBCs with the rare Gerbich-negative
muscle and nerve disorders that, together with the serologic phenotypes Ge: –2, –3, 4 and Ge: –2, –3, –4. The phenotypic
and hematologic picture, are collectively known as the relationship between Gerbich and Kell is not understood. The
McLeod syndrome, one of the neuroacanthocytosis syn- umbrella term Kmod is used to describe other phenotypes with
dromes. McLeod individuals develop a slow, progressive very weak Kell expression, often requiring adsorption-elution
form of muscular dystrophy between ages 40 and 50 years tests for detection. As a group, these RBCs have a reduced
and cardiomegaly (leading to cardiomyopathy). The associ- amount of Kell glycoprotein and enhanced Kx expression.
ated neurological disorder presents initially as areflexia (a Some Kmod individuals make an antibody that resembles
lack of deep tendon reflexes) and progresses to choreiform anti-Ku but does not react with other Kmod RBCs (unlike anti-
movements (well-coordinated but involuntary movements). Ku made by Ko individuals).
These individuals also have elevated serum creatinine phos- Patients with autoimmune hemolytic anemia, in which the
phokinase levels of the MM type (cardiac/skeletal muscle) autoantibody is directed against a Kell antigen, may have de-
and carbonic anhydrase III levels. pressed expression of that antigen. Antigen strength returns
In 1971, Giblett and colleagues made an association to normal when the anemia resolves and the DAT becomes
between the rare Kell phenotypes, including the McLeod negative. This phenomenon appears to be more common in
phenotype, and the rare X-linked chronic granulomatous the Kell system than in others.1
disease (CGD).36 CGD is characterized by the inability of Finally, RBCs may appear to acquire Kell antigens.
phagocytes to make NADH oxidase, an enzyme important McGinnis and coworkers38 described a K– patient who
in generating H2O2, which is used to kill ingested bacteria. acquired a K-like antigen during a Streptococcus faecium
Afflicted children can die at an early age from overwhelm- infection. Cultures containing the disrupted organism con-
ing infections if not treated. Not all males with the McLeod verted K– cells to K+, but bacteria-free filtrates did not.
phenotype have CGD, nor do all patients with CGD have
the McLeod phenotype. Autoantibodies
The expression of Kx in women who are carriers of the
McLeod phenotype follows the Lyon hypothesis, which Most Kell system autoantibodies are directed against unde-
states that in early embryo development, one X chromosome fined high-prevalence Kell antigens, but identifiable autoan-
randomly shuts down in female cells that have two. All cells tibodies to K, Kpb, and K13 have been reported. Mimicking
descending from the resulting cell line express only the allele specificities have also been reported, such as when an appar-
on the active chromosome. Hence, McLeod carriers exhibit ent anti-K is eluted from DAT+ K– RBCs and the anti-K in
two RBC populations: one having Kx and normal Kell anti- the eluate can be adsorbed onto K– RBCs.
gens, the other having the McLeod phenotype and acantho-
cytosis. The percentage of McLeod phenotype RBCs in The Duffy (008) System
carriers varies from 5% to 85%.1
McLeod males with CGD make anti-Kx + Km (sometimes The Duffy blood group system was named for Mr. Duffy, a
called anti-KL), which reacts strongly with Ko RBCs, weaker multiply transfused hemophiliac who in 1950 was found to
with normal Kell phenotype RBCs, and not at all with have the first described example of anti-Fya. One year later,
McLeod phenotype RBCs. Anti-Km is made by McLeod the antibody defining its antithetical antigen, Fyb, was found
males without CGD. There are rare reports of a McLeod male in the serum of a woman who had had three pregnancies.
without CGD who has made anti-Kx + Km.37 The expression In 1955, Sanger and colleagues39 reported that the major-
of Kell antigens on RBCs with common, McLeod, and Ko ity of African Americans tested were Fy(a–b–). The gene
phenotypes is summarized in Table 8–17. responsible for this null phenotype was called Fy. FyFy
appeared to be a common genotype in blacks, especially in
Altered Expressions of Kell Antigens Africa; the gene is exceedingly rare in whites.
In 1975, it was observed that Fy(a–b–) RBCs resist infec-
Weaker than normal Kell antigen expression is associated tion in vitro by the monkey malaria organism Plasmodium
with the McLeod phenotype and the suppression by the Kpa knowlesi. It was later shown that Fy(a–b–) RBCs also resist
gene (cis-modified effect) on Kell antigens. Depressed Kell infection by Plasmodium vivax (one of the organisms causing

Table 8–17 Expression of Kell Antigens on RBCs With Common, Ko, and McLeod Phenotypes
Phenotype RBC Antigen Expression Possible Antibody
Kell Antigens Km Kx

Common k, Kpb, Jsb, K11 . . . Normal Weak Alloantibody

Ko None None Increased Anti-Ku

McLeod Trace k, Kpa, Jsb, K11 . . . None None Anti-Kx + Km (CGD): Anti-Km (non-CGD)
6888_Ch08_173-211 29/10/18 5:27 PM Page 198

198 PART II Blood Groups and Serologic Testing

malaria in humans).39 This discovery provides an explana- react with enzyme-treated RBCs, this is a helpful technique
tion for the predominance of the Fy(a–b–) phenotype in per- to identify other antibodies when multiple antibodies are
sons originating from West Africa. present.
Antibodies to other antigens in the Duffy blood group Some examples of anti-Fya and anti-Fyb show dosage,
system, Fy3, Fy5, are rarely encountered. RBCs that are reacting more strongly with RBCs that have a double dose
Fy(a–b–) are also Fy: –3, –5. Fy5 is also not present on Rhnull than RBCs from heterozygotes. It must be remembered that
RBCs, regardless of the Fya or Fyb status of those RBCs. The some reagent RBCs that appear to be from homozygotes
Duffy blood group system is designated by the symbol FY or (and have a double dose of either Fya or Fyb) may actually
008 by the ISBT. be from heterozygotes if they are from black donors; a silent
allele, Fy, is commonly found in blacks. For example,
Fya and Fyb Antigens Fy(a+b–) RBCs will have a double dose of Fya if they are
from a white FyaFya donor but will have a single dose of
The Duffy antigens most important in routine blood bank Fya if they are from a black donor who is genetically FyaFy.
serology are Fya and Fyb. They can be identified on fetal Additional phenotypic markers commonly found in black
RBCs as early as 6 weeks gestational age and are well devel- donors can give a clue to the possible presence of the silent
oped at birth. There are about 13,000 to 14,000 Fya or Fyb Fy allele: Ro, S–s–, V+VS+, Js(a+), Le(a–b–).
sites on Fy(a+b–) and Fy(a–b+) RBCs, respectively; there are Anti-Fya and anti-Fyb have been associated with acute and
half that number of Fya sites on Fy(a+b+) RBCs.8 The anti- delayed HTRs. Once the antibody is identified, Fy(a–) or
gens have not been found on platelets, lymphocytes, mono- Fy(b–) blood must be given; finding such units in a random
cytes, or granulocytes, but they have been identified in other population is not difficult. For example, one in three random
body tissues, including brain, colon, endothelium, lung, units of blood is Fy(a–) and one in five random units of
spleen, thyroid, thymus, and kidney cells.10 The prevalence blood is Fy(b–). Anti-Fya and anti-Fyb are associated with
of the common phenotypes in the Duffy system are given in HDFN that ranges from mild to severe.
Table 8–18. The disparity in distribution in different races is Rare autoantibodies with mimicking Fya and Fyb speci-
notable. ficity have been reported—for example, anti-Fyb that can be
Fya and Fyb antigens are destroyed by common prote- adsorbed onto and eluted from Fy(a+b–) RBCs. Issitt and
olytic enzymes such as ficin, papain, bromelin, and chy- Anstee1 suggest that these may represent alloantibodies with
motrypsin, and by ZZAP (which contains either papain or “sloppy” specificity made early in an immune response.
ficin in addition to DTT); they are not affected by DTT alone,
AET, or glycine-acid EDTA treatment. Neuraminidase may Biochemistry
reduce the molecular weight of Fya and Fyb, but it does not
destroy antigenic activity and neither does purified trypsin.
Advanced Concepts
Anti-Fya and Anti-Fyb Enzymes, membrane solubilization methods, immunoblot-
ting, radiolabeling, and amino acid sequencing have all
Anti-Fya is a common antibody and is found as a single been used to study the biochemistry of Duffy antigens.1
specificity or in a mixture of antibodies. Anti-Fya occurs Duffy antigens reside on a glycoprotein of 336 amino acids
three times less frequently than anti-K. Anti-Fyb is 20 times that has a relative mass of 36 kD and two N-glycosylation
less common than anti-Fya and often occurs in combination sites (Fig. 8–9).40 The glycoprotein is predicted to traverse
with other antibodies. The antibodies are usually IgG and the cell membrane seven times and has two predicted disul-
react best at the antiglobulin phase. Rare examples of anti- fide bridges.
Fya and anti-Fyb bind complement. A few examples are The amino acid at position 42 on the Duffy glycoprotein
saline agglutinins. Antibody activity is enhanced in a low- defines the Fya and Fyb polymorphism: Fya has glycine, and
ionic strength medium. Because anti-Fya and anti-Fyb do not Fyb has aspartic acid. The Fy3 epitope, as defined by mon-
oclonal antibody, is on the third extracellular loop, and Fy6
appears to involve amino acids 19 through 25.40
The Duffy glycoprotein is a member of the superfamily
Table 8–18 Prevalence of Common Duffy
of chemokine receptors and is known as the atypical
Phenotypes chemokine receptor 1 (ACKR1, previously known as
African DARC). Thus, in addition to being a receptor for the
Americans Chinese malaria parasite P. vivax, the Duffy glycoprotein binds a va-
Phenotype Whites(%) (%) (%) riety of proinflammatory cytokines.
Fy(a+b–) 17 9 90.8

Fy(a+b+) 49 1 8.9 Genetics


Fy(a–b+) 34 22 0.3 In 1968, the Duffy gene (ACKR1, formally known as DARC)
Fy(a–b–) Very rare 68 0 was linked to a visible, inherited abnormality of chromosome 1,
thus becoming the first human gene to be assigned to a
6888_Ch08_173-211 29/10/18 5:27 PM Page 199

Chapter 8 Blood Group Terminology and Common Blood Groups 199

Fy6
Fya/Fyb Fy3 antigenic determinant or precursor common to both Fya and

[
NH2 Fyb and was called Fy3. Unlike Fya and Fyb, the Fy3 antigen

[
S SS is not destroyed by enzymes.

S
Anti-Fy3 is a rare antibody made by Fy(a–b–) individuals
who lack the Duffy glycoprotein. The Fy(a–b–) phenotype
has been found in whites, Cree Indian families, and blacks.1
Blacks with the Fy(a–b–) phenotype rarely make anti-Fy3.
COOH Some blacks who make anti-Fy3 initially make anti-Fya.8

Figure 8–9. Proposed structure for the Duffy protein. Disulfide bonds probably Fy5 Antigen and Antibody
link the NH2 terminal domain and the third loop and the first and second loop.
In 1973, Colledge and coworkers42 discovered anti-Fy5 in
the serum of an Fy(a–b–) black child who later died of
specific chromosome. The gene ACKR1 is located near the leukemia. Initially it was thought to be a second example of
centromere on the long arm of chromosome 1 at position anti-Fy3, because it reacted with all Fy(a+) or Fy(b+) RBCs
1q21-q22.4 The Fy locus is syntenic to the Rh locus, which but not with Fy(a–b–) cells. The antibody differed in that it
is located near the tip of the short arm; that is, they are on reacted with the cells from an Fy(a–b–)Fy:–3 white female,
the same chromosome, but they are far enough apart that but it did not react with Fy(a+) or Fy(b+) Rhnull RBCs and
linkage cannot be demonstrated and serologically they reacted only weakly with Fy(a+) or Fy(b+) –D– RBCs.
appear to segregate independently. Sometimes, sera containing anti-Fy5 also contain anti-Fya.
There are three common alleles at the Fy locus: Fya, Fyb, Several examples of anti-Fy5 have been reported in multiply
and Fy. Fya and Fyb encode the antithetical antigens Fya and transfused Fy(a–b–) patients with sickle cell disease with a
Fyb, respectively, and Fy is a silent allele and is the major mixture of other antibodies.
allele in blacks. The Fy gene in Fy(a–b–) blacks is predomi- The molecular structure of Fy5 is not known, but it ap-
nantly an Fyb variant with a change in the promoter region pears to be the result of interaction between the Rh complex
(GATA) of the gene, which disrupts the binding site for and the Duffy glycoprotein. People who are Fy(a–b–) or
mRNA transcription in the RBC.10 Consequently, Fy(a–b–) Rhnull do not make Fy5 antigen and are at risk of making the
blacks do not express Fyb on their RBCs but express Fyb in antibody, although few do. Like Fy3, Fy5 is not destroyed by
other tissues. The presence of Fyb in tissues presumably pre- enzymes.
cludes the recognition of Fyb as foreign; thus, no anti-Fyb is
made by these individuals. Molecular testing for the GATA The Kidd (009) System
mutation is helpful for transfusion management for patients
with sickle cell disease. A molecular analysis of Fy(a–b–) The Kidd blood group is a simple and straightforward
whites revealed different mutations. These individuals carry system consisting of only three antigens. In 1951, Allen and
no Duffy protein on their RBCs or on other tissues and thus colleagues43 reported finding an antibody in the serum of
can form anti-Fyb and anti-Fy3. Mrs. Kidd, whose infant had HDFN. The antibody, named
Typing for Duffy antigens has been performed on the anti-Jka (for the infant John Kidd), reacted with 77% of
RBCs of chimpanzees, gorillas, and old- and new-world Bostonians. Its antithetical partner, Jkb, was found 2 years
monkeys. The results suggest that Fy3 developed first, then later. The null phenotype Jk(a–b–) was described in 1959.
Fyb, and that Fya arose during human evolution.10 The propositus made an antibody to a high-prevalence
antigen called Jk3, which is present on any RBC positive
Fyx for Jka or Jkb. No other antigens associated with the Kidd
system have been described.
Fyx was described in 1965 as a new allele at the Fy locus. It The Kidd system is designated by the symbol JK or 009
does not produce a distinct antigen but rather is an inherited by the ISBT. It has special significance to routine blood bank-
weak form of Fyb that reacts with some examples of anti-Fyb. ing because of its antibodies, which can be difficult to detect
Fyx has been described in white populations. Individuals and are a common cause of HTRs.
with Fyx may type Fy(b–), but their RBCs adsorb and elute
anti-Fyb. They also have depressed expression of their Fy3 Jka and Jkb Antigens
and Fy5 antigens. The decreased expression of Fyb due to
Fyx appears to be related to a reduced amount of Duffy gly- Jka and Jkb are antigens commonly found on RBCs of most
coprotein on the surface of RBCs.41 There is no anti-Fyx. individuals. Table 8–19 summarizes the prevalence of the
four known phenotypes. There are notable differences in
Fy3 Antigen and Antibody antigen frequency among various races: 91% of blacks and
77% of whites are Jk(a+); 57% of blacks and only 28% of
In 1971, anti-Fy3 was found in the serum of an Fy(a–b–) whites are Jk(b–).
white Australian female. It reacted with all RBCs tested Jka and Jkb antigens are well developed on the RBCs of
except those of the Fy(a–b–) phenotype. Because it was an neonates. Jka has been detected on fetal RBCs as early as
inseparable anti-FyaFyb, it was thought to react with an 11 weeks; Jkb has been detected at 7 weeks.8 Although this
6888_Ch08_173-211 29/10/18 5:27 PM Page 200

200 PART II Blood Groups and Serologic Testing

Table 8–19 Prevalence of Kidd Phenotypes plasma) and using polyspecific reagents with both anti-IgG
and anti-complement can be helpful in these situations.1
Whites Blacks Asians The titer of anti-Jka or anti-Jkb quickly declines in vivo.
Phenotype (%) (%) (%) A strong antibody identified following a transfusion reaction
Jk(a+b–) 28 57 23 may be undetectable in a few weeks or months.1 This con-
firms the need to check blood bank records for previously
Jk(a+b+) 49 34 50
identified antibodies before a patient is transfused. It is
Jk(a–b+) 23 9 27 equally important to inform the patient that he or she has
such an antibody and to provide a wallet card or other doc-
Jk(a–b–) Exceedingly Exceedingly 0.9 Polynesians
umentation that notes the specificity in case the patient is
rare rare
transfused elsewhere.
The decline in antibody reactivity and the difficulty in
detecting Kidd antibodies are reasons why they are a com-
mon cause of HTRs, especially of the delayed type. Although
early development of Kidd antigens contributes to the intravascular hemolysis has been noted in severe reactions,
potential for HDFN, anti-Jka and anti-Jkb are only rarely coated RBCs more often are removed extravascularly. The
responsible for severe HDFN. Jk(a+b–) RBCs carry 14,000 rate of clearance of incompatible RBCs can vary but is
antigen sites per cell.8 The Kidd antigens are not very usually rapid.
immunogenic. Contrary to their hemolytic reputation in transfusion,
Kidd antigens are not denatured by papain or ficin; treat- most Kidd antibodies are only rarely associated with severe
ment of RBCs with enzymes generally enhances reactivity cases of HDFN.44
with Kidd antibodies. Kidd antigens are also not affected
by chloroquine, DTT, AET, or glycine-acid EDTA. The anti- Biochemistry
gens are not found on platelets, lymphocytes, monocytes,
or granulocytes.
Advanced Concepts
Anti-Jka and Anti-Jkb Heaton and McLoughlin45 reported in 1982 that Jk(a–b–)
Kidd antibodies have a notorious reputation in the blood RBCs resist lysis in 2M urea, a solution commonly used
bank. They demonstrate dosage, are often weak, and are to lyse RBCs in a sample before it is used in some auto-
found in combination with other antibodies, all of which mated platelet-counting instruments. Urea crosses the RBC
make them difficult to detect. membrane, causing an osmotic imbalance and an influx
Anti-Jka is more frequently encountered than anti-Jkb, but of water, which rapidly lyses normal cells. With Jk(a+) or
neither antibody is common. The antibodies are usually IgG Jk(b+) RBCs, lysis in 2M urea occurs within 1 minute; with
(antiglobulin reactive) but may also be partly IgM and are Jk(a–b–) cells, lysis is delayed by 30 minutes.8
made in response to pregnancy or transfusion. The predicted Kidd glycoprotein has 389 amino
The ability of Kidd antibodies to show dosage can con- acids with 10 membrane-spanning domains and two
found inexperienced serologists. Many anti-Jka and anti-Jkb N-glycosylation sites, one of which is extracellular on the
react more strongly with RBCs that carry a double dose of third extracellular loop (Fig. 8–10). The glycoprotein is
the respective antigen and may not react with Jk(a+b+) a urea transporter.
RBCs. An anti-Jka that reacts only with Jk(a+b–) RBCs can
give inconclusive panel results and appear compatible with Genetics
Jk(a+b+) cells. Readers are urged to rule out anti-Jka and
anti-Jkb only with Jk(a+b–) and Jk(a–b+) panel cells, respec- The Jk locus is on chromosome 18 at position 18q11-q12.4
tively, and to type all crossmatch-compatible units with com- The gene SLC14A1 (for solute carrier family 14, member 1)
mercial antisera. To ensure that antisera can indeed detect
weak expressions of the antigen, Jk(a+b+) RBCs should be
tested in parallel as the positive control.
Antibody reactivity can also be enhanced by using LISS Jka/Jkb
or PEG (to promote IgG attachment), by using four drops of
serum instead of two in a saline tube method (to increase
the antibody-to-antigen ratio), or by using enzymes such as
ficin or papain. In vitro hemolysis can sometimes be ob-
served with enzyme-treated RBCs if serum is tested; antigen
dose may influence this hemolytic activity.1
Many examples of the Kidd antibodies bind complement. NH2 COOH
Rare examples are detected only by the complement they Figure 8–10. Proposed structure for Kidd protein. One of two proposed
bind (i.e., they are nonreactive in antiglobulin phase of test- N-glycans is extracellular and is located on the third extracellular loop; the Jka/Jkb
ing using anti-IgG reagents). Testing serum (rather than polymorphism is located on the fourth extracellular loop.
6888_Ch08_173-211 29/10/18 5:27 PM Page 201

Chapter 8 Blood Group Terminology and Common Blood Groups 201

is a member of the urea transporter gene family. The gene Like other Kidd antibodies, anti-Jk3 reacts optimally by an
is organized into 11 exons. The Jka/Jkb polymorphism is antiglobulin test, and the reactivity is enhanced with enzyme
associated with an amino acid substitution at position 280, pretreatment of the RBCs.
predicted to be located on the fourth extracellular loop Anti-Jk3 has been associated with severe immediate and
of the glycoprotein. Molecular studies have demonstrated delayed HTRs and with mild HDFN. Compatible units
the silent Jk allele can arise from mutations in both the are best found by typing siblings or searching the rare-
Jka and Jkb alleles. Jka and Jkb are inherited as codominant donor files.
alleles.
Autoantibodies
Jk(a–b–) Phenotype and the Recessive Allele, Jk
Autoantibodies with Kidd specificity (anti-Jka, anti-Jkb, and
People with the null Jk(a–b–) phenotype lack Jka, Jkb,
and anti-Jk3) are rare, but they have been associated with
the common antigen Jk3. Although very rare, the Jk(a–b–) autoimmune hemolytic anemia.1 As with other blood
phenotype is most abundant among Polynesians, and it groups, Kidd autoantibodies may have mimicking specificity
has also been identified in Filipinos, Indonesians, and or may be associated with depressed antigen expression.
Chinese.1 The null phenotype has also been reported in
several European families (Finnish, French, Swiss, and The Lutheran (005) System
English) and in the Mato Grosso Indians of Brazil. The
In 1945, anti-Lua was found (and described in detail a year
delayed lysis of Jk(a–b–) RBCs in 2M urea has proved an
later) in the serum of a patient with lupus erythematosus,
easy way to screen families and populations for this rare
following the transfusion of a unit of blood carrying the
phenotype.
corresponding low-prevalence antigen.46 (This patient also
No clinical abnormalities have been associated with
made anti-c, anti-N, the first example of anti-CW, and anti-
the Jk(a–b–) phenotype to date. Several unrelated Jk(a–b–)
Levay, now known as anti-Kpc!) The new antibody was
individuals had normal blood urea nitrogen, creatinine, and
named Lutheran for the donor; the donor’s last name was
serum electrolytes, but studies on two individuals with
Lutteran but the donor’s blood sample was incorrectly
this phenotype showed a marked defect in their ability to
labeled.2 In 1956, Cutbush and Chanarin47 described
concentrate urine.
anti-Lub, which defined the antithetical partner to Lua.
Family studies show that most Jk(a–b–) nulls are
The blood group system appeared complete until 1961,
homozygous for the rare “silent” allele Jk. Parents of JkJk
when Crawford and colleagues48 described the first
offspring and children of JkJk parents type Jk(a+b–) or
Lu(a–b–) phenotype. Unlike most null phenotypes at the
Jk(a–b+) but never Jk(a+b+), because they are genetically
time, this one demonstrated dominant inheritance. In
JkaJk or JkbJk. Their RBCs also demonstrate a single dose
1963, the Lu(a–b–) phenotype inherited as a recessive
of Jka or Jkb antigen in titration studies.
silent allele was described.
Twenty-four antigens are part of the Lutheran system.
Jk(a–b–) Phenotype and the Dominant
Most of these antigens are high prevalence; four sets of anti-
In(Jk) Allele
gens are antithetical. Many of the antigens were associated
Another genetic explanation for the Jk(a–b–) phenotype is with the Lutheran system when their corresponding anti-
association with a dominant gene called In(Jk), for “in- bodies were nonreactive with the rare Lu(a–b–) RBCs. All
hibitor,” that shows a dominant pattern of inheritance within are summarized in Table 8–20. The ISBT designation of the
a Japanese family analogous to the inhibitor gene responsible Lutheran blood group system is LU or 005.
for the dominant type Lu(a–b–) phenotype in the Lutheran Blood bankers seldom deal with the serology of the
blood group system.8,10 Dominant type Jk(a–b–) RBCs ad- Lutheran blood group system. The antigens are either high
sorb and elute anti-Jk3 and anti-Jka or anti-Jkb (depending prevalence, so only a few people lack the antigen and can
on which genes were inherited), indicating that the antigens make an alloantibody, or very low prevalence, so that only a
are expressed but only very weakly. Individuals with the few people are ever exposed. Consequently, the antibodies
dominant type Jk(a–b–) phenotype do not make anti-Jk3. are seen infrequently, and there is not much data on the clin-
Family studies show that the In(Jk) gene does not reside at ical significance of Lutheran antibodies.
the Jk locus. The molecular basis is unknown. Although the antigens have been detected on fetal RBCs
as early as 10 to 12 weeks of gestation, they are poorly de-
Anti-Jk3 veloped at birth. As a result, HDFN is rare and only mild.10
Lutheran antigens have not been detected on platelets, lym-
Alloanti-Jk3 is an IgG antiglobulin-reactive antibody that phocytes, monocytes, or granulocytes. However, Lutheran
looks like an inseparable anti-JkaJkb. Because panel cells are glycoprotein is widely distributed in tissues: brain, lung,
Jk(a+) or Jk(b+), anti-Jk3 reacts with all RBCs tested except pancreas, placenta, skeletal muscle, and hepatocytes (espe-
the autocontrol. Most blood banks do not have the rare cells cially fetal hepatic epithelial cells).8 The presence of
needed to confirm anti-Jk3; however, they can easily deter- Lutheran glycoprotein on placental tissue may result in
mine its most probable specificity by means of antigen typ- adsorption of maternal antibodies to Lutheran antigens, thus
ing. The individual making the antibody will type Jk(a–b–). decreasing the likelihood of HDFN.10
6888_Ch08_173-211 29/10/18 5:27 PM Page 202

202 PART II Blood Groups and Serologic Testing

Table 8–20 Lutheran System Antigens


Conventional Name Prevalence (%) Year Discovered Comments
Lua 8 whites 1945 Antithetical to Lub
5 blacks
Lub 99.8 1956 Antithetical to Lua
Lu3 >99.9 1963
Lu4 >99.9 1971
Lu5 >99.9 1972
Lu6 >99.9 1972 Antithetical to Lu9
Lu7 >99.9 1972
Lu8 >99.9 1972 Antithetical to Lu14
Lu9 2 1973 Antithetical to Lu6
Lu11 >99.9 1974
Lu12 >99.9 1973
Lu13 >99.9 1983
Lu14 2.4 1977 Antithetical to Lu8
Lu16 >99.9 1980
Lu17 >99.9 1979
Aua 80 whites 1961 Antithetical to Aub
Aub 50 whites 1989 Antithetical to Aua
Lu20 >99.9 1992
Lu21 >99.9 2002
LURC >99.9 2009
LUIT High 2016
LUGA High 2016
LUAC High 2016
LUBI High 2016

Lutheran antigens are resistant to the enzymes ficin and Lutheran antigen expression is variable from one individual
papain and to glycine-acid EDTA treatment but are destroyed to another. The number of Lub sites per RBC is low, estimated
by treatment with the enzymes trypsin and α-chymotrypsin. to be from 1,640 to 4,070 on Lu(a–b+) RBCs and from 850 to
Most Lutheran antibodies do not react with RBCs treated 1,820 on Lu(a+b+) RBCs.8
with the sulfhydryl reagents DTT and AET.
Anti-Lua
Lua and Lub Antigens
Most examples of anti-Lua are IgM naturally occurring saline
Lua and Lub are antigens produced by allelic codominant agglutinins that react better at room temperature than at
genes. The prevalence of common phenotypes is listed in 37°C. A few react at 37°C by indirect antiglobulin test. Some
Table 8–21. Most individuals are Lu(b+); 8% of whites and are capable of binding complement, but in vitro hemolysis
5% of blacks are Lu(a+).10 has not been reported.
6888_Ch08_173-211 29/10/18 5:27 PM Page 203

Chapter 8 Blood Group Terminology and Common Blood Groups 203

Table 8–21 Prevalence of Common Lua/Lub NH2


Lu21
Lutheran Phenotypes Lu5
Lu17
Phenotype Most Populations (%) Lu12 V V
Lu (a+b–) 0.15 LURC
Lu4 LUGA
Lu (a+b+) 7.5 Lu8/Lu14 LUAC V V
Lu16
Lu (a–b+) 92.35
Lu20 C C
Lu (a–b–) Very rare Lu6/Lu9

Lu7 C C

Lu14
Anti-Lua often goes undetected in routine testing Aua/Aub
C C
LUBI
because most reagent RBCs are Lu(a–). Anti-Lua is more
likely encountered as an incompatible crossmatch or dur-
ing an antibody workup for another specificity. Experi-
enced technologists recognize Lutheran antibodies by their
characteristic loose, mixed-field reactivity in a test tube.
Examples of anti-Lua that react only at temperatures below Lu B-CAM
37°C are clinically insignificant. There are no documented
cases of immediate HTRs; there are only rare and mild Figure 8–11. The two structures encoded by the Lu gene: the longer Lu glyco-
delayed HTRs due to anti-Lua.6 protein and the shorter basal cell adhesion molecule (B-CAM). The five extracellular
disulfide-bonded domains are homologous to immunoglobulin variable (V) or con-
stant (C) domains.
Anti-Lub

Although the first example of anti-Lub was a room tempera- variable or constant domains. The Lutheran proteins are
ture agglutinin, and IgM and IgA antibodies have been multifunctional adhesion molecules that bind laminin,
noted, most examples of anti-Lub are IgG and reactive at notably in sickle cell disease.49
37°C at the antiglobulin phase. The antibody is made in The molecular basis for the four pairs of antithetical
response to pregnancy or transfusion. antigens and several of the high-prevalence antigens
Alloanti-Lub reacts with all cells tested except the au- has been determined through the creation of Lutheran
tocontrol, and reactions are often weaker with Lu(a+b+) glycoprotein mutants and subsequent sequencing of the
RBCs and cord RBCs. Anti-Lub has been implicated with exons encoding the extracellular domains. For most of
shortened survival of transfused cells and post-transfusion the antigens, expression was caused by a single nucleotide
jaundice, but severe or acute hemolysis has not been polymorphism, resulting in single amino acid changes on
reported. the protein level.50

Biochemistry
Genetics
Advanced Concepts The LU gene BCAM is located on chromosome 19 at position
The Lutheran antigens are located on a type 1 transmem- 19q13.2, along with genes that govern expression of several
brane protein. The protein exists in two forms as a result of blood group antigens (H, Se, Le, LW, Oka).4 A linkage
alternative RNA splicing: the longer Lu glycoprotein and between Lu and the Se gene (FUT2) was the first example of
the shorter basal cell adhesion molecule (B-CAM). The autosomal linkage described in humans.
longer 85-kD protein contains 597 amino acids with five ex-
tracellular domains, a hydrophobic transmembrane domain Lu(a–b–) Phenotypes
of 19 amino acids, and a cytoplasmic domain of 59 amino
acids (Fig. 8–11). The smaller isoform (78 kD) is identical Three genetic explanations for the Lu(a–b–) phenotype have
except for a shorter cytoplasmic domain of 59 amino acids. been described. These are summarized in Table 8–22.
The external portion consists of five disulfide-bonded
Dominant Type Lu(a–b–)
domains. The Lutheran glycoproteins belong to the im-
munoglobulin superfamily of proteins; the repeating extra- The first Lu(a–b–) family study was reported by the proposi-
cellular domains are homologous to immunoglobulin tus herself.48 Because the phenotype was seen in successive
generations in 50% of her family members and others, and
6888_Ch08_173-211 29/10/18 5:27 PM Page 204

204 PART II Blood Groups and Serologic Testing

Table 8–22 Summary of Lu(a–b–) Phenotypes


Mode of Other RBC Antigens
Inheritance Gene Responsible Lu Antigens Affected Make Anti-Lu3?
Dominant KLF151 Extremely weak Reduced P1, i, Inb, AnWj, No
MER2, CD44
Not at Lu locus
Yes
Recessive Lu None Not affected
No
X-linked GATA154 Extremely weak Not affected

because null individuals passed normal Lutheran genes to Recessive X-Linked Inhibitor Type
their offspring, the expression of Lutheran was thought to An Lu(a–b–) phenotype in a large Australian family did not
be suppressed by a rare dominant regulator gene later called fit either the dominant or recessive inheritance patterns. All
In(Lu) for “inhibitor of Lutheran.” Recently, mutations in the Lu(a–b–) family members were male and carried trace
gene for erythroid Krüppel-like factor (EKLF), a transcrip- amounts of Lub detected by adsorption-elution. The pattern
tion factor, were shown to be associated with the In(Lu) of inheritance suggested an X-borne inhibitor to Lutheran.
phenotype in 21 of 24 In(Lu) individuals studied.51 In all Hemizygosity for a mutation in the X-linked gene for
cases, the mutated KLF1 allele occurred in the presence of a the major erythroid transcription factor GATA-1 has been
normal KLF1 allele. The authors of this study concluded shown to result in this X-linked inheritance of Lu(a–b–) phe-
that the In(Lu) phenotype is caused by inheritance of a loss- notype.54 There have been no other families reported with
of-function mutation on one allele of KLF1. The original this rare X-linked Lu(a–b–) phenotype.10
proband for the In(Lu) phenotype was recently shown to
have the responsible KLF1 mutation.52 Anti-Lu3
Dominant type Lu(a–b–) RBCs carry trace amounts of
Lutheran antigens as shown by adsorption-elution studies. Anti-Lu3 is a rare antibody that reacts with all RBCs except
For example, the RBCs of a person with the dominant type Lu(a–b–) RBCs. The antibody looks like inseparable anti-
Lu(a–b–) who inherited two normal Lub genes will type Luab and recognizes a common antigen, Lu3, that is present
Lu(a–b–) with routine methods but will adsorb and elute whenever Lua or Lub is present (much like the Jk3 associa-
anti-Lub. Because individuals with the dominant type tion with Jka and Jkb). Anti-Lu3 is usually antiglobulin reac-
Lu(a–b–) RBCs have normal Lutheran antigens, they do tive. This antibody is made only by individuals with the
not make anti-Lu3. recessive type of Lu(a–b–).
In addition to reduced expression of Lutheran antigens,
dominant type Lu(a–b–) RBCs also can have reduced expres- Applications to Routine Blood Banking
sion of CD44 and a weak expression of P1, i, AnWj, MER2,
and Inb blood group antigens. The major blood group systems outside of ABO and Rh
become important only after patients develop unexpected
Recessive Type Lu(a–b–) antibodies. Then a fundamental knowledge of antibody char-
In some families, the Lu(a–b–) phenotype demonstrates re- acteristics, clinical significance, and antigen frequency is
cessive inheritance, the result of having two rare silent alleles needed to help confirm antibody specificity and to select ap-
LuLu at the Lutheran locus. The parents and offspring of propriate units for transfusion.
these nulls may type Lu(a–b+), but dosage studies and titers Only a few antibody specificities are commonly seen: an-
show them to carry a single dose of Lub. tibodies to M, P1, and I react at room temperature and are
Unlike the dominant type, people with recessive Lu(a–b–) considered clinically insignificant; antibodies to K, S, s, Fya,
RBCs truly lack all Lutheran antigens (i.e., they have the Fyb, Jka, and Jkb react in the antiglobulin phase and are clin-
null phenotype) and can make an inseparable anti-Luab ically significant. These and selected others are summarized
called anti-Lu3. They also have normal antigen expression in Table 8–23.
of P1, i, and the other antigens that are weakened with the Not all antibody problems are easily solved; panel reac-
dominant type. This distinction emphasizes the importance tions are sometimes inconclusive. As described in this chap-
of testing an antibody against recessive Lu(a–b–) RBCs ter, the existence of silent, regulator, and inhibitor genes can
before defining the specificity phenotypically related to affect antigen expression. Hopefully the reader will find that
Lutheran. the information in this chapter provides a starting point for
Different inactivating mutations were recently reported for serologic problem-solving. For further detailed information
three individuals with the recessive Lu(a–b–) type.53 In all about the RBC antigens and antibodies described here, see
three individuals, the mutations would result in a truncated Daniels.8 Resolution of antibody problems involving the un-
glycoprotein that would not be integrated in the RBC surface usual specificities described here may require the assistance
membrane. of an immunohematology reference laboratory.
6888_Ch08_173-211 29/10/18 5:27 PM Page 205

Chapter 8 Blood Group Terminology and Common Blood Groups 205

Table 8–23 Summary of Antibody Characteristics


Bind In Vitro Compatible In
Antibody Reactivity Enzymes Complement Hemolysis HTR HDFN U.S. Population (%)
≤RT 37 AHG

M* Most Few Few Destroy No No Few Mild— 22


severe

N* Most Few Few Destroy No No Rare Moderate 28

S Some Some Most Variable effect Some No Yes Mild 45 W


6B

s Few Few Most Variable effect Rare No Yes Mild— 11 W


severe
6B

U Rare Some Most No change No No Yes Mild— <1 B


severe

P1* Most Some Rare Enhance Rare Rare Rare No 21

PP1Pk† Most Some Some Enhance Most Most Yes Mild <0.1

P‡ Most Some Some Enhance Most Some Yes Mild— <0.1


severe

I* Most Few Few Enhance Most Few Rare No See text

i* Most Few Few Enhance Most Few No Mild See text

K Some Some Most No change Rare No Yes Mild— 91


severe

k Few Few Most No change No No Yes Mild 0.2

Kpa Some Some Most No change No No Yes Mild 98

Kpb Few Few Most No change No No Yes Mild <0.1

Jsa Few Few Most No change No No Yes Moderate 100 W


80 B

Jsb No No Most No change No No Yes Mild— 1B


moderate

Fya Rare Rare Most Destroy Rare No Yes Mild— 34 W


severe

Fyb Rare Rare Most Destroy Rare No Yes Mild 17 W


77 B

Jka‡ Few Few Most Enhance Yes§ Some Yes Mild 24

Jkb‡ Few Few Most Enhance Yes§ Some Yes Mild 26 W


52 B

Lua Most Few Few Variable effect Some No No Mild 92

Lub Few Few Most Variable effect Some No Yes Mild 0.2

B = blacks; HDFN = hemolytic disease of the fetus and newborn; HTR = hemolytic transfusion reactions; ≤RT = room temperature or colder; W = whites
*Usually clinically insignificant.
†Potent hemolysins may be associated with early abortions.
‡Associated with severe delayed HTR.
§Provided IgM is present.
6888_Ch08_173-211 29/10/18 5:27 PM Page 206

206 PART II Blood Groups and Serologic Testing

CASE STUDIES
Case 8-1

A 30-year-old female, pregnant with her second child has a routine ABO, Rh, and prenatal antibody screen with the
following results.
CABO/Rh
Anti-A Anti-B Anti-D A1 Cells B Cells Interpretation
4+ 0 3+ 0 4+

Antibody Screen
D C E c e f M N S s Lua Lub P1 Lea Leb K k Fya Fyb Jka Jkb IS 37C PEG IAT
1 + + 0 0 + 0 + 0 + 0 0 + + + 0 + + + 0 0 + 0 NH 2+
2 + 0 + + 0 0 0 + 0 + 0 + + 0 + 0 + 0 + + 0 0 NH 0
NH = No hemolysis
Panel Results
D C E c e f M N S s Lua Lub P1 Lea Leb K k Fya Fyb Jka Jkb IS 37C PEG IAT
1 + + 0 0 + 0 + 0 + 0 0 + + + 0 + + + 0 0 + 0 NH 2+
2 + + 0 0 + 0 0 + 0 + 0 + + 0 + 0 + 0 + + 0 0 NH 0
3 + 0 + + 0 0 + + + + 0 + 0 + 0 0 + + 0 0 + 0 NH 0
4 + 0 0 + + + + 0 + 0 0 + + 0 0 0 + 0 0 + 0 0 NH 0
5 0 + 0 + + + + + 0 + 0 + + 0 + 0 + + 0 + + 0 NH 0
6 0 0 + + + + + + 0 + 0 + 0 0 + 0 + + + + + 0 NH 0
7 0 0 0 + + + 0 + 0 + 0 + + + 0 0 + 0 + 0 + 0 NH 0
8 0 0 0 + + + + 0 + + 0 + 0 0 + + 0 + + + + 0 NH 2+
9 0 0 0 + + + 0 + + 0 0 + 0 0 + 0 + + + + + 0 NH 0
10 0 0 0 + + + + 0 + + + + + + 0 0 + + 0 + 0 0 NH 0
11 + 0 + + 0 0 + + + + 0 + + 0 + + + 0 + + + 0 NH 2+
AC 0 NH 0
1. What is the patient’s ABO, Rh type, and antibody screen?
2. What is the unexpected alloantibody identified in the panel?
3. How early in fetal development can the antigen be detected on fetal RBCs?
4. Can the titer predict the severity of HDFN? Why or why not?

Case 8-2 (Advanced)

A 65-year-old male had a routine “type and screen” ordered for a scheduled hip replacement. He has a history of transfusion
20 years ago following a car accident.
ABO/Rh
Anti-A Anti-B Anti-D A1 Cells B Cells Interpretation
0 0 3+ 4+ 4+
Antibody Screen
D C E c e f M N S s Lua Lub P1 Lea Leb K k Fya Fyb Jka Jkb IS 37C PEG IAT
1 + + 0 0 + 0 + 0 + 0 0 + + + 0 + + + 0 0 + 0 NH 3+
2 + 0 + + 0 0 0 + 0 + 0 + + 0 + 0 + 0 + + 0 0 NH 3+
NH = No hemolysis
6888_Ch08_173-211 29/10/18 5:27 PM Page 207

Chapter 8 Blood Group Terminology and Common Blood Groups 207

Panel Results
D C E c e f M N S s Lua Lub P1 Lea Leb K k Fya Fyb Jka Jkb IS 37C PEG IAT
1 + + 0 0 + 0 + 0 + 0 0 + + + 0 + + + 0 0 + 0 NH 3+
2 + + 0 0 + 0 0 + 0 + 0 + + 0 + 0 + 0 + + 0 0 NH 3+
3 + 0 + + 0 0 + + + + 0 + 0 + 0 0 + + 0 0 + 0 NH 3+
4 + 0 0 + + + + 0 + 0 0 + + 0 0 0 + 0 0 + 0 0 NH 3+
5 0 + 0 + + + + + 0 + 0 + + 0 + 0 + + 0 + + 0 NH 3+
6 0 0 + + + + + + 0 + 0 + 0 0 + 0 + + + + + 0 NH 3+
7 0 0 0 + + + 0 + 0 + 0 + + + 0 0 + 0 + 0 + 0 NH 3+
8 0 0 0 + + + + 0 + + 0 + 0 0 + + 0 + + + + 0 NH 3+
9 0 0 0 + + + 0 + + 0 0 + 0 0 + 0 + + + + + 0 NH 3+
10 0 0 0 + + + + 0 + + + + + + 0 0 + + 0 + 0 0 NH 3+
11 + 0 + + 0 0 + + + + 0 + + 0 + + + 0 + + + 0 NH 3+
AC 0 NH 0
Reactivity could be due to a high incidence antibody or multiple antibodies.
• Patient’s phenotype: C-c+E-e+; K-k+; Fy(a-b-); Jk(a+b+); Le(a-b-); M+N-S-s-.
• Genotype testing shows the presence of the GATA gene.

1. What is the patient’s ABO, Rh type, and antibody screen?


2. Does the antibody appear to be auto or allo and why?
3. What is the suspected ethnicity of the patient based on the phenotype results?
4. Based on the phenotype results, what high-prevalence antigen should be considered?
5. What specialized testing could be used to determine the significance of the Fy(a-b-) phenotype?
6. If rare high incidence cells are not available to rule out additional allow antibodies, what technique could be
performed?
7. If an underlying anti-N was detected, should it be considered to be potent?

SUMMARY CHART
Blood Group Terminology Lewis Blood Group
 The ISBT terminology for RBC surface antigens pro-  Lewis blood group antigens are not synthesized by the
vides a standardized numeric system for naming RBCs. These antigens are adsorbed from plasma onto
authenticated antigens that is suitable for electronic the RBC membrane.
data-processing equipment. This terminology was not  The Le gene codes for L-fucosyltransferase, which adds
intended to replace conventional terminology. L-fucose to type 1 chains.
 In the ISBT classification, RBC antigens are assigned a  The Le gene is needed for the expression of Lea sub-
six-digit identification number: the first three digits stance, and Le and Se genes are needed to form Leb
represent the system, collection, or series, and the sec- substance.
ond three digits identify the antigen. All antigens are  The lele genotype is more common among blacks than
catalogued into one of the following four groups: among whites and results in the Le(a–b–) phenotype.
• A blood group system if controlled by a single gene  Lewis antigens are poorly expressed at birth.
locus or by two or more closely linked genes
• A collection if shown to share a biochemical, sero-  Lewis antibodies are generally IgM (naturally occur-
logic, or genetic relationship ring) made by Le(a–b–) individuals.
• The high-prevalence series (901) if found in more  Lewis antibodies are frequently encountered in preg-
than 90% of most populations nant women.
• The low-prevalence series (700) if found in less than  Lewis antibodies are not considered significant for
1% of most populations transfusion medicine.
Continued
6888_Ch08_173-211 29/10/18 5:27 PM Page 208

208 PART II Blood Groups and Serologic Testing

SUMMARY CHART—cont’d
The P Blood Group  Anti-S and anti-s are IgG antibodies, reactive at 37°C
and the antiglobulin phase. They may bind comple-
 The P blood group consists of the biochemically re-
ment and have been associated with HDFN and HTRs.
lated antigens P, P1, Pk, PX2, NOR, and LKE.
 P1 antigen expression is variable; P1 antigen is poorly
 The S–s–U– phenotype is found in blacks.
developed at birth.  Anti-U is usually an IgG antibody and has been asso-
ciated with HTRs and HDFN.
 Anti-P1 is a common naturally occurring IgM antibody
in the sera of P1– individuals; it is usually a weak, cold- The Kell Blood Group System
reactive saline agglutinin and can be neutralized with
soluble P1 substance found in hydatid cyst fluid.  The Kell blood group antigens are well developed at
birth and are not destroyed by enzymes.
 Anti-PP1Pk is produced by the rare p individuals early
in life without RBC sensitization and reacts with all  The Kell blood group antigens are destroyed by DTT,
RBCs except those of other p individuals. Antibodies ZZAP, and glycine-acid EDTA.
may be a mixture of IgM and IgG, efficiently bind com-  Excluding ABO, the K antigen is rated second only to
plement, may demonstrate in vitro hemolysis, and can D antigen in immunogenicity.
cause severe HTRs. Anti-PP1Pk is associated with  The k antigen is high prevalence.
spontaneous abortions.  Anti-K is usually an IgG antibody reactive in the
 Alloanti-P is found as a naturally occurring alloanti- antiglobulin phase and is made in response to preg-
body in the sera of Pk individuals and is clinically sig- nancy or transfusion of RBCs; it has been implicated
nificant. Anti-P has also been associated with in severe HTRs and HDFN.
spontaneous abortion.  The McLeod phenotype, affecting only males, is de-
 Autoanti-P is most often the specificity associated with scribed as a rare phenotype with decreased Kell system
the cold-reactive IgG autoantibody in patients with antigen expression. The McLeod syndrome includes
paroxysmal cold hemoglobinuria (PCH). the clinical manifestations of abnormal RBC morphol-
 The autoanti-P of PCH usually does not react by rou- ogy, compensated hemolytic anemia, and neurological
tine tests but is demonstrable as a biphasic hemolysin and muscular abnormalities. Some males with the
only in the Donath-Landsteiner test. McLeod phenotype also have the X-linked chronic
granulomatous disease.
The I and i Antigens
The Duffy Blood Group System
 I and i antigens are not antithetical; they have a recip-
rocal relationship.  Fya and Fyb antigens are destroyed by enzymes and
 Most adult RBCs are rich in I and have only trace ZZAP; they are well developed at birth. The Fy(a–b–)
amounts of i antigen. phenotype is prevalent in blacks but virtually nonex-
istent in whites.
 At birth, infant RBCs are rich in i; I is almost unde-
tectable; over the next 18 months of development, the  Fy(a–b–) RBCs resist infection by the malaria organism
infant’s RBCs will convert from i to I antigen. P. vivax.
 Anti-I is typically a benign, weak, naturally occurring,  Anti-Fya and anti-Fyb are usually IgG antibodies and
saline-reactive IgM autoagglutinin, usually detectable react optimally at the antiglobulin phase of testing;
only at 4°C. both antibodies have been implicated in delayed HTRs
and HDFN.
 Pathogenic anti-I is typically a strong cold autoagglu-
tinin that demonstrates high-titer reactivity at 4°C and The Kidd Blood Group System
reacts over a wide thermal range (up to 30° to 32°C).
 Patients with M. pneumoniae infections may develop  Anti-Jka and anti-Jkb may demonstrate dosage, are
strong cold agglutinins with autoanti-I specificity. often weak, and are found in combination with other
antibodies; both are typically IgG and reactive in the
 Anti-i is a rare IgM agglutinin that reacts optimally at
antiglobulin phase of testing.
4°C; potent examples may be associated with infec-
tious mononucleosis.  Kidd system antibodies may bind complement and are
made in response to foreign RBC exposure during
The MNS Blood Group System pregnancy or transfusion.
 Anti-M and anti-N are cold-reactive saline agglutinins
 Kidd system antibodies are a common cause of delayed
HTRs.
that do not bind complement or react with enzyme-
treated cells; both anti-M and anti-N may demonstrate  Kidd system antibody reactivity is enhanced with en-
dosage. zymes, LISS, and PEG.
6888_Ch08_173-211 29/10/18 5:27 PM Page 209

Chapter 8 Blood Group Terminology and Common Blood Groups 209

SUMMARY CHART—cont’d
The Lutheran Blood Group System response to foreign RBC exposure during pregnancy
or transfusion.
 Lua and Lub are antigens produced by codominant
alleles; they are poorly developed at birth.  The Lu(a–b–) phenotype is rare and may result from
three different genetic backgrounds; only individuals
 Anti-Lua may be a naturally occurring saline agglutinin
with the recessive type Lu(a–b–) can make anti-Lu3.
that reacts optimally at room temperature.
 Anti-Lub is usually an IgG antibody reactive at
the antiglobulin phase; it is usually produced in

7. A type 1 chain has:


Review Questions
a. The terminal galactose in a 1-3 linkage to subterminal
1. The following phenotypes are written incorrectly except N-acetylglucosamine
for: b. The terminal galactose in a 1-4 linkage to subterminal
N-acetylglucosamine
a. Jka+
c. The terminal galactose in a 1-3 linkage to subterminal
b. Jka+
N-acetylgalactosamine
c. Jka(+)
d. The terminal galactose in a 1-4 linkage to subterminal
d. Jk(a+)
N-acetylgalactosamine
2. Which of the following characteristics best describes
8. Which of the following best describes Lewis antigens?
Lewis antibodies?
a. The antigens are integral membrane glycolipids
a. IgM, naturally occurring, cause HDFN
b. Lea and Leb are antithetical antigens
b. IgM, naturally occurring, do not cause HDFN
c. The Le(a+b–) phenotype is found in secretors
c. IgG, in vitro hemolysis, cause hemolytic transfusion
d. None of the above
reactions
d. IgG, in vitro hemolysis, do not cause hemolytic trans- 9. Which of the following genotypes would explain RBCs
fusion reactions typed as group A Le(a+b–)?
3. The Le gene codes for a specific glycosyltransferase that a. A/O Lele HH Sese
transfers a fucose to the N-acetylglucosamine on: b. A/A Lele HH sese
c. A/O LeLe hh SeSe
a. Type 1 precursor chain
d. A/A LeLe hh sese
b. Type 2 precursor chain
c. Types 1 and 2 precursor chains 10. Anti-LebH will not react or will react more weakly with
d. Either type 1 or type 2 in any one individual but not which of the following RBCs?
both a. Group O Le(b+)
4. What substances would be found in the saliva of a group b. Group A2 Le(b+)
B secretor who also has Lele genes? c. Group A1 Le(b+)
d. None of the above
a. H, Lea
b. H, B, Lea 11. Which of the following best describes MN antigens and
c. H, B, Lea, Leb antibodies?
d. H, B, Leb a. Well developed at birth, susceptible to enzymes, gen-
5. Transformation to Leb phenotype after birth may be as erally saline reactive
follows: b. Not well developed at birth, susceptible to enzymes,
generally saline reactive
a. Le(a–b–) to Le(a+b–) to Le(a+b+) to Le(a–b+)
c. Well developed at birth, not susceptible to enzymes,
b. Le(a+b–) to Le(a–b–) to Le(a–b+) to Le(a+b+)
generally saline reactive
c. Le(a–b+) to Le(a+b–) to Le(a+b+) to Le(a–b–)
d. Well developed at birth, susceptible to enzymes, gen-
d. Le(a+b+) to Le(a+b–) to Le(a–b–) to Le(a–b+)
erally antiglobulin reactive
6. In what way do the Lewis antigens change during
12. Which autoantibody specificity is found in patients with
pregnancy?
paroxysmal cold hemoglobinuria?
a. Lea antigen increases only
a. Anti-I
b. Leb antigen increases only
b. Anti-i
c. Lea and Leb both increase
c. Anti-P
d. Lea and Leb both decrease
d. Anti-P1
6888_Ch08_173-211 29/10/18 5:27 PM Page 210

210 PART II Blood Groups and Serologic Testing

13. Which of the following is the most common antibody 22. A patient with an M. pneumoniae infection will most
seen in the blood bank after ABO and Rh antibodies? likely develop a cold autoantibody with specificity to
a. Anti-Fya which antigen?
b. Anti-k a. I
c. Anti-Jsa b. i
d. Anti-K c. P
d. P1
14. Which blood group system is associated with resistance
to P. vivax malaria? 23. Which antigen is destroyed by enzymes?
a. P a. P1
b. Kell b. Jsa
c. Duffy c. Fya
d. Kidd d. Jka
15. The null Ko RBC can be artificially prepared by which
References
of the following treatments?
a. Ficin and DTT 1. Issitt PD, Anstee, DJ. Applied blood group serology. 4th ed.
Durham (NC): Montgomery Scientific; 1998.
b. Ficin and glycine-acid EDTA 2. Garratty G, Dzik W, Issit PD, et al. Terminology for blood group
c. DTT and glycine-acid EDTA antigens and genes—historical origins and guidelines in the
d. Glycine-acid EDTA and sialidase new millennium. Transfusion. 2000;40:477-89.
3. Storry JR, Castilho L, Daniels G, et al. International Society of
16. Which antibody does not fit with the others with respect Blood Transfusion working party on red cell immunogenetics
to optimum phase of reactivity? and terminology: report of the Seoul and London meetings.
a. Anti-S ISBT Sci Ser. 2016;11:118-22.
4. isbtweb.org [Internet]. Amsterdam (Netherlands): International
b. Anti-P1 Society of Blood Transfusion [cited 2018 March]. Available from:
c. Anti-Fya http://www.isbtweb.org/working-parties/red-cell-immunogenetics-
d. Anti-Jkb and-blood-group-terminology/
5. Daniels, GL, Fletcher, A, Garratty, G, et al: Blood group termi-
17. Which of the following Duffy phenotypes is prevalent nology 2004: from the International Society of Blood Transfu-
in blacks but virtually nonexistent in whites? sion committee on terminology for red cell surface antigens.
a. Fy(a+b+) Vox Sang. 2004;87:304-16.
6. Fung MK, Eder AF, Spitalnik SL, Westhoff CM, editors. Tech-
b. Fy(a–b+) nical manual. 19th ed. Bethesda (MD): AABB; 2017.
c. Fy(a–b–) 7. Mourant AE. A “new” human blood group antigen of frequent
d. Fy(a+b–) occurrence. Nature. 1946;158:237-38.
8. Daniels G. The human blood goups. 3rd ed. Oxford: Blackwell
18. Antibody detection cells will not routinely detect which Science; 2013.
antibody specificity? 9. Grubb R. Correlation between Lewis blood group and secretor
a. Anti-M character in man. Nature. 1948;162:933.
10. Reid ME, Lomas-Francis C, Olsson ML. The blood group anti-
b. Anti-Kpa gen facts book. 3rd ed. San Diego (CA): Academic Press; 2012.
c. Anti-Fya 11. Levine P, Bobbitt OB, Waller RK, et al. Isoimmunization by a
d. Anti-Lub new blood factor in tumor cells. Proc Soc Exp Biol Med.
1951;77:403-5.
19. Antibodies to antigens in which of the following blood 12. Sanger, R. An association between the P and Jay systems of
groups are known for showing dosage? blood groups. Nature. 1955;176:1163-4.
a. I 13. Matson GA, Swanson J, Noades J, et al. A “new” antigen and
antibody belonging to the P blood group system. Am J Hum
b. P Genet. 1959;11:26-34.
c. Kidd 14. Arndt PA, Garratty G, Marfoe RA, et al. An acute hemolytic
d. Lutheran transfusion reaction caused by an anti-P1 that reacted at 37°C.
Transfusion. 1998;38:373-7.
20. Which antibody is most commonly associated with 15. Hellberg A, Poole J, Olsson ML. Molecular basis of the globo-
delayed hemolytic transfusion reactions? side-deficient Pk blood group phenotype. J Biol Chem.
a. Anti-s 2002;277:29455-9.
16. Yoshida H, Ito K, Kusakari T, et al. Removal of maternal anti-
b. Anti-k bodies from a woman with repeated fetal loss due to P blood
c. Anti-Lua group incompatibility. Transfusion. 1994;34:702-5.
d. Anti-Jka 17. Tippett P, Sanger R, Race RR, et al. An agglutinin associated
with the P and the ABO blood group systems. Vox Sang.
21. Anti-U will not react with which of the following RBCs? 1965;10:269-80.
a. M+N+S+s– 18. Storry JR, Castilho L, Chen Q, et al. ISBT International Society
b. M+N–S–s– of Blood Transfusion Working Party on red cell immunogenet-
ics and terminology: report of the Seoul and London meetings.
c. M–N+S–s+ ISBT Science Series. 2016;(11):118-22.
d. M+N–S+s+
6888_Ch08_173-211 29/10/18 5:27 PM Page 211

Chapter 8 Blood Group Terminology and Common Blood Groups 211

19. Lund N, Olsson ML, Ramkumar S, et al. The human Pk histo- 37. Bansal I, Jeon H, Hui SR, et al. Transfusion support for a patient
blood group antigen provides protection against HIV-1 infec- with McLeod phenotype without chronic granulomatous disease
tion. Blood. 2009;113:4980-91. and with antibodies to Kx and Km. Vox Sang. 2008;94:216-20.
20. Marsh WL, Jenkins WJ. Anti-i: a new cold antibody. Nature. 38. McGinnis MH, MacLowry JD, Holland PV. Acquisition of
1960;188:753. K:1-like antigen during terminal sepsis. Transfusion. 1984;
21. Yu L, Twu Y, Chang C, et al. Molecular basis of the adult i phe- 24:28-30.
notype and the gene responsible for the expression of the 39. Sanger R, Race RR, Jack J. The Duffy blood groups of New
human blood group I antigen. Blood. 2001;98:3840-5. York Negroes: the phenotype Fy(a–b–). Br J Haematol. 1955;
22. Yu L, Twu Y, Chou M, et al. The molecular genetics of the 1:370-4.
human I locus and molecular background explain the partial 40. Pogo AO, Chaudhuri A. The Duffy protein: a malarial and
association of the adult i phenotype with congenital cataracts. chemokine receptor. Semin Hematol. 2000;37:122-9.
Blood. 2003;101:2081-8. 41. Yazdanbakhsh K, Rios M, Storry JR, et al. Molecular mecha-
23. Curtain CC, Baumgarten A, Gorman J, et al. Cold haemagglu- nisms that lead to reduced expression of Duffy antigens. Trans-
tinins: unusual incidence in Melanesian populations. Br J fusion. 2000;40:310-20.
Haematol. 1965;11:471-9. 42. Colledge KI, Pezzulich M, Marsh WL. Anti-Fy5, an antibody
24. Booth PB, Jenkins WL, Marsh WL. Anti-IT: a new antibody of disclosing a probable association between the Rhesus and
the I blood group system occurring in certain Melanesian sera. Duffy blood group genes. Vox Sang. 1973;24:193-9.
Br J Haematol. 1966;12:341-4. 43. Allen FH, Diamond LK, Niedziela B. A new blood group
25. Garratty G, Petz LD, Walerstein RO, et al. Autoimmune antigen. Nature. 1951;167:482.
hemolytic anemia in Hodgkin’s disease associated with anti-IT. 44. Fernando M, Martinez-Cañabate S, Luna I, et al. Severe he-
Transfusion. 1974;14:226-31. molytic disease of the fetus due to anti-Jkb [letter]. Transfusion.
26. Greenwalt TJ, Sasaki T, Sanger R, et al. An allele of the S(s) 2008;48:402-403.
blood group genes. Proc Natl Acad Sci. 1954;40:1126-9. 45. Heaton DC, McLoughlin K. Jk(a–b–) red blood cells resist urea
27. Fukuda M. Molecular genetics of the glycophorin A gene clus- lysis. Transfusion. 1982;22:70-1.
ter. Semin Hematol. 1993;30:138-51. 46. Callender STE, Race RR. A serological and genetical study of
28. Palacajornsuk P. Review: molecular basis of MNS blood group multiple antibodies formed in response to blood transfusion
variants. Immunohematology. 2006;22:171-82. by a patient with lupus erythematosus diffusus. Ann Eugen.
29. Reid ME, Storry JR, Ralph H, et al. Expression and quantitative 1946;13:102.
variation of the low-incidence blood group antigen He on some 47. Cutbush M, Chanarin I. The expected blood-group antibody,
S–s– red cells. Transfusion. 1996;36:719-24. anti-Lub. Nature. 1956;178:855-6.
30. Darnborough J, Dunsford I, Wallace JA. The Ena antigen 48. Crawford MN, Greenwalt TJ, Sasaki T, et al. The phenotype
and antibody. A genetical modification of human red cells Lu(a–b–) together with unconventional Kidd groups in one
affecting their blood grouping reactions. Vox Sang. 1969;17: family. Transfusion. 1961;1:228-32.
241-55. 49. Eyler CE, Telen MJ. The Lutheran glycoprotein: a multifunc-
31. Furuhjelm U, Myllylä G, Nevanlinna HR, et al. The red cell tional adhesion receptor. Transfusion. 2006;46:668-77.
phenotype En(a-) and anti-Ena: serological and physiochemical 50. Karamatic Crew V, Green C, Daniels G. Molecular bases of the
aspects. Vox Sang. 1969;17:256-78. antigens of the Lutheran blood group system. Transfusion.
32. Jung HH, Danek A, Frey BM. McLeod syndrome: a neuro- 2003;43:1729-37.
haematological disorder. Vox Sang. 2007;93:112-21. 51. Singleton, BK, Burton, NM, Green, C, et al: Mutations in
33. Marsh WL, Nichols ME, Øyen R, et al. Naturally occurring EKLF/KLF1 form the molecular basis of the rare blood group
anti-Kell stimulated by E. coli enterocolitis in a 20-day-old In(Lu) phenotype. Blood. 2008;112:2081-8.
child. Transfusion. 1978;18:149-54. 52. Keller J, Vege S, Horn T, et al. Novel mutations in KLF1
34. Daniels G, Hadley A, Green CA. Causes of fetal anemia in encoding the In(Lu) phenotype reflect a diversity of clinical
hemolytic disease due to anti-K [letter]. Transfusion. 2003; presentations. Transfusion. 2018;58:196-99.
43:115-6. 53. Karamatic Crew, V, Mallinson, G, Green, C, et al: Different in-
35. Allen FH, Krabbe SMR, Corcoran PA. A new phenotype activating mutations in the LU genes of three individuals with
(McLeod) in the Kell blood group system. Vox Sang. the Lutheran-null phenotype. Transfusion. 2007;47:492-8.
1961;6:555-60. 54. Singleton, BK, Roxby, DJ, Stirling, JW, et al. A novel GATA1
36. Giblett ER, Klebanoff SJ, Pincus SH, et al. Kell phenotypes in mutation (Stop414Arg) in a family with the rare X-linked blood
chronic granulomatous disease: a potential transfusion hazard group Lu(a-b-) phenotype and mild macrothrombocytic
[letter]. Lancet. 1971;1:1235-6. thrombocytopenia. Br J Haematol. 2013;161:139-42.
6888_Ch09_212-231 31/10/18 9:48 AM Page 212

Chapter 9
Uncommon Blood Groups
Regina M. Leger, MSQA, MT(ASCP)SBB, CMQ/OE(ASQ)
and Angela K. Novotny MT(ASCP)SBBCM

Introduction The Gill (029) System ISBT 700 Series


The Diego (010) System The Rh-associated Glycoprotein (030) ISBT 901 Series
The Yt (011) System System Emm Antigen
The Xg (012) System The FORS (031) System AnWj Antigen
The Scianna (013) System The JR (032) System Sda Antigen
The Dombrock (014) System The LAN (033) System PEL Antigen
The Colton (015) System The VEL (034) System ABTI Antigen
The Landsteiner-Wiener (016) System The CD59 (035) System MAM Antigen
The Chido/Rodgers (017) System The Augustine (036) System HLA Antigens on RBCs
The Gerbich (020) System ISBT Blood Group Collections Applications to Routine Blood Banking
The Cromer (021) System Cost Collection 205 Case Studies
The Knops (022) System Ii Collection 207 Case 9-1
The Indian (023) System Er Collection 208 Case 9-2
The Ok (024) System Globoside Collection 209 Summary Chart
The Raph (025) System Collection 210 (Unnamed) Review Questions
The John Milton Hagen (026) System MN CHO Collection 213 References

OBJECTIVES
Antigen Characteristics
1. List the “uncommon” blood group systems with the appropriate gene and antigen prevalence, including the null phenotype
if applicable.
2. List the blood group collections, including antigen prevalence.
3. List the low- and high-prevalence antigens.
4. Identify different antigens that have varying prevalence within different ethnic groups.
5. Explain the prevalence of Dia with South, Central, and North American native populations and the consequent higher preva-
lence of the Di(b–) phenotype in those populations.
6. Describe the relationship between the GPA and Wrb expression.
7. Describe the RBC membrane defect seen in paroxysmal nocturnal hemoglobinuria (PNH) and how this relates to the Yt,
Dombrock, Cromer, CD59, and Emm antigens.
8. Describe the phenotypic relationship of Gy(a−) to the other phenotypes in the Dombrock system.
9. Describe the phenotypic relationship between LW and Rh.
10. List the Gerbich-negative phenotypes and the antibodies that can be made by each.
11. For the antigen described in this chapter, List the antigens that are denatured by routine blood bank enzymes and antigens
whose reactivity with antibody is enhanced with enzymes.

212
6888_Ch09_212-231 31/10/18 9:48 AM Page 213

Chapter 9 Uncommon Blood Groups 213

OBJECTIVES—cont’d
12. List which antigens are destroyed by treatment with DTT (dithiothreitol).
13. Explain how the age of RBCs affects the Knops antigens.
14. Describe the relationship between RHAG and the Rh antigens.
15. Explain the original hypothesis that the Forssman glycolipidis was an A subgroup.
16. Describe the Cost collections RBC expression variations between individuals and the subsequent difficulty in identifying
antibodies to these antigens.

Antibody Characteristics
17. List the characteristics of the antibodies to system, collection, and series antigens described in this chapter.
18. List the antibodies in the Dombrock system that are clinically significant for transfusion.
19. Describe how cord RBCs and DTT-treated RBCs can be used to differentiate anti-LW and anti-D.
20. Describe how pooled plasma can be used to aid in the identification of anti-Ch and anti-Rg.
21. Identify the common age group of individuals with anti-JMH and how it relates to antigen expression.
22. Describe the clinical significance of anti-Lan.
23. Describe the association between the immunoglobulin class, ability to activate complement, and in vitro and in vivo hemolysis
of anti-Vel.
24. Describe the importance of the antiglobulin crossmatch as it relates to low-prevalence antibodies.

Clinical Significance and Disease Association


25. Correlate the system, collection, and series antibodies with hemolytic transfusion reactions (HTR) and hemolytic disease of
the fetus and newborn (HDFN).
26. Identify which antigens are expressed on placental tissue and the role that plays in HDFN.
27. Describe the role that the carrier molecule for Knops (CR1) plays in processing immune complexes and pathogens for
clearance from the circulation.
28. Describe the relationship between MER2 antigen and renal disease.
29. Relate how age and expression of the JMH antigen relates to autoantibodies.
30. Explain the role of the Forssman antigen as it relates to pathogens.
31. Identify the antigen (AnWj) used by Haemophilus influenza as a receptor to enter RBCs.

Introduction The Diego (010) System


This chapter covers the “uncommon” blood group systems, The Diego system is composed of 22 antigens: three sets of
collections, and low- and high-prevalence series not cov- high and low pairs of antithetical antigens—Dia/Dib, Wra/Wrb,
ered in Chapter 8. Most of the information covered is con- and Wu/DISK—and 16 low-prevalence antigens.1–3 The
sidered advanced concepts and is used routinely only by an system was named after the first antibody maker in a
advanced immunohematology reference laboratory tech- Venezuelan family during an investigation of HDFN. The
nologist or individuals studying for the Specialist in Blood Diego system has the ISBT designation DI and the system
Bank Technology exam. With the advances in genomic test- number is 010.
ing, new antigens are continually being discovered and re- The Diego antigens are carried on band 3, a major integral
classified. Because most of the antigens discussed are either RBC membrane glycoprotein with about one million copies
of high or low prevalence, the corresponding antibodies de- per RBC. Band 3 is also known as the red blood cell anion
tected are rarely encountered. It is important for clinical exchanger (AE1) or solute carrier family-4 anion exchanger,
laboratorians to know that these “uncommon” antigens/ member 1 (SLC4A1). The protein crosses the membrane
antibodies exist and have a resource for information con- multiple times, and both the amino- and carboxyl-terminal
cerning their reactivity and clinical significance as they per- domains are in the cytoplasm. A large N-glycan on the fourth
tain to transfusion and hemolytic disease of the fetus and external loop carries over half the RBC A, B, H, and I blood
newborn (HDFN). By being aware of the “uncommon” group antigens. The long amino-terminal domain of band 3
antigen/antibodies, technologists will be better prepared interacts with ankyrin and protein 4.2 of the membrane
when they encounter one of these uncommon antibodies. skeleton. The gene encoding band 3 and the Diego antigens,
6888_Ch09_212-231 31/10/18 9:48 AM Page 214

214 PART II Blood Groups and Serologic Testing

SLC4A1, consist of 20 exons and is located on chromosome The Yt antigens are antithetical and represent an amino
17 at position 17q21.31.4 acid substitution on the glycosylphosphatidylinositol (GPI)-
Reported in 1955, anti-Dia had caused HDFN in a linked RBC glycoprotein acetylcholinesterase (AChE). AChE
Venezuelan baby. Anti-Dib was described 2 years later. Dia is is an important enzyme participating in neurotransmission,
rare in most populations but is polymorphic in people of but the function of RBC-bound AChE is not known. The
Mongoloid ancestry. In South American Indians, the preva- gene ACHE is located on chromosome 7 at position 7q22.4
lence of Dia can be as high as 54%.3 Dia is also present in the Yt antigens are variably sensitive to ficin and papain, are
North and Central American native populations but is sur- sensitive to DTT, and are resistant to glycine-acid EDTA
prisingly rare in Canada and among the Alaskan Inuit.2 The treatment. The antigens are developed at birth but are
prevalence of Dib is generally greater than 99% but is 96% expressed weaker on cord RBCs than on adult RBCs and are
in Native Americans. The Dia/Dib polymorphism is located absent from RBCs of people with paroxysmal nocturnal
on the last (seventh) extracellular loop of the protein. The hemoglobinuria (PNH) III.2
Di(a–b–) phenotype has not been reported. Anti-Yta and anti-Ytb are IgG and are stimulated by preg-
Wra, a low-prevalence antigen, and the antithetical, high- nancy or transfusion. Anti-Yta is not an uncommon antibody,
prevalence Wrb are associated with an amino acid substitu- so it appears that Yta is reasonably immunogenic. However,
tion on the fourth external loop, close to the insertion point Ytb appears to be a poor immunogen, as the antibody is rare.
of the protein into the RBC membrane. However, expression Yt antibodies have not caused HDFN. Some examples of
of Wrb requires the interaction of band 3 and normal GPA anti-Yta have been shown to be clinically significant for
of the MNS blood group system. GPA-deficient RBCs are transfusion whereas others have not.2 Monocyte phagocyto-
Wr(a–b–).2 sis assays (MMA) may be helpful in determining clinical sig-
Localization of Dia and Dib antigens to band 3 enabled nificance of anti-Yta.
many low-prevalence antigens to be assigned to the Diego
system: Wda, Rba, WARR, ELO, Wu, Bpa, Moa, Hga, Vga, Swa, The Xg (012) System
BOW, NFLD, Jna, KREP, Tra, Fra, and Sw1.
Diego antigens are expressed on RBCs of newborns. The Anti-Xga was discovered in 1962 in the serum of a multiply
antigens are resistant to treatment with ficin and papain, transfused man. The antibody detected an antigen with a
DTT, and glycine-acid EDTA, with the exception of Bpa, higher prevalence in females than in males. Family studies
which is sensitive to papain.2,3 were used to confirm the antigen Xga expression was con-
Diego system antibodies are sometimes IgM, but are usu- trolled by an X-linked gene. The antigen was named after
ally IgG, reactive in the antiglobulin phase of testing. Both the X chromosome and g for “Grand Rapids,” where the pa-
anti-Dia and anti-Dib have caused hemolytic transfusion re- tient was treated.3
actions (HTRs) and HDFN.2,3 Anti-Wra is a relatively com- The Xg system is designated by the symbol XG and num-
mon antibody in donors and patients; some are directly ber 012. There are two antigens in the Xg system: Xga and
agglutinating, but most require the antiglobulin phase of CD99. The gene XG encoding Xga is located on the X chro-
testing to be detected. Anti-Wra has caused severe HTRs. mosome at position Xp22.32.4 The gene responsible for
Only a few examples of alloanti-Wrb in individuals with CD99, MIC2, is located on the X chromosome at Xp22.2 and
Wr(a–b–) RBCs have been described, so information about on the Y chromosome at Yp11. CD99 became part of the Xg
clinical significance is insufficient. Autoanti-Wrb is relatively system because the MIC2 and XG genes are adjacent and ho-
common in the serum of patients with warm autoimmune mologous and two CD99– Japanese individuals were found
hemolytic anemia. Little or no data are available on the clin- with anti-CD99. Xga has a phenotypic relationship to CD99:
ical significance of antibodies to the low-prevalence Diego all Xg(a+) individuals have a high expression of CD99 on
antigens, with the exception of anti-ELO, which has caused their RBCs, and all Xg(a–) females have a low expression of
severe HDFN.2 CD99; however, 68% of Xg(a–) males have a high expression
and 32% have a low expression of CD99.2 Both Xga and CD99
The Yt (011) System escape X chromosome inactivation. The Xg glycoprotein
crosses the RBC membrane once, with the amino terminus
Two antigens make up the Yt system, which was named in directed externally. There are approximately 9,000 copies of
1956 for the first antibody maker and used the last letter Xga per RBC.3
“t” in the patient’s last name, which was Cartwright.5 Ap- The prevalence of Xga is 66% in males and 89% in females.
parently “why T” became “Yt.”3 Yta is the high-prevalence Because males have only one X chromosome, Xg(a+) males
antigen in all populations; Ytb is the low-prevalence antigen are hemizygotes. Females, having two X chromosomes, can
found in about 8% of whites and 21% to 26% of Israelis, be homozygotes or heterozygotes. However, homozygosity
but it is not found in Japanese.2,3 Three phenotypes are ob- for the gene does not directly correlate to RBC antigen
served: the common Yt(a+b–) and Yt(a+b+) and the rare strength.6 Cord RBCs express Xga weakly. Weak expression
Yt(a–b+). The Yt(a–b–) phenotype has not been reported. of Xga is seen on RBCs from some adult females, but weak
The Yt system has the ISBT designation YT and system expression on RBCs from adult males is rare.3 The antigen is
number 011. sensitive to ficin and papain but resistant to DTT treatment.
6888_Ch09_212-231 31/10/18 9:48 AM Page 215

Chapter 9 Uncommon Blood Groups 215

Anti-Xga is usually IgG; some examples are naturally The high-prevalence antigens Gya and Hy were both de-
occurring. Anti-Xga has not been implicated in HDFN or scribed in 1967. RBCs from whites who are Gy(a–) were
as a cause of HTRs. Only a few examples of anti-CD99 found to be Hy–, but Hy– RBCs from blacks were weakly
have been reported so little is known about the clinical Gy(a+). The high-prevalence antigen Joa was described in
significance. 1972, and the phenotypic relationship to Gya and Hy was
later shown: Gy(a–) RBCs or Hy– RBCs are also Jo(a–). In
The Scianna (013) System 1995, it was reported that Gy(a–) RBCs were also Do(a–b–).9
The Gy(a–) phenotype is the Dombrock null. The Dombrock
The Scianna blood group system, ISBT symbol SC and num- system currently has five additional high-prevalence antigens,
ber 013, currently consists of seven antigens. The system is DOYA, DOMR, DOLG, DOLC, and DODE.4 Two rare
named after the first antibody maker. In 1962, a new high- phenotypes in blacks (and not found in whites) are Hy–
prevalence antigen was named Sm; 1 year later, a new low- Jo(a–) and Hy+w Jo(a-)
prevalence antigen, Bua, was found. After it was confirmed The Dombrock antigens are carried on a mono-ADP-
that these two antigens were antithetical, the Scianna blood ribosyltransferase 4 (ART4) attached to the RBC membrane
group system was established in 1974, and the two antigens by a GPI anchor. The gene ART4 encoding the Dombrock
were renamed Sc1 and Sc2, respectively. The prevalence glycoprotein is located on chromosome 12 at position
of Sc2 in Northern Europeans is 1% but is higher in the 12p13-p12.4
Mennonite population.3 The Dombrock antigens are resistant to ficin, papain, and
In 1980, an individual in the Marshall Islands in the South glycine-acid EDTA and are sensitive to 0.2 M DTT treatment.
Pacific was found with the Sc:–1,–2 phenotype. He had made The antigens are present on cord RBCs, but are absent from
an antibody to an unknown high-prevalence antibody. PNH III RBCs. The Doa and Dob antigens are poor immuno-
The antibody was named anti-Sc3; separable anti-Sc1 and gens and the antibodies are rarely found as single specifici-
anti-Sc2 were not identified. The very rare Sc:–1,–2,–3 phe- ties; Gya, however, is highly immunogenic.3 Due to the
notype is the Scianna null type. Three examples of anti-Sc3, scarcity of anti-Doa and anti-Dob as single specificities,
nonreactive with Sc:–1,–2, were found to be incompatible molecular testing is used to predict RBC antigen status.
with the RBCs of the other anti-Sc3 makers, indicating the Anti-Doa and anti-Dob have caused delayed HTRs but no
existence of additional high-prevalence antigens.7 clinical HDFN. The Dombrock antibodies are usually IgG,
The SC gene ERMAP is located on chromosome 1 at po- reacting optimally with enzyme-treated RBCs. These anti-
sition 1p34.2.4 The product of the gene is a protein called bodies are usually weakly reactive and disappear, both fac-
erythroid membrane-associated protein (ERMAP), which is tors making them difficult to identify.
an RBC adhesion protein. Once location of Scianna to
ERMAP was found, other antigens were assigned to the The Colton (015) System
system. The low-prevalence antigen Rd became Sc4. Sc5
(STAR), Sc6 (SCER), and Sc7 (SCAN) are all high-prevalence The Colton blood group system, ISBT symbol CO and num-
antigens.8 ber 015, consists of four antigens. The system was named in
The Scianna antigens are resistant to ficin and papain.The 1967 for the first antibody maker; it should have been named
antigens are also resistant to DTT treatment, except for Sc2, Calton, but the handwriting on the tube was misread!3 The
which has variable reactivity after DTT-treatment.3 The anti- high- and low-prevalence antithetical antigens are Coa and
gens are expressed on cord RBCs. Cob, respectively. The Cob antigen is present in about 10%
Alloantibodies to Scianna antigens are rare, and little is of most populations.3 The third antigen, Co3, is present on
known about their clinical significance. They are usually IgG all RBCs except those of the very rare Co(a–b–) phenotype.
and reactive in the antiglobulin phase of testing. None have Co4, a high-prevalence antigen, has been identified on RBCs
been reported to cause a severe HTR. Only mild HDFN has from two individuals with the Co(a–b–) phenotype.10
been reported, except for one severe case for anti-Rd, for The Colton antigens are carried on an integral membrane
which the baby required exchange transfusion. Autoantibod- protein, aquaporin 1 (AQP1), which accounts for 80% of
ies to Sc1 and Sc3 have been reported. water reabsorption in the kidneys.3 The glycoprotein crosses
the RBC membrane multiple times. The gene AQP1 is located
The Dombrock (014) System on chromosome 7 at position 7p14.4 The Colton antigens
are expressed on RBCs of newborns and are resistant to treat-
The Dombrock blood group system, designated by the ISBT ment with ficin and papain, chloroquine, and DTT.
with the symbol DO and number 014, was named for the Antibodies are usually IgG and are enhanced with enzyme-
first antibody maker, Mrs. Dombrock (anti-Doa), found in treated RBCs. Anti-Coa is often seen as a single specificity
1965. Anti-Dob, which recognizes the antithetical antigen, and has been reported to cause HTRs and HDFN.11 Anti-Cob
was identified in 1973. The prevalence of the three resulting appears more often with other specificities but has also
phenotypes, Do(a+b–), Do(a+b+), and Do(a–b+), varies in caused HTRs and mild HDFN.3 Anti-Co3, which reacts with
different populations. In whites, they are 18%, 49%, and all Co(a+) and Co(b+) RBCs, has been reported to cause mild
33%, respectively. HTR and severe HDFN.3,12
6888_Ch09_212-231 31/10/18 9:48 AM Page 216

216 PART II Blood Groups and Serologic Testing

The Landsteiner-Wiener (016) System and not at all with Rhnull RBCs. A weak anti-LW may react
only with D+ RBCs and may appear to be anti-D unless en-
The Landsteiner-Wiener blood group system, ISBT symbol hancement techniques are used. This is because there are
LW and number 016, had its origins along with the discovery more LW antigen sites on D+ RBCs from adults than on
of the D antigen of the Rh blood group system. In 1940, D– RBCs.3 However, anti-LW reacts equally well with cord
Landsteiner and Wiener reported that an antibody produced RBCs regardless of their D type. Distinguishing anti-LW from
in rabbits (and later, guinea pigs) after injection with RBCs anti-D is most easily accomplished by testing DTT-treated
of rhesus monkeys reacted with 85% of human RBCs.13 This D+ RBCs: the D antigen is not denatured by DTT, so anti-D
antibody was called anti-Rh for anti-rhesus. The reactivity would still be detected; however, LW antigen is destroyed by
of anti-Rh was similar to the human antibody reported DTT, so anti-LW would no longer react. LW antigens are
by Levine and Stetson in 1939 in a woman who delivered a resistant to treatment of RBCs with enzymes and glycine-
stillborn infant and had a transfusion reaction to the blood acid EDTA.
donated by her husband.14 Both sera identified the same The structure that carries the LW antigens is a glycopro-
population of Rh+ and Rh– RBCs. The two antibodies were tein known as intracellular adhesion molecule 4 (ICAM-4),
later shown to be different in several studies; in the 1960s, a member of the immunoglobulin superfamily. The LW
the human anti-Rh was renamed anti-D (but called anti-Rho glycoprotein is part of the band 3/Rh macrocomplex;15 Rhnull
by some workers), and the rabbit anti-Rh was called anti-LW RBCs lack the LW glycoprotein. The LW gene ICAM4 is
in honor of Landsteiner and Wiener. Examples of human located on chromosome 19 at position 19p13.2.4
anti-LW were subsequently described. LW antigens may be depressed during pregnancy and in
There are three LW antigens: LWa, LWab, and LWb. The some diseases, such as lymphoma and leukemia.3 Autoanti-
first two, LWa and LWab, are common, high-prevalence anti- LW made by these patients can appear to be an alloantibody;
gens, and LWb is of low prevalence, found in less than 1% as the antigen strength returns, the antibody diminishes.
of most Europeans but in 6% of Finns.3 LWab was originally Autoanti-LW is also common in serum from patients with
defined by the antibody made by an individual with an in- warm autoimmune hemolytic anemia. No anti-LW has been
herited LW(a–b–) phenotype. Terminology for LW antigens shown to cause serious HDFN or transfusion reactions.
has evolved as more information became available. The ISBT Many patients with anti-LW have successfully been trans-
has used LW5, LW6, and LW7 as the antigen numbers for fused with D– RBCs. Very few examples of alloanti-LWab
LWa, LWab, and LWb, respectively, to prevent confusion with have been described, e.g., the antibodies of the only known
obsolete terminology that used LW1-LW4 to designate phe- propositi with an inherited LW(a–b–) phenotype.2
notypes. The null phenotype is LW(a–b–); in one individual
who made anti-LWab (Mrs. Big), this phenotype resulted The Chido/Rodgers (017) System
from a 10-base pair deletion in exon 1 of an LWa gene,
which introduced a premature stop codon. Rhnull RBCs also The Chido/Rodgers blood group system, designated by the
type LW(a–b–) and are considered to be the only true LW– ISBT with symbol CH/RG and number 017, was named after
RBCs because they fail to elicit the formation of anti-LW in the first two antibody producers, Ch for Chido and Rg for
animals, whereas injection of Mrs. Big’s LW(a–b–) RBCs into Rodgers. These two antibodies were described in 1967 and
guinea pigs caused the formation of anti-LW. The LW phe- 1976, respectively. Serologically, they were both character-
notypes are shown in Table 9–1. ized as nebulous because antigen strength on different sam-
As was shown by the similarity in reactivity of the original ples of RBCs was variable.12 It was also appreciated that both
animal anti-Rh and the human anti-Rh (later called anti-D), anti-Ch and anti-Rg could be neutralized by plasma.
there is a phenotypic relationship between the D antigen and Ch and Rg antigens are not intrinsic to the RBC mem-
anti-LW. Anti-LW usually reacts strongly with D+ RBCs, brane. Rather, they are on the fourth component of comple-
weakly (and sometimes not at all) with D– RBCs from adults, ment (C4), and are adsorbed onto RBCs from plasma.12 The

Table 9–1 LW Phenotypes


Phenotype Reactivity With Prevalence
Anti-LWa Anti-LWb Anti-LWab Most Europeans Finns

LW(a+b–) + – + 97% 93.9%

LW(a+b+) + + + 3% 6%

LW(a–b+) – + + Rare 0.1%

LW(a–b–) Big – – –

LW(a–b–) Rhnull – – –
6888_Ch09_212-231 31/10/18 9:48 AM Page 217

Chapter 9 Uncommon Blood Groups 217

C4 glycoprotein has two isoforms: C4B carries the Ch anti- Americans, Japanese, and Polynesians.2,3 Few individuals
gens, and C4A expresses Rg antigens. The genes C4A and with the Ge:–2,–3,–4 phenotype have been reported; all were
C4B at two closely linked loci located on chromosome 6 at English or North American. The RBCs of Ge null phenotype
position 6p21.3 encode these isoforms.4 The Chido/Rodgers are elliptocytic and these individuals may have a mild
system consists of nine antigens: Ch1 to Ch6, Rg1, and Rg2 anemia.2
are all high prevalence; WH has a prevalence of about 15%.3 Gerbich antigens are expressed at birth. RBCs of the
Differentiation of the determinants is not made for routine Gerbich or Leach phenotypes have weak expression of Kell
serology. blood group antigens, and some anti-Vel fail to react with
Ch is present in 96% to 98% of most populations.3,12 Rg Ge:–2,–3,4 RBCs.3 Gerbich antigens are resistant to treat-
is present in 97% to 98% of most populations.3,12 The anti- ment with DTT and glycine-acid EDTA; Ge2 and Ge4 are
gens are destroyed by ficin and papain but are resistant to ficin and papain sensitive, but Ge3 is ficin resistant.
treatment with DTT and glycine-acid EDTA.3 Some Gerbich antibodies may be IgM, but most are IgG.
Anti-Ch and anti-Rg are usually IgG and react weakly, Gerbich antibodies are sometimes clinically significant for
often to moderate or high titration endpoints. Neutralization transfusion and sometimes not. Gerbich antibodies can be
of anti-Ch and anti-Rg with pooled plasma is often used as eluted from DAT+ cord bloods, but only three cases of
part of the identification of these antibodies in a patient’s serious HDFN due to anti-Ge3 have been reported, and two
serum. Anti-Ch and anti-Rg are clinically insignificant for were children of the same mother.16,17 In these cases, the
transfusion.12 severe anemia was late onset, after birth, associated with in-
hibition of erythroid cell growth; in one case there was also
The Gerbich (020) System early onset of hemolysis. Therefore, babies born to mothers
with anti-Ge3 should be monitored for several weeks after
The Gerbich blood group system was named in 1960 after birth for anemia.
Mrs. Gerbich, the first antibody producer. Gerbich became Anti-Ge2 is the most common of the Gerbich antibodies.2
a system in 1990, designated by the ISBT as GE and num- It is the antibody made by the Ge:–2,3,4 phenotype individ-
ber 020. There are currently six high-prevalence Gerbich uals, but it is also the more common antibody made by the
antigens (Ge2, Ge3, Ge4, GEPL, GEAT, and GETI) and five Ge:–2,–3,4 phenotype and Ge:–2,–3,–4 phenotype individ-
low-prevalence antigens (Wb, Lsa, Ana, Dha, and GEIS). uals. Anti-Ge3 is less frequently made by the Ge:–2,–3,4 and
The antigens are carried on sialoglycoprotein structures Ge:–2,–3,–4 phenotype individuals. Anti-Ge4 is a very rare
GPC and GPD. The glycoproteins help to maintain the RBC antibody.
membrane integrity through interaction with protein band
4.1 and p55, and because they are rich in sialic acid, they The Cromer (021) System
contribute to the net negative charge of the RBC membrane
(as do GPA and GPB of the MNS system). There are about In 1965, an antibody was found in a black prenatal patient,
135,000 copies of GPC and 50,000 copies of GPD per Mrs. Cromer, that reacted with all RBCs except her own and
RBC.3 GPC and GPD are both encoded by the GYPC gene, two siblings. It was originally thought her antibody recog-
located on chromosome 2 at position 2q14.3. nized an antigen antithetical to Goa of the Rh blood group
There are three Gerbich-negative phenotypes in which the system. The antibody was named anti-Cra in 1975.2,3
RBCs lack one or more of the high-prevalence antigens: The antigens of the Cromer system are carried on decay-
Ge:–2,3,4 (Yus type), Ge:–2,–3,4 (Gerbich type), and accelerating factor (DAF, CD55), a complement-regulatory
Ge:–2,–3,–4 (Leach type). The Leach type is the Gerbich null protein. The gene CD55 is located on chromosome 1 at
phenotype. These are summarized in Table 9–2. Outside of position 1q32.4 The glycoprotein encoded by the gene is
Papua, New Guinea, and Melanesia, the Gerbich-negative attached to the RBC membrane through a GPI linkage.
phenotypes are very rare, but they have been found in PNH III RBCs are deficient in DAF so they also lack Cromer
diverse populations. The Yus phenotype has been found in antigens.
Mexicans, Israelis, Mediterraneans, and others but has not The Cromer system has 19 antigens listed on the current
been found in Papua, New Guinea.2,3 The Gerbich pheno- ISBT website.4 Of these, 15 have been classified as high-
type is polymorphic in certain areas of Papua, New Guinea, prevalence antigens and 3 low-prevalence antigens.18,19
and has also been found among Europeans, Africans, Native All these antigens are absent from Inab phenotype RBCs,
the Cromer null phenotype, which is very rare. CROK
(CROM19) appears to be a novel Inab-like phenotype with a
Table 9–2 Gerbich-Negative Phenotypes mutation that affects the expression of the Cromer antigens.20
The Cr(a–) phenotype is typically found in blacks and is not
Type Antibody found in whites. Three Cromer antigens, Tca, Tcb, and Tcc, are
Ge: –2, 3, 4 Yus Anti-Ge2 antithetical; Tca is the high-prevalence antigen and the
other two are low prevalence. WESa (low prevalence) and
Ge: –2, –3, 4 Gerbich Anti-Ge2 or anti-Ge3 WESb (high prevalence) are also antithetical.3 The Dra
Ge: –2, –3, –4 Leach type Anti-Ge2 or anti-Ge3 antigen is of high prevalence; Dr(a–) RBCs have weakened
expression of all other high-prevalence Cromer antigens due
6888_Ch09_212-231 31/10/18 9:48 AM Page 218

218 PART II Blood Groups and Serologic Testing

to a markedly reduced copy number of DAF. The Dr(a–) phe- The Indian (023) System
notype has been found only among Jews from Bakhara and
in Japanese.3 The Indian blood group system was named because the first
Cromer system antigens are resistant to treatment with In(a+) individuals were from India. There are now five anti-
ficin and papain but are destroyed by α-chymotrypsin, which gens in the system, designated IN and number 023 by the
is used to distinguish specificities in this system from other ISBT. The antigen Ina was reported in 1973 and is present
blood group antibodies. Cromer antigens are weakened with on RBCs of 4% of Indians, 11% of Iranians, and 12% of
DTT treatment and are resistant to glycine-acid EDTA. Arabs.3 Inb is the antithetical high-prevalence antigen. These
Antibodies in the Cromer system are usually IgG, but do and three other high-prevalence antigens are located on
not cause HDFN. DAF is strongly expressed on placental CD44, an adhesion molecule. The gene CD44 is located on
tissue and will adsorb Cromer antibodies.3,18,19 Anti-Cra and chromosome 11 at position 11p13.4 The extremely rare
anti-Tca have been implicated in HTRs, but other examples In(a–b–) phenotype has been found in only one individual
of these specificities have not caused clinical reactions after who presented with congenital dyserythropoietic anemia
transfusion of incompatible units.18 Anti-IFC is the antibody and whose RBCs also typed Co(a–b–).22
made by individuals with the Cromer null Inab phenotype. CD44 is reduced on RBCs of dominant type Lu(a–b–) in-
dividuals. Ina and Inb are weakly expressed on cord RBCs
The Knops (022) System and are sensitive to treatment with ficin, papain, and DTT
but are resistant to glycine-acid EDTA.3
There are nine antigens in the Knops blood group system, Antibodies are usually IgG and reactive in the antiglobu-
designated by the ISBT as KN and number 022. The system lin phase of testing and do not bind complement. Positive
was established when the antigens were shown to be located DATs but no clinical HDFN have been reported for anti-Ina
on complement receptor 1 (CR1) and was named after and anti-Inb; decreased cell survival with anti-Ina and an
Mrs. Knops, the first antibody maker. The gene CR1 is immediate HTR due to anti-Inb have been reported.3
located on chromosome 1at position 1q32.2.4
Except for the low-prevalence antigens Knb and McCb, The Ok (024) System
the Knops antigens have a prevalence of more than 90% in
most populations; however, ethnic differences exist. Sla is Currently, there are three high-prevalence antigens in the Ok
present on RBCs of only about 60% of African Americans. system, designated by the ISBT symbol OK and number 024.
Among West Africans, Sla has a prevalence of 30% to 38%, Anti-Oka was identified in 1979 and was named after the
and KCAM has a prevalence of only 20%.21 The antithetical antibody maker, Mrs. Kobutso. Because Ko was already in use,
pairs of antigens are Kna and Knb, McCa and McCb, and Sla the first two letters were switched to Ok.23 Her parents were
and Vil. Knops antigens are weakly expressed on cord RBCs cousins from a small Japanese island. Two of three siblings of
and weaken upon storage of adult RBCs (e.g., older units of the proposita were also Ok(a–).2 RBCs from 400 individuals
blood). The antigens are weakened by treatment with ficin from the same island were all Ok(a+).12 Two additional anti-
and papain and are destroyed by DTT; the antigens are re- gens, OKGV and OKVM, received their names because of the
sistant to glycine-acid EDTA.3 amino acid changes G to V and V to M, respectively.
Serologically, these antigens have been grouped together The OK antigens are carried on CD147, or basigin, a mem-
because their corresponding antibodies demonstrate variable ber of the immunoglobulin superfamily that mainly functions
reactions, are not neutralized by pooled normal serum (un- as receptors and adhesion molecules. Basigin is a receptor
like anti-Ch and anti-Rg), and are difficult to adsorb and essential for invasion by Plasmodium falciparum.3 The OK
elute.21 Antibody reactivity is enhanced with longer incuba- gene BSG is on chromosome 19 at position 19p13.3.4
tion (e.g., 1 hour, at 37°C). Weak and variable reactivity is Oka is well developed on RBCs from newborns and is
due to variable expression of CR1 on different samples of resistant to treatment with ficin and papain, DTT, and
RBCs. The “Helgeson phenotype” represents the serologic glycine-acid EDTA.3
null phenotype for the Knops blood group; these RBCs type The original anti-Oka was IgG, reactive in the antiglobulin
Kn(a–b–), McC(a–), Sl(a–), and Yk(a–) because of the low phase of testing. At least one other example of the antibody
copy number of CR1, but they are not truly devoid of Knops has been found. The antibody caused reduced survival of
antigens.3,21 Cr-labeled Ok(a+) RBCs injected into the original antibody
The antibodies are usually IgG, reactive in the antiglobu- maker, suggesting clinical significance; an in vitro monocyte-
lin phase of testing, but are clinically insignificant for both monolayer assay also predicted clinical signficance.12 There
transfusion and HDFN. Of the Knops antibodies, anti-Kna is have been no reports of HDFN caused by anti-Oka, but this
frequently found in multiply transfused individuals and mul- may be due to the rarity of the antibody.
tispecific sera; anti-Sla is more frequently found in blacks.
CR1 binds the complement component fragments C3b The Raph (025) System
and C4b and processes immune complexes for transporta-
tion to the liver and spleen and subsequent clearance from The only antigen in the Raph system is MER2, which was
the circulation.2,21 CR1 has also been identified as a receptor originally defined by two monoclonal antibodies; it has
for several pathogenic organisms. since been recognized by human polyclonal antibodies. The
6888_Ch09_212-231 31/10/18 9:48 AM Page 219

Chapter 9 Uncommon Blood Groups 219

antigen name is derived from monoclonal, and Eleanor phenotypes. Rather, the patient’s JMH– status is acquired and
Roosevelt, the laboratory where the antibody was produced. can be transient.12 It is widely accepted that JMH levels de-
When MER2 was raised to system status, the system was cline during the later years of life, sometimes to the point of
named Raph for the first patient to make the alloanti-MER2. not being detected serologically.12 Once JMH expression is
MER2 is encoded by the gene CD151 located on chromo- reduced, anti-JMH can be made. In some cases with anti-
some 11 at position 11p15.5.4 JMH, the DAT is positive and some JMH is detected on the
Three examples of alloanti-MER2 were found in Jews patient’s RBCs. This autoanti-JMH with a positive DAT has
originating from India and living in Israel; two were related. never been associated with autoimmune RBC destruction.12
All three had end-stage renal disease. A fourth example of JMH is weakly expressed on cord RBCs and is destroyed
anti-MER2 was found in a healthy Turkish blood donor. by treating RBCs with ficin and papain and DTT; the antigen
Blood samples from these four individuals were studied. It is resistant to treatment with glycine-acid EDTA.3
was shown that MER2 is located on CD151, a tetraspanin, Anti-JMH is usually IgG (predominantly IgG4 in acquired
which appears to be essential for the assembly of basement JMH-negative people).3 The antibodies are often high titer
membranes in the kidney and skin.24 but weakly reactive, even when tested without dilution, and
The polymorphism identified by the monoclonal antibod- they are not neutralized with pooled plasma. JMH antibodies
ies indicated that 8% of the English blood donor population are generally considered clinically insignificant. Rare exam-
is MER2–. Subsequent studies suggest the antigen-negative ples of alloanti-JMH in individuals whose RBCs express vari-
status represents the low end of antigen expression on ant forms of CD108 may be clinically significant.12
RBCs.24 MER2 is abundant on platelets and is expressed
on erythroid precursors of individuals with either MER2+ The Gill (029) System
or MER2– RBCs. MER2 expression decreases over time with
increasing maturation of erythroid cells.24 It has been sug- Anti-GIL was first identified in 1980, but the antigen was not
gested that most people whose RBCs type MER2– have raised to system status until 2002 when it was shown that
MER2 expressed on other cells and tissues and will not make GIL is genetically discrete from all other blood group systems.
anti-MER2, and those individuals who have made alloanti- The ISBT Gill system symbol is GIL and number 029. There
MER2 are true MER2–. Those individuals who have made is only one antigen, GIL.
anti-MER2 showed mutations in CD151. In the patients with This antigen is found on the glycerol transporter aqua-
renal disease, a truncated CD151 protein would be predicted, porin 3 (AQP3), a member of the major intrinsic protein fam-
but in patients without renal disease, mutations would result ily of water and glycerol channels. The gene AQP3 is located
in an altered CD151 that appears to be present and func- on chromosome 9 at position 9p13.4 The GIL– phenotype
tional at the cell surface.24,25 results from a frameshift and a premature stop codon.
The antigen is resistant to treatment with ficin and papain Reactivity with anti-GIL is enhanced with ficin and
but is sensitive to treatment with trypsin, a-chymotrypsin, papain treatment of RBCs; the antigen is resistant to DTT
pronase, and AET. and glycine-acid EDTA treatment.
Little is known about the clinical significance of anti- RBCs of two babies born to mothers with anti-GIL had a
MER2, but one patient with the antibody showed signs of a positive DAT but no clinical HDFN.2 One example of anti-
transfusion reaction after transfusion with 3 units. As 8% of GIL was found following a hemolytic transfusion reaction.
the population is expected to have MER2– RBCs, it would
be prudent to transfuse with crossmatch-compatible units.25 The Rh-Associated Glycoprotein (030)
System
The John Milton Hagen (026) System
The Rh-associated glycoprotein system symbol is RHAG and
JMH is a high-prevalence antigen. Numerous examples of anti- number 030.1 The Rh-associated glycoprotein (RhAG) does
JMH have been seen, especially in patients 50 years and older. not have Rh blood group antigens; however, its presence in
In 1978, a large number of samples with this antibody was a complex with the Rh proteins is essential for Rh antigen
characterized and the antibody was named anti-JMH for the expression. Absence of RhAG due to inactivating mutations
first antibody maker, John Milton Hagen. The system (ISBT in RHAG results in the Rhnull phenotype; some missense
symbol JMH and number 026) was established after it was mutations in RHAG result in the Rhmod phenotype.27 Unlike
shown that the JMH protein is the GPI-linked glycoprotein the RhD and RhCcEe proteins, RhAG is glycosylated on the
CD108 and the gene SEMA7A was cloned. The gene SEMA7A first extracellular loop, carrying ABH antigens. RhAG is
is located on chromosome 15 at position 15q22.3-q23.4 encoded by the gene RHAG, located on chromosome 6 at
Five other antigens were added to the system, JMH2 position 6p12.3.4
through JMH6; these are JMH variants associated with amino Four antigens have been assigned to the RHAG system:
acid substitutions in the protein.26 JMH1 represents the anti- Duclos (high prevalence), Ola (very rare, low prevalence),
gen recognized by antibodies made by individuals lacking the high prevalence DSLK (for Duclos-like), and RHAG4.
the JMH protein. Homozygosity for the allele encoding Ola is associated
Most examples of anti-JMH are not found in patients who with the Rhmod phenotype. Absence of Duclos and DSLK is
lack the JMH protein or who have one of the variant JMH associated with U– Rhnull or U– Rhmod phenotypes.
6888_Ch09_212-231 31/10/18 9:48 AM Page 220

220 PART II Blood Groups and Serologic Testing

Clinical significance of anti-Duclos, anti-Ola and anti- anti-Jra have received Jr(a+)-incompatible units for transfu-
DSLK is unknown because of the rarity of the antibodies. sion and have had no ill effect, but in other cases, incompat-
Anti-RHAG4 has caused a single case of severe HDFN, but ible transfusions have resulted in HTRs.
significance for transfusion is unknown.3
The LAN (033) System
The FORS (031) System
Lan was first named in 1961 after the first antigen-negative
In 1987, the Forssman glycosphingolipid was first thought proband, Langereis, who made anti-Lan. The Lan antigen, a
to be a subgroup of A called Apae. In 2011, it was shown to high-incidence antigen, was raised from the 901 series of
be unrelated to the ABO system and renamed FORS in honor high-incidence antigens to system status in 2012 when the
of John Forssman, who first discovered the antigen. The ISBT ABCB6-null allele was demonstrated with the Lan– pheno-
symbol is FORS and the number 31; there is only one antigen, type. The ISBT Lan system symbol is LAN and number 033.
FORS1. The gene GBGT1 is located on chromosome 9 There is only one antigen, Lan, with an occurrence of greater
at position 9q34.13-q34.3.4 The GBGTI gene produces than 99% in all populations. The Lan– phenotype occurs in
glycosyltransferase, which causes the formation of the Forss- about 1 in 20,000 people.
man glycolipid by the addition of N-acetylgalactosamine The Lan gene, ABCB6, located on chromosome 2 at posi-
(GalNAc) to the P antigen. The GalNAc terminal sugar’s tion 2q36, encodes another of the ATP-binding cassette
cross-reactivity with some polyclonal anti-A explains transporters.4 ABCB6 functions in heme synthesis with the
the original hypothesis that the Forssman glycolipid was ATP-dependent uptake of heme and porphyrins into mito-
an A subgroup. Group O RBCs expressing FORS1 do not chondria. In individuals with the Lan– phenotype, other
react with Dolichos biflorus or monoclonal anti-A.3 FORS1+ porphyrin transporters are thought to compensate. The pro-
donor RBCs may react in a crossmatch due to naturally tein is widely expressed in heart, skeletal muscles, fetal liver,
occurring anti-FORS1 in the plasma of ABO-compatible eye, mitochondrial membrane, and Golgi apparatus. Cord
FORS1– individuals. RBCs have a stronger reaction with monoclonal anti-Lan
The Forssman glycolipid is not normally expressed on than do adult cells.2
RBCs, having an occurrence in Caucasians of less than 0.1%.2,3 Reactivity with anti-Lan is resistant to ficin and papain
Healthy and malignant human tissue such as gastric and treatment of RBCs; the antigen is also DTT and EDTA/
colonic mucosa, lung, and kidney have been reported to ex- glycine-acid resistant.3 Anti-Lan is formed due to exposure
press the Forssman glycolipid. The Forssman glycolipid can via transfusion or pregnancy and has not been known to
serve as a receptor for pathogens such as Escherichia coli, mak- be naturally occurring. Lan antibodies are IgG reacting at
ing one believe that human cells that express the FORS1 may the antiglobulin phase of testing with some antibodies
have an increased susceptibility to E. coli infection. known to bind complement. Anti-Lan is considered clini-
Reactivity with anti-FORS1 is enhanced with ficin and cally significant; the first anti-Lan caused an immediate
papain treatment; the antigen is resistant to DTT and HTR. Lan– RBCs should be selected for transfusion. Anti-
glycine-acid EDTA treatment. The antibody is mostly IgM Lan has not been known to cause severe HDFN. In two re-
with optimal reactivity at room temperature or 4°C, with an ported cases, the newborns of mothers with anti-Lan had a
unknown clinical significance. positive DAT2.

The JR (032) System The Vel (034) System


Jra is a high-prevalence antigen in most populations; the Anti-Vel was first described in 1952 and was named after the
Jr(a–) phenotype is found more commonly in Japanese. JR individual in whom the first antibody was identified. Vel, a
was assigned a system symbol JR and number 32 in 2012.3 high-prevalence antigen, was raised to system status in 2013
The gene ABCG2 is located on chromosome 4 at position when absence of SMIM1, a single-pass integral membrane
4q22.1.4 The encoded protein, ABCG2, is a member of the protein, was shown to produce the Vel– phenotype. The ISBT
adenosine triphosphate (ATP) binding cassette transporters Vel system symbol is VEL and number 034. There is one
broadly distributed throughout the body. It is involved in antigen, Vel. No molecular evidence has been discovered to
multidrug resistance in tumor cells, presenting a problem in link ABTI to the Vel system despite the phenotypic associa-
chemotherapy.3 There is only one antigen, Jra, in the JR sys- tion demonstrated with serologic testing.1 The gene SMIM1
tem. The first anti-Jra, named for the antibody maker Rose is located on chromosome 1 at position 1p36.32.4
Jacobs, was described in 1970; several examples have since Anti-Vel is characterized by its ability to activate comple-
been found. The antigen is fully developed at birth and is re- ment and cause in vitro and in vivo hemolysis. The antibody
sistant to treatment with ficin and papain, DTT, and glycine- is most often IgG but can be IgM, and it has caused severe, im-
acid EDTA. mediate HTRs.2,12 Anti-Vel has also caused one case of severe
Anti-Jra is usually IgG. Clinical significance of anti-Jra is HDFN in which the mother had previously been transfused.3
not well established due to the rare occurrence of the anti- Vel antigen expression is weak on cord RBCs and can
body. Anti-Jra has caused severe HDFN in some cases, in- vary from person to person on RBCs of adults. RBCs with
cluding two fatal cases of HDFN.28 Some patients with weak expression of Vel can be mistyped as Vel–. This is one
6888_Ch09_212-231 31/10/18 9:48 AM Page 221

Chapter 9 Uncommon Blood Groups 221

of the reasons why anti-Vel can be a difficult antibody to work The Augustine (036) System
with and identify. Reactivity with anti-Vel can be enhanced
with enzyme-treated RBCs. The antigen is also resistant to Anti-Ata was first described in 1967 in the serum of a black
glycine-acid EDTA and DTT treatment, though some exam- woman named Mrs. Augustine.12 Ata is a high-prevalence
ples of anti-Vel do not react with DTT-treated RBCs.29 antigen, and all At(a–) individuals have been black. The ISBT
blood group system status was assigned in 2015 with a sym-
The CD59 (035) System bol of AUG, number 036, and two antigens, AUG1 and
AUG2 (Ata). The antigen defined by the antibody produced
In 2014, the blood group number 035 was assigned to the with the null phenotype is AUG1. The AUG gene, SLC29A1,
CD59 system based on a CD59-deficient child who formed located on chromosome 6 at position 6p21.1, encodes the
an alloantibody with CD59 specificity. The gene CD59 is equilibrative nucleoside transporter 2 (ENT2).4
located on chromosome 11 at position 11p13 and has only The antigens are fully developed at birth and are resistant
one antigen, CD59.1.4 Both the blood group system and to treatment with ficin and papain, DTT, and glycine-acid
symbol are CD59.1 EDTA. Anti-Ata is usually IgG, reactive in the antiglobulin
CD59 is a GPI-linked complement-regulatory glycopro- phase of testing, and has caused severe HTRs and one re-
tein also known as the membrane inhibitor of lysis (MIRL). ported mild case of HDFN.2,3 A new low-prevalence antigen
CD59 plays a key role in protecting against complement- has been provisionally assigned AUG3; the corresponding
regulated hemolysis by binding to C8 and C9 thus interfer- antibody caused severe HDFN.31
ing with the formation of the membrane-attach complex
(MAC).2 Paroxysmal nocturnal hemoglobinuria (PNH) is ISBT Blood Group Collections
an acquired hemolytic anemia caused by a mutation in the
GPI-linker gene. PNH patients are deficient in all GPI-linked Collections are antigens that have a biochemical, serologic,
proteins, including CD59. or genetic relationship but do not meet the criteria for a
Congenital CD59-deficient individuals (i.e., who do not system.10 Most the antigens in the blood group collections
have PNH) have been identified among Japanese, North are either high or low prevalence. Antigens classified as a
Africans, Jews, and Turks. The one example of anti-CD59.1 collection are assigned a 200 number. Some of the previously
was IgG, which showed increased reactivity with enzyme- established collections have been made obsolete as the
treated RBCs and was nonreactive with DTT-treated RBCs.30 criteria have been met to establish a system or incorporate
Patients with CD59 deficiency show PNH-like symptoms, antigens into an existing system. See Table 9–3 for a listing
including hemolysis and strokes, but also neuropathy. of the ISBT collections.

Table 9–3 International Society of Blood Transfusion Blood Group Collections


Collection Antigen
Number Name Symbol Number Symbol Incidence %

205 Cost COST 205001 Csa 95


205002 Csb 34

207 Ii I 207002 I

208 Er ER 208001 Era >99


208002 Erb <1
208003 Er3 >99

209 GLOB 209003 LKE 98

210 210001 Lec 1


210002 Led 6

213 MN CHO 213001 Hu


213002 M1
213003 Tm
213004 Can
213005 Sext
213006 Sj
6888_Ch09_212-231 31/10/18 9:48 AM Page 222

222 PART II Blood Groups and Serologic Testing

Cost Collection 205 antigens, Era and Er3. The third antigen is a low-prevalence
antigen, Erb. Anti-Er3 is produced by the presumed null
The Cost collection, formally referred to as Cost-Sterling, phenotype Er(a−b−). The Er antigens are resistant to
was established in 1988. It was named after the two enzyme and DTT treatment, but sensitive to glycine acid/
patients (Copeland and Sterling) who made anti-Csa.3 EDTA treatment. The antibodies are IgG reacting at the
Five antigens formally in the Cost collection have been antiglobulin phase of testing and do not fix complement.
placed into the Knops system once it was established that Limited data exist on the clinical significance of Er anti-
they are carried on complement receptor 1 (CR1). Unlike bodies. Transfusion of Er(a+) RBCs to a patient with anti-
the Knops system antigens (which are all located on CR1), Era resulted in a positive DAT but no signs of hemolysis.2
the Cost collection, although serologically similar to the A patient with anti-Er3 showed mild hemolysis following
Knops system antigens (especially Yka), is not located transfusion of an incompatible unit. Monocyte-monolayer
on CR1. It has been detected on three of four examples of assays (MMA) suggest anti-Era is not clinically significant,
the Helgeson phenotype, the Knops system serologic null whereas anti-Er3 had potential significance.2 The Er
phenotype.2 The relationship between the Cost collection antigens are expressed on cord cells, and anti-Era and −Erb
and the Knops system is still unknown. The Cost collection have caused a positive DAT in the newborn but no clini-
currently consists of two antithetical antigens, Csa and Csb. cally significant HDFN.
The Csa antigen is a high-prevalence antigen with a
frequency of 95% in the black population and greater than Globoside Collection 209
98% in most populations.3 The Csb antigen is a polymor-
phic antigen with a frequency of 34% in most populations. The Globoside collection currently consists of one high-
Both antigens are thought to be expressed on cord cells prevalence antigen, LKE.1,4 The GLOB collection and how it
with a slightly weaker expression of Csa. Both the Csa and relates to the P1PK (003) and Globoside (028) blood group
Csb antigens demonstrate a resistance to enzyme treatment systems is covered in Chapter 8.
and varying effects to treatment with DTT.3 The Cost col-
lection antibodies are IgG reacting at antiglobulin phase of Collection 210 (Unnamed)
testing, and have not been found to be clinically significant.
The antibodies are difficult to identify due to the varying Collection 210 consist of two antigens, Lec and Led, which
RBC expression from person to person. are glycosphingolipid adsorbed onto RBCs. Lec and Led
are precursors to the Lewis antigens and demonstrate
Ii Collection 207 increased expression in Le(a−b−) individuals.2 The antigen
names are misleading because neither Lec nor Led are
The Ii collection was first assigned in 1990 and consisted of produced by a Lewis gene transferase. Lec is a low-
two antigens, I and i. In 2002, the I antigen was promoted prevalence antigen occurring in 1% of most populations.
to a blood group system 027, leaving i the sole antigen in the Anti-Lec agglutinates Le(a−b−) RBCs from nonsecretors.
collection. The genetic basis of the i expression is currently Both humans and goats are known to make anti-Lec.3 Led
unknown.3 The i antigen occurs on unbranched carbohy- is a polymorphic antigen occurring in 6% of most popula-
drate chains and is the precursor for the I antigen. Along tions.3 Anti-Led agglutinates Le(a−b−) RBC from secretors,
with RBCs, the I and i antigens are found on most human but not from Le(a−b−) nonsecretors.2 The discovery of
cells and on soluble glycoproteins in body fluids. The anti-Led was the result of injecting goats with saliva from
i antigen is strongly expressed on cord cells with only trace a Le(a−b+) individual.2
amounts on adult RBCs. The i adult phenotype is rare and
usually has anti-I present. The i antigen is resistant to both MN CHO Collection 213
enzyme and chemical treatments. The i antigen may increase
in expression during hemopoietic stress due to rapid pro- The MN CHO collection currently consists of six polymor-
duction of RBCs. The i expression varies with the different phic antigens: Hu, M1, Tm, Can, Sext, and Sj. The antigens
Lu(a−b−) phenotypes. The dominant Lu(a−b−) has a decreased are associated with the M or N antigen in the MNS (002)
expression of the i antigen; whereas, the X-linked Lu(a−b−) system and are expressed on GPA with altered levels of sialic
has an enhanced i expression. Anti-i is usually IgM reacting acid (NeuNAc) or GlcNAc. All the antigens are sensitive
at room temperature or 4°C, some may bind complement. to ficin/papain, with some demonstrating resistance to
Anti-i is not known to cause transfusion reactions and rarely α-chymotrypsin.3 All the antibodies to the MN CHO collec-
causes HDFN. Autoanti-i is a cold agglutinin associated with tion react optimally at room temperature.
infectious mononucleosis and some lymphoproliferative The Hu antigen (213001) was formerly called Hunter
disorders that occasionally can cause hemolysis.2,3 after the donor of the RBCs used to immunize rabbits in
1934. The frequency of the Hu antigen varies within differ-
Er Collection 208 ent ethnicities: 1% Caucasians, 7% African Americans, and
22% West Africans.3 The Hu antigen is predominantly an
The Er collection was established as a collection in 1990 altered GPAN with a strong link to the Sext antigen, which
with three antigens. Two of the antigens are high-prevalence is also in the MN CHO collection. Enzyme treatment shows
6888_Ch09_212-231 31/10/18 9:48 AM Page 223

Chapter 9 Uncommon Blood Groups 223

sensitivity to ficin/papain and trypsin and resistance to Center, Stenbar and James, whose RBCs demonstrated
α-chymotrypsin. Antibodies to the Hu antigen have only strong agglutination with the serum.3 The frequency of the
been demonstrated in immunized rabbits. Sj antigen is 2% in whites and 4% in blacks.3 The antigen
The M1 antigen (213002) was reported in 1960 and is is predominantly associated with GPAN and sensitive to
predominantly an altered GPAM. Enzyme treatment of the ficin/papain.
M1 antigen shows sensitivity to ficin/papain and trypsin and
resistance to α-chymotrypsin. The M1 antigen is expressed ISBT 700 Series
only on M+ RBCs, but M1 is not an enhanced expression
of the M antigen. Some anti-M only react with M1+ RBCs. Antigens in the 700 series of low-prevalence antigens repre-
The majority of anti-M1 are made by M– individuals, but a sent those with a prevalence of less than 1% of most random
few cases of anti-M1 have been found in M+N+ individuals. populations. The responsible gene for these antigens is un-
The separation of anti-M and anti-M1 by differential adsorp- known; therefore,the antigens cannot be placed in a system.
tion has not been successful. When the genetic basis of the antigens in the 700 Series is
In 1965, the Sheerin antigen was renamed Tm (213003). identified and the antigen placed into a blood group system,
The logic behind the Tm naming was that “T” was next in the series number becomes obsolete. See Table 9–4 for cur-
line after the S and s, and since the “T” was already associ- rent 700 series antigens.4
ated with polyagglutination, the letter “m” was included due The low-prevalence antigen and antibody discovery
to the association with the MN system. The Tm antigen is is usually unexpected. Some examples of how the low-
predominantly an altered GPAN with a frequency of 25% prevalence antigen/antibodies were detected are (1) an
in whites and 31% in blacks.3 Enzyme treatment of the unknown maternal antibody causing HDFN, (2) an unex-
Tm antigen shows sensitivity to ficin/papain and trypsin and plained incompatible crossmatch, and (3) unexplained
resistance to α-chymotrypsin. positive reactivity with commercial human source antisera
The Can antigen (213004) was reported in 1979 and containing an unknown low-prevalence antibody. Low-
named after the first antigen-positive proband, Canner. An- prevalence antibodies are commonly found in serum that
tibodies to the Can antigen react with a higher percentage of contains multiple antibodies, especially antibodies to other
African Americans (60%) than with Caucasians (27%). Of low-prevalence antigens. A healthy donor, Mrs. Tillett, had
the Can+ RBCs, 87% of them were M+. The Can antigen is serum that contained 22 low-prevalence antibodies along
predominantly an altered GPAM that is sensitive to ficin/ with 8 unknown specificities.2
papain and trypsin and resistant to α-chymotrypsin. Seventeen antigens currently make up the 700 series of
The Sext antigen (213005) was reported in 1974 and the ISBT classification: By, Chra, Bi, Bxa, Toa, Pta, Rea, Jea, Lia,
named after the individual who produced the antibody. It is Milne, RASM, JFV, Kg, Jones, HJK, HOFM, and REIT. Of
found in only 24% of N+ black individuals. The Sext antigen these, anti-JFV, -Kg, -Jones, -HJK, and -REIT have all caused
is predominantly an altered GPAN with a strong link to the HDFN. A case of severe HDFN that required three intrauter-
Hu antigen. The Sext antigen is sensitive to ficin/papain, ine transfusions was seen in a neonate due to the presence
trypsin, and salidase. of anti-HJK. HDFN caused by anti-Kga and -REIT required
The Sj antigen (213006) was reported in 1968 when an exchange transfusion. Other cases of HDFN ranged in sever-
anti-Tm serum was shown to have an additional specificity. ity resulting in a positive DAT, phototherapy, and/or trans-
Sj was named after two employees at the New York Blood fusions.2 Finding compatible blood for transfusion is not

Table 9–4 International Society of Blood Transfusion Antigens of Low (700 Series) Prevalence
Number Name Symbol Number Name Symbol
700002 Batty By 700039 Milne

700003 Christiansen Chra 700040 Rasmussen RASM

700005 Biles Bi 700044 JFV

700006 Box Bxa 700045 Katagiri Kg

700017 Torkildsen Toa 700047 Jones JONES

700018 Peters Pta 700049 HJK

700019 Reid Rea 700050 HOFM

700021 Jensen Jea 700054 REIT

700028 Livesay Lia


6888_Ch09_212-231 31/10/18 9:48 AM Page 224

224 PART II Blood Groups and Serologic Testing

difficult because of the low prevalence of the antigen, making Israeli women and one Arab-Israeli family demonstrated the
compatible blood readily available. As with any low-prevalence AnWj– phenotype. The AnWj antigen is not expressed on
antigen, there is a risk of transfusing an incompatible unit cord cells and has varying expression in adults. The RBCs
when a potent low-prevalence antibody is present and the of the dominant In(Lu) phenotype demonstrate a very
crossmatch is not carried through to the antiglobulin weak expression of the AnWj antigen. The AnWj antigen is
phase of testing (e.g., when electronic or immediate spin resistant to ficin/papain, trypsin, and α-chymotrypsin, and
crossmatch is used). is variable with DTT. Haemophilus influenza uses the AnWj
antigen as a receptor to enter RBCs. Anti-AnWj is an IgG
ISBT 901 Series antibody demonstrating reactivity at antiglobulin phase
of testing; most anti-AnWj are autoantibodies due to a tran-
Unlike low-prevalence antigens, finding compatible units sient suppression of the AnWj antigen. Only two examples
negative for antigens in the 901 series can be challenging. of alloanti-AnWj have been reported in two Israeli women,
Antigens in the 901 series of high-prevalence antigens with only one described to cause a severe transfusion reac-
represent those with a prevalence of more than 90% of tion. Since most of the anti-AnWj are autoantibodies
most random populations. The responsible gene for the with antigen suppression, transfusion of AnWj+ units may
antigen is unknown, and the antigen cannot be placed in be tolerated.
a system. Six antigens currently make up the 901 series of
the ISBT classification: Emm, AnWj, Sda, PEL, ABTI, and Sda Antigen
MAM. See Table 9–5 for current 901 series antigens.4 The
difficulty in identifying and finding compatible units The Sda antigen is a high-prevalence carbohydrate antigen
makes antibodies to the high-prevalence antigens a poten- named for Sid, who was the head of the maintenance
tial transfusion risk. department at the Lister Institute in London. His RBCs
had been used for many years as a panel donor, and they
Emm Antigen reacted strongly with examples of a new antibody. The sol-
uble form of Sda is Tamm-Horsfall glycoprotein found in
The Emm antigen was first reported in 1987 and named after urine. The antigen is not expressed on RBCs of newborns
the first identified antigen-negative proband. Six probands but is in their saliva, urine, and meconium. The strength
with the negative phenotype have been identified. The anti- of Sda on adult RBCs varies and is markedly reduced in
gen is expressed on cord cells and is resistant to all enzyme pregnancy. The antigen is found on 91% of RBC samples,
and chemical treatments. Since the Emm antigen is carried and Sda substance is found in 96% of urine samples. Only
on a GPI-linked protein and paroxysmal nocturnal hemo- 4% of people are Sd(a–).3 Strong examples of Sda are noted
globinuria (PNH) patients are deficient in all GPI-linked as Sd(a++).
proteins, the Emm antigen is not present on PNHIII RBCs.3 Anti-Sda can naturally occur (i.e., without known stim-
Anti-Emm has been identified to be both IgG and IgM react- ulation by transfusion or pregnancy) in the sera of individ-
ing at both 4°C and antiglobulin phase of testing with a uals who are Sd(a–). Anti-Sda is usually an IgM agglutinin
higher occurrence of IgG antibodies. Some anti-Emm have that is reactive at room temperature, but it can be detected
been shown to bind complement. Six of the seven probands in the indirect antiglobulin test and does not react with
were examples of a naturally occurring anti-Emm in non- cord RBCs. Reactivity is described as small, refractile
transfused males.3 (shiny) agglutinates in a sea of free RBCs when viewed
microscopically. Because the soluble antigen is present
AnWj Antigen in urine of Sd(a+) individuals and in Guinea pig urine,
neutralization of the refractile agglutinates by urine is
In 1982, an alloantibody to an antigen called Anton was a technique used to identify anti-Sda. The Sda antigen is
identified and followed by identification in 1983 of an resistant to treatment with ficin, papain, DTT, and glycine-
autoantibody to an antigen call Wj. In 1985, it was shown acid EDTA. Reactivity of the antibody is enhanced with
that they were the same antigen and renamed AnWj. Two enzyme-treated RBCs. Anti-Sda is generally considered

Table 9–5 International Society of Blood Transfusion Antigens of High (901 Series) Prevalence
Number Name Symbol Number Name Symbol
901008 Emm 901014 PEL

901009 Anton AnWj 901015 ABTI

901012 Sid Sda 901016 MAM


6888_Ch09_212-231 31/10/18 9:48 AM Page 225

Chapter 9 Uncommon Blood Groups 225

clinically insignificant for transfusion, though there are two Monocyte-monolayer assays (MMA) suggest a potentially
reports of transfusion reactions associated with the trans- shortened survival of transfused MAM+ cells.2
fusion of Sd(a++) RBCs.
HLA Antigens on RBCs
PEL Antigen
HLA class I antigens (HLA-A, -B, and -C) are present on
The high-prevalence PEL antigen was first identified in all nucleated cells. Since mature RBCs are not nucleated,
1980 and given the name PEL in 1996 after the first negative they generally do not have detectable levels of HLA anti-
proband. The PEL-negative phenotype has been identified gens; thus HLA antigens are not considered a blood group
in two French-Canadian families. The PEL antigen is ex- antigen. The name “Bg” (Bga, Bgb, and Bgc) was given to
pressed on cord cells and is resistant to both enzyme and HLA class I antigens that are detectable on mature RBCs.
chemical treatment. Anti-PEL are presumed to be IgG react- Bga represents HLA-B7, Bgb represents HLA-B17, and Bgc
ing at the antiglobulin phase of testing. Transfusion of represents HLA-A28.32 Not all individuals with the corre-
51Cr-labeled RBCs have shown reduced survival.3 No signs sponding HLA antigens on their lymphocytes express the
of HDFN were noted for the baby of the original PEL- Bg antigen on their RBCs. The strength of Bg antigens can
negative propand.2 vary with each donation, making a constant prediction of
expression on panel RBCs difficult. HLA antigens on RBCs
ABTI Antigen are not destroyed by enzymes or DTT/AET treatment.
However, chloroquine or EDTA/glycine-HCL can be used
In 1996, the high-prevalence ABTI antigen was reported to remove the HLA antigen from RBCs. Due to the in-
with the detection of anti-ABTI in three multiparous women creased expression of HLA class I antigens on platelets, Bg
of an inbred Israeli-Arab family. The ABTI– phenotype antibodies can be adsorbed from the serum/plasma using
was also reported in one Bavarian and one German indi- platelet concentrate. Bg antibodies are usually considered
vidual. The expression on cord RBCs was presumed to be insignificant, but although rare, Bg antibodies have been
the mechanism of immunization. Since Vel– RBCs have reported to cause immediate and delayed HTRs.2,33 HLA
demonstrated a weak expression of the ABTI antigen, and antibodies have not been implicated in HDFN.2 HLA anti-
ABTI– RBCs have weak expression of Vel, the antigen was bodies play a significant role in transfusion-associated
placed in the VEL collection. In 2013, Vel was assigned acute lung injury (TRALI).
system status (034) with the linkage to the SMIMI gene.
At that time, no known molecular evidence linked ABTI Applications to Routine Blood Banking
to SMIM1, the single-pass integral membrane protein as-
sociated with the Vel gene SMIM1. Since no relationship The uncommon antigens and antibodies covered in this
to the gene SMIMI could be made, ABTI was returned to chapter may rarely be encountered in day-to-day serology
the 901 series and the VEL collection was made obsolete.1 testing due to the prevalence (either low or high) of the
The ABTI antigen is resistant to enzymes and chemical antigen. When an uncommon antibody is detected, under-
treatment. Anti-ABTI are IgG reacting at the antiglobulin standing antigen and antibody characteristics and how they
phase of testing. Anti-ABTI has not demonstrated evidence relate to frequency, ethnicity, enzymes, and chemical treat-
of HDFN. The clinical significance of anti-ABTI for ments is critical in the identification process. Antibodies to
transfusion is unknown since there are no clinical data low-prevalence antigens are most often detected through
available.3 unexpected incompatible crossmatches or cases of HDFN.
The selection of units does not present a problem due to
MAM Antigen large numbers of crossmatch-compatible units available,
but proper identification of the low-prevalence antigens can
The high-prevalence antigen MAM was first reported be challenging when one is confronted with an unexplained
in 1993 and assigned to the 901 series in 1999. Four HDFN. Antibodies to high-prevalence and other uncom-
MAM– probands have been reported. All the probands are mon antigens are easy to detect but difficult to work with
thought to have formed anti-MAM through exposure dur- since most blood banks do not have the antigen-negative
ing pregnancy because none of the probands had a history reagent RBCs needed to exclude other alloantibodies, nor
of transfusion.2,3 The MAM antigen is expressed on cord do they have typing reagents to phenotype the patient’s
cells, lymphocytes, granulocytes, monocytes, and probably RBCs or have an inventory of rare units. Due to the vari-
platelets. One of the babies of the probands had severe ability in clinical significance of the antibodies to uncom-
HDFN requiring intrauterine transfusion. The babies of mon antigens and their relationship to transfusion and
the other three probands demonstrated a positive DAT HDFN, proper identification and understanding the rele-
with no HDFN. It is thought that anti-MAM may also vance of the antibodies is critical to good patient care. Few
cause neonatal thrombocytopenia.3 The MAM antigen is technologists will have the opportunity to identify uncom-
resistant to both enzyme and chemical treatment. Anti- mon antibodies, but this chapter will provide a reference
MAM are IgG, reacting at the antiglobulin phase of testing. for all technologists when the rare antibody is detected.
6888_Ch09_212-231 31/10/18 9:48 AM Page 226

226 PART II Blood Groups and Serologic Testing

CASE STUDIES
Case 9-1

A type and screen is ordered on a 40-year-old female pregnant with her third child for a scheduled C-section.
Anti-A Anti-B Anti-D A1 Cells B Cells Interpretation
0 4+ 3+ 4+ 0
Antibody Screen
D C E c e f M N S s Lua Lub P1 Lea Leb K k Fya Fyb Jka Jkb IS PEG IAT
1 + + 0 0 + 0 + 0 + 0 0 + + + 0 + + + 0 0 + 0 1+
2 + 0 + + 0 0 0 + 0 + 0 + + 0 + 0 + 0 + + 0 0 0
Please note: There is no 37C reading when PEG is used.
Panel
D C E c e f M N S s Lua Lub P1 Lea Leb K k Fya Fyb Jka Jkb IS PEG IAT
1 + + 0 0 + 0 + 0 + 0 0 + + + 0 + + + 0 0 + 0 0
2 + + 0 0 + 0 0 + 0 + 0 + + 0 + 0 + 0 + + 0 0 0
3 + 0 + + 0 0 + + + + 0 + 0 + 0 0 + + 0 0 + 0 0
4 + 0 0 + + + + 0 + 0 0 + + 0 0 0 + 0 0 + 0 0 0
5 0 + 0 + + + + + 0 + 0 + + 0 + 0 + + 0 + + 0 0
6 0 0 + + + + + + 0 + 0 + 0 0 + 0 + + + + + 0 1+
7 0 0 0 + + + 0 + 0 + 0 + + + 0 0 + 0 + 0 + 0 1+
8 0 0 0 + + + + 0 + + 0 + 0 0 + + 0 + + + + 0 0
9 0 0 0 + + + 0 + + 0 0 + 0 0 + 0 + + + + + 0 0
10 0 0 0 + + + + 0 + + + + + + 0 0 + + 0 + 0 0 0
11 + 0 + + 0 0 + + + + 0 + + 0 + + + 0 + + + 0 0
AC 0 0
1. What is the patient’s ABO, Rh type, and antibody screen results?
2. Have all of the common alloantibodies been ruled out?
3. Chloroquine treatment of cells 6 and 7 removes reactivity. What is the likely antibody?
4. What type of adsorption can also remove reactivity?
5. What is the clinical significance for HTR or HDFN?
6. How should RBC units be selected if this patient requires transfusion during or after her surgery?
7. What type of transfusion reaction can occur when this antibody is present in the donor’s plasma?

Case 9-2

One unit of blood was ordered for transfusions on a 75-year-old male with myelodysplastic syndrome with a history of
multiple blood transfusions within the last 3 months with no history of unexpected alloantibodies.
ABO/Rh
Anti-A Anti-B Anti-D A1 Cells B Cells Interpretation
0 0 0 4+ 4+
Antibody Screen
D C E c e f M N S s Lua Lub P1 Lea Leb K k Fya Fyb Jka Jkb IS PEG IAT
1 + + 0 0 + 0 + 0 + 0 0 + + + 0 + + + 0 0 + 0 0
2 + 0 + + 0 0 0 + 0 + 0 + + 0 + 0 + 0 + + 0 0 0
Unit was crossmatched at immediate spin and found to be compatible.
Patient reported back pain and feeling of impending doom. A transfusion reaction evaluation was initiated with the
following results:
• Clerical check: no discrepancy
• ABO check: no discrepancy
• Direct antiglobulin test: negative with IgG and C3
• Hemolysis check: hemolysis present in EDTA specimen
6888_Ch09_212-231 31/10/18 9:48 AM Page 227

Chapter 9 Uncommon Blood Groups 227

Panel
D C E c e f M N S s Lua Lub P1 Lea Leb K k Fya Fyb Jka Jkb IS PEG IAT
1 + + 0 0 + 0 + 0 + 0 0 + + + 0 + + + 0 0 + 0 0
2 + + 0 0 + 0 0 + 0 + 0 + + 0 + 0 + 0 + + 0 0 0
3 + 0 + + 0 0 + + + + 0 + 0 + 0 0 + + 0 0 + 0 0
4 + 0 0 + + + + 0 + 0 0 + + 0 0 0 + 0 0 + 0 0 0
5 0 + 0 + + + + + 0 + 0 + + 0 + 0 + + 0 + + 0 0
6 0 0 + + + + + + 0 + 0 + 0 0 + 0 + + + + + 0 0
7 0 0 0 + + + 0 + 0 + 0 + + + 0 0 + 0 + 0 + 0 0
8 0 0 0 + + + + 0 + + 0 + 0 0 + + 0 + + + + 0 0
9 0 0 0 + + + 0 + + 0 0 + 0 0 + 0 + + + + + 0 0
10 0 0 0 + + + + 0 + + + + + + 0 0 + + 0 + 0 0 0
11 + 0 + + 0 0 + + + + 0 + + 0 + + + 0 + + + 0 0
AC 0 0
Additional testing on transfused unit:
• Direct antiglobulin test on the suspected unit was negative.
• AHG crossmatch of the suspected unit was 4+.
1. What is the patient’s ABO, Rh type, and antibody screen results?
2. What additional testing should be performed on the transfused unit suspected in the transfusion reaction?
3. Would you suspect the antibody specificity to be due to a high-prevalence or low-prevalence antigen?
4. Which of the following antigens is likely the suspected cause of the hemolytic transfusion reaction?
a. Knb
b. Wra
c. Wrb
d. Gya
5. What would be required if an additional unit was requested?

SUMMARY CHART
Blood Group Terminology  Wrb expression requires the presence of a normal GPA
(MNS system); alloanti-Wrb is extremely rare.
 In the ISBT classification, all antigens are catalogued
into one of the following four groups:  Yt antigens are GPI-linked RBC glycoproteins that are
• A blood group system if controlled by a single gene absent in individuals with PNH.
locus or by two or more closely linked genes  Anti-Yta is a fairly common antibody to a high-prevalence
• A collection if shown to share a biochemical, sero- antigen that is sometimes clinically significant and some-
logic, or genetic relationship times insignificant.
• The high-prevalence series (901) if found in more  The Xga antigen is found on the short arm of the
than 90% of most populations X chromosome and is of higher prevalence in females
• The low-prevalence series (700) if found in less than (89%) than in males (66%). Although it is usually IgG;
1% of most populations some examples are naturally occurring.
 Anti-Xga has not been implicated in HDFN or as a
“Uncommon” Blood Groups and Antigens cause of HTRs.
 The Diego system antigens are located on a major RBC  Antibodies to Scianna system antigens are rare and lit-
protein, band 3, also known as the RBC anion ex- tle is known about their clinical significance. The rare
changer (AE1). null phenotype, Sc:–1,–2,–3, has been observed in the
 Anti-Dia, anti-Dib, and anti-Wra are generally considered Marshall Islands and New Guinea.
to be clinically significant; all have caused severe HTRs  In addition to the Doa and Dob antigens, the Gya, Hy,
and HDFN. Anti-Wra is a relatively common antibody. and Joa antigens are assigned to the Dombrock system.
Continued
6888_Ch09_212-231 31/10/18 9:48 AM Page 228

228 PART II Blood Groups and Serologic Testing

SUMMARY CHART—cont’d
Anti-Doa and anti-Dob have caused HTRs but no clin-  Anti-Vel is most often IgG but can be IgM, and it has
ical HDFN; these antibodies are usually weak and caused severe immediate HTRs and HDFN. When
difficult to identify. serum is tested, anti-Vel characteristically causes in
 The Colton system is composed of the antithetical vitro hemolysis.
Coa and Cob antigens as well as the high-prevalence  Anti-Ata has been found only in blacks; the antibody
Co3 antigen; the antigens are carried on aquaporin 1, is usually IgG and has caused severe HTRs.
an RBC water channel. The Colton antibodies have  Anti-Jra is found more commonly in Japanese, but clin-
caused HTRs and HDFN. ical significance is not well established, since it is a rare
 LW has a phenotypic relationship with the D antigen; antibody; it has caused HTRs and fatal cases of HDFN.
Rhnull RBCs type LW(a–b–).  The CD59 antigen plays a key role in protecting
 Anti-LW reacts strongly with D+ RBCs and can look against complement-regulated hemolysis by binding
like anti-D. DTT treatment of test RBCs will distinguish to C8 and C9.
between these two antibodies because the LW antigen  Collections are antigens that have a biochemical, sero-
is denatured by DTT, but the D antigen is not. In other logic, or genetic relationship but do not meet the
words, anti-LW does not react with DTT-treated D+ criteria for a system.
RBCs but anti-D does.  Cost, Ii, Er, Globoside, Unnamed, and MN CHO are all
 The antigens in the Chido/Rodgers system are located blood group collections.
on the complement fragments C4B and C4A, respec-  The MN CHO antigens are associated with the M or
tively, that are adsorbed onto RBCs from plasma. N antigen in the MNS (002) system and are expressed
 The clinically insignificant anti-Ch and anti-Rg react on GPA with altered levels of sialic acid (NeuNAc) or
weakly, often to moderate or high-titer endpoints in GlcNAc.
the antiglobulin test, and may be tentatively identified  Low-prevalence antigen/antibodies are usually de-
by plasma inhibition methods. tected when either an unknown maternal antibody
 Gerbich-negative phenotypes are very rare outside of causing HDFN is detected or an unexplained incom-
Papua, New Guinea. patible crossmatch is found.
 The Gerbich or Leach phenotypes have weak expres-  Low-prevalence antibodies are commonly found in
sion of Kell blood group antigens. serum that contains multiple antibodies.
 Gerbich antibodies are sometimes clinically signifi-  Finding compatible blood for patients with low-
cant for transfusion and sometimes insignificant. prevalence antibodies for transfusion is not difficult
Three cases of serious HDFN due to anti-Ge3 have due to the low prevalence of the antigen making com-
been reported, associated with late onset anemia patible blood readily available.
(after birth).  Unlike low-prevalence antigens, finding compatible
 The Cromer antigens are carried on the decay-acceler- units negative for antigens in the 901 series can be
ating factor (DAF) and are distributed in body fluids challenging.
and on RBCs, WBCs, platelets, and placental tissue.  Six antigens currently make up the 901 series of the ISBT
 The rare anti-Cra and anti-Tca have been found only classification: Emm, AnWj, Sda, PEL, ABTI, and MAM.
in black individuals; some examples have caused  Anti-Sda has characteristic shiny and refractile agglu-
HTRs. tinates under the microscope and is inhibited with
 The Knops antigens are located on complement receptor urine from Sd(a+) individuals and guinea pigs.
1 (CR1). Knops antibodies are clinically insignificant  Haemophilus influenza uses the AnWj antigen as a
and have weak and nebulous reactivity at the antiglob- receptor to enter RBCs.
ulin phase; they are not inhibited by plasma.
 The difficulty in identifying and finding compatible
 The Ina antigen is more prevalent in Arab and Iranian units makes antibodies to the high-prevalence antigens
populations. Ina and Inb antigen expression is de- a potential transfusion risk.
pressed on the dominant type Lu(a–b–) RBCs.
 HLA antigens are not considered a blood group antigen.
 JMH antibodies most often occur in individuals with
 Chloroquine or EDTA/glycine-HCL can be used to
acquired JMH– status. Anti-JMH in these individuals
remove the HLA antigens from RBCs.
is not clinically significant.
6888_Ch09_212-231 31/10/18 9:48 AM Page 229

Chapter 9 Uncommon Blood Groups 229

8. The following antibodies are generally considered


Review Questions
clinically insignificant because they have not been
associated with causing increased destruction of RBCs,
1. The antibody to this high-prevalence antigen demon-
HDFN, or HTRs.
strates mixed-field agglutination that appears shiny and
refractile under the microscope. a. Anti-Doa and anti-Coa
b. Anti-Ge3 and anti-Wra
a. Vel
c. Anti-Ch and anti-Kna
b. JMH
d. Anti-Dib and anti-Yt
c. Jra
d. Sda 9. Which antigen is the receptor for Haemophilus influenza?
2. What red blood cell treatment can be used to differentiate a. AnWj
between anti-D and anti-LW? b. PEL
c. FORS
a. Ficin
d. Kna
b. Trypsin
c. DTT 10. Which antigen is not absent or is weakened on RBCs of
d. Papain individuals with PNH?
3. Which of the following has been associated with causing a. Yta
severe immediate HTRs? b. Cra
c. CD59
a. Anti-JMH
d. Coa
b. Anti-Lub
c. Anti-Vel 11. Which of the following blood groups is carried on a
d. Anti-Sda structure that helps to maintain the RBC membrane
integrity through interaction with protein band 4.1?
4. Which of the following antibodies would more likely be
found in a black patient? a. Di
b. Kn
a. Anti-Cra
c. Ge
b. Anti-Ata
d. Vel
c. Anti-Hy
d. All of the above 12. What is the name of the Knops system serologic null
phenotype?
5. Which of the following antigens is not in a blood group
system? a. Gregory
b. Leach
a. Doa
c. Helgeson
b. LKE
d. McLeod
c. JMH
d. Kx 13. Which antigen when absent produces a null in the
Dombrock system?
6. A weakly reactive antibody with a titer of 128 is neutral-
ized by plasma. Which of the following could be the a. Hy
specificity? b. Joa
c. Dob
a. Anti-JMH
d. Gya
b. Anti-Ch
c. Anti-Kna 14. Which antigens are strongly expressed on placental
d. Anti-Kpa tissue, allowing for the adsorption of antibodies?
7. An antibody reacted with untreated RBCs and DTT- a. Cromer
treated RBCs but not with ficin-treated RBCs. Which b. Knops
of the following antibodies could explain this pattern of c. Diego
reactivity? d. Vel

a. Anti-JMH 15. Which antigen was returned to the 901 series because
b. Anti-Yta there was no determined linkage to the SMIM1 gene?
c. Anti-Cra a. JMH
d. Anti-Ch b. Ata
c. ABTI
d. MAM
6888_Ch09_212-231 31/10/18 9:48 AM Page 230

230 PART II Blood Groups and Serologic Testing

16. The FORS blood group system was first thought to 8. Flegel WA, Chen Q, Reid ME, Martin J, Orsini LA, Poole J,
be part of what system due to the addition of et al. SCER and SCAN: two novel high-prevalence antigens in
the Scianna blood group system. Transfusion. 2005;45:1940-44.
N-acetylgalactosamine (GalNAc) to the P antigen? 9. Banks JA, Hemming N, Poole, J. Evidence that the Gya, Hy and
a. ABO Joa antigens belong to the Dombrock blood group system. Vox
b. Lewis Sang. 1995;68:177-82.
c. P1PK 10. Daniels GL, Fletcher A, Garratty G, Henry S, Jørgensen J,
Judd WJ, et al. Blood group terminology 2004: from the Inter-
d. Globoside
national Society of Blood Transfusion committee on terminol-
17. What glycophorin expresses the MN CHO collection ogy for red cell surface antigens. Vox Sang. 2004;87:304-16.
11. Michalewska B, Wielgos M, Zupanska B, Bartkowiak R. Anti-
antigens that are associated with altered levels of sialic Coa implicated in severe haemolytic disease of the foetus and
acid (NeuNAc) or GlcNAc? newborn. Transfus Med. 2008;18:71-73.
a. GPA 12. Issitt PD, Anstee DJ. Applied blood group serology. 4th ed.
b. GPB Durham (NC): Montgomery Scientific; 1998.
13. Landsteiner K, Wiener AS. An agglutinable factor in human
c. GPC
blood recognized by immune sera for rhesus blood. Proc Soc
d. GPD Exp Biol. 1940;43:223.
14. Levine P, Stetson RE. An unusual case of intragroup agglutina-
18. What techniques can be used to remove the reactivity of
tion. J Am Med Assn. 1939;113:126-27.
Bg antigens? 15. Bruce LJ. Hereditary stomatocytosis and cation leaky red
a. EDTA/glycine-HCL cells—recent developments. Blood Cells Mol Dis. 2009;42:
b. Platelet adsorption 216-22.
16. Arndt PA, Garratty G, Daniels G, Green CA, Wilkes AM, Hunt P,
c. Chloroquine treatment
et al. Late onset neonatal anaemia due to maternal anti-Ge:
d. All of the above possible association with destruction of erythroid progenitors.
Transfus Med. 2005;15:125-32.
19. ABTI was thought to be classified with which antigen 17. Blackall DP, Pesek GD, Montgomery MM, et al. Hemolytic
prior to it gaining system status? disease of the fetus and newborn due to anti-Ge3: combined
a. Jra antibody-dependent hemolysis and erythroid precursor cell
b. FORS1 growth inhibition. Am J Perinatol. 2008;25:541-45.
18. Storry JR, Reid ME. The Cromer blood group system: a review.
c. Vel
Immunohematology. 2002;18:95-103.
d. Lan 19. Hue-Roye K, Lomas-Francis C, Belaygorod L, Lublin DM,
Barnes J, Chung A, et al. Three new high-prevalence antigens
20. The Jr(a–) phenotype is found more commonly in: in the Cromer blood group system. Transfusion. 2007;47:
a. Japanese. 1621-29.
b. African Americans. 20. Yahalom V, Finkel L, Poole J, Crew V, Chezar J, Akaria L, et al.
c. South American Indians. CROK—a novel mutation of the Cromer blood group system.
Vox Sang. 2012 (Suppl 1);103:212.
d. Caucasians. 21. Moulds JM. The Knops blood group system: a review. Immuno-
hematology. 2010;26:2-7.
References 22. Parsons SF, Jones J, Anstee DJ, Judson PA, Gardner B, Wiener E,
et al. A novel form of congenital dyserythropoietic anemia
1. Storry JR, Castilho L, Chen Q, Daniels G, Debomme G, Flegel associated with deficiency of erythroid CD44 and a unique
WA, et al. International Society of Blood Transfusion Working blood group phenotype [In(a–b–), Co(a–b–)]. Blood. 1994;
Party on red cell immunogenetics and terminology: report of the 83:860-68.
Seoul and London meetings ISBT Science Series 2016;11:118-22. 23. Garratty G, Dzik W, Issit PD, Lublin DM, Reid ME, Zelinski T.
2. Daniels G. The human blood groups. 3rd ed. Oxford: Blackwell Terminology for blood group antigens and genes—historical
Science; 2013. origins and guidelines in the new millennium. Transfusion.
3. Reid ME, Lomas-Francis C, Olsson ML. The blood group anti- 2000;40:477-89.
gen facts book. 3rd ed. San Diego (CA): Elsevier, New York, NY.; 24. Karamatic Crew V, Burton N, Kagan A, Green CA, Levene C,
2012. Flinter F, et al. CD151, the first member of the tetraspanin
4. Red cell immunogenetics and blood group terminology. Am- (TM4) superfamily detected on erythrocytes, is essential for
sterdam (Netherlands): International Society of Blood Transfu- the correct assembly of human basement membranes in kidney
sion [cited 2018 May]. Available from: http://www.isbtweb.org/ and skin. Blood. 2004;104:2217-23.
working-parties/red-cell-immunogenetics-and-blood-group- 25. Karamatic Crew V, Poole J, Long S, Warke N, Colavecchia C,
terminology/ Burton N, et al. Two MER2-negative individuals with the same
5. Eaton BR, Morton JA, Pickles MM, White KE. A new antibody, novel CD151 mutation and evidence for clinical significance of
anti-Yta, characterizing a blood group antigen of high incidence. anti-MER2. Transfusion. 2008;48:1912-16.
Br J Haematol. 1956;2:333-41. 26. Seltsam A, Strigens S, Levene C, Yahalom V, Moulds M, Moulds JJ,
6. Byrne KM, Byrne PC. Review: other blood group systems— et al. The molecular diversity of Sema7A, the semaphorin
Diego, Yt, et al, Xg, Scianna, Dombrock, Colton, Landsteiner- that carries the JMH blood group antigens. Transfusion.
Wiener, and Indian. Immunohematology. 2004;20:50-58. 2007;47:133-46.
7. Devine P, Dawson FE, Motschman TL, Skradski KJ, Orsini L, 27. Tilley L, Green C, Poole J, Gaskell A, Ridgwell K, Burton NM,
Moulds JJ, et al. Serologic evidence that Scianna null (Sc:–1,–2) et al. A new blood group system, RHAG: three antigens result-
red cells lack multiple high-frequency antigens. Transfusion. ing from amino acid substitutions in the Rh-associated glyco-
1988;28:346-49. protein. Vox Sang. 2010;98:151-59.
6888_Ch09_212-231 31/10/18 9:48 AM Page 231

Chapter 9 Uncommon Blood Groups 231

28. Castilho L, Reid ME. A review of the JR blood group system. 31. Millard GM, McGowan EC, Wilson B, Martin JR, Spooner M,
Immunohematology 2013;29:63-8. Morris S, et al. A proposed new low-frequency antigen in the
29. Rainer T, Israel B, Caglioti S, Figueroa D. The effects of dithio- Augustine blood group system associated with a severe case of
threitol-treated red blood cells with anti-Vel [abstract]. Trans- hemolytic disease of the fetus and newborn. Transfusion
fusion. 2004;44:122A. 2018;58:1320-22.
30. Weinstock C, Anliker M, von Zabern I. CD59: a long-known 32. Fung MK, Eder AF, Spitalnik SL, Westhoff CM (eds). Technical
complement inhibitor has advanced to a blood group system. Manual. 19th ed. Bethesda, MD: AABB; 2017.
Immunohematology 2015;31:145-51.
6888_Ch10_232-255 29/10/18 4:57 PM Page 232

Chapter 10
Detection and Identification of Antibodies
Susanne Bishop, MS, MLS (ASCP)CMSBBCM and
Kathleen S. Trudell, MLS (ASCP)CMSBBCM

Introduction Neutralization Case 10-4


Antibody Detection Adsorption Case 10-5
Tube Method Direct Antiglobulin Test and Elution Summary Chart
Other Methods (Gel Technology, Techniques Review Questions
Solid-Phase Technology) Temperature-Dependent References
Interpretation of Results Methods
Limitations pH Method
Antibody Identification Organic Solvent Methods
Patient History Antibody Titration
Reagents Providing Compatible Blood
Exclusion Products When an Alloantibody
Is Present
Evaluation of Panel Results
Case Studies
Additional Techniques for Resolving
Antibody Identification Case 10-1
Selected Cell Panels Case 10-2
Enzymes Case 10-3

OBJECTIVES
1. Differentiate between the following antibodies: expected and unexpected, immune, naturally occurring, passive, autoantibody,
alloantibody, warm and cold.
2. Explain what factors make an antibody clinically significant.
3. Describe which patient populations require an antibody screen.
4. Select appropriate cells to include in a screen cell set.
5. Interpret the result of an antibody screen.
6. List the limitations of the antibody screen.
7. Interpret the results of an antibody identification panel.
8. Summarize the exclusion and inclusion methods.
9. Correlate knowledge of the serologic characteristics of commonly encountered antibodies with antibody identification panel
findings.
10. Identify the situations in which additional panel cells should be tested and select appropriate cells.
11. Describe antigen-typing techniques.
12. Calculate the number of red blood cell (RBC) units that must be antigen-tested to fulfill a provider’s request for crossmatch.
13. Explain the principles behind enzyme and neutralization techniques.
14. Given a patient’s history and initial results, choose the correct method for performing an adsorption.

232
6888_Ch10_232-255 29/10/18 4:57 PM Page 233

Chapter 10 Detection and Identification of Antibodies 233

OBJECTIVES—cont’d
15. Describe elution methods and give an example of when each would be used.
16. Outline the procedure for determining the antibody titer level, including reporting of results.
17. Given a patient scenario, identify additional steps that could be employed to resolve a complex antibody identification
problem.

Introduction certain populations require antibody screen testing. The


AABB’s Standards for Blood Banks and Transfusion Services
The detection of antibodies directed against red blood cell requires the use of an antibody screen to detect clinically sig-
(RBC) antigens is critical in pretransfusion compatibility nificant antibodies in allogeneic blood donors and in patients
testing. It is one of the principal tools for investigating as part of pretransfusion compatibility testing for whole
potential hemolytic transfusion reactions and immune blood, RBCs, and granulocyte components.2 Although not
hemolytic anemias. In addition, it aids in detecting and mon- an AABB requirement, an antibody screen is also included
itoring patients who are at risk of delivering infants with in standard prenatal testing for obstetric patients to evaluate
hemolytic disease of the fetus and newborn (HDFN). the risk of HDFN and to assess the mother’s candidacy for
The focus of antibody detection methods is on “irregular” Rh-immune globulin (RhIg) prophylaxis.3 An antibody
or “unexpected” antibodies, as opposed to the “expected” screen may additionally be performed when evaluating the
antibodies of the ABO system. The unexpected antibodies compatibility of allogeneic hematopoietic progenitor cell
of primary importance are the immune alloantibodies, (HPC) and bone marrow donors with the intended trans-
which are produced in response to RBC stimulation through plant recipient.4
transfusion, transplantation, or pregnancy. Other unexpected
antibodies may be “naturally occurring” (i.e., produced with- Tube Method
out RBC stimulation). Naturally occurring alloantibodies
form as a result of exposure to environmental sources, such The traditional testing method used to detect clinically sig-
as pollen, fungus, and bacteria, which have structures similar nificant antibodies is the IAT performed in a test tube. In this
to some RBC antigens. A third category of unexpected anti- method, RBC reagents (suspended at a concentration be-
bodies are passively acquired antibodies. Passively acquired tween 2% and 5% in a preservative diluent), enhancement
antibodies are antibodies produced in one individual and reagent (if desired), and AHG reagent are used to sensitize
then transmitted to another individual via plasma-containing reagent RBCs with the patient’s antibodies, followed by
blood components or derivatives such as intravenous im- formation of visible RBC agglutinates.
munoglobulin (IVIG). A fourth category of unexpected an-
Method Overview
tibodies are autoantibodies. Autoantibodies are antibodies
directed against antigens expressed on one’s own RBCs and In the test tube method, the patient’s serum is mixed with
generally react with all RBCs tested. reagent RBCs that have known antigen content (Fig. 10–1).
The presence of naturally occurring alloantibodies, pas- This test may include an immediate spin (IS) phase to detect
sively acquired antibodies, and autoantibodies may compli- antibodies reacting at room temperature. Performance of the
cate the detection and identification of clinically significant, IS phase is, however, not required and may lead to the de-
immune alloantibodies. Clinically significant alloantibodies tection of clinically insignificant cold antibodies. The test
are those that cause decreased survival of RBCs possessing must include a 37°C incubation phase; during this phase,
the target antigen. These antibodies are typically IgG anti- IgG antibodies, if present in the patient’s serum, will sensitize
bodies that react at 37°C and/or at the antihuman globulin any reagent RBCs that possess the target antigen. Enhance-
(AHG) phase of the indirect antiglobulin test (IAT). ment reagent may be added to the test tube prior to incubation
When an unexpected antibody is detected in the antibody at 37°C to increase the degree of sensitization. Depending on
screen, an antibody identification panel is performed to de- the enhancement added, the tubes may be centrifuged and
termine the specificity of the antibody (or antibodies) pres- observed for hemolysis and agglutination following this in-
ent. Once identified, the antibody’s clinical significance can cubation phase; tubes only need to be observed for hemoly-
be ascertained and the appropriate transfusion considerations sis when fresh serum and reagent RBCs that do not contain
put into place. This chapter describes antibody detection and EDTA are used because complement is necessary for hemol-
identification methods, the resolution of complex antibody ysis to occur. To observe for hemolysis, the tube is carefully
cases, and transfusion considerations when an auto- and/or removed from the centrifuge so as not to dislodge the RBC
alloantibody is present. button. The supernatant is observed for pink or red discol-
oration. To observe for agglutination, the tube is gently tilted
Antibody Detection or rolled to dislodge the cell button. The degree of reactivity,
which is determined after all the RBCs have been dislodged
Only a small percentage of the general population (between from the bottom of the test tube, is graded as 0 (no aggluti-
0.8% and 2%) has detectable RBC alloantibodies.1 However, nation present) to w+ (agglutination barely visible to the
6888_Ch10_232-255 29/10/18 4:57 PM Page 234

234 PART II Blood Groups and Serologic Testing

Immediate Spin phase 37°C phase AHG phase


(optional) Centrifuge Centrifuge
Centrifuge Observe for Observe for
Observe for hemolysis hemolysis and hemolysis and
and agglutination agglutination agglutination

2 drops Add enhancement reagent Wash 3 to 4 times Add 2 drops Confirm negative
patient’s plasma (optional) with normal saline AHG reagent reactions with Coombs’
⫹ Incubate at 37°C Unbound antibodies Sensitized RBCs control cells
1 drop If antibodies present, removed cross-linked by
screen cell reagent RBC sensitization occurs antibodies in AHG

Figure 10–1. Steps for performing the tube antibody screen test.

naked eye) to 4+ (one solid agglutinate) (see Color Plate 1). or three (R1R1, R2R2, rr) cells, each having a unique combi-
The tubes are then washed with 0.9% saline a minimum of nation of antigens. For blood donor antibody screening, it is
three times to remove all antibodies that remain unbound. acceptable to use a pooled screening reagent that contains
AHG reagent, also known as Coombs’ serum, is added to RBCs from at least two different individuals; a pooled RBC
each tube. The tubes are centrifuged and examined for screening reagent cannot be used for patient antibody
hemolysis, if applicable, and agglutination. If reading for he- screening. Since pooled reagent RBCs contain more than one
molysis is applicable (i.e., fresh serum and reagent RBCs that RBC population, reactions should be carefully observed for
do not contain EDTA are used), it may appear as a loss of mixed-field agglutination, as only one RBC in the pool may
cell button mass. If the RBCs are coated with IgG antibodies, express the target antigen.
the anti-IgG antibody in the AHG reagent will create a bridge Within the reagent set or pooled reagent, there should be
between sensitized reagent RBCs, resulting in observable one cell that is positive for each of the following antigens: D,
agglutination. If there are no antibodies directed against any C, c, E, e, K, k, Fya, Fyb, Jka, Jkb, Lea, Leb, P1, M, N, S, and s.
of the antigens present on the reagent RBCs, the RBCs will Other antigens will be expressed as well. An antigen profile
not be sensitized, and there will be no agglutination. De- sheet, detailing which known antigens are present, will accom-
pending on the enhancement media used, the agglutination pany each set of screen cells or pooled reagent. These profiles
reactions may be observed macroscopically only or may in- are lot-specific and should not be interchanged. Figure 10–2
clude a microscopic examination. All negative tests must shows an example of a three-cell profile.
have Coombs’ control cells (also known as check cells) Ideally, there will be homozygous expression of many
added to confirm the validity of the negative result. of the antigens within the screen cell set/pooled reagent,
Use of the tube test remains popular due to the flexibility allowing for detection of antibodies that show dosage. A
of the test system, the availability of required testing equip- cell with homozygous antigen expression is from an indi-
ment, and the relative low cost. The disadvantages include vidual who inherited only one allele at a given genetic locus.
the instability of the reactions and subjective nature of grad- Therefore, the cell surface has a “double dose” of that anti-
ing by the technologist, the amount of hands-on time for the gen. A cell with heterozygous antigen expression is from
technologist, and problems related to the failure of the wash- an individual who inherited two different alleles at a given
ing phase to remove all unbound antibody. genetic locus. When this occurs, the alleles “share” the
available antigen sites on the cell surface. In Figure 10–3,
RBC Reagents the pair of chromosomes on the left possess the same allele,
The RBC reagents used in antibody screen testing come from thus resulting in an RBC with homozygous antigen expres-
group O individuals who have been typed for the most com- sion. The pair of chromosomes on the right possess different
mon and the most significant RBC antigens. Group O cells alleles, thus resulting in an RBC with heterozygous antigen
are used so that anti-A and anti-B will not interfere in the de- expression.
tection of antibodies to other blood group systems. The Certain antibodies, such as those of the Kidd system, may
screen cells used for patient antibody screening are manu- only be detected when tested against RBCs expressing one
factured as a set and are comprised of two (R1R1 and R2R2) allele (homozygous). Antibodies that react more strongly
6888_Ch10_232-255 29/10/18 4:57 PM Page 235

Chapter 10 Detection and Identification of Antibodies 235

Cell D C c E e Cw K k Kpa Kpb Jsa Jsb Fya Fyb Jka Jkb Lea Leb P1 M N S s Lua Lub Xga

R1R1 + + 0 0 + 0 0 + 0 + 0 + + + + 0 + 0 + + + + + 0 + +

R2R2 + 0 + + 0 0 + + 0 + 0 + + 0 0 + 0 + 0 0 0 + 0 0 + +

rr 0 0 + 0 + 0 0 + 0 + 0 + 0 + + + 0 + + + + 0 + 0 + +

Figure 10–2. Antigen profile of three-cell screen set.

and polyethylene glycol (PEG). For a detailed discussion


of these enhancement reagents, refer to Chapter 5 “The
Antiglobulin Test.”

AHG Reagents
AHG reagent (monospecific or polyspecific) is added to/
included in the test system as it allows for agglutination of
incomplete antibodies. Monospecific AHG reagent contains
Figure 10–3. Homozygous inheritance versus heterozygous inheritance.
only one antibody specificity, either anti-IgG or antibodies
to complement components C3 and C4 or C3b and C3d.
with cells having homozygous antigen expression are said to Polyspecific AHG reagent, which is also called polyvalent or
show dosage. Box 10–1 lists the antibodies that may show broad spectrum Coombs’ serum, contains antibodies to both
dosage, and Table 10–1 provides examples of RBC phenotypes, IgG and complement components. Polyspecific AHG reagent
comparing homozygous and heterozygous antigen expression. is not often used when performing antibody detection testing
since the presence of anticomplement in the AHG reagent
Enhancement Reagents may lead to the detection of clinically insignificant anti-
Various enhancement reagents, or potentiators, may be bodies. Relatively few examples of clinically significant anti-
added to the test system prior to the 37°C incubation phase bodies react with complement alone; antibodies to some
to increase sensitivity. Inclusion/addition of an enhancement Kidd blood group system antigens are the exception, how-
reagent may also allow for a shortened incubation time. En- ever, and react better or only with anticomplement.5 For a
hancement reagents used in IAT antibody detection testing detailed discussion of these reagents, including the use of
include 22% albumin, low ionic strength solution (LISS), monospecific versus polyspecific AHG in the IAT, refer to
Chapter 5, “The Antiglobulin Test.”
Any tube test that is negative after adding the AHG
reagent should be controlled by adding Coombs’ control
BOX 10–1 cells, which are Rh-positive RBCs that have been coated with
anti-D. In a negative antiglobulin test, these antibody-coated
Common Blood Group Systems With Antibodies cells will react with the anti-IgG in the AHG reagent that re-
That Exhibit Dosage mains free in the test tube, resulting in visible agglutination.
• Rh (except D) The addition of the Coombs’ control cells proves adequate
• Kidd washing was performed to remove unbound antibodies be-
• Duffy fore the AHG reagent was added; the AHG reagent was added
• MNSs to the test tube; and the AHG reagent was working properly.
• Lutheran If the Coombs’ control cells fail to agglutinate, the antibody
screen must be repeated from the beginning.

Table 10–1 Examples of RBC Phenotypes From Homozygous and Heterozygous Individuals
Phenotype Antigens Present Homozygous vs. Heterozygous
Jk(a–b+) Jkb only Homozygous

Jk(a+b+) Both Jka and Jkb Heterozygous

Fy(a+b–) Fya only Homozygous

Fy(a+b+) Both Fya and Fyb Heterozygous

M+ N- M only Homozygous

M+N+ Both M and N Heterozygous


6888_Ch10_232-255 29/10/18 4:57 PM Page 236

236 PART II Blood Groups and Serologic Testing

Other Methods (Gel Technology, Solid-Phase target antigen is found on more than one screen cell, or
Technology) when the patient’s serum contains an autoantibody. A sin-
gle antibody specificity should be suspected when all
The antibody screen is also routinely performed using gel screen cells yielding a positive reaction react at the same
technology or solid-phase technology. phase(s) and strength. Multiple antibodies are most likely
For a detailed discussion of how these methods are used present when screen cells react at different phases or
for antibody screening at the IAT phase of testing, refer to strengths. Autoantibodies should be suspected when the
Chapter 5, “The Antiglobulin Test”; for the method princi- autologous control or DAT is positive and all screen cells
ple, refer to Chapter 12, “Blood Bank Testing Technologies tested yield a positive reaction. Figure 10–4 provides sev-
and Automation”; and for reagent RBC requirements refer to eral examples of antibody screen results with possible
the aforementioned “RBC Reagents” section in this chapter. causes.
4. Is hemolysis or mixed-field agglutination present?
Interpretation of Results Certain antibodies, such as anti-Lea, anti-Leb, anti-PP1Pk,
and anti-Vel, are known to cause in vitro hemolysis in the
A positive test result at any phase of testing indicates the
presence of complement. Mixed-field agglutination is as-
need for antibody identification studies. Evaluation of anti-
sociated with anti-Sda and Lutheran antibodies.
body screen results, including the autologous control if
5. Are the cells truly agglutinated, or is rouleaux present?
tested at this time, and patient history (i.e., age, sex, race,
Serum from patients with altered albumin-to-globulin
diagnosis, pregnancy and transfusion history, current med-
ratios (e.g., patients with multiple myeloma) or from
ications, and intravenous solutions) can provide clues and
those who have received high molecular weight plasma
give direction for identification and resolution of the anti-
expanders (e.g., dextran) may cause nonspecific aggrega-
body or antibodies present. In complex cases, patient history
tion of RBCs, known as rouleaux. Rouleaux is not a sig-
can be particularly valuable. The investigator should con-
nificant finding in antibody screening tests; however, it is
sider the following questions:
easily confused with antibody-mediated agglutination.
1. In what phase(s) did the reaction(s) occur? Knowing the following characteristics of rouleaux helps
Antibodies of the IgM class react best at room tempera- in differentiating between rouleaux and agglutination:
ture or lower and are capable of causing agglutination of • Cells have a “stacked coin” appearance when viewed
saline-suspended RBCs (immediate spin reaction). Anti- microscopically (see Color Plate 1).
bodies of the IgG class react best at the AHG phase. Of • Rouleaux is observed in all tests containing the patient’s
the commonly encountered antibodies, anti-N, anti-I, and serum, including the autologous control and the reverse
anti-P1 are frequently IgM, whereas those directed against ABO grouping.
Rh, Kell, Kidd, Duffy, and Ss antigens are usually IgG. • Rouleaux does not interfere with the AHG phase of
Lewis and M antibodies may be IgG, IgM, or a mixture testing in tube method or solid-phase technology
of both. because the patient’s serum is washed away prior to the
2. Is the autologous control negative or positive? addition of the AHG reagent.
The autologous control is the patient’s RBCs tested against • Unlike agglutination, rouleaux disperses by adding one
the patient’s serum in the same manner as the antibody to three drops of saline to the test tube when perform-
screen. A positive antibody screen and a negative autolo- ing tube testing.
gous control indicates that an alloantibody has been de-
tected. If the autologous control is positive, transfusion Limitations
history needs to be evaluated. If the patient has not been
transfused within the last 3 months, a positive autologous Screening reagents and methods are designed to detect as
control may indicate the presence of autoantibodies or an- many clinically significant antibodies as possible and to
tibodies to medications. If the patient has been recently avoid detecting antibodies that are clinically insignificant.
transfused (i.e., within the previous 3 months), the posi- When using a three-cell screen set, a negative result with all
tive autologous control, which may exhibit mixed-field three cells gives the technologist 95% confidence that no
appearance, may be caused by alloantibodies coating the clinically significant antibodies are present.
circulating donor RBCs. Evaluation of samples with a pos- Despite this, there are limitations to the antibody screen.
itive autologous control or positive direct antiglobulin The screen will not detect antibodies when the antibody titer
test (DAT) result is often complex and may require a great has dropped below the level of sensitivity for the screening
deal of time and experience on the part of the investigator. method employed. One study, which reviewed antibodies de-
Some technologists choose to omit the autologous control tected over a 20-year period, showed that 26% of antibodies
when performing the antibody screen and include it only became undetectable over time, with a median time of
when performing antibody identification studies. 7 months.6 Another study of antibodies formed in response
3. Did more than one screen cell sample react? If so, did to transfusion found that two-thirds of antibodies were no
they react at the same strength and phase(s)? longer detectable 5 years after formation.7 The screen also
More than one screen cell may be positive when the pa- cannot detect antibodies directed against low-prevalence
tient has multiple antibodies, when a single antibody’s antigens that are not present on any of the RBCs in the screen
6888_Ch10_232-255 29/10/18 4:57 PM Page 237

Chapter 10 Detection and Identification of Antibodies 237

Results Possible Interpretation

cell IS 37°C AGT (poly) 1. Single alloantibody


SC I neg neg neg 2. Two alloantibodies, antigens only
present on cell II
SC II neg neg 2+
auto neg neg neg 3. Probably lgG alloantibody

cell IS 37°C AGT (poly) 1. Multiple alloantibodies


SC I neg 1+ 3+ 2. Single alloantibody (dosage)
SC II neg neg 1+ 3. Probable lgG alloantibody
auto neg neg neg

cell IS 37°C AGT (poly) 1. Single or multiple antibodies


SC I 1+ neg neg 2. Probably lgM alloantibody
SC II 2+ neg neg
auto neg neg neg

cell IS 37°C AGT (poly) 1. Multiple alloantibodies, warm and


cold
SC I 2+ neg 1+
2. Potent cold alloantibody binding
SC II 3+ 1+ 2+
complement in AGT
auto neg neg neg

cell IS 37°C AGT (poly) 1. Single warm alloantibody, antigen


present on both cells
SC I neg neg 1+
2. Antibody to high-prevalence antigen
SC II neg neg 1+
3. Complement binding by a cold
auto neg neg neg alloantibody not detected at lS

cell IS 37°C AGT (poly) 1. Warm alloantibodies


SC I neg neg 3+ 2. Transfusion reaction
SC II neg neg 3+ 3. Probable lgG alloantibody
auto neg neg 3+ 4. Warm autoantibody

Figure 10–4. Examples of antibody screen results with possible causes.

cell set. Lastly, antibodies showing dosage may not be detected used in antibody detection testing. Further additional testing
if none of the screen cells have homozygous expression of the may be required based on the initial, additional testing results.
target antigen.
Patient History
Antibody Identification
Information concerning the patient’s age, sex, race, diagno-
Once an antibody has been detected in the antibody screen, sis, transfusion and pregnancy history, medications, and in-
additional testing/review is necessary to identify the antibody travenous solutions may provide valuable clues in antibody
and determine its clinical significance. In general, reviewing identification studies, particularly in complex cases. Know-
patient history and testing an antibody identification panel, ing the patient’s race can be beneficial, as some antibodies
along with an auto control or DAT (if not previously per- are associated with a particular race. For example, anti-U is
formed), comprises initial, additional testing/review. When more frequently associated with persons of African descent
performing initial antibody identification panel testing, the because most U-negative individuals are found in this pop-
method(s) and test phases used should be the same as those ulation. Knowledge of transfusion and pregnancy history is
6888_Ch10_232-255 29/10/18 4:57 PM Page 238

238 PART II Blood Groups and Serologic Testing

also beneficial, as patients who have been exposed to non- the antigens on each cell and providing a place to record re-
self RBCs via transfusion or pregnancy are more likely to actions accompanies each panel (Fig. 10–5). The profile
have produced immune antibodies. Naturally occurring anti- sheet will also often indicate the presence of rare cells, which
bodies (e.g., anti-M, anti-Leb) should be suspected in patients are positive for low-prevalence antigens or negative for high-
with no transfusion or pregnancy history. In addition, know- prevalence antigens. As with the screen cells, the profile
ing if a patient has received IVIG, RhIg, and antilymphocyte sheet is lot-specific and should not be interchanged with that
globulin is beneficial as these medications may passively trans- of another panel.
fer antibodies such as anti-A or anti-B, anti-D, and antispecies
antibodies, respectively. This will result in the presence of Exclusion
an unexpected serum antibody that is likely to confound the
interpretation of antibody identification. When interpreting panel results, the first step is to exclude
The patient’s history is especially important when the au- antibodies that could not be responsible for the reactivity
tologous control or DAT is positive. Certain infectious and seen. To perform exclusions or “rule-outs,” the RBCs that
autoimmune disorders are associated with production of gave a negative reaction in all phases of testing are examined;
RBC autoantibodies, and some medications are known to the antigens on these negatively reacting cells are probably
cause positive DATs. Furthermore, in a patient transfused not the antibody’s target. Generally, it is advisable to perform
within the past 3 months, a positive DAT result may indicate this rule-out technique only if there is homozygous expres-
a delayed hemolytic transfusion reaction. sion of the antigen on the cell. This avoids excluding a weak
Information regarding recent transfusions is also impor- antibody that is showing dosage. Exceptions are made for
tant when antigen-typing the patient’s RBCs. Antigen-typing low-prevalence antigens that are rarely expressed homozy-
results must be interpreted carefully when the patient has re- gously, such as K, Kpa, Jsa, and Lua. In the sample panel
cently received a transfusion, because positive reactions may shown in Figure 10–6, cell numbers 1, 3, 4, 6, 7, 10, and 11
be caused by the presence of donor RBCs remaining in the reacted positively and cannot be used for exclusions. Cell
patient’s circulation. Positive reactions caused by donor RBCs numbers 2, 5, 8, and 9 reacted negatively and can therefore
usually show mixed-field agglutination, but this depends on be used to perform exclusions. Cell number 2 can be used
how recent the transfusion was given and how much blood to exclude D, C, e, Cw, K, Kpb, Jsb, Fyb, Jka, P1, M, S, Lub,
was transfused. If mixed-field agglutination is present, results and Xga. Cell number 5 can be used to exclude k, Jkb, and
should be interpreted with caution. Leb. Cell number 8 can be used to exclude c and Lea and cell
number 9 can be used to exclude N and s. Antibodies that
Reagents could not be excluded and hence may be present include
anti-E, anti-Kpa, anti-Jsa, anti-Lua, and anti-Fya.
An antibody identification panel is a collection of 11 to
20 group O reagent RBCs, with various antigen expression. Evaluation of Panel Results
The pattern of antigen expression should be diverse so that
it will be possible to distinguish one antibody from another After each negatively reacting cell has been evaluated, the
and should include cells with homozygous expression of Rh, remaining antigens that were not excluded should be exam-
Duffy, Kidd, and MNSs antigens. A profile sheet specifying ined to see if the pattern of reactivity matches a pattern of

Donor Cell
D C c E e Cw K k Kpa Kpb Jsa Jsb Fya Fyb Jka Jkb Lea Leb P1 M N S s Lua Lub Xga
number

R1R1 1 + + 0 0 + 0 0 + 0 + 0 + + + + 0 0 + + + + + + 0 + +

R1wR1 2 + + 0 0 + + + + 0 + 0 + + 0 0 + 0 + 0 + 0 + + 0 + +

RzR1 3 + + 0 + + 0 0 + 0 + 0 + 0 + + + 0 + + 0 + 0 + 0 + +

R2R2 4 + 0 + + 0 0 0 + 0 + 0 + + 0 + + 0 + + + 0 + 0 0 + +

r’r 5 0 + + 0 + 0 0 + 0 + 0 + 0 0 0 + + 0 + + 0 + + 0 + 0

r”r 6 0 0 + + + 0 0 + 0 + 0 + + 0 0 + 0 0 + 0 + 0 + 0 + +

rr 7 0 0 + 0 + 0 + + 0 + 0 + + 0 0 + 0 + + 0 + 0 + 0 + +

Rr 8 0 0 + 0 + 0 0 + 0 + 0 + 0 + + 0 0 + 0 + 0 + 0 0 + +

Rr 9 0 0 + 0 + 0 0 + 0 + 0 + + 0 + 0 0 + + + + 0 + 0 + 0

R1r 10 + + + 0 + 0 0 + 0 + 0 + + 0 + + 0 + + + 0 0 + 0 + +

R0r 11 + 0 + 0 + 0 0 + 0 + 0 + + + 0 + 0 + + + 0 0 + 0 + +

Patient
Cells

Figure 10–5. Antibody identification profile sheet: + indicates the antigen is present on the cell, 0 indicates the antigen is not present on the cell.
6888_Ch10_232-255 29/10/18 4:57 PM Page 239

Chapter 10 Detection and Identification of Antibodies 239

Donor Cell
D C c E e Cw K k Kpa Kpb Jsa Jsb Fya Fyb Jka Jkb Lea Leb P1 M N S s Lua Lub Xga IS 37 AHG CC
number

R1R1 1 + + 0 0 + 0 0 + 0 + 0 + + + + 0 0 + + + + + + 0 + + 0 0 2+

R1wR1 2 + + 0 0 + + + + 0 + 0 + 0 + + 0 0 0 + + 0 + 0 0 + + 0 0 0 3+

RzR1 3 + + 0 + + 0 0 + 0 + 0 + + 0 + + 0 + + 0 + 0 + 0 + + 0 0 3+

R2R2 4 + 0 + + 0 0 0 + 0 + 0 + + 0 + + + 0 + + 0 + 0 0 + + 0 0 3+

r’r 5 0 + + 0 + 0 0 + 0 + 0 + 0 0 0 + 0 + + + 0 + + 0 + 0 0 0 0 3+

r”r 6 0 0 + + + 0 0 + 0 + 0 + + + 0 + 0 0 + 0 + 0 + 0 + + 0 0 2+

rr 7 0 0 + 0 + 0 + + 0 + 0 + + + 0 + 0 + + 0 + 0 + 0 + + 0 0 2+

rr 8 0 0 + 0 + 0 0 + 0 + 0 + 0 + + 0 + 0 0 + 0 + 0 0 + + 0 0 0 3+

r’r” 9 0 + + + + 0 0 + 0 + 0 + 0 + + 0 0 + + 0 + 0 + 0 + 0 0 0 0 3+

rr 10 0 0 + 0 + 0 0 + 0 + 0 + + 0 + + 0 + + + 0 0 + 0 + + 0 0 3+

R0r 11 + 0 + 0 + 0 0 + 0 + 0 + + + 0 + 0 + + + 0 0 + 0 + + 0 0 2+

Patient
0 0 0 3+
Cells

Figure 10–6. Sample panel demonstrating anti-Fya.

antigen-positive cells (inclusion technique). Evaluation of Phase of reactivity may be helpful in establishing the
panel results should be carried out in a logical step-by-step clinical significance of an antibody. IgM antibodies,
method to ensure proper identification and to avoid missing which are usually not significant, most often react at the
antibody specificities that may be masked by other antibod- immediate spin phase, room temperature or colder. Clin-
ies. A logical approach to antibody identification is outlined ically significant IgG antibodies are most often detected
below, using a series of questions and the example illustrated during the AHG phase. Some potent IgG antibodies,
in Figure 10-6. such as D, E, and K, may become evident following the
37°C incubation phase. Table 10–2 summarizes the op-
1. Do all of the positive cells react to the same degree? timal phase of reactivity for the more commonly encoun-
Carefully grading the observed reactions may aid in an- tered alloantibodies.
tibody identification. The strength of the reaction does 3. Does the serum reactivity match any of the remaining
not indicate the significance of the antibody, only the specificities? When a single alloantibody is present, the
amount of antibody available to participate in the reac- pattern of reactivity usually matches a pattern of antigen
tion. A stronger reaction may be due to dosage (cells expression exactly. In our example (Fig. 10–6), the serum
with homozygous antigen expression reacting more reactivity matches the Fya pattern exactly. The serum gave
strongly than cells with heterozygous antigen expres- positive results with all Fya-positive cells (1, 3, 4, 6, 7,
sion). Different reaction strengths could also indicate the 10, and 11) and negative results with all of the Fya-negative
presence of more than one antibody. A cell that possesses cells (2, 5, 8, and 9). The reason for the variation in
more than one of the target antigens may react more strength in the AHG phase is due to dosage; all cells yield-
strongly than a cell possessing only one of the target anti- ing 2+ reactions were heterozygous for Fya, and all cells
gens. A third possibility is an antigen with variable ex- yielding 3+ reactions were homozygous for Fya.
pression. I, P1, Lea, Leb, Vel, Ch/Rg, and Sda antigens are 4. Are all commonly encountered RBC antibodies excluded?
expressed more strongly on some RBCs than on others; As previously mentioned, in this example (Fig. 10–6),
hence, antibodies to these antigens may react more anti-E, anti-Kpa, anti-Jsa, and anti-Lua cannot be excluded.
strongly with one panel cell than another. In the example None of the RBCs on this panel are positive for Kpa, Jsa,
presented in Figure 10–6, the strengths of the reactions or Lua (all are low-prevalence antigens), so they are not
were 2+ and 3+. Based on the variation in reaction the cause of the observed reactions. Antibodies to low-
strength, multiple antibodies or a single antibody show- prevalence antigens are uncommon because the chance of
ing dosage should be considered. being exposed to the antigen is low; therefore, it may not
2. Do all of the positive cells react at the same phase, or be necessary to pursue additional testing to exclude these
do any react at different or multiple phases? Reactions specificities. In contrast, if a commonly encountered anti-
of certain cells at one phase and different cells at another body is not ruled out, it is important to test selected cells
phase may indicate the presence of multiple antibodies. that will rule out the presence of the antibody. In this ex-
Cells that react at multiple phases may also be a sign of ample, additional testing should be performed to exclude
an antibody showing dosage, with the cells having ho- anti-E. A negative result when testing an E+ e– Fya- RBC
mozygous antigen expression reacting at an earlier phase cell would exclude anti-E (see the “Selected Cell Panels”
than cells with heterozygous antigen expression. section in this chapter).
6888_Ch10_232-255 29/10/18 4:57 PM Page 240

240 PART II Blood Groups and Serologic Testing

Table 10–2 Optimal Phase of Reactivity for Some Common Alloantibodies


Immediate Spin (Room
Phase Temperature) 37°C Incubation Antiglobulin Phase
Antibodies Lea, Leb Potent cold (IgM) antibodies (especially Rh antibodies
those causing hemolysis)

M, N Some warm antibodies, if high in titer Kell


(e.g., D, E, and K)

Lua Duffy

P1 Kidd

S,s

Lub

If the pattern of reactivity is not discernable and all Testing of RBCs selected from other panels is necessary
commonly encountered antibodies have been excluded, when inadequate numbers of antigen-positive or antigen-
consider the possible presence of an antibody previously negative cells have been tested initially. In our example
known as an HTLA (high titer, low avidity) antibody or (Fig. 10-6), the patient’s serum reacted with seven
an antibody to Bg antigen (HLA Class I antigen) ex- Fya-positive cells (1, 3, 4, 6, 7, 10, and 11), but did not
pressed on RBCs. Resolution of HTLA antibody problems react with four Fya-negative cells (2, 5, 8, and 9). Thus,
is discussed briefly in the case studies at the end of this additional cells need not be tested (once anti-E has been
chapter. excluded) as the 95% confidence level has been met, and
5. Is the autologous control (last row in panel antigen the identification of anti-Fya is conclusive.
profile) positive or negative? In the example shown 7. Does the patient lack the antigen corresponding to the
(Fig. 10-6), the autocontrol is negative, indicating that antibody? Individuals cannot make alloantibodies to anti-
the positive reactions are caused by an alloantibody, not gens they possess on their own RBCs; therefore, the last
by an autoantibody. The presence of autoantibodies may step in identification studies is to test the patient’s RBCs
mask the presence of alloantibodies, and complicates for the corresponding antigen. A negative result is ex-
the process of antibody identification. Resolution of au- pected, providing additional evidence that identification
toantibody problems is discussed briefly in the case results are correct. If the patient’s RBCs are positive for
studies at the end of this chapter. the corresponding antigen, misidentification of the anti-
6. Is there sufficient evidence to prove the suspected anti- body or a false-positive typing are the most likely expla-
body? Conclusive antibody identification requires testing nations. Antigen-typing can also help resolve complex
the patient’s serum with enough antigen-positive and cases, as it may eliminate possible specificities. For
antigen-negative RBC samples to ensure that the pattern example, an R1R1, K-negative, Fy(a–b+), Jk(a+b+),
of reactivity is not the result of chance alone. Testing the M+N+S+s+ patient could only form anti-c, anti-E, anti-K,
patient’s serum with at least three antigen-positive and and anti-Fya. It is not practical to do extended typing on
three antigen-negative cells (also known as the 3 and all patients with antibodies; however, judicious use of this
3 rule) will result in a probability (P) value of 0.05.8 A procedure can be useful, especially in patients who
P value is a statistical measure of the probability that a cer- chronically receive transfusions (e.g., patients with sickle
tain set of events will happen by random chance. A P value cell disease or thalassemia) and are consequently at risk
of 0.05 or less is required for identification results to be con- for alloimmunization.
sidered valid, and it means that there is a 5% (1 in 20)
chance that the observed pattern occurred for reasons other Phenotyping may be complicated when a patient has a pos-
than a specific antibody reacting with its corresponding itive DAT or has been transfused in the last 3 months. When
antigen. Stated another way, it means that the interpretation the IgG DAT is positive, antibody bound to the RBCs prevents
of the data will be correct 95% of the time. When multiple the typing serum from reacting with the corresponding anti-
antibodies are present, the 3 and 3 rule must be applied gen, if present. When AHG reagent is added to the test system,
to each specificity separately. For example, if anti-K and it will react with the bound antibody detected in the DAT,
anti-E are both suspected, the 3 and 3 rule would be ful- yielding false-positive results. Consequently, typing serum that
filled if three K–E– cells reacted negatively, three K+E– cells employs the IAT should not be used in this instance unless
reacted positively, and three K–E+ cells reacted positively. the coating antibody can be successfully removed (eluted)
Other researchers have derived formulas where a P value of prior to testing. Two reagents useful in stripping antibody from
0.05 is fulfilled with two positive cells and three negative the RBC surface, while leaving the membrane intact to allow
cells9 or two positive cells and two negative cells.10 phenotyping, are acid glycine/EDTA (EGA) and chloroquine
6888_Ch10_232-255 29/10/18 4:57 PM Page 241

Chapter 10 Detection and Identification of Antibodies 241

diphosphate.11 In the EGA method, washed RBCs are incu- In addition, molecular testing is advantageous because there
bated with reagent (disodium EDTA and glycine-HCL) at are no limitations due to the lack of rare antisera or the ex-
room temperature for 2 minutes. Following incubation, TRIS pense in purchasing these reagents.21 There are, however, lim-
base is added to the tube. The RBCs are then washed three itations associated with RBC genotype testing. Limitations
times with saline, followed by DAT testing. If the DAT is neg- associated with molecular technologies include the following:
ative, then the RBCs can be used for IAT phenotype testing, cost; regulatory approval of methods; inaccurate prediction of
with the exception of Kell antigens; Kell antigens are dena- RBC antigen expression (the genotype may not accurately pre-
tured when using this method, so RBCs cannot be typed reli- dict the RBC phenotype); not all blood group antigens are the
ably for these antigens. If the DAT is positive after performance consequence of SNPs, thus requiring the use of more sophis-
of the procedure, the EGA-treated RBCs can be treated one ticated testing methods or testing algorithms; and the presence
additional time with EGA using the aforementioned steps.12 of mixed DNA populations in certain transplant recipients.21,22
If the DAT remains positive following the second EGA treat-
ment, the RBCs cannot be used in IAT phenotype testing. In Additional Techniques for Resolving
the chloroquine diphosphate method, washed RBCs are incu- Antibody Identification
bated with the reagent at room temperature for 30 minutes to
2 hours or for 5 to 30 minutes at 37°C; incubation time and There may be times when the initial antibody identification
number of incubations will depend on the results of the post- panel does not reveal a clear-cut specificity. When multiple
treatment DAT. Following incubation, the cells are then washed specificities remain following the exclusion and inclusion
to remove the chloroquine diphosphate. This step is followed process, additional testing is necessary.
by performance of the DAT. When the treated cells yield a neg-
ative DAT, they may be phenotyped using IAT typing sera.13 Selected Cell Panels
Recently transfused patients present a different challenge. Perhaps the simplest step to take is to test additional cells
When phenotyping a recently transfused patient, mixed-field from a different panel or panels. The cells selected for testing
reactions may be observed since two RBC populations are should have minimal overlap in the antigens they possess.
present (i.e., autologous patient RBCs and transfused donor Figure 10–7 shows the results of an initial panel in which
RBCs). If the donor RBCs express the antigen being tested anti-E, anti-Kpa, anti-Jsa, anti-Fya, and anti-Jkb are not ex-
for (i.e., corresponding antigen), the typing serum will react cluded. The lower section of the figure shows the results of
with those RBCs but not the autologous patient RBCs. To get a selected cell panel that could be used to differentiate be-
an accurate phenotype of autologous patient RBCs, reticu- tween anti-E, Anti-Fya, and anti-Jkb. Based on the results of
locyte typing can be performed. For this technique, the pa- the selected cell panel, anti-Jkb appears to be present; se-
tient’s RBCs are drawn into microhematocrit tubes and lected cell number 1 eliminates anti-Fya; and selected cell
centrifuged. Because reticulocytes are less dense than mature number 3 eliminates anti-E. As mentioned previously, find-
RBCs, the patient’s reticulocytes should be at the top of the
ing cells that are positive for low-prevalence antigens such
RBC layer. These cells can then be harvested from the
as Kpa and Jsa may not be possible, as is the case in this ex-
microhematocrit tubes and used for antigen-typing.14 When ample. Consequently, anti-Kpa and anti-Jsa cannot be ex-
performing antigen-typing on cells separated by this method, cluded or included with the selected cell panel; however,
it is important to observe reactions for the presence of none of the panel or selected cells that reacted express Kpa
mixed-field reactivity. If mixed-field reactivity is observed, or Jsa, so the reactivity observed with these cells is not at-
the cells should not be used for phenotype testing since tributable to either antibody.
donor cells are still present. Selected cell panels are also useful when a patient has a
Positive and negative control cells should be tested with known antibody and the technologist is attempting to deter-
each antisera used for antigen-typing, on the day of use. The mine if additional antibodies are present. Figure 10–8 is an
positive control cell should have heterozygous antigen ex- example of a selected cell panel for a patient with a history
pression to ensure that the antiserum has the sensitivity to of anti-Fya. Since it is not necessary to demonstrate the pres-
detect small quantities of the antigen. The negative control ence of anti-Fya, the panel cells selected are negative for Fya
cell should lack the target antigen to confirm reactivity with but possess the other common, significant antigens. In this
only the target antigen. example, all of the selected cells were negative, thus the re-
Phenotyping is routinely performed using tube, solid-phase maining major clinically significant antibody specificities
adherence,15 or gel methods.16 Phenotyping can also be per- have been excluded.
formed using flow cytometry, although this method is not rou-
tinely used. Usage of molecular technologies, which determine Enzymes
the RBC genotype by examining DNA for single-nucleotide
polymorphisms (SNPs), is becoming more prevalent in trans- Proteolytic enzymes (ficin, papain, bromelin, and trypsin)
fusion medicine and donor testing.17–20 Advantages of molec- modify the RBC surface by removing sialic acid residues and
ular testing include the ability to screen for multiple antigens by denaturing or removing glycoproteins. This modification
at once and to screen large numbers of donors in a relatively results in the destruction of certain antigens and the en-
short amount of time. There is also no interference from a re- hanced expression of others. Because of this effect, the use
cent transfusion, and DAT-positive specimens can be tested. of enzyme-treated panel cells may aid in identification when
6888_Ch10_232-255 29/10/18 4:57 PM Page 242

242 PART II Blood Groups and Serologic Testing

Initial Panel Results

Donor Cell
D C c E e Cw K k Kpa Kpb Jsa Jsb Fya Fyb Jka Jkb Lea Leb P1 M N S s Lua Lub Xga IS 37 AHG CC
number

R1r 1 + + + 0 + 0 0 + 0 + 0 + + + + + 0 + + + + + + 0 + + 0 0 1+

R1wR1 2 + + 0 0 + + + + 0 + 0 + 0 + + 0 0 0 + + 0 + 0 0 + + 0 0 0 3+

R2R2 3 + 0 + + 0 0 0 + 0 + 0 + + 0 + + 0 + + 0 + 0 + 0 + + 0 0 1+

R0r 4 + 0 + 0 + 0 0 + 0 + 0 + + 0 + + + 0 0 + 0 + + 0 + 0 0 0 1+

r’r 5 0 + + 0 + 0 0 + 0 + 0 + 0 0 + 0 0 + + + 0 + + 0 + 0 0 0 0 3+

r”r 6 0 0 + + + 0 0 + 0 + 0 + + + 0 + 0 + + 0 + 0 + 0 + + 0 0 2+

rr 7 0 0 + 0 + 0 + + 0 + 0 + + + + + 0 + 0 + + 0 + 0 + + 0 0 1+

rr 8 0 0 + 0 + 0 0 + 0 + 0 + 0 + + 0 + 0 + + + + 0 + + 0 0 0 0 3+

r’r” 9 0 + + + + 0 0 + 0 + 0 + 0 + + 0 0 + + 0 + 0 + 0 + + 0 0 0 3+

rr 10 0 0 + 0 + 0 0 + 0 + 0 + + 0 + + 0 + + + 0 + 0 0 + + 0 0 1+

R1r 11 + + + 0 + 0 0 + 0 + 0 + + + 0 + 0 + + + + + + 0 + + 0 0 2+

Patient
0 0 0 3+
Cells

Selected Cell Panel

Donor Cell
D C c E e Cw K k Kpa Kpb Jsa Jsb Fya Fyb Jka Jkb Lea Leb P1 M N S s Lua Lub Xga IS 37 AHG CC
number

R1R1 1 + + 0 0 + 0 + + 0 + 0 + + 0 + 0 0 + + 0 + + + 0 + 0 0 0 0 3+

rr 2 0 0 + 0 + 0 0 + 0 + 0 + 0 + 0 + + 0 + 0 + + 0 0 + + 0 0 2+

R2R2 3 + 0 + + 0 0 0 + 0 + 0 + 0 + + 0 0 + 0 + 0 + 0 0 + 0 0 0 0 3+

Figure 10–7. Initial and selected cell panels.

Donor Cell PEG/


D C c E e Cw K k Kpa Kpb Jsa Jsb Fya Fyb Jka Jkb Lea Leb P1 M N S s Lua Lub Xga CC
number AHG

R1R1 1 + + 0 0 + 0 0 + 0 + 0 + 0 + + 0 0 + + + + + 0 0 + + 0 2+

R1wR1 2 + + 0 0 + + 0 + + + 0 + 0 + + + 0 + + + 0 + 0 0 + 0 0 2+

R2R2 3 + 0 + + 0 0 + + 0 + 0 + 0 + 0 + 0 0 0 0 + 0 + 0 + + 0 2+

r’r 4 0 + + 0 + 0 0 + 0 + 0 + 0 0 + + + 0 + + 0 + + 0 + + 0 2+

Patient
0 2+
Cells

Figure 10–8. Selected cell panel for a patient with known antibody.

multiple antibodies appear to be present or help narrow enzyme-treated cells may be stronger or present at an earlier
down possible specificities when a high-prevalence antibody phase of testing (i.e., enhanced by enzymes); remain un-
appears to be present. Table 10–3 reviews how proteolytic changed (i.e., not affected by enzymes); be nonreactive or give
enzymes affect some common antigens. weaker reactions (destroyed by enzymes). When performing
Enzymes may be utilized in place of enhancement media exclusions, cells that are nonreactive with the enzyme-treated
(e.g., LISS, PEG) in a one-stage enzyme test method. In this cells, but reactive with the untreated cells, cannot be used to
method, untreated panel RBCs, enzyme, and patient serum exclude specificities destroyed by enzymes. Figure 10–9 pro-
are all added to the test tube prior to incubation. A second, vides an example of an enzyme-treated panel.
more sensitive method (two-stage method), requires enzyme
treatment of panel RBCs prior to the addition of patient Neutralization
serum to the test tube.
When using enzyme-treated cells as an aid in antibody Substances in the body and in nature have antigenic struc-
identification, compare the pre- and postenzyme treatment re- tures similar to certain RBC antigens. Consequently, these
actions whenever possible. Reactions observed with the substances can be used to neutralize the corresponding RBC
6888_Ch10_232-255 29/10/18 4:57 PM Page 243

Chapter 10 Detection and Identification of Antibodies 243

Table 10–3 The Effect of Proteolytic Table 10–4 Sources of Substances for
Enzymes on Select Antigen- Neutralization of Certain
Antibody Reactions Antibodies
Enhanced Inactivated Source of Neutralizing
Antibody Substance
Rh Duffy
Anti-P1 Hydatid cyst fluid, pigeon droppings,
Kidd MNS turtledoves’ egg whites
Lewis Xga Anti-Lewis Plasma or serum, saliva
P1 Anti-Chido, Serum (contains complement)
anti-Rodgers
I
Anti-Sda Urine
ABO
Anti-I Human breast milk

antibodies in serum, allowing for exclusion of the remaining results, it appears anti-Leb and at least one other additional
antibodies or confirmation that a particular antibody is pres- antibody are present. When the serum is incubated with
ent. This technique is especially helpful when multiple an- Lewis substance, positive reactions are seen only with cells 2,
tibodies are suspected and one of the suspected antibodies 7, and 10, which matches the pattern of anti-Kell.
appears to be an antibody that can be neutralized. Table 10–4
lists some of the antibodies that can be neutralized and the Adsorption
source of the corresponding neutralizing substances. Of
the substances listed, Lewis and P1 substances are available Antibodies may be removed or adsorbed from serum by in-
commercially. cubating the specimen with the corresponding antigen, thus
To perform neutralization, the patient’s serum is first in- allowing the antibody to bind to that antigen. The adsorbent
cubated with the neutralizing substance, allowing the soluble used in an adsorption procedure is typically RBCs but may
antigens in that substance to bind with the corresponding be another antigen-bearing substance. In the adsorption
antibody, if present. An antibody identification panel is then method, the antigen-antibody complex is composed of solid
performed using the treated serum. Use of a control (saline precipitates and is separated from the adsorbed serum by cen-
and serum) is also required to prove that loss of reactivity is trifugation. Once the desired antibody has been completely
due to neutralization and not to dilution of the antibody by adsorbed from the specimen, the adsorbed serum can be
the added substance. In Figure 10–10, neat serum reacted tested against RBC panel cells for the presence of unadsorbed
with cells 1, 2, 4, and 7 through 11. Based on the panel alloantibodies.

Donor Cell PEG/ Ficin/


D C c E e Cw K k Kpa Kpb Jsa Jsb Fya Fyb Jka Jkb Lea Leb P1 M N S s Lua Lub Xga AHG CC AHG CC
number

R1r 1 + + + 0 + 0 0 + 0 + 0 + + + + + 0 + + + + + + 0 + + 1+ 0 3+

R1wR1 2 + + 0 0 + + + + 0 + 0 + 0 + + 0 0 + + + 0 + 0 0 + + 3+ 2+

R2R2 3 + 0 + + 0 0 0 + 0 + 0 + + 0 0 + 0 + + 0 + 0 + 0 + + 0 3+ 0 3+

R0r 4 + 0 + 0 + 0 0 + 0 + 0 + + 0 + + + 0 0 + 0 + + 0 + 0 0 3+ 0 3+

r’r 5 0 + + 0 + 0 0 + 0 + 0 + 0 0 0 + 0 0 + + 0 + + 0 + 0 0 3+ 0 3+

r”r 6 0 0 + + + 0 0 + 0 + 0 + + + 0 + 0 + 0 0 + 0 + 0 + + 1+ 0 3+

rr 7 0 0 + 0 + 0 + + 0 + 0 + + + + + 0 + + + + 0 + 0 + + 2+ 2+

rr 8 0 0 + 0 + 0 0 + 0 + 0 + 0 + + 0 + 0 + + + + 0 0 + 0 2+ 0 3+

r’r” 9 0 + + + + 0 0 + 0 + 0 + 0 + + 0 0 + + 0 + 0 + 0 + + 2+ 0 3+

rr 10 0 0 + 0 + 0 0 + 0 + 0 + + 0 + + 0 + + + 0 + 0 0 + + 0 3+ 0 3+

R1r 11 + + + 0 + 0 0 + 0 + 0 + + + 0 + 0 + + + + + + 0 + + 1+ 0 3+

Patient
0 3+ 0 3+
Cells

Figure 10–9. Enzyme panel.


244
PART II
6888_Ch10_232-255 29/10/18 4:57 PM Page 244

Control: Serum +
Donor Cell PEG/ Serum + Lewis
D C c E e Cw K k Kpa Kpb Jsa Jsb Fya Fyb Jka Jkb Lea Leb P1 M N S s Lua Lub AHG CC CC CC
number Saline Subst.
PEG/AHG PEG/AHG

R1R1 1 + + 0 0 + 0 0 + 0 + 0 + + + + + 0 + + + + + + 0 + 2+ 2+ 0 3+

R1wR1 2 + + 0 0 + + + + 0 + 0 + 0 + + 0 0 + + + 0 + 0 0 + 3+ 3+ 2+

R2R2 3 + 0 + + 0 0 0 + 0 + 0 + + 0 0 + + 0 + 0 + 0 + 0 + 0 3+ 0 3+ 0 3+

R0r 4 + 0 + 0 + 0 0 + 0 + 0 + + 0 + + 0 + 0 + 0 + + 0 + 2+ 2+ 0 3+
Blood Groups and Serologic Testing

r’r 5 0 + + 0 + 0 0 + 0 + 0 + 0 0 0 + + 0 + + 0 + + 0 + 0 3+ 0 3+ 0 3+

r”r 6 0 0 + + + 0 0 + 0 + 0 + + + 0 + 0 0 0 0 + 0 + 0 + 0 3+ 0 3+ 0 3+

rr 7 0 0 + 0 + 0 + + 0 + 0 + + + + + 0 + + + + 0 + 0 + 3+ 3+ 2+

rr 8 0 0 + 0 + 0 0 + 0 + 0 + 0 + + 0 0 + + + + + 0 + + 2+ 2+ 0 3+

r’r” 9 0 + + + + 0 0 + 0 + 0 + 0 + + 0 0 + + 0 + 0 + 0 + 2+ 2+ 0 3+

rr 10 0 0 + 0 + 0 + + 0 + 0 + + 0 + + + 0 + + 0 + 0 0 + 2+ 2+ 2+

R1r 11 + + + 0 + 0 0 + 0 + 0 + + + 0 + 0 + + + + + + 0 + 2+ 2+ 0 3+

Patient
0 3+ 0 3+ 0 3+
Cells

Figure 10–10. Neutralization using Lewis’s substance in a serum containing anti-Leb and anti-Kell.
6888_Ch10_232-255 29/10/18 4:57 PM Page 245

Chapter 10 Detection and Identification of Antibodies 245

Commercial Reagents for Adsorption serum can be used in antibody identification testing. It is not
Human platelet concentrate is used to adsorb Bg-like anti- unusual for three to six aliquots of autologous RBCs to be
bodies from serum. In this method, the HLA antigens present used for autoadsorption. With some potent warm autoanti-
on platelets bind the HLA-related Bg antibodies,23 leaving bodies, the technologist may be unable to remove the autoan-
other specificities in the serum. Antibody identification can tibody completely. In this instance, reactivity will persist with
then be performed using the adsorbed serum. most if not all cells tested; hence, exclusion of alloantibodies
Rabbit erythrocyte stroma (RESt), which possesses I, H, using the adsorbed serum cannot be performed.
and IH-like structures, performs a similar function with Figure 10–11 illustrates steps in the autoadsorption
these cold-reacting autoantibodies. In this method, incubat- process. In the first image, the patient’s RBCs have been
ing the patient’s serum at 4°C with RESt will remove these coated with autoantibody in vivo. Autoantibody can also be
insignificant antibodies, which may interfere with the detec- seen free in the serum, along with an alloantibody. In the
tion of clinically significant, warm-reacting antibodies. Other second image, the autoantibody bound to the patient’s RBCs
antibody specificities, with the exception of anti-B and in vivo has been removed (post-treatment). Following this
anti-P1, remain unaffected by RESt adsorption; RESt also step, the RBCs are incubated with the patient’s serum (third
possesses structures similar to the B and P1 antigens and can image). The free autoantibody present in the patient’s serum
adsorb these two specificities as well.24 Because RESt may begins to adsorb onto the patient’s post-treated RBCs and is
adsorb anti-B, reverse grouping and crossmatching with removed from the serum. In the last image, the autoantibody
RESt-adsorbed serum should not be performed. has once again coated (in vitro) the patient’s RBCs, leaving
only alloantibody in the serum. The adsorbed serum can now
RBC Adsorption be used in antibody identification testing to reveal the alloan-
tibody that had previously been masked by the autoantibody.
Autoantibodies are commonly removed using RBC adsorp- Figure 10–12 shows an example of an antibody identification
tion techniques so that any underlying, clinically significant panel before and after warm autoadsorption. Anti-K is appar-
antibodies can be detected. When autoantibodies are present, ent in the postadsorption panel.
RBC adsorptions can be performed using allogeneic or au-
tologous RBCs; autologous RBCs can only be used when the Allogeneic Adsorption. When the volume of autologous RBCs is
patient has not been transfused within the last 3 months insufficient or when a patient has been recently transfused
(donor cells may adsorb alloantibodies) and the volume of (i.e., within the last 3 months), phenotypically matched or
RBCs is adequate. RBCs used to perform autoantibody ad- differential adsorptions may be employed instead of autoad-
sorption can be enzyme treated prior to adsorption to en- sorption to remove autoantibodies from patient serum; autol-
hance adsorption of the autoantibody. ogous adsorption is preferred over allogeneic adsorption for
Alloantibodies can also be removed using allogeneic RBC the removal of autoantibodies, since allogeneic RBCs have the
adsorption. Removal of alloantibodies using allogeneic RBC potential to adsorb an antibody to a high-prevalence antigen.
adsorption is not commonly performed and is usually only
employed to exclude common specificities when a previ- Phenotypically Matched Adsorption. If the patient’s phenotype is
ously identified, high-prevalence alloantibody is present known or can be determined (i.e., not transfused within the
or when multiple, common alloantibodies, of which one is last 3 months and DAT-negative RBCs are available), then
usually of higher prevalence (e.g., e antigen, G antigen), have phenotypically matched RBCs can be used for adsorption in
been previously identified or are thought to be present. RBCs place of autologous cells. For example, if a patient types as
used to perform alloantibody adsorption may or may not be R1R1, K–, Fya+, Fyb+, Jka–, Jkb+, S+, s–, then he or she may
enzyme treated and will depend on the specificity of the an- form anti-E, anti-c, anti-K, anti-Jka, and anti-s. Based on the
tibody being adsorbed. patient’s phenotype, the phenotypically matched donor cells
used for adsorption must be negative for E, c, K, Jka, and
Autoadsorption. Perhaps the simplest method for removing au- s antigens in order for those antibodies to remain in the
toantibodies is adsorption using the patient’s own RBCs (au- adsorbed serum.
tologous adsorption). The autologous RBCs are first washed
thoroughly to remove unbound antibody. The cells may then Differential Adsorption. Differential adsorption can be performed
be treated to remove any autoantibody coating the RBCs. when a patient’s phenotype is unknown and performing
Next, the RBCs are incubated with the patient’s serum for antigen-typing is not an option (i.e., positive DAT or transfused
30 to 60 minutes. The temperature of incubation will depend within the last 3 months). For this method, group O cells with
on the thermal range of the autoantibody being removed, the following phenotypes are used: R1R1, R2R2, and rr. One of
generally 4°C for cold-reacting autoantibodies and 37°C for these cells must also be negative for K, another negative for
warm-reacting autoantibodies. During incubation, the sample Jka, and another negative for Jkb. Prior to adsorption, the cells
should be mixed periodically. The serum is then harvested should be treated with an enzyme to render them negative
and should be tested against DAT-negative autologous cells. for antigens of the Duffy and MNSs systems; testing of the
If reactivity is observed with the DAT-negative autologous enzyme-treated RBCs should be performed prior to adsorp-
cells, further adsorption, using a new aliquot of autologous tion to ensure these antigens were destroyed. Following
RBCs, is required. If no reactivity is observed, then the au- adsorbing cell treatment, an aliquot of the patient’s serum is
toantibody has been completely adsorbed and the adsorbed mixed with an aliquot of each adsorbing cell and incubated
6888_Ch10_232-255 29/10/18 4:57 PM Page 246

246 PART II Blood Groups and Serologic Testing

RBC RBC RBC RBC

Initial sample. Patient’s cells have Adsorption step. Adsorption is complete.


Autoantibody (triangle) been treated to remove Treated patient cells are All of the autoantibody
has coated patient’s bound autoantibody incubated with patient has been removed from
RBCs in vivo, and is free from the cells. This is serum. Autoantibody the patient’s serum. The
in the serum. Alloantibody performed prior to present in the serum absorbed serum may be
(square) is also present performing the begins to absorb onto harvested and tested to
in the serum. adsorption step. the patient’s cells. identify the alloantibody
Alloantibody remains that is present.
in the serum.

Figure 10–11. Steps for performing an autoadsorption.

Donor Cell Peg/ Absorbed


D C c E e Cw K k Kpa Kpb Jsa Jsb Fya Fyb Jka Jkb Lea Leb P1 M N S s Lua Lub Xga CC CC
number IgG serum

R1R1 1 + + 0 0 + 0 0 + 0 + 0 + + 0 + + + 0 + + + + + 0 + + 2+ 0 3+

R1wR1 2 + + 0 0 + + + + 0 + 0 + + + 0 + 0 + + + 0 + + 0 + + 3+ 2+

R2R2 3 + 0 + + 0 0 0 + 0 + 0 + 0 + 0 + 0 + + + + + 0 0 + + 2+ 0 3+

R0r 4 + 0 + 0 + 0 0 + 0 + 0 + 0 0 + + 0 + + + 0 + 0 0 + 0 2+ 0 3+

r’r 5 0 + + 0 + 0 0 + 0 + 0 + + 0 + 0 0 0 0 0 + 0 + 0 + 0 2+ 0 3+

r”r 6 0 0 + + + 0 + + 0 + 0 + + + + + 0 + + + + + + 0 + + 3+ 2+

rr 7 0 0 + 0 + 0 0 + 0 + 0 + + + + + + 0 0 + + 0 + 0 + + 2+ 0 3+

rr 8 0 0 + 0 + 0 0 + 0 + 0 + 0 + 0 + 0 + + + 0 0 + 0 + + 2+ 0 3+

rr 9 0 0 + 0 + 0 0 + 0 + 0 + 0 + + 0 0 + 0 + + + + 0 + 0 2+ 0 3+

rr 10 0 0 + 0 + 0 + + 0 + 0 + 0 + + + 0 + + 0 + 0 + 0 + + 3+ 2+

R0r 11 + 0 + 0 + 0 0 + 0 + 0 + + + 0 + 0 + + 0 + 0 + 0 + + 2+ 0 3+

Patient 2+
Cells

Figure 10–12. Panel using warm autoadsorption technique.

at the temperature dictated by the thermal range of the au- Alteration in pH


toantibody for 30 to 60 minutes. The serum is then harvested Most antibodies react best at a neutral pH between 6.8 and
and should be tested against the corresponding adsorbing 7.2;25 however, some examples of anti-M demonstrate en-
cell. If reactivity is observed with the corresponding adsorb- hanced reactivity at a pH of 6.5.26 Acidifying the test system
ing cell, further adsorption, using a new aliquot of RBCs, is may aid in distinguishing anti-M from other antibodies.
required. If no reactivity is observed, antibody identification
panels can be performed separately on each aliquot of ad- Direct Antiglobulin Test and Elution
sorbed serum. See Chapter 21, “Autoimmune Hemolytic Ane- Techniques
mias” for a more complete discussion.
Allogeneic adsorption may also be performed when mul- Detection of antibodies coating RBCs is valuable when inves-
tiple, common alloantibodies are present in order to sepa- tigating suspected hemolytic transfusion reactions, HDFN,
rate the specificities or when a patient has a known and autoimmune and drug-induced hemolytic anemias. The
antibody to a high-prevalence antigen. In either case, the DAT is used to detect in vivo sensitization of RBCs. In the tube
adsorbing cell(s) must be antigen-positive for one sus- method, the patient’s RBCs are washed thoroughly to remove
pected specificity but negative for others. Following ad- any unbound antibody, and then AHG reagent is added. If IgG
sorption, the serum is tested to see which, if any, additional antibodies or complement are coating the RBCs, agglutination
alloantibodies are present. will be observed. If neither is present, no agglutination will be
6888_Ch10_232-255 29/10/18 4:57 PM Page 247

Chapter 10 Detection and Identification of Antibodies 247

observed. Coombs’ control cells are added to validate the neg- antibodies together.31,32 Elution methods that use organic
ative test. The DAT may also be performed using solid-phase solvents are time-consuming, and the chemicals pose several
adherence and gel methods. health and safety hazards; hence they are rarely used in the
When the IgG DAT is positive, the next step is to dissociate clinical laboratory.
the antibodies, using an elution method, from the RBC surface
to allow for identification; elution methods also concentrate
Antibody Titration
the bound antibodies. Methods used to dissociate the bound
antibody change the thermodynamics of the environment, Once an antibody is identified, it is sometimes useful (e.g.,
change the attractive forces between antigen and antibody, or pregnancy, certain types of antibodies) to quantify the
change the structure of the RBC surface. Elution methods amount or concentration that is present. This can be accom-
commonly used in the clinical laboratory include heat, Lui plished by performing an antibody titration. Techniques that
freeze-thaw, and acid. Once an elution method has been per- employ flow cytometry, radioimmunoassay, or enzyme-linked
formed, the resulting solution, known as an eluate, can be immunoassay can also be used; however, these techniques
tested against an RBC panel to identify the antibody(-ies) are not readily available in every laboratory.
present. When testing the eluate, the last wash must be tested To perform antibody titration, two-fold serial dilutions of
in parallel to detect the presence of unbound antibody. The serum are prepared and tested against reagent RBCs that ex-
last wash should be nonreactive. If the last wash is reactive, press the target antigen. The titer level is the reciprocal of
the eluate results are invalid. the greatest dilution in which agglutination of 1+ or greater
is observed. A score may also be assigned, based on the
Temperature-Dependent Methods strength of reactivity. Each reaction is given a value, and the
score is determined by adding up the individual values (see
The simplest elution methods involve changing the tempera-
Table 10–5).
ture of the antigen-antibody environment; the temperature
After determining the initial titer, the specimen should be
can be increased to 56°C (heat method) or decreased to –18°C
frozen. When new specimens are submitted for titration test-
or colder (Lui freeze-thaw method). Both of these methods
ing, the initial specimen should be tested in parallel to control
are total elution methods, which means the RBCs are de-
variability among technologists’ technique (e.g., pipetting,
stroyed by the process and cannot be used for further test-
grading reactions) and the relative strength of the target anti-
ing.27,28 Temperature-dependent elutions are best at detecting
gen on the reagent RBCs used in testing. A comparison of the
IgG antibodies directed against antigens of the ABO system.
current specimen’s results and the initial specimen’s current
results should be made. A four-fold or greater increase in titer
pH Method
(reactivity in two or more additional tubes) or an increase in
A common, relatively quick and easy total elution method for score of 10 or more is considered to be clinically significant.
the detection of non-ABO IgG antibodies is acid elution.29,30 Table 10–5 shows an example of titer-level results that indi-
In this method, the washed antibody-coated cells are mixed cate a significant increase in antibody levels.
with a glycine acid solution at a pH of 3. The antigen-antibody When performing the antibody titer, careful preparation
bond is disrupted, and the antibody is released into the acidic of dilutions is necessary. Contamination from a tube with a
supernatant. The supernatant is harvested, and the pH is neu- higher antibody concentration can lead to falsely elevated
tralized so that antibody identification testing can take place. titer-level results. Changing pipette tips between each tube
when preparing the dilutions and interpreting or reading
Organic Solvent Methods from the most diluted tube to the least diluted can help avoid
this problem.
Several organic solvents, including dichloromethane, xylene, The phenotype of the RBCs selected for use in titration
and ether, have been used in total elution methods to detect testing should be consistent throughout the series of titer-
non-ABO IgG antibodies. These solvents act on the lipids in level studies. If a cell with homozygous antigen expression
the RBC membrane to reduce surface tension and lead to the was used for the initial titer, then all subsequent titer speci-
reversal of the van der Waals forces that hold antigens and mens should be tested against a homozygous cell. The

Table 10–5 Titer and Score of Anti-D*


Dilution 1:2 1:4 1:8 1:16 1:32 1:64 1:128 Titer and Score
Previous sample 2+ 2+ 1+ 0 0 0 0 Titer 8

Score 8 8 5 0 0 0 0 Score 21

Current sample 3+ 3+ 2+ 2+ 1+ 0 0 Titer 32

Score 10 10 8 8 5 0 0 Score 41

*Score values: 4+ = 12, 3+ = 10, 2+ = 8, 1+ = 5


6888_Ch10_232-255 29/10/18 4:57 PM Page 248

248 PART II Blood Groups and Serologic Testing

method used must also be consistent. It has been reported taking this approach, it is important to remember that units
that titers using the gel method are more sensitive than those that appear crossmatch compatible, but type antigen-positive
using the tube method and therefore result in a higher titer cannot be given to the patient. If specimen quantity is limited
level.33 The technologist must ensure the test systems are as or if the antibody is no longer detectable in the serum, units
identical as possible in order to make valid comparisons be- must be antigen-typed first, then crossmatched.
tween samples. Knowing the frequency of the antigen(s) in the population
Titer-level studies are useful in monitoring obstetric pa- is helpful when determining the number of units that should
tients who have an IgG antibody that may cause HDFN. An be antigen-typed to find a sufficient number to fulfill the cross-
increase in antibody titer level during pregnancy suggests that match request. To determine the number of units to test, divide
the fetus is antigen-positive and therefore at risk of develop- the number of units requested by the frequency of antigen-
ing HDFN. An increasing titer level may indicate more inva- negative individuals in the population. For example, if a cross-
sive testing needs to be performed. An antibody titer may also match for 2 units of RBCs is requested on a patient who has
be used to help differentiate immune anti-D from passively anti-E, the calculation would be 2 (units requested)/0.70 (the
acquired anti-D due to RhIg administration. The titer level frequency of E-negative individuals). The result is 2.8, meaning
associated with RhIg administration is rarely above 4.34 that 3 units would need to be typed for the E antigen in order
Performing a titer is one way to confirm the presence of an- to theoretically find 2 E-negative units. When multiple speci-
tibodies that have previously been known as HTLA (high titer, ficities are present, the frequencies of antigen-negative individ-
low avidity) antibodies. These antibodies, directed against uals in the population are multiplied together. For example, if
high-prevalence antigens, are observed at the AHG phase of 2 E-negative, c-negative units are required, the calculation
testing with weakly positive reactions. The weak reactions per- would be 2/(0.70 × 0.20) = 14.3. Fourteen or 15 units would
sist through extensive dilutions (as high as 2,048).35 Examples theoretically have to be antigen-typed for E and c in order to
of these antibodies include anti-Ch, anti-Rg, anti-Csa, anti-Yka, find 2 units that are negative for both antigens.
anti-Kna, anti-McCa, and anti-JMH. These antibodies are not When the prevalence (i.e., high prevalence) or number of
usually clinically significant, but may mask antibodies that are antigens that must be negative makes it difficult to find suit-
clinically significant. able units, rare donor registries may be consulted. These reg-
istries maintain a list of rare donors and can put out a request
Providing Compatible Blood Products to blood donor facilities for rare units.
When an Alloantibody Is Present In certain cases, knowing the donor’s race is helpful when
selecting units for antigen testing because the frequency of
The relative difficulty in providing compatible blood products some antigens varies between races. For example, when
is determined by the frequency of the antigen in the popula- searching for units that are Fya-negative, it may be prudent
tion and by the clinical significance of the antibody. If the an- to screen black donors, as 90% will be negative for Fya com-
tibody does not cause decreased survival of antigen-positive pared with only 34% of whites.
RBCs, then use of random blood products that are crossmatch- Some transfusion medicine experts have proposed that
compatible is acceptable. Examples of such antibodies include certain patient populations, particularly sickle cell and beta-
anti-M, anti-N, anti-P1, anti-Lea, and anti-Leb.36 thalassemia patients, receive units that are phenotypically
When a patient specimen contains a clinically significant matched even if they have not formed any alloantibodies.38,39
antibody(-ies) or a patient has a history of a clinically signif- These repeatedly transfused patient populations seem more
icant antibody(-ies), units for transfusion must be antigen- likely to make alloantibodies than the general population,
negative. The crossmatch technique must also demonstrate hence the proactive approach. Another approach is to trans-
compatibility at the AHG phase.37 If the patient specimen is fuse units that are phenotypically matched for Rh antigens
plentiful, the technologist may choose to crossmatch units, (C, c, E, and e) and the Kell antigen, as these antigens are
then antigen-type those that are crossmatch compatible. If the most immunogenic.40

CASE STUDIES
Case 10-1

Multiple Antibodies
Suspect multiple antibodies when all or most of the screen and panel cells are positive, but reactions are at different
strengths or in different phases and the autocontrol is negative. When using inclusion techniques, it may appear that no
single antibody accounts for all of the positive reactions observed.
Resolution may include performing a selected cell panel to exclude certain specificities. Several sets of selected cells may
need to be tested to narrow the list of possible antibodies (refer to Fig. 10–7 for an example of a selected cell panel). Alter-
natively, the use of enzyme-treated panel RBCs may aid in the identification of antibodies if one of the suspected antigens is
destroyed by enzymes and another suspected antigen exhibits enhanced expression or is unaffected by enzyme treatment
(refer to Fig. 10–9). In other cases, neutralization or adsorption techniques may prove useful in separating multiple antibodies.
6888_Ch10_232-255 29/10/18 4:57 PM Page 249

Chapter 10 Detection and Identification of Antibodies 249

Refer to Figure 10-9 to answer the following questions:


1. Looking at the original testing (PEG/AHG column), is there reactivity that suggests that this specimen may contain
more than one antibody?
2. Of the specificities not excluded in the original testing, which antigens are destroyed by enzymes and which are
enhanced?
3. Comparing the original reactions to those of the enzyme-treated panel, which antibodies appear to be present in this
specimen?
4. What additional testing could be performed to eliminate anti-C?

Case 10-2

Antibody to a High-Prevalence Antigen


High-prevalence antigens are those that are present in almost all individuals. Suspect an antibody to a high-prevalence
antigen when all screen and panel cells are positive, with reactions in the same phase and at the same strength, along
with a negative autocontrol (refer to Fig. 10–13). If a panel cell is negative for a high-prevalence antigen it is usually
indicated on the antigen profile sheet or on the manufacturer’s extended typing list, which usually accompanies the
panel set.
Among the antibodies included in this category are the so-called HTLA antibodies. It is not necessary to determine the
specificity of an HTLA antibody, but removing these antibodies is usually necessary to identify any underlying alloanti-
bodies. Some HTLA antibodies, notably anti-Ch and anti-Rg, may be neutralized by normal serum, which contains com-
plement. Routine blood bank enzymes will destroy anti-Ch, anti-Rg, and anti-JMH, whereas anti-Kna and anti-McCa are
destroyed by dithiothreitol (DTT).
Finding compatible blood for patients with an antibody to a high-prevalence antigen may be a challenge. Autologous
donations should be encouraged. Other sources of antigen-negative blood may include family members and the rare donor
registry. Fortunately, because these antigens do occur so frequently, it is rare to find a patient with an antibody to one of
them. Refer to Figure 10–13.
1. What antibody appears to be present in this specimen?
2. What additional testing needs to be performed to confirm this identification?
3. What additional patient information would be useful when resolving a case such as this?

Donor Cell Peg/


number D C c E e Cw K k Kpa Kpb Jsa Jsb Fya Fyb Jka Jkb Lea Leb P1 M N S s Lua Lub Xga IgG CC

R1R1 1 + + 0 0 + 0 0 + 0 + 0 + + 0 + + + 0 + + + + + 0 + + 2+
R1wR1 2 + + 0 0 + + + + 0 + 0 + + + 0 + 0 + + + 0 + + 0 + + 2+
R2R2 3 + 0 + + 0 0 0 + 0 + 0 + 0 + 0 + 0 + + + + + 0 0 + + 2+
R0 r 4 + 0 + 0 + 0 0 + 0 + 0 + 0 0 + + 0 + + + 0 + 0 0 + 0 2+
r’r 5 0 + + 0 + 0 0 + 0 + 0 + + 0 + 0 0 0 0 0 + 0 + 0 + 0 2+
r”r 6 0 0 + + + 0 + + 0 + 0 + + + + + 0 + + + + + + 0 + + 2+
rr 7 0 0 + 0 + 0 0 + 0 + 0 + + + + + + 0 0 + + 0 + 0 + + 2+
Cob +
rr 8 0 0 + 0 + 0 0 + 0 + 0 + 0 + 0 + 0 + + + 0 0 + 0 + + 2+
rr 9 0 0 + 0 + 0 0 + 0 + 0 + 0 + + 0 0 + 0 + + + + 0 + 0 2+
rr
Yt(a) – 10 0 0 + 0 + 0 + + 0 + 0 + 0 + + + 0 + + 0 + 0 + 0 + + 0 3+

R 0r 11 + 0 + 0 + 0 0 + 0 + 0 + + + 0 + 0 + + 0 + 0 + 0 + + 2+
Patient 0 3+
Cells

Figure 10–13. Panel performed on a patient with an antibody to a high-prevalence antigen.


6888_Ch10_232-255 29/10/18 4:57 PM Page 250

250 PART II Blood Groups and Serologic Testing

Case 10-3

Antibody to a Low-Prevalence Antigen


Low-prevalence antigens are antigens that occur infrequently (generally less than 1%) in the population.41 Hence, anti-
bodies to these antigens are uncommon because exposure to the antigen is rare. Antibodies to these antigens should be
suspected when an antiglobulin crossmatch is incompatible and the antibody screen is negative and when other reasons
for reactivity, such as ABO incompatibility or positive donor DAT, have been eliminated. These antibodies may also be
suspected when an infant has a positive DAT, there is no known ABO blood group discrepancy between mother and infant,
and the mother has a negative antibody screen. Because these antigens are infrequent, finding antigen-negative units for
crossmatch is usually not difficult. Refer to Figure 10–14.
1. What low-prevalence antigen is found on cell 4?
2. What additional testing should be performed to confirm the identity of the antibody?

Case 10-4

Cold-Reacting Autoantibodies
Most adult sera contain low titers of cold-reacting autoantibodies, most notably autoanti-I, autoanti-H, and autoanti-IH.
These antibodies are usually IgM and of no clinical significance. They are troublesome in that they may interfere with the
detection of clinically significant alloantibodies, resulting in prolonged workups and delayed transfusions.
Cold-reacting autoantibodies may be suspected when the screen cells/panel cells and the autocontrol are all positive at
the immediate spin phase, and reactivity gets weaker or disappears with incubation at 37°C (refer to Fig. 10–15).
Certain cold autoantibodies activate complement and may be detected at the AHG phase when using complement-
containing AHG reagent. Very strong cold autoantibodies may also be detected at the AHG phase when using monospecific
anti-IgG AHG reagent. As a result, cold autoantibodies may appear as weakly reacting IgG antibodies in the aforementioned
instances.
Although it is not usually necessary to determine the specificity of the cold autoantibody, testing against additional cells
may confirm its presence. Cord blood cells that lack the I antigen are of particular value for this purpose. A cold panel consisting
of group O adult cells (H and I antigens), group O cord cells (H and i antigens), group A1 adult cells (I antigen), and an auto-
control may be tested at 4°C to determine the specificity of the cold autoantibody (refer to Fig. 10–15 and Fig. 10–16).
1. Why is a cold autoantibody suspected in this case?
2. Why may a technologist test cord blood cells in a case such as this?
3. How might a technologist avoid interference from cold autoantibodies?

Donor Cell LISS/


D C c E e Cw K k Kpa Kpb Jsa Jsb Fya Fyb Jka Jkb Lea Leb P1 M N S s Lua Lub Xga IS 37°C CC
number AHG

R1R1 1 + + 0 0 + 0 0 + 0 + 0 + + 0 + + + 0 + + + + + 0 + + 0 0 0 3+

R1wR1 2 + + 0 0 + + + + 0 + 0 + + + 0 + 0 + + + 0 + + 0 + + 0 0 0 3+

R2R2 3 + 0 + + 0 0 0 + 0 + 0 + 0 + 0 + 0 + + + + + 0 0 + + 0 0 0 3+

R0r 4 + 0 + 0 + 0 0 + 0 + + + 0 0 + + 0 + + + 0 + 0 0 + 0 0 1+ 2+

r’r 5 0 + + 0 + 0 0 + 0 + 0 + + 0 + 0 0 0 0 0 + 0 + 0 + 0 0 0 0 3+

r”r 6 0 0 + + + 0 + + 0 + 0 + + + + + 0 + + + + + + 0 + + 0 0 0 3+

rr 7 0 0 + 0 + 0 0 + 0 + 0 + + + + + + 0 0 + + 0 + 0 + + 0 0 0 3+

rr 8 0 0 + 0 + 0 0 + 0 + 0 + 0 + 0 + 0 + + + 0 0 + 0 + + 0 0 0 3+

rr 9 0 0 + 0 + 0 0 + 0 + 0 + 0 + + 0 0 + 0 + + + + 0 + 0 0 0 0 3+

rr 10 0 0 + 0 + 0 + + 0 + 0 + 0 + + + 0 + + 0 + 0 + 0 + + 0 0 0 3+

R0r 11 + 0 + 0 + 0 0 + 0 + 0 + + + 0 + 0 + + 0 + 0 + 0 + + 0 0 0 3+

Patient
0 0 0 3+
Cells

Figure 10–14. Panel performed on a patient with an antibody to a low-prevalence antigen.


6888_Ch10_232-255 29/10/18 4:57 PM Page 251

Chapter 10 Detection and Identification of Antibodies 251

Donor Cell LISS/


D C c E e Cw K k Kpa Kpb Jsa Jsb Fya Fyb Jka Jkb Lea Leb P1 M N S s Lua Lub Xga IS 37 CC
number AHG

R1R1 1 + + 0 0 + 0 0 + 0 + 0 + + 0 + + + 0 + + + + + 0 + + 2+ w+ 0 3+

R1wR1 2 + + 0 0 + + + + 0 + 0 + + + 0 + 0 + + + 0 + + 0 + + 2+ w+ 0 3+

R2R2 3 + 0 + + 0 0 0 + 0 + 0 + 0 + 0 + 0 + + + + + 0 0 + + 2+ w+ 0 3+

R0r 4 + 0 + 0 + 0 0 + 0 + 0 + 0 0 + + 0 + + + 0 + 0 0 + 0 2+ w+ 0 3+

r’r 5 0 + + 0 + 0 0 + 0 + 0 + + 0 + 0 0 0 0 0 + 0 + 0 + 0 2+ w+ 0 3+

r”r 6 0 0 + + + 0 + + 0 + 0 + + + + + 0 + + + + + + 0 + + 2+ w+ 0 3+

rr 7 0 0 + 0 + 0 0 + 0 + 0 + + + + + + 0 0 + + 0 + 0 + + 2+ w+ 0 3+

rr 8 0 0 + 0 + 0 0 + 0 + 0 + 0 + 0 + 0 + + + 0 0 + 0 + + 2+ w+ 0 3+

rr 9 0 0 + 0 + 0 0 + 0 + 0 + 0 + + 0 0 + 0 + + + + 0 + 0 2+ 1+ 0 3+

rr 10 0 0 + 0 + 0 + + 0 + 0 + 0 + + + 0 + + 0 + 0 + 0 + + 2+ 1+ 0 3+

R0r 11 + 0 + 0 + 0 0 + 0 + 0 + + + 0 + 0 + + 0 + 0 + 0 + + 2+ 1+ 0 3

I neg Cord 0 0 0 3+
Cell

Patient 2+ 1+ 0 3+
Cells

Figure 10–15. Panel performed on a patient with a cold autoantibody.

CELL IS RT 18°C 4°C


A1 0 0 1+ 2+
A2 1+ 2+ 4+ 4+
B 0 0 2+ 3+
O adult 2+ 3+ 4+ 4+
O cord 0 0 1+ 2+

Figure 10–16. Cold antibody screen performed on an AB patient with Auto 0 0 1+ 2+


autoanti-IH.

Case 10-5

Warm-Reacting Autoantibodies
Workups for warm-reacting autoantibodies are some of the most challenging to perform. Warm autoantibodies are un-
common, but they may occur secondary to chronic lymphocytic leukemia, lymphoma, systemic lupus erythematosus,
and other autoimmune diseases or because of drug therapy. Suspect a warm autoantibody when all cells, including the
autocontrol, are reactive at the AHG phase and at the same strength. If an underlying alloantibody is present, cells that
possess that antigen may react stronger than those that are antigen-negative. It is of primary importance to remove the
warm autoantibody to detect any underlying alloantibodies. It is not necessary to determine the specificity of the autoan-
tibody, although many seem to be directed against Rh antigens.
Warm autoantibodies are IgG antibodies that coat the patient’s RBCs in vivo and are present as free antibody in the pa-
tient’s serum once all the corresponding antigen sites (i.e., the antigen sites the autoantibody is directed against) on the
patient’s RBCs have been bound. Adsorption techniques are used to remove the antibody from the serum.
Finding RBC units that are compatible with a patient who has a warm autoantibody may be difficult, since the autoan-
tibody frequently interferes with this testing. If after adsorption no clinically significant antibodies are detected, random
donor units can be chosen for transfusion. Crossmatch testing protocols for these units will vary based on facility protocol.
Protocols include, but are not limited to the following: IS crossmatch testing using neat serum only (IAT crossmatch
testing not performed); an electronic crossmatch only (IAT crossmatch testing not performed); or IS crossmatch testing
using neat serum and IAT crossmatch testing using adsorbed serum. If clinically significant antibodies are present,
antigen-negative units must be crossmatched at IS and IAT.
6888_Ch10_232-255 29/10/18 4:57 PM Page 252

252 PART II Blood Groups and Serologic Testing

Refer to Figure 10–12. This panel was performed on a male lymphoma patient who was last transfused 2 years ago. He
has not received any transplants and is not currently on any medications.
1. What does the pattern of reactivity in the original panel (PEG/IgG column) suggest?
2. How can you differentiate this pattern from that of an antibody to a high-prevalence antigen?
3. What type of adsorption procedure is recommended in this case?
4. Are any alloantibodies present in this specimen?
5. How would you prepare RBC products for this patient?

SUMMARY CHART
 The purpose of the antibody screen is to detect unex-  Coombs’ control cells are reagent RBCs coated with
pected antibodies, which are found in approximately human IgG antibody, which are added to all AHG-
0.8% to 2% of the general population. The antibodies negative tube tests to ensure adequate washing
may be classified as immune (the result of RBC stimu- occurred and that the AHG reagent is present and
lation in the patient), passive (transferred to the patient functional in the test system.
through blood products or derivatives), or naturally oc-  Gel and solid-phase adherence methods are alterna-
curring (the result of environmental factors). Antibod- tives to tube testing. These methods may be automated
ies may also be classified as alloantibodies, directed at to increase efficiency.
foreign antigens, or autoantibodies, directed at one’s  The antibody exclusion method rules out possible
own antigens. antibodies based on antigens that are present on
 A clinically significant antibody is one that results in negatively reacting cells.
the shortened survival of RBCs possessing the target  Conclusive antibody identification is achieved when the
antigen. These antibodies are typically IgG antibodies serum is reactive with at least three antigen-positive cells
that react best at 37°C or in the AHG phase of testing (i.e., reagent cells that express the corresponding anti-
and are known to cause hemolytic transfusion reac- gen) and negative with at least three antigen-negative
tions and/or HDFN. cells (i.e., reagent cells that do not express the correspond-
 Screen cells are commercially prepared group O RBC ing antigen). This can also be achieved using two
suspensions obtained from individual donors who are antigen-positive cells and three antigen-negative cells or
phenotyped for the most commonly encountered and two antigen-positive cells and two antigen-negative cells.
clinically significant RBC antigens.  The DAT detects RBCs that were sensitized with anti-
 RBCs from a homozygous individual have a double body in vivo. Elution methods are used to free anti-
dose of a single antigen, which results from the inher- body from the cell surface to allow for identification.
itance of two genes that code for the same antigen,  The relative quantity of an antibody can be determined
whereas heterozygous individuals carry only a single by testing serial two-fold dilutions of serum against
dose each of two antithetical antigens (each gene codes antigen-positive RBCs; the reciprocal of the highest serum
for a different antigen.) dilution showing agglutination is the antibody titer.
 Antibodies in the Kidd, Duffy, Lutheran, Rh, and MNSs  The calculation used to determine the number of
blood group systems show dosage and yield stronger random donor units that should be antigen-typed in
reactions against RBCs with homozygous expression order to theoretically provide the requested number of
of their corresponding antigen. antigen-negative RBC units for patients with an anti-
 Enhancement reagents, such as LISS and PEG, are so- body involves dividing the number of antigen-negative
lutions added to serum and cell mixtures prior to in- units requested by the frequency of antigen-negative
cubation at 37ºC to promote antigen-antibody binding individuals in the donor population.
or agglutination.
6888_Ch10_232-255 29/10/18 4:57 PM Page 253

Chapter 10 Detection and Identification of Antibodies 253

4. The physician has requested 2 units of RBCs for patient


Review Questions
DB, who has two antibodies, anti-L and anti-Q. The fre-
quency of antigen L is 45%, and the frequency of antigen
1. Based on the following phenotypes, which pair of cells
Q is 70% in the donor population. Approximately how
would make the best screening cells?
many units will need to be antigen-typed for L and Q to
a. Cell 1: Group A, D+C+c–E–e+, K+, Fy(a+b–), Jk(a+b–), fill the request?
M+N–S+s–
a. 8
Cell 2: Group O, D+C–c+E+e–, K–, Fy(a–b+), Jk(a–b+),
b. 12
M–N+S–s+
c. 2
b. Cell 1: Group O, D–C–c+E–e+, K–, Fy(a–b+), Jk(a+b+),
d. 7
M+N–S+s+
Cell 2: Group O, D+C+c–E–e+, K–, Fy(a+b–), Jk(a+b–), 5. Anti-Sda has been identified in patient ALF. What sub-
M–N+S–s+ stance would neutralize this antibody and allow detection
c. Cell 1: Group O, D+C+c+E+e+, K+, Fy(a+b+), Jk(a+b+), of other alloantibodies?
M+N–S+s+ a. Saliva
Cell 2: Group O, D–C–c+E–e+, K–, Fy(a+b–), Jk(a+b+), b. Hydatid cyst fluid
M+N+S–s+ c. Urine
d. Cell 1: Group O, D+C+c–E–e+, K+, Fy(a–b+), Jk(a–b+), d. Human breast milk
M–N+S–s+
Cell 2: Group O, D- C–c+E+e–, K–, Fy(a+b–), Jk(a+b–), 6. Patient JM appears to have a warm autoantibody. She was
M+N–S+s– transfused 2 weeks ago. What would be the next step per-
formed to identify any alloantibodies that might be in her
2. Antibodies are excluded using RBCs that are homozygous serum?
for the corresponding antigen because: a. Acid elution
a. Antibodies may show dosage b. Warm autoadsorption using autologous cells
b. Multiple antibodies may be present c. Warm differential adsorption
c. It results in a P value of 0.05 for proper identification d. RESt adsorption
of the antibody
d. All of the above 7. What is the titer and score for this prenatal anti-D titer?
(Refer to Fig. 10–17.)
3. A request for 8 units of RBCs was received for patient LF. a. Titer = 64; score = 52
The patient has a negative antibody screen, but 1 of the b. Titer = 1:32; score = 15
8 units was 3+ incompatible at the AHG phase. Which of c. Titer = 64; score = 21
the following antibodies may be the cause? d. Titer = 32; score = 52
a. Anti-K
b. Anti-Lea
c. Anti-Kpa
d. Anti-Fyb

Figure 10–17. Anti-D titer results for


Question 7. Saline
Dilution 1:1 1:2 1:4 1:8 1:16 1:32 1:64 1:128 1:256 1:512 1:1024 control
Results 4+ 4+ 3+ 2+ 1+ 1+ 0 0 0 0 0 0
6888_Ch10_232-255 29/10/18 4:57 PM Page 254

254 PART II Blood Groups and Serologic Testing

For Questions 8 through 10, refer to Figure 10–18. 11. Which of the following methods may be employed to re-
move IgG antibodies that are coating a patient’s red
8. Select the antibody(ies) most likely responsible for the
blood cells?
reactions observed.
a. Adsorption
a. Anti-E and anti-K
b. Elution
b. Anti-Fya
c. Neutralization
c. Anti-e
d. Titration
d. Anti-Jkb
12. A technologist has decided to test an enzyme-treated
9. What additional cells need to be tested to be 95% confi-
panel of RBCs against a patient’s serum. Which of the
dent that the identification is correct?
following antibody pairs could be separated using this
a. Three e-negative cells that react negatively and one
technique?
additional e-positive cell that reacts positively
a. Anti-Jka and anti-Jkb
b. One additional E-positive, K-negative cell to react
b. Anti-S and anti-Fya
positively and one additional K-positive, E-negative
c. Anti-D and anti-C
cell to react positively
d. Anti-Jka and anti-Fya
c. Two Jkb homozygous positive cells to react posi-
tively and one Jkb heterozygous positive cell to react 13. An antibody demonstrates weak reactivity at the AHG
negatively phase when the tube method is used with no enhance-
d. No additional cells are needed ment reagent and monospecific anti-IgG AHG reagent.
When repeating the test, which of the following actions
10. Using the panel (Fig. 10–18), select cells that would make
may increase the strength of the positive reactions?
appropriate controls when typing for the C antigen.
a. Adding an enhancement reagent, such as LISS or PEG
a. Cell number 1 for the positive control and cell num-
b. Decreasing the incubation time from 30 minutes to
ber 2 for the negative control
10 minutes
b. Cell number 1 for the positive control and cell num-
c. Employing the prewarm technique
ber 6 for the negative control
d. Decreasing the incubation temperature to 18°C
c. Cell number 2 for the positive control and cell num-
ber 4 for the negative control
d. Cell number 4 for the positive control and cell num-
ber 5 for the negative control

Donor Cell
D C c E e Cw K k Kpa Kpb Jsa Jsb Fya Fyb Jka Jkb Lea Leb P1 M N S s Lua Lub Xga IS 37 AHG CC
number

R1R1 1 + + 0 0 + 0 0 + 0 + 0 + + + + 0 0 0 + + + + + 0 + + 0 0 0 3+

R1wr 2 + + + 0 + + + + 0 + 0 + + 0 0 + 0 0 + + 0 + + 0 + + 0 0 3+

R1R1 3 + + 0 0 + 0 0 + 0 + 0 + 0 + + + 0 0 + 0 + 0 + 0 + + 0 0 0 3+

R2R2 4 + 0 + + 0 0 0 + 0 + 0 + + 0 + + + 0 + + 0 + 0 0 + + 0 2+ 3+

r’r 5 0 + + 0 + 0 0 + 0 + 0 + 0 0 0 + + 0 0 + 0 + + 0 + 0 0 0 0 3+

r”r” 6 0 0 + + 0 0 0 + 0 + 0 + + 0 0 + 0 0 0 0 + 0 + 0 + + 0 2+ 3+

rr 7 0 0 + 0 + 0 + + 0 + 0 + + 0 0 + 0 + + 0 + 0 + 0 + + 0 0 3+

rr 8 0 0 + 0 + 0 0 + 0 + 0 + 0 + + 0 0 0 + + 0 + 0 0 + + 0 0 0 3+

r’r” 9 0 0 + 0 + 0 0 + 0 + 0 + + 0 + 0 0 + + + + 0 + + + 0 0 0 0 3+

R1r 10 + + + 0 + 0 0 + 0 + 0 + + 0 + + 0 + + + 0 0 + 0 + + 0 0 0 3+

R0r 11 + 0 + 0 + 0 0 + 0 + 0 + + + 0 + 0 + + + 0 0 + 0 + + 0 0 0 3+

Patient
0 0 0 3+
Cells

Figure 10–18. Panel for Question 8.


6888_Ch10_232-255 29/10/18 4:57 PM Page 255

Chapter 10 Detection and Identification of Antibodies 255

References 22. Hillyer CD, Shaz BH, Winkler AM, Reid M. Integrating molec-
ular technologies for red blood cell typing and compatibility
1. Tormey CA, Fisk J, Stack G. Red blood cell alloantibody fre- testing into blood centers and transfusion services. Transfus
quency, specificity, and properties in a population of male mil- Med Rev. 2008;22(2):117-32.
itary veterans. Transfusion. 2008;48(10):2069-76. 23. Aster RH, Miskovich BH, Rodey GE. Histocompatibility anti-
2. AABB. Standards for blood banks and transfusion services. gens of human plasma. Localization to HLD-3 lipoprotein frac-
31st edition. Bethesda (MD): AABB; 2018. tion. Transplantation. 1973;16(3):205-10.
3. Judd WJ, Luban NL, Ness PM, Silberstein LE, Stroup M, Wid- 24. Yuan S, Fang A, Davis R, Siplon CJ, Goldfinger D. Immunoglob-
mann FK. Prenatal and perinatal immunohematology: recom- ulin M red blood cell alloantibodies are frequently adsorbed by
mendations for serologic management of the fetus, newborn rabbit erythrocyte stroma. Transfusion. 2010;50(5):1139-43.
infant, and obstetric patient. Transfusion. 1990;30(2):175-83. 25. South, SF. Use of the direct antiglobulin test in routine testing.
4. ISCT-EBMT. International standards for cellular therapy prod- Current application and interpretation of the direct antiglobulin
uct collection, processing, and administration accreditation test. Wallace ME, Levitt JS, editors. Arlington (VA): American
manual. 5th ed. Omaha, (NE), Barcelona, (Spain): Foundation Association of Blood Banks; 1988. p. 25.
for the Accreditation of Cellular Therapy/Joint Accreditation 26. Issitt PD, Anstee DJ. Applied blood group serology. 4th ed.
Committee ISCT-EBMT; 2012. Durham (NC): Montgomery Scientific Publications; 1998. p. 470.
5. Issitt PD, Anstee DJ. Applied blood group serology. 4th ed. 27. Landsteiner K, Miller CP. Serologic studies on the blood of pri-
Durham (NC): Montgomery Scientific Publications; 1998. p. 132. mates: II. The blood group of anthropoid apes. J Exp Med.
6. Schonewille H, Haack HL, van Zijl, AM. RBC antibody persis- 1925;42(6):853-62.
tence. Transfusion. 2000;40(9):1127-31. 28. Feng CS, Kirkley KC, Eicher CA, de Jongh DS. The Lui elution
7. Tormey CA, Stack G. The persistence and evanescence of blood technique: a simple and efficient method for eluting ABO an-
group alloantibodies in men. Transfusion. 2009;49(3):505-12. tibodies. Transfusion. 1985;25(5):433-4.
8. Fisher, RA. Statistical methods and scientific inference. 2nd ed. 29. Rekkvig OP, Hannestad, K. Acid elution of blood group anti-
Edinburgh (Scotland): Oliver and Boyd; 1959. bodies from intact erythrocytes. Vox Sang. 1977:33(5):280-5.
9. Harris RE, Hochman HG. Revised p values in testing blood 30. Judd WJ, Johnson ST, Storry J. Judd’s methods in immunohe-
group antibodies. Transfusion. 1986;26(6):494-9. matology. 3rd ed. Bethesda (MD): AABB Press; 2008. p. 126.
10. Kanter MH, Poole G, Garratty G. Misinterpretation and mis- 31. van Oss CJ, Absolom DR, Neumann AW. The “hydrophobic
application of p values in antibody identification: the lack of effect”: Essentially a van der Waals interaction. Colloid Polym
value of a p value. Transfusion. 1997;37(8):816-22. Sci. 1980:258(4):424-7.
11. Method 2-20. Dissociating IgG by chloroquine for antigen test- 32. van Oss CJ, Absolom DR, Neumann AW. Applications of net
ing of red cells with a positive DAT. In: Fung MK, Eder AF, Spi- repulsive van der Waals forces between different particles,
talnik SL, Westhoff CM (eds). Technical Manual. 19th ed. macromolecules, or biological cells in liquids. Colloid Polymer
Bethesda, MD: AABB; 2017. Sci. 1980:1(1):45-56.
12. Judd WJ, Johnson ST, Storry J. Judd’s methods in immunohe- 33. Ciavarella D, Pate L, Sorenson E. Serologic comparisons of anti-
matology. 3rd ed. Bethesda (MD): AABB Press; 2008. p. 166. body detection and titration methods: LISS, PEG, gel [abstract].
13. Judd WJ, Johnson ST, Storry J. Judd’s methods in immunohe- Transfusion. 2002;42 Suppl:S108.
matology. 3rd ed. Bethesda (MD): AABB Press; 2008. p. 169. 34. Kennedy MS, Delaney M, Scrape S. Perinatal issues in transfu-
14. Method 2-22. Separating transfused from autologous red cells sion practice. In: Fung MK, editor. Technical Manual. 18th ed.
by simple centrifugation. In: Fung MK, Eder AF, Spitalnik SL, Bethesda (MD): AABB; 2014. p. 566.
Westhoff CM (eds). Technical manual. 19th ed. Bethesda 35. Schulman IA, Petz LD. Red cell compatibility testing: clinical
(MD): AABB; 2017. significance and laboratory methods. In: Petz, LD, Swisher SN,
15. Morelati F, Revelli N, Musella A, Paccapelo C, Villa M, Drago F, Kleinman S, editors. Clinical practice of transfusion medicine.
et al. Automated red cell phenotyping [abstract]. Transfusion. 3rd ed. New York: Churchill Livingstone; 1996. p. 199-244.
2001;41Suppl:S110. 36. Downes KA, Shulman IA. Pretransfusion testing. In: Fung MK,
16. Lowe L. Use of MTS IgG gel cards for red blood cell antigen editor. Technical manual. 18th ed. Bethesda (MD): AABB; 2014.
typing [abstract]. Transfusion. 2002;42 Suppl:S109. p. 381.
17. Bennett PR, Le Van Kim C, Dolon Y, Warwick RM, Cherif- 37. AABB. Standards for blood banks and transfusion services.
Zahar B, Fisk NM, et al. Prenatal determination of fetal RhD 30th ed. Bethesda (MD): AABB; 2016.
type by DNA amplification. N Engl J Med. 1993;329(9):607-10. 38. Tahhan HR, Holbrook CT, Braddy LR, Brewer LD, Christie JD.
18. Palacajornsuk P, Halter C, Isakova V, Tarnawski M, Farmer J, Antigen-matched donor blood in the transfusion management of
Reid M, et al. Detection of blood group genes using multiplex patients with sickle cell disease. Transfusion. 1994;34(7):562-9.
SNaPshot method. Transfusion. 2009;49(4):740-9. 39. Matteocci A, Palange M, Dionisi MG, Castagna K, Collaretti A,
19. Wagner FF, Bittner R, Petershofen EK, Doescher A, Müller TH. Mazzoli C, et al. Retrospective study on the red cell alloimmu-
Cost-efficient sequence-specific priming–polymerase chain re- nization after multiple blood transfusions [abstract]. Transfu-
action screening for blood donors with rare phenotypes. sion. 2001;(41 Suppl):S112.
Transfusion. 2008;48(6):1169-73. 40. Vichinsky EP, Luban NL, Wright E, Olivieri N, Driscoll C,
20. Karpasitou K, Drago F, Crespiatico L, Paccapelo C, Truglio F, Pegelow CH, et al. Prospective RBC phenotype matching in a
Frison S, et al. Blood group genotyping for Jka/Jkb, Fya/Fyb, S/s, stroke-prevention trial in sickle cell anemia: a multicenter
K/k, Kpa/Kpb, Jsa/Jsb, Coa/Cob, and Lua/Lub with microarray transfusion trial. Transfusion. 2001;41(9):1086-92.
beads. Transfusion. 2008;48(3):505-12. 41. Coghlan G. Antibodies to low-incidence antigens. Transfus
21. Keller, MA. The role of red cell genotyping in transfusion med- Apher Sci. 2009;40(3):199-202.
icine. Immunohematology. 2015;31(2):49-52.
6888_Ch11_256-267 31/10/18 9:50 AM Page 256

Chapter 11
Pretransfusion Testing
Laurie A. Wolf, MLS(ASCP)SBBCM

Introduction Selection Criteria Issuance of Blood Components


Regulatory and Accreditation Platelets, Thawed Plasma, The Future of Transfusion Medicine
Requirements and Cryoprecipitate Conclusion
Specimen Collection Crossmatch Tests Case Studies
Positive Patient Identification and Serologic Crossmatch Case 11-1
Specimen Labeling Computer Crossmatch Case 11-2
Specimen Requirements Special Circumstances Summary Chart
Compatibility Tests Emergent Transfusions Review Questions
Review of Records Massive Transfusions References
Recipient Testing Neonatal Transfusions
Selection of Donor Units Intrauterine Transfusions
Donor Unit Testing

OBJECTIVES
1. Describe positive patient identification and specimen labeling requirements.
2. Describe the specimen requirements for pretransfusion testing.
3. Describe the importance of comparing historical records with current results.
4. List the tests required in compatibility tests for recipients.
5. List criteria for appropriate selection of donor units.
6. Compare serologic and computer crossmatching procedures.
7. Resolve incompatibilities in crossmatch tests.
8. Explain compatibility tests and appropriate blood component selection in special circumstances.
9. Describe the required steps in blood component issuance.
10. Discuss the future of transfusion medicine.

Introduction Regulatory and Accreditation


Requirements
Pretransfusion testing can be defined as the use of serologic
principles and tests to ensure compatibility and prevent an All blood component manufacturers and distributors as well
immune-mediated hemolytic transfusion reaction. Prior to as medical laboratories in the United States are regulated by
this chapter, topics discussed included fundamental con- the federal government. The Food and Drug Administration
cepts, blood group serology, and specific serologic testing (FDA) defines blood components as pharmaceuticals because
procedures. This chapter focuses on pretransfusion activities, of the intent of their use to cure, mitigate, treat, or prevent
including proper specimen collection, pretransfusion and disease.1,2 Under the auspices of the Clinical Laboratory Im-
compatibility tests, special clinical situations, and patient provement Amendments of 1988 (CLIA ’88), the Centers for
safety. Medicare and Medicaid (CMS) have the authority to regulate

256
6888_Ch11_256-267 31/10/18 9:50 AM Page 257

Chapter 11 Pretransfusion Testing 257

pretransfusion testing.2 Facilities providing blood compo- the United States, clerical errors and ABO-incompatible trans-
nents and laboratory services are subject to inspections by ei- fusions continue to occur and have potentially serious, clin-
ther entity at any given time. An in-depth review of regulatory ical consequences.4,5 According to the 2016 Serious Hazards
requirements is addressed in detail in Chapter 27. of Transfusion Report (SHOT),4 74.5% of hemovigilance re-
In addition to maintaining regulatory compliance, many ports analyzed were related to error incidents. In addition,
facilities participate in quality driven programs to achieve 264 potential ABO-incompatible transfusions were reported
accreditation. The American Association of Blood Banks, now as near misses. Three ABO-incompatible transfusions were
known simply as AABB, is an accrediting organization whose reported, with two resulting in major morbidity. A study of
goals include enhancing the quality and safety of services pro- transfusion errors in the state of New York over a 10-year
vided by blood banks and transfusion services.3 Table 11–1 period demonstrated that erroneous administrations occurred
outlines the steps in pretransfusion testing.3 Adequate pre- with a frequency of 1 in 19,000 units of red blood cells
transfusion testing does not guarantee normal survival of trans- administered.6 Errors include phlebotomy and testing errors,
fused red blood cells in the recipient’s circulation, but strict administration to the wrong recipient, or issuance of the
adherence to all facets of pretransfusion testing and donor unit wrong donor unit. Transfusion errors continue to be a signif-
selection is imperative to ensure safe transfusion therapy. icant risk necessitating systems to prevent failures in patient
identification, specimen labeling, and administration.
Specimen Collection In order to prevent patient misidentification and admin-
istration failures, most institutions require patients to wear
Facilities must strictly follow policies and procedures to a facility-generated ID wristband upon admission until dis-
identify patients and collect properly labeled specimens to charge. This wristband must contain two unique identifiers,
mitigate misidentification errors and improve patient safety. typically the patient’s first and last names and date of birth
or medical record number. The Joint Commission’s National
Positive Patient Identification and Specimen Patient Safety Goals require two unique identifiers to iden-
Labeling tify patients and ensure that correct medicine and treatment
are administered.7 Other information such as driver’s license
While pulmonary complications are the leading cause of
number, social security number, and photographic ID can
death associated with transfusion in the United Kingdom and
be used for further verification. If the patient does not have
a wristband or if the patient’s identity is unknown, some
form of positive identification must be attached to the pa-
Table 11–1 Steps in Pretransfusion Testing
tient. Acceptable alternatives to wristbands are ankle bands
Steps in Pretransfusion Testing or labels affixed to an exposed part of the patient’s body
Request for transfusion such as the head or torso of a draped surgical patient. For a
patient whose identity is unknown, a facility-generated fic-
Identification of transfusion recipient and blood specimen collected titious name with unique number identifier can be assigned
Testing of transfusion recipient’s blood specimen: and later merged with the patient’s information.
• Evaluation of specimen for testing suitability Prior to specimen collection, the phlebotomist must
• ABO group confirm the wristband information is accurate and contains
• Rh type (Weak D testing is optional when testing the patient) all required information. Common practice includes asking
• Screening for unexpected antibodies to red cell antigens the patient to state his or her name and date of birth, if they
• Antibody identification if unexpected antibodies are detected are coherent. Requests for testing must be authorized by an
• Comparison of current and previous test results, and any discrepan-
cies shall be investigated and appropriate action taken before a unit ordering provider and include two unique identifiers to
is issued for transfusion positively identify the patient.3 Wristband information and
requests must be legible and indelible. Any discrepancies
Donor RBC unit testing: between the two sets of information must be completely
• ABO group confirmation and Rh type confirmation for Rh-negative resolved prior to collecting the pretransfusion specimen.
RBC units
Once collected, the specimen must be accurately labeled at
Donor red blood cell unit selection: the bedside. Patient information on the label, wristband, and
• Selection of components of ABO group and Rh type that are com- request must be in complete agreement. Furthermore, speci-
patible with the transfusion recipient and with any unexpected men draw date and time and the collecting individual must be
allogeneic antibodies traceable back to the collection of the patient sample.3 Standard
Compatibility testing (crossmatch): operating procedures must be adhered to when the collected
• Serologic specimen is received into the laboratory. Testing personnel
• Computer or electronic must confirm that the labeling information on the collected
sample is complete, accurate, and in accordance with the test-
Labeling of blood or blood components with the recipient’s identifying
information and issue ing requested. If misidentification of the sample is suspected
for any reason, the specimen should be recollected.
6888_Ch11_256-267 31/10/18 9:50 AM Page 258

258 PART II Blood Groups and Serologic Testing

Systems such as barcode readers and radio frequency


identification (RFID) are available and can be implemented
as part of a facility-wide patient safety program. Commer-
cially manufactured identification systems using preprinted
tags and numbers (Figs. 11–1, 11–2, and 11–3) are useful as
well. Regardless of system, it is crucial that protocols are in
place to positively identify patients and that phlebotomy
procedures are defined. All personnel involved must be
trained and have competency assessed.

Specimen Requirements

Either anticoagulated or clotted specimens are acceptable for


pretransfusion testing.2 Anticoagulated specimens are often
preferred due to ease of handling. Red blood cells from an
anticoagulated sample are ideal for preparing a uniform cell
suspension for testing. Clotted red blood cells may require
additional washing steps to minimize interference in test
interpretations. In addition, serum may contain small fibrin
clots that can be difficult to distinguish from true agglutina-
tion. Anticoagulated specimens can inactivate complement- Figure 11–2. Confirm that sample and requisition form agree.
binding antibodies due to calcium chelation disrupting the
complement cascade. Despite this possibility, the benefits of
using anticoagulated specimens outweigh this potential risk.
Intravenous line (IV) contamination is unacceptable, and
the use of hemolyzed samples should be avoided. IV contam-
inants from the infusion fluid dilute otherwise detectable
antibodies in the specimen and may produce erroneous results.
Specimens should be collected from the arm opposite the
IV catheter and never above it. If venous access is limited and
the specimen must be collected from the IV line, the infusion
fluid must be stopped and the line flushed. It is recommended
that an amount of blood equal to three or more times the dead
space of the catheter be discarded.8 The use of hemolyzed spec-
imens can mask antigen-antibody–mediated hemolysis and
create difficulties in evaluating test results. If a new specimen
cannot be obtained, results must be interpreted carefully.

Figure 11–3. Commercially manufactured identification systems. Pretransfusion


comparison of recipient armband and donor unit bag.

The age of the specimen used in pretransfusion testing is


crucial. If a patient has been transfused or pregnant within
the past 3 months or if the patient’s history is unknown, a
specimen must be collected every 3 days for antibody screen-
ing and compatibility testing. The date of the collection
is considered day 0, and the specimen expires at midnight
on day 3.3 For example, if a specimen was collected on a
Monday (day 0), it may be used for transfusion on Tuesday
(day 1), Wednesday (day 2), and Thursday (day 3) until
11:59 p.m. The 3-day testing window minimizes the risk
Figure 11–1. Commercially manufactured identification systems. Compare of an adverse reaction due to an emerging alloantibody in a
preprinted label with the patient armband. recipient that has been recently transfused. Some facilities
6888_Ch11_256-267 31/10/18 9:50 AM Page 259

Chapter 11 Pretransfusion Testing 259

extend this testing window in surgical patients who have anti-B for detecting the antigens on the red blood cells
been scheduled in advance to prevent delays on the day of and A1 and B cells for confirming the naturally occurring
the procedure. However, the transfusion and pregnancy his- antibodies in the plasma. All ABO discrepancies must be
tories must be known with certainty and neither can have resolved in order to transfuse appropriately. If an ABO
occurred in the previous 3 months.2 discrepancy cannot be resolved prior to the need for trans-
Pretransfusion specimens and donor unit segments are fusion, group O red blood cells should be selected. For
typically stored at 2° to 8°C and must be retained in the a detailed review of the ABO blood group system and
event that additional testing is warranted. The patient sam- discrepancy resolution, refer to Chapter 6.
ple and a segment from the donor unit must be retained
post-transfusion for at least 7 days.3 This allows for the in- Rh Typing
vestigation of any adverse events related to transfusion, such Rh typing is performed to detect the presence or absence of
as a delayed hemolytic transfusion reaction. the D antigen. Licensed anti-D reagents are monoclonal
blends of IgM and IgG components.2 IgM promotes direct
Compatibility Tests agglutination of red blood cells at room temperature,
whereas IgG is utilized in the indirect antiglobulin test (IAT)
Compatibility tests are performed to prevent an immune- for the detection of the weak D antigen. An Rh control is
mediated hemolytic transfusion reaction. Compatibility tests indicated when spontaneous agglutination of red blood
include: determining the recipient’s ABO group and Rh type, cells is suspected, such as in patients typing as AB-positive.
screening for any unexpected antibodies, and crossmatching In addition, an Rh control must be tested in parallel with
the donor unit with the recipient’s plasma. anti-D reagent in the indirect antiglobulin test for weak
D determination. If the Rh control is positive at any phase
Review of Records of testing, the tests results are invalid and additional testing
Current results such as ABO group and Rh type, detection of is required. The test for weak D is optional in transfusion
clinically significant antibodies, significant adverse events, recipients but is required for donor units during manufac-
and special transfusion requirements must be compared to turing.2,3 For a detailed review of the Rh blood group system
historical records.3 Patient identification and testing errors and typing reagents, refer to Chapter 7.
may be found with the comparison of historical and current Rh-negative recipients should receive Rh-negative red
ABO/Rh results. For patients with no historical records, many blood cells to prevent the production of anti-D alloantibod-
facilities have implemented an additional specimen draw for ies. Because weak D red blood cells are a result of a reduced
a blood type confirmation. Additionally, many antibody titers number of antigen sites, patients possessing weak D antigens
decrease to undetectable levels, making a thorough history are not at risk for developing anti-D. Patients with the partial
review essential. Between 30% and 35% of antibodies reach D phenotype, however, possess hybrid genes resulting in the
undetectable levels within 1 year, and nearly 50% become loss of D epitopes on the antigen.10 Typically, these patients
undetectable after 10 years.9 Any discrepancy between his- type Rh-positive and their partial D phenotype is unknown
torical records and current test results must be resolved prior until the development of an anti-D alloantibody. Partial
to transfusion. D phenotypes are common in the African American commu-
nity and create challenges in transfusion.
Recipient Testing
Antibody Screening
ABO grouping, Rh typing, and screening for unexpected Antibodies to blood group antigens must be detected and
antibodies must be performed within 3 days prior to the their clinical significance assessed. Clinically significant an-
scheduled transfusion except in the previously mentioned tibodies have been associated with red blood cell destruc-
situations for which the pretransfusion testing window may tion, hemolytic transfusion reactions, and hemolytic
be extended.3 An accurate medical history including current disease of the fetus and newborn (HDFN). Commercially
medications, recent blood transfusions, and previous preg- prepared 2-cell and 3-cell antibody screening sets are avail-
nancies may be helpful in explaining any unusual results. able and must meet licensing requirements. The FDA man-
All tests should be performed according to the reagent man- dates that red blood cells used in antibody detection tests
ufacturer’s instructions. Serologic reactions must be graded must express the following antigens: D, C, E, c, e, K, k, Fya,
and recorded immediately. Refer to Color Plate 1 to aid in Fyb, Jka, Jkb, Lea, Leb, P1, M, N, S, and s.11 Reagent red
the grading of reactions. blood cells cannot possibly contain every blood group anti-
gen. Low-incidence antigens such as V, Cw, Kpa, or Jsa are
ABO Grouping not required because they rarely cause development of the
The ABO blood group continues to be the most important corresponding antibody.
blood group in transfusion and transplantation due to The incidence of alloimmunization in all transfusion re-
immunogencity.10 ABO grouping requires licensed reagents cipients is approximately 2% to 5% and significantly higher
and can be performed using tubes, column-agglutination in multiply transfused populations.12 Typically, clinically
technology, or in microtiter plates used in solid-phase red cell significant antibodies react at 37°C and are detectable at the
adherence tests. Required reagents include anti-A and antiglobulin phase of testing. However, clinically significant
6888_Ch11_256-267 31/10/18 9:50 AM Page 260

260 PART II Blood Groups and Serologic Testing

antibodies in the early stages of development or that are ABO grouping, Rh typing, a weak D test if the initial Rh typ-
primarily of the IgM isotype may be detected at lower tem- ing result is negative, and a screen for antibodies to common
peratures such as in the room temperature phase of testing. blood group antigens. Because the weak D antigen has the
Regardless of the phase in which the clinically significant ability to stimulate anti-D production, blood donations that
antibody is detected, transfusion of antigen-negative red test weak D-positive must be clearly labeled as Rh-positive
blood cells is the transfusion requirement. Transfusion to prevent accidental transfusion to Rh-negative recipients.
requirements vary among facilities when clinically insignif- In addition, blood suppliers are required to test for infectious
icant antibodies are detected. Most consider donor units disease markers to prevent disease transmission. Required
that are crossmatch compatible by the indirect antiglobulin infectious disease testing on donor units includes HBV DNA,
test (IAT) to be safe.13 Table 11–2 outlines common blood HBsAG, HBc antibodies, HCV antibodies, HCV RNA, HIV1/2
group antibodies according to their preferred phase of reac- antibodies, HIV-1 RNA, HTLV I/II antibodies, WNV RNA,
tivity and clinical significance. and a serologic test for syphilis. In addition, each donor must
Many testing methods are available, including tube tests be tested at least once for antibodies to Trypanosoma cruzi.3
with or without enhancement, column-agglutination tech- Recently, the FDA provided guidance and recommended uni-
nology, and solid-phase red blood cell adherence tests with versal testing for the Zika virus. Once acceptability criteria
effectiveness varying between each. Tube testing allows are met, the units are distributed to transfusion facilities.
for the chemical modification of the patient’s red blood To ensure proper labeling and testing of the donor unit,
cells or plasma and the flexibility of testing at various phases. the transfusing facility is required to confirm the ABO group-
Column-agglutination technology and solid-phase red blood ing on all units and the Rh typing on all Rh-negative units
cell adherence tests are available as automated testing plat- upon receipt from the supplier. Access to the donor unit is
forms, creating standardization and workload efficiency. obtained through one of the integrally attached segments.
All tests should be performed with licensed reagents accord-
Selection of Donor Units ing to the manufacturer’s instructions. Any discrepancies
must be reported to the collecting facility.3
A common goal shared by blood suppliers and transfusion
services is to provide a safe and efficacious blood component Selection Criteria
that benefits the intended recipient. A thorough review of
donor selection and blood component preparation is covered ABO/Rh
in succeeding chapters. ABO antigens on the donor unit red blood cells must be
compatible with the naturally occurring anti-A and/or
Donor Unit Testing
anti-B in the recipient’s plasma. If whole blood is the only
Blood suppliers are required to perform a battery of tests on option, the product and the recipient must be ABO identical
blood components prior to distribution. Tests performed are due to the plasma portion of the blood component. However,
most whole blood collections are further processed into in-
dividual components such as packed red blood cells and
plasma, allowing for flexibility in donor unit selection. Refer
to Table 11–3 for ABO group selection of red blood cells for
Table 11–2 Antibodies to Blood Group
transfusion.
Antigens
Rh-negative red blood cells may be given to Rh-positive re-
Blood Blood Group Optimal Clinical cipients. However, this limited resource should be reserved for
Group Antigens Technique Significance Rh-negative recipients, especially women of childbearing age,
Rh D, C, E, c, e, f, V, Cw IAT Significant to prevent alloimmunization. Approximately 80% of healthy,
Rh-negative recipients transfused Rh-positive red blood cells
Kell K, k, Kpa, Kpb, Jsa, Jsb IAT Significant produce anti-D antibodies.14 In emergent situations where Rh-
Duffy Fya, Fyb IAT Significant negative inventory is limited, the risks associated with not

Kidd Jka, Jkb IAT Significant

Lewis Lea RT, 37 C, IAT Rarely Significant Table 11–3 ABO Group Selection Order for
Transfusion of RBCs
Leb RT, 37 C, IAT Insignificant
Recipient 1st 2nd 3rd 4th
P1 P1 RT Insignificant ABO Group Choice Choice Choice Choice
M 4 C, RT, IAT Rarely Significant AB AB A B O
N 4 C, RT, IAT Insignificant A A O
MNSs
S RT, IAT Significant B B O
s IAT Significant O O
6888_Ch11_256-267 31/10/18 9:50 AM Page 261

Chapter 11 Pretransfusion Testing 261

transfusing may outweigh the risks of potential alloimmuniza- Immediate Spin Crossmatch
tion. Facilities must have policies to address these special cir- If no clinically significant antibodies are detected and there
cumstances, which usually include approval from the medical is no history of antibody, a serologic test to detect ABO
director and notification of the ordering provider. Refer to the incompatibility is sufficient.3 Recipient plasma is mixed
Emergent Transfusions section for more details. with cells from the donor unit, immediately centrifuged, and
Antigen-Negative observed for agglutination and/or hemolysis. Absence of
either indicates ABO compatibility, whereas positive results
If a patient has a history of, or has detectable clinically
require further investigation.
significant antibodies, donor units selected for transfusion
Incompatible immediate spin crossmatches may occur
must be negative for the corresponding antigen(s). In certain
for many reasons and must be resolved prior to issuing units
clinical situations, it might be necessary to phenotypically
for transfusion. Possible reasons for incompatibilities and
match red blood cells in order to prevent antibody produc-
subsequent resolutions include the following:
tion or decrease the need for costly workups in the future.
Predicted phenotypes by DNA analysis are affordable and 1. Incorrect ABO grouping of recipient or of donor unit
useful in the selection of donor units. For example, the rate selected
of alloimmunization is approximately 30% in patients with • Repeat ABO grouping of the recipient specimen and
sickle cell disease who have been chronically transfused.12 confirm with a new specimen, if necessary.
Limited red blood cell matching for the Rh, Kell, and Duffy • Verify the donor unit is ABO-compatible with the
antigens has proven to be effective in decreasing the rate of recipient and repeat donor confirmation testing, if
new antibody formation to 0.5% in patients with sickle cell necessary.
disease.15 In multiple myeloma patients treated with daratu- 2. Cold-reactive allo- or autoantibody in the plasma not
mumab, phenotypically matched units for all common blood detected in antibody detection tests
group antigens may decrease the need for costly serologic • If the antibody screen was negative, perform antibody
workups. Daratumumab is an anti-CD38 therapeutic agent identification at room temperature since the incompat-
that reacts with CD38 on reagent red blood cells. The inter- ibility is detectable at that phase of testing. Include
ference makes exclusion of alloantibodies in antibody iden- an autocontrol to distinguish between a cold-reactive
tification procedures impossible without the treatment of alloantibody versus autoantibody.
DTT or trypsin. The recommendation by the AABB is to • Prewarming of the test system may be indicated to
obtain a phenotype by DNA and match red blood cells for circumvent the reactivity in the presence of a cold-
all potential clinically significant alloantibodies to avoid the reactive antibody. Select donor units based upon the
need for extended workups.16 antibody identified and the clinical significance
assessment.
Platelets, Thawed Plasma, and Cryoprecipitate • Depending upon the recipient’s blood type, it may be
useful to include A1, A2, and B cells when testing the
No compatibility testing is required for the transfusion of plasma to rule out the presence of immune-mediated
platelets, thawed plasma, and cryoprecipitate.17 Either a cur- or passively acquired ABO antibodies. A thorough stem
rent or a historical ABO grouping is sufficient. Rh typing is cell or solid organ transplantation history may be help-
ignored because these components contain very few red ful in resolving this type of discrepancy.
blood cells as a result of collection methods or frozen 3. Abnormalities in the recipient’s plasma
storage. ABO-identical platelet units should be selected • Rouleaux mimics agglutination and may be present in
whenever possible to ensure adequate platelet survival. diseases such as multiple myeloma. See Color Plate 1
Plasma units should be compatible with the recipient’s ABO for the characteristic “stacking of coins” observed in
red blood cell antigens. However, many facilities provide rouleaux formation.
group A thawed plasma emergently to trauma patients with • Rouleaux is detectable only when plasma is present.
an unknown blood type due to the lack of group AB plasma Perform saline replacement and repeat the immediate
availability.18 If ABO-identical or ABO-compatible compo- spin crossmatch. Saline replacement technique involves
nents are unavailable, incompatible plasma should be limited removing the plasma from the immediate spin cross-
to prevent hemolysis in the recipient. Refer to the Emergent match and replacing with saline. Rouleaux will disap-
Transfusions section for more details. pear while true agglutination will not.
Crossmatch Tests
Antiglobulin Crossmatch
Serologic Crossmatch Once a clinically significant antibody has been detected, an
antiglobulin crossmatch is required. The donor unit selected
A serologic crossmatch consists of mixing recipient plasma should be antigen-negative for the corresponding alloantibody
with cells directly obtained from the donor unit to detect identified. The antiglobulin crossmatch consists of an imme-
ABO or blood group antibody incompatibilities. A nonreac- diate spin crossmatch with the recipient’s plasma and cells
tive serologic crossmatch indicates the donor unit is com- from the donor unit. The test system is then incubated at
patible and safe for transfusion. 37°C and completed with the antiglobulin test. Antiglobulin
6888_Ch11_256-267 31/10/18 9:50 AM Page 262

262 PART II Blood Groups and Serologic Testing

crossmatching may be performed in tube tests, typically with 4. Donor unit has a positive direct antiglobulin test (DAT).
the addition of LISS or PEG enhancements or in column- • Perform a DAT on the donor unit. A positive DAT
agglutination and solid-phase red cell adherence tests. Observ- should be reported to the collecting facility.
able reactivity and/or hemolysis at any phase of testing indicate
Refer to Table 11–4 for additional information about
incompatibility. Subsequent testing should include an anti-
causes of positive pretransfusion test results for additional
globulin testing phase and the addition of an autocontrol.
information.
Possible reasons for incompatibilities include the following:
1. New alloantibody is present in recipient’s plasma. Computer Crossmatch
• Perform antibody identification using the method
at which the incompatibility was detected to identify ABO compatibility can be verified electronically via a vali-
additional antibody specificities. dated, on-site computer system provided acceptable criteria
2. Alloantibody to a low-incidence present is on the donor have been met. Electronic crossmatching eliminates the
unit red blood cells. need for a serologic crossmatch, which reduces sample
• Perform more extensive antibody identification to volume requirements and testing time. Two determinations
detect antibody to low-incidence antigens. of the recipient’s ABO grouping must be on file, one of
3. Warm-reactive autoantibody is present in the recipient’s which is from the current specimen collection. The confir-
plasma. matory ABO grouping may be from historical records or
• Autoantibodies present in the plasma may require from an additional collection. The recipient must not have
adsorptions to circumvent reactivity. detectable clinically significant antibodies or a history of
• Donor units may be tested with adsorbed plasma and antibodies. The laboratory information system (LIS) must
a release signed by the ordering provider. The waiver contain donor unit information to include the donation
is necessary because of the antibody interference caus- identification number, component name, ABO group,
ing an incompatible crossmatch interpretation. Rh type, donor confirmation typing, and the interpretation

Table 11–4 Causes of Positive Pretransfusion Tests*


Negative Antibody Screen, Incompatible Immediate Spin Crossmatch

• Donor red blood cells are ABO incompatible.

• Donor red blood cells are polyagglutinable.

• Anti-A1 is the serum of an A2 or A2B individual.

• Other alloantibodies reactive at room temperature (e.g., anti-M).

• Rouleaux formation.

• Cold autoantibodies (e.g., anti-I).

• Passively acquired anti-A or anti-B.

Negative Antibody Screen, Incompatible Antiglobulin Crossmatch

• Donor red blood cells have a positive direct antiglobulin test.

• Antibody reacts only with red blood cells having strong expression of a particular antigen (e.g., dosage) or variation in antigen strength (e.g., P1).

• Antibody to a low-incidence antigen on the donor red blood cells.

• Passively acquired anti-A or anti-B.

Positive Antibody Screen, Compatible Crossmatches

• Autoanti-IH (autoanti-H) or anti-LebH and non-group O units are selected.

• Antibodies dependent on reagent red blood cell diluent.

• Antibodies demonstrating dosage and donor red blood cells are from heterozygotes (i.e., expressing a single dose of antigen).

• Donor unit is lacking corresponding antigen.

Positive Antibody Screen, Incompatible Crossmatches, Negative Autocontrol

• Alloantibody(-ies)
6888_Ch11_256-267 31/10/18 9:50 AM Page 263

Chapter 11 Pretransfusion Testing 263

Table 11–4 Causes of Positive Pretransfusion Tests*—cont’d


Positive Antibody Screen, Incompatible Crossmatches, Positive Autocontrol, Negative Direct Antiglobulin Test

• Antibody to ingredient in enhancement media or enhancement-dependent autoantibody.

• Rouleaux formation.

Positive Antibody Screen, Incompatible Crossmatches, Positive Autocontrol, Positive DAT

• Alloantibody causing either a delayed serologic or hemolytic transfusion reaction.

• Passively acquired autoantibody (e.g., intravenous immune globulin).

• Cold- or warm-reactive autoantibody.

• Rouleaux formation.

*Causes depend on serologic methods used.


Reprinted with permission from Roback, J (ed): Technical Manual, 16th ed. American Association of Blood Banks, Bethesda, MD, 2008.

of compatibility with the recipient. A method must exist to


verify that data entry is correct prior to the release of blood
components. In addition, the LIS must contain logic that
alerts the user of any discrepancies between recipient
records and the donor unit.3 Figure 11–4 illustrates the No STOP—No
Crossmatch
steps required for the electronic crossmatch. requested?
crossmatch
requested

Special Circumstances
Yes

Emergent Transfusions Collect a current


specimen and Current
No ABO/Rh and
Prior to the completion of pretransfusion testing, urgent sit- perform the
ABO/Rh and antibody screen
uations may arise when it is medically necessary to transfuse antibody on file?
blood components. The transfusion service must have poli- screen tests
cies addressing the selection of appropriate donor units to Yes
prevent transfusion delays. If the blood type of the recipient Perform a second
ABO/Rh type on a
is unknown, group O red blood cells are required. ABO No new specimen or
Two ABO/Rh
group-specific red blood cells may be issued once a blood types on file? repeat the ABO/Rh
type is obtained.3 Rh-negative red blood cell units are pre- testing on the
same specimen
ferred but may be limited to women of childbearing age in Yes for confirmation
order to conserve inventory.
Issuing group A thawed plasma to traumatically injured
patients where ABO grouping is unknown is becoming wide- Resolve the Yes Any ABO/Rh
ABO/Rh
spread practice in the United States. Group A thawed plasma discrepancy
discrepancies?
is readily available at most facilities, and its use in the initial
resuscitation showed no increase in mortality.18 Emergently No
released units must clearly indicate that compatibility testing
has not been completed at the time of issue. Pretransfusion
Positive antibody
testing must be completed expeditiously and any incompat- screen or history of Yes Not eligible
ibility reported to the medical director. An emergency release for electronic
a clinically significant
crossmatch
waiver signed by the ordering provider accepting responsi- antibody?
bility for the transfusion due to critical needs is required.
No
Massive Transfusions

Massive transfusion is defined as the administration of Perform the


8 to 10 red blood cell units to an adult patient in less than electronic
crossmatch
24 hours or acute administration of 4 to 5 units within
1 hour.2 The American College of Surgeons recommends
transfusion of red blood cells, thawed plasma, and platelets in Figure 11–4. Electronic crossmatch requirements.
6888_Ch11_256-267 31/10/18 9:50 AM Page 264

264 PART II Blood Groups and Serologic Testing

a ratio of 1:1:1 for effective blood component resuscitation.19 The transfusionist is responsible for confirming the in-
The goal of treatment is to restore blood volume rapidly to tended recipient’s identification and the donor unit informa-
a level adequate to maintain hemostasis, oxygen-carrying tion at the bedside. A two-person verification process must
capacity, oncotic pressure, and biochemical parameters.20 If be utilized prior to initiating the transfusion. An automated
pretransfusion testing has not been completed prior to a mas- identification system such as barcoding may be used if two
sive transfusion, emergency release procedures must be fol- individuals are not available.7 Blood components are admin-
lowed. Special transfusion requirements, such as providing istered according to the facility’s policy, with any adverse
antigen-negative or irradiated units, should be met if time per- event related to the transfusion investigated.
mits. If the risk of delaying transfusion outweighs the special
requirement need, the medical director should be contacted. The Future of Transfusion Medicine
The future of transfusion medicine will focus on patient blood
Neonatal Transfusions
management (PBM) programs and personalized medicine.
Pretransfusion testing may be abbreviated in infants less than PBM is an evidence-based approach to reducing the need for
4 months of age. For ABO grouping, only anti-A and anti-B blood components and reducing health-care costs.21 PBM is
reagents are required. Either the infant’s or mother’s plasma the concept that the detection and treatment of anemia, the
may be used for antibody screening and any necessary com- assessment of potential blood loss, and the use of restrictive
patibility testing. If a clinically significant antibody is identi- transfusion triggers can decrease the need for blood compo-
fied, the donor unit selected must be either antigen-negative nent transfusion.22 In 1999, a multicenter, randomized clinical
for the corresponding antibody or antiglobulin crossmatch trial was published in the New England Journal of Medicine an-
compatible until the antibody is no longer demonstrable in alyzing transfusion requirements in critical care patients. The
the infant’s plasma. If an infant has a negative antibody screen TRICC trial explored whether restrictive or liberal transfu-
and will only receive group O red blood cell transfusions, re- sions in critical patients resulted in different outcomes. The
peat testing may be omitted for the remainder of the hospital recommendation of this study was that thresholds for trans-
admission. Non-group O red blood cells may be issued to a fusion of critically ill patients be lowered from 10 to 7 g/dL,
non-group O infant. However, an antiglobulin test to detect which further supports PBM strategies.23
passively acquired maternal anti-A and/or anti-B is required.3 Personalized medicine involves individualization of treat-
ment for patients. Advances in blood group genomics has
Intrauterine Transfusions contributed to the ability to provide this service in transfu-
sion medicine. Phenotypically matching red blood cell units
Intrauterine transfusions (IUT) are indicated in severe cases to prevent alloimmunization in patients with underlying dis-
of fetal anemia. Intrauterine transfusions are performed by in- eases such as sickle cell disease and warm autoimmune he-
serting a needle into the umbilical vein using high-resolution molytic anemia is becoming the standard of care. In a world
sonography to guide the procedure. Fetal samples may be where extensive travel is expected, a national database to in-
obtained for testing to include blood typing, direct anti- clude individual patient information would be beneficial in
globulin test, antigen typing, and bilirubin levels. Typically, providing personalized care. The database should include
the fetal blood type is unknown, and group O Rh-negative any alloantibodies identified, predicted phenotypes by DNA,
red blood cells should be selected. The unit should be fresh and any special transfusion requirements such as irradiation,
(<7 days old), leukocyte-reduced, irradiated to prevent trans- IgA deficiency, and CMV status.
fusion-associated graft-versus-host disease, and negative for
hemoglobin associated with sickling.2 In cases in which ma- Conclusion
ternal antibody is present, the unit should be antigen-negative
and crossmatch compatible at the antiglobulin phase of The transfusion service partners with almost all clinical areas
testing. to ensure safe transfusions. This partnership is responsible
for all aspects of transfusion, including specimen collection,
pretransfusion and compatibility testing, blood administra-
Issuance of Blood Components tion, and the investigation of any adverse event. Following
Donor units must be labeled appropriately at the time the correct procedures and employing any new technology
of issue. A form or label must be attached to the unit that will ensure the best possible patient safety outcomes.
contains the intended recipient’s two unique identifiers, the
donation identification number or pool number, and an in-
terpretation of compatibility.3 The donor unit, the attached CASE STUDIES
form, and historical records must be compared for accuracy
Case 11-1
and compatibility by transfusion service personnel. All
special transfusion requirements must be met and the unit A bone marrow transplant patient arrives in the emer-
visually inspected for any abnormalities such as hemolysis gency department with a hemoglobin of 5 g/dL and
or the presence of clots. Blood components must not be requires a red blood cell transfusion. The patient’s blood
issued until all discrepancies have been resolved.
6888_Ch11_256-267 31/10/18 9:50 AM Page 265

Chapter 11 Pretransfusion Testing 265

type cannot be determined due to his transplant status. Two units are requested to be held for a C-section surgery.
The antibody screen is negative. The patient’s blood type is O-positive. The antibody screen
is positive and the patient’s plasma contains anti-K. The
1. What is the ABO/Rh of the donor unit selected for
delivered infant’s blood type is O-negative.
transfusion?
2. What type of crossmatch do you perform? 1. What units should be selected for transfusion for the
mother?
Case 11-2 2. If the neonate requires transfusion, what unit should
be selected?
A pregnant woman with ruptured membranes is admitted,
and the ordering provider orders a type and crossmatch.

SUMMARY CHART
 Clerical error and ABO incompatibility are implicated  Emergency transfusions:
in fatal transfusion reactions. • Select uncrossmatched, group O, Rh-negative or
 Pretransfusion specimens, labels, and testing requests Rh-positive red blood cell units based upon patient’s
must contain two unique identifiers. sex and age.
 Writing must be legible and indelible.  Massive transfusions:
 Specimen draw date and time and the collecting indi- • The American College of Surgeons recommends
vidual must be traceable back to the collection of the transfusion of red blood cells, thawed plasma, and
patient sample. platelets in a ratio of 1:1:1.
 Pretransfusion specimens must be collected within  Neonatal transfusions:
3 days of the scheduled transfusion. • Select a group O, Rh-compatible red blood cell unit.
• Perform an antibody screen and any required com-
 Review of records prior to transfusion is essential for
patibility testing with either an infant’s or mother’s
safety.
specimen.
 Selection of donor unit is based on ABO grouping,
 Intrauterine transfusion:
Rh typing, current and historical antibody screening
• Select a group O, Rh-negative red blood cell unit
results, and any special requirements.
that is fresh, leukocyte-reduced, irradiated, negative
 Perform immediate spin, antiglobulin, or computer for sickling hemoglobin, and antigen-negative, if
crossmatch based on current or historical serologic applicable.
results.
 Platelets, thawed plasma, and cryoprecipitate transfu-
sions require a historical or current ABO grouping
only. No compatibility testing required.

3. How many days must a pretransfusion specimen and


Review Questions
donor unit segments be retained post-transfusion?
1. Which is not included on a properly labeled specimen? a.3 days
b.7 days
a. Two unique patient identifiers
c.14 days
b. Date and time of draw
d.1 month
c. Phlebotomist’s initials
d. Patient’s home address 4. If a blood type cannot be resolved, what ABO group
should be selected for a red blood cell transfusion?
2. How many days before a pretransfusion specimen expires?
a.Group A
a. 3 days
b.Group B
b. 7 days
c.Group O
c. 14 days
d.Group AB
d. 1 month
6888_Ch11_256-267 31/10/18 9:50 AM Page 266

266 PART II Blood Groups and Serologic Testing

5. Which antibody specificity is not required in antibody 12. A mother, 30 weeks’ pregnant, has anti-K with a titer of 32.
detection tests? An intrauterine red blood cell transfusion is indicated. The
a. K donor unit selected should be all of the following except:
b. Cw a. O-negative
c. Fya b. K-negative
d. S c. Positive for sickling hemoglobin
d. Irradiated
6. A patient has a history of anti-Jka. The antibody screen is
currently negative. Which red blood cell unit should be 13. A patient with sickle cell disease is B-positive with a pos-
selected, and what type of crossmatch should be per- itive antibody screen. The antibody identified is anti-D,
formed? and the autocontrol is negative. What is a possible
a. Jk(a-) red blood cells, computer crossmatch explanation?
b. Jk(a-) red blood cells, antiglobulin crossmatch a. The patient is weak D-positive
c. Jk(a-) red blood cells, immediate spin crossmatch b. Autoantibody is present
d. ABO-compatible because the antibody screen is c. Patient possesses the partial D phenotype
negative d. The patient has a positive DAT

7. Which is not true of rouleaux formation?


References
a. Mimics agglutination
b. Appears like a “stacking of coins” 1. 21 U.S.C. § 321, U.S. Code. Title 21. Food and Drugs.
2. Fung M, Eder AF, Spitalnik SL, Westhoff CM eds. Technical
c. Can be seen in the antiglobulin test manual. 19th ed. Bethesda (MD): AABB; 2017.
d. Can be dispersed by saline 3. Ooley PW. Standards for blood banks and transfusion services.
31st ed. Bethesda (MD): AABB; 2018.
8. A patient’s blood type is AB-negative, but there are no 4. Poles Debbi, Paula Bolton-Maggs. Annual SHOT Report 2016
AB-negative red blood cell units available. What donor [Internet]. Manchester (UK): Royal College of Pathologists;
units could be selected? 2017. Available from: www.shotku.org.
a. A-negative 5. United States Food and Drug Administration. Fatalities reported
to FDA following blood collection and transfusion annual
b. O-positive summary for FY2015 [Internet]. Washington (DC): U.S. Food
c. B-positive and Drug Administration, 2016. Available from: https://www
d. All of the above .fda.gov/downloads/BiologicsBloodVaccines/SafetyAvailability/
ReportaProblem/TransfusionDonationFatalities/UCM518148.pdf.
9. A patient requires 15 units of thawed plasma for 6. Linden JV, Wagner K, Voytovich AE, Sheehan J. Transfusion
an apheresis procedure. The patient’s blood type is error in New York State: an analysis of 10 years’ experience.
O-negative. What donor units could be selected? Transfusion. 2000;40:1207-13.
7. The Joint Commission. 2017 National patient safety goals.
a. O-negative Oakbrook (IL): The Joint Commission; 2017. Available from
b. AB-positive https://www.jointcommission.org/hap_2017_npsgs/.
c. A-negative 8. ClinLabNavigator.com [Internet]. [updated 2013 Jan 13; cited
d. All of the above 2017 Apr 6]. Kansas: Guidelines for Detecting IV Contamination
of Blood Samples. Available from: http://www.clinlabnavigator
10. The American College of Surgeons recommends trans- .com/guidelines-for-detecting-iv-contamination-of-blood-samples
fusion of red blood cells, thawed plasma, and platelets .html.
in what ratio for a massive transfusion? 9. Ramsey G, Smietana SJ. Long-term follow-up testing of red cell
alloantibodies. Transfusion 1994;34:122.4.
a. 2 units of red blood cells for every unit of platelets 10. Dean L. Chapter 5, The ABO blood group. In: Blood groups
b. 1 unit of red blood cells to 1 unit of thawed plasma and red cell antigens [Internet]. Bethesda (MD): National Cen-
to 1 unit of platelets ter for Biotechnology Information (US); 2005. Available from:
c. 1 unit of red blood cells to 3 units of thawed plasma https://www.ncbi.nlm.nih.gov/books/NBK2267/.
11. Code of Federal Regulations (CFR), Title 21, Food and Drugs.
d. It’s an emergency. Give the surgeon whatever she Part 660, § 33, revised April 1, 2016.
wants 12. Campbell-Lee SA, Kittles RA. Red blood cell alloimmunization
in sickle cell disease: listen to your ancestors. Transfus Med
11. A patient’s antibody screen was positive and an anti-c Hemotherapy. 2014;41:431-35.
was identified. Antiglobulin crossmatches were per- 13. Makroo RN, Arora B, Bhatia A, Chowdhry M, Luka RN. Clini-
formed with c-negative units and 1 of the 6 units was cal significance of antibody specificities to M, N and Lewis
incompatible. What should be performed to resolve the blood group system. Asian J Transfus Sci. 2014;8(2):96-9.
incompatible crossmatch? doi:10.4103/0973-6247.137442.
14. Körmöczi GF, Mayr WR. responder individuality in red blood
a. Give O-negative red blood cells cell alloimmunization. Transfus Med Hemother. 2014;41:
b. Retype the incompatible unit for the c antigen 446-51.
c. Perform a DAT on the incompatible unit 15. Skeate R, Goldman M. Phenotype matching for sickle cell patient:
d. Perform additional identification testing to include a review and recommendation for transfusion practice. Canadian
Blood Services [Internet]. 2014. Available from: https://professional
low-specificity antigens education.blood.ca/en/transfusion/publications/phenotype-
e. b, c, and d matching-sickle-cell-patients-review-and-recommendations.
6888_Ch11_256-267 31/10/18 9:50 AM Page 267

Chapter 11 Pretransfusion Testing 267

16. Markowitz M, Regan D. Association Bulletin #16-02 [Inter- trauma/tqip/massive%20transfusion%20in%20trauma%20


net]. Bethesda (MD): American Association of Blood Banks: guildelines.ashx.
January 15, 2016. Available from: https://www.aabb.org/ 20. Trauma.org. Transfusion for massive blood loss—managing
programs/publications/bulletins/Documents/ab16-02.pdf. massive transfusion events [Internet]. Trauma.org; 2006. Avail-
17. American Association of Blood Banks. Circular of information able from: http://www.trauma.org/index.php/main/article/376/.
for the use of human blood and blood components [Internet]. 21. AABB. Patient blood management. Bethesda (MD): AABB;
Revised October 2017. Available from: http://www.aabb.org/ 2017. Available from: http://www.aabb.org/pbm/Pages/default
tm/coi/Pages/default.aspx. .aspx.
18. Dunbar NM, Yazer MH; Biomedical Excellence for Safer Trans- 22. Spahn DR, Theusinger OM, Hofmann A. Patient blood
fusion (BEST) Collaborative and the STAT Study Investigators. management is a win-win: a wake-up call. Br J Anaesth.
Safety of the use of group A plasma in trauma: the STAT study. 2012;108(6):889-92.
Transfusion. 2017;57:1879-84. doi:10.1111/trf.14139. 23. Hebert P, Wells G, Blajchman M, Marshall J, Martin C,
19. Cryer HG. ACS TQIP massive transfusion in trauma guidelines. Pagliarello G, et al. A multicenter, randomized, controlled clin-
Chicago (IL): American College of Surgeons; 2015. Available ical trial of transfusion requirements in critical Care. New Engl
from: https://www.facs.org/~/media/files/quality%20programs/ J Medi. 340(13);1999:1056.
6888_Ch12_268-280 29/10/18 11:30 AM Page 268

Chapter 12
Blood Bank Testing Technologies
and Automation
Phyllis S. Walker, MS, MT(ASCP)SBB and Denise Harmening, PhD, MT(ASCP)

Introduction Immucor Solid-Phase Red Cell Test Reactions and Procedures


Column Agglutination Technology (CAT) Adherence (SPRCA) Quality Control
Introduction Bio-Rad Solid-Phase Protein A Technology Sensitivity and Specificity
History History Summary Chart
Applications Applications and Principles Review Questions
Principle Advantages and Disadvantages References
Advantages and Disadvantages Automation
Automation Comparison of CAT, SPRCA, and Protein A
Solid-Phase Technology Technologies
Introduction Equipment

OBJECTIVES
1. Explain the principles of CAT, solid-phase red cell adherence (SPRCA), and protein A technologies.
2. Describe the test reactions and how the results are interpreted for each technology.
3. List the advantages and disadvantages of each technology.
4. Describe the automated equipment that is available for each technology.
5. Compare CAT, SPRCA, and protein A technologies in terms of equipment, test reactions, procedures, sensitivity, specificity,
and quality control.

Introduction immunoassay to perform red blood cell (RBC) typing and


antibody screening tests. In 1988, Yves Lapierre,4 working
The earliest reference to “antibodies” came in 1890 when with DiaMed AG in Murten, Switzerland, reported using
Emil von Behring and Shinasabura Kitasato published a column agglutination technology (CAT) to perform RBC
paper describing the use of serum from animals immunized typing and antibody screening tests.
against diphtheria to cure animals infected with the disease. As personnel shortages and cost containment became
In 1901, von Behring was awarded the Nobel Prize for this important issues in the clinical laboratory, automation
work.1 Blood group antibodies were first reported by Land- offered a real solution. Large chemistry, hematology, and
steiner in 1901,2 and he subsequently received the Nobel immunology laboratories were the first to be automated,
Prize for this discovery in 1930. Historically, agglutination but transfusion services were hampered by the complexity
testing was performed on slides, on tiles, and in test tubes. of their procedures and the subjectivity of their test in-
As time passed, test tube techniques became the routine terpretation. Because cost containment forced many labo-
method for performing ABO and Rh typing, direct anti- ratories to rely on less-experienced and/or rotating staff,
globulin testing (DAT), antibody detection, antibody iden- solid-phase and CAT methods were especially appealing.
tification, and compatibility testing. Tube testing remained They offered simplicity, accuracy, reproducibility, and a less
the standard of practice until the 1980s. In 1978, Richard subjective endpoint. With the introduction of commercial
Rosenfield and coworkers3 first reported using a solid-phase reagents for performing these techniques, immunohematology

268
6888_Ch12_268-280 29/10/18 11:30 AM Page 269

Chapter 12 Blood Bank Testing Technologies and Automation 269

laboratories began adopting these alternatives to routine DG 8 Gel contains eight columns. Both companies offer
serologic testing. automated equipment for their products.
Increased health-care management and greater regula-
tory demands provided the incentives for transfusion ser- Applications
vices to seek automation. Automation in blood centers was
first applied to infectious disease tests, which are less com- ID-MTS and DG Gel 8 are currently cleared by the FDA in
plex and have less subjective endpoints. However, with the the United States for ABO forward and reverse grouping,
automation of solid-phase and CAT techniques, transfusion Rh typing, DAT, antibody screen, antibody identification,
services can enjoy the full benefits of walk-away automa- antibody titration, antigen typing, and compatibility testing.
tion. Automated equipment provides the level of quality The ID-MTS card for forward/reverse grouping contains
assurance required by current regulatory standards. Barcod- anti-A, -B, -D, Ctrl, and two buffered columns. Microtubes
ing reduces identification errors by providing accurate with buffered gel are used for ABO reverse grouping. The
patient and reagent identification. Standardized techniques Rh typing card uses microtubes filled with gel containing
reduce the opportunities for human errors and allow the anti-D. The Rh phenotype card contains gels that contain
staff to perform multiple tasks simultaneously. anti-D, anti-C, anti-E, anti-c, anti-e, and a control. Microtubes
filled with gel containing anti-IgG are used for compatibility
Column Agglutination testing, antibody detection, and antibody identification.
Technology (CAT)
Principle
Introduction The gel test, which is performed in a specially designed
Dr. Yves Lapierre of Lyon, France, investigated gelatin, microtube, is a hemagglutination test based on controlled
acrylamide gel, and glass beads as he searched for a way to centrifugation of RBCs through a dextran-acrylamide gel that
trap red blood cell (RBC) agglutinates during standardized contains predispensed reagents. Each microtube is composed
sedimentation and centrifugation. Gel particles provided the of an upper reaction chamber that is wider than the tube
ideal material for trapping the agglutinates, and this discovery itself and a long, narrow portion referred to as the column.
led to a patented process that is the basis of the gel-based In the gel test, a plastic card with microtubes is used instead
tests. In addition, it was discovered that antiglobulin tests of test tubes. A gel card measures approximately 5 × 7 cm
can be performed without multiple saline washes to remove and consists of six or eight columns. Each microtube con-
unbound immunoglobulin. Red blood cells and plasma are tains predispensed gel, diluent, and reagents, if applicable.
allowed to interact in a chamber that has an air bubble barrier Measured volumes of RBCs and serum or plasma are
to keep unbound plasma from neutralizing the antiglobulin dispensed into the reaction chamber of the microtube
reagent that is located in the lower gel column. Because un- (Figs. 12–1, 12–2, 12–3, 12–4). If appropriate, the card is
bound plasma and antiglobulin reagent are prevented from incubated and then centrifuged.
mixing, antiglobulin control cells are not needed to confirm The reaction chamber is where RBCs may be sensitized
the presence of antiglobulin reagent in negative tests. Com- (antigen-antibody binding) during incubation. The column
pared with traditional tube technology, the gel-based tests of each microtube contains dextran-acrylamide gel particles
provide a more stable endpoint and more reproducible suspended in a diluent with reagents added, if applicable.
results—eliminating the variability associated with the phys- The shape and length of the column provides a large surface
ical resuspension of RBC buttons after centrifugation and the
subjectivity in interpretating hemagglutination reactions.

History

The gel test was developed in Europe by Yves Lapierre at


DiaMed AG (Murten, Switzerland). Two companies are cur-
rently licensed by the U.S. Food and Drug Administration
(FDA) to manufacture and distribute gel-based tests in the
United States. In September 1994, Ortho Clinical Diagnostics
(Pompano Beach, Florida) began distributing the ID-MTS
Gel through Micro Typing Systems (MTS), Inc. In March
2002, Ortho Clinical Diagnostics, Inc. acquired MTS, and its
gel-based test has been named the ID-Micro Typing System
(ID-MTS).5 In July 2013, the FDA granted Grifols Diagnos-
tics Solutions, Inc.6 a license to manufacture and distribute
DG Gel 8 cards,6 a form of CAT, in the United States. One
major difference between these two products is the number Figure 12–1. Grifols DG Gel 8 ABO/Rh + Kell card. (Courtesy of Grifols Diag-
of columns. The ID-MTS contains six columns, whereas the nostic Solutions, Inc.)
6888_Ch12_268-280 29/10/18 11:31 AM Page 270

270 PART II Blood Groups and Serologic Testing

Figure 12–4. Grifols DG Spin for centrifuging DG Gel® cards. (Courtesy of


Grifols Diagnostic Solutions, Inc.)

Figure 12–2. Grifols manual station for preparing gel tests. (Courtesy of Grifols
Diagnostic Solutions, Inc.)

Figure 12–5. Ortho Gel Matrix (close-up). (Courtesy of Ortho Clinical Diagnos-
tics, Inc.)
Figure 12–3. Grifols DG Therm for incubating DG Gel® cards. (Courtesy of
Grifols Diagnostic Solutions, Inc.)
remain near the top of the column, with a few agglutinates
staggered below the thicker band. The majority of aggluti-
area for prolonged contact of the RBCs with the gel particles nates are observed in the top half of the column. In a
during centrifugation. 2+ reaction, RBC agglutinates are distributed throughout the
The gel particles make up 75% of the gel-liquid mixture upper and lower halves of the column, with a few aggluti-
that is preloaded by the manufacturer into each microtube. nates at the bottom of the microtube. In a 1+ reaction, RBC
The gel particles are porous, and they serve as a reaction agglutinates are predominantly seen in the lower half of the
medium and filter, sieving the RBC agglutinates according column, with some RBCs at the bottom of the microtube.
to size during centrifugation. Large agglutinates are trapped These reactions may be weak, with only a few agglutinates
at the top of the gel and are not allowed to travel through remaining in the area just above the RBC pellet at the bottom
the gel during the centrifugation of the card. Agglutinated of the microtube. In a negative reaction, the RBCs form a
RBCs remain trapped in the gel, while unagglutinated RBCs well-delineated pellet at the bottom of the microtube. The
travel unimpeded through the length of the microtube, area above the RBC pellet is clear and free of agglutinates. In
forming a pellet at the bottom following centrifugation a mixed-field (mf) reaction, the reaction is characterized by
(Fig. 12–5). a layer of agglutinated RBCs at the top of the column and a
Agglutination reactions in the gel test are graded from 1+ pellet of unagglutinated cells at the bottom of the microtube.
to 4+ and mixed-field, similar to the reactions in the test tube False-positive mixed-field reactions may be caused when
hemagglutination technique. In the CAT test, a 4+ reaction is incompletely clotted serum is used in the CAT test. Fibrin
characterized by a solid band of agglutinated RBCs at the top strands in such serum may trap unagglutinated RBCs, form-
of the column. Usually, no RBCs are visible at the bottom of ing a thin line at the top of the column. Other unagglutinated
the column. In a 3+ reaction, most of the agglutinated RBCs cells pass through the column during centrifugation and
6888_Ch12_268-280 29/10/18 11:31 AM Page 271

Chapter 12 Blood Bank Testing Technologies and Automation 271

travel to the bottom of the microtube. Before interpreting a Standardization is one of the major advantages, inasmuch
reaction as mixed-field, the clinical history of the patient as there is no tube shaking to resuspend the RBC button.
should be considered. For example, recently transfused Tube-shaking technique varies among technologists and may
patients or bone marrow transplant recipients are expected result in variations in the reading, grading, and interpretation
to have mixed populations of RBCs, and their RBCs com- of the test. The CAT technique provides stable, well-defined
monly produce mixed-field reactions. Hemolysis is seen as endpoints of the agglutination reaction that can be observed
clear red liquid in the chamber. or reviewed for up to 3 days. It includes simple standardized
CAT tests may be read manually. Grifols offers the DG procedures, no wash step, and no need for antiglobulin
Reader, an instrument that can interpret gel reactions and control cells. These factors combine to produce more objec-
record the gel test results. The DG Reader identifies the DG tive, consistent, and reproducible interpretation of the test
Gel cards by bar code, digitizes and processes the image with results. Because the CAT technology offers objective, consis-
Diana System software, interprets the results, and records tent results, it is ideally suited to individuals who have been
and stores the data for traceability (Figs. 12–6 and 12–7). cross-trained to work in the blood bank. Other advantages
Ortho offers the ORTHO Workstation, a compact and include the decreased sample volume needed for testing and
light-weight instrument that weighs approximately 24 lbs. the enhanced sensitivity and specificity of the results. Finally,
The ORTHO Workstation integrates ID-MTS Gel cards, a CAT offers improved productivity when compared with
20-card capacity incubator, and a 10-card capacity cen- traditional tube testing, especially when it is combined with
trifuge. All available ID-MTS Gel cards can be used on the automation.
ORTHO Workstation. The major disadvantages of the CAT technology are the
sample restrictions and the need for special equipment.
Advantages and Disadvantages Hemolyzed or grossly icteric blood samples should not be
used because the results may be difficult to interpret visually.
CAT technology, which is applicable to a broad range of blood Grossly lipemic samples containing particulates may clog the
bank tests, offers several advantages over routine tube testing. column, as indicated by diffuse blotches of red blood cells.
Such samples must be clarified by centrifugation or filtration
and retested. Rouleaux caused by serum or plasma with ab-
normally high concentrations of protein (such as in patients
with multiple myeloma or Waldenstrom’s macroglobuline-
mia or from patients who have received plasma expanders
of high molecular weight) may produce hazy reactions or
false-positive results (Fig. 12–8).
Both ID-MTS and DG Gel 8 tests require special incuba-
tors and centrifuges to accommodate the microtube cards,
and a specific pipette must be used to measure and dispense
the 25 µL of plasma or serum and the 50 µL of 0.8% RBCs
into the reaction chambers of the microtubes. Finally, the
calibration of the special pipette must be checked on a
regular schedule.

Automation

Figure 12–6. Grifols DG Reader. (Courtesy of Grifols Diagnostic Solutions, Inc.) In the United States, automated equipment for CAT sero-
logic testing is available from both Ortho Clinical Diag-
nostics, Inc.5 and Grifols Diagnostic Solutions, Inc.6 The
tests that have been automated include ABO grouping,

Figure 12–7. ORTHO Workstation. (Courtesy of Ortho Clinical Diagnostics, Inc.) Figure 12–8. Grifols DG pipette. (Courtesy of Grifols Diagnostic Solutions, Inc.)
6888_Ch12_268-280 29/10/18 11:31 AM Page 272

272 PART II Blood Groups and Serologic Testing

Rh testing, antibody screening, antibody identification,


and compatibility testing.
Grifols offers two automated instruments: the smaller
analyzer, Wadiana, is a compact fully automated instrument
that offers medium throughput, with a maximum capacity
of 48 samples. The Wadiana has bidirectional connectivity
to the laboratory information systems (LIS). All available DG
Gel 8 cards can be tested on the Wadiana, either in batch
mode or individually, as needed. To eliminate waste, partially
used gel cards can be stored for later use (Fig. 12–9).
The larger automated analyzer, Erytra, is a high-throughput
instrument, with a maximum capacity of 96 samples and
400 gel cards. The Erytra has a space-saving vertical design
with a footprint similar to benchtop models. An auto shaker
for red blood cell reagents replaces the need for stir balls.
Similar to the Wadiana, the Erytra has bidirectional connec-
tivity with the LIS, and it can test all available DG Gel
8 cards. Finally, samples can be continuously loaded and
processed as they enter the laboratory (Fig. 12–10).
Ortho Clinical Diagnostics, Inc. offers the ORTHO VISION
Analyzer, a benchtop instrument that automates the full range
of immunohematology testing including serial dilutions for
titration studies and selected cell panels, which helps to
virtually eliminate the need for manual testing, even specialty
testing. The ORTHO VISION Analyzer provides continuous,
random access, as well as batch mode testing. The instrument
has the capacity for 42 patient samples, 120 ID-MTS Gel
Cards, and 33 vials (3 mL) or 18 vials (10 mL) of reagents.
Two centrifuges can spin 10 ID-MTS Gel Cards per centrifuge
(Fig. 12–11).
Figure 12–10. Grifols Erytra®. (Courtesy of Grifols Diagnostic Solutions, Inc.)

Solid-Phase Technology

Introduction
Solid-phase immunoassays have been used for many years
in immunology and chemistry laboratories. In these test sys-
tems (immunoassays), one of the test reactants (either anti-
gen or antibody) is bound to a solid support (usually a
microtiter well) before the test is started. The ability of plas-
tics, such as polystyrene, to absorb proteins from solution
and to bind them irreversibly makes plastic microplate wells

Figure 12–11. ORTHO VISION Analyzer. (Courtesy of Ortho Clinical Diagnostics,


Inc.)

ideal for solid-phase serologic assays. In the United States,


two solid-phase technologies are commercially available for
use in transfusion medicine laboratories. Immucor, Inc. uses
antigens in the form of red blood cell stroma in plastic
microplates to “capture” the antibody. Bio-Rad, Inc. uses
protein A to “capture” IgG-coated red blood cells, that is,
Figure 12–9. Grifols Wadiana®. (Courtesy of Grifols Diagnostic Solutions, Inc.) direct and indirect antiglobulin tests.
6888_Ch12_268-280 29/10/18 11:31 AM Page 273

Chapter 12 Blood Bank Testing Technologies and Automation 273

Immucor Solid-Phase Red Cell Adherence are bound to the immobilized target antigen, that is, the
(SPRCA) sandwich technique. If antibody is attached to the antigen
during the incubation phase, the indicator cells form a
History monolayer of RBCs. If no antibody is present, nothing is at-
In 1986, Beck and coworkers reported using SPRCA to detect tached to the antigen, and the indicator cells form a clearly
RBC antigens and antibodies.7 The first SPRCA commercial delineated button at the center of the microplate well during
assays were manufactured by Immucor, Inc.8 under the trade centrifugation.
name of Capture for the detection of RBC and platelet In Figure 12–12, provided by Immucor, Inc.,8 is an
antibodies. Currently, Capture immunoassays are available outline of the steps in the procedure and a grading chart that
to detect antibodies to RBCs, platelets, and cytomegalovirus compares the endpoints of test tubes and solid phase.
(CMV). Laboratory equipment that is needed to perform SPRCA
testing includes a microplate incubator, a microplate washer,
Applications and a centrifuge that is capable of holding 96-well microplates
SPRCA assays are available in two formats. In one format, or strip wells. Also, an illuminated surface for reading manual
the target antigen (RBCs, platelets, platelet proteins, etc.) microplates is very desirable (Fig. 12–13).
is added by the user to the microplate wells before starting
the test. Capture-R Select, for the identification of RBC anti- Advantages and Disadvantages
bodies, and Capture-P, for the detection of platelet antibod- The major advantage of Immucor SPRCA technology is stan-
ies, are examples of this format. In the other format, the dardization. SPRCA provides stable, well-defined endpoints,
target antigen (RBC stroma, platelet glycoproteins, or which makes cross-training of technologists with rotating
CMV) is bound to the microplate test wells during the man- schedules easy. No predilution of reagents is required, and it
ufacturing process. Capture-R Ready-Screen/Capture-R is possible to test hemolyzed, lipemic, or icteric samples.
Ready-ID and Capture-P Ready-Screen are examples of this LISS in the test process provides enhanced sensitivity for de-
format. tecting weak antibodies. Also, the LISS used in the Immucor
For red blood cell testing, SPRCA assays are currently Capture technology changes color when serum/plasma is
approved by the FDA for antibody screening, antibody iden- added, a safety feature that ensures the patient sample was
tification, weak D testing, IgG autologous control, and com- added to the test system when manual testing is performed.
patibility testing. Ready-Screen assays use microwells that Finally, because the reagent red blood cells in SPRCA are
are preloaded with reagent cell stroma: antibody screening dried and precoated onto the surface of the wells, the test
cells; either a set of two cells (I and II), a set of three cells, a plates have a long shelf life.
set of four cells, or a pool of two cells. The two-, three-, and Similar to the other automated technologies, the major
four-cell sets may be used for antibody detection in transfu- disadvantage of SPRCA is the need for specialized equip-
sion recipients. Pooled cells are used for donor antibody ment, that is, a microplate centrifuge, a 37°C incubator for
detection when increased sensitivity is undesirable. Reagent microplates, and a light source for reading the final results.
panels of preloaded cells are also available for RBC antibody While a manual station is available for solid-phase testing,
identification. When it is desirable to test selected cells, in- users report that the user’s technique affects the outcome of
tact reagent RBCs can be added by the user to chemically the testing, especially the manual handling of the test strips.
modified microwells. In addition, the increased sensitivity may also be a disadvan-
tage inasmuch as SPRCA may detect weak autoantibodies
Principle that other systems miss.
Immucor SPRCA assays may be performed with either plasma
or serum, but plasma is preferable. When clotted samples are Automation
incompletely clotted, the serum is difficult to remove during Full automation of the Capture solid-phase technology is
the wash cycle; allowing residual unbound serum to clot and available from Immucor, Inc.,8 using two FDA-approved,
make the endpoint of the test unreadable. For best results, walk-away instruments. The smaller instrument, Echo, can
the manufacturer recommends adding a pH-stabilizing buffer be continuously loaded and unloaded. It holds 20 samples,
to the isotonic saline for washing the microplates. (A suitable 16 reagents, and 32 microstrips. All Capture assays can be
buffer is available from the manufacturer.) performed on Echo.8 Echo provides multiple quality control
To perform Immucor SPRCA testing, patient serum or features, including barcode readers to ensure proper identi-
plasma and low-ionic-strength saline (LISS) are added to fication of samples and reagents, liquid level sensors, clot
microwells that are coated with the target antigen. The detectors, controlled temperature settings, and incubation
microplates are incubated at 37°C, allowing time for possible timers (Fig. 12–14).
antibodies to attach to the antigens in the well. After incu- The larger instrument, NEO, is also fully automated.8
bation, the wells are washed with pH-buffered isotonic saline Both Echo and NEO use hemagglutination to perform blood
to remove unbound serum proteins. Next, anti-IgG–coated typing and Capture assays to perfom antibody screening,
indicator RBCs are added, and the microwells are cen- antibody identification, weak D testing, IgG autologous
trifuged. Centrifugation forces the indicator RBCs into close control, and compatibility testing. NEO has a capacity of
contact with IgG antibodies from the test serum/plasma that 15 microplates and 224 samples, with continuous access
6888_Ch12_268-280 29/10/18 11:31 AM Page 274

274 PART II Blood Groups and Serologic Testing

Figure 12–12. Immucor Capture® Tech-


nology Testing Procedure and Grading Chart.
(Courtesy of Immucor, Inc.)

Figure 12–14. Immucor Echo® instrument. (Courtesy of Immucor, Inc.)

Figure 12–13. Manual Capture® workstation. (Courtesy of Immucor, Inc.)


Bio-Rad Solid-Phase Protein A
Technology
for loading and unloading. The NEO has an automated
microplate washer, an incubator with six-ambient and eight History
37°C bays, a centrifuge module, and an image analysis reader
with two charged-coupled device cameras for interpreting In 1992, Biotest AG developed a test in Europe for detecting
test results (Fig. 12–15). IgG antibodies using microplate wells that are coated with
6888_Ch12_268-280 29/10/18 11:31 AM Page 275

Chapter 12 Blood Bank Testing Technologies and Automation 275

Figure 12–15. Immucor NEO® instrument. (Courtesy of Immucor, Inc.)

protein A. Protein A is a component of the cell wall of Staphy-


lococcus aureus that has a very high affinity for the Fc portion
of most immunoglobulin classes.9 In 2005, the U.S. FDA ap-
proved the distribution of the Solidscreen II assay in the United
States, and in 2010, Bio-Rad Laboratories, Inc.10 acquired this
test from Biotest AG. Erytype S and Solidscreen II are reagents Figure 12–16. Bio-Rad Erytype® S Blood Group System. (Courtesy of Bio-Rad
that are currently available from Bio-Rad for use in their auto- Laboratories, Inc.)
mated instrutment, TANGO infinity.10

Applications and Principles If antibodies are attached to the red blood cells, the coated
cells are captured by protein A on the surface of the microwell
The Erytype S10 blood group system is a hemagglutination to form a homogenous layer on the bottom of the micowell
microplate method that uses microplate wells precoated with (positive reaction). If no antibodies are attached to the RBCs,
blood-grouping monoclonal antibodies. Agglutination reac- a compact red blood cell button is formed in the center of the
tion results, and the agglutination patterns are captured and well (negative reaction). The reaction patterns are recorded
interpreted by the TANGO infinity10 automation platform. as a digital image and interpreted by the TANGO infinity,10
Assays include forward and reverse ABO group, Rh type, and using decision logic programmed into the instrument. Posi-
K (Kell) phenotype (Fig. 12–16). tive and negative controls are performed each day before test-
The Solidscreen II assay10 provides a standardized method ing, whenever reagents are changed, or after service/repair of
for antiglobulin testing. The assay combines traditional the TANGO infinity10 (Fig. 12–17).
low-ionic-strength test methods with a unique solid-phase
format. Unlike other solid-phase methods that use red blood Advantages and Disadvantages
cell stroma, the Solidscreen II assays use a microwell coated
with protein A, which has a high affinity for the Fc portion The Solidscreen II assay10 provides a standardized method
of IgG. The system uses the same liquid reagent red blood for performing antiglobulin testing using a unique solid-
cells and low-ionic-strength solution used in tube methods phase capture method. In 2004, Heintz et al. showed that
for antibody screening and identification. To perform an the detection of wanted antibodies by TANGO optimo 10 (the
indirect test, 50 µL of patient serum/plasma, typing serum, predecessor of the TANGO infinity instrument10) was similar
or control reagents are combined in a microwell with a to Capture-R Ready-Screen.11 Because the assay uses micro-
1% suspension of reagent red blood cells (antibody screen/ wells that are coated with protein A, the microplates have a
identification), patient or donor cells (antigen typing or au- long product and on-board shelf life. The assay uses intact
tocontrol), or donor red blood cells (crossmatch), depending red blood cells as opposed to red blood cell fragments, which
on which test is being performed. A low-ionic-strength are used in other solid-phase methods.
solution (MLB 2) is added to the microwells, and the con- Solidscreen II10 requires specialized equipment, the
tents of the microwell are mixed and allowed to incubate TANGO infinity.10 In the United States, the TANGO infinity10
at 37°C. For both direct and indirect tests, the mixture is is not cleared by the FDA to test enzyme-treated or frozen/
then centrifuged, the supernatant is aspirated, and the deglycerolized cells, although such cells can be used outside
microwell is washed with phosphate buffered saline (PBS). the United States. Finally, the Solidscreen II assay10 is only
For antiglobulin tests,100 µL of anti-IgG anti-human globulin FDA licensed for use with anti-IgG reagent, and it does not
(AHG) is added; the microwell is remixed and centrifuged. detect complement-coated cells as a positive result. Outside
6888_Ch12_268-280 29/10/18 11:31 AM Page 276

276 PART II Blood Groups and Serologic Testing

Figure 12–18. Bio-Rad TANGO infinity® instrument. (Courtesy of Bio-Rad


Laboratories, Inc.)

Equipment

CAT requires a special incubator and a special centrifuge


to spin the cards. The solid-phase technology requires a
Figure 12–17. Bio-Rad Solidscreen® II Methodology. (Courtesy of Bio-Rad Lab-
special centrifuge to spin microplates, a 37°C incubator for
oratories, Inc.) microplates, and a light source for reading the final results is
desirable as well. Automated and semiautomated equipment,
approved by the FDA, is currently available for both CAT and
the United States, both polyspecific AHG and anti-IgG are SPRCA technologies. Automated equipment, approved by the
available for use on the TANGO infinity.10 While many of the FDA, is currently available for the protein A technology.
TANGO reagents used for antibody detection and identifica- All of the technologies improve safety and decrease haz-
tion can be used off-line in traditional tube tests, there is no ardous waste, inasmuch as the use of plastic eliminates the
manual Solidscreen II10 method to use as a backup in the danger associated with broken glass and miniaturized reac-
United States. Outside the United States, a manual worksta- tion chambers reduce the quantity of hazardous waste.
tion is available for use with the Solidscreen II10 method.
Test Reactions and Procedures
Automation
All technologies provide stable, reproducible endpoints. CAT
Solidscreen II10assays are performed in an automated system, uses a special pipette to precisely measure the quantity of test
the TANGO infinity.10 The instrument performs blood group- cells and sera, whereas manual SPRCA technology does not
ing, antibody screening and identification, autocontrol, direct require a special pipette. SPRCA uses conventional drops of
antiglobulin testing, and weak D and cord blood testing. The cells and sera. By standardizing the reactants in the assay and
instrument verifies reagent and sample identity and dispenses eliminating variation in the tube-shaking technique, all of
precise volumes of reagents, providing confidence in the accu- these technologies eliminate the subjectivity associated with
racy of the test results. Minimal preparation is required to op- interpreting the endpoint of test tube agglutination tests.
erate the instrument because it has on-board optimal storage CAT is read as 4+, 3+, 2+, 1+, mixed-field (mf), or negative,
conditions with a capacity of 27 reagents and 120 microplate and it is based on agglutination reactions. The ease of quanti-
strips. The TANGO infinity10 system provides random access tating reactions with CAT is an advantage when evaluating an
with a capacity to load up to 120 samples (Fig. 12–18). antibody identification for multiple antibodies or dosage
effects. Mixed-field agglutination produces a characteristic pat-
Comparison of CAT, SPRCA, tern in CAT. In this technology, the agglutinated population of
and Protein A Technologies RBCs is trapped in the column, and the unagglutinated popu-
lation is pelleted at the bottom of the microtube following
In 2008, Butch published an article in Immunohematology that centrifugation. The ability to recognize mixed-field reactions
compares the automated methods that were available in the is particularly valuable in evaluating a possible transfusion re-
transfusion service at that time.12 This section compares the action or the survival of a minor population of transfused cells.
latest developments in CAT, SPRCA, and protein A technolo- The solid-phase assays are read as 4+, 3+, 2+, 1+; however,
gies in terms of equipment, procedures, test reactions, sensi- mixed-field reactions may give an NTD (No Test Detected) re-
tivity, specificity, quality control, and automation. Table 12–1 sult. All of the automated instruments, Erytra, ORTHO VISION
compares the procedural steps of the CAT and SPRCA manual Analyzer, Echo, NEO, and TANGO infinity, interpret the results
technologies, and Table 12–2 compares the features of the as positive or negative and grade the reaction strength (0 to 4+).
three technologies for automated testing. The endpoints of all of the technologies are stable, and they
6888_Ch12_268-280 29/10/18 11:31 AM Page 277

Chapter 12 Blood Bank Testing Technologies and Automation 277

Table 12–1 Procedural Steps of CAT and SPRCA Manual Techniques


CAT Solid-Phase Test
Capture-R Ready-Screen
Procedural Steps ID-MTS DG Gel 8 Capture-R Ready-ID

1. Add RBCs to wells or tubes Yes N/A

2. Centrifuge plate to form monolayer N/A N/A

3. Wash away unbound RBCs N/A N/A

4. Add test serum/plasma Yes Yes

5. Incubation 15 min 15 min


6. Wash cycle N/A Yes

7. Add indicator cells N/A Yes

8. Centrifuge 10 min 2 min

ID-MTS Gel test (Ortho Clinical Diagnostics, Inc.); DG Gel 8 Cards Gel test (Grifols Diagnostic Solutions, Inc.); Capture solid-phase (Immucor, Inc.)
RBC = red blood cells; min = minutes; N/A = not applicable
Table 12–1 is used by permission from F.A. Davis, Inc.

Table 12–2 Comparison of Automated Technologies


CAT SPRCA Protein A
Tests and Features ORTHO VISION Wadiana, Erytra Immucor Echo, Immucor NEO TANGO infinity

ABO forward Yes Yes Yes

ABO reverse Yes Yes Yes

Rh Yes Yes Yes

Antibody screen Yes Yes Yes

Antibody identification Yes Yes Yes

Crossmatch Yes Yes Yes

Direct antiglobulin test ID-MTS detects IgG, C3d No C3d detection No C3d detection

Autocontrol (IgG) Yes Yes Yes

Washing required No Yes Yes

pH of wash critical N/A Yes Yes

Centrifugation critical Yes Yes Yes

Detects IgG, IgM IgG IgG, IgM

Media LISS LISS LISS

Incubation time 15 minutes 15 minutes 20 minutes

Minimum tests 6 or 8 columns per card ID = 1 test per strip 8 tests per strip
3-cell screen = 2 tests per strip

Sensitivity (antibodies detected) ↑ LISS (Tube) ↑ LISS (tube) ↑ LISS (tube)


CAT ↓ SPRCA, protein A SPRCA, protein A ↑ CAT Protein A, SPRCA ↑ CAT

Specificity
Clinically significant antibodies Similar to SPRCA, protein A Similar to CAT, protein A Similar to CAT, SPRCA
Insignificant/nonspecific antibodies CAT ↓ SPRCA, protein A CAT ↓ SPRCA, protein A CAT ↓ protein A, SPRCA
Continued
6888_Ch12_268-280 29/10/18 11:31 AM Page 278

278 PART II Blood Groups and Serologic Testing

Table 12–2 Comparison of Automated Technologies—cont’d


CAT SPRCA Protein A
Tests and Features ORTHO VISION Wadiana, Erytra Immucor Echo, Immucor NEO TANGO infinity

Stability Yes (2–3 days) Yes (2 days) Yes


Standardized reproducible results Yes Yes Yes
Quantitation of reactions Neg, 1+ to 4+ Neg, 1+ to 4+ Neg, 1+ to 4+
Mixed-field No mixed-field No mixed-field
Quality control Daily QC Daily QC Daily QC
LISS color change
+s, +w, neg controls

Safety increased over tubes? Yes Yes Yes

Hazardous waste decreased? Yes Yes Yes

Automation (FDA approved) Yes Yes Yes


ORTHO VISIONTM Analyzer IMMUCOR Echo® BIO-RAD
TANGO optimo®
GRIFOLS Wadiana® IMMUCOR NEO® BIO-RAD
GRIFOLS Erytra® TANGO infinity®

N/A = not applicable; IgG = immunoglobulin G; IgM = immunoglobulin M; ↑ = increased; ↓ = decreased; LISS = low-ionic-strength saline; SPRCA = solid-phase red blood cell
adherence; DTT = dithiothreitol; +s = strong positive control; +w = weak positive control; neg = negative control

can be read 2 to 3 days after the test is performed. Such stability the LISS used in the manual SPRCA tests is formulated to de-
is a distinct advantage when less-experienced technologists are tect the addition of serum/plasma by a color change from pur-
cross-trained to work in the transfusion service. All of the au- ple to blue when plasma is added. This feature protects the user
tomated instruments capture a digital image in high resolution from failing to add plasma to a well during manual testing.
of the endpoint for review and storage by the user. Whenever
the interpretation of an assay is uncertain, the test results can Sensitivity and Specificity
be retained until a supervisor can review them.
The procedures in all of the technologies are performed Before CAT and solid-phase assays were licensed by the FDA
in subsequent steps: pipetting, incubating, spinning, and for use in the United States, studies were performed to show
reading. In CAT, these steps are performed automatically that the new technologies were as effective in detecting
by the ORTHO VISION analyzer,5 as well as the Grifols antibodies as traditional test tube methods.
Wadiana, and Erytra analyzers.6 The washing cycle step has Ideally, a technique should detect antibodies that are con-
been eliminated with CAT. In the SPRCA technology, these sidered potentially clinically significant and avoid detecting
steps are performed automatically by the Immucor Echo and antibodies that are considered clinically insignificant, such as
NEO analyzers,8 and in the protein A assay, the steps are per- benign autoantibodies and cold agglutinins. Studies have
formed automatically by the TANGO infinity analyzer. The shown that although these technologies are all LISS-based,
endpoint of all of the assays detects IgG, and it is possible to they are all more sensitive than LISS test tube technique.13,14
perform an autocontrol to detect IgG-coated cells; however, Also, LISS-dependent reactivity (artifact) in some patients can
an autocontrol cannot be substituted for a DAT unless the interfere with all of these technologies. Differences in sensi-
test detects both IgG- and C3d complement–coated cells. tivity have been reported when CAT is compared with the Im-
mucor SPRCA. Ramsey et al.15 reported that Immucor SPRCA
Quality Control was more sensitive than CAT for detecting RBC alloantibod-
ies, particularly for detecting antenatal RhIg in Rh-negative
In addition to routine quality control (QC) for the 37°C incu- mothers. However, they also reported that Immucor SPRCA
bator, centrifuge, pipette tips, and pipette dispenser used in had a higher rate of nonspecific reactions. These conclusions
testing, these technologies have other QC features. The CAT were confirmed by Garozzo et al.16 and Yamada et al.17
test uses special dispensers to prepare the RBC suspensions Weisbach et al. reported that Immucor SPRCA had a high rate
and special pipettes to add a measured volume of plasma/ of nonspecific reactions than the Bio-Rad Solidscreen II.18
serum and RBCs. Each lot number of cards and diluent should Because these assays are designed to detect IgG, they
be tested on the day of use to confirm that the test cards and avoid detecting clinically insignificant IgM antibodies such
the diluted reagent RBCs are reacting as expected. In addition, as cold agglutinins. However, they may fail to detect
6888_Ch12_268-280 29/10/18 11:31 AM Page 279

Chapter 12 Blood Bank Testing Technologies and Automation 279

clinically significant IgM antibodies that are forming during a 2016, Kay et al.21 reported that SPRCA detected anti-Jka that
primary immune response. CAT detects IgM antibodies when was missed by the gel technique. Barker et al.22 reported a
incompatible cells form a lattice in the reaction chamber and delayed transfusion reaction caused by anti-Fya that was not
are trapped at the top of the column during centrifugation. In- detected in the pretransfusion sample by LISS, PEG, or gel
creased sensitivity is an advantage when a weak, clinically tests. The post-transfusion sample demonstrated anti-Fya using
significant antibody is present, but it is a disadvantage when only PEG-AHG technique; LISS-AHG and gel were negative.
weak autoantibodies are present. In a review article that compared antibody screening
Finally, the question of specificity is complex. All of the methods, Casina23 sums up these experiences by concluding
technologies demonstrated the ability to detect the majority of that there is no one method that will detect all clinically
wanted antibodies before they were licensed by the FDA; how- important antibodies.
ever, there are many isolated reports of method-dependent Automated methods are being adopted throughout the
antibodies that are only demonstable by one technique. Combs world, in the United States as well as Europe and Asia. For
and Bredehoeft19 reported missing 11 antibodies in nine more information, see an article by Delaflor-Weiss and
patients (three anti-K, two anti-c, two anti-E and -Fya, one Chizhavsky24 describing a 3-year experience in a community
anti-D, and one anti-Jka) using the gel technique. Callahan hospital in the United States, and an article by Bajpai et al.25
et al.20 reported a severe transfusion reaction caused by describing automation in immunohematology in New Delhi,
anti-Jkb that was only demonstrable by SPRCA. Similarly, in India.

SUMMARY CHART
 Advantages of CAT and solid-phase technologies over • In SPRCA tests, the target antigen is affixed to the
routine tube testing are as follows: bottom of the microplate wells.
• Standardization, which means there is no tube • If the test plasma contains antibodies to the antigen,
shaking or resuspension of an RBC button to cause they attach to the fixed antigen, and indicator cells
subjectivity in the interpretation of the test. detect the attached antibodies by forming a mono-
• Stability, which means there are well-defined end- layer of RBCs.
points of the reaction. • If the test plasma contains no antibodies to the anti-
• Decreased sample volume is needed for testing. gen, there is no attachment to the fixed antigen, and
• Enhanced sensitivity and specificity are present. the indicator cells form a clearly delineated button
 CAT test points to remember: at the center of the microplate well.
• The principle of the CAT test is hemagglutination. • Solid-phase reactions are stable for observation or
• In the CAT test, RBCs and serum or plasma are al- review for 2 days.
lowed to incubate together in a reaction chamber. • Solid-phase technology is currently approved for an-
• Following incubation, controlled centrifugation tibody screening, antibody identification, and com-
drives the RBCs through a specially designed micro- patibility testing.
tube filled with beads of dextran-acrylamide gel. • Advantages of solid-phase technology include the
• Agglutinated cells remain at the top of the tube or ease of use, because no predilution of reagents is
are trapped in the gel, depending on the size of the required, and the ability to test hemolyzed, lipemic,
agglutinates. or icteric samples.
• Unagglutinated cells move through the gel to the • Enhanced sensitivity increases the detection of weak
bottom of the tube. alloantibodies.
• The gel test reactions are stable for observation or • The major disadvantage of solid-phase technology
review for 2 to 3 days. is the need to purchase special equipment: a cen-
• CAT technology is currently approved for ABO for- trifuge that can spin microplates, a 37°C incubator
ward and reverse grouping, Rh typing, DAT, antibody for microplates, and a light source for reading the
screening, antibody identification, and compatibility final results.
testing.  Solid-phase protein A technology points to remember:
• The major disadvantage of the CAT technology is • IgG antibodies are captured in microwells that are
the need to purchase special equipment: a centrifuge coated with protein A.
to accommodate the microtube cards used for test- • Solidscreen II, which uses solid-phase protein A
ing and a pipette for dispensing plasma or serum technology, is an assay that uses traditional antiglob-
and RBC suspensions into the reaction chambers of ulin technique.
the microtubes. • Solid-phase protein A testing is available in the
 SPRCA assay points to remember: United States only as an automated technology on
• The principle of SPRCA is based on solid-phase the TANGO infinity instrument.
technology.
6888_Ch12_268-280 29/10/18 11:31 AM Page 280

280 PART II Blood Groups and Serologic Testing

Proceedings of the Plenary Session, 25th Congress, Interna-


Review Questions tional Society Blood Transfusion. Paris; 1978.
4. Lapierre Y, Rigal D, Adam J, Josef D, Meyer F, Greber S, et al.
1. The endpoint of the CAT test is detected by: The gel test: a new way to detect red cell antigen-antibody
reactions. Transfusion. 1990;30(2):109-13.
a. Agglutination. 5. Ortho Clinical Diagnostics, 1001 Route 202, Raritan, NJ 08869,
b. Hemolysis. Telephone: 800-421-3311, website: www.orthoclinical.com
c. Precipitation. 6. GRIFOLS USA, LLC, 2410 Lillyvale Avenue, Los Angeles, CA
d. Attachment of indicator cells. 90032-3514, Telephone: 888-474-3657, website: www.grifolsusa
.com
2. The endpoint of the SPRCA test is detected by: 7. Beck ML, Plapp FV, Sinor LT, Rachel JM. Crit Rev Clin Lab Sci.
a. Agglutination. 1986; 22(4): 317-42.
8. Immucor, Inc. 3130 Gateway Drive, Norcross, GA 30091-5625,
b. Hemolysis. Telephone: 855-466-8267, website: www.immucor.com
c. Precipitation. 9. King BF, Wilkinson BJ. Binding of human immunoglobulin G
d. Attachment of indicator cells. to protein A in encapsulated Staphylococcus aureus. Infect
Immun. 1981;33:666-72.
3. The endpoint of the solid-phase protein A assay is: 10. Bio-Rad Laboratories, Inc. 5731 W. Las Positas Blvd, Pleasanton,
a. Agglutination. CA 94588, Telephone: 925-474-9074, website: www.biorad.com
b. Hemolysis. 11. Heintz M, Bahl M, Mann J, Yohannes M, Levitt J. Evaluation
of blood group antisera and antibody screening with the
c. Precipitation. TANGOTM automated blood bank analyzer. Transfusion. 2004;
d. Attachment of cells to microwell. 44:125A.
12. Butch SH. Automation in the transfusion service. Immunohe-
4. Protein A captures antibodies by binding to the:
matology. 2008;24:86-92.
a. Fab portion of immunoglobulin. 13. Novaretti MCZ, Jens Silveira E, Filho EC, Dorlhiac-Llacer PE,
b. Fc portion of immunoglobulin. Chamone DAF. Comparison of tube and gel techniques for
c. Surface of test cells. antibody identification. Immunohematology. 2000;16:138-41.
14. Weisbach V, Kohnhäuser T, Zimmermann R, Ringwald J,
d. Surface of indicator cells. Strasser E, Zingsem J, Eckstein R. Comparison of the perfor-
5. Mixed-field reactions can be observed in: mance of microtube column systems and solid-phase systems
and the tube low-ionic-strength solution additive indirect
a. Gel. antiglobulin test in the detection of red cell alloantibodies.
b. SPRCA. Trans Med. 2006;16:276-84.
c. Protein A technology. 15. Ramsey G, Sumugod RD, Garland FD, Lindholm PF, Masarik
d. None of the automated technologies. SR. Automated solid-phase RBC antibody screening in a trans-
fusion service. Transfusion. 2005;45:123A.
6. An advantage for both CAT and solid-phase technology is: 16. Garozzo G, Licitra V, Criscione R, Comitini N, Noto C,
Lomagno R, Ruta D, Spadola G, Zago V, Bonomo P. A comparison
a. No cell washing steps. of two automated methods for the detection and identification of
b. Standardization. red blood cell alloantibodies. Blood Transfus. 2007;5:33-40.
c. Use of IgG-coated control cells. 17. Yamada C, Serrano-Rahman L, Vasovic LV, Mohandas K,
d. Specialized equipment. Uehinger J. Antibody identification using both automated
solid-phase red cell adherence assay and a tube polyethylene
7. A disadvantage for both CAT and solid-phase technol- glycol antiglobulin method. Transfusion. 2008;48:1693-98.
ogy is: 18. Weisbach V, Teufel M, Bredehoeft A, Strobel J, Eckstein R.
DGTI, Transfus Med Hemother. 2008:35(Suppl 1):67.
a.Decreased sensitivity. 19. Combs MR, Bredehoeft SJ. Selecting an acceptable and safe anti-
b.Inability to test hemolyzed, lipemic, or icteric samples. body detection test can present a dilemma. Immunohematol.
c.Inability to detect C3d complement–coated cells. 2001;17:86-89.
d.Large sample requirement. 20. Callahan DL, Kennedy MS, Ranalli MA, Waheed A, Nicol KK.
Delayed hemolytic transfusion reaction caused by Jkb antibody
8. A safety feature in the SPRCA test is: detected by only solid phase technique. Transfusion. 2000;
a. Air bubble barrier. 40:113S.
21. Kay B, Poisson JL, Tuma CW, Shulman IA. Anti-Jka that are
b. Viscous barrier. detected by solid-phase red blood cell adherence but missed
c. Color change of the LISS. by gel testing can cause hemolytic transfusion reactions. Trans-
d. Use of IgG-coated control cells. fusion. 2016;56:2973-79.
22. Barker JM, Scillian J, Spindler BJ, Cruz MC. A delayed transfu-
References sion reaction due to anti-Fya not detected by LISS-tube or gel
techniques. Transfusion. 2004;44:119A.
1. Behring E, Kitasato S. Ueber das Zustandekommen der Diphtherie- 23. Casina TS. In search of the holy grail: comparison of antibody
Immunitat und der Tetanus-Immunitat bei thieren. Deutsche screening methods. Immunohematology. 2006;22:196-202.
medizinsche Wochenschrift. 1890;16:1113-14. 24. Delaflor-Weiss E, Chizhevsky V. Implement of gel testing for
2. Landsteiner K. Ueber Agglutinationserscheinunger normalen antibody screening and identification in a community hospital,
menchlichen Blutes. Wiener Wochenschrift. 1901;46:1132-34. a 3-year experience. Lab Med. 2005;36:489-92.
3. Rosenfield RE, Kochwa SE, Kaczera Z. Solid phase serology 25. Bajpai M, Kaur R, Gupta E. Automation in immunohematology.
for the study of human erythrocyte antigen-antibody reactions. Asian J Transfus Sci. 2012;6:140-44.
Transfusion Practices

Chapter 13
Donor Selection
Virginia Hughes, PhD, MT(ASCP)SBB

Introduction Whole Blood Collection


Governing Agencies Donor Identification HCV
U.S. Food and Drug Administration Aseptic Technique HIV
AABB Collection Procedure HTLV-1/11
College of American Pathologists Postdonation Inst uctions WNV
Donor Selection Donor Reactions Syphilis
Registration Mild T. cruzi (Chagas Disease)
Medical History Questionnaire Moderate Zika and Ebola
Medical History Questions Severe Case Studies
The Physical Examination Hematomas Case Study 13-1
Informed Consent Donor Rec rds Case Study 13-2
Autologous Donation Donor rocessing Summary Chart
Directed Donation ABO/Rn Review Questions
Apheresis Donation References

OBJECTIVES
1. Identify the organizations that regulate or accredit the immunohematology laboratory.
2. State the acceptable levels for the following tests in allogeneic and autologous donation:
• Weight
• Temperature
• Hemoglobin
• Hematocrit
3. Differentiate between acceptable donation, and temporary, indefinite, and permanent deferral given various medical conditions.
4. Differentiate among the four different types of autologous donations.
5. Describe the procedure for a whole blood donation, including arm preparation, blood collection, and postphlebotomy care
instructions for the donor.
6. Differentiate among mild, moderate, and severe reactions, and state recommended treatments for each.
7. List the tests required for allogeneic, autologous, and apheresis donation.
Continued

281
6888_Ch13_281-306 29/10/18 5:43 PM Page 282

282 PART III Transfusion Practices

OBJECTIVES—cont’d
8. State the acceptable interval of donation for allogeneic, autologous, and apheresis donors.
9. Discuss record-keeping procedures for allogeneic, autologous, and apheresis donors.
10. Compare and contrast donor reentry protocols for donors who previously tested positive for human immunodeficiency virus
(HIV), hepatitis C virus (HCV), hepatitis B virus (HBV).
11. Describe a hematoma and how it is treated.
12. Discuss how a human is infected with West Nile virus (WNV) and how donors are tested.
13. Compare and contrast Creutzfeldt-Jakob disease (CJD) and variant Creutzfeldt-Jakob disease (vCJD) regarding age of onset
and immunohistochemistry.
14. Define prion.
15. Identify endemic countries for malaria and donor regulations affecting U.S. military personnel.
16. State deferral criteria for Babesia, Zika, and Ebola.
17. List donor screening and confirmation tests for human T-cell lymphotrophic virus type I/II (HTLV-I/II).
18. State a contraindication for autologous donation.
19. State serologic tests required for directed donations.

Introduction regulating the collection of blood and blood components


used for transfusion and for the manufacture of pharma-
The selection of potential blood donors has become more ceuticals derived from blood and blood components. CBER
rigorous in the last 5 years with cogent additions to the develops and enforces quality standards, inspects blood
medical history questionnaire and serologic testing. The establishments, and monitors reports of errors, accidents,
transfusion of blood to a patient in need is safer than it has and adverse clinical events. In the early 1990s, the FDA
ever been. However, safety and therapeutic benefit are began to treat blood establishments the same as any drug
dependent on guidelines established by the U.S. Food and manufacturer, requiring strict compliance with all aspects
Drug Administration (FDA) and AABB and are carried out of transfusion medicine, including donor selection and
by the qualified and trained personnel in blood collection screening. In addition, the FDA establishes and maintains
centers. This chapter will explore the donor history and the regulations and inspects blood collection and process-
physical examination qualifications regarding allogeneic, ing centers. The FDA is also responsible for licensing the
autologous, and apheresis donors in addition to the testing collection and processing facilities, the blood products and
requirements for each of those donations. derivatives, and the reagents used in the processing and
testing of those products (see Chapter 27 “Transfusion
Governing Agencies Safety and Federal Regulatory Requirements”). The FDA
provides recommendations and requirements regarding in-
Guidelines and regulations for donor selection and serologic fectious diseases as well as potential emerging diseases.
testing are written by the FDA and AABB. While the purview Blood establishments may adapt stricter requirements than
of these two entities may overlap in some areas of donor those published by the FDA as written in the blood center’s
collection and donor testing, their institutional goals and standard operating procedure (SOP).
national donation guidelines provide an effective framework
of processes utilized by all blood donor centers across the AABB
United States. The College of American Pathologists (CAP)
conducts inspections of all clinical laboratory departments, The AABB, formerly known as the American Association of
including transfusion services, awarding accreditation to Blood Banks, was established in 1947. It is an international
facilities that are deemed to be compliant with established association of blood centers, transfusion and transplantation
standards. services, and individuals involved in transfusion medicine.
It provides a voluntary inspection and accreditation program
U.S. Food and Drug Administration for its member institutions that meet the requirements of the
Centers for Medicare and Medicaid Services (CMS) and the
The U.S. FDA is a regulating agency. Its regulations for Clinical Laboratory Improvement Amendments of 1988
donor screening are outlined in the Code of Federal Regu- (CLIA). The mission of the AABB is to establish and provide
lations (CFR), Title 21, parts 211, 600-799. Under the the highest standard of care for patients and donors in all
auspices of the FDA, blood is regarded both as a biologic aspects of transfusion medicine. The AABB has published
and a drug. In 1988, the Center for Biologics Evaluation books on transfusion medicine throughout its existence; two
and Research (CBER) was formed. CBER is responsible for resources that are vital to donor screening procedures are the
6888_Ch13_281-306 29/10/18 5:43 PM Page 283

Chapter 13 Donor Selection 283

AABB Standards for Blood Banks and Transfusion Services and


the AABB Technical Manual. The specific guidelines for BOX 13–1
donor screening are discussed in these publications and will Minimum Age to Donate Allogeneic Whole Blood
be referred to throughout this chapter.
Alabama 16 Nebraska 16
Alaska 16 Nevada 16
College of American Pathologists Arizona 16 New Hampshire 16
Arkansas 16 New Jersey 16
The CAP also provides a voluntary inspection and accredi-
California 16 New Mexico 17
tation program for its member institutions. Since most trans-
Colorado 16 New York 16
fusion services are part of the clinical laboratory departments
Connecticut 17 North Carolina 16
in a hospital, those services are included in a CAP inspection.
Delaware 17 North Dakota 16
As with the AABB inspections, CAP inspections are approved
District of Columbia 17 Ohio 16
by CMS and meet the CLIA requirements.
Florida 17 Oklahoma 16
Georgia 16 Oregon 16
Donor Selection
Hawaii 16 Pennsylvania 16
Donor selection encompasses the medical history require- Idaho 16 Puerto Rico 16
ments for the donor, the (partial) physical examination, and Illinois 16 Rhode Island 16
serologic testing of the donor blood. Any one of these areas Indiana 16 South Carolina 16
may preclude a potential donor from the donation process. Iowa 16 South Dakota 16
The medical history information and physical examination Kansas 16 Tennessee 16
are designed to answer two questions: (1) Will a donation Kentucky 16 Texas 16
of approximately 450 mL of whole blood be harmful to the Louisiana 16 Utah 16
donor? (2) Could blood drawn from this donor at this time Maine 16 Vermont 16
potentially transmit a disease to the recipient? Maryland 16 Virgin Islands 16
Massachusetts 16 Virginia 16
Registration Michigan 16 Washington 16
Minnesota 16 West Virginia 16
As outlined in the AABB Standards, blood collection facilities Mississippi 16 Wisconsin 16
must confirm donor identity and link the donor to existing Missouri 16 Wyoming 16
donor records. Most facilities require a photographic identi- Montana 16
fication such as a driver’s license, passport, or school identi-
fication card. In addition, to prevent an ineligible donor from Parental/guardian consent required for 16-year-old.

donating again, every donor must be checked against a per-


manent record of previously deferred donors. The following
is a list of information used by the collection facility in the blood as a means of getting an HIV test. At some point in
registration process and is kept on record by use of a single the interview process, donors must sign a statement doc-
record form or electronically: umenting that they have given consent to the donation.
This is typically done at the end of the donor history
• Name (first, last, MI) questionnaire (DHQ).
• Date and time of donation Additional information: The following additional infor-
• Address mation may be helpful in some cases:
• Telephone • The name of the patient for whom the blood is intended
• Gender (self-identified and self-reported regarding trans- (directed donation)
gender donors)1 • Race of the donor for matching specific phenotypes
• Age or date of birth: The minimum age for an allogeneic • Cytomegalovirus (CMV) status (some patient groups,
donation is greater than or equal to 16 years depending such as neonates, require CMV-negative blood in cer-
on individual state requirements (Box 13-1).2 There is no tain circumstances).
upper age limit. For autologous donation (donating blood
to be used for oneself), there is no age restriction; how- Medical History Questionnaire
ever, each donor-patient must be evaluated by the blood
bank medical director. Obtaining an accurate medical history of the donor is essen-
• Consent to donate: Donors should be informed of the tial to ensure protection of the donor and benefit to the re-
procedure for donating blood and its potential risks. They cipient. A standardized medical history questionnaire, or
must also be given educational materials informing them donor history questionnaire (DHQ), was developed by a task
of the signs and symptoms associated with HIV (human force that included representatives from the AABB, the FDA,
immunodeficiency virus) infection and AIDS (acquired and the blood and plasma industry. The questionnaire was
immune deficiency syndrome), behaviors that put them designed to be self-administered by the donor but, if pre-
at high risk of infection, and a caution not to donate ferred, may be administered by a trained donor historian.
6888_Ch13_281-306 29/10/18 5:43 PM Page 284

284 PART III Transfusion Practices

Self-administered questionnaires must be reviewed by a temporary deferral, the donor cannot donate for a specified
trained personnel before completing the screening process time period, such as 2 weeks from receipt of the polio
and prior to collecting blood. The interviewer should be vaccine. In an indefinite deferral the donor is prohibited
familiar with the questions, and the interview should be con- from donating blood to another person for an unspecified
ducted in a secluded area of the blood center or donor site. period of time; in a permanent deferral the donor may never
The questions are designed so that a simple yes or no can be donate blood to another person. A donor may also be
answered and elaborated, if indicated. The medical history is deferred based on the mini-physical exam, for example,
conducted on the same day as the donation. There are three abnormal hemoglobin value. The current DHQ is depicted
types of deferrals: temporary, indefinite, and permanent. In in Figure 13–1.

Full-Length Donor History Questionnaire [DHQ]


YES NO
Are you
1. Feeling healthy and well today?
2. Currently taking an antibiotic?
3. Currently taking any other medication for an infection?
4. Have you taken any medications on the Medication Deferral List in the time frames indicated?
(Review the Medication Deferral List)
5. Have you read the educational materials today?
In the past 48 hours
6. Have you taken aspirin or anything that has aspirin in it?
In the past 8 weeks have you
7. Donated blood, platelets, or plasma?
8. Had any vaccinations or other shots?
9. Had contact with someone who was vaccinated for smallpox in the past 8 weeks?
In the past 16 weeks
10. Have you donated a double unit of red cells using an apheresis machine?
In the past 12 months have you
11. Had a blood transfusion?
12. Had a transplant such as organ, tissue, or bone marrow?
13. Had a graft such as bone or skin?
14. Come into contact with someone else’s blood?
15. Had an accidental needle stick?
16. Had sexual contact with anyone who has HIV/AIDS or has had a positive test for the HIV/AIDS virus?
17. Had sexual contact with a prostitute or anyone else who takes money or drugs or other payment for sex?
18. Had sexual contact with anyone who has ever used needles to take drugs or steroids, or anything not
prescribed by their doctor?
19. Male donors: Had sexual contact with another male?
20. Female donors: Had sexual contact with a male who had sexual contact with another male in the past 12
months?
21. Had sexual contact with a person who has hepatitis?
22. Lived with a person who has hepatitis?
23. Had a tattoo?
24. Had ear or body piercing?
25. Had or been treated for syphilis or gonorrhea?
26. Been in juvenile detention, lockup, jail, or prison for more than 72 consecutive hours?
In the past 3 years have you
27. Been outside the United States or Canada?
From 1980 through 1996,
28. Did you spend time that adds up to 3 months or more in the United Kingdom?
(Review list of countries in the UK)
29. Were you a member of the U.S. military, a civilian military employee, or a dependent of a member of the
U.S. military?
Continued

Figure 13–1. Revised donor history questionnaire.


6888_Ch13_281-306 29/10/18 5:43 PM Page 285

Chapter 13 Donor Selection 285

Full-Length Donor History Questionnaire — Continued


YES NO
From 1980 to the present, did you
30. Spend time that adds up to 5 years or more in Europe? (Review list of countries in Europe.)
31. Receive a blood transfusion in the United Kingdom or France?
Have you EVER
32. Female donors: Been pregnant or are you pregnant now?
33. Had a positive test for the HIV/AIDS virus?
34. Used needles to take drugs, steroids, or anything not prescribed by your doctor?
35. Received money, drugs, or other payment for sex?
36. Had malaria?
37. Had Chagas disease?
38. Had babesiosis?
39. Received a dura mater (or brain covering) graft or xenotransplantation product?
40. Had any type of cancer, including leukemia?
41. Had any problems with your heart or lungs?
42. Had a bleeding condition or a blood disease?
43. Have any of your relatives had Creutzfeldt-Jakob disease?
Use this area for additional questions

Figure 13–1. cont’d

Medical History Questions • Are you currently taking an antibiotic or taking any
other medications for an infection? Donors currently
The following is a summary of DHQ questions with elabo-
taking antibiotics for an infection or for prophylaxis after
ration and explanation. For more in-depth information on
dental surgery may be deferred temporarily until the
the questionnaire and interpretation of the questions please
donor has completed the prescribed medication regimen
refer to the AABB Technical Manual and the FDA website.
and the infection has cleared up. Donors who have taken
• Are you feeling healthy and well today? Donors should tetracycline or other antibiotics used to treat acne are ac-
appear to be in good health without obvious signs or ceptable for donation. All drugs and medications must be
symptoms of colds, flu, or other illness. cleared by the blood collection facility or blood bank
6888_Ch13_281-306 29/10/18 5:43 PM Page 286

286 PART III Transfusion Practices

medical director. If the donor’s response is yes, the inter- • In the past 6 weeks, have you been pregnant or are you
viewer must investigate further. Types and descriptions pregnant now? Female donors should be temporarily de-
of deferrals are listed in Box 13–2. ferred for 6 weeks following termination of pregnancy.2
• Are you taking or have you ever taken any medications Exceptions can be made by the blood bank medical
on the Medication Deferral List? The Medication Deferral director for an autologous donation if complications are
List (Fig. 13–2) was developed along with the DHQ, and it anticipated at delivery. A first-trimester or second-
is recommended that it be used in conjunction with the trimester abortion or miscarriage is not cause for defer-
DHQ. It can be found on the FDA website along with other ral. A 12-month deferral would apply if the woman
donor history documents. Each facility’s medical director received a transfusion during her pregnancy.
is responsible for determining if any other medications re- • In the past 8 weeks, have you donated blood, platelets,
quire a deferral. Most centers have a predetermined list of or plasma? In the past 16 weeks have you donated a
medications and their deferral requirements. double unit of red blood cells (RBCs) using an apheresis
• Have you read the educational materials? Prior to begin- machine? The time interval between allogeneic whole
ning the DHQ, prospective donors must be provided infor- blood donations is 8 weeks or 56 days. If the prospective
mation about the collection procedure and any risks donor has participated in an apheresis donation (platelets,
involved. Prospective donors must also be informed (in a leukocytes, granulocytes), then at least 48 hours must pass
language they can understand) about high-risk behavior re- before donating whole blood. The FDA limits platelet
lated to the AIDS virus and must be given the opportunity apheresis procedures to no more than 24 in a calendar
to ask questions regarding any aspect of the collection pro- year, unless approved by the blood bank medical director.
cedure. The donor needs to acknowledge that he or she has An apheresis donor may only donate twice in a period of
read and understands all of the material. This is generally 7 days and only once in 7 days for a double or triple
done by having the donor sign a statement indicating they apheresis donation. Infrequent plasma apheresis requires
have read and understand the materials, have answered the a 4-week deferral. An infrequent plasma apheresis can be
health history questions honestly, have been informed of the defined as undergoing the procedure no more frequently
risks of donation, and give their consent to the donation. than once every 4 weeks. In contrast, a serial plasma
• In the past 48 hours, have you taken aspirin or anything apheresis program is structured so that the donor under-
with aspirin in it? Donors who have taken piroxicam, goes the procedure more frequently than once every
aspirin, or anything with aspirin in it within 3 days of 4 weeks. The deferral time for a double RBC apheresis
donation may not be a suitable donor for platelet apheresis donation is 16 weeks due to the additional volume of
as these medications inhibit platelet function. There is no RBCs that are donated.
restriction for whole blood donation. However, if whole • In the past 8 weeks, have you had any vaccinations
blood is collected, random donor platelets may be pre- or other shots? Have you had contact with someone
pared but may not be used as sole source of platelets if the who had a smallpox vaccination? If a potential donor has
donor answered yes to this question. received a live attenuated or bacterial vaccine such as
measles (rubeola), mumps, oral polio, typhoid, or yellow
fever, there is a 2-week deferral; if the donor has received
a live attenuated vaccine for German measles (rubella) or
BOX 13–2
chickenpox, there is a 4-week deferral. However, there is
Types of Deferral no deferral for toxoids or killed or synthetic viral, bacte-
Temporary Deferral: Prospective donor is unable to donate blood rial, or rickettsial vaccines such as diphtheria, hepatitis A,
for a limited period of time. hepatitis B, influenza, Lyme disease, pneumococcal
EXAMPLE: Donor has received a blood transfusion; defer for polysaccharide, polio injection (Salk), anthrax, cholera,
12 months from date of transfusion. Donor received vaccination for pertussis, plague, paratyphoid, rabies, Rocky Mountain
yellow fever; defer for 2 weeks from date of vaccination. spotted fever, tetanus, or typhoid injection if the donor is
Indefinite Deferral: Prospective donor is unable to donate
blood for someone else for an unspecified period of time due to symptom-free and afebrile. Deferral for smallpox vaccina-
current regulatory requirements. This donor would not be able to tion is 14 to 21 days or until the scab has fallen off. In ad-
donate blood until the current requirement changes. These donors dition, donors who have been in close contact with
may be eligible to donate autologous blood. someone who was recently vaccinated are at risk of possi-
EXAMPLE: Donor states they have lived in England for 1 year in ble infection as well. The FDA defines close contact as
1989; defer indefinitely.
Permanent Deferral: Prospective donor will never be eligible exposure to the vaccination site or bandages, clothing,
to donate blood for someone else. These donors may be eligible to towels, or bedding that have been in contact with the vac-
donate autologous blood. Some permanent deferrals may result cination site. An additional question that could be posed
from the testing performed on a previous donation. to the prospective donor is “If you have had the smallpox
EXAMPLE: Donor states that he or she has hepatitis C; defer vaccine, has the scab fallen off by itself?”3 Plasma products
permanently.
From: Donor History Questionnaire user brochure, FDA May 2008. from recipients who have received the smallpox vaccine
Available at www.fda.gov. need not be quarantined as the vaccinia virus is inacti-
vated in the manufacturing process.
6888_Ch13_281-306 29/10/18 5:43 PM Page 287

Chapter 13 Donor Selection 287

MEDICATION DEFERRAL LIST


Please tell us if you are now taking or if you have EVER taken any of these medications:
Proscar© (finasteride) − usually given for prostate gland enlargement

Avodart© (dutasteride) − usually given for prostate enlargement

Propecia© (finasteride) − usually given for baldness

Accutane© (Amnesteem, Claravis, Sotret, isotretinoin) − usually given for severe acne

Soriatane© (acitretin) – usually given for severe psoriasis

Tegison© (etretinate) – usually given for severe psoriasis

Growth Hormone from Human Pituitary Glands − used usually for children with delayed or impaired growth

Insulin from Cows (Bovine, or Beef, Insulin) − used to treat diabetes

Hepatitis B Immune Globulin – given following an exposure to hepatitis B.


NOTE: This is different from the hepatitis B vaccine which is a series of 3 injections given over a 6 month period to prevent future
infection from exposures to hepatitis B.

Plavix (clopidogrel) and Ticlid (ticlopidine) – inhibits platelet function; used to reduce the chance for heart attack and stroke.

Feldene – given for mild to moderate arthritis pain

Experimental Medication or Unlicensed (Experimental) Vaccine – usually associated with a research protocol

IF YOU WOULD LIKE TO KNOW WHY THESE MEDICINES AFFECT YOU AS A BLOOD DONOR, PLEASE KEEP READING:

• If you have taken or are taking Proscar, Avodart, Propecia, Accutane, Soriatane, or Tegison, these medications can cause
birth defects. Your donated blood could contain high enough levels to damage the unborn baby if transfused to a pregnant woman.
Once the medication has been cleared from your blood, you may donate again. Following the last dose, the deferral period is one
month Proscar, Propecia and Accutane, six months for Avodart and three years for Soriatane. Tegison is a permanent deferral.

• Growth hormone from human pituitary glands was prescribed for children with delayed or impaired growth. The hormone was
obtained from human pituitary glands, which are found in the brain. Some people who took this hormone developed a rare nervous
system condition called Creutzfeldt-Jakob Disease (CJD, for short). The deferral is permanent.

• Insulin from cows (bovine, or beef, insulin) is an injected material used to treat diabetes. If this insulin was imported into the
US from countries in which “Mad Cow Disease” has been found, it could contain material from infected cattle. There is concern
that "Mad Cow Disease" is transmitted by transfusion. The deferral is indefinite.

• Hepatitis B Immune Globulin (HBIG) is an injected material used to prevent infection following an exposure to hepatitis B. HBIG
does not prevent hepatitis B infection in every case, therefore persons who have received HBIG must wait 12 months to donate
blood to be sure they were not infected since hepatitis B can be transmitted through transfusion to a patient.

• Feldene is a non-steroidal anti-inflammatory drug that can affect platelet function. A donor taking Feldene will not be able to
donate platelets for 2 days; however, its use will not affect whole blood donations.

• Plavix and Ticlid are medications that can decrease the chance of a heart attack or stroke in individuals at risk for these
conditions. Since these medications can affect platelets, anyone taking Plavix or Ticlid will not be able to donate platelets for
14 days after the last dose. Use of either medication will not prohibit whole blood donations.

• Experimental Medication or Unlicensed (Experimental) Vaccine is usually associated with a research protocol and the effect
on blood donation is unknown. Deferral is one year unless otherwise indicated by Medical Director.

Figure 13–2. Medication deferral list. (U.S. Food and Drug Administration, “Guidance for industry: Implementation of acceptable full-length Donor History Questionnaire
and accompanying materials for use in screening donors of blood and blood components,” October 2006.)

• In the past 12 months have you had a blood transfusion; • In the past 12 months have you come in contact with
a transplant such as organ, tissue, or bone marrow; or a someone else’s blood or had an accidental needle-stick
graft such as bone or skin? Donors who during the pre- injury? Had a tattoo? Had an ear or body piercing?
ceding 12 months have received a transfusion of blood or Deferral is 12 months due to potential exposure to sub-
its components or other human tissues (organ, tissue, stances known to be sources of bloodborne pathogens.
bone marrow transplant, or bone or skin graft) known to Exposure is assumed if the blood came in contact with an
be possible sources of bloodborne pathogens should be open wound or any nonintact skin or mucous membranes
deferred for 12 months from the time of receiving the (e.g., nose, mouth, eyes). Skin-penetrating injuries from
blood product or graft.2 instruments, equipment, needles, and so forth that are
6888_Ch13_281-306 29/10/18 5:43 PM Page 288

288 PART III Transfusion Practices

nonsterile and contaminated with blood or body fluids risk factor was aligned to the epidemiology for each infection
other than the donor’s own are also cause for deferral. This validating the current donor deferral criteria. A donor that
includes tattoos, permanent makeup, and ear and body had sex with an HIV-positive partner was associated with a
piercings unless applied by a state-regulated organization 132-fold increase in risk of being HIV positive, and a history
where sterile needles and ink are not reused. of MSM contact was associated with a 62-fold increase in risk
• In the past 12 months, have you had sexual contact with of being HIV positive.6 The REDS III study evaluated opin-
anyone who has HIV/AIDS or has had a positive test for ions from the MSM community through surveys that were
HIV/AIDS? Deferral is 12 months from the time of sexual web based. The qualitative data revealed that 90% of MSM
contact with a person with clinical or laboratory evidence thought that the donation deferral should change and that
of HIV infection or who is at high risk for infection. 59% of MSM said they would comply with the change to
• In the past 12 months, have you had sexual contact with a 1-year deferral. Some participants thought the policy
a prostitute or anyone else who takes money or drugs or was discriminatory toward MSM. The prevalence of HIV in-
other payment for sex? Deferral is 12 months from the fection in MSM who classified themselves as blood donors
time of sexual contact. is 0.25%.7 Lastly, countries such as Australia that also had
• In the past 12 months, have you had sex with anyone a volunteer blood donor program and an MSM population,
who has ever used a needle to take drugs or steroids or were analyzed. Australia changed from a lifetime deferral
anything not prescribed by their doctor? In the past to a 1-year deferral, resulting in no change in risk to their
12 months, have you ever had sex with anyone who has blood supply 5 years before the change and 5 years after the
hemophilia or has used clotting factor concentrates? The change in policy.8 Other countries that have adopted a
FDA mandates persons who have had sex with any person 1-year deferral policy include Argentina, Brazil, Hungary,
who is a past or present IV drug user should be deferred Japan, Sweden, and the United Kingdom. Ultimately, the de-
for 12 months; additionally, persons who have had sex cision to adopt a time-based deferral policy since last sexual
with any person with hemophilia or a related blood encounter involving MSM was postulated to be most effec-
disorder who has received factor concentrates should be tive in aligning with current serologic testing and epidemi-
deferred for 12 months. ology of the HIV virus. The 1977 benchmark had become
• Male donors: Have you had sexual contact with another less effective over time, and the once-pondered option to
male in the past 12 months? Meeting all other donor eligi- eliminate all HIV-related deferrals and rely just on current
bility requirements, males who have had sex with another testing was rejected because of possible new cases of HIV
male (MSM) may donate whole blood if they have not had that may go undetected.1
sex with another male in the past year.1
• Female donors: Have you had sexual contact with a
In 1985, the FDA began instituting an indefinite deferral male who has ever had sexual contact with another
for MSM since 1977, even if the sexual contact only happened male? Women who have had sex with an MSM in the past
once, to safeguard the public from transfusion-transmitted 12 months should be deferred for 12 months.1
HIV (TT-HIV). However, with more sensitive testing protocols • In the past 12 months, have you had sexual contact with
and viral inactivation measures, the Department of Health and a person who has hepatitis? Have you lived with a person
Human Services initiated public discussion toward a revision who has hepatitis? Sexual contact or living with a person
of policy. Multiple factors were involved, including operational (close contact) who has acute or chronic hepatitis B (test
assessment, DHQ studies, the Retrovirus Epidemiology Donor positive for HBsAg or HBV) or who has symptomatic hep-
Study II (REDS II), REDS III, and supportive data from other atitis C or other hepatitis virus requires a 12-month deferral
countries. Through operational assessment, it was determined following discontinuation of being in close contact.
that quarantine release errors involving HIV contributed min- • In the past 12 months, have you been treated for syphilis
imally to the risk of TT-HIV.4 This has been largely due to or gonorrhea? Prospective donors with a history of syphilis
computerized inventory management in which a barcode or gonorrhea or treatment for either, a reactive screening
reader is utilized to identify quarantined units. A DHQ study test for syphilis, or where no confirmatory test was per-
was conducted in 2011 in which donors who had sex with the formed, should be deferred for 12 months after completion
opposite sex and MSM were interviewed. It was found that of therapy. The agent that causes syphilis, Treponema
answering questions related to sexual behavior was more pallidum, may live for 1 to 5 days in cold storage so that
effective when they approached the question from the stand- only a fresh unit of RBCs may transmit infection. At room
point of their blood being safe. Participants in the study also temperature, however, this agent thrives very well, placing
recommended shorter donor educational materials and the platelet concentrates above RBCs as potentially supporting
option to answer the question with “I don’t know.”5 transfusion-transmitted syphilis. To date, however, only
The REDS II study evaluated four viral markers (hepatitis B three cases of transfusion-transmitted syphilis have been
virus [HBV], hepatitis C virus [HCV], human T-cell lym- documented. Donor centers must use a screening test for
photropic virus I, II [HTLV-I, II], and HIV) in the context of T. pallidum that has been licensed by the FDA.9
behavioral risk factors associated with donations that were • Have you been in juvenile detention, lockup, or prison
positive for one or more of the viruses. A key finding of this for more than 72 hours? Deferral is 12 months from the
pilot study was that for each viral infection, the behavioral last date of the incarceration.
6888_Ch13_281-306 29/10/18 5:43 PM Page 289

Chapter 13 Donor Selection 289

• In the past 3 years, have you been outside of the United a malaria-endemic area after residence for 3 years
States or Canada? This question is general and is used consecutively in a nonendemic country, that donor is
to detect a donor who may have an increased risk of deferred for 1 year from the time that they return to the
exposure to malaria, Creutzfeldt-Jakob disease or variant nonendemic country.”5
Creutzfeldt-Jakob disease (CJD or vCJD), and more re- • CJD and vCJD: Creutzfeldt-Jakob disease and variant
cently, leishmaniasis. Creutzfeldt-Jakob disease are members of a group of
• Malaria: “Prospective donors who have been diagnosed neurological disorders known as the transmissible
with malaria are deferred for 3 years after becoming spongiform encephalopathies (TSE) or prion diseases,
asymptomatic, irrespective of the receipt of antimalarial which affect sheep, cows, and humans. CJD results in
prophylaxis.”2 “Individuals who have lived longer than progressive dementia and spongiform alterations in the
5 consecutive years in countries considered malaria- brain and is rapidly fatal. CJD may be transmitted by
endemic by the Malarial Branch, Centers for Disease corneal transplants, human dura mater grafts, pituitary-
Control and Prevention, U.S. Department of Health and derived human growth hormone, and neurosurgical
Human Services (Box 13–3) are deferred for 3 years instruments.10 Humans can also be infected by eating
after departure from malaria-endemic countries.”2 In beef contaminated with prions, abnormal proteins
addition, those individuals who have lived longer than found in the brain tissue of diseased cattle, which
5 years in malaria-endemic countries and who have led to the term “mad cow disease.” Prions are highly
been a resident of nonendemic countries for less than resistant to heat and ultraviolet light, exposures that
3 consecutive years are deferred for 3 years. “After the effectively kill bacteria and viruses. In TSE, prions
3-year deferral period, the donor may be eligible to do- induce abnormal folding of normal proteins, leading to
nate, provided the donor has been free from malaria neurological symptoms and death.11 When humans
during this period and meets all other donor eligibility ingest contaminated beef from a diseased cow they can
criteria.”5 “Individuals who have traveled to an area develop vCJD. As of 2015, 228 patients have been
where malaria is endemic are deferred for 12 months diagnosed with clinical vCJD. These cases involved the
after departure from the malaria-endemic area.”2 “If a following countries: United Kingdom, United States,
prior resident of a malaria-endemic country returns to Canada, France, Austria, Belgium, Czech Republic,

BOX 13–3

Countries Endemic for Malaria


Afghanistan Dominican Republic Madagascar Sierra Leone
Angola Ecuador Malawi Solomon Island
Argentina El Salvador Malaysia Somalia
Azerbaijan Equatorial Guinea Mali South Africa
Bangladesh Eritrea Mauritania South Sudan
Belize Ethiopia Mayotte Sri Lanka
Benin French Guiana Mexico Sudan
Bhutan Gabon Mozambique Suriname
Bolivia Georgia Myanmar Swaziland
Botswana Ghana Namibia Tajikistan
Brazil Guatemala Nepal Thailand
Burkina Faso Guinea Nicaragua The Gambia
Burundi Guinea-Bissau Niger Timor-Leste
Cambodia Guyana Nigeria Togo
Cameroon Haiti Pakistan Turkey
Cape Venda Honduras Panama Uganda
Central Africa Republic India Papua New Guinea Uzbekistan
Chad Indonesia Paraguay Vanuatu
Colombia Iran Peru Venezuela
Comoros Iraq Philippines Vietnam
Congo Kenya Rwanda Yemen
Costa Rica Korea Sao Torne and Principle Zambia
Côte d’lvoire Kyrgyzstan Saudi Arabia Zimbabwe
Democratic Republic of the Congo Lao Senegal
Djibouti Liberia
6888_Ch13_281-306 29/10/18 5:43 PM Page 290

290 PART III Transfusion Practices

Denmark, Finland, Germany, Greece, Ireland, Italy, United Kingdom or France since 1980 should be indef-
Liechtenstein, Luxembourg, the Netherlands, Poland, initely deferred as a blood donor.
Portugal, Spain, Switzerland, Sweden, Israel, and Japan. Have you ever:
Box 13–4 compares and contrasts CJD and vCJD • Had a positive test for HIV/AIDS virus? Indefinite
processes. deferral is required for any person with clinical or lab-
A permanent deferral is indicated for anyone diag- oratory diagnosis of HIV infection.
nosed with CJD or vCJD or if the donor received a dura • Used needles to take drugs, steroids, or anything not
mater transplant or pituitary growth hormone from a prescribed by your doctor? Donors’ arms should be
human cadaver. An indefinite deferral is given to any- checked for evidence of scars or punctures indicating
one who is a blood relative of someone diagnosed with the administration of self-injected drugs or for presence
CJD or vCJD. If the donor spent 3 or more months in of any skin lesions in the venipuncture site. Donors
the United Kingdom from 1980 to 1996 and/or 5 years with evidence of past or present nonprescription drug
or more in France from 1980 to the present, the donor use should be indefinitely deferred.
is indefinitely deferred. For persons who served in • Used clotting factor concentrates? Prospective donors
the U.S. military, an indefinite deferral is given for with hemophilia or other bleeding disorders and who
those who spent 6 months or more in Northern Europe receive clotting factor concentrates should be deferred
(Germany, UK, Belgium, the Netherlands) from 1980 to from donating blood or components unless otherwise
1990 or spent 6 months or more from 1980 to 1996 in approved by the medical director because there is a high
Greece, Turkey, Spain, Portugal, or Italy. If a donor re- risk of bleeding following venipuncture. Receipt of
ceived a blood transfusion in the United Kingdom from clotting factor concentrates, such as receipt of blood
1980 to the present, the donor is indefinitely deferred; transfusions, mandates a 12-month deferral.
if a donor received bovine insulin since 1980, the donor • Had hepatitis? Donors with a positive test for HBsAg
is indefinitely deferred unless the bovine insulin did not should be permanently deferred. If a donor reacted
come from the United Kingdom.10 positively for anti-HBc on more than one occasion, then
• Leishmaniasis: Leishmania spp. are intracellular proto- they must be indefinitely deferred. Donors are also in-
zoan parasites that cause leishmaniasis. They are en- definitely deferred if they had a positive HBV nucleic
demic in tropical and subtropical areas in the Middle acid testing (NAT).1 If a donor has a clinical history or
East, Mediterranean coast, Africa, Central and South diagnosis of viral hepatitis at age 11 or later, then the
America, and Asia. The parasite is transmitted to donor must be permanently deferred. If, on the other
humans by the sand fly (Phlebotomus) infected with hand, the donor was diagnosed with viral hepatitis be-
Leishmania. Clinical manifestations range from cuta- fore age 11, the donor should be temporarily deferred
neous lesions to visceral infections that may be fatal. and blood and blood components collected from the
There have been 15 cases of transfusion-transmitted donor must not be used for further manufacture until
leishmaniasis reported in the literature, primarily the clinical circumstances are thoroughly reviewed by
involving Leishmania donovani.12 In 2003, with the medical personnel.13
deployment of military and National Guard troops in • Had Chagas disease? Had babesiosis? A history of
Iraq, a 12-month deferral from the date of departure Chagas disease or babesiosis is cause for indefinite defer-
from the area was instituted for any military personnel ral. Chagas disease, also known as American trypanoso-
stationed in Iraq and any civilian and contract person- miasis, is caused by the protozoan parasite Trypanosoma
nel who have visited the country. cruzi. The vector responsible for transmitting the parasite
• Received a blood transfusion in the UK or France: is the hematophagous bug belonging to the family Redu-
Those who have received a transfusion of blood, vidae. Transmission occurs when mucous membranes
platelets, plasma, cryoprecipitate, or granulocytes in the or breaks in the skin are contaminated with the feces of

BOX 13–4

Comparison of CJD and vCJD


CJD vCJD
Age of onset Later in life Early in life
Median age of death 68 years 28 years
Sensory symptoms Later in illness Early in illness
Median duration of illness 4 months 13 months
Immunohistochemistry No abnormal accumulation of prion protein in Abnormal accumulation of prion protein in
lymphoid tissue lymphoid tissue

CJD= Creutzfeldt-Jakob disease; vCJD= variant Creutzfeldt-Jakob disease


6888_Ch13_281-306 29/10/18 5:43 PM Page 291

Chapter 13 Donor Selection 291

infected hematophagous bugs. Chagas disease is endemic


in parts of Central and South America and Mexico, where BOX 13–5
an estimated 11 million persons are infected with T. cruzi. Current List of Countries with HIV-1 Group O Risk
Approximately 300,000 persons residing in the United
Benin
States and Canada may be infected with T. cruzi.14 Chagas
Cameroon
disease may be transmitted congenitally by breastfeeding,
Central African Republic
organ transplants, or blood transfusion. In the United
Chad
States and Canada, there have been seven documented
Congo
cases of transfusion-associated transmission of Chagas
Equatorial Guinea
disease in the past 20 years, all occurring in immunosup-
Gabon
pressed recipients.
Kenya
There are 70 known species of Babesia and at least
Niger
five are identified as infecting humans. Babesia microti
Senegal
utilizes the vector Ixodes scapularis to infect the human
Togo
and transmit the parasite. The Babesia organism pen-
Zambia
etrates the erythrocyte where the trophozoite multi-
plies; upon lysis, the RBC merozoites are released into
the blood where they are free to infect other RBCs.
Transfusion-associated infection with Babesia carries or other medical treatment with a blood product, they
an incubation period of 2 to 8 weeks;15 symptoms may should be deferred indefinitely.
include malaise, fatigue, anorexia, arthralgia, nausea, • Had sexual contact with anyone who was born or has
vomiting, abdominal pain, and fever reaching temper- lived in Africa? The FDA recommends indefinite defer-
atures of 40°C. ral for any prospective donor who indicates having had
• Received a dura mater (or brain covering) graft? Due sex with a person who was born in any of the African
to an increased risk of exposure to CJD or vCJD, countries (Box 13-5) targeted with increased risk of
prospective donors with a history of a dura mater graft HIV-1 group O infection. Moreover, if the collecting
require a permanent deferral. facility has implemented testing for HIV-1/2 or testing
• Had any type of cancer, including leukemia? A history that has been licensed for use in detecting antibodies to
of cancer, leukemia, or lymphoma is generally a cause HIV-1 group O, the questions concerning residence in
for indefinite deferral. Any donor presenting with a or travel to specific African countries or sexual contact
history of cancer should be evaluated by the blood with persons from those countries may be omitted. In
bank medical director. The exceptions include basal or addition, donors who were previously deferred prior to
squamous cell cancer, carcinoma in situ of the cervix, the availability of the HIV-1 group O testing may be
and papillary thyroid carcinoma that has been surgically reentered provided it has been at least 1 year since the
removed. last potential exposure to HIV-1 group O and the
• Had any problems with your heart or lungs? A history screening test results are nonreactive.16
of cardiovascular, coronary, or rheumatic heart disease • Have any of your relatives had Creutzfeldt-Jakob
is usually a cause for deferral; however, in the absence disease (CJD)? The FDA recommends prospective
of disability or restrictions by the patient’s physician, donors with a family history of CJD be indefinitely
the donor may be accepted on a case-by-case basis deferred unless the diagnosis of CJD was confidently
by the blood bank medical director. Active pulmonary excluded. If the donor was deferred due to family his-
tuberculosis or other active pulmonary disease is cause tory, he or she may be reentered if the diagnosis of CJD
for deferral. in the family member(s) is confidently excluded or CJD
• Had a bleeding condition or a blood disease? Donors in- in the family member(s) is iatrogenic, or if appropriate
dicating a history of a bleeding problem following surgery, laboratory testing such as gene sequencing shows the
invasive dental procedures, cuts, or abrasions must be donor does not have a mutation found in association
further evaluated by the blood bank medical director. Dis- with familial CJD.10
eases of the blood such as hemophilia, von Willebrand • Have you ever received a xenotransplantation? While
disease, sickle cell anemia, thalassemia, Kaposi’s sarcoma, nonliving biologics or materials from nonhuman ani-
polycythemia, or a history of receiving clotting factor con- mals such as porcine heart valves and porcine insulin
centrates are causes for indefinite deferral. are acceptable, a donor is permanently deferred if the
• Been in Africa? HIV-1 group O virus is endemic to cen- donor received cells, tissues, or organs from a nonhu-
tral and west regions of Africa. Anyone who was born man animal source.
or lived in any of the African countries on the FDA list
of countries (Box 13–5) with HIV-1 group O risk since The Physical Examination
1977 should be deferred indefinitely. In addition, if the
prospective donor has traveled to any of the countries The donor center representative evaluates the prospective
on the list since 1977 and received a blood transfusion donor with regard to general appearance, weight, temperature,
6888_Ch13_281-306 29/10/18 5:43 PM Page 292

292 PART III Transfusion Practices

hemoglobin, and presence of skin lesions. A blood bank physi- • Skin lesions: Prior to donation, the donor’s arms should
cian should be available to evaluate any special considerations. be inspected for skin lesions. Evidence of skin lesions
(e.g., multiple puncture marks) is cause for indefinite de-
• General appearance: The donor center representative
ferral. Skin disorders that are not cause for deferral include
should observe the prospective donor for presence of ex-
poison ivy and other rashes; these, however, should not
cessive anxiety, drug or alcohol influence, or nervousness.
be present in the area of the venipuncture site and may
If possible, this should be done in a gentle manner so as
need to be evaluated by a blood bank physician.
to not deter the donor from future donations.
• Weight: All donors should weigh at least 110 lbs.2 Stan-
Informed Consent
dards mandates a maximum of 10.5 mL of blood/kg of
donor weight for whole blood collection, inclusive of pilot AABB Standards mandates that informed consent of allo-
tubes for testing. If the donor weighs less than 100 pounds, geneic, autologous, and apheresis donors be obtained before
the amount of blood collected must be proportionately donation. The donor must be informed of the risks of the
reduced as well as that of the anticoagulant. The following procedure and of the tests that are performed to reduce
formulas can be used to calculate the adjusted volume the risk of infectious disease transmission to the recipient.
of blood to be collected and anticoagulant (CPD and The donor must be able to ask questions concerning any
CPDA-1) to be used. element of the collection or testing process. If the donor is
Volume to collect = (donor’s weight in kg/50) × 450 mL. a minor or is unable to comprehend the informed consent
Volume to collect/450 × 63 mL = reduced volume of anti- protocol, applicable state law provisions will intercede. An
coagulant. example of an informed consent is shown in Figure 13–3.
63 mL – above calculated volume = amount of solution to
be removed. Autologous Donation
• Temperature: Standards mandates the donor temperature
must be less than or equal to 37.5°C or 99.5°F. Donors are An autologous donor is one who donates blood for his or
asked not to drink coffee or hot beverages while waiting her own use; thus, such a donor is referred to as the donor-
to donate, as this may sometimes affect their temperature. patient. Most autologous blood is used to treat surgical blood
Oral temperatures that are lower than normal are not loss in very specific situations where there is a reasonable
cause for deferral. opportunity to avoid homologous transfusions or when
• Hemoglobin: The donor’s hemoglobin level should be compatible allogeneic blood is not available. The potential
greater than or equal to 12.5 g/dL and the hematocrit level advantage of using autologous blood over allogeneic blood
greater than or equal to 38% for women in allogeneic includes a decreased risk of disease transmission, transfusion
donations and greater than or equal to 13.0 g/dL (hemo- reactions, and alloimmunization. However, there is still a risk
globin) and 39% (hematocrit) for men. For autologous of bacterial contamination, circulatory overload, cytokine-
donation, the hemoglobin and hematocrit level should be mediated reactions, and misidentification of the product
greater than or equal to 11.0 g/dL and 33%, respectively. or patient. The patients for whom autologous donation
The methods used for measuring hemoglobin include or transfusion holds the greatest advantage are those with
copper sulfate or point-of-care instruments using spec- very rare blood types and those with multiple antibodies for
trophotometric methodology. A hematocrit or packed cell which compatible units in the general blood supply may be
volume can be determined manually by centrifugation. difficult or impossible to find.
The earlobe should not be used for hemoglobin or hemat- The disadvantages of autologous donation or transfusion
ocrit determination.2 include a higher cost due to added administrative processes

Donor Release:
I have read and I understand the information contained in the Responsibilities of a Blood Donor and Important Information About
Donating Blood Information sheet. I understand that if my behavior or physical condition puts me at risk to transmit AIDS or any other
disease, I should not donate blood. The medical history I have given is truthful and accurate to the best of my knowledge. I have not
participated in activities considered high risk for acquiring and transmitting AIDS.
I give my permission for all laboratory testing necessary to provide safe blood to the recipient including tests to detect exposure to
hepatitis, AIDS, and other transfusion-transmitted diseases. Some of these tests may be unlicensed and for research only. Some
positive test results are routinely reported to the State Health Department as required by law for notification of sexual partners. I realize
that LifeSouth is not a testing center and that by giving blood I am not guaranteed that disease testing will be performed.
Although donating blood is normally a pleasant experience, I realize that short-term side effects can occur, such as bruising, dizziness,
or fainting, and that, in rare cases, a donor may experience infection or nerve damage.
I voluntarily donate my blood to the LifeSouth Community Blood Centers to be used as the blood center deems advisable.

Donor: ______________________________________________________ Date: _______________________________________

Figure 13–3. Sample informed consent.


6888_Ch13_281-306 29/10/18 5:43 PM Page 293

Chapter 13 Donor Selection 293

and special labeling requirements to ensure that units Most autologous donor units are a standard 450 or
get transfused to the proper patient. It should be noted that 500 mL ± 10%; however, if the donor weighs less than
there is a high percentage of wasted units (30% to 50%), 50 kg (110 lbs), the total amount of blood collected must
because patients end up not requiring any or all of the be reduced proportionately. If the amount of blood to be
units donated.17 In addition, the AABB Standards2 does not collected is determined to be 300 to 405 mL for a 450-mL
permit “crossing over” of unused autologous units into the bag or 333 to 449 in a 500-mL bag, the unit must be labeled
general inventory, except in exceptional circumstances. The as low volume.2 There is no requirement to reduce the
decision must be approved by the medical director on a case- volume of anticoagulant-preservative solution; however,
by-case basis. the plasma is not suitable for transfusion. If the unit must
Although the use of autologous blood has decreased in be less than 300 mL, then the anticoagulant-preservative
the past several years, it is still a viable and common alter- solution must be adjusted and approval by the blood bank
native therapy for select patients. There are various methods medical director is required. See the earlier section “The
and techniques for obtaining autologous blood, including: Physical Examination” for formulas to calculate the volume
and anticoagulant adjustments.
Preoperative collection. Testing requirements for autologous donor units are
Acute normovolemic hemodilution. somewhat less stringent than with allogeneic units. The col-
Intraoperative collection. lecting facility must determine the ABO and Rh of the blood,
Postoperative collection. but antibody screening is optional. If the collecting and the
transfusing facilities are the same, then viral marker testing
Preoperative Collection is not required; however, if they are different, the collecting
Preoperative collection occurs during the 5 to 6 weeks facility must test for HBV DNA, HBsAg, anti-HBc, anti-HCV,
immediately preceding a scheduled, elective surgical proce- HCV RNA, anti-HIV-1/2, HIV-1 RNA, anti-HTLV-I/II, WNV
dure unless the red blood cells and plasma are scheduled to RNA, and STS on at least the first unit collected from the
be frozen. Procedures that typically might use preoperative donor in every 30-day period. If any of the markers yield a
autologous blood include orthopedic procedures, vascular positive or reactive result, the patient’s physician and trans-
surgery, cardiac or thoracic surgery, and radical prostatec- fusing facility must be notified of the result.2
tomy. Although some women do participate in an autologous The transfusing facility must reconfirm the ABO and
collection program during pregnancy for unforeseen com- Rh of the unit, but a crossmatch is optional. However, an
plications, blood is seldom needed except in cases in which immediate spin crossmatch would be a good safety check
the mother has multiple antibodies to high-frequency anti- confirming that the selected unit is identified properly.
gens or risk for placenta previa or intrapartum hemorrhage. Units collected for autologous use must be labeled appro-
The decision to use preoperative autologous blood re- priately. The label should include the patient’s full name,
quires both an order from the patient’s physician (Fig. 13–4) unique identifying number (UIN), expiration date of the
and an approval from the blood bank medical director. The unit, and the name of the facility where the donor-patient
maximal surgical blood order schedule (MSBOS) can provide will be transfused. The label must also clearly state “For
guidance for surgical procedures to estimate the number of Autologous Use Only” (Fig. 13–5). Autologous units also
units needed for transfusion. The last blood collection generally have a distinct green label and tag (Fig. 13–6). This
should occur no later than 72 hours (3 days) before the is done both to ensure the unit is linked correctly with the
scheduled surgery to allow for volume replacement. donor-patient and to make the blood bank technologists
Medical history and physical exam requirements for aware that certain patients have autologous units on the shelf
autologous donation are less stringent than those for allo- that must be transfused before allogeneic units; more specif-
geneic donations. There is no minimum or maximum age ically, the oldest units should be transfused first. Blood banks
requirement; however, the donor must be able to tolerate the should have a system in place to ensure autologous units are
donation, and for younger donors, most centers limit the age selected first.
to children whose veins can accommodate the phlebotomy
needle and who can understand the procedure. The mini- Acute Normovolemic Hemodilution
mum hemoglobin/hematocrit level is 11.0 g/dL and 33%, Acute normovolemic hemodilution (ANH) results in the
respectively. The donor-patient’s temperature should not be collection of whole blood with the concurrent infusion of
elevated or indicate any sign of possible infection. Medical crystalloid or colloid solutions, thus maintaining a normal
history questions can be limited to those needed to ensure blood volume but decreasing the patient’s hematocrit. The
the safety of the donor, including cardiac history, bleeding ratio of replacement is 3:1 for crystalloids and 1:1 for
disorders, major illness, previous donor reactions, fainting colloids. The number of units collected depends on the
problems, and so forth. In addition, there should be questions patient’s ability to tolerate the decrease in hemoglobin/
to rule out the possibility of bacteremia in the donor-patient. hematocrit. A limited hemodilution will reduce the hemat-
This would include information on current medications, ocrit to 28%; severe dilution will reach 21% or less.18 It is
antibiotics, recent infections, fever, recent minor procedures recommended that the patient start with a hemoglobin of
such as dental procedures, gastrointestinal problems, or at least 12.0 g/dL. The procedure is performed in surgery
diarrhea. immediately prior to beginning the surgical procedure and
6888_Ch13_281-306 29/10/18 5:43 PM Page 294

294 PART III Transfusion Practices

*An appointment is recommended for this service. Please contact your local blood bank for information.

Branch Location:
Reset Form

1221 Northwest 13th Street


Gainesville, Florida 32601
(352) 334-1000 Request For Autologous Collections

Patient's Name SS# Birth Date

Patient's Phone Number Patient's Address

Date of Surgery/Transfusion Diagnosis Hospital

If patient is also under treatment for, or has any, preexisting medical condition(s), please indicate below.

, MD Condition

, MD Condition

, MD Condition

Please list all medications prescribed for this patient:

LifeSouth Community Blood Centers will process and store this blood for subsequent replacement transfusion. The number of units drawn
and the interval between donations will be determined by the patient's hemoglobin and anticipated blood requirements and will be at the
discretion of the blood center's Medical Director. Units will be held only until outdate (42 days from the date drawn) unless specific
arrangements are made with the blood center prior to that date. Blood should not be drawn from the donor-patient within 72 hours of the
time of anticipated surgery or transfusion. A cardiac release is required for all patients with a history of cardiac problems; units will not be
collected until the blood center has received written release from the patient's cardiologist.
Physicians will be notified in writing of abnormal test results as soon as possible. It is the responsibility of the ordering physician to notify
the patient.

Physicians, please complete:


I have explained the reasons for and risks involved in autologous collection to this patient and have considered
the need for iron supplementation:

#Units
• Packed Cells Signature, MD Date
• Whole Blood Physician (printed name)
• Fresh Frozen Plasma Phone
• Cryoprecipitated AHF

.............................................................................................. Blood Center Use Only .....................................................................................

Verbal Order Taken by Date

Donation Date Unit Number Donation Date Unit Number

Donation Date Unit Number Donation Date Unit Number

Apr 99 ver 1.0

Figure 13–4. Autologous donation permission form.

is managed by anesthesiology. The blood is collected in stan- maintaining the viability of both platelets and coagulation
dard blood bags containing anticoagulant or preservative factors.
and is stored in the room at room temperature. The blood Blood units are reinfused in the reverse order of collec-
is normally reinfused to the patient during or immediately tion so that the last unit reinfused carries the highest hemat-
following the surgery, but within 8 hours of collection, thus ocrit level. Generally, because the blood does not leave the
6888_Ch13_281-306 29/10/18 5:43 PM Page 295

Chapter 13 Donor Selection 295

See circular of information for for contamination of the surgical site by bowel contents,
indications, contraindications,
cautions and methods of infusion amniotic fluid, urine, clotting agents, and so forth, or
AUTOLOGOUS DONOR where there is a risk of bacterial contamination or activa-
This product may transmit infectious agents.
Caution: Federal law prohibits dispensing
tion of coagulation factors.
without a prescription. As with ANH, the blood generally does not leave the OR;
PROPERLY IDENTIFY INTENDED RECIPIENT
however, if some of the blood is to be stored postoperatively,
DONOR NAME
it must be labeled with the patient’s full name, UIN, date and
time of collection, and with “For Autologous Use Only.” The
DONOR SIGNATURE blood may be stored at room temperature for up to 6 hours
or at 1°C to 6°C for up to 24 hours,19 as long as the latter
Social Security # Date of Birth
temperature has begun within 4 hours from the end of
collection. Hospitals should establish their own policies
Figure 13–5. Autologous donation label.
and procedures for this type of collection.

Postoperative Blood Salvage


FOR AUTOLOGOUS USE ONLY Postoperative blood salvage is collected from a drainage tube
Unit # placed at the surgical site. It is reinfused, with or without
Patient processing, via a microaggregate filter to screen out any
Hospital debris. This blood is characterized as being dilute, partially
SSN or Hospital # hemolyzed, and defibrinated. It is recommended that no
Date needed
more than 1,400 mL be reinfused.18 Procedures that have
Signature
LifeSouth Community Blood Centers, Gainesville, FL 32601
used postoperative blood collection include orthopedic (e.g.,
arthroplasty) and cardiac surgeries. Blood must be reinfused
Figure 13–6. Autologous donation tag. within 6 hours of collection or it is to be discarded. Hospitals
should establish their own policies and procedures for this
type of collection.
operating room (OR), the units are not tested, labeled, or The advantage of this procedure is that it has a very low
tracked, and the blood bank is not involved. However, if the cost, and it can be used in conjunction with other autologous
blood is not transfused during surgery, it can be stored for blood collection procedures to avoid the need for allogeneic
up to 24 hours in a monitored blood bank refrigerator if blood transfusions. Because the blood does not leave the pa-
refrigerated within the first 8 hours. If the units leave the tient’s bedside, there is no risk of the blood being transfused
OR and are stored, they must be appropriately tested and to the wrong person. The disadvantage of postoperative
labeled as with preoperative autologous units. salvage is that it generally does not yield a large volume of
blood and by itself would not produce enough blood for
Intraoperative Collection
the patient’s needs. In addition, it carries a significant risk of
Intraoperative autologous collection involves collecting producing febrile nonhemolytic transfusion reactions due to
shed blood from the surgical site; processing the blood the presence of activated coagulation factors, fibrin degrada-
through an instrument that washes it with saline to remove tion products, and cytokines.
tissue debris, free hemoglobin, and plasma that may contain The blood bank is typically not responsible for managing
activated coagulation factors; concentrating the residual ANH intraoperative recovery and postoperative salvage
RBCs to a hematocrit of 50% to 60%; and then reinfusing collection procedures in most settings. However, the AABB
those cells immediately. This process is repeated continually and the College of American Pathologists (CAP) have rec-
during the surgical procedure. This type of collection has ommendations and guidelines that require the blood bank
been used in cardiothoracic, major orthopedic, and cardiac medical director or supervisor to assist in the development
surgeries, in addition to vascular surgeries, such as liver and implementation of the programs. This includes assisting
transplantation. The advantage of using intraoperative with the development of procedures, maintaining the equip-
autologous blood collection is that it may be used in cases ment, monitoring quality control, training personnel, and
where preoperative donation is not possible due to the assessing competency.
urgency of the surgery, or when the patient cannot be sched-
uled for multiple preoperative donations. In addition, the Directed Donation
risk of misidentifying the patient and blood product is min-
imized, and there are no labeling, testing, and storing costs. A directed donation is a unit collected under the same re-
The disadvantages include the high cost of the instru- quirements as those for allogeneic donors, except that the
mentation involved and training of the personnel to run unit collected is directed toward a specific patient. Often,
the instrument. In addition, frequently the amount of when a friend or family member needs blood, the donor cen-
blood that is collected is not sufficient for the patient’s ter will accommodate these directed donations so that the
total needs, and allogeneic blood may still need to be required testing may be done as soon as possible; if the blood
given. The procedure is contraindicated if there is potential is compatible, it can be used by the patient. The tag for the
6888_Ch13_281-306 29/10/18 5:43 PM Page 296

296 PART III Transfusion Practices

directed unit is a distinct color (e.g., yellow, salmon) to dif- affected. In addition, donors on Plavix (clopidogrel) or Ticlid
ferentiate it from autologous tags (Fig. 13–7). If the donor is (ticlopidine) should be deferred for 14 days after the last
a blood relative, the unit must be irradiated to prevent trans- dose,2 which is the time required for the medication to clear
fusion-associated graft-versus-host disease (TA-GVHD) the system and the platelet function to return to normal. If
so that viable T cells from the donor that enter the patient’s the donor has donated whole blood, or if 100 mL or more of
circulation do not mount an attack against the patient’s cells red blood cells were not able to be returned to the donor
and tissue. A system should be in place to ensure directed on the previous apheresis procedure, the donor must be de-
units from blood relatives are irradiated. ferred for 8 weeks to allow time for the donor to replenish
the lost RBC mass. Certain exceptions can be made for com-
Apheresis Donation pelling medical reasons, provided a physician certifies that
the donor will not be harmed, and the blood bank medical
Apheresis collection is an effective mechanism for collecting director agrees.
a specific blood component while returning the remaining Although a platelet count is not required on the first
whole blood components back to the patient. Most apheresis donation, it is required if the interval between donations is
instruments use an automated cell separator device whose less than 4 weeks; in that case, the platelet count must be
centrifugal force separates blood into components based on above 150,000/µL.19 The total amount of plasma that can
differences in density. (Refer to Chapter 18,”Apheresis.”) be removed along with the platelets is limited to 500 mL
Apheresis can be used to collect platelets, plasma, white (600 mL for donors weighing more than 175 lbs) or the
blood cells (leukocytes), red blood cells, and stem cells. It amount of plasma stated in the directions circular for the
is designed to collect large volumes of the intended compo- automated instrument being used for the collection. Each
nent and is the only effective method for collecting leuko- apheresis platelet unit is required to contain at least 3 × 1011
cytes and stem cells. The donor requirements for apheresis platelets. The automated instruments used today are very
donation are generally the same as for whole blood donation; efficient, and a single donor collection frequently yields a
however, there are some differences, depending on the com- high enough platelet count to allow the collection to be split
ponent that is to be collected. As with whole blood donation, into two or three individual products (refer to Chapter 18).
the process is regulated by the FDA, and both the AABB and Donor reactions to plateletpheresis collections are most com-
the American Society for Apheresis (ASFA) provide compre- monly a reaction to the citrate or anticoagulant used in the
hensive guidelines and standards. procedure. Vasovagal and hypovolemic reactions are very
rare but have been reported.
Plateletpheresis
Testing requirements are the same as for other allogeneic
Today the majority (more than 75%) of platelet transfusions blood products, including ABO group/Rh type, antibody
are pheresis-derived platelets.18 An apheresis platelet unit is screen, and infectious disease markers. If the donor is do-
equivalent to six to eight random donor platelets, so a single nating repeatedly for a specific patient, repeat testing need
product is a typical therapeutic dose for most adult patients. only be done every 30 days. In addition, the platelet count
This significantly reduces the recipient’s donor exposure, on each product is determined and recorded but does not
makes routine leukoreduction of the product practical, and have to be recorded on the product label. If there are visible
allows compatibility matching of the donor and patient pos- RBCs in the finished product, the volume of RBCs must be
sible in cases where the patient has become alloimmunized determined. If the product contains more than 2 mL of RBCs,
and refractory to random donor platelets. Plateletpheresis a pilot sample must be attached to the product and used by
donors may donate more frequently than donors of whole the transfusing facility to crossmatch the product with the
blood (8 weeks or 56 days). The interval between platelet- intended recipient. FDA guidelines require that the donation
pheresis donations is at least 2 days, not to exceed more than records of regular plateletpheresis donors be reviewed by a
twice a week or more than 24 times a year.2 physician at least every 4 months.20
Donors who have ingested aspirin, Feldene, or aspirin-
containing medications should be deferred for 48 hours Plasmapheresis
because these medications interfere with platelet adhesion Plasma was the first product to be collected by apheresis
and all of the platelets in a given therapeutic dose would be methods and was primarily used as a method for collecting
“source plasma,” which is further manufactured into plasma
derivatives. Today plasma apheresis is also used to collect
DIRECT DONATION transfusable fresh-frozen plasma. Plasmapheresis donors are
Unit # Component classified as either “infrequent/occasional” or “serial,” de-
Patient pending on the frequency of donations. An infrequent donor
Patient Blood Type D.O.B undergoes no more than one procedure in a 4-week period.2
Hospital
Serial donors may donate more frequently than 4 weeks
SSN or Hospital #
Date needed but no more than every 48 hours and no more than two
LifeSouth Community Blood Centers, Gainesville, FL 32601 donations in a 7-day period.
The donor requirements for both infrequent and serial
Figure 13–7. Directed donation tag. donors are the same as for allogeneic whole blood donors,
6888_Ch13_281-306 29/10/18 5:43 PM Page 297

Chapter 13 Donor Selection 297

although serial donors have some additional requirements to of these growth factors is that they can produce four to eight
protect the donor from excessive red blood cell and plasma times the volumes of cells in each collection compared with
protein loss. The RBC loss must not exceed 25 mL/week or other agents. In addition, these growth factors appear to be
200 mL in an 8-week period. As with any apheresis proce- quite well tolerated by the donor. Each collection facility
dure, if the donor’s red blood cells cannot be returned, the should develop policies outlining maximal doses of stimu-
donor must be deferred for 8 weeks before returning to a lating agents used in leukapheresis with appropriate con-
plasmapheresis program. At the start of a serial apheresis traindications for donors.
program and at 4-month intervals, the donor must be tested Each leukapheresis product must be tested for ABO group
for total serum/plasma protein levels and quantitative im- and Rh type as well as the HLA type of the leukocytes. Most
munoglobulin levels, and protein electrophoresis must be leukocyte concentrates are contaminated with some RBCs.
performed. The levels must remain in the normal range, or As stated for platelet concentrates, if the amount of contam-
the donor must be removed from the program until the levels inating RBCs exceeds 2 mL, the product should be cross-
return to normal. matched with the recipient,2 and a pilot tube sample must
If the plasmapheresis procedure is being performed man- accompany the product.
ually, there must be a mechanism in place to ensure positive
donor and product identification, as well as safe return of Double RBC Apheresis
the donor’s cells. There should be two separate forms of iden- In the 1990s, double units of red blood cells were collected
tification so that both the donor and the phlebotomist can using the apheresis equipment. The FDA finalized the guid-
verify the ID. Full names, signatures, unique numbers, or ance recommendations for this procedure in 2001. Double
pictures may all be used. RBC apheresis can be used to collect either allogeneic or au-
tologous units. The donor must meet the requirements for
Leukapheresis
whole blood donation and the recommendations established
Apheresis is the only effective method for collecting leuko- by the equipment manufacturer. The volume of red blood
cytes or, more specifically, granulocytes. The therapeutic cells removed from the donor should not yield a donor
effectiveness of granulocyte transfusions is still somewhat hematocrit of less than 30% or a hemoglobin of less than
controversial, but it has been shown to be effective in specific 10 g/dL after volume replacement.2
cases. A typical therapeutic dose is at least 1 × 1010 granulo- Donors participating in double RBC apheresis programs
cytes each day for 5 consecutive days.19 are deferred for 16 weeks following successful completion
In order to collect a large enough volume of leukocytes of the donation procedures. They should not participate in
(more than 1 × 1011 granulocytes), the donor must be given platelet or plasma apheresis during that period. If the pro-
certain drugs or sedimenting agents, and specific informed cedure is discontinued prior to completion and the total
consent must be obtained from the donor prior to adminis- RBC loss is less than 200 mL, the donor can donate again
tering these drugs. AABB Standards states that any of these within 8 weeks, provided all other donation criteria are met.
drugs or agents used to facilitate leukapheresis will not be If the RBC loss is greater than 200 mL but less than 300 mL,
used on donors whose medical history suggests that such a the donor should be deferred for 8 weeks. If the total RBCs
drug will exacerbate previous disease. lost is greater than 300 mL, the donor must be deferred for
One of the drugs typically given is hydroxyethyl starch the full 16 weeks.19 For more detail on apheresis, refer to
(HES), which is a common sedimenting agent. It enhances Chapter 18.
the separation of the white blood cells from the red blood
cells during centrifugation, which increases the amount of
Whole Blood Collection
leukocytes collected and decreases the amount of RBC
contamination in the final product. The disadvantage is Once the donor has satisfied requirements of the screening
that HES is a colloid; it expands the donor’s blood volume process and has been registered, whole blood collection can
and remains in the circulation for extended periods of proceed. This procedure must be performed only by trained
time. As a result, caution must be taken to control the personnel working under the direction of a qualified licensed
amount of HES given to a donor and its accumulation in physician. This section describes donor identification, asep-
the donor’s circulation. tic technique, venipuncture, collection of pilot tubes and
Corticosteroids such as prednisone or dexamethasone can whole blood unit, postdonation instructions, and adverse
also be used. These drugs are given to the donor prior to the donor reactions.
collection procedure. They work by pulling the granulocytes
from the marginal pool into the general circulation, thus Donor Identification
increasing the supply of cells available for collection. Careful
scrutiny must be used to obtain an accurate health history A numeric or alphanumeric system is used to link the donor
on the donor to identify any medical condition that could to the donor record, pilot tubes, blood container, and all
be exaggerated by the presence of corticosteroids. components made from the original collection. Care must
Finally, the newest agents are growth factors. Now that be taken to avoid duplicate numbers, voided numbers, or
recombinant hematopoietic growth factors are available, they other mistakes in the labeling process. These issues must be
are being used in leukapheresis procedures. The advantages investigated and kept on record.
6888_Ch13_281-306 29/10/18 5:43 PM Page 298

298 PART III Transfusion Practices

AABB Standards require that the trained phlebotomist


must review the donor record and ensure that the donor BOX 13–7
name and identification numbers match.2 The phlebotomist Whole Blood Collection Procedure
should ask the donor to state or spell his or her name. At
1. Confirm donor identity, and make the donor as comfortable as
this time, the phlebotomist can attach all labels to blood possible.
bags, donor record, and pilot tubes. 2. Using a tourniquet or blood pressure cuff, select a large, firm
vein in the antecubital space that is free of any skin lesions or
Aseptic Technique scarring. Inspect both arms.
3. Prepare the site using an FDA-approved cleansing method.
For blood collection, most blood centers use an iodine com- When finished, cover site with sterile gauze.
pound such as PVP-iodine or polymer iodine complex. 4. Inspect the blood bags for any defects or discoloration.
Using a tourniquet or blood pressure cuff, the venipuncture 5. Ensure the balance system is adjusted to the volume being
site is identified, and the area is scrubbed at least 4 cm in all drawn; ensure counterbalance is level. Place hemostats on the
directions from the site for a minimum of 30 seconds. The tubing to prevent air from entering the line.
area is then covered with a dry sterile gauze pad until the 6. Reapply tourniquet or blood pressure cuff (40 to 60 mm Hg) to
venipuncture is performed. Donors who are allergic or sen- increase distention of the vein.
sitive to iodine compounds may use chlorhexidine gluconate 7. Uncover the sterile gauze, and perform the venipuncture immedi-
and isopropyl alcohol. All methods must be approved by the ately. Check the position of the needle, and tape the tubing to the
donor’s arm to hold the needle in place. Cover with a sterile gauze.
FDA (Box 13–6).
8. Release the hemostat, and ask the donor to open and close the
hand every 10 to 12 seconds during collection procedure.
Collection Procedure
9. Reduce the pressure on the cuff to approximately 40 mm Hg.
The collection procedure for whole blood is outlined in 10. Continue to monitor the patient throughout the entire collection
Box 13–7. process. The donor should never be left unattended. Mix blood
and anticoagulant periodically during the procedure (e.g., every
45 seconds).
11. When the primary bag has tripped the scale, the donor can stop
BOX 13–6 squeezing and tubing can be clamped. A unit containing a vol-
ume of 405 to 550 mL should weigh between 429 and 583 g, plus
Arm Preparation Methods the weight of the container and anticoagulant. The conversion
1.06 g/mL is used to convert grams to milliliters. If the volume
Method I collected is in the low volume range (300 to 404 mL in a 450-mL
1. Scrub the site (2 × 2 inches) for 30 seconds using an aqueous collection or 333 to 449 mL in a 500-mL collection), the unit must
iodophor scrub solution (0.7%). Iodophor is a polyvinyl be labeled as a “low volume unit,” and fresh-frozen plasma (FFP)
pyrrolidone-iodine or poloxamer iodine complex. cannot be made from this unit as it would not contain adequate
2. Apply iodophor complex and let stand for 30 seconds. Use a con- levels of coagulation factors.
centric spiral motion, starting in the center and moving outward. 12. Before the needle is removed from the donor’s arm, pilot tubes
Do not go back to the center. Removal of the iodophor solution is are filled. The pressure is reduced to 20 mm Hg or less, and de-
not necessary. pending on the tubing of the bag, the tubes are filled.
3. Site is now ready for venipuncture. Cover with sterile gauze until • In-line needle. A hemostat or metal clip is used to seal tubing
ready for needle insertion. distal to the needle. The connector is opened, the needle is in-
serted into the pilot tube, the hemostat is removed, and tubes
4. If the donor bends the arm, or the prepared site is touched with
are filled. The donor needle can now be removed.
the fingers or other nonsterile object, the arm preparation must
• Straight-tubing assembly. Place hemostats approximately four
be repeated.
segments from the needle. Tighten the loose knot made previ-
Method II ously in the tubing; release the hemostats, and strip a segment
of tubing between knot and needle. Secure hemostat and cut
1. Apply a minimum of 1 mL of One Step Gel directly to the
tubing in stripped area of segment. Fill required tubes by re-
venipuncture site by squeezing the gel bottle directly over the
leasing hemostats. This is an open system, and appropriate
intended area.
biohazard precautions should be followed. Reapply hemostats
2. Using a sterile applicator and while holding it at an approximate and remove needle from donor’s arm.
30° angle, begin scrubbing in a circular motion in about a 1-inch
13. Once the needle has been removed from the donor’s arm, apply
area directly over the venipuncture site. Continue this for a mini-
pressure over gauze, and ask the donor to raise his or her arm,
mum of 30 seconds.
continuing to exert pressure over the site. When the bleeding
3. After scrubbing for 30 seconds, use the same applicator to begin has stopped, the donor can lower his or her arm, and an appro-
at intended site of venipuncture, and move gradually outward in priate bandage can be applied.
concentric circles to form a total prepared area measuring at least
14. The needle assembly should be discarded into an appropriate
3 inches in diameter.
biohazard receptacle. The tubing should be stripped to allow
4. Using a second sterile applicator and starting from the center of proper mixing of anticoagulant/preservative with blood.
the 3-inch prepared area, remove excess gel by moving gradually
15. Heat-seal the filled tubing into segments, and apply an appropri-
outward in concentric circles.
ate identification label to one segment and detach from the
5. Allow site to air-dry according to manufacturer’s instructions. blood bag for storage. Place blood at the appropriate tempera-
6. If the donor bends the arm or the prepared site is touched with ture. Units in which platelets will be made must be maintained at
the fingers or with any other nonsterile object, the arm prepara- room temperature (20°C to 24°C) until the platelet concentrate
tion must be repeated. has been prepared; all others can be stored at 1°C to 6°C.
6888_Ch13_281-306 29/10/18 5:43 PM Page 299

Chapter 13 Donor Selection 299

Postdonation Instructions Severe

Most donor reactions will occur during or shortly after the A donor experiencing convulsions defines a severe reaction.
donation. It is recommended that donors remain in an area Convulsions can be caused by cerebral ischemia, marked
where they can be observed by the donation center staff and hyperventilation, or epilepsy. The former is associated with
be given instructions to follow for the next 24 hours. Most vasovagal syncope or reduced blood flow to the brain owing
blood centers have a designated postdonation area where to the shock symptoms, and hyperventilation is caused by
donors can sit and replenish their fluids. An example of post- marked depletion of carbon dioxide. The following should
donation instructions is shown in Figure 13–8. be followed by the donor room personnel:
1. Call for help immediately and notify the blood bank
Donor Reactions
physician
Most donors tolerate the withdrawal of a unit of blood with- 2. Try and restrain the donor to prevent injury to self or
out incident; however, in the event an adverse reaction does others
occur, the donor room staff must be well trained and able to 3. Ensure an adequate airway
react immediately to the donor’s needs. Donor reactions In the event of cardiac or respiratory difficulties, the
cover a wide spectrum, from nervousness and hematomas at donor room staff should perform CPR until medical help
the phlebotomy site to convulsions and loss of conscious- arrives.
ness. The donor staff should be trained in CPR. Reactions
can generally be divided into three categories: mild, moder- Hematomas
ate, and severe.
A hematoma is a localized collection of blood under the skin,
Mild resulting in a bluish discoloration. It is caused by the needle
going through the vein, with subsequent leakage of blood
Reactions in this category encompass one or more of the into the tissue. If a hematoma develops, the following
following: syncope or fainting, nausea or vomiting, hyper- instructions apply:
ventilation, twitching, and muscle spasm. Syncope may be
idiopathic or may be brought about by the sight of blood. 1. Remove the tourniquet and needle from donor’s arm
The donor may show signs of sweating, dizziness, pallor, or 2. Apply pressure with sterile gauze pads for 7 to 10 min-
convulsions. The following instructions apply for a donor utes, with the donor raising his or her arm above the
who has fainted: heart
3. Apply ice to the area for 5 minutes
1. Remove the tourniquet and withdraw needle
2. Place cold compresses on the donor’s forehead Donor Records
3. Raise the donor’s legs above the level of the head
4. Loosen tight clothing and secure airway Donor records must be retained by the blood collection
5. Monitor vital signs facility as mandated by the FDA and AABB. There must be a
system to ensure that the donor’s confidentiality is not com-
Donors who are extremely nervous may exhibit sudden promised and that donor records are not altered. There must
twitching or muscle spasms. If this happens, try to disengage be policies on record-keeping and storage, and the blood
the hyperventilation sequence by conversing with the donor bank staff should be well trained on these policies and pro-
and having the donor breathe into a paper bag, if necessary. It cedures. The minimum retention time for donor records
is not advised to give oxygen to these donors. If the donor starts varies from 5 to 10 years to indefinitely. Table 13–1 lists
to feel nauseated or vomits, the following instructions apply: minimum retention times for various donor records.
1. Instruct the donor to breathe slowly
2. Apply cold compresses to the forehead Donor Processing
3. Turn the donor’s head to one side and provide an appro-
The donor unit collected must be tested and processed by
priate receptacle
blood bank technologists before it can be made available for
4. The donor may be given water after vomiting has ceased
transfusion. The tests performed on donor blood include
those detailed in the following sections.
Moderate

A moderate reaction can include any of the reactions listed ABO/Rh


above in addition to loss of consciousness. The donor may
Testing for the donor’s ABO group must include both
have a decreased pulse rate, may hyperventilate, and may ex-
forward and reverse grouping. The ABO group must be de-
hibit a fall in systolic pressure to 60 mm Hg. The following
termined by testing the donor RBCs with anti-A and anti-B
instructions apply:
reagents and by testing the donor serum or plasma with
1. Check vital signs frequently reagent A1 cells and B cells. (Refer to Chapter 6, “The ABO
2. Administer 95% oxygen and 5% carbon dioxide Blood Group System.”)
6888_Ch13_281-306 29/10/18 5:43 PM Page 300

300 PART III Transfusion Practices

LifeSouth Community Blood Centers


1221 NW 13th Street Post Donation Recommendations
Gainesville, FL 32601

Date: Location: Branch:

Donor Recommendations
Please read the following instructions and sign at the bottom.

1. Contact the blood center:

• If any illness arises after your donation.

• If you develop a headache and fever of 101oF or higher within 14 days following your donation or if you become diagnosed with
West Nile Virus infection.

• If you have any questions about your donation or if you remember something about your medical or personal history that may
affect your blood donation.

2. Do not smoke for one-half hour.

3. Eat and drink something before leaving.

4. Do not leave until released by the donor technician.

5. Drink more fluids (nonalcoholic) than usual in the next four hours, especially fruit juices.

6. Leave the bandage on for a few hours, and then you may remove it. If there is any bleeding from the puncture site, raise arm, and
apply direct pressure.

7. If you feel faint or dizzy, either lie down or sit down with head between the knees.

8. Do not perform strenuous activities or engage in critical work where safety requires your maximum abilities.

9. If any symptoms persist, either return to blood center or see your doctor.

Signatures

Thank you for your blood donation.

Figure 13–8. Sample postdonation instructions.


6888_Ch13_281-306 29/10/18 5:43 PM Page 301

Chapter 13 Donor Selection 301

Table 13–1 Retention of Donor Records in the tube system, cells sensitized with IgG are added to all
negative tubes after the AHG phase. If the gel system or an
Retention automated instrument is used, the manufacturer’s guidelines
Donor Record Time (Years) must be followed (see Chapter 12, “Blood Bank Testing Tech-
Donor ABO/Rh 10 nologies and Automation”).
Donor antibody screen 10
HBV
Informed consent for donation 10
Approximately 240 million people are chronically infected
Medical director approval for donation interval 5 with HBV, necessitating sensitive testing methods for the ear-
Physical examination 10 liest detection of infection. In 2016, the FDA recommended
the implementation of HBV DNA to testing regimens already
Medical history information 10 in place, namely anti-HBc (IgG and IgM) and HBsAg, in
Identification number of donor unit 10 order to reduce the residual risk of HBV transmission from
donor to recipient. A donor’s blood may be used for transfu-
Viral marker testing results 10 sion if all three screening tests are nonreactive. If any one of
Quarantine of donor unit 10 the three assays are reactive, the donor’s blood must not be
used for transfusion.21 A donor who was repeatedly reactive
Repeat testing of donor blood 10 with anti-HBc on more than one occasion may be considered
ID of donor-processing tech 10 for reentry if after 8 weeks the HBsAg, anti-HBc, and HBV
NAT were negative.21
Plt count for frequent platelet apheresis 10

Sedimenting agent of leukapheresis 10 HCV

Notification of abnormal results 10/indefinite The hepatitis C virus (HCV) was identified in 1988 and was
initially referred to as non-A, non-B (NANB) hepatitis.
Modified from AABB Standards, American Association of Blood Banks, Bethesda, MD Screening tests for anti-HCV involve enzyme immunoassay
2009, p. 74.
(EIA) and chemiluminescent immunoassay (ChLIA) meth-
ods. In 1999, NAT for HCV RNA was introduced, and in
The donor’s Rh type should be determined by testing 2002 the tests were licensed by FDA and mandated for rou-
with anti-D reagent at the immediate spin phase. In the tine donor screening in addition to the EIA or ChLIA meth-
event the initial testing is negative, a test for weak D must ods. NAT is able to detect small amounts of viral nucleic acid
be performed. This involves a 37°C incubation and an an- in blood before antibodies or viral proteins such as HCV core
tihuman globulin (AHG) phase. If both the immediate spin antigen are detectable by current methods. Implementation
and weak-D test results are negative, label the donor as of HCV NAT effectively reduced the window period for
Rh-negative; however, if any part of the testing yields a detection of HCV by approximately 70%, from a mean of
positive test for D, the donor unit should be labeled as 82 days to 25 days.22
Rh-positive. (Refer to Chapter 7, “The Rh Blood Group Confirmatory methods include the RIBA (recombinant
System.”) Some automated testing systems do not require immunoblot assay) and HCV RNA. The test is reported as
an antiglobulin test phase for the detection of the weak-D positive, negative, or indeterminate. An individual who is
variants. positive by RIBA is considered to have the HCV antibody
and would be indefinitely deferred as a blood donor. Donors
Antibody Screen who repeatedly test reactive for HCV screening tests must be
deferred, and the components or products prepared are
AABB Standards requires that donors with a history of preg- discarded.
nancy or transfusion be tested for unexpected antibodies to In the reentry protocol for HCV, the FDA utilizes two
RBC antigens. Most blood centers choose to perform the an- groups of donors. One group is identified as being NAT
tibody screen test on all donors, not just those with a history reactive with a negative anti-HCV test and the second group
of pregnancy or transfusion. Unlike the antibody screen as NAT nonreactive or not done, anti-HCV repeat reactive,
performed on a recipient prior to transfusion, which uses and RIBA indeterminant, negative, or not done. After a
two or three individual reagent cells, the testing of donor follow-up of 6 months and using HCV NAT and anti-HCV,
units generally uses a pooled cell reagent. This reagent con- if the NAT results are reactive and anti-HCV repeat reactive,
tains two or three individual cells pooled into a single the donor is deferred permanently. Likewise, if the NAT is
reagent. The pooled screening cell reagent contains all of the reactive and anti-HCV negative, the donor is permanently
required antigens and is capable of detecting the presence of deferred. If the NAT is nonreactive and anti-HCV repeat
the clinically significant alloantibodies. (Refer to Chapter 10, reactive, the donor is deferred and followed up at a later
“Detection and Identification of Antibodies.”) A control time. Finally if the NAT is nonreactive and anti-HCV nega-
system must be in place for the method used. For example, tive, the donor may be reentered for a future donation.22
6888_Ch13_281-306 29/10/18 5:43 PM Page 302

302 PART III Transfusion Practices

HIV recombinant env protein rgp46II. There is no approved pro-


tocol for donor reentry involving HTLV-infected donors.
All donor units must be screened for the presence of the
human immunodeficiency virus (HIV-1/2) antibody using WNV
an FDA-approved method and HIV-1 RNA. If the initial
screening tests are negative, the unit is suitable for transfu- West Nile virus (WNV) is an arthropod-borne virus involv-
sion; if it is positive, the test must be repeated in duplicate. ing the natural reservoir host (birds) and the intermediate
If any one of the duplicate tests is reactive, the unit must host (mosquitos), which can infect a human. Humans are
be discarded as well as any in-date components from prior considered dead-end hosts because the level of viremia is
donations. Screening tests include EIA, ChLIA, and NAT. inadequate to infect a mosquito.24 WNV was first detected
Minipool (MP)-NAT is run using 16 to 24 donation samples in the United States in 1999 in New York City.25 Symptoms
per pool. If the pool is nonreactive all units may be released of disease include headache, myalgia, arthralgia, rash, or
for transfusion. If the pool is reactive, then units are tested gastrointestinal symptoms. Testing for WNV began in 2003,
individually using NAT. Any reactive units identified for and in 2009 the FDA published their final guidelines for
HIV-1 should undergo a confirmation test. Confirmation testing whole blood and component donor samples. It is rec-
tests for HIV include the Western blot and the immunoflu- ommended that units be tested year-round using either
orescence assay (IFA). Results are expressed as positive, aMP-NAT or individual donor (ID-NAT) method. MP-NAT
negative, or indeterminate. uses pools of 6 to 16 donor samples and is routinely used
For donor reentry, the FDA utilizes three possible groups. when risk of WNV infection in the geographical area is low,
Group 1 is NAT reactive, anti-HIV-1/2 negative, and HIV-1 for example, during the winter months. A switch to the
EIA negative. Group 2 is NAT nonreactive or not done, anti- ID-NAT format is recommended when the risk is high. This
HIV-1/2 repeat reactive, HIV-1 WB or IFA indeterminate, generally coincides with mosquito season during the sum-
unreadable, negative, or not done. Group 3 is NAT nonre- mer months. Each blood collection facility must define the
active or not done, anti-HIV-1/2 negative, HIV-1 EIA repeat trigger point and have established procedures to switch from
reactive, neutralization test positive or indeterminate. If a MP-NAT to ID-NAT testing and when to switch back again.
donor is classified as being in one of these groups, the donor If an MP is reactive, each sample in the pool should be
is followed up in 8 weeks and tested using HIV-1 NAT and retested individually. Those that remain nonreactive may be
an anti-HIV-1/2 test. The donor is permanently deferred if released for transfusion. Any individual units with reactive
the NAT is reactive and the anti-HIV-1/2 is repeat reactive test results should be discarded, and the donor should be
or the NAT is reactive and the anti-HIV-1/2 test is negative. deferred from further donations for 120 days. In addition, any
The donor is deferred and followed up if the NAT is nonre- currently in-date products from the same donor during the
active and the anti-HIV-1/2 is repeat reactive. The donor is previous 120 days should be retrieved and quarantined. The
reentered for future donation if the NAT is nonreactive and donor should be notified and counseled. Repeat testing with
the anti-HIV-1/2 is negative.22 the same or another individual NAT test and a test for WNV
antibodies is recommended.
HTLV-I/II
Syphilis
The HTLV-I virus, or human T-cell lymphotrophic virus
type I, is the causative agent of adult T-cell leukemia and has A serologic test for syphilis is required by both the AABB and
been associated with a neurological disorder called HTLV- the FDA. Screening tests include the rapid plasma reagin
associated myelopathy. HTLV-II has been shown to have (RPR) and the Venereal Disease Research Laboratory (VDRL)
about 60% homology23 with type I and is prevalent among test. Both tests are based on reagin, or antibody directed to-
intravenous drug users in the United States. Persons can con- ward cardiolipin particles. Cardiolipin-like antibodies have
tract both viruses from transfusion via infected lymphocytes. been documented in persons with untreated syphilis infec-
Screening for HTLV-I began in 1988; a combined HTLV-I/II tions. Antibody will agglutinate cardiolipin carbon particles
assay was approved 10 years later in 1998. Screening test in the form of visible flocculation.
methodologies include both EIA and ChLIA. In recipients of The confirmatory test for syphilis is the FTA-ABS or
blood infected by HTLV-I and II, 20% to 60% will develop fluorescent treponemal antibody absorption test. In this
infection. Myelopathy can occur in a person infected with test, indirect immunofluorescence is used to detect anti-
these viruses as a result of blood transfusion. bodies to the spirochete T. pallidum, the agent that causes
As with the other viral markers, a donation that is re- syphilis. If the STS or VDRL screening test is reactive or
peatedly reactive may not be used for transfusion. IFA and indeterminate, the components from that donation must
Western blot can be used as confirmatory tests; however, be discarded and the donor deferred unless a confirmatory
indeterminate results may hinder discrimination between FTA-ABS test is nonreactive. If the FTA-ABS test is non-
HTLV-I and HTLV-II. In 2014, the FDA approved the MP reactive, the donor may be reentered and the components
Diagnostics HTLV Blot 2.4 (MP Biomedicals, Santa Ana, labeled as reactive with the screening test. The donor of a
California), which can determine which virus type is confirmed reactive test may be reentered into the donor
present in donor plasma. It is an EIA that utilizes an HTLV-I pool after 12 months and documentation of completion
specific recombinant env protein rgp46-I and an HTLV-II of treatment. The FDA revised the guideline of testing of
6888_Ch13_281-306 29/10/18 5:43 PM Page 303

Chapter 13 Donor Selection 303

blood donors for syphilis in 2015. Blood centers must use transmission unless they can be screened by NAT or treated
an FDA licensed, approved, or cleared donor screening test with pathogen-reduction technology (PRT). The Cobas Zika
for potential blood donors stating that “it can no longer ID-NAT (Roche Diagnostics, Indianapolis, Indiana) test has
support a policy for the use of diagnostic syphilis tests for been employed in Puerto Rico to detect Zika viral RNA in
use as a donor screening test” enforcing the requirements plasma specimens from blood donors.26
of 21 CFR 1271.80.9 Ebola is caused by infection of the Ebola virus, which is
The spirochete that causes syphilis, T. pallidum, cannot part of the Filoviridae family. The virus was first described
survive more than 72 hours in citrated blood stored at in 1976 near the Ebola River in the Congo. Transmission
1°C to 6°C, which would make platelets the only component occurs via exposure to blood or body fluids, contaminated
capable of transmitting infection. A serologic test for syphilis needles, and infected primates. The incubation period ranges
is also done because the disease is characterized as being from 2 to 21 days. Symptoms include fever, headache,
sexually transmitted and places the donor at higher risk for muscle and abdominal pain, fatigue, diarrhea, vomiting, and
possible exposure to hepatitis and HIV. unexplained hemorrhaging. Like Zika, there is no FDA-
approved vaccine to prevent Ebola. In 2014, an outbreak
T. cruzi (Chagas Disease) occurred in West Africa with nearly 29,000 suspected cases
and 11,325 related deaths. The CDC has characterized Ebola
Chagas disease is a parasitic infection that is endemic to as a Category A bioterrorism agent/disease. The FDA has rec-
Mexico and Central and South America. The infection is ommended the DHQ should be updated in assessing donors
generally mild, but it persists, without symptoms, through- having a history of Ebola infection or disease, as well as a
out the infected individual’s life and can be transmitted history of residence or travel in the past 8 weeks to a country
through blood transfusions. The FDA first recommended associated with widespread transmission or in urban areas
testing donors for antibodies to T. cruzi in 1989, but with uncertain control mechanisms. In addition, the DHQ
it wasn’t until December of 2007 that an EIA test was should include questions assessing a history of close contact
licensed. In 2007, some donor centers began testing, and in the past 8 weeks with an individual confirmed to have
in 2010 the FDA released their final recommendations for Ebola infection or disease or an individual who is suspected
donor screening.14 as having Ebola. Close contact would include health-care
All donors (allogeneic and autologous) are to be tested workers and individuals with a history of sexual contact in
once. If the results are nonreactive, the donor need not be the past 8 weeks with a person known to have recovered
retested with each donation. This was reasoned because most from Ebola. Regarding deferral periods, the FDA recom-
donors who are infected in the United States have a chronic mends an indefinite deferral for a donor with a history of
infection that was acquired when residing in a country where Ebola virus infection or disease as well as an 8-week deferral
the disease is endemic. Repeat reactive donors must be from the date of his or her departure from an area with wide-
deferred indefinitely and the products quarantined and spread transmission of Ebola virus, be it residence or travel.27
destroyed. No approved confirmatory tests are available,
so there are no mechanisms for reentry for deferred donors.
The donor should be notified and counseled, and a “look-
CASE STUDIES
back” procedure should be performed, looking back to all
donations from that donor for the previous 10 years. If the Case Study 13-1
donor had been previously tested with a licensed test, the
look-back should include 12 months from the last nonreac- A 79-year-old woman was admitted to a community
tive test. hospital for sepsis/anemia in January 2010. Transfusion
history included 2 units of leukocyte-reduced red blood
Zika and Ebola cells (LR RBC) on August 4, 2009; 6 units of LR RBCs
in December, 2009; and 2 units of LR RBCs in January,
Zika is an RNA virus and spread by the bite of an infected 2010. Parasitemia was observed on her peripheral blood
Aedes mosquito, namely A. aegypti and A. albopictus. It is part smear on January 28, 2010. Serology confirmed the
of the Flaviviridae family. Currently, there is no vaccine for parasitemia was due to Babesia with a titer greater
Zika. The virus can also be transmitted via blood transfusion, than 1024. The woman resided in the northeast region
from mother to fetus, and by sexual intercourse. Symptoms of the United States.
include fever, rash, headache, joint and muscle pain, and
redness of eyes. Zika has been linked to microcephaly and 1. Is the transfusion history consistent with the incuba-
Guillain-Barre syndrome. Regarding blood donation, the tion period for B. microti?
FDA recommends potential donors who have been infected 2. What symptoms are associated with Babesia infection?
with Zika not donate blood for 4 weeks. This includes those The blood supplier conducted a look-back study to
who have demonstrated symptoms or had sexual contact identify if any of the donors had risk factors associated
with people who have traveled to countries associated with with Babesia or had serologic or symptomatic evidence of
transmission. Moreover, no blood should be used from areas infection. It was determined four of the donors had a
where there is active viral transmission. Blood collections negative titer for B. microti using IFA, one was not tested,
should cease in areas of the United States with active Zika
6888_Ch13_281-306 29/10/18 5:43 PM Page 304

304 PART III Transfusion Practices

and one donor had an IgG titer of 1024 for B. microti. The was 102.9°F, and there was noticeable fluid in his right
blood center permanently deferred the donor with the eye. Vital signs and oxygen saturation were normal, as
titer of 1024. were examinations of his lungs, heart, and kidneys. A
CBC showed 4,900/µL leukocytes, hemoglobin 12 g/dL,
3. Is the blood center’s action in line with current re-
hematocrit 37%, platelet count 255,000/µL. The WBC
quirements for deferral?
differential showed 61% neutrophils, 34% lymphocytes
4. Should there be a deferral for the donor that was not
with moderate atypical forms, and 5% monocytes. Trypo-
tested?
mastigotes of T. cruzi were found on his peripheral blood
Case Study 13-2 smear. An ELISA test revealed a T. cruzi antibody titer
of 128. The patient was placed on nifurtimox therapy for
A 22-year-old Caucasian male from Utah traveled to 3 months.
Honduras as part of his 1-year mission with the Church of
1. Upon returning to the United States in 11 months,
Latter Day Saints. A month after arriving in Tegucigalpa,
could this patient donate whole blood?
he complained of swelling in his right eye, severe headache,
2. What disease is caused by T. cruzi?
fever, and malaise. Upon admission to the ER. his fever

SUMMARY CHART
 An allogeneic blood donor should weigh at least 110 lbs  Predeposit autologous donation refers to blood for the
(50 kg). donor-patient that is drawn before an anticipated
 The minimum hemoglobin for a female whole blood transfusion for surgery and stored until used.
donor is 12.5 g/dL.  An autologous donor must have a hemoglobin of at
 The minimum hemoglobin for a male whole blood least 11.0 g/dL and a hematocrit of at least 33%.
donor is 13.0 g/dL.  A person who has received a nonhuman animal trans-
 Persons who have had a blood transfusion are deferred plant of cells or tissues is permanently deferred from
for 12 months owing to risk of exposure to hepatitis, donating whole blood.
HIV, or other viral diseases.  Males who have had sex with another male (MSM) in
 The interval between whole blood donations is the past 12 months are deferred for 1 year.
8 weeks or 56 days.  Acute normovolemic hemodilution takes place in the
 The interval between plasmapheresis, plateletpheresis, operating room when 1 to 3 units of whole blood are
or leukapheresis is at least 2 days. collected and the patient’s volume is replaced with
 Attenuated live viral vaccines such as smallpox, colloid or crystalloid, then reinfused during surgical
measles, mumps, yellow fever, and influenza carry a procedure, starting with the first unit collected.
2-week deferral.
 A blood donor that has a positive serologic test for
syphilis is deferred for 12 months.

2. Which of the following would be cause for deferral for a


Review Questions
male donor?
1. Which of the following information is not required for a. Temperature of 99.2°F
whole blood donation? b. Hematocrit of 37%
c. Spent 2 weeks in the United Kingdom in 1998
a.Name
d. Weighs 80 kg
b.Address
e. Received a blood transfusion 2 years ago
c.Transfusion history
d.Sex
e.Date of Birth
6888_Ch13_281-306 29/10/18 5:43 PM Page 305

Chapter 13 Donor Selection 305

3. Which of the following would be cause for a permanent 11. Which of the following donors would be rejected for
deferral? whole blood donation?
a. Received a dura mater graft 9 months ago a. A male who had sex with another male in 1988
b. Received hepatitis B immune globulin b. A female who had sex with a male in 1992
c. Is currently on warfarin c. A male who had sex with another male last month
d. Diagnosis of babesiosis d. A female who had sex with a male 9 months ago
e. Traveled to Senegal 2 years ago
12. What does “infrequent” refer to when talking about a
4. Immunization for rubella would result in a temporary plasmapheresis program?
deferral for: a. Donating no more frequently than once every 4 weeks
a. 4 weeks. b. Donating once a year
b. 8 weeks. c. Donating once every 6 months
c. 6 months. d. Donating no more frequently than once every 8 weeks
d. 1 year.
13. A patient is having an exploratory laparotomy performed
e. 3 years.
and donated blood for use in the patient’s upcoming
5. Which of the following donors is acceptable? surgery. Three units were collected, with the last unit
a. Donor who had a first-trimester abortion 4 weeks ago collected 2 days before surgery. Given this information,
b. Donor whose husband is a hemophiliac who regu- can the patient undergo surgery as planned?
larly received cryoprecipitate before 1989 a. Yes
c. Donor who was treated for gonorrhea 6 months ago b. No
d. Donor who had a needle-stick injury 10 months ago
14. Which of the following refers to a temporary deferral?
6. Which of the following tests is not required as part of a. Donor received varicella zoster live attenuated vaccine
the donor-processing procedure for allogeneic donation? b. Donor had a confirmed positive test for HBsAg
a. ABO c. Donor has a history of CJD
b. Rh d. Donor was diagnosed with babesiosis
c. STS
15. Which of the following carries a 12-month deferral?
d. Anti-HTLV-I
e. Anti-CMV a. Donor received Hepatitis B immune globulin
b. Donor received pituitary growth hormone from
7. How long must a 2-unit RBC donor wait before donating another human
red blood cells again? c. Donor received the MMR vaccine
a. 8 weeks d. Donor spent 10 years in Africa
b. 16 weeks
c. 6 months References
d. 12 months
1. Food and Drug Administration. Guidance for industry. Revised
8. What is the deferral period for Plavix? recommendations for reducing the risk of human immunodefi-
a. 14 days after last dose ciency virus transmission by blood and blood products.
Rockville (MD): US Health and Human Services CBER; 2015.
b. 1 month after last dose 2. AABB. Standards for blood banks and transfusion services.
c. 12 months after last dose 31th ed. Bethesda (MD): AABB; 2018.
d. 48 hours after last dose 3. Food and Drug Administration. Guidance for industry. Recom-
mendations for deferral of donors and quarantine and retrieval
9. All of the following records must be kept for 10 years, of blood and blood products in recent recipients of smallpox
except: vaccine (vaccinia virus) and certain contacts of smallpox vaccine
a. Unique ID of each unit. recipients. Rockville (MD): US Health and Human Services
CBER; 2002.
b. Donor consent. 4. AABB. AABB on quarantine release errors [Internet]. White
c. Request for blood or blood component. paper. Bethesda (MD): AABB; 2013. Available from: http://
d. A signed statement from requesting physician for www.aabb.org/research/whitepapers/documents/quarantine-
emergency release. release-errors-white-paper.pdf#search=qre.
5. Advisory Committee on Blood and Tissue Safety and Availability.
10. What is the causative agent of Chagas disease? Results from the Uniform Donor History Questionnaire
a. Trypanosoma cruzi [Internet]. Washington (DC): Office of the HIVAIDS an Infec-
tious Disease Policy; 2013. http://www.hhs.gov/bloodsafety/
b. Yersinia pestis advisorycommittee/acbtsa_201312meeting_agenda.html.
c. Treponema pallidum 6. Custer B, Kessler D, Vahidnia F, Leparc G, Krysztof DE, Shaz B,
d. Plasmodium falciparum et al. NHLBI Retrovirus Epidemiology Donor study II (REDS II),
6888_Ch13_281-306 29/10/18 5:43 PM Page 306

306 PART III Transfusion Practices

risk factors for retrovirus and hepatitis virus infections in 16. Food and Drug Administration. Guidance for industry. Recom-
accepted blood donors. Transfusion. 2014, doi: 10.1111/trf mendations for management of donors at increased risk for
.12951. human immunodeficiency virus type 1 (HIV-1) group O infec-
7. Advisory Committee on Blood and Tissue Safety and Availability. tion. Rockville (MD): US Health and Human Services CBER;
NHLBI Recipient Epidemiology and Donor Study III (REDS III). 2010.
Noncompliance with the men who have sex with men (MSM) 17. Jabbour N. Transfusion free medicine and surgery. Oxford:
deferral among U.S. male blood donors, Blood Donation Rules Blackwell Publishing; 2005, p. 91.
Opinion Study (BloodDROPs); 2014. Web cast. Available from: 18. Kreimeier U, Messmer K. Perioperative hemodilution. Transfus
http://webcast.nccsite.com/nih/2016. Apher Sci. 2002;27(1):59-72.
8. Seed CR, Kiely P, Law M, Keller AJ. No evidence of a significantly 19. Fung, MK, Eder AF, Spitalnik SL, Westhoff CM (eds). Technical
increased risk of transfusion-transmitted human immunodefi- manual. 19th ed. Bethesda (MD): AABB;2017.
ciency virus infection in Australia subsequent to implementing 20. Food and Drug Administration. Guidelines for the collection
a 12 month deferral for men who have had sex with men. Trans- of platelets. Rockville (MD): US Health and Human Services
fusion. 2010;50:2722-30. CBER; 1988.
9. Food and Drug Administration. Guidance for industry. Use of 21. Food and Drug Administration. Guidance for industry. Use of
donor screening tests to test donors of human cells and nucleic acid tests to reduce the risk of transmission of hepatitis
tissues—cellular and tissue based products for infection with b virus from donors of human cells, tissues, and cellular and
Treponema pallidum (Syphilis). Rockville (MD): US Health and tissue-based products. Rockville (MD): US Health and Human
Human Services CBER; 2015. Services CBER; 2016.
10. Food and Drug Administration. Guidance for industry. Revised 22. Food and Drug Administration. Guidance for industry. Nucleic
preventative measures to reduce the possible risk of transmis- acid testing (NAT) for human immunodeficiency virus type 1
sion of CJD and vCJD by blood and blood products. Rockville (HIV-1) and hepatitis c virus (HCV): testing, product disposi-
(MD): US Health and Human Services CBER; 2016. tion, and donor deferral and reentry. Rockville (MD): US
11. Concha-Marambio L, Pritzkow S, Moda F, Ironside JW, Schulz Health and Human Services CBER; 2010.
PE, Soto C. Detection of prions in blood from patients with 23. Food and Drug Administration Guidance for industry. Donor
variant Creutzfeldt-Jakob disease. Sci Transl Med. 2016; screening for antibodies to HTLV-II. Rockville (MD): US Health
8(370):183. and Human Services CBER; 1997.
12. Cohen C, Corazza F, De Mol P, Brasseur D. Leishsmaniasis 24. Food and Drug Administration. Guidance for industry. Use of
acquired in Belgium. Lancet. 1991;338:128. nucleic acid tests to reduce the risk of transmission of West
13. U.S. Food and Drug Administration. Vaccines, Blood & Biologics Nile Virus from donors of human cells, tissues, and cellular
[Internet]. Rockville (MD): FDA. Donating Blood Questions and and tissue-based products (HCT/P). Rockville (MD): US Health
Answers. Reasons for other deferral explained. Available from: and Human Services CBER; 2013.
Fda.gov/biologicsbloodvaccines/bloodbloodproducts/questions 25. Nash D, Mostashari F, Fine A, Miller J, O’Leary D, Murray K
aboutblood/donatingblood et al. The outbreak of West Nile virus infection in New York
14. Food and Drug Administration. Guidance for industry. Use City area in 1999. N Engl J Med. 2001;344:1807-14.
of serological tests to reduce the risk of transmission of 26. Chevalier MS, Biggerstaff BJ, Basavaraju SV, et al. Use of donor
Trypanosoma cruzi infection in whole blood and blood compo- screening data to estimate Zika virus incidence, Puerto Rico,
nents intended for transfusion. Rockville (MD): US Health and April-August 2016. Emerg Inf Dis. 2017;23(5):790-5.
Human Services CBER; 2010. 27. Food and Drug Administration. Guidance recommendations
15. Carnevale J, Feller R, Shalvoy M. Transfusion-transmitted for assessment of blood donor eligibility, donor deferral and
babesiosis during total hip arthroplasty. Int Orthop. 2015; blood product management in response to Ebola virus.
38(9):852-5. Rockville (MD): US Health and Human Services CBER; 2017.
6888_Ch14_307-332 29/10/18 5:43 PM Page 307

Chapter 14
Transfusion-Transmitted Diseases
JoAnn Christensen, MS, MT(ASCP)SBB and Elizabeth F. Williams, MHS,
MT(ASCP)SBB

Introduction Zika Virus Transfusion-Associated Parasites


Donor Testing Profile Babesia microti
Transfusion-Associated Hepatitis Other Viruses Trypanosoma cruzi
Hepatitis A Cytomegalovirus Malaria (Plasmodium Species)
Hepatitis B Epstein-Barr Virus Prion Disease
Hepatitis C Parvovirus B19 Creutzfeldt-Jakob Disease
Hepatitis D Human Herpesvirus 6 and Human Pathogen Inactivation
Hepatitis E Herpesvirus 8 Plasma Derivatives
Hepatitis G Virus Emerging Viruses Cellular Components
HIV Types 1 and 2 Chikungunya Virus Quarantine and Recipient Tracing
Profile Dengue Virus (DENV) (Look-Back)
Human T-Cell Lymphotropic Viruses Ebola Virus Summary Chart
Types I/II (HTLV-I/II) Bacterial Contamination Review Questions
Profile Overview References
West Nile Virus Syphilis
Profile Tick-Borne Bacterial Agents

OBJECTIVES
1. Describe the pathology, epidemiology, laboratory testing, and prophylaxis/treatment of the following diseases: hepatitis A
through G, HIV 1 and 2, human T-cell lymphotropic viruses I and II, and West Nile virus (WNV).
2. Explain the implications of the following diseases for blood transfusions: Epstein-Barr virus, cytomegalovirus (CMV),
parvovirus B19, herpesvirus 6 and 8, dengue virus, chikungunya virus, Zika virus, Ebola virus, general bacterial contamination,
syphilis, Babesia microti, Trypanosoma cruzi, malaria (Plasmodium species), Leishmania donovani, and Creutzfeldt-Jakob
disease and variant Creutzfeldt-Jakob disease.
3. Describe procedures for look-back and recipient follow-up.
4. Describe pathogen inactivation for plasma and cellular components.

Introduction Donor Testing


Blood is a lifesaving resource. In the United States, blood com- Once a donor passes the medical screen and donor question-
ponents are subjected to rigorous testing that makes them naire, required serologic testing is performed for hepatitis B
extremely safe and renders the likelihood of a transfusion- surface antigen (HBsAg), antibody to hepatitis B core antigen
transmitted disease (TTD) very small. However, bacterial, (anti-HBc), antibody to hepatitis C virus (anti-HCV), anti-
viral, parasitic, and prion pathogens constantly evolve, and bodies to human immunodeficiency virus (anti-HIV 1/2),
if not detected in the testing process, can cause harm and antibody to human T-cell lymphotropic virus types I and II
even death. (anti-HTLV-I/II), and syphilis. Each donor must also be

307
6888_Ch14_307-332 29/10/18 5:43 PM Page 308

308 PART III Transfusion Practices

tested at least once for antibodies to Trypanosoma cruzi. Re- hepatitis viruses.2 The hepatitis viruses affect the liver as the
quired nucleic acid testing (NAT) is performed for: HBV primary clinical manifestation. Hepatitis viruses can be
DNA, HIV-I RNA, HCV RNA, WNV RNA, and ZIKV RNA transmitted through the fecal-oral route or parenterally
(Table 14–1).1,2 (through contact with blood and other body fluids).
As current test methods are extremely sensitive, confir- Hepatitis A virus (HAV) and hepatitis E virus (HEV) are
matory tests are used to detect false-positives. These tests, mainly transmitted through the fecal/oral route. Hepatitis B
which vary by the disease, include polymerase chain reaction virus (HBV), hepatitis C virus (HCV), hepatitis D virus
(PCR), Western blot (WB), and radioimmunoprecipitation (HDV), and hepatitis G virus (GBV-C or HGV) are primarily
assay (RIPA). transmitted parenterally.2
Surrogate markers (alanine aminotransferase [ALT] and
anti-HBc) were used in the past to detect non-A, non-B hepa- Hepatitis A
titis. Due to the sensitivity of the current testing for HCV, ALT
is no longer required; however, blood centers are continuing The hepatitis A virus (HAV) belongs to the Picornaviridae
to perform both tests. ALT testing has continued because the family of viruses and is a small, nonenveloped, single-
European Union requires it for recovered plasma. The Food stranded RNA enterovirus.
and Drug Administration (FDA) recommends performance of
Clinical Manifestations and Pathology
anti-HBc as an additional screen for HBV.2
Many other organisms may be transfusion transmitted; Symptoms, if they occur at all, generally appear abruptly
however, tests for them are not required in the blood screen- and last fewer than 2 months, but may persist for as long
ing process. These include other viruses such as Epstein- as 6 months in some individuals.3 They may include nau-
Barr virus (EBV), CMV, parvovirus B19 (B19), Ebola virus, sea, vomiting, anorexia, fatigue, fever, jaundice, dark urine,
dengue virus, chikungunya virus, bacteria (now considered and abdominal discomfort. Less than 10% of children
to be the leading cause of death from transfusion),2 parasites under 6 years will develop jaundice. Jaundice is more com-
such as Babesia microti, malaria, Leishmania donovani, and mon in older children and adults, occurring in 40% to 50%
prion diseases. of children between 6 and 14 years and 70% to 80% of
individuals older than 14 years.4 The disease is rarely fatal.
Transfusion-Associated Hepatitis Fulminant, cholestatic, or relapsing hepatitis may occur.
Symptoms usually resolve within 3 weeks and are generally
Hepatitis is a generic term describing inflammation of self-limiting.5 In rare cases, a patient may develop fulmi-
the liver. Symptoms typically include jaundice, dark urine, nant liver failure.
hepatomegaly, anorexia, malaise, fever, nausea, abdominal
pain, and vomiting. The clinical symptoms of hepatitis range Epidemiology and Transmission
from being asymptomatic to death. Transmission is primarily through the fecal-oral route—
It can be caused by many things, including viruses, bac- spread through water, food, and person-to-person contact.
teria, noninfectious agents such as chemicals (including Poor hygiene and poor sanitation contribute to the spread
drugs and alcohol), ionizing radiation, and autoimmune of HAV. Because young children are generally asymptomatic,
processes.2 EBV, CMV, parvovirus, and herpes simplex virus the disease is predominantly spread from person to person
(HSV) can cause hepatitis as a complication, but because it within the household. Other individuals at risk are those
is not the primary disease, these viruses are not considered who travel to countries where HAV is endemic, are users of
illegal drugs, have clotting factor disorders, work with non-
human primates, and are men who have sex with men.5 A
Table 14–1 Disease Transmission risk factor cannot be identified in 46% of cases.6
Prevention—Required Tests The incubation period for HAV is 28 days on average, and
Diseases Required Tests the peak viremic period occurs 2 weeks before the onset of
the elevation of liver enzymes or the appearance of jaundice.5
Hepatitis B HBsAg, IgM, and IgG antibody to HBc, HBV DNA Transmission of HAV by clotting factor concentrates treated
Hepatitis C IgG antibody to HCV, HCV RNA
with solvent or detergent pathogen process has been re-
ported.7 Rates in the United States have dropped following
HIV IgM and IgG antibody to HIV-1/2, HIV-1 RNA introductions of the Hep A vaccine in 1995 and universal in-
HTLV IgG antibody to HTLV-I/II
fant vaccination in 2006. In 2014 there were a reported 1,239
acute new cases in the United States7 (Table 14–2). Out-
Syphilis IgG or IgM antibody to T. pallidum antigens breaks of acute hepatitis A involving men who have sex with
or nontreponemal serologic test for syphilis men have been reported in European countries.6
West Nile virus WNV RNA
Laboratory Diagnosis
Trypanosoma cruzi IgG antibody to T. cruzi (one-time testing) Most of the virus is shed in the feces during the incubation
Zika virus ZIKV RNA period and declines to low levels by the onset of symptoms
(Fig. 14–1). The presence of IgM anti-HAV antibody is
6888_Ch14_307-332 29/10/18 5:43 PM Page 309

Chapter 14 Transfusion-Transmitted Diseases 309

Table 14–2 Disease Burden From Hepatitis A, B, and C in the United States
Hepatitis A Hepatitis B Hepatitis C
2013 2014 2015 2013 2014 2015 2013 2014 2015

Number of acute cases reported 1,781 1,239 1,390 3,050 2,791 3,370 2,138 2,194 2,436

Rate of infection per 100,000 population 0.6 0.4 0.4 1.0 0.9 1.1 0.7 0.7 0.8

Number of persons with chronic infections No chronic disease 850,000–2.2 million 2.7–3.9 million

Data from Centers for Disease Control (www.cdc.gov/hepatitis/Statistics/).

Markers in HAV Infection Clinical Manifestations and Pathology


The clinical picture of HBV infection is highly variable. Most
Icterus people with acute illness will recover with no liver damage,
Enzymes 15% to 25% of chronically infected persons develop liver dis-
ease, and an estimated 3,000 persons in the United States die
HAV from HBV-related illness per year.4 The individual may be
(Stool)
Anti-HAV completely asymptomatic or may present with typical signs
Exposure Anti-HAV
IgG of disease, including jaundice, dark urine, hepatomegaly,
IgM anorexia, malaise, fever, nausea, abdominal pain, and vom-
iting. For children younger than 5 years, fewer than 10%
5 20 30 show signs of jaundice and clinical illness. However, infec-
Days Years tions acquired at birth or between ages 1 to 5 years result in
Figure 14–1. Markers in acute HAV infection. The typical pattern of HAV infection a chronic infection 90% and 30% of the time, respectively.
includes early shedding of virus in the stool, appearance of IgM anti-HAV, and For those older than 5 years, up to 50% will have clinical
immunity on recovery. illness, but only 2% to 10% will develop a chronic infection.
Chronic infections are associated with cirrhosis and liver
cancer. Hepatitis B related mortality rate for 2014 was
required for diagnosis of hepatitis A. IgM antibodies are 0.5 deaths per 100,000 population.4
detectable at or prior to the onset of clinical illness and de-
Epidemiology and Transmission
cline in 3 to 6 months. IgG antibodies to HAV appear soon
after IgM and may persist for years after the infection.4 HBV is transmitted through exposure to bodily fluids con-
taining the virus from an infected individual. Concentrations
Prophylaxis and Treatment of the virus are at high levels in blood, serum, and wound
A vaccine to HAV was licensed in the United States in 1995. exudates; at moderate levels in semen, vaginal fluids, and
Use of the vaccine was initially recommended for at-risk saliva; and at low levels in urine, feces, sweat, tears, and
populations. In 2006 use of the vaccine was recommended breast milk. Transmission may be sexual, parenteral, or peri-
for all children ≥1 years of age as well as at-risk populations. natal.8 Percutaneous transmission may occur through needle
The number of cases has decreased 90.8% from 2000 stick (drug use, occupational hazard, acupuncture, tattooing,
(13,397 cases reported) to 2014 (1,239 cases reported).4 or body piercing), hemodialysis, human bite, transfusion
The vaccine is produced from inactivated HAV. It is believed of unscreened blood or blood products, or sharing razors.
that the risk is low for pregnant women and that no special Permucosal transmission can occur through sexual inter-
precautions should be taken for immunocompromised per- course or vertically from mother to infant (transplacental or
sons.5 Other prevention methods include improvement in through breast milk).8 According to the Surveillance for Viral
water purification, good hygiene, and improved sanitation. Hepatitis Report of 2014, higher rates of hepatitis B continue
Immune globulin can be used preexposure to protect those among adults, especially among males between the ages of
traveling to high HAV-endemic areas or postexposure to pre- 30 to 49 years.4
vent infection in those exposed within a family, after an out-
Laboratory Diagnosis
break at a day-care center, or from a common source of
exposure such as a restaurant. Immune globulin should be HBV consists of several proteins or antigens to which the
used postexposure within 2 weeks for maximum protection.7 body can make antibodies (Fig. 14–2). A surface antigen
protein, HBsAg, is on the outer envelope of the virus. It can
Hepatitis B also be found floating free in the plasma. Antibodies can be
produced to two proteins within the core: hepatitis B core
Hepatitis B virus (HBV) is a partially double-stranded circu- antigen (HBcAg) and hepatitis B envelope antigen (HBeAg).
lar DNA virus of the Hepadnaviridae family.4 Viral replication levels of these markers along with the
6888_Ch14_307-332 29/10/18 5:43 PM Page 310

310 PART III Transfusion Practices

Dane Core Coat has been diagnosed, family members should be tested. If un-
particle particle protein infected, they should be vaccinated. If infected, they should
(HBcAg) (HBsAg)
(HBeAg)
be treated.
Detergent
Hepatitis C
+
Treatment
28nm Hepatitis C virus (HCV) is a member of the Flaviviridae virus
42nm 200 x 22nm
family and is caused by a virus with an RNA genome.8
SDS Clinical Manifestations and Pathology
The incubation period of HCV is 2 to 26 weeks. The average
+ incubation period is 7 to 8 weeks, followed by seroconver-
sion occurring in 8 to 9 weeks.9 Most HCV cases are asymp-
DNA Polymerase
activity
tomatic. Some cases report nonspecific symptoms such as
anorexia, malaise, fatigue, or abdominal pain. Most sympto-
Figure 14–2. Diagram of the intact Dane particle (HB virion) as seen by electron matic cases are very mild. Of HCV-infected individuals,
microscopy. Detergent treatment disrupts the particle into a core particle and outer 75% to 85% become chronic carriers, 60% to 70% develop
coat protein, releasing DNA (double- and single-stranded) and DNA polymerase
chronic liver disease, 5% to 20% develop cirrhosis over a
activity.
period of 20 to 30 years, and 1% to 5% will die from cirrhosis
or liver cancer. Of the three types of hepatitis (A, B, C),
host’s production of IgM or IgG antibodies are all used to hepatitis C accounted for the highest rate of death due to
make an initial diagnosis and follow the course of infection hepatitis in 2014 (5.0 deaths per 100,000 population).4
(Table 14–3).5
HBV DNA is the first marker to appear and can be Epidemiology and Transmission
detected by polymerase chain reaction (PCR) testing before Because most HCV cases are asymptomatic, the worldwide
HBsAg reaches detectable levels. HBsAg is detectable 2 to incidence is unknown. There was a 2.6-fold increase of
12 weeks postexposure during the acute stage and becomes reported acute hepatitis C cases from 2010 (850 cases) to
undetectable in 12 to 20 weeks after development of anti- 2014 (2,194). It is estimated that there are 2.7 to 3.9 million
HBsAg (Fig. 14–3). If the patient develops chronic HBV, the chronic carriers in the United States.4 In 2014 the rate of
level of HBsAg remains high. HBsAg can be used to monitor reported acute cases was 0.7 per 100,000 population (see
the stages of HBV from the acute, active infection to recov- Table 14–2). The risk of post-transfusion HCV has declined
ery or a chronic infection. It is also used to screen donor dramatically since the introduction of testing.
blood.8 HCV can be transmitted percutaneously through needle
HBeAg appears after the HBsAg and, in recovering stick, hemodialysis, human bite, transplant or transfusion,
patients, disappears before HBsAg. In chronic patients, it or by acupuncture, tattooing, or body piercing. It can also
remains elevated. HBeAg is present during the time of active be transmitted permucosally through sexual intercourse,
replication of the virus and is considered a marker of high contact with an infected toothbrush or razor, or perina-
infectivity.9 tally. Hepatitis C is transmitted mainly by exposure to
HBcAg is present in the serum but is undetectable. How- contaminated blood, with IV drug use being the main
ever, IgM anti-HBc is the first antibody to appear, and it per- source of infection. Recent increased cases have been re-
sists for about 6 months. Appearance of this antibody ported from the HIV-positive men who have sex with men
indicates current or recent acute infection.9 population.4

Prophylaxis and Treatment Laboratory Diagnosis


The donor questionnaire is used to identify individuals at Diagnosis of HCV is difficult. Not only are symptoms so
risk for HBV infection. For those who are not eliminated by mild in acute cases as to make separation of acute HCV
that process, testing for HBsAg, anti-HBc, and HBV DNA is from chronic HCV difficult, but also separating HCV from
required.10 other forms of liver disease is not easy. Diagnosis depends
An HBV vaccine was licensed in 1981 and introduced in on biochemical changes suggestive of HCV, detection of
1982. Hepatitis vaccination programs will eliminate domestic HCV RNA or anti-HCV in serum, or a known exposure to
HBV transmission, and the increased vaccination of adults the virus.
with risk factors will help accelerate the progress toward Today, anti-HCV testing via enzyme immunoassay (EIA)
elimination.4 or chemiluminescent immunoassay (ChLIA) methodology
Hepatitis B immune globulin (HBIG) injections and the and HCV RNA testing are performed on all donor units. For
vaccine given soon after exposure or within 12 hours of supplemental confirmatory testing, the recombinant im-
birth, if the mother is infected, may prevent infection. Three munoblot assay (RIBA) is no longer available; however, blood
other treatments licensed by the FDA are interferon (IFN)- centers may obtain FDA approval to use an alternate testing
α-2b,11 lamivudine, and adefovir dipivoxil.12 Once a patient pathway, including line immunoblot8 (see Table 14–3).
6888_Ch14_307-332 29/10/18 5:43 PM Page 311

Chapter 14 Transfusion-Transmitted Diseases 311

Table 14–3 Molecular and Serologic Tests in the Diagnosis of Viral Hepatitis
Virus Test Reactivity Interpretation
Anti-HBc
HBV DNA HBsAg Total IgM Anti-HBs HBeAg* Anti-HBe

+ + +/– +/– – +/– – Early acute HBV infection/chronic carrier

+ + + + – + – Acute infection

+/– – + + – +/– +/– Early convalescent infection/possible


early chronic carrier

+/_ + + – – +/– +/– Chronic carrier

– – + – + – +/– Recovered infection


– – – – + – – Vaccination or recovered infection

– – + – – – – Recovered infection? False-positive?

+ – – – – – – Window period

HDV RNA HBsAG Anti-HBc Anti-HBs Anti-HDV

+ + + _ + Acute or chronic HDV infection

– – + + + Recovered infection

Anti-HCV Recombinant
Antigens

Screening
HCV RNA EIA 5-1-1 c100-3 c33c c22-3

+/– + Not available Possible acute or chronic HCV infection

– + – – – – False-positive

+/– + + + – – Possible false-positive (if RNA is


negative); possible acute infection
(if RNA is positive)†

+/– + – – + + Early acute or chronic infection (if RNA


is positive); false-positive or late recov-
ery (if RNA is negative)†

+ + + + + + Acute or chronic infection

– + +/– +/– + + Recovered HCV†

HAV RNA Anti-HAV

Total IgM
+ + + Acute HAV

– + – Recovered HAV/vaccinated

HEV RNA Anti-HEV

Total IgM Acute HEV

+ + +

+ + – Recovered HEV

Taken from AABB Technical Manual, 15th ed., pp. 670–671. Bethesda, Maryland, 2005, with permission.
HBsAg = hepatitis B surface antigen; anti-HBc = antibody to hepatitis B core antigen; anti-HBs = antibody to HBsAg; HBeAg = hepatitis B envelope antigen; anti-HDV = antibody to
hepatitis D virus; anti-HAV = antibody to hepatitis A virus; anti-HCV = antibody to hepatitis C virus; anti-HEV = antibody to hepatitis E virus
*Those with HBeAg are more infectious and likely to transmit vertically.
†Anti-5-1-1 and anti-c100-3 generally appear later than anti-c22-3 and anti-c33c during seroconversion and may disappear spontaneously during immunosuppression or after

successful antiviral therapy.


6888_Ch14_307-332 29/10/18 5:43 PM Page 312

312 PART III Transfusion Practices

HBsAg Anti-HBs Hepatitis E


HBeAg Anti-HBc IgM
Anti-HBc IgG
Anti-HBe
Hepatitis E (HEV) is a member of the Caliciviridae family of
nonenveloped RNA viruses. It is rare in the United States;
however, it is the leading cause of hepatitis in the United
LFT Kingdom.14 Recent studies have reported the rate of 1:9,500
SYMP donors in the United States are HEV RNA positive.14 As in
HAV, HEV is spread through the fecal-oral route, usually
through contaminated drinking water in developing coun-
tries. A carrier state does not develop after the acute, usually
self-limiting illness. Chronic HEV infection has been re-
ported in patients on immunosuppressive therapy following
solid organ transplantation.15

Clinical Manifestations and Pathology


Weeks after exposure Symptoms are the same as for any hepatitis. Generally, these
cases are short-lived but can be prolonged. HEV causes an
Figure 14–3. Markers in HBV infection.
acute, self-limiting hepatitis that may last from 1 to 4 weeks
in most people.15

Prophylaxis and Treatment Epidemiology and Transmission


Currently there is no HCV vaccine. Prevention consists of HEV genotypes (G) 1, 2, and 4 (usually occurring in devel-
worldwide screening of blood and blood products; de- oping countries) and HEV G3 (usually occurring in devel-
struction or sterilization of needles and surgical or dental oped countries) are responsible for acute, sporadic cases
instruments; universal precautions; and education about of infection that can be short-lived or prolonged. The fecal-
the risks. oral route is the most common form of transmission for G1
Optimal therapy for chronic HCV is now considered to and G2. Transmission route for G3 and G4 are foodborne.14
be pegylated IFN and ribavirin combination.13 In a small Most cases in the United States are G3 contracted through
study by Gerlach and colleagues,13 50% of patients with eating undercooked pork or venison.15 HEV G1, G2, and G4
acute HCV spontaneously and permanently cleared the virus do not progress to a chronic state. Cases of chronic HEV G3
within the first 3 to 4 months. Patients who were still viremic infection have been reported.14 HEV is a major cause of
at 3 months and were treated at that point had an 80% clear- hepatitis globally and is becoming a concern for transfusion
ance rate. However, most cases were not symptomatic and transmission as more cases of transfusion-associated infec-
therefore were not noticed and treated until the patient was tions are reported in developed as well as underdeveloped
in the chronic phase. Treatment with INF-α and ribavirin in countries. Some countries have introduced HEV screening
these chronic cases achieved only a 30% to 54% sustained for blood donations.14
viral clearance.13
Laboratory Diagnosis
Hepatitis D Both IgM and IgG antibody to HEV (anti-HEV) may occur
following HEV infection. The titer of IgM anti-HEV de-
Hepatitis D (HDV) is a defective, single-stranded RNA virus clines rapidly during early convalescence; IgG anti-HEV
that is found only in patients with HBV infection. It requires persists and appears to provide at least short-term protec-
HBsAg in order to synthesize an envelope protein and repli- tion against disease (see Table 14–3). Antibodies are usually
cate. It was previously called the delta antigen. If HBV and identified using highly sensitive enzyme immunoassays
HDV are contracted concurrently, this coinfection, as com- that are recombinant and synthetic HEV antigens.9 Nucleic
pared with HBV alone, appears to cause a more severe acute acid testing for HEV RNA is used by research labs but is
disease, with a higher risk of fulminant hepatitis (2% to 20%) not available for commercial use. There are also no FDA-
but a lesser risk of developing chronic hepatitis. approved serologic methods approved for diagnostic use in
Those at highest risk of infection are IV drug users. This the United States.15
infection can also be transmitted sexually. It is believed that
20 million people are infected worldwide, but the number Prophylaxis and Treatment
of new infections appears to be declining, most likely due to Meat such as pork, venison, and shellfish must be thor-
the implementation of the Hep B vaccine.8 oughly cooked and water supplies must be cleaned and
HDV is detected by testing for IgM or IgG anti-HDV or sewage disposal handled properly to prevent HEV infection.
HDAg and HDV RNA in the serum. Tests for HDV are not Currently, no commercially available vaccine exists for the
required for blood donations. If a donor has HBV, the unit will prevention of hepatitis E. Ribavirin has been shown to be
not be used for transfusion. As HDV cannot exist without effective for treatment of chronic HEV, but its effectiveness
HBV, testing for HBV will eliminate any infections with HDV. for treatment of acute infection is not clear.14
6888_Ch14_307-332 29/10/18 5:43 PM Page 313

Chapter 14 Transfusion-Transmitted Diseases 313

Hepatitis G Virus HIV is a retrovirus that is spherical in shape, with an ap-


proximate diameter of 100 nm. It consists of an envelope of
GB virus C (GBV-C) and hepatitis G virus (HGV) are two glycoproteins, core proteins, and an inner core of viral RNA
genotypes of the same enveloped RNA virus that belongs to and reverse transcriptase. Infection with the virus causes a
the Flaviviridae family. Approximately 1% to 2% of U.S. slowly progressing immune disorder. The causative viruses,
donors have tested positive for HGV, making this virus more HIV-1 and HIV-2, are similar in structure, varying primarily
common than HCV.16 However, recent reports do not impli- in the envelope proteins (Fig. 14–4 and Table 14–4). Almost
cate GBV-C/HGV as a cause of hepatitis. all cases in the United States result from infection with the
HIV-1 virus. HIV-2 is prevalent in West Africa but is very
Clinical Manifestations and Pathology
rarely diagnosed in the United States; when it is diagnosed
Although acute, chronic, and fulminant hepatic failure cases in the United States, it is usually linked to an association
have been associated with GBV-C/HGV, there are other studies with West Africa.23
that do not implicate GBV-C/HGV. One article showing a
strong association with fulminant hepatitis dealt with a certain Profile
mutated strain of GBV-C/HGV.17 In another article by Halasz,18
33 GBV-C/HGV individuals were identified who had no coin-
fection with other known hepatitis viruses. No evidence of liver Clinical Manifestations and Pathology
disease, clinical or biochemical, was found. In fact, there is Primary infection with HIV may be asymptomatic or may re-
some evidence that patients with HIV who have a coinfection sult in a mild, chronic lymphadenopathy with symptoms
with HGV have a slower progression to AIDS.16 Overall data similar to those seen in infectious mononucleosis. Symptoms
do not support GBV-C/HGV as a major cause of liver failure. may occur within 6 to 12 weeks of infection and persist for
a few days to 2 weeks. HIV enters the cell by the binding of
Epidemiology and Transmission the virus glycoprotein 120 with cell surface receptors. Cells
HGV is transmitted by the bloodborne route. Parental trans- possessing these receptors include CD4+ lymphocytes,
mission through contaminated blood and the presence of the macrophages, and other antigen-presenting cells. The disease
virus in bile, stool, and saliva suggest transmission through may have a long, clinical latency period with the absence of
the fecal-oral and respiratory routes.9 It has been found in clinical symptoms. During this period, antibody concentra-
20% to 24% of intravenous drug users and in higher rates tion and viral load reach equilibrium. As the viral load in-
among people with HIV.19 Vertical or perinatal transmission creases and the CD4 count decreases, the patient progresses
from mother to child has been documented.20 toward clinical AIDS. When the CD4 count is less than
Most adult infections appear to be transient, with viral 200/µL, the patient is classified as having clinical AIDS. The
clearance followed by antibody to the viral envelope (E2)
production. Vertical or perinatal infections and other infec-
tions established early in life can last for years but do not HIV-2
HIV-1
cause liver disease.18,20

Laboratory Diagnosis gp120 gp41 gp125 gp36


Reverse transcription polymerase chain reaction (RT-PCR) RNA p26 RNA
p24
for GBV-C/HGV-RNA is used to diagnose a current, ongoing
infection. Anti-E2 along with a negative PCR for GBV-C/
RT RT
HGV-RNA indicates a past infection and recovery. Individu-
als who never develop the GBV-C/HGV E2 antibody are still Figure 14–4. Schematic representation of human immunodeficiency virus
infected.21 These assays are first generation, and the evalua- genomes, HIV-1 and HIV-2. RT = reverse transcriptase.
tion has not been completed on the sensitivity and specificity
of these assays.18
Table 14–4 Components of the HIV Virus
Prophylaxis and Treatment
Bands Observed
Interferon-α treatment has been used with conflicting
results. In most cases, the level of the GBV-C/HGV-RNA Gene HIV-1 HIV-2 Protein
returned to normal levels once therapy was discontinued. Gag p18, p24, p15 p16, p26, p55 Core
Only a small percentage of cases with low pretreatment viral
loads had a predictable sustained response.22 Pol p31 Endonuclease

p51, p65 p68 Reverse transcriptase


HIV Types 1 and 2
Env gp41 gp36 Transmembrane protein
HIV-1 and HIV-2 are well recognized as the etiologic agents gp120, gp160 gp140, gp125 Envelope unit
of AIDS. AIDS was first diagnosed in 1981, but the causative
agent was not identified until 1984. gp = glycoprotein (number indicates molecular weight); p = protein
6888_Ch14_307-332 29/10/18 5:43 PM Page 314

314 PART III Transfusion Practices

resultant immunodeficiency allows the onset of opportunis-


tic infections such as Pneumocystis carinii pneumonia, p24
Anti-p24
Kaposi’s sarcoma (KS), fungal infections, and a host of oth- Anti-gp41,
ers. About 50% of patients do not progress to clinical AIDS 120, 160
for 10 years or more.

Epidemiology and Transmission


The number of cases of HIV infection rose rapidly in the
1980s after identification of the disease. After the incorpo-
ration of combination retroviral drug therapy in treatment
protocols in the late 1990s, there was a significant reduction
in the reported numbers of new AIDS cases and deaths.
Reduction of death rates has resulted in an increased number
of persons living with AIDS. The Centers for Disease Control
and Prevention (CDC) estimates that 1.1 million adults and
adolescents were living with diagnosed and undiagnosed Infection Acute Asymptomatic ARC AIDS
6 wks- months
HIV infection at the end of 2011 in the United States.23 2 mo to years
HIV is transmitted through sexual contact with an in-
fected person, use of contaminated needles during drug use, Figure 14–5. Pattern of serologic markers detected in HIV infection. ARC =
and very rarely through transfusion of blood or blood com- AIDS-related complex.
ponents. Congenital transmission may also occur. High-risk
populations include men who have sex with men and HIV-2 enzyme-linked immunoassay, and a rapid diagnostic test
IV drug users. The blood supply is most at risk from those used for HIV-1 and HIV-2 differentiation.24 HIV-1 RNA detec-
individuals who have been recently infected with the virus tion by NAT was introduced in 1999 as an investigational new
but have not yet produced antibodies. drug (IND). In 2002 the first NAT test for HIV-1 RNA was ap-
It was recognized over a decade ago that transfusion of proved (licensed) for use in the United States. HIV-1 RNA test-
blood and components from HIV-infected individuals may ing closed the window period between time of infection and
result in HIV infection in the recipient and development of the detection of antibody to 7 to 10 days.24 False-positive test
transfusion-associated AIDS. HIV infection may occur after results for either antibody and/or NAT may allow for donor
receiving a single contaminated unit of whole blood or its reentry.25 The risk of HIV-1 infection through transfusion is
components. Albumin and immune globulins have not been 1:1,000,000 per unit transfused.24
reported to transmit HIV. Blood donor screening practices
have dramatically reduced the incidence of transfusion- Prophylaxis and Treatment
related transmission, but the possibility of transmitting In 2005, the CDC revised recommendations for routine test-
HIV remains when the donor has not yet seroconverted and ing and has implemented an initiative aimed at reducing
the level of virus in the blood is low. This subpopulation of barriers to early diagnosis of HIV infection entitled Advanc-
HIV-infected persons who are unaware of their positive ing HIV Prevention: New Strategies for a Changing Epi-
serostatus may pose the greatest risk to the blood supply. demic. This initiative also aims to increase access to medical
care, treatment, and prevention for those persons living with
Laboratory Diagnosis HIV.26 To reduce perinatal transmission, the CDC recom-
The use of very sensitive serologic testing in screening the mends routine HIV testing of all pregnant women and
blood supply has resulted in an extremely low risk of HIV screening of all neonates whose mothers have not been
transmission. The pattern of serologic markers detected in tested.
HIV infection is shown in Figure 14–5. The window period Treatment with highly active antiretroviral therapy has
is that time after infection but before antibody or antigen is lengthened life and improved quality of life for those infected
detectable by currently available testing procedures. It is pos- with HIV. Use of this therapy has resulted in the most stable
sible for a donation to be infectious but to test negative for HIV morbidity and mortality rates since 1998.27
HIV-1/2 antibodies when the donor is in the window period.
Antibodies are detectable at about 22 days after infection. Human T-Cell Lymphotropic Viruses
In 1985, using an EIA to screen all blood donations for Types I/II (HTLV-I/II)
antibodies to HIV-1 was instituted. Testing for HIV-2 was
initiated in 1992. EIA is used for the qualitative detection of HTLV-I and HTLV-II are RNA retroviruses. HTLV-I causes a
HIV-1/2 in a ChLIA test. Positive screening tests are repeated T-cell proliferation with persistent infection.28 Once the RNA
in duplicate, and if at least one of the duplicates also tests pos- has been transcribed into DNA, it is integrated randomly into
itive, a confirmatory test is performed. The confirmation of the host cell’s genome.29 Once integrated into the DNA, the
HIV-1/2 is performed using one or a combination of tests. provirus can either complete its replication cycle or remain
These include HIV-1 indirect immunofluorescence assay (IFA), latent for many years.
6888_Ch14_307-332 29/10/18 5:43 PM Page 315

Chapter 14 Transfusion-Transmitted Diseases 315

Profile whereas HTLV-II is seen in some Native American popula-


tions.28,31 Indications from the high numbers of carriers and
Clinical Manifestations and Pathology low numbers of individuals diagnosed with actual disease are
HTLV-I was the first retrovirus to be associated with a human that most carriers are asymptomatic their entire lives.28,29
disease. That association was with adult T-cell lymphoma/ The risk of HTLV-I/II transmission through transfusion is
leukemia (ATL), a highly aggressive, mature T-cell non- less than 1:2,000,000 units transfused.24
Hodgkin’s lymphoma with a leukemic phase.30 ATL does not
respond well to chemotherapy, and mean survival time with Laboratory Diagnosis
acute ATL is less than 1 year. Immunodeficiency similar to Because the majority of carriers are asymptomatic, diagnosis
that of patients with AIDS develops, making the ATL patient is based on seroconversion after exposure. In the United
susceptible to other hematologic malignancies.29 HTLV-I is States, FDA-licensed EIA using whole virus lysates from both
also associated with the progressive neurological disorder viruses is used to detect both anti-HTLV-I and anti-HTLV-II.
known as HTLV-I-associated myelopathy or tropical spastic With 60% homology between HTLV-I and HTLV-II, antibody
paraparesis (HAM/TSP). A few case reports in the literature cross-reactivity makes it difficult to distinguish between the
suggest that HTLV-II may impact the development of neuro- two viruses. Results are reported as reactive or negative for
logical diseases, including HAM, but subsequent studies have HTLV-I/II. For first-time reactive donors testing is repeated
failed to support this with convincing evidence.31 using a second licensed HTLV-I/II screening test and with a
Epidemiological data suggest blood donors infected with licensed Western blot test. There are no NAT tests available
HTLV-I or HTLV-II have an excess of infectious syndromes, for HTLV-I/II and no FDA guidance for donor reentry.24
such as pneumonia, bronchitis, and urinary infections.32 The AABB33 and FDA have published guidelines on the
HTLV-I is associated with uveitis and infective dermatitis of use of the unit for transfusion and donor notification as
children, Sjögren’s syndrome, polymyositis, and facial nerve shown in Figure 14–6. The guidelines state that if the donor
palsy.9 is repeatedly reactive by the test of record (original EIA)
but negative by the second licensed EIA of a different type
Epidemiology and Transmission (different manufacturer), the donor is still eligible for dona-
HTLV-I is transmitted vertically (breastfeeding), sexually tion. The donor can continue to donate as long as the test-
(transmission from male to female more common), and of-record EIA is negative on the next donation. If the donor
parenterally (blood transfusion or IV drug abuse).9 Because is repeatedly reactive by test of record on two separate occa-
recipients of RBCs, platelets, and whole blood, but not fresh- sions or on the same donation by the test-of-record assay and
frozen plasma, have seroconverted, it is believed that trans- the different manufacturer’s EIA, the donor is indefinitely
mission requires introduction of infected living white blood deferred.
cells (WBCs).28,29 This theory is supported by the fact that
units stored for at least 7 days before transfusion are less Prophylaxis and Treatment
likely to transmit the virus. ATL does not respond well to treatment. However, early
There appears to be a strong correlation between disease treatment with corticosteroids appears to have some effect
development and host factors such as cytotoxic T lympho- on HAM/TSP. There is no treatment for chronic or advanced
cytes and HLA types. Susceptibility to ATL seems to correlate disease.28 The best prophylaxis is to prevent exposure. How-
with polymorphisms of the tumor necrosis factor α (TNF-α) ever, as the majority of infected individuals are asympto-
that result in an increased production of TNF-α.29 In indi- matic, it is difficult to prevent spread to an uninfected
viduals with HAM/TSP, both cellular and humoral immune individual vertically or sexually. Infected mothers should not
responses are increased as compared with those of asympto- breastfeed.
matic carriers and seronegative controls.31 However, ATL
seems to occur in persons who were infected as infants, with West Nile Virus
a latent period of approximately 67 years, whereas HAM/TSP
is generally seen in individuals who are infected in childhood West Nile virus (WNV) is a member of the Flavivirus family
or as an adult, with a variable latency as short as weeks to and is a human, avian, and equine neuropathogen. It is a
months. There is a 40% to 60% probability of seroconversion single-stranded RNA lipid-enveloped virion that is common in
within 51 days following an infected blood transfusion.28 For Africa, West Asia, and the Middle East. WNV is a member of
both diseases, the host’s ability to keep the proviral load low the Japanese encephalitis virus antigenic complex that includes
correlates with asymptomatic carriers.29,31 St. Louis encephalitis virus prevalent in the Americas, Japanese
Worldwide, it is estimated that 10 to 20 million people are encephalitis virus prevalent in East Asia, and Murray Valley
infected with HTLV-I and HTLV-II.29,31 It is endemic in parts encephalitis virus and Kunjin virus prevalent in Australia.34
of southern Japan, central and West Africa, the Caribbean, WNV was first documented in the Western Hemisphere
the Middle East, Melanesia, Papua New Guinea, the Solomon when 149 cases were reported in New York in 1999. In the
Islands, and in Australian aborigines. In the United States, United States, reported cases of WNV reached 9,862 cases
HTLV-I and HTLV-II are seen primarily in IV drug users. with 264 reported deaths in 2003. By 2015, WNV cases num-
HTLV-I is also seen in immigrants from endemic areas, bered 2,175, and 146 deaths were reported.35
6888_Ch14_307-332 29/10/18 5:43 PM Page 316

316 PART III Transfusion Practices

First Donation or
Previous Donation Previous Donation
Negative Repeat Reactive

Current EIA Current EIA

repeat Negative or repeat Negative or


reactive non-repeat reactive non-repeat
reactive reactive
2nd licensed 2nd licensed
EIA Unit ok to EIA unit ok to use
use donor not
notified, but
negative reactive negative reactive remains in
surveillance
category
unit destroyed unit unit destroyed unit
donor not destroyed destroyed
notified, but donor notified;
entered into indefinite
surveillance confirmatory deferral confirmatory
category tests* tests*

Not performed performed performed performed

donor notified
indefinite deferral neg, or Ind pos

Donor Donor donor donor


notified notified counseling notified
indefinite permanent indefinite
Ind - indeterminant; pos - positive; neg - negative deferral deferral deferral
EIA - enzyme immunosorbent assay
*confirmatory tests - western blot; immunoblot;
recombinant immunoprecipitation assay

Figure 14–6. Flowchart for HTLV-I/II testing.

Profile and then bite humans. Although at least 65 species of


mosquitoes have been found to carry WNV, the genus Culex
Clinical Manifestations and Pathology is the chief vector.39 The infection in humans has an incuba-
WNV is usually subclinical but may cause a mild flulike dis- tion period of approximately 3 to 14 days following the
ease. However, the strain in the United States is often associated mosquito bite, with symptoms lasting 3 to 6 days. Other
with more severe disease. In 2002, outbreaks determined animals can become infected, including horses, cats, dogs,
by antibody screening found that 20% to 30% of infected bats, chipmunks, skunks, squirrels, and domestic birds and
individuals exhibited symptoms ranging from mild fever and rabbits. Although mosquito bites are the most common route
headache to extensive rash, eye pain, vomiting, inflamed of infection, there is a slight risk of contracting WNV from
lymph nodes, prolonged lymphocytopenia, muscle weakness, blood components, organ transplants, pregnancy, and breast
disorientation, and even acute flaccid paralysis and po- milk. Following the introduction of blood donor NAT screen-
liomyelitis.36 The association of paralysis and poliomyelitis ing for WNV RNA in 2003, there have been 14 cases of
with WNV is recent. It is a peripheral demyelinating process transfusion-associated WNV infection reported in the Amer-
similar to Guillain-Barré syndrome.37 WNV is capable of cross- ican Red Cross system; translating to a risk of transmission
ing the blood-brain barrier and can cause what is known as of WNV through transfusion at 1 per 84,000,000 units
West Nile encephalitis, West Nile meningitis, or West Nile donated.24
meningoencephalitis.38 Approximately 1 in 150 infections re-
sults in severe neurological disease that may cause permanent Laboratory Diagnosis
neurological impairment, with encephalitis reported more Viremia usually lasts approximately 6 days and peaks around
often than meningitis. The risk of severe neurological disease the onset of symptoms. Once clinical symptoms occur, the
increases markedly for anyone over the age of 50 years.38 IgM WNV-specific antibody titer increases and the virus con-
centration in the bloodstream decreases. Until July 2003,
Epidemiology and Transmission diagnosis depended on the clinical findings and specific
Birds are the primary amplifying hosts in a mosquito-bird- laboratory tests. IgM antibody-capture enzyme-linked im-
mosquito cycle. Incidentally, mosquitoes feed off infected birds munosorbent assay (ELISA) was the method used to detect
6888_Ch14_307-332 29/10/18 5:43 PM Page 317

Chapter 14 Transfusion-Transmitted Diseases 317

IgM antibody to the WNV in serum and cerebrospinal fluid infected individuals experience symptoms. ZIKV has been
(CSF). In the 1999 and 2000 outbreak in New York, 95% of associated with severe neurological complications including
all infected patients for whom CSF was tested had a demon- increased rates of microcephaly and fetal brain anomalies
strable IgM antibody. However, because all Flaviviruses are during pregnancy and Guillain-Barre syndrome.43
antigenically similar, cross-reactivity has been observed in
testing persons who have been vaccinated for a Flavivirus, Epidemiology and Transmission
such as yellow fever or Japanese encephalitis, or who have ZIKV was first described in a rhesus monkey in 1947. Human
been recently infected with another Flavivirus, such as illness due to ZIKV was confirmed in cases in Nigeria in
St. Louis encephalitis or dengue fever. The plaque reduction 1953. Outbreaks of ZIKV infections occurred in Micronesia
neutralization test is the most specific test for arthropod- and in French Polynesia in 2007 and 2013, respectively. By
borne Flaviviruses and helps to distinguish false-positive IgM 2015, ZIKV cases were reported in the Americas with cases
antibody-capture ELISA from cross-reactivity.40 reported in the United States from Puerto Rico. In July of
Clinically, the serologic tests for IgM antibodies to WNV 2016, the first cases of ZIKV infection were reported on the
using ELISA can be used for testing symptomatic patients. continental United States in Florida.44 In 2016, ZIKV infec-
Because the virus is in the bloodstream before either symp- tions became a nationally notifiable condition and subject to
toms or antibodies develop, blood screening tests for WNV surveillance by the Centers for Disease Control and Preven-
that identify the virus itself were needed. In June 2003, two tion (CDC). Provisional 2016 data indicate that there were
commercial WNV-screening NATs were distributed, and im- 5,102 symptomatic (excluding congenital disease cases)
plementation of donor blood testing began “under phase III ZIKV cases. Approximately 95% of the reported cases
investigational new drug (IND) FDA approval.”41 As of involved travelers returning from affected areas.45 The CDC
July 14, 2003, all civilian blood donations were being established a U.S. Zika pregnancy registry for surveillance
screened by NAT. Units are initially screened individually or data on congenital disease cases. Data gathered in 2016
in pools of 6 or 16, depending on the kit manufacturer. through June of 2017 include a reported 1,687 completed
Individual samples are tested if the pool is positive with pregnancies and eight pregnancy losses (with birth defects).
NAT.41 Testing facilities must establish policies and proce- Eighty-eight of the 1,687 completed pregnancies resulted in
dures to define when pools versus individual NAT testing a live-born infant with birth defects.46
will be performed based on threshold levels during seasonal
activity.41 From 2003 through 2016 the Red Cross has de- Laboratory Diagnosis
tected approximately 3,500 WNV-infected donors.24 Approximately 80% of ZIKV infections are asymptomatic.
WNV has become a major area of focus for transfusion When symptoms do occur, the period of viremia without
safety, especially since there have been large outbreaks in the symptoms ranges from 3 to 12 days.44 ZIKV RNA has been
United States when comparing WNV with other transmissi- found in asymptomatic blood donors, and there have been
ble diseases tested by NAT testing. There is a similar interval reported cases of probable transmission of ZIKV through
in which it is detected, but when comparing testing by the transfusion.44 The FDA has established ZIKV as a transfusion-
minipool method, WNV has a much shorter duration of transmitted infection. In February of 2016, the FDA and
viremia.42 CDC issued guidance documents making recommenda-
Prophylaxis and Treatment tions for reducing the risk of ZIKV transmission through
transfusion. The February guidance included recommen-
Individuals should avoid mosquitoes and wear mosquito re-
dations to defer donors for 28 days following travel to
pellant and appropriate clothing if they are going to be in a
countries with active ZIKV transmission as well as donors
mosquito-infested area. Once infected, there is no licensed
who had sexual contact with men who traveled to those
treatment, only supportive therapy. Research is ongoing for
countries. The FDA guidance also included recommenda-
the use of ribavirin, interferon-α,36 and West Nile immune
tions for areas of active ZIKV transmission to import blood
globulin to treat WNV. Having survived the illness, a person
from nonactive areas or to test donor blood for ZIKV RNA
is immune for life.
using NAT or to use blood components that were pathogen
Zika Virus reduced using an FDA-approved technology. In August of
2016, the FDA released an updated guidance for the in-
Zika virus (ZIKV) is an arbovirus in the Flavividae family, dustry requiring testing of all donations with an individual
genus Flavivirus. ZIKV is transmitted by the Aedes aegypti and donor nucleic acid test for ZIKV under an investigational
Aedes albopictus mosquitoes. Cases of transmission through new drug application.44 By the end of 2016, all donor
intrauterine, perinatal, laboratory-acquired, sexual- and blood collected in the United States is tested for ZIKV RNA
transfusion-associated transmission have been reported.43 using NAT. NAT-reactive donor blood is further tested
using a polymerase chain reaction (PCR) diagnostic test
Profile and antibody testing.45 Antibody test platforms include
enzyme-linked immunoabsorbant (ELISA) and immuno-
Clinical Manifestations and Pathology fluorescence assays. These tests have a low specificity with
ZIKV disease symptoms include fever, headache, rash, and noted cross-reactivity with antibodies directed against
muscle and joint pain. It is estimated that less than 20% of other flaviviruses.43
6888_Ch14_307-332 29/10/18 5:43 PM Page 318

318 PART III Transfusion Practices

Prophylaxis and Treatment Epidemiology and Transmission


Similar to WNV infections there is no medicine available Transmission occurs from person to person through contact
for treatment of ZIKV infection. Avoidance of mosquito with infected body fluids, which may include urine, semen,
bites through use of insect repellent is recommended saliva, blood, cervical secretions, and breast milk. CMV is
to reduce the risk of infection in areas of active ZIKV the most frequently transmitted virus from mother to fetus.50
transmission.47
Laboratory Diagnosis
Other Viruses Antibodies formed to CMV last a lifetime and can be de-
tected by ELISA. Other laboratory tests include PCR, fluo-
Cytomegalovirus rescence assays, indirect hemagglutination, and latex
agglutination. If the patient is symptomatic, active infection
Cytomegalovirus (CMV) is a member of the herpesvirus can be detected by viral culture of urine, throat swabs, and
group and is found in all geographic locations and socio- tissue samples.48
economic groups, with a higher prevalence in developing
countries. In areas with lower socioeconomic conditions, Prophylaxis and Treatment
the prevalence approaches 100%. Of adults in the United Currently, there is no treatment for CMV for a healthy indi-
States, 50% to 85% have been exposed to CMV by the age vidual. Infants are being evaluated using antiviral drug ther-
of 40 years.48 apy, and ganciclovir is being used for patients with depressed
immunity.
Clinical Manifestations and Pathology
Blood and blood components are not universally screened
When exposure occurs after birth to an individual with a for CMV because of the generally benign course of this dis-
competent immune system, there are generally few symp- ease and the high percentage of virus carriers. To prevent
toms. Rarely, mononucleosis-like symptoms with fever and CMV transmission, leukocyte-reduced blood or blood from
mild hepatitis occur. Once an individual is exposed, CMV seronegative donors may be used. Leukoreduction using
can remain latent in the tissues and leukocytes for years, high-efficiency filters such that the final level of leukocytes
with reactivation occurring from a severe immune system is less than or equal to 5 × 106 leukocytes per component
impairment.48 appears to work well with high-risk neonates (weighing less
Those at the highest risk of a CMV infection are fetuses and than 1,200 grams) and transplant recipients.2 In an AABB
immunocompromised individuals receiving allogeneic mar- bulletin, prestorage leukoreduction was encouraged rather
row transplants or solid organ transplants. CMV-seronegative than bedside leukoreduction.
recipients transplanted with CMV-seronegative allogeneic
marrow are at risk if they receive untested or non–WBC- Epstein-Barr Virus
reduced blood components. CMV-seronegative women who
become infected in the first two trimesters have up to a Epstein-Barr virus EBV is a ubiquitous member of the her-
40% chance of delivering an affected infant, many of whom pesvirus family. As many as 95% of the adult population in
will have clinically apparent disease. Intrauterine transfusions the United States have been exposed to the virus by the age
with CMV-positive components is also a high risk to the of 40 years and maintain an asymptomatic latent infection
fetus.49 in B lymphocytes for life. Infections occurring in infants or
Individuals at moderate risk are recipients of solid organ young children are usually asymptomatic. In adolescence
transplants, persons with HIV, and individuals who may re- and young adulthood, EBV causes infectious mononucleosis
quire an allogeneic marrow transplant in the future. When in 30% to 50% of patients.51
the individual becomes immunosuppressed, a reactivation Although transfusion transmission is rarely an individual’s
of a latent infection is possible, resulting in a clinically ap- first exposure to the virus and reactivation usually occurs
parent infection.49 only in immunocompromised individuals, there are a few
Low-birth-weight neonates and autologous marrow recip- cases in the literature of transfusion-associated EBV. EBV is
ients are considered to be at low risk. Preterm, multitrans- not detected by current practices and could cause severe con-
fused neonates weighing less than 1,200 grams are currently sequences in immunocompromised patients, particularly
considered to be at a lower risk than once considered as a organ transplant patients.52
result of better transfusion techniques and management EBV has been called the “kissing disease” because the
of their condition. However, leukocyte-reduced or CMV- virus usually replicates in the cells of the oropharynx, pos-
negative units reduce the risk of CMV infection in these low- sibly in infected B cells. The virus is shed in the saliva and is
birth-weight neonates.49 The neonate may be exposed at the most frequently associated with infectious mononucleosis.
time of delivery, through breastfeeding, or through contact EBV was first discovered in 1964 in Burkitt’s lymphoma
with seropositive individuals. Approximately 1% of all new- cells.53 Since then it has been associated with many illnesses
borns are infected with CMV, but most are asymptomatic at besides infectious mononucleosis and cancers such as na-
birth.50 The fetus that is exposed to the mother’s reactivation sopharyngeal carcinoma, non-Hodgkin’s lymphoma, oral
of the virus during pregnancy rather than a primary expo- hairy leukoplakia in AIDS patients, T-cell lymphomas, and
sure rarely has any damage.50 Hodgkin’s disease.9
6888_Ch14_307-332 29/10/18 5:43 PM Page 319

Chapter 14 Transfusion-Transmitted Diseases 319

Parvovirus B19 Disease transmission of parvovirus B19 is rare. Very high


levels of parvovirus B19 (up to 1012 IU/mL) in plasma of
Human B19 parvovirus (B19) is a small, nonenveloped acutely infected asymptomatic donors may pose a greater
virus.2 It causes a common childhood illness called “fifth dis- risk for plasma derivatives. This is due to the pooling of
ease” and is usually transmitted through respiratory secre- larger plasma units by manufacturers when processing these
tions. Fifth disease presents with a mild rash described as products.57 There have been no confirmed reports that
“slapped cheek” when occurring on the face and a lacy red immunoglobulin and albumin products have transmitted
rash when occurring on the trunk and limbs. Approximately parvovirus B19 infection.57
50% of adults have been exposed as children or adolescents
and have protective antibodies. Primary infection in an adult Human Herpesvirus 6 and Human Herpesvirus 8
is usually asymptomatic, but a rash or joint pain and swelling
may occur transiently.54 Human herpesvirus 6 (HHV-6) is a very common virus that
As in all viral infections, the virus must enter the cell causes a lifelong infection. Seroprevalence approaches 100%
through a specific cell receptor. B19 parvovirus enters the in some populations. The virus replicates in the salivary
red blood cell (RBC) via the P antigen and replicates in the gland and then remains latent in lymphocytes, monocytes,
erythroid progenitor cells.54 The cytotoxicity of erythroid and perhaps other tissues.
precursors can lead to serious illness in individuals In childhood, HHV-6 causes roseola infantum, also
with chronic hemolytic anemia, such as sickle cell disease known as “exanthem subitum” or “sixth disease.” Symptoms
and thalassemia, who may have a transient aplastic crisis. are those of a mild, acute febrile disease. In immunocompe-
Severe RBC aplasia or chronic anemia may manifest in tent adults, it is very rare to find infection or reaction from
patients with chronic or acquired immunodeficiency or sites other than the salivary gland where secretions from
malignancies or in organ transplant recipients. Hydrops saliva are a known source of transmission. Side effects are
fetalis and fetal death can occur when the virus is transmit- not common but can include lymphadenopathy and fulmi-
ted during pregnancy.54 nant hepatitis.
The viremic stage occurs shortly after infection. A donor HHV-6 has been associated with a number of diseases
would be asymptomatic but capable of transmitting the virus other than roseola infantum, such as multiple sclerosis and
during this period. This is a concern for donor centers lymphoproliferative and neoplastic disorders. There is no
because the rate of seroconversion is high after exposure. evidence to support a TTD association; with the high level
In one study, B19 DNA was found in approximately 1 out of of seropositivity in the population, blood components are
800 donations. B19 is very resistant to heat and detergent not being tested for HHV-6.30
and has been found through PCR to be present in plasma HHV-8 is another human herpesvirus. Unlike HHV-6, it
components. Because of the lack of inactivation in the is not common in the population but has been seen in
manufacturing process, B19 has been implicated in several Africa.58 Only 3% of donors are seropositive in the United
studies, with transmission through factor concentrates and, States.59 It is associated with several diseases that generally
in one study, through an antithrombin III concentrate.55 affect the immunosuppressed patient. These include Kaposi’s
There has been ongoing regulatory concern about the sarcoma (KS), primary effusion lymphoma, and multicentric
safety of plasma derivatives that has led some manufacturers Castleman’s disease. Spread is generally through sexual con-
and regulatory authorities to require B19 DNA qualification tact. However, in KS patients, 30% to 50% have circulating
testing of plasma and release testing of manufactured lots.56 lymphocytes harboring HHV-8, which lends support to the
The FDA recommends manufacturers of plasma-derived premise that exposure to HHV-8 could be transfusion-
products to engage in practices that will reduce the time associated. There has been no evidence to support this to
between product collection and process testing in order for date.59 In post-transplant patients who develop KS, it ap-
the collection establishments to be notified of positive test pears to be due to reactivation. The transmission of HHV-8
results within the in-date period of any blood components has been associated with organ transplants and injection
that are intended for transfusion.57 drug use.58
In a study by Weimer and colleagues,55 plasmas were
tested for B19 DNA levels by PCR, and those with high titers Emerging Viruses
were eliminated from the pool used in the manufacture of
antithrombin III (ATIII). After the manufacturing process, Chikungunya Virus (CHIKV)
PCR was used to compare ATIII concentrates made from a
pool in which high titers were excluded and ATIII concen- CHIKV is a small enveloped single-stranded RNA virus of
trates that were made from a pool that had not been tested the family Togaviridae. The virus is vector borne, transmitted
for B19 DNA. None of the concentrates manufactured through mosquitoes mainly from the Aedes family.60 Primary
with high-titer plasma eliminated from the pool were PCR- disease symptoms include high fever, severe joint pain,
positive, whereas 66% of the concentrates that were not headache, muscle pain, rash, and leukopenia. Joint pain may
tested prior to manufacturing were PCR-positive. This indi- persist for months in 10% to 15% of those infected.60 Menin-
cates the effectiveness of eliminating high-titer plasmas in goencephalitis has been described during an outbreak on Re-
reducing the B19 DNA to undetectable levels. union Island in 2005 to 2007.60 During the Reunion Island
6888_Ch14_307-332 29/10/18 5:43 PM Page 320

320 PART III Transfusion Practices

outbreak the estimated risk of viremic donations was 132 to There are no FDA-approved therapeutics or vaccines for
1,500 per 100,000 donations.60 Despite the relative high risk, EVD. Due to the severity of EVD and the possibility of asymp-
there were no reported transfusion-associated cases. In 2016 tomatic viremia, the FDA has determined that Ebola virus
there were 175 CHIKV cases in the United States and 171 in meets the criteria of a transfusion-transmitted infection.64
U.S. territories reported to the CDC.61 Of the cases reported FDA guidelines published in January of 2017 include recom-
in the United States, all were reported from travelers from mendations that donor history questionnaires include assess-
affected areas. The U.S. territory of Puerto Rico reported 170 ment of donors for a history of Ebola virus infection or
locally acquired CHIKV infections.61 There is no medication disease, assessment of donors with a history of travel in the
for treatment of the disease and no vaccine for prevention. past 8 weeks to a country with widespread transmission of
Similar to WNV and ZIKV, avoidance of mosquito bites is EVD or cases in urban areas with uncertain control measures,
the recommendation to reduce risk of infection. Theoreti- a history of contact in the past 8 weeks with a person known
cally, CHIKV could be spread through transfusion, but there to have Ebola virus infection, a history of sexual contact with
have been no reported cases of CHIKV transmission through persons known to have recovered from EVD, and a history
transfusion even during outbreaks.60 of notification by a public health authority that he or she may
have been exposed in the past 8 weeks.64 Donors must be in-
Dengue Virus (DENV) definitely deferred if they have a history of Ebola virus infec-
tion or disease. Eight-week deferral periods are required from
DENV is a small enveloped single-strand RNA virus in the the date of the donor’s departure from a country with wide-
genus Flavivirus.62 The virus is vector borne by the mos- spread transmission of Ebola virus, after the last date of donor
quitoes Aedes aegypti and Aedes albopictus. Transmission contact with a person infected with Ebola virus, after the last
of DENV through transfusion and organ transplantation date of donor sexual contact with a person known to have re-
has been reported.62,63 Symptoms of disease usually occur covered from EVD, or after notification of exposure from a
4 to 7 days following the bite of an infected mosquito. public health authority.64
Illness usually presents with onset of fever, rash, a severe
headache, lumbosacral aching pain followed by muscle Bacterial Contamination
pain, bone pain, anorexia, nausea, vomiting, and weak-
ness. Severe dengue is rare and includes capillary leakage Overview
sequelae leading to shock with a subset demonstrating
hemorrhagic manifestations.62 It is estimated that 40% of As the infection risk for other diseases has decreased due to
the world’s population live in endemic areas, and the better donor testing, bacterial contamination has come to the
World Health Organization (WHO) estimates that 50 to forefront and has become a great concern as a transfusion-
100 million infections occur each year, with 22,000 deaths. transmitted disease.66 Although the incidence of transfusion-
Nearly all reported DENV infection cases in the continen- associated bacterial sepsis is low, the morbidity and mortality
tal United States are associated with travelers from rates are high. Common sources of bacterial contamination
endemic areas. There have been outbreaks reported in the are from donor skin or from asymptomatic donor blood.2
U.S. territory of Puerto Rico, and DENV is considered Platelets have been the most frequent source of septic trans-
endemic in that area as well as in the Virgin Islands and fusion reactions because room temperature storage promotes
most United States–affiliated Pacific Islands.63 There is cur- bacterial growth. In 2004, bacterial contamination screening
rently no FDA or AABB guidance for donor deferral and no methods were implemented, resulting in lower numbers of
tests licensed for donor testing. Risk of DENV infection can reported cases of transfusion-related fatalities due to bacterial
be reduced by mosquito control and avoidance. Vaccines are contamination. Although the percentage of fatalities due to
in clinical trials, and there are no medicines for treatment bacterial contamination has decreased since 2004, reported
of DENV illness other than those used in supportive care.62 cases to the FDA have essentially leveled off between fiscal
year 2005 and fiscal year 2015. Bacterial contamination
Ebola Virus accounted for 13% of confirmed or possible transfusion-
related fatalities in fiscal year 2005 and approximately 13.5%
Ebola virus is a member of the family Filoviridae and can in fiscal year 2015.67
cause severe hemorrhagic fever. Ebola virus disease (EVD)
has historically been associated with high mortality rates. Clinical Manifestations and Pathology
Symptoms of EVD include fever, severe headache, muscle The most common signs and symptoms of transfusion-
pain, weakness, followed by diarrhea, vomiting, and abdom- associated sepsis are rigors, fever, and tachycardia.68 Other
inal pain. Diffuse hemorrhage has been reported. Symptoms symptoms may include shock, low back pain, disseminated
occur most often within 4 to 10 days following infection and intravascular coagulation (DIC), and an increase or decrease
generally within 21 days. In the 2014 outbreak in West Africa, in systolic blood pressure.2 Of all the blood components,
5% reported symptom onset at >21 days.64 Ebola virus RNA platelets are associated with the highest risk of sepsis and
has been detected in semen of EVD survivors for up to 199 fatality.69 Sepsis due to platelets can occur hours after
days following symptom onset.65 There have also been the transfusion, and the connection may be unrecognized.
isolated reports of asymptomatic Ebola virus infection.64 This may be because many patients who receive platelets
6888_Ch14_307-332 29/10/18 5:43 PM Page 321

Chapter 14 Transfusion-Transmitted Diseases 321

are already immunosuppressed because of their condition within 24 hours after collection and has drafted new guid-
or treatment, and the sepsis may be attributed to the ance for the field.69
immunosuppression.
Prophylaxis and Treatment
Epidemiology and Transmission The 31st edition of the AABB Standards states that “the blood
Bacterial contamination usually originates with the donor, bank or transfusion service shall have methods to detect bac-
either through skin contamination at the phlebotomy site or teria or use pathogen reduction technology in all platelet
an asymptomatic bacteremia. It may also occur through con- components.”1
tamination during processing.2 Bacterial contamination is Use of apheresis platelets rather than pooled whole
the most common risk of infection due to transfusion of blood–derived platelets from multiple donors reduces the in-
blood. Bacterial contamination of platelets occurs in approx- cidence of contamination occurring during phlebotomy.
imately 1 of 2,000 to 3,000 platelet transfusions.70 Staphylo- However, apheresis platelets cannot meet all platelet trans-
coccus epidermidis or Staphylococcus aureus are the most fusion needs, and whole blood–derived platelets are still
common bacterial contaminants of blood.70 needed. Therefore, proper arm preparation for phlebotomy
According to the CDC,71 Yersinia enterocolitica is the most is of paramount importance. Standard 5.6.2 in the 31st edi-
common isolate found in RBC units, followed by the tion of Standards states that the use of green soap will no
Pseudomonas species. Taken together, these two account for longer be allowed.1 Improved bacterial disinfection has been
more than 80% of all bacterial infections transmitted by correlated with the use of an iodine-based scrub. In donors
RBCs. In a study by Kunishima and colleagues,72 Propioni- allergic to iodine, chlorhexidine or double isopropyl alcohol
bacterium acnes, a common isolate of human skin, was the skin disinfectant may be used.74
most common bacterial contaminant in RBCs. It is a slow- Phlebotomy diversion consists of collecting the first 20 to
growing anaerobic bacteria that can go unrecognized if tested 30 mL of blood in a separate container to be used for testing.
in aerobic conditions or by using short-term bacterial cul- This reduces the quantity of skin contaminants entering the
tural methods. Although P. acnes has been implicated in only unit during phlebotomy and appears to be very effective in
a few cases of transfusion-related sepsis, studies are needed reducing Staphylococcus species contamination. Several
to confirm long-term safety as it has been associated with blood bag manufacturers have developed systems with a di-
sarcoidosis. version pouch.
Leukoreduction of units can be helpful in removing
Laboratory Diagnosis phagocytized bacteria along with the leukocytes. Some ad-
Before the unit of RBCs or platelets is issued, the unit should vocate this for RBCs to reduce Yersinia contamination. Other
be inspected for discoloration (dark purple or black), which methods under consideration listed in the AABB Technical
strongly indicates contamination. The unit may have no vis- Manual include endotoxin assays, detection of by-products
ible evidence of contamination at the time of issue. However, of bacterial metabolism, NAT, and pathogen inactivation
clots in the unit and hemolysis may also indicate contami- methods.30
nation. Because the bacteria in the unit consume the oxygen, Water baths used in a blood bank can have high bacterial
the cells may lyse, resulting in discoloration in the unit as counts unless disinfected frequently. An overwrap is recom-
compared with the segments that remain normal in color.30 mended for any components placed in the water bath, with
To detect bacterial contamination, both the donor blood inspection of the outlet ports before use.
component and the recipient’s blood should be tested. It is If transfusion-associated sepsis is suspected, treatment
better to test the component itself and not the segments, as should begin immediately without waiting for laboratory
they may be negative.2 The FDA has cleared two culture confirmation. Treatment should include IV antibiotics and
methods for quality-control monitoring of bacterial contam- necessary therapy for whatever symptoms are present, such
ination in platelets. Both methods can be used for leukocyte- as shock, renal failure, and DIC.
reduced apheresis platelets, whereas only one can be used
for leukocyte-reduced whole blood–derived platelets. Syphilis
Bacterial screening of platelets was implemented in the
United States from 2003 to 2004. This has reduced the risk Treponema pallidum, the causative agent of syphilis, is a spiro-
of transfusing contaminated platelets to patients. Between chete. It is usually spread through sexual contact but can be
2004 and 2006, the American Red Cross documented a transmitted through blood transfusions. In 2015 in the
residual risk of clinically relevant septic shock reactions United States, 23,872 primary and secondary cases of
of 1 in 74,807 and a fatality rate of at least 1 in 498,711 syphilis were reported.75 The rates reported between 2000
with platelets that were distributed (to outside facilities) and 2015 showed increasing numbers primarily due to in-
after routine bacteria detection by culture techniques.73 creases of reported cases in men who have sex with men.75
This shows a 50% reduction in reported reactions and The standard serologic tests for syphilis (STS) usually do
fatalities in a 10-month period after bacteria screening was not detect a donor in the spirochetemia phase who has not
implemented. yet seroconverted. Spirochetemia is short, and seroconver-
The FDA has approved a psoralen/UV irradiation–based sion usually occurs after this phase. The last reported case
pathogen reduction method for use on apheresis platelets of transfusion-transmitted syphilis was 1996.76 Despite the
6888_Ch14_307-332 29/10/18 5:43 PM Page 322

322 PART III Transfusion Practices

low sensitivity and positive predictive value of STS testing, such as the MO1-type Babesia.79 Babesia infection may also
the 30th edition of the AABB Standards continues to require be acquired by blood transfusion and solid organ transplant.
the STS.1 FDA guidelines issued in September of 2014 also Estimates are that between 70 and 100 cases of transfusion-
still recommend STS testing of donor blood largely due to transmitted Babesia (TTB) have occurred over the last
the reported higher rates of HIV-, HCV-, HBV-, HBsAg-, and 30 years in the United States, with at least 12 fatalities in
HTLV-positive donations of donors with positive syphilis test transfusion recipients diagnosed with babesiosis.80,81 In a
results.76 5-year period including fiscal year 2011 through fiscal year
Polymerase chain reaction followed by Southern blotting 2015, there were 18 transfusion-related fatalities related to
and a labeled probe have been used to confirm the presence contaminated blood products reported to the CDC. Three of
of treponemal antigen. The test is capable of detecting as few the reported fatalities were due to contamination of trans-
as one treponeme in CSF. Nontreponemal EIAs, fluorescent fused red blood cells with B. microti.82
treponemal antibody absorption (FTA-ABS), T. pallidum im-
Clinical Manifestations and Pathology
mobilization (TPI), and T. pallidum hemagglutination (TPHA)
are the methodologies utilized. Blood donations that are reac- Most cases of babesiosis are asymptomatic. Symptomatic
tive may not be used unless a confirmatory test such as FTA patients usually develop a malaria-type illness characterized
is determined to be nonreactive.30 Donors that have confirmed by fever, chills, lethargy, and hemolytic anemia. The risk for
positive results can be reinstated for donation after 12 months developing severe complications, which include renal failure,
with documentation of treatment.76 DIC, and respiratory distress syndrome, increases for elderly,
asplenic, or immunocompromised patients. Reported incu-
Tick-Borne Bacterial Agents bation periods for symptomatic patients range from 1 to
8 weeks after transfusion; therefore, it is important that
Lyme disease, Rocky Mountain spotted fever (RMSF), and physicians consider babesiosis when diagnosing a febrile
ehrlichiosis are all bacterial diseases spread by a tick bite. illness following a transfusion.83
Lyme disease is caused by the spirochete Borrelia burgdorferi,
and RMSF (Rickettsia rickettsii) and ehrlichiosis (Ehrlichia Epidemiology and Transmission
species) are caused by bacteria that are obligate intracellular Areas of the United States, such as the northeast, mid-
pathogens. Atlantic, and upper midwestern states are said to have
endemic transmission.79 The incidence is higher during
Transfusion-Associated Parasites the spring and summer months, which corresponds to the
increase in tick activity and outdoor recreation of humans.
At least three parasites have been associated with transfusion- Persons infected with Babesia may not have clinical signs
associated infections: Babesia microti, Trypanosoma cruzi, of illness for an extended time. Infected persons who
and malaria (Plasmodium species). Several additional para- donate blood during the asymptomatic period pose the
sites have been identified in association with transfusion- greatest risk to the blood supply, as they probably have
associated disease. These include Leishmania species, other infectious organisms circulating in their bloodstreams.
Trypanosoma species, Toxoplasma gondii, and the microfilarial Units of packed RBCs (liquid stored and frozen deglyc-
parasites. Most of these infections occur on rare occasions erolized) and platelet units, which contain RBCs, have been
and typically involve patients who are severely immunocom- associated with transmission.80 B. microti can survive in
promised. The risk for acquiring a blood transfusion contain- refrigerated, uncoagulated blood for 21 to 35 days. There
ing these parasites may be underreported in endemic areas were 63 transfusion-transmitted babesiosis cases in the
but has always been very low in the United States. However, United States between 2004 and 2008.79
in October 2003, the AABB put forth a recommendation to
blood collection facilities that all individuals who had been Laboratory Diagnosis
in Iraq should be deferred for 1 year from the last date of Prompt diagnosis is essential, as Babesia responds well to
departure. This was done after cases of leishmaniasis were antibiotic therapy but can be fatal in certain risk groups if
reported in personnel stationed in Iraq. not properly treated. There is no specific test to diagnose an
infection with B. microti. Thick and thin blood smears
Babesia microti stained with Giemsa or Wright stain can be examined for in-
traerythrocytic organisms. A single negative smear does not
Babesiosis, a zoonotic disease, is usually transmitted by the rule out an infection. Serologic studies such as immunoflu-
bite of an infected deer tick. Infection is caused by the proto- orescence assays can be used to detect circulating antibody.79
zoan parasite, Babesia, which infects the RBCs. Most human Currently there is no licensed screening test for blood
cases of Babesia infection that occur in the United States are donors. IND protocols for antibody and DNA testing are
caused by the B. microti parasite.77 There have been reported being used by several blood centers.84
cases of simultaneous transmission of B. microti and Borrelia
burgdorferi, the causative agent of Lyme disease, because the Prophylaxis and Treatment
tick vectors are the same for the two organisms.78 Other re- Babesiosis can be effectively treated with antibiotic therapy.
ported species are Babesia duncani, formally called WA1-type There is no specific drug of choice, but quinine and
Babesia, CA1-type Babesia, and Babesia divergens–like agents clindamycin are very effective. In addition, the combination
6888_Ch14_307-332 29/10/18 5:43 PM Page 323

Chapter 14 Transfusion-Transmitted Diseases 323

of atovaquone and azithromycin can be as effective in pa- disease can be transmitted congenitally, transplacentally, or
tients without a life-threatening illness.84 Apheresis has through solid organ transplantation.89
also been successful in patients who fail to respond to an- Screening for Chagas disease was implemented in January
tibiotic therapy.85,86 2007 using an FDA-approved ELISA test.89
There is no test currently available to screen for asymp-
tomatic carriers of Babesia. Many blood banks have added Laboratory Diagnosis
questions to their donor questionnaire that address topics Acute Chagas disease is diagnosed by detecting the organ-
such as living in an endemic area and previous Babesia in- ism in the patient’s blood. Blood smears stained with
fection. Some blood banks have chosen to defer individuals Giemsa or Wright stain may be examined for the charac-
who reside in areas that are heavily tick-infested in the sum- teristic C- or U-shaped trypomastigote (Fig. 14–7). Antico-
mer months.84 This practice may have little value, as donors agulated blood or the buffy coat may also be evaluated for
may remain asymptomatic for months after exposure to the motile organisms.
organism, and there have been transfusion-transmitted cases Chronic Chagas disease is diagnosed serologically. Such
reported in nonendemic areas. Donors with a history of testing includes complement fixation, immunofluorescence,
babesiosis should be deferred from donating blood for an in- and ELISA. False-positive reactions are common; therefore,
definite period of time.1 Because B. microti can be transmit- it is recommended that patient specimens be analyzed using
ted by blood donated from asymptomatic donors, effective more than one assay. Trypomastigotes are rare or absent in
measures for preventing transmission are needed. The AABB the peripheral blood during the chronic phase, so examina-
Transfusion Transmitted Disease (TTD) Committee has pri- tion of blood smears is not useful.
oritized babesiosis as an agent for which there is a critical
need for the development and implementation of an inter- Prophylaxis and Treatment
vention to reduce transfusion-associated infection.84 National screening of the blood supply was initiated in 2007.
Since that time, more than 10,000 donors with T. cruzi in-
Trypanosoma cruzi fection have been identified.89 The FDA approved a second
test to screen blood, tissue, and organ donors in April 2010.
T. cruzi is a flagellate protozoan that is the etiologic agent of
The test, called the Abbott Prism Chagas, is highly sensitive
Chagas disease (American trypanosomiasis). It is estimated
and specific for the detection of antibodies to T. cruzi.90
that 300,000 people are infected within the United States.87
The AABB TTD Committee has given T. cruzi an orange
The disease is naturally acquired by the bite of a reduviid bug,
category rating.56 An orange category agent is considered a
thus making it a zoonotic infection. Insect transmission is the
low scientific or epidemiological risk regarding blood
most common mode of infection, but the organism has also
saftey.56 T. cruzi was assigned a moderate rating by the TTD
been transmitted by blood transfusion and organ transplants.
Committee based on public and regulatory attention to in-
Clinical Manifestations and Pathology troducing blood donor screening.56
In the United States, medication for Chagas disease may
The acute phase of Chagas disease is initiated when the
organism enters the host. The reduviid bug bite produces a be obtained only by contacting the CDC.
localized nodule, referred to as a “chagoma.” The chagoma
is usually painful and may take up to 3 months to heal. Clin- Malaria (Plasmodium Species)
ical symptoms may be mild or absent; therefore, many cases Malaria, another intraerythrocytic protozoan infection,
are not diagnosed until the chronic phase of the disease. may be caused by several species of the genus Plasmodium
Symptoms include anemia, weakness, chills, intermittent
fever, edema, lymphadenopathy, myocarditis, and gastroin-
testinal symptoms. Death may occur within a few weeks or
months after initial infection.
Following the acute phase, the disease may enter a latent
phase, which can last up to 40 years.88 During this phase,
the patient is usually asymptomatic but has parasites circu-
lating in the bloodstream. Transfusion-associated Chagas
disease is most likely to occur during this phase.
Chagas disease usually progresses to the chronic phase
years or decades after the acute phase.88 In the chronic
phase, the organism begins to cause damage to cardiac tissue,
thus causing cardiomyopathy.

Epidemiology and Transmission


Chagas disease is endemic in Central and South America
and some areas of Mexico. Cases have also been reported in
the southern United States.89 Infected individuals pose a risk
of infecting the recipients of their donated blood. Chagas Figure 14–7. T. cruzi trypomastigote.
6888_Ch14_307-332 29/10/18 5:43 PM Page 324

324 PART III Transfusion Practices

(Plasmodium malaria, Plasmodium falciparum, Plasmodium appropriate therapy and decrease the chance of resistance
vivax, and Plasmodium ovale). Natural transmission occurs by the organism.
through the bite of a female Anopheles mosquito, but in- Some individuals have a natural immunity to certain
fection may also occur following transfusion of infected species of malaria caused by a genetic alteration in their
blood. Malaria is very rare in the United States. One hun- RBCs. These include persons who have sickle cell anemia or
dred and one cases of transfusion-transmitted malaria have trait, G6PD deficiency, or RBCs that lack the Duffy blood
been reported from 1990 to 2005, making the incidence group antigen.
rate of 0.1 cases per 1 million units transfused.91
Prion Disease
Clinical Manifestations and Pathology
Symptoms include fever, chills, headache, anemia, hemoly- Creutzfeldt-Jakob Disease
sis, and splenomegaly. There may be variations in symptoms
among the different species of Plasmodium. Malaria often Creutzfeldt-Jakob disease (CJD) is one of the transmissible
mimics other diseases, and its diagnosis is often delayed due spongiform encephalopathies (TSEs). These are rare diseases
to lack of suspicion in nonendemic areas. characterized by fatal neurodegeneration that results in
sponge-like lesions in the brain. Although a definitive diag-
Epidemiology and Transmission nosis can be made only at autopsy, neurological signs and
Malaria is endemic in tropical and subtropical areas and in symptoms and disease progression are used to make a pre-
West Africa. The World Health Organization (WHO) esti- liminary diagnosis. Animals, such as sheep, goats, cattle,
mated that in 2008 malaria caused 190 to 311 million clini- cats, minks, deer, and elk, and humans can be affected by
cal episodes and 708,000 to 1,003,000 deaths.92 Many people TSE. In humans, sporadic CJD is the most common form,
associate malaria with a history of traveling to an endemic representing 85% to 90% of all cases, generally occurring in
area. However, other transmission modes are possible, late middle age (average age 60 years). An inherited form
including blood transfusions and congenital infection. due to a gene mutation accounts for another 5% to 10% of
Transfusion-associated malaria is acquired by receiving cases, and iatrogenic CJD acquired through contaminated
blood products from an asymptomatic carrier. Plasmodium can neurosurgical equipment, cornea or dura mater transplants,
survive in blood components stored at room temperature or or human-derived pituitary growth hormones accounts for
4°C for at least a week, and deglycerolized RBCs can transmit less than 5% of cases. The sporadic, inherited, and iatrogenic
disease. CJD are considered the classic CJD.94 In 1996, a variant form
of CJD (vCJD) affecting younger individuals was noted, and
Laboratory Diagnosis epidemiological evidence linked vCJD to bovine spongiform
Examination of thick and thin blood smears is performed to encephalopathy, possibly from eating contaminated beef. Of
diagnose infection with malaria. Although each species of the 129 cases reported from 1996 to 2002, most were in the
Plasmodium varies morphologically, diagnosis can be quite United Kingdom.94
difficult. Depending on the species of Plasmodium and the The causative agent of all TSEs is believed to be a prion,
stage of the parasite’s life cycle, timing is crucial when eval- which is described as a self-replicating protein. It does not
uating the blood smear. A single negative smear does not rule contain nucleic acid but is formed when the confirmation of
out a diagnosis of malaria. the normal cell surface glycoprotein, the prion protein, is
changed to an abnormal form. This abnormal form accumu-
Prophylaxis and Treatment lates in the brain and makes the brain tissue highly infectious.
A practical or cost-effective serologic test to screen asymp- It is resistant to inactivation by heat, radiation, and formalin.94
tomatic donors does not exist. According to FDA guidance The median duration of illness for vCJD is 13 to
documents, persons who have traveled to an endemic area 14 months.94 However, the incubation period in humans
are deferred for 1 year, and those who have had malaria or varies from 4 to 20 years and may eventually prove to be
who have immigrated from or lived in an endemic area are longer in some cases.
deferred for 3 years.93 There is no epidemiological evidence linking classic CJD
Chloroquine is generally effective for chemoprophylaxis to TTD. However, in vCJD cases, prion particles have been
and treatment of all four species of Plasmodium, except found in lymphoreticular tissues, including the tonsils,
P. vivax acquired in Indonesia or Papua New Guinea, which spleen, and lymph nodes. As blood is intimately involved
is best treated with atovaquone-proguanil, with mefloquine with the lymphoreticular system, concerns arose regarding
or quinine plus tetracycline or doxycycline as alterna- the ability of vCJD individuals to transmit the prion to re-
tives.92 Therapy has become more complicated due to the cipients of blood or blood products.94
increase in resistance of P. falciparum and, more recently, Currently, there is no reliable diagnostic test that can de-
P. vivax to chloroquine. It is important for the physician tect asymptomatic individuals. Therefore, deferral of donors
to carefully evaluate the species of Plasmodium causing with connection to the United Kingdom and parts of Europe
the illness, the estimated parasitemia, and the patient’s as well as other associated risk factors is used to prevent
travel history. This information is necessary to prescribe transmission.94
6888_Ch14_307-332 29/10/18 5:43 PM Page 325

Chapter 14 Transfusion-Transmitted Diseases 325

Pathogen Inactivation Cellular Components

The safety of the blood supply in the United States has im- Pathogen inactivation using psoralen activated by ultraviolet
proved greatly over the years, with improved screening of light has been tested with platelet concentrates. It has been
donors and testing of the blood product. However, pathogen shown to inactivate cell-associated viruses, cell-free viruses,
inactivation methods have been developed to account for and selected prokaryotic organisms. Whether this process
residual risks associated with serologic window periods, will work against intracellular bacterial organisms has not
virus variants, and laboratory errors and for organisms for been established. There are three licensed platelet pathogen
which testing is not performed routinely.73 The possibility reduction systems that are currently in use in the United
of newly emerging pathogens also exists, as evidenced States and Canada: the Cerus Corporation’s INTERCEPT
by WNV, DENV, ZIKV, and DENV, that can be transmitted Blood System, CaridianBCT Biotechnologies’ Mirasol PRT,
by blood. and the MacoPharma’s Theraflex UV.62 These companies also
have processes for pathogen reduction in plasma.56
Plasma Derivatives CaridianBCT Biotechnologies is currently using a photo-
chemical process for red blood cell pathogen reduction. This
Heat inactivation, the first pathogen inactivation interven- system incorporates riboflavin and UV light.56 A process for
tion, has been used since 1948 to treat albumin.3 Even RBCs that uses a chemical cross-linker specific for nucleic
before the introduction of third-generation testing for acid is being designed by Cerus Corporation.56
HBsAg, heat inactivation prevented the transmission of Limitations of pathogen reduction systems include agents
HBV. The transmission of viruses or bacteria has been pre- with intrinsic resistance, such as prions, some bacterial spores,
vented due to albumin’s pasteurization method (60°C for and nonenveloped viruses.56 Some viruses may not be inacti-
10 hours).30 vated if they are of high titer, including B19V and HBV.56
In 1973, third-generation assays for HBsAg were li-
censed. Only one case of HBV transmission by immune Quarantine and Recipient Tracing
globulin was ever documented before then. Intramuscular (Look-Back)
immune globulin has never transmitted HIV or HCV. All
immunoglobulin plasma pools were screened for HBsAg, All blood banks and transfusion services are required to have
and only those that were negative were used. Viral inacti- a process to detect, report, and evaluate any complication of
vation included cold-ethanol (Cohn-Oncley) fractionation transfusion, including recipient development of HBV, HCV,
and anion-exchange chromatography (for one IV im- HIV, or HTLV. There must be an established method to notify
munoglobulin). However, in 1994, the FDA required viral donors of any abnormality with the predonation evaluation,
clearance processing or proof of absence of HCV by NAT laboratory testing, or recipient follow-up. A report should
testing because of outbreaks of HCV from anion-exchange be submitted to the collecting agency when the recipient of
chromatography in Ireland and Germany, countries that did a blood component develops a TTD.1
not use a viral clearance procedure. NAT is now used in the Current donations that test positive for HBV, HCV, HIV, or
processing of all source plasmas.30 HTLV cannot be used for transfusion.1 All prior donations
Coagulation factors had a high rate of viral transmission from these donors become suspect. The timeline and stan-
until the early 1980s. Chronic hepatitis was the biggest prob- dards using the look-back procedure to identify recipients of
lem until HIV emerged. More than 50% of all hemophiliacs a component from the implicated donation or other donations
receiving concentrates became infected with HIV. Since by the same donor differ depending on the disease. Any prior
1987, these clotting factors have become very safe due to im- components still in-date must be quarantined, and the dispo-
plementation of a variety of virus inactivation steps, and sition depends on results of licensed supplemental tests.2
there have been no cases of HIV transmission. Today, all If on recipient follow-up it is noted that a patient devel-
manufacturers use methods that either remove the virus or oped HBV, HCV, HIV, or HTLV after receiving a single unit
inactivate it. The lipid-enveloped viruses—HIV, HBV, HCV, from one donor, that donor is permanently deferred. If the
HTLV, EBV, CMV, HHV-6, and HHV-8—are all inactivated by recipient received donations from several donors, all donors
use of organic solvents and detergents. This process is not do not have to be excluded. These implicated donors may
effective with non–lipid-enveloped viruses such as HAV and be called in for retesting. If a donor has been implicated in
parvovirus B-19.30,95 more than one case of TTD, this donor should be retested
The current risk of enveloped virus transmission is very and possibly permanently deferred.2 Once a donor has been
low because of the combination of procedures such as heat implicated in a TTD, other recipients of a component from
treatment, solvent and detergent treatment, and nanofiltra- the suspected donor should be contacted. The donor must
tion.3 These methods are often used in combination during be placed on the appropriate donor deferral list if subsequent
the manufacturing process. With the exception of one case tests are positive.2 Donors who have been permanently de-
of HCV transmission in IV immune globulin in 1994, there ferred due to positive test results must be notified of the fact.
have been no cases of HBV, HCV, or HIV since 1985 by any Notification and a thorough explanation of the positive test
U.S. licensed plasma derivative.95 results and their implications must be given to the donor.2
6888_Ch14_307-332 29/10/18 5:43 PM Page 326

326 PART III Transfusion Practices

Follow-up testing should be performed by the donor’s own abnormalities must be transmitted to the patient and the
physician.2 patient’s physician.1
Autologous donations positive for HBV, HCV, HIV, HTLV, Any fatalities due to a TTD must be reported to the direc-
or syphilis can be used. If they are not transfused at the col- tor of the Center for Biologics Evaluation and Research
lecting facility, the collecting facility must notify the trans- within 1 working day, followed by a written report within
fusion service. Testing must be repeated every 30 days 7 days.96 Table 14–5 summarizes the laboratory tests for
on at least the first unit to be shipped. Information about transfusion-transmitted diseases.

Table 14–5 Summary of Laboratory Tests for Transfusion-Transmitted Diseases


Date Test Confirmatory Risk of
Disease Testing Implemented Method Test Method Transmission
Hepatitis B Virus (HBV) B surface antigen 1971 ChLIA Antigen neutralization
(HBsAq)

Hepatitis B core 1986 ChLIA Ultrasensitive HBV


antibody (HBc) DNA detection by
PCR

HBV DNA 2009 NAT All TMA-reactive do- 1 in 800,000 and 1 in


nations confirmed 1,000,00029
by PCR

Human T-Lymphotropic Qualitative 1998 ChLIA Second licensed HTLV-I less than 1 in
Virus (HTLV-I/II) antibody screening test 2 million29
detection for and WB
HTLV-II not yet
both HTLV-I and
proven unequivo-
HTLV-II in a
cally to be of signifi-
combined test
cant clinical concern

Hepatitis C Virus (HCV) Antibody testing 1990 ELISA, ChLIA Second licensed
screening test

HCV RNA 1999 NAT (using NAT 1 in 1,000,00029


TMA in
minipools
of 16…)

Human Immunodeficiency Antibody testing 1985 EIA, ChLIA One or a combination


Viruses, Types 1 and 2 of HIV-1 IFA and
(HIV 1, 2) HIV-2 EIA (a rapid
diagnostic test used
for HIV-1 and HIV-2
differentiation)

NAT 1999 NAT (using 1 in 1,000,00029


TMA in
minipools
of 16…)

Chagas Disease (T. cruzi) Antibody testing— 2007 ELISA, ChLIA ESA 20 reported cases in
qualitative the literature,
detection of worldwide
antibodies to
T. cruzi

Syphilis (T. pallidum) Antibody testing— 1950s Agglutination EIA, as well as a 0 (no cases of
qualitative assay test for reagin (a transfusion-
screening test protein-like sub- transmitted
detects pres- stance that is pres- syphilis recorded
ence of ent during acute in last 50 years)
antibodies to infection and for
T. pallidum several months
following resolution
of infection)
6888_Ch14_307-332 29/10/18 5:43 PM Page 327

Chapter 14 Transfusion-Transmitted Diseases 327

Table 14–5 Summary of Laboratory Tests for Transfusion-Transmitted Diseases—cont’d


Date Test Confirmatory Risk of
Disease Testing Implemented Method Test Method Transmission
West Nile Virus (WNV) WNV RNA 2003 NAT (same 1 in 84 million
detection type of assay
as used for
HBV, HIV-1,
and HCV)

Babesiosis (Babesia Antibody testing– 2012 IFA PCR and WB


microti) IND protocols

NAT–IND 2012 NAT PCR and WB 1 in 100,000


protocols

Zika Virus Zika virus RNA– 2016 NAT PCR and antibody Not yet determined
IND protocols testing

ChLIA = chemiluminescent immunoassay; NAT = nucleic acid testing; PCR = polymerase chain reaction; TMA = transcription-mediated amplification; ELISA = Enzyme-Linked,
Immunosorbent Assay Test System; WB = Western Blot Assay; EIA = Enzyme Immunoassay; IFA = Indirect Immunofluorescence Assay; ESA = Enzyme Strip Assay. (American Red
Cross, Infectious Disease Testing. Available from: http://www.redcrossblood.org/learn-about-blood/what-happens-donated-blood/blood-testing.)

SUMMARY CHART
 The first and most important step in ensuring that  Transfusion-associated CMV infection is a concern
transfused blood will not transmit a pathogenic virus for seronegative allogeneic organ transplant recipi-
is careful selection of the donor. ents and fetuses. Reactivation of a latent infection
 HAV is usually spread by the fecal-oral route in com- can occur when an individual becomes severely
munities where hygiene is compromised. immunocompromised.
 On infection with HBV, the first positive test is HBV  The risk of CMV infection for low-birth-weight
NAT and the first serologic marker to appear is HBsAg, neonates is not as great as it was in the past due to bet-
followed by HBeAg and IgM anti-HBc within the first ter transfusion techniques and management of their
few weeks of exposure. conditions.
 HBIG is an immune globulin prepared from persons  The WB confirmation test detects the presence of anti-
with a high titer of anti-HBs and is used to provide pas- HIV and determines with which viral proteins the an-
sive immunity to health-care workers and others who tibodies react.
are exposed to patients with HBV infection.  The window period for HIV can be shortened by using
 A combined vaccine for HAV and HBV is available to the polymerase chain reaction, which detects HIV in-
provide immunity. fection before tests for antigen or antibody are positive.
 HDV infection is common among drug addicts and can  Bacterial contamination is the most frequent cause of
occur simultaneously with HBV infection; diagnosis transfusion-transmitted infection.
depends on finding anti-HDV or HDV RNA in the  Because routine screening for parasitic infections is not
serum. currently available, many blood banks have added
 Of all HCV infections, 60% to 70% are asymptomatic. questions to their donor questionnaire that address
With the implementation of NAT testing for HCV, the topics associated with risk for parasitic infection.
window period has been reduced to 10 to 30 days.  Pathogen inactivation methods are in use for plasma
 HCV is the leading cause of liver transplants in the and platelet products and under development for red
United States. cell products. These methods remove or reduce the
 HEV is an emerging agent associated with transmission residual risk of transfusion-associated disease due to
through transfusion and is the leading cause of hepa- the window period, virus variants, laboratory mistakes,
titis in the United Kingdom. and new, emerging diseases.
 Diagnosis of HIV-1 and HIV-2 infection is dependent  Look-back is a process mandated by the FDA that di-
on the presence of antibodies to both envelope and rects collection facilities to notify donors who test pos-
core proteins; HIV-positive persons with fewer than itive for viral markers, to notify prior recipients of the
200 CD4+ T cells per microliter are considered to have possibility of infection, and to quarantine or discard
AIDS in the absence of symptoms. implicated components currently in inventory.
6888_Ch14_307-332 29/10/18 5:43 PM Page 328

328 PART III Transfusion Practices

9. HBV is transmitted most frequently:


Review Questions
a. By needle sharing among IV drug users
1. The fecal-oral route is common in transmitting which of b. Through blood transfusions
these hepatitis viruses? c. By unknown methods
d. By sexual activity
a. HAV and HEV
b. HBV and HCV 10. Which of the following is the most common cause of
c. HDV chronic hepatitis, cirrhosis, and hepatocellular carci-
d. HGV noma in the United States?
2. Which of the following is the component of choice for a a. HAV
low-birth-weight infant with a hemoglobin of 8 g/dL if b. HBV
the mother is anti-CMV negative? c. HCV
d. HDV
a. Whole blood from a donor with anti-CMV
b. RBCs from a donor who is anti-CMV negative 11. The first retrovirus to be associated with human dis-
c. Leukoreduced platelets ease was:
d. Solvent detergent–treated plasma a. HCV
3. Which of the following is an FDA-licensed screening test b. HIV
for HCV? c. HTLV-I
d. WNV
a. NAT + anti-HBc
b. RIBA 12. All of the following statements are true concerning
c. Lymph node biopsy WNV except:
d. HCV RNA a. 1 in 150 infections results in severe neurological
4. Currently, which of the following does the AABB con- disease
sider to be the most significant infectious threat from b. Severe disease occurs most frequently in the over-
transfusion? 50 age group
c. Deaths occur more often in those over 65 years who
a. Bacterial contamination
present with encephalitis
b. CMV
d. Fatalities occur in approximately 38% of infected
c. Hepatitis
individuals
d. HIV
13. The primary host for WNV is:
5. Which of the following is the most frequently transmitted
virus from mother to fetus? a. Birds
b. Horses
a. HIV
c. Humans
b. Hepatitis
d. Bats
c. CMV
d. EBV 14. Tests for WNV include all of the following except:
6. Jaundice due to HAV is seen most often in the: a. ELISA
b. NAT
a. Adolescent
c. Plaque reduction neutralization test
b. Adult
d. Immunofluorescent antibody assay
c. Child younger than 6 years old
d. Newborn 15. Individuals exposed to EBV maintain an asymptomatic
latent infection in:
7. Currently, steps taken to reduce transfusion-transmitted
CMV include: a. B cells
b. T cells
a.Plaque reduction neutralization test
c. All lymphocytes
b.NAT testing
d. Monocytes
c.Leukoreduction
d.Minipool screening 16. Fifth disease is caused by:
8. HBV remains infectious on environmental surfaces for 1: a. CMV
b. EBV
a. Day
c. Parvovirus B19
b. Week
d. HTLV-II
c. Month
d. Year
6888_Ch14_307-332 29/10/18 5:43 PM Page 329

Chapter 14 Transfusion-Transmitted Diseases 329

17. Transient aplastic crisis can occur with: 25. Which disease is naturally caused by the bite of a deer
a. Parvovirus B19 tick?
b. WNV a. Chagas disease
c. CMV b. Babesiosis
d. EBV c. Malaria
d. Leishmaniasis
18. Reasons why syphilis is so rare in the U.S. blood supply
include all of the following except:
a. 4°C storage conditions References
b. Donor questionnaire 1. American Association of Blood Banks. Standards for blood
c. Short spirochetemia banks and transfusion services. 31st ed. Bethesda (MD): AABB;
d. NAT testing 2018.
2. Fung MK, Eder AF, Spitalnik SL, Westhoff CM (eds). Technical
19. Nucleic acid amplification testing for HIV was instituted manual. 19th ed. Bethesda (MD): American Association of
in donor testing protocols to: Blood Banks; 2017.
3. Cuthbert JA. Hepatitis A: old and new. Clin Microbiol Rev.
a. Identify donors with late-stage HIV who lack 2001;14(1):38-58.
antibodies 4. Centers for Disease Control and Prevention [Internet]: Atlanta
b. Confirm the presence of anti-HIV in asymptomatic (GA): The CDC [cited 2017 Jul 13]. Surveillance for viral
HIV-infected donors hepatitis—United States, 2014. Available from: www.cdc.gov/
hepatitis/statistics/2014survaillance/index.htm.
c. Reduce the window period by detecting the virus 5. Centers for Disease Control and Prevention [Internet]: Atlanta
earlier than other available tests (GA): The CDC [cited 2017 May 15]. Hepatitis A – questions
d. Detect antibodies to specific HIV viral proteins, and answers for health professionals. Available from: www
including anti-p24, anti-gp41, and anti-gp120 .cdc.gov/hepatitis/hav/havfaq.htm#general.
6. Freidl GS, Sonder GJ, Bovee LP, Friesema IH, van Rljckevorsel RL,
20. Screening for HIV is performed using the following Ruijs WL, et al. Hepatitis A outbreak among men who have sex
technique: with men (MSM) predominantly linked with the EuroPride in
the Netherlands. July 2016 to February 2017. Euro Surveil.
a. Radio immunoassay 2017;22(8):30468.
b. WB 7. Centers for Disease Control and Prevention [Internet]: Atlanta
c. Immunofluorescent antibody assay (GA): The CDC [cited 2017 May 11]. Statistics and surveillance.
d. NAT Available from: www.cdc.gov/hepatitis/statistics/indexhtm.
8. Bishop ML, Fody EP, Schoeff LE. Clinical chemistry tech-
21. The first form of pathogen inactivation was: niques, principles and correlations. 6th ed. Baltimore (MD):
a. Chemical Lippincott Williams & Wilkins; 2010.
9. Miller LE. Serology of viral infections. In: Stevens CD, editor.
b. Heat Clinical immunology and serology: a laboratory perspective.
c. Cold-ethanol fractionation 2nd ed. Philadelphia: FA Davis Company; 2003. p. 324.
d. Anion-exchange chromatography 10. Food and Drug Administration [Internet]: Rockville (MD):
FDA [cited 2017 May 13]. Guidance for industry: use of
22. What is the most common parasitic complication of nucleic acid tests on pooled and individual samples from donors
transfusion? of whole blood and blood components, including source
a. Babesia microti plasma, to reduce the risk of transmission of hepatitis B virus.
Available from: www.fda.gov/BiologicsBlood Vaccines/Guidance
b. Trypanosoma cruzi ComplianceRegulatoryInformation/Guidances/Blood/ucm32750
c. Plasmodium species .htm.
d. Toxoplasma gondii 11. Lox ASF, McMahon BF. AASLD practice guidelines: chronic
hepatitis B. Hepatology. 2001;34:1225.
23. Which organism has a characteristic C- or U-shape on 12. Marcellin P, Chang T, Lim SG, Tong MJ, Sievert W, Shiffman ML,
stained blood smears? et al. Adefovir dipivoxil for treatment of hepatitis B antigen-
a. Trypanosoma cruzi positive chronic hepatitis B. N Engl J Med. 2003;348:808.
13. Gerlach JT, Diepolder HM, Zachoval R, Gruener NH, Jung MC,
b. Plasmodium vivax Ulsenheimer A, et al. Acute hepatitis C: high rate of both spon-
c. Plasmodium falciparum taneous and treatment-induced viral clearance. Gastroenterol-
d. Babesia microti ogy. 2003;125:80-8.
14. Ankcorn MJ, Tedder RS. Hepatitis E the current state of play.
24. Which transfusion-associated parasite may have asymp- Transfus Med. 2017;27(2):84-95.
tomatic carriers? 15. Centers for Disease Control and Prevention [Internet]: Atlanta
a. Babesia microti (GA): The CDC [cited 2010 Aug 9] Viral hepatitis–Hepatitis E
virus. Available from: www.cdc.gov//hepatitis/HEV?HEVfaq
b. Trypanosoma cruzi .htm#section1.
c. Plasmodium species 16. Alter HJ, Nakatsuji Y, Melpolder J, Wages J, Wesley R, Shih JW,
d. All of the above et al. The incidence of transfusion associated hepatitis G virus
6888_Ch14_307-332 29/10/18 5:43 PM Page 330

330 PART III Transfusion Practices

infection and its relation to liver disease. N Engl J Med. cases and deaths reported to CDC by year and clinical presen-
1997;336:747. tation 1999–2015. Available from: www.cdc.gov/westnile/
17. Heringlake S, Osterkamp S, Trautwein C, Tillmann HL, Boker K, resources/pdfs/data/1-WNV-Disease-Cases-by-year_1999-
Muerhoff S, et al. Association between fulminant hepatic failure 2015_07072016.pdf.
and a strain of GB virus C. Lancet. 348:1996;1626. 36. Prowse CV. An ABC for West Nile virus. Transfus Med.
18. Halasz R, Weiland O, Sallberg M. GB virus C/hepatitis G virus. 2003;13:1.
Scand J Infect Dis. 2001;33:572. 37. Centers for Disease Control and Prevention. Acute flaccid
19. Nunnari G, Nigro L, Palermo F, Attanasio M, Berger A, Doerr HW, paralysis syndrome associated with West Nile virus infection—
et al. Slower progression of HIV-1 infection in persons with Mississippi and Louisiana, July–August 2002. MMWR. 2002;
GB virus C coinfection correlates with an intact T-helper 51:825.
1 cytokine profile. Ann Intern Med. 2003;139:26. 38. Centers for Disease Control and Prevention [Internet]: Atlanta
20. Zanetti AR, Tanzi E, Romano L, Principi N, Zuin G, Minola E, (GA): The CDC [cited 2010 Aug 13]. West Nile virus:
et al. Multicenter trial on mother-infant transmission of Overview of West Nile virus. Available from: www.cdc.gov/
GBV-virus. J Med Virol. 1998;54:107. ncidod/dvbid/westnile/qa/overview.htm.
21. Sathar MA, Soni PN, York D. GB virus C/hepatitis G virus 39. Centers for Disease Control and Prevention [Internet]: Atlanta
(GBV-C/HGV): still looking for a disease. Int J Exper Pathol. (GA): The CDC [cited 2017 Jul 14]. West Nile virus in the
2000;81:305. United States: guidelines for surveillance, prevention and con-
22. Jarvis LM, Bell H, Simmonds P, Hawkins A, Hellum K, Harthug S, trol. Available from: www.cdc.gov/westnile/resources/pdfs/
et al. The effect of treatment with alpha-interferon on hepatitis WNVGuidelines.pdf.
G/GBV-C viraemia. The CONSTRUCT Group. Scand J Gas- 40. Petersen LR, Marfin AA. West Nile virus: a primer for clini-
troenterol. 1999;33:195. cians. Ann Intern Med. 2002;137:173.
23. Centers for Disease Control and Prevention [Internet]: At- 41. Food and Drug Administration [Internet]: Rockville (MD): FDA.
lanta (GA): The CDC [cited 2017 Jul 14]. HIV-2 infection Guidance for industry: use of nucleic acid tests to reduce the risk
surveillance—United States, 1987–2009. Available from: www of transmission of west Nile virus from donors of whole blood
.cdc.gov/mmwr/preview/mmwrhtml/mm6029a3.htm. Accessed and blood components intended for transfusion. Available from:
July 14, 2017. www.fda.gov/BiologicsBloodVaccines/GuidanceCompliance
24. American Red Cross [Internet]: Washington (DC): American RegulatoryInformation/UCM189464.pdf
Red Cross [cited 2017 May 10]. Infectious disease testing. 42. Busch MP, Caglioti S, Robertson EF, McAuley JD, Tobler LH,
Available from: www.redcross/blood.org/learn-about-blood/ Kamel H, et al. Screening the blood supply for West Nile virus
blood-testing. RNA by nucleic acid amplification testing. N Engl J Med.
25. Food and Drug Administration [Internet]: Rockville (MD): FDA 2005;353:460–7.
[cited 2017 May 13]. Guidance for industry: nucleic acid testing 43. Jimenez A, Shaz BH, Bloch E. Zika virus and the blood supply:
(NAT) for human immunodeficiency virus type 1 (HIV-1) and what do we know? Transfu Med Rev. 2017;31:1-10.
hepatitis C virus (HCV): testing, product disposition, and donor 44. Food and Drug Administration [Internet]: Rockville (MD): FDA
deferral and reentry. Available from: www.fda.gov/Biologics- [cited 2017 May 13]. FDA guidance for the industry: recommen-
BloodVaccines/GuidnaceComplianceRegulatoryInfromation/ dations for donor screening, deferral, and product management
Guidances/Blood/ucm210270.pdf. to reduce the risk of transfusion-transmission of zika virus.
26. Centers for Disease Control and Prevention [Internet]: Atlanta Available from: www.fda.gov.BiologicsBloodVaccines/Guidance
(GA): The CDC [cited 2010 Aug 11] . Advancing HIV preven- ComplianceRegulatoryInformation/Guidances/Blood/ucm518213
tion: new strategies for a changing epidemic—United States, .pdf.
2005. Available from: www.cdc.gov/hiv/topics/prev_prog/AHP/ 45. Centers for Disease Control and Prevention [Internet]: Atlanta
default.htm. (GA): The CDC [cited 2017 May 30]. Zika virus 2016 case
27. Centers for Disease Control and Prevention [Internet]: Atlanta counts in the US. Available from: www.cdc.gov/zika/reporting/
(GA): The CDC [cited 2017 Jul 14]. Vital signs: HIV diagnosis, 2016-case-counts.html.
care and treatment among persons living with HIV–United States, 46. Centers for Disease Control and Prevention [Internet]: Atlanta
2011. Available from: www.cdc.gov/mmwr/preview/mmwrhtml/ (GA): The CDC [cited 2017 Jul 15]. Zika virus outcomes of
mm6347a5.htm. pregnancies with laboratory evidence of possible zika virus in-
28. Manns A, Hisada M, La Grenade L. Human T-lymphotropic fection in the United States and the US territories. Available
virus type I infection. Lancet. 1999;353:1951. from: www.cdc.gov/zika/reporting/preganancy-outsomes.html.
29. Matsuoka M. Human T-cell leukemia virus type I and adult 47. Centers for Disease Control and Prevention [Internet]: Atlanta
T-cell leukemia. Oncogene. 2003;22:5131. (GA): The CDC [cited 2017 Jul 15]. Zika virus prevention.
30. Roback JD, editor. Technical manual. 16th ed. Bethesda (MD): Available from: www.cdc.gov/zika/prevention/index.html.
American Association of Blood Banks; 2008. 48. Centers for Disease Control and Prevention [Internet]: Atlanta
31. Nagai M, Osame M. Human T-cell lymphotropic virus type I (GA): The CDC [cited 2017 Jul 15]. Cytomegalovirus (CMV)
and neurological diseases. J Neurovirol. 2003;9:228. and congenital CMV infection. Available from: www.cdc.gov/
32. Murphy EL, et al. Increased prevalence of infectious diseases cmv/overview.html.
and other adverse outcomes in human T lymphotropic virus 49. Zlemann M, Thiele T. Transfusion-transmitted CMV infection—
types I- and II-infected blood donors. J Infect Dis. 1997; current knowledge and future perspectives. Transfus Med.
176:1468. 2017 Aug;27(4):238-48.
33. American Association of Blood Banks [Internet]: Bethesda (MD): 50. Ely JW, Yankowitz J, Bowdler NC. Evaluation of pregnant
American Association of Blood Banks [cited 2010 Aug 11]. Dual women exposed to respiratory viruses. Am Fam Physician.
enzyme immunoassay (EIA) approach for deferral and notifica- 2000 May 15;61(10)3065-74.
tion of anti-HTLV-I/II EIA reactive donors. Association Bulletin 51. Centers for Disease Control and Prevention [Internet]: Atlanta
#99–9. Available from: www.aabb.org/members_only/archives/ (GA): The CDC [cited 2010 Aug 16]. Epstein-Barr virus and
association_bulletins/ab99–9.htm. infectious mononucleosis. May 16, 2006. Available from:
34. Petersen LR, Marfin AA, Gubler DJ. West Nile virus. JAMA. www.cdc.gov/ncidod/diseases/ebv.htm.
2003;290:524. 52. Tattevin P, Cremieux AC, Descamps D, Carbon C. Transfusion-
35. Centers for Disease Control and Prevention [Internet]: Atlanta related infectious mononucleosis. J Infect Dis 34:777,
(GA): The CDC [cited 14 Jul 2017]. West Nile virus disease 2002.
6888_Ch14_307-332 29/10/18 5:43 PM Page 331

Chapter 14 Transfusion-Transmitted Diseases 331

53. Epstein MA, Achong BG, Barr YM. Virus particles in cultured risk. Available from: http://www.fda.gov/downloads/Biologics-
lymphoblasts from Burkitts’s lymphoma. Lancet. 1964;1:702. BloodVaccines/GuidanceComplianceRegulatoryInformation/
54. American Association of Blood Banks [Internet]: Bethesda Guidances/Blood/UCM425952.pdf.
(MD): American Association of Blood Banks [cited 2017 70. Centers for Disease Control and Prevention [Internet]: Atlanta
Jul 15] Human parvovirus B19. Available from: www.aabb.org/ (GA): The CDC [cited 2017 Jul 15]. Blood safety overview.
tm/eid/Documents/Human-Parvovirus-B19.pdf. Available from: https://www.cdc.gov/bloodsafety/bbp/diseases-
55. Weimer T, Streichert S, Watson C, Groner A. High-titer screen- organisms.html#bcbp.
ing PCR: a successful strategy for reducing the parvovirus B19 71. Centers for Disease Control and Prevention. Red blood cell
load in plasma pools for fractionation. Transfusion. 2001; transfusions contaminated with Yersinia enterocolitica—United
41:1500. States, 1991–1996, and initiation of a national study to detect
56. Stramer SL, Hollinger FB, Katz LM, Kleinman S, Metzel PS, bacteria-associated transfusion reactions. MMWR, 1997;
Gregory KR, et al. Emerging infectious disease agent supple- 46:553.
ment and their potential threat to transfusion safety. Transfu- 72. Kunishima S, Inoue C, Kamiya T, Ozawa K. Presence of Propi-
sion. 2009;49: Suppl 2:1S-29S. onibacterium acnes in blood components. Transfusion.
57. Food and Drug Administration [Internet]: Rockville (MD): 2001;41:1126.
FDA. Guidance for industry: nucleic acid testing (NAT) to 73. Eder AF, Kennedy JM, Dy BA, Notari EP, Weiss JW, Fang CT,
reduce the possible risk of parvovirus B19 transmission by et al. Bacterial screening of apheresis platelets and the residual
plasma-derived products. Available from: www.FDA.gov/ risk of septic transfusion. The American Red Cross Experience
BiologicsBlood Vaccines/Guidance Compliance Regulatory (2004–2006). Transfusion. 2007;47:1134-42.
Information/Guidance/Blood/ucm071592.htm. 74. de Korte D, Marcelis JH, Verhoeven AJ, Soeterboek AM. Diver-
58. Hladik W, Dollard SC, Mermin J, Fowlkes AL, Downing R, sion of first blood volume results in a reduction of bacterial
Amin MM, et al. Transmission of human herpesvirus-8 by contamination for whole blood collections. Vox Sang. 2002;
blood transfusion. N Engl J Med. 2006;355:1331-8. 83:13.
59. Pellett PE, Wright DJ, Engels EA, Ablashi DV, Dollard SC, 75. Centers for Disease Control and Prevention [Internet]: Atlanta
Forghani B, et al. Multicenter comparison of serologic assays (GA): The CDC [cited 2017 Jul 15]. 2015 Sexually transmitted
and estimation of human herpesvirus 8 seroprevalence among diseases surveillance. Syphilis. Available from: https://www
US donors. Transfusion. 2003;43:1260. .cdc.gov/std/stats15/syphilis.htm.
60. American Association of Blood Banks [Internet]: Bethesda 76. Food and Drug Administration [Internet]: Rockville (MD):
(MD): American Association of Blood Banks [cited 2017 FDA [cited 2017 Jul 15]. Guidance for industry: recommenda-
Jul 14]. Chikungunya virus. Available from: www.aabb.org/ tions for screening, testing and management of blood donors
tm/eid/documents/chikungunya-virus.pdf. and blood and blood components based on screening tests for
61. Centers for Disease Control and Prevention [Internet]: Atlanta syphilis. Available from: www.fda.gov/BiologicsBloodVaccines/
(GA): The CDC [cited 2017 Jul 15]. Chikungunya virus 2016 GuidancesComplianceRegulatoryInforamtion/Guidances?blood/
provisional data for the United States. Available from: ucm411780.htm.
https://www.cdc.gov/chikungunya/geo/united-states-2016 77. Centers for Disease Control and Prevention [Internet]:
.html. Atlanta (GA): The CDC [cited 2010 Aug 24]. Babesiosis fact
62. American Association of Blood Banks [Internet]: Bethesda sheet. Available from: www.cdc.gov/babesiosis/factsheet.htm/
(MD): American Association of Blood Banks [cited 2017 #intro.
Jul 14]. Dengue viruses. Available at: www.aabb.org/tm/eid/ 78. Mahon CR, Lehman DC, Manuselis G Textbook of diagnostic
documents/dengue-viruses.pdf. microbiology. St. Louis (MO): Saunders Elsevier; 2007.
63. Centers for Disease Control and Prevention [Internet]: Atlanta 79. Food and Drug Administration [Internet]: Rockville (MD): FDA
(GA): The CDC [cited 2017 Jul 15]. Dengue epidemiology. [cited 2010 Aug 24]. July 26–27, 2010: Blood Products Advisory
Available from: www.cdc.gov/dengue/epidemiology/index Committee Meeting presentations. Available from: www.FDA
.html. .gov/advisory/committees/committeesmeeting-materials/Blood
64. Food and Drug Administration [Internet]: Rockville (MD): Vaccines and other Biologics/BloodProductsAdvisoryCommittee/
FDA [cited 2017 May 15]. Guidance for industry: recommen- UCM221857.htm.
dations for assessment of blood donor eligibility, donor deferral 80. Leiby DA. Transfusion-transmitted Babesia spp.: bull’s-eye on
and blood product management in response to Ebola virus, Babesia microti. Clin Microbiol Rev. 2011;24(1):14–28.
Available from: http://www.fda.gov/BiologicsBloodVaccines/ 81. Stramer SL, Hollinger FB, Katz LM, Kleinman S, Metzel PS,
GuidanceComplianceRegulatoryInformation/Guidances/default Gregory KR, et al. Emerging infectious disease agents and their
.htm. potential threat to transfusion safety. Transfusion. 2009;49
65. Centers for Disease Control and Prevention [Internet]: Atlanta Suppl 2:1S–29S.
(GA): The CDC [cited 2017 Jul 15]. Review human to human 82. Food and Drug Administration [Internet]: Rockville (MD):
transmission of Ebola virus. Available from: https://www.cdc FDA [cited 2017 Jul 15]. Fatalities reported to FDA following
.gov/vhf/ebola/transmission/human-transmission.html. blood collection and transfusion annual summary for FY2015.
66. Brecher ME, Hay SN. Bacterial contamination of blood prod- Available from: www.fda.gov/downloads/BiologicsBloodVaccines/
ucts. Clin Microbiol Rev. 2005;18(1):195–204. SafetyAvailability/ReportaProblem/TransfusionDonationFatalities/
67. Food and Drug Administration [Internet]: Rockville (MD): UCM518148.pdf.
FDA [cited 2017 Jul 14]. Fatalities reported to FDA following 83. Herwaldt BL, Linden J, Bosseman E, Young C, Olkowska D,
blood collections and transfusion. Annual summary FY2015. Wilson M. Transfusion-associated babesiosis in the United States:
Available from: https://www.fda.gov/downloads/BiologicsBlod a description of cases. Ann Intern Med. 2011;155:509-519.
Vaccines/SafetyAvailability/ReportaProblem/TranfusionDontaion 84. American Association of Blood Banks [Internet]: Bethesda
Fatalities/ucm. (MD): American Association of Blood Banks [cited 2017
68. Kuehnert MJ, Roth VR, Haley NR, Gregory KR, Elder KV, Jul 15]. AABB Association Bulletin #14-05. Available from:
Schreiber GB, et al. Transfusion-transmitted bacterial infection www.aabb.org/programs/publications/bulletins/Documents/
in the United States, 1998 through 2000. Transfusion. 2001;41; ab14-05.pdf.
1493. 85. Jocoby GA, Hunt JV, Kosinski KS, Demirjian ZN, Huggins C,
69. Food and Drug Administration [Internet]: Rockville (MD): Etkind P, et al. Treatment of transfusion-transmitted babesiosis
FDA [cited 2017 Jul 15]. Draft guidance for industry: bacterial by exchange transfusion. New Engl J Med. 1980;303:1098.
6888_Ch14_307-332 29/10/18 5:43 PM Page 332

332 PART III Transfusion Practices

86. Evenson DA, Perry E, Kloster B, Hurley R, Stroncek DF. Ther- 93. Food and Drug Administration [Internet]: Rockville (MD):
apeutic apheresis for babesiosis. J Clin Apher. 1998;13:32. FDA [cited 2017 Jul 15]. Guidance for industry: recommenda-
87. Bern C, Montgomery SP. An estimate of the burden of Chagas tions for donor questioning, deferral, reentry and product
disease in the United States. Clin Infect Dis. 2009;49e:52-54. management to reduce the risk of transfusion-transmitted
88. Pan AA, Winkler MA. The threat of Chagas’ disease in transfu- malaria. Available from: www.fda.gov/BiologicsBloodVaccines/
sion medicine. Lab Med. 1997;28:269. GuidanceComplianceRegulatoryInformation/Guidances/Blood/
89. Food and Drug Administration [Internet]: Rockville (MD): ucm365191.htm.
FDA [cited 2017 Jul 15]. Draft guidance for industry: amend- 94. Food and Drug Administration [Internet]: Rockville (MD):
ment to “guidance for industry: use of serological tests to FDA [cited 2017 Jul 15]. Guidance for industry: revised
reduce the risk of transmission of Trypanosoma cruzi infection preventive measures to reduce the possible risk of transmission
in whole blood and blood components intended for transfu- of Creutzfeldt-Jakob disease and variant Creutzfeldt-Jakob
sion.” Available from: www.fda.gov/BiologicsBloodVaccines/ disease by blood and blood products. Available from: www
GuidanceComplianceRegulatoryInformation/Guidances/Blood/ .fda.gov/BiologicsBloodVaccines/GuidanceComplianceRegulatory
ucm528600.pdf. Information/Guidances/Blood/UCM307137.pdf.
90. Food and Drug Administration [Internet]: Rockville (MD): 95. Tabor E. The epidemiology of virus transmission by plasma de-
FDA. FDA approves Chagas disease screening test for blood tis- rivatives: Clinical studies verifying the lack of transmission of
sue & organ donors. Available from: www.FDA.gov/newsevents/ hepatitis B and C viruses and HIV type 1. Transfusion. 1999:
newsroom/PressAnnouncements/ucm210429.htm. 39:1160.
91. Holtzclaw A, Mrsic Z, Managloanog J, Calvano T, Colombo C. 96. Food and Drug Administration [Internet]: Rockville (MD): FDA
Transfusion-transmitted malaria not preventable by current [cited 2010 Sept 3]. Transfusion/donation fatalities. Available
blood donor screening guidelines: a case report. Transfusion. from: www.fda.gov/BiologicsBloodVaccines/SafetyAvailability/
2016;56:2221-2224. ReportaProblem/transfusiondonationFatalities/default.htm.
92. Griffith KS, Lewis LS, Mali S, Parise ME. Treatment of malaria
in the United States: a systemic review. JAMA. 2007:297(20)
2264-77.
6888_Ch15_333-354 31/10/18 9:51 AM Page 333

Chapter 15
Component Preparation
Heather M. Vaught, BS, MLS(ASCP)CMSBBCM

Introduction Red Blood Cells Factor VIII Concentrates (FVIII)


Component Preparation Platelets Factor IX Concentrates
Equipment in the Component Leukoreduction Factor XIII Concentrates
Manufacturing Laboratory Plasma Immune Serum Globulin
Documentation of Manufacturing Cryoprecipitated Antihemophilic Normal Serum Albumin (NSA)
Steps Factor/Cryo-Poor Plasma Plasma Protein Fraction
Centrifuges Granulocyte Concentrates Rho (D) Immune Globulin
Plasma Expressors Additional Manufacturing Synthetic Volume Expanders
Scales Irradiation Antithrombin III Concentrates
Tubing Sealers Washing Labeling of Components
Sterile Connection Devices Aliquots Summary Chart
Plasma Freezers Pooling Review Questions
Storage Devices Frozen, Deglycerolized RBCs References
Blood Product Manufacturing and Quality Plasma Derivatives
Control Activated Factor VII (Factor VIIa)
Whole Blood

OBJECTIVES
1. Identify the method of preparation, storage conditions, shelf life, and quality control requirements for the following:
• Whole blood
• Red blood cells, including irradiated, leukoreduced, and saline washed
• Random and apheresis platelets, including irradiated, leukoreduced, and washed
• Pooled random platelets
• Frozen, deglycerolized RBCs
• Fresh-frozen plasma and plasma frozen within 24 hours
• Liquid plasma
• Cryoprecipitate and prepooled cryoprecipitate
• Granulocyte concentrates
• Factor concentrates, including activated factor VII, VIII, IX, and XIII concentrates
• Rho(D) immunoglobulin
• Normal serum albumin
• Immune serum globulin
• Plasma protein fraction
• Antithrombin III concentrates

333
6888_Ch15_333-354 31/10/18 9:51 AM Page 334

334 PART III Transfusion Practices

Introduction should include who performed each critical task, the equip-
ment that was used, and if preparing time-sensitive compo-
For the first 300 or more years of transfusion history, blood nents, the date and time that the task was completed. The
was acquired, stored, and transfused as whole blood. In the BECS also assists with preventing components from being
1950s, the introduction of the plastic bag to replace break- shipped before the completion of infectious disease testing
able glass bottles and the development of refrigerated cen- and electronic quarantine of components that are later found
trifuges brought forth the dawn of a new age in transfusion to be unacceptable (e.g., postdonation information from
medicine: component therapy.1 donor). To comply with U.S. Food and Drug Administration
(FDA) and AABB requirements, manufacturing records must
Component Preparation be retained and protected from accidental destruction.
A single blood donation can provide transfusion therapy to Centrifuges
multiple patients in the form of red blood cells (RBCs),
platelets, fresh-frozen plasma, and cryoprecipitate. Other Component manufacturing centrifuges are typically either
products such as derivatives of plasma (e.g., immune serum large floor units or large tabletop units that can spin a maxi-
globulin) also benefit patients with various diseases or con- mum of 6 to 12 units of whole blood at once. Component man-
ditions. This section describes the manufacturing process of ufacturing centrifuges have programmable speed (RPM) and
all components used in transfusion therapy. time settings that can be changed based on the components
Components may be prepared either by collecting whole that are being manufactured. The centrifugation profile used
blood and transforming into components using centrifuga- will depend on the types of components that are to be manu-
tion or by collecting targeted components using apheresis, factured. Table 15–1 summarizes the AABB-recommended
which involves centrifugation of whole blood at the donor’s centrifuge conditions to manufacture red blood cells, plasma,
bedside and the return of blood fractions that are not col- platelets, and cryoprecipitate.
lected. Often, more of the target component can be collected
by a single apheresis donation than can be obtained from col- Plasma Expressors
lecting whole blood. Collection profiles include apheresis
platelets (one donation can collect up to three adult doses of Plasma expressors are mechanical devices that apply pres-
platelets, or 18 times the quantity that can be harvested from sure to the blood bag, which allows blood components
a single whole blood donation), apheresis plasma (one dona- to flow from one bag to another by way of the integrated
tion can collect up to 1 L of plasma, which is approximately tubing system. Basic expressors are nothing more than
three to four times the volume that can be harvested from a spring-loaded Plexiglas. Automated expressors are also
single whole blood donation), apheresis double-red blood commercially available, such as the Compomat G5 (Fenwal),
cells (one donation can collect up to 2 units of red blood which boast barcode scanners, pneumatic pumps, inte-
cells, double what can be harvested from a single whole grated scales, and BECS interfaces. While these devices are
blood donation), or a combination, such as platelets and more costly than the manual counterparts, they tend to
plasma, platelets and red blood cells, or plasma and red blood cite more consistent product quality, higher plasma yield,
cells. For more on apheresis collections, see Chapter 18, better ergonomics, and a more effective use of the work-
“Apheresis.” force as justification for their cost. The two types of devices
can be seen in Figure 15–1.
Equipment in the Component Manufacturing
Laboratory Scales

Equipment used in the component manufacturing laboratory Some components, like plasma, must be labeled within
is intended for one of three functions: manufacture, quality 10% of the actual volume of the component. Scales should
control, or storage. The order of steps in the manufacturing be validated to ensure that they produce accurate and reli-
process will be influenced by the bag used for blood collec- able results. Quality control of each scale using calibrated
tion and the types of end components that are to be manu-
factured. The anticoagulant/preservative, additive (if present),
Table 15–1 AABB-Recommended
number of satellite bags and their composition, and presence
Centrifuge Conditions
or absence of an inline leukoreduction filter will dictate the
exact steps required to transform donated whole blood into Components Prepared Spin Conditions
components.
Red blood cells + plasma 5,000 × g, 5 minutes

Documentation of Manufacturing Steps Red blood cells + platelet-rich plasma 2,000 × g, 3 minutes

Whether documented on manual forms or in a blood estab- Harvesting platelets or cryoprecipitate 5,000 × g, 7 minutes
from plasma
lishment computer system (BECS), the component manu-
facturing laboratory must have a mechanism for capturing g = units of gravity. Please note: The RPM is based on your centrifuge’s rotor diameter
critical steps in the manufacturing process. Documentation to achieve the relative centrifugal force (g).
6888_Ch15_333-354 31/10/18 9:51 AM Page 335

Chapter 15 Component Preparation 335

solid. To maximize throughput in the freezing process and


to enhance clotting factor recovery, rapid freezing devices
may be utilized. Blast freezers use circulating cooled air to
rapidly freeze plasma units that are placed flat on a tray. In
contrast, immersion baths circulate an antifreeze and water
mixture around protective pockets, into which the plasma
units are placed either standing upright or lying flat.

Storage Devices

Blood component storage devices should be carefully selected


Figure 15–1. Comparison of automated Compomat G5 (Fresenius Kabi) (left) and validated to ensure that they are capable of maintaining
to a manual plasma expressor (right).
the FDA-regulated storage temperatures. Temperatures must
be continuously monitored, recorded at least every 4 hours,
standard weights should be performed periodically to and the device should alert the user if an unacceptable tem-
ensure that the scale is performing within specifications. perature condition occurs. Storage devices include refrigera-
tors, freezers, and platelet agitators. An optional piece of
Tubing Sealers equipment is an environmental chamber that maintains
platelet storage at 20° to 24°C. This type of device can be
The tubing that connects blood bags is usually made of omitted if the room in which platelets are stored is constantly
polyvinyl chloride (PVC). PVC will melt upon exposure to monitored and maintained at 20° to 24°C and there is an
radio frequency energy. Blood component tubing sealers, adequate location to store platelets should problems with the
sometimes incorrectly referred to as “heat sealers,” use a com- facility’s HVAC arise.
bination of targeted radio frequency energy and pressure to
melt and seal the tubing. The tubing is placed in a channel, Blood Product Manufacturing
and a trigger is activated when the user is ready to make the and Quality Control
seal. The seal usually includes an exceptionally thin band that
can be pulled or twisted apart. When making segments on Whole Blood
red blood cell components, the seal is usually placed at strate-
Whole blood is collected in a ratio of 14 mL of anticoagulant-
gic locations such that each segment contains a serial number.
preservative for every 100 mL of whole blood targeted for col-
Once the segments are separated from the RBCs, this serial
lection. Depending on the collection system used, a whole
number provides positive linkage of the segment and the RBC
blood component typically contains either 450 mL (±10%)
unit from which it came. When making a series of seals along
of whole blood with 63 mL of anticoagulant-preservative or
a section of tubing, it is critical to start at the closed end and
500 mL (±10%) of whole blood with 70 mL of anticoagulant-
work toward the open container to avoid pressure buildup
preservative, collected from blood donors with a minimum
within the tubing with each seal that is made, which will
hematocrit of 38%. Occasionally, units of other volumes are
eventually cause the tubing to rupture during sealing.
collected; the collection volume is stated on the label. Whole
blood transfusions provide both oxygen-carrying capacity
Sterile Connection Devices
and volume expansion. However, this same benefit can be
Sterile connection devices (SCDs) allow two separate blood obtained with concentrated red blood cells and either a
bags to be connected via their PVC tubing without breaching manufactured volume expander such as saline or albumin
the integrity of either container. Each piece of tubing is placed or frozen plasma that would contain viable clotting factors.
in a channel and clamped. The SCD heats a single-use copper Although whole blood has not been used widely for trans-
wafer to a sterile temperature, then the wafer slices through fusion since the adoption of universal component therapy
the tubing. The tubing is realigned while the wafer is held in in the 1950s, the U.S. military has published literature on
place; once the two pieces of tubing are in the desired position, the successful use of whole blood in trauma,2–4 which has
the wafer is removed. The melted tubing is pressed together inspired some hospitals to transfuse whole blood during
and allowed to cool. The tubing is checked for proper align- civilian trauma.5 By far, most whole blood donations are
ment, then the sealed section is pinched to open the tubing manufactured into components, red blood cells, platelets,
channel. The welded tubing is inspected again for any evi- plasma, cryoprecipitate, or some combination of these com-
dence of leaking or bubbles indicating air contamination. ponents. If the donation remains as whole blood, it must be
When the SCD is used to connect two containers, the expira- stored at 1° to 6°C, and the shelf life is dependent on the
tion of the original/target component is maintained. anticoagulant/preservative used. Whole blood collected in
acid-citrate-dextrose formula A (ACD-A), citrate-phosphate-
Plasma Freezers dextrose (CPD), or citrate-phosphate-double dextrose (CP2D)
has a shelf life of 21 days. Whole blood collected in citrate-
To freeze plasma, liquid components may be placed in a phosphate-dextrose-adenine (CPDA-1) has a shelf life of
standard –18°C or colder freezer and allowed to freeze until 35 days.
6888_Ch15_333-354 31/10/18 9:51 AM Page 336

336 PART III Transfusion Practices

Quality Control components from whole blood using centrifugation. Red


Quality-control indicators for this component are not rou- blood cells must be stored at 1° to 6°C. In the absence of
tinely measured. Process control measures during blood additive solution, RBCs have the same shelf life as whole
collection, including quality control of hemoglobinometers blood, either 21 (CPD, CP2D, ACD-A) or 35 (CPDA-1)
used to determine donor hemoglobin/hematocrit and scales days. The addition of additive solution to the prepared
used to weigh the collected component and determine the RBCs extends the shelf life to 42 days. Currently, licensed
collection endpoint, will ensure that the whole blood com- anticoagulant-preservative and additive combinations in
ponent meets the characteristics previously described. the United States include CPD with AS-1 or AS-5, CP2D
with AS-3, and ACD-A with AS-3. In addition to these for-
Red Blood Cells mulations, novel anticoagulants are being developed and
designed to mitigate the red blood cell storage lesion,
RBCs are separated from whole blood by centrifugation, as discussed in Chapter 1, “Red Blood Cell and Platelet
apheresis, or less commonly, sedimentation. Apheresis col- Preservation: Historical Perspectives and Current Trends.”
lections are discussed in Chapter 18. Although RBCs may
be prepared from whole blood donations at any time during Quality Control
the whole blood shelf life, they are typically prepared Red blood cells without additive solution must have a fin-
shortly after donation to allow the manufacture of platelet ished hematocrit of less than 80%. Component hematocrit
concentrates, frozen plasma, or cryoprecipitate, which can be measured with a commercial hematology analyzer or
must be prepared within designated time periods after col- manually using a capillary tube spun in a microhematocrit
lection, usually 8 to 24 hours. If additive solutions (AS) are centrifuge. Red blood cells with additive solution are not
employed, as much of the plasma is removed as possible, required to undergo hematocrit testing, as the addition of
and the AS must be added to the RBC component within 100 mL or more of additive solution will result in a compo-
3 days of collection, resulting in a finished product with a nent with a hematocrit of approximately 55%.
hematocrit of 55% to 65%. If an additive solution is not AABB Standards for Blood Banks and Transfusion Services
used, the volume of plasma removed is targeted such that state that red blood cells collected by apheresis must contain
the finished RBC product has a hematocrit of 65% to 80%. a mean of at least 60 g of hemoglobin or 180 mL of RBC vol-
RBC components typically have a final red cell volume of ume, and 95% of units tested must contain greater than 50 g
160 to 275 mL or 50 to 80 g of hemoglobin suspended in of hemoglobin or 150 mL of RBC volume, unless other pa-
the residual plasma and/or additive solution.6 Box 15–1 and rameters are specified in the collection device’s FDA-approved
Figure 15–2 outline a general procedure for manufacturing instructions for use.7 Apheresis component hematocrit or

BOX 15–1

Procedure for Whole Blood Fractionation Using Centrifugation


1. If whole blood filter is present for leukoreduction, filter the whole 7. Completing RBC manufacturing:
blood unit following the manufacturer’s instructions. a. Follow the manufacturer’s instructions for adding the attached
2. Load whole blood units into the blood component centrifuge’s additive solution to the RBCs if applicable.
swinging bucket apparatus, ensuring opposing units are balanced. b. If desired, the RBC may be leukoreduced if not already done; use
3. Centrifuge whole blood: a sterile connection device to attach a leukoreduction filter or
a. RBC and plasma only: Hard spin (see Table 15–1) at 4°C. use the filter integrally attached to the collection bag. Complete
b. RBC and PRP: Light spin (see Table 15–1) at 20° to 24°C. filtration following the filter manufacturer’s instructions.
4. At the end of the centrifuge cycle, remove the units from the c. Store the RBCs at 1° to 6°C.
centrifuge and place into a plasma expressor. 8. If manufacturing platelets:
5. Express the plasma into the attached satellite bag. a. Place balanced loads of PRP into the blood component centrifuge’s
a. If more than one satellite bag is present, apply hemostats so that swinging bucket apparatus and spin at a hard-spin setting.
the plasma will flow into only one bag. b. Once the centrifuge cycle has stopped, remove the PRP units and
b. If additive solutions are to be added to the red cell product, place into the plasma expressor.
remove virtually all of the plasma (90% to 95% hematocrit). c. Remove the temporary closure and allow plasma to drain into
c. If no additive solution is to be added, then use a method to empty satellite bag and apply the hemostat when the approxi-
ensure that the final RBC product hematocrit is less than 80%. mate target platelet volume, usually 45 to 70 mL, is achieved.
6. When the appropriate amount of plasma has been removed, apply d. Place the platelet bag on a scale and adjust the volume if necessary.
hemostats to the satellite bag. Always ensure the satellite bags have e. Use the tubing sealer to seal and detach the platelet and plasma
the same donor number as the primary bag. components.
a. If making platelets, then place a temporary closure on the tubing, f. Allow the platelets to rest at room temperature for approximately
such as a rubber band or plastic clip. Seal and detach the PRP 1 hour before placing on the agitator at 20° to 24°C.
bag with an empty satellite bag from the RBCs. 9. The plasma is stored depending on the product desired (FFP, liquid
b. If not making platelets, seal and detach the plasma from the rest plasma, etc.).
of the bags.
6888_Ch15_333-354 31/10/18 9:51 AM Page 337

Chapter 15 Component Preparation 337

Whole Blood Collection

Yes
Filter whole blood Whole Blood
Filtration?

Yes Plt No
Soft spin Manufacturing? Hard spin

Separate RBC and PRP Separate RBC and plasma


RBCs PRP

Add additive if desired


Add additive if desired Hard spin PRP

Express plasma
Yes RBC
Filter RBCs Filtration? Plt Plasma Plasma RBCs

No Let rest ~1 hour Freeze plasma


RBC Yes
Filtration? Filter RBCs
Store at 1°–6°C
Store at –18°C or colder
No
Store at 1°–6°C
Store at 20°–24°C,
agitate
Yes
Cryo? Thaw at 4°C

Hard spin thawed plasma

No Express cryo poor plasma

Pool cryo if desired

Store at –18°C Freeze cryo/CPP


or colder

Figure 15–2. Overview of the process for manufacturing whole blood into components.

hemoglobin may also be measured with automated or manual by a single nontraumatic venipuncture, and the concentrate
methods. must be prepared within 8 hours of collection. Platelets from
donors who are taking a platelet-inhibiting drug such as
Platelets aspirin may not be used as the sole source of platelets for a
patient; therefore, donor centers that make platelets from
Platelet components can be produced from whole blood whole blood donations must have a mechanism to identify
donations by apheresis. Apheresis platelet collection is such donors so that they may be excluded from platelet
discussed in Chapter 18. Analysis of blood collection and production or they may be pooled with other acceptable
manufacturing trends in the United States indicates that products,9 Box 15–2. Whereas platelets derived from whole
apheresis platelet collections are increasing, while the man- blood are typically called random-donor platelets (RDPs)
ufacture of platelets from whole blood is decreasing.8 Whole the platelet product obtained from an apheresis donation is
blood used to prepare platelet concentrates must be drawn referred to as single-donor platelets (SDPs) because each
6888_Ch15_333-354 31/10/18 9:51 AM Page 338

338 PART III Transfusion Practices

BOX 15–2

Procedure for Preparing Random-Donor Platelets and Plasma from Whole Blood
1. Whole blood should be: 9. Allow the platelets to rest undisturbed for 1 to 2 hours at 20° to 24°C
a. From a nontraumatic venipuncture. or until all platelet clumps have been resuspended in the residual
b. Collected less than 8 hours ago. plasma. Be sure the platelet button is covered with the plasma.
Gentle manipulation can be used if needed.
c. Maintained at 20° to 24°C before and during platelet preparation.
10. Weigh the plasma bag and determine the volume. Record the vol-
d. A closed, multibag system. ume on the bag.
2. Set the centrifuge temperature to a range of 20° to 24°C at a soft- 11. Freeze the plasma. The plasma must be frozen within 8 hours
spin setting (see Table 15–1). or 24 hours, depending on which frozen plasma product is to be
3. Express the platelet-rich plasma into one of the satellite bags. If not made. The plasma can be labeled as FFP, plasma frozen within
using an additive solution, enough plasma must remain on the RBCs 24 hours (PF24), or liquid plasma. (Note: For FFP prepared from
to ensure 80% or less hematocrit. WB collected in ACD, the plasma must be frozen within 6 hours.)
4. Seal the tubing between the RBC and the plasma. Disconnect the 12. Once the platelet concentrate has rested, the unit should be placed
RBC, leukoreduced if indicated, and store it at 4°C. on an agitator to maintain gentle agitation during storage at 20°
5. Recentrifuge the platelet-rich plasma at 20° to 24°C using a heavy to 24°C.
spin (see Table 15–1). This will separate the platelets from the 13. Platelet concentrate shelf life is 5 days from the date of collection.
plasma. If the system is opened, transfusion must occur within 4 hours from
6. Express the majority of the plasma into the second satellite bag, the time the closed system was disrupted.
leaving approximately 50 to 70 mL on the platelets. The volume- 14. All of the units for a single platelet dose (typically 4 to 8 units for
to-bag surface area ratio and platelet concentration is important an adult) can be pooled into a single bag before transfusion. Once
to maintain the pH above 6.2 during storage. pooled, the product must be transfused within 4 hours of pooling if
7. Seal the tubing between the bags and separate. prepared in an open system. The pooled unit must be given a
8. Inspect the platelet components for RBC contamination and disposi- unique pool number, which must be placed on the label.
tion per facility policy.

component contains one adult dose from a single donor. causing the platelets to pellet at the bottom of the container
Both types of platelets have advantages and disadvantages so that the supernatant plasma can be removed. In other
that should be considered for specific patient populations. countries, such as Canada and much of Europe, RDPs are
RDPs are relatively inexpensive to manufacture. Collection manufactured using a buffy coat method.10 In this process,
bag systems that include a platelet storage container are the whole blood is centrifuged at a hard-spin profile, most
slightly more expensive than collection bag systems not of the plasma is removed, then the leukocyte and platelet-
intended for platelet manufacturing because the platelet rich buffy coat is harvested. The buffy coat can either be
storage container must be made of a plastic that allows for collected with or without plasma. Several buffy coat concen-
oxygen exchange to maintain the appropriate pH throughout trates are pooled together, and a synthetic medium, such as
the storage period. The second spin also adds expense in the a platelet additive solution, may be added to the pool. The
form of additional labor to manipulate the bags and harvest pool is centrifuged at the soft-spin setting, harvesting the
the additional components. Apheresis collections tend to be platelets with either the residual plasma or the synthetic
very expensive, with an upfront cost to purchase and main- medium.
tain the apheresis equipment, specialized disposable kits, Platelets are stored at 20° to 24°C with constant agitation,
and solutions for each donation, and staff labor to monitor and therefore they are the blood component that is most
the donation for up to 2 hours. RDPs may be maintained as likely to grow bacteria if inadvertently contaminated. Bacte-
individual components for small-volume transfusions or ria may be introduced into a platelet product either by inef-
pooled together for adult transfusions. SDPs are generally in- fective skin cleaning prior to phlebotomy or a low-level
dicated for patients who are unresponsive to random platelets donor bacteremia that is not identified during health history
due to HLA alloimmunization or to limit the platelet expo- review or physical examination of the donor.11 Bacterial
sure from multiple donors, but they must be divided into contamination in platelets was once estimated to occur in
aliquots if small-volume transfusions are required. Due to approximately 1 in 2,000 to 1 in 3,000 components.12 To
centrifuge conditions, RDPs are prone to RBC contamination, address this risk, the AABB Standards for Blood Banks and
whereas apheresis platelets contain very few RBCs. Transfusion Services requires blood banks and transfusion
Manufacturing RDPs is accomplished using two methods services to have methods in place to detect bacteria or use
throughout the world. In the United States, the platelet-rich pathogen reduction technology (PRT) in all platelet compo-
plasma (PRP) method is employed, where the whole blood nents.13 Two devices are approved by the FDA to identify
is centrifuged at a slower speed and shorter time, allowing contaminated platelets. One system, Haemonetics eBDS, uses
the platelets to remain suspended in the plasma. PRP is har- oxygen consumption as a surrogate marker for bacterial con-
vested from the red blood cells, then centrifuged at higher tamination.14 The eBDS process requires an aliquot of the
RCF (relative centrifugal force) for longer periods of time, platelet product to be transferred into a pouch containing
6888_Ch15_333-354 31/10/18 9:51 AM Page 339

Chapter 15 Component Preparation 339

pelleted growth medium. The aliquot is incubated at 35°C shelf life. The draft guidance allows for day 4 and 5 testing
for at least 18 hours, then a probe is used to aspirate the head- to be waived if treated using pathogen reduction technology
space air in the pouch and measure the oxygen content. If (PRT; see Chapter 1), but it requires PRT-treated platelets to
the oxygen content is reduced, then the product is flagged be transfused within 5 days, as none of the PRT systems have
as potentially containing bacteria. Platelets are considered FDA approval for 7-day labeling.
released for transfusion at the time that the test is read. In
contrast, the BacT/ALERT 3D (bioMerieux) system utilizes Quality Control
plastic blood culture bottles containing a liquid growth Whole blood–derived platelet concentrates should contain
medium and a chromogenic carbon dioxide sensor.15 An at least 5.5 × 1010 platelets. Apheresis or single-donor
aliquot of the platelet product is aseptically transferred to platelets contain at least 3 × 1011 platelets (therapeutic equiv-
bottles (aerobic, anaerobic, or both) and placed into an alent of four to six random-donor platelets). Both methods
incubation chamber that uses light reflection to detect a color must maintain a pH of greater than or equal to 6.2 through
change in the chromogenic carbon dioxide sensor. Measure- the end of the labeled storage period. Automated hematology
ments are taken every 10 minutes. If there is no growth in a analyzers are typically used to determine the number of
facility-defined time period (usually 12–24 hours), then platelets per microliter, which is multiplied by the total com-
the platelet products are considered released for transfusion. ponent volume to determine the yield. Many commercial
The culture bottles remain in the incubation chamber and devices are available to determine platelet pH.
continue to be monitored until outdate of the platelet prod- Due to the increased prevalence of apheresis collections,
uct. If a slow-growing organism is present, then it will be the FDA published a guidance document in December
detected later in the culture process and the associated prod- 2007 addressing quality control requirements for apheresis
ucts may be retrieved from the hospital. If the products have platelets.21 The guidance document requires statistically
already been transfused, then the patient(s) can be monitored sound sample sizes based on 95% confidence that 75% of
closely and/or treated prophylactically to prevent further components will meet the target platelet yield and a sample
complications. Currently, the INTERCEPT® Blood System is size based on 95% confidence that 95% of components will
the only FDA approved technology for the manufacture of meet the target pH. The guidance document suggests mini-
certain “pathogen-reduced” apheresis platelet and plasma mum sample sizes of 11 platelets per month for platelet yield
products. and 60 platelets per month for pH, with no failures allowed
In 2005, the FDA approved the PASSPORT (Post Ap- (see discussion on leukoreduction in this chapter for addi-
proval Surveillance Study of Platelet Outcomes, Release tional information). A similar requirement does not exist
Tested) study, which allowed the outdate platelet products for RDPs; therefore most facilities sample a fixed number or
to extend from 5 to 7 days.16 To participate in the study, percent per month for platelet yield and pH.
donor centers had to utilize the BacT/ALERT 3D system to
monitor platelets using aerobic and anaerobic culture bottles Leukoreduction
throughout the entire 7 days of storage and perform post-
market surveillance testing by collecting a new sample from Leukocyte reduction, or leukoreduction, is the removal of
any outdated platelet products on the 8th day and incubating white blood cells (WBCs) from blood or blood components
for an additional 7 days to look for bacterial growth. A total prior to transfusion. The FDA first published recommenda-
of 388,903 apheresis platelets were accrued during the study tions for leukoreduction in a memorandum issued in 1996.22
period (September 2005 to January 2008). The repeat testing Under those recommendations, leukoreduced blood compo-
revealed an unacceptably high risk of bacterial contamina- nents were to contain less than 5.0 × 106 residual white
tion in the units, and the study and the use of 7-day stored blood cells per each whole blood, red blood cells, or aphere-
apheresis platelets was stopped based upon the results. sis platelet, and less than 8.3 × 105 residual WBCs per
In 2011, the FDA approved the Verax Platelet PGD test each platelets derived from whole blood. In addition, at least
(Verax Biomedical) as a safety measure to detect bacterial 85% of the original component must be recovered after
contamination in platelets.17 Subsequently, in 2015, the FDA leukoreduction. Leukoreduction of whole blood and blood
granted 510(k) clearance to two major apheresis platelet bag components has been shown to reduce recurrent febrile non-
manufacturers to extend platelet dating to 7 days if the hemolytic transfusion reactions, reduce alloimmunization to
platelet products were tested within 24 hours before transfu- leukocyte antigens that may complicate care of patients who
sion using a point-of-issue safety device intended to detect undergo transplantation or chronic transfusion therapy, and
bacterial contamination.18,19 These approvals were followed protect against transmission of cytomegalovirus (CMV) to
in March 2016 with a draft guidance from the FDA outlining patients at increased risk of CMV disease.23 Leukoreduction
the agency’s proposed next steps to reduce the risk of adverse performed shortly after collection, usually within 72 hours,
reactions to platelet transfusion due to bacterial contamina- is considered prestorage leukoreduction. Products that are
tion20 (U.S. Department of Health and Human Services, Food not leukoreduced early in their shelf life may also be leuko-
and Drug Administration, Center for Biologics Evaluation reduced at the patient’s bedside by passing the component
and Research). The draft guidance supports not only PGD through a filter during infusion. Recent reports indicate that
testing to extend outdate to 7 days, but it also requires testing over 84% of whole blood/RBCs and over 86% of apheresis
for all platelet products transfused on days 4 and 5 of their platelets transfused in the United States are leukoreduced
6888_Ch15_333-354 31/10/18 9:51 AM Page 340

340 PART III Transfusion Practices

and that 73% of transfusing facilities reported that their Apheresis platelet collection methods are configured
policy is to exclusively transfuse leukoreduced blood com- to collect a leukocyte-poor product without the need for
ponents.8 Of the almost 12 million leukoreduced blood com- filtration, which would also be considered prestorage
ponents transfused in 2011, only 1.1% were leukoreduced leukoreduction.
at the bedside; the remaining 98.9% were leukoreduced
either before or during storage. The impetus for prestorage Quality Control Testing
leukoreduction involved biological response modifiers WBCs typically cannot be counted for quality control (QC)
(BRMs) released from leukocytes during storage of the com- testing using routine hematology analyzers because the
ponent that were found to promote febrile transfusion reac- expected concentration is below the instruments’ level of
tions. Examples of BRMs include proinflammatory cytokines linearity or detectability. WBCs can be manually counted
(interleukin-1, interleukin-6, and tumor necrosis factor) and using a specialized hemocytometer called a Nagoette cham-
complement fragments (C5a and C3a).24 Bedside filtration ber, which is designed to count WBCs at exceptionally
has been associated with precipitous hypotension in the low levels. Alternatively, WBCs may be counted using flow
transfusion recipient, particularly in patients on medications cytometry.
that inhibit the angiotensin-converting enzyme (ACE
inhibitors). This infrequent yet serious adverse effect is not
associated with prestorage leukocyte reduction. Removing Advanced Concepts
leukocytes by centrifugation or filtration just before trans- In the early period after adoption of leukoreduction, facil-
fusion of blood should prevent reactions that are caused ities typically sampled either a fixed number or a percent-
by leukocyte antibodies in a patient’s plasma and leukocytes age of blood components to ensure compliance with the
present in the transfused blood; however, it would not guidelines. In December 2007 and September 2012, the
prevent reactions caused by BRMs that originate from FDA published Guidance for Industry documents outlining
the leukocytes present in the component during storage. the agency’s current thinking on the topic of apheresis
Studies suggest that the age of the RBC unit is a predictor of platelet collection and leukoreduction, respectively.20,25
a febrile reaction and that the cytokine involvement may be The main focus of these documents was to introduce sta-
cumulative.24 tistical methods for validation and quality control of the
Leukoreduced RBCs and whole blood are prepared using leukoreduction process, to make a distinction between
special filters that achieve at least a 99.9% (a 2 to 4 log) re- process failures and nonprocess failures, and to introduce
moval of leukocytes by employing multiple layers of polyester the concept of donor-specific factors, including the pres-
or cellulose acetate nonwoven fibers that trap leukocytes and ence of hemoglobin S, that should be identified in the
platelets but allow RBCs to flow through. If the collection bag donor population to prevent leukoreduction failures. To
does not have a filter integrally attached by the manufacturer, determine how many products needed to be evaluated
then a filter may be attached to the red blood cells after per month, the population must first be defined, then the
manufacturing. Whether to use a bag with an integrated filter number of products within that population must be deter-
versus one that must be individually attached is a financial mined. The guidance document has two recommended
decision by the collecting facility. When using bags with an two-stage statistical models: hypergeometric and binomial.
inline filter, then a filter must be purchased for each donation The binomial model assumes an infinite number of com-
and discarded if the donation is not successful or if the prod- ponents prepared and dictates that if 60 products per
uct is damaged during manufacturing. When the filter is month are tested and no failures are observed, then it has
attached after RBCs are manufactured, the collection facility been proven with 95% confidence that 95% of the products
purchases filters only for components that successfully make meet the defined criteria. If one failure is observed in this
it through the manufacturing process. There is additional cost infinite population, then the failure must be investigated
associated with sterile connection, including the disposable to determine if the process failed. If the failure was due to
wafer and labor costs of the individuals performing the con- a factor that is known to impact performance, such as the
nection that must be considered. There are two major filter presence of HgbS, then the failed product can be excluded
types available in prestorage leukoreduction. In the first from the population and a replacement component can be
method, whole blood is passed through the leukoreduction tested. If the failure cannot be explained as a nonprocess
filter before centrifugation. RBCs and plasma can then be failure, then 74 consecutive components must be tested. If
prepared, but platelets are typically also trapped in the filter no additional failures are observed, then the monthly QC
and thus random-donor platelets cannot be manufactured. In has been successful. If additional failures are observed, then
the second method, the whole blood is centrifuged, the a full investigation must be completed to identify and
plasma is removed from the whole blood unit, and then rectify the part of the process that failed. Alternatively, if
the packed cells are passed through the leukoreduction the facility would prefer to allow one process failure during
filter. Today, many institutions maintain 100% of their red the sampling period without requiring additional testing,
blood cell inventory as prestorage leukoreduced products. then an initial population size of 94 components could be
Historically, other methods to achieve RBC leukoreduction selected. If two failures would be observed in the initial
include centrifugation and washing, or freezing, thawing, and population, then 75 components would need to be tested.
deglycerolizing.
6888_Ch15_333-354 31/10/18 9:51 AM Page 341

Chapter 15 Component Preparation 341

Smaller population sizes can use the hypergeometric collection device’s labeled indications. Many platforms
model, for which the number of products tested is based have options for both FFP and PF24 labeling.
on the number of components prepared. For example, a FFP will contain the maximum levels of both stable and
population size of 100 requires only 45 products to be labile clotting factors, about 1 international unit (IU) per
tested with zero failures observed instead of 60, making milliliter. PF24 contains all stable proteins found in FFP, has
the hypergeometric method preferable when fewer com- normal levels of factor V, and has only slightly reduced levels
ponents are produced. In large populations, the hyperge- of factor VIII. Factor levels in LP are poorly characterized in
ometric model converges with the binomial model. A this product and will vary depending on when and how it
population of 400 requires at least 55 components to be was manufactured.6
tested, and a population size of 4,000 requires 58 compo- Both PF24 and FFP are thawed at temperatures between
nents to be tested. 30° and 37°C or in an FDA-approved microwave device. If a
Statistical sample sizes were seen as exceptionally bur- water bath is used, then the product must be placed in a pro-
densome in the blood community. Facilities had previously tective lining or overwrap so that the ports of the unit are
been testing a fixed number or percentage of their collec- not contaminated by contact with the water. Once thawing
tions, which was often as few as four products per month or is complete, the product may be stored at 1° to 6°C for up to
1% of the products made. Under the new guidance, facilities 24 hours. If not transfused within the initial 24-hour period,
with small collection numbers would have to test substan- the thawed plasma may be stored for up to 5 days, but the
tially more products on a monthly basis. For example, if a product label must be changed to “thawed plasma” because
facility collected on average 30 single platelet donations it can not maintain therapeutic levels of the labile clotting
per month, then they would have to test 23 products (77%) Factor V and Factor VIII.
per month and find no products with a WBC above the The volume of a unit of FFP or PF24 from apheresis may
acceptable yield. If a product was found to have an elevated be as much as 800 mL. Large apheresis plasma donations
WBC count, then 100% of the products collected that month may also be divided into smaller doses that are similar in
would need to be tested. Also included in this guidance range to whole blood–derived FFP. A single unit of FFP or
was the requirement to separate single, double, and triple PF24 from whole blood collection may contain anywhere
donations into three different populations, requiring all from 200 to 375 mL, depending on the original collection
centers to test at least 69 products per month (23 singles, volume, the donor’s hematocrit, and any additional pro-
23 doubles, and 23 triples). Facilities that collect large num- cessing. For example, if a 500-mL bag is used, the actual
bers of components potentially observed reduced QC re- blood collection volume may be anywhere from 450 to
quirements; for example, a large facility producing 10,000 550 mL (±10% of the target volume). If the maximum
leukoreduced red blood cells per month and testing 1% of 550 mL was collected and the donor had the minimum-
all components prepared would perform quality control test- allowed hematocrit of 38%, then the collected blood is
ing on 100 components per month. Under the new guid- 62% plasma, or 310 mL. Upon centrifugation, the antico-
ance, they would be required to test only 60 per month. agulant will be expressed with the plasma, adding 70 mL
These savings were usually offset by additional platelet to the product volume for a theoretical yield of 380 mL. In
testing. some donor groups, finger-stick hemoglobin readings have
been shown to overestimate venous hemoglobin values;26
therefore, the percentage of any given donation that is
Plasma plasma may be slightly more than expected. In addition, it
is not usually possible to harvest all of the plasma from the
Plasma from a whole blood donation can be manufactured donation, and several milliliters are usually retained on the
into a variety of different components. The plasma may be RBC component. In contrast, a 450-mL donation from a
frozen within 8 hours and labeled as fresh-frozen plasma male with a hematocrit of 55% contains only 45% plasma,
(FFP) or frozen within 24 hours and labeled as plasma frozen or 202.5 mL. The 450-mL collection bag contains an
within 24 hours (PF24). Plasma frozen within 8 hours can additional 63 mL of anticoagulant, for a total of 265.5 mL.
be further manufactured into cryoprecipitate and cryo-poor If the whole blood was centrifuged at a light-spin setting
plasma, which will be discussed later in this chapter. Frozen to harvest platelets, then the initial plasma harvest will be
plasma is stored at –18°C or colder for 1 year or at –65°C or suboptimal and an additional 45 to 70 mL of plasma vol-
below for 7 years with FDA approval. Plasma from whole ume will be diverted into the platelet product. All of these
blood donations may also remain in a liquid state, stored factors could result in a plasma yield of less than 200 mL.
at 1° to 6°C, and be labeled as liquid plasma (LP). LP expires One unit of plasma contains approximately 300 mg
5 days after the whole blood shelf life from which it was fibrinogen per 100 mL6 and 1 unit of activity per milliliter
collected. For example, CPD whole blood has a 21-day shelf of each of the stable clotting factors. FFP also contains the
life, therefore CPD LP has a shelf life of 26 days, whereas same level (1 unit/mL) of factors V and VIII. PF24 is known
CPDA-1 whole blood has a 35-day shelf life, and CPDA-1 LP to have reduced levels of labile clotting factors, especially
has a shelf life of 40 days. factor VIII; however, the differences are mostly statistically
Apheresis plasma must be frozen within the time frame insignificant or still maintain hemostatic activity well above
specified and stored under conditions specified by the the minimum required for safe surgical hemostasis.27
6888_Ch15_333-354 31/10/18 9:51 AM Page 342

342 PART III Transfusion Practices

Quality Control In recent years, cryoprecipitate has also been used to


Neither the FDA nor AABB mandate quality control testing make fibrin glue, a substance composed of cryoprecipitate
for plasma components. Because each product is labeled with (fibrinogen) and topical thrombin. Generally, 1 to 2 units of
the volume, scales must be calibrated and tested to ensure cryoprecipitate are thawed and drawn into a syringe. Topical
accuracy. When performing manual entry of plasma volumes thrombin with or without calcium is drawn into a second
into a BECS that are automatically printed onto the label, the syringe. The contents of the two syringes are simultaneously
laboratory should have practical checks in place to ensure applied to the bleeding surface, where fibrinogen is con-
that a miskeyed entry does not result in a product being mis- verted to fibrin.28 Fibrin glue is also available as commer-
labeled. Manual labeling processes should also include steps cially prepared products that are viral inactivated and
to verify accurate labeling and avoid transposition errors. licensed to control bleeding in cardiovascular surgeries.
CPP is also thawed at 30° to 37°C, but it is stored at
Cryoprecipitated Antihemophilic Factor/ refrigerated temperatures and transfused within 24 hours
Cryo-Poor Plasma unless relabeled as thawed plasma, cryoprecipitate reduced.
Relabeled plasma may be transfused up to 5 days after it was
Cryoprecipitated antihemophilic factor (AHF) is the cold- thawed. CPP is most often used for transfusion or plasma
precipitated concentration of factor VIII, also known as the exchange in patients with Thrombotic Thrombocytopenia
antihemophilic factor. The process for isolating factor VIII Purpura (TTP) who have been initially treated using FFP
also harvests fibrinogen, factor XIII, von Willebrand factor without an adequate response, but it could also be used as a
(vWF), cryoglobulin, and fibronectin.6 Cryoprecipitate- source of those factors that remain after cryoprecipitate is
reduced plasma (cryo-poor plasma or CPP) is the plasma from harvested.
which the cryoprecipitate concentrate has been harvested. A procedure for producing cryoprecipitate and cryo-poor
After removing the cryoprecipitate, CPP contains the resid- plasma is outlined in Box 15–3.
ual albumin; factors II, V, VII, IX, X, XI; and ADAMTS13.
CPP cannot be used as a substitute for FFP, PF24, or thawed Quality Control
plasma.6 Cryoprecipitate and CPP are prepared from FFP Cryoprecipitate contains at least 80 units of AHF activity and
that is thawed at 1° to 6°C, which prevents resuspension of at least 150 mg of fibrinogen.29 Factor VIII and fibrinogen
cold-insoluble clotting factor proteins. The thawed plasma activity assays are performed by hemostasis reference labo-
is centrifuged at 4°C at a hard-spin setting, then the super- ratories. Quality control is not indicated for cryo-poor
natant plasma is expressed, leaving a button of cryoprecipi- plasma.
tate in 10 to 25 mL of plasma. Once the centrifuge is opened,
cryoprecipitate must be harvested by expressing the super- Granulocyte Concentrates
natant plasma and refrozen within 1 hour. Each unit (U) of
cryoprecipitate is commonly prepared from 1 unit of fresh- For information on granulocyte collection, see information on
frozen plasma (FFP). An adult dose of cryoprecipitate is con- leukapheresis in Chapter 18, “Apheresis.” A granulocyte com-
sidered 10 units. To maximize the time available to transfuse ponent should contain at least 1 × 1010 granulocytes, which
cryoprecipitate once thawed, some donor centers manufac- can be measured using an automated hematology analyzer as
ture prestorage pooled cryoprecipitate. Working within the long as the results are within the linearity limits. Dilutions
1-hour refreezing time frame, multiple single cryoprecipitate may have to be considered at higher WBC concentrations.
components are pooled together using a sterile connection Table 15–2 provides a comprehensive list of blood com-
device to maintain a closed system. The number of compo- ponent characteristics.
nents may vary by manufacturer, and the number of compo-
nents in the pool will be listed on the label. CPP may be Additional Manufacturing
refrozen up to 24 hours after harvesting. Both cryoprecipitate
and CPP are stored at –18°C or below for up to 1 year from Irradiation
the whole blood collection date.
Cryoprecipitate should be thawed quickly at 30° to 37°C Irradiation of cellular blood components (red blood cells,
and stored at room temperature (22° to 24°C) until trans- platelets, and granulocytes) is indicated to prevent the
fused. Once thawed, prestorage pooled cryoprecipitate and development of transfusion-associated graft-versus-host
single units of cryoprecipitate must be transfused within disease (TA-GVHD). Patients at risk of TA-GVHD include
6 hours. Cryoprecipitate pooled in the transfusion service immunocompromised patients who are receiving a bone
laboratory using an open system must be transfused within marrow or stem cell transplant and fetuses undergoing
4 hours. Cryoprecipitate is indicated in the treatment of an intrauterine transfusion. Irradiation is also indicated for
factor XIII deficiency, as a source of fibrinogen for hypofi- recipients of components collected from a blood relative or
brinogenemia, and as a secondary line of treatment for clas- HLA-matched donors.
sic hemophilia (hemophilia A) and von Willebrand’s disease. For a detailed discussion on the clinical implications of
Cryoprecipitate should not be used to treat hemophilia A or TA-GVHD, see Chapter 17, “Adverse Effects of Blood Trans-
von Willebrand’s disease if virus-inactivated or recombinant fusion.” Briefly, TA-GVHD occurs when donor (graft) T lym-
factor preparations are available.6 phocytes escape destruction by the patient’s (host’s) immune
6888_Ch15_333-354 31/10/18 9:51 AM Page 343

Chapter 15 Component Preparation 343

BOX 15–3

Procedure for Production of Cryoprecipitate


1. The venipuncture must be nontraumatic. 6. Express the supernatant plasma into the attached satellite bag. The
2. The whole blood can be cooled before and during production be- cryoprecipitate will be a small white mass in the original plasma
cause platelets are not usually produced along with cryoprecipitate. bag. Leave only 10 to 20 mL of plasma on the precipitate.
The volume of plasma required to remain on the RBCs and the 7. If preparing prestorage pooled cryoprecipitate, identify compatible
platelet concentrate would reduce the amount of plasma available components for pooling and combine at this time.
for cryoprecipitate production enough to significantly reduce the 8. Refreeze the cryoprecipitate immediately. Time elapsed should be
final AHF activity in the precipitate. At least 200 mL of plasma no more than 1 hour at room temperature. A delay in refreezing or
(205 g) should be used to ensure that the final product will contain exposure of the unit to elevated temperatures during processing
at least 80 AHF units. will significantly decrease the factor VIII activity level in the final
3. The plasma must be stored at –18°C or below within 8 hours of product. The centrifuge temperature must be at 1° to 6°C, and it is
collection and within 1 hour from the time freezing was initiated. better if the centrifuge cups are well chilled.
4. The second stage of cryoprecipitate preparation begins by allowing 9. The final product should be placed in a protective container be-
the frozen plasma to thaw slowly in the refrigerator at 1° to 6°C . cause of the brittle nature of the plastic bag at freezer tempera-
This takes 14 to 16 hours when plasma is thawed in a standard tures. Store at –18°C or colder up to 12 months from the date of
blood bank refrigerator. If a circulating cryoprecipitate thaw bath whole blood collection.
(4°C water bath) is used, the thawing time is reduced to about 10. If the supernatant plasma is refrozen at –18°C, it must be labeled
2 to 4 hours. The endpoint is when the plasma becomes slushy. as “plasma cryoprecipitate reduced.”
5. Centrifuge the plasma at 1° to 6°C for a hard spin.

Table 15–2 Blood Component Characteristics


Storage
Component Shelf Life Temp. Quality Control Volume Dosage
Whole blood (WB) CPD 21 d 1°–6°C N/A 450–500 mL ↑ hgb 1 g/dL
CPDA-1 35 d ↑ hct 3%
CP2D 21 d

Whole blood irradiated Original expiration or 1°–6°C 25 Gy to center of canister 450–500 mL ↑ hgb 1 g/dL
28 d post-irradiation
↑ hct 3%

RBCs CPD 21 d 1°–6°C No additive: hct ≤ 80% 250–300 mL ↑ hgb 1 g/dL


CPDA-1 35 d ↑ hct 3%
CP2D 21 d Additive: N/A
ACD 21 d
AS 42 d

RBC aliquots Closed system: same 1°–6°C varies 10 mL/kg


Open system: 24 hr ↑ hgb 2 g/dL

RBC irradiated Original outdate or 1°–6°C 25 Gy to center of canister 250–300 mL ↑ hgb 1 g/dL
28 d post-irradiation
↑ hct 3%

RBC leukoreduced Closed system: same 1°–6°C <5 × 106 WBCs ≥ 85% RBC 250–300 mL ↑ hgb 1 g/dL
recovery
Open system: 24 hr ↑ hct 3%

Washed RBCs 24 hr 1°–6°C hct 70–80% 180 mL ↑ hgb 1 g/dL


↑ hct 3%

Frozen RBCs 10 years ≤ –65°C

RBC deglycerolized 24 hr 1°–6°C 80% RBC recovery 180 mL ↑ hgb 1 g/dL


<1% glycerol ↑ hct 3%

Platelets, whole-blood 5–7 d 20°–24°C ≥5.5 × 1010 plts 50–70 mL ↑5k–10k/µL


derived (RD)
pH ≥ 6.2
Continued
6888_Ch15_333-354 31/10/18 9:51 AM Page 344

344 PART III Transfusion Practices

Table 15–2 Blood Component Characteristics—cont’d


Storage
Component Shelf Life Temp. Quality Control Volume Dosage
Platelets, apheresis (SD) 5–7 d 20°–24°C ≥ 3 × 1011 plts 200–400 mL ↑30k–60k/µL
pH ≥ 6.2
Platelets, irradiated 5d 20°–24°C 25 Gy to center of canister Same Same
Platelets, pooled 4 hr 20°–24°C pH ≥ 6.2 Varies Varies
Platelets, leukoreduced 5d 20°–24°C pH ≥ 6.2 RD: 40–70 mL RD: ↑ 5–10k/µL
RD: <8.3 × 105 WBCs SD: 200–400 mL SD: ↑ 30–60k/µL
SD: <5 × 106 WBCs

FFP 1 yr ≤–18°C 200–380 mL ↑Factor 20%–30%


7 yr ≤–65°C 200–1,000 mL 10–20 mL/kg
AFFP 1 yr ≤–18°C
PF24 1 yr ≤–18°C 200–380 mL Same
7 yr ≤–65°C
LP 5 days after WB 1–6°C 200–380 mL Not well
expires liquid characterized
Cryoprecipitate Frozen: 1 yr ≤–18°C FVIII: 80 IU 10–25 mL ↑Fibrinogen
Fibrinogen: 150 mg 5–10 mg/dL
Thawed: 6 hr 20°–24°C
Pooled (open): 4 hours
Cryo-reduced plasma 1 yr ≤18°C 200–380 mL
Granulocytes 24 hr 20°–24°C ≥1.0 × 1010 200–600 mL 1–2 × 1010/infusion
four daily doses

Granulocytes, irradiated 24 hr 20°–24°C ≥1.0 × 1010 200–600 mL Same

hgb = hemoglobin; hct = hematocrit; plts = platelets; WB = whole blood; SD = single donor; RD = random donor

system and subsequently proliferate and mount an immune device is sent through an irradiation cycle, and the minimum
response against the patient’s tissues. In immunocompro- and maximum radiation dose is determined. For irradiation
mised patients, donor T lymphocytes escape destruction devices using a radioactive source, the amount of exposure
because the patient’s immune system lacks the resources nec- time required to achieve the minimum radiation exposure
essary to detect and destroy the foreign cells. When a patient increases as the radioactive source decays.
receives blood from a relative or an HLA-matched donor, the Each facility should have a protocol and procedure for
donor cells may be haploidentical to the patient cells and irradiating blood components, training of personnel using
therefore not express antigens that would trigger the patient’s the irradiator, and issuing of irradiated components. The
immune system to recognize the donor cells as foreign. Irra- expiration date of irradiated RBCs is 28 days from the time
diation damages the nucleic acid of the donor T lymphocytes of irradiation or the original outdate, whichever is sooner.
and therefore makes them unable to proliferate and cause The expiration date of platelets and granulocytes are not
disease. impacted by irradiation.
Both the FDA and AABB recommend a minimum dose of
gamma irradiation of 25 Gy to the central portion of the Washing
blood unit, with no less than 15 Gy delivered to any part
of the blood unit.28 Irradiation may be achieved by using Red blood cells prepared using the methods previously
either a radioactive source (cesium-137 or cobalt-60) or described in this chapter are stored for up to 42 days
x-ray. To confirm a product was irradiated, a radiochromic suspended in a solution that contains additive solution (in-
film label is affixed to the component before it is placed into cluding saline, amino acids, and sugars) in addition to a
the metal canister of the irradiator. Darkening of the film small volume of residual plasma and anticoagulant that is
confirms irradiation requirements. Irradiation devices also not removed during manufacturing. Platelets are also
require semiannual or annual dose delivery verification using stored in a mixture of donor plasma and anticoagulant
an outside source. To determine dose delivery, a detection solution, with or without additive. The process of removing
6888_Ch15_333-354 31/10/18 9:51 AM Page 345

Chapter 15 Component Preparation 345

the extracellular solution and replacing it with normal after irradiation has been shown to reduce the extracellular
saline is called washing. potassium levels.33
Automated washing devices are commercially available.
In the absence of an automated device, it is possible to wash Aliquots
components manually using a centrifuge. The process would
include adding the wash solution to the component, cen- Aliquoted blood products are often transfused during the
trifuging, then manually expressing the supernatant into a neonatal period or in infants younger than 4 months of age
waste bag. due to their small blood volume. Indications for small-volume
Washed RBCs are stored at 1° to 6°C for up to 24 hours. transfusion in this patient population include anemia caused
Washed platelets are stored at 20° to 24°C and must be trans- by spontaneous fetomaternal or fetoplacental hemorrhage,
fused within 4 hours. twin-twin transfusion, obstetric accidents, and internal
There are three main indications for washing. hemorrhage. Blood drawn from infants for laboratory testing
(iatrogenic anemia) also may result in a neonatal transfusion
Allergic Reactions if more than 10% of the blood volume has been lost due to
Red blood cell or platelet components may be washed for phlebotomy, which may occur as frequently as every 2 days
patients who have severe allergic reactions to proteins found for infants who require close monitoring due to their fragile
in the residual plasma. Washing components is usually re- clinical condition.34 Transfusions for neonates are typically
served for patients whose allergic reactions are anaphylactic dosed at 10 to 15 mL/kg, therefore requiring only small
or anaphylactoid in nature and cannot be effectively miti- volumes of blood products (10 to 25 mL) per dose. Several
gated with medication prior to transfusion. Some patients aliquots may be prepared from a single-donor unit.
who are IgA deficient may develop anti-IgA, which may To prepare aliquots of red blood cells or platelets, a small
stimulate an allergic transfusion reaction when the patient container is attached to the finished component using a ster-
is exposed to normal plasma containing IgA.30 In others, the ile connection device. An aliquot is prepared by zeroing the
source of the reaction is not easily explained. scale using an empty container, then slowly adding product
to the final container until the target volume is delivered.
Removal of Antibody Aliquots of plasma are typically prepared before initially
When antigen-negative RBCs or platelets are collected from a frozen, stored frozen at –18°C or below for up to 1 year, and
mother in cases of hemolytic disease of the fetus and newborn thawed when needed.
or neonatal alloimmune thrombocytopenia, the mother’s RBCs The precise volume of blood desired can be aspirated into
or platelets lack the required antigen but contain a consider- a syringe through a large-bore needle inserted through an
able dose of antibody. Washing the RBCs or platelets allows injection site coupler. The filled syringe should be closed
transfusion of the needed component without exposing the securely with a sterile cap and labeled with expiration date,
fetus or newborn to additional antibody. Another scenario patient identification, volume transfused, preservative, and
when antibody removal is desired is when group O RBCs, ABO type. In some institutions, the blood bank will issue the
which contain small amounts of anti-A, anti-B, and anti-A,B blood aliquot to the neonatal ICU or floor where the syringe,
are transfused to treat cases in which ABO antibodies cause if used, will be filled by the patient care staff.
hemolytic disease of the fetus and newborn. Red blood cell aliquots prepared using a closed system
maintain the outdate of the original RBC component. If the
Removal of Other Substances RBC aliquot is prepared using an open system, it must be
A third indication for washing cellular components is to transfused within 24 hours. RBC transfusions of 10 mL/kg
remove other substances of clinical concern. One example in a unit with a hematocrit level of 80% should raise the he-
is to remove potassium and avoid hyperkalemic cardiac ar- moglobin by 3 g/dL.29 Use of an RBC component in additive
rhythmias or arrest due to potassium leakage in RBC com- solution with a lower hematocrit would raise the hemoglo-
ponents. At plasma potassium levels of 7.5 mEq/L and bin by a smaller increment. Aliquots prepared using a closed
higher, conduction of electrical signals through the cardiac system retain the outdate of the original RBC component.
muscle is depressed.31 Extracellular potassium levels in- Because CPDA-1 red blood cell components contain no
crease over time in stored RBC units, which have been additive solution, this anticoagulant has historically been
documented to contain as high as 60 mEq/L potassium after popular for neonatal transfusions. However, recent literature
32 days of storage,32 and irradiated RBCs have been docu- has shown that additive solutions, including AS-3, which
mented to have extracellular potassium levels as high as contains a higher dose of mannitol than any other additive
31 mEq/L as soon as 2 days after irradiation. In large pa- solution, are safe for neonatal transfusion.35
tients, small patients with normal compensatory functions, As with RBCs, platelet transfusions for neonates require
and patients receiving transfusions via peripheral access, the only small volumes, and several aliquots may be prepared
potassium load is either diluted or neutralized without any from a single unit with the use of a sterile docked/connected
adverse effects. In small patients for whom transfusions are quadruple bag. This system can increase the number of
administered via central venous catheter, the high potassium transfusions an infant can receive from one donor, limiting
content has been documented to cause cardiac arrhythmias, the number of donor exposures, or it can be used to supply
cardiac arrest, and even death. Washing RBC components transfusions to several neonates or infants, providing better
6888_Ch15_333-354 31/10/18 9:51 AM Page 346

346 PART III Transfusion Practices

utilization of the product during the short dating period.


Transfusion of platelet concentrates is indicated for neonates BOX 15–4
whose counts fall below 50,000/mL and who are experienc- Preparation of Platelet Aliquots for Neonates
ing bleeding. Factors that may be associated with thrombo-
1. Select a unit to be aliquoted. Either AB or group-specific units
cytopenia include immaturity of the coagulation system, should be selected. CMV-negative or volume-reduced platelets
platelet dysfunction, increased platelet destruction, dilution may be requested by the physician. For volume-reduced platelets,
effect secondary to massive transfusion, or exchange trans- when infants cannot tolerate large intravenous infusions of
fusion and intraventricular hemorrhage. Either random or plasma, stored platelets are centrifuged and the plasma is re-
apheresis platelets may be transfused and should increase moved. The platelets remain undisturbed at room temperature
for 20 to 60 minutes before being resuspended in residual
the platelet count by 50,000 to 100,000, given a dose of 5 to plasma. Platelets should rotate for at least 30 minutes at room
10 mL/kg.28 Unless quality control testing is performed on temperature following the room temperature rest.
platelet aliquots to ensure that an adequate pH is maintained, 2. Remove cap from stopcock apparatus and attach to end of blood
platelet aliquots should not be stored in aliquot bags or sy- set. Use a 170- to 260-mm filter.
ringes any longer than necessary. In addition, when remov- 3. Remove second cap from stopcock and cap from syringe to be
ing aliquots from an apheresis platelet container, the original filled and place them on a piece of sterile gauze. Attach syringe to
component must still conform to the bag manufacturer’s stopcock.
specifications for minimum storage volume. 4. Close the roller clamps at the spike end of the blood set above
the filter and spike the platelet unit.
In addition to preparing aliquots for small patients, blood
5. Open the roller clamp on the spike and draw the platelets
components may be divided to support infusion at slower through the filter and into the syringe. Remove the syringe from
rates in patients who are susceptible to transfusion-associated the filter set and force the excess air back out of the syringe.
circulatory overload (TACO), which is a leading cause of 6. Label the syringe properly. Indicate volume of aliquot on platelet
transfusion-related morbidity and mortality, accounting product label.
for 22% of the transfusion-related fatalities reported to the 7. The expiration is 4 hours from the time the unit was spiked if
U.S. FDA from 2010 to 2014.36 Some risk factors for TACO using an open system.
include diagnoses such as congestive heart failure, chronic
pulmonary disease,37 and chronic renal failure.38 Other TACO
risk factors are more opportunistic in nature, for example,
patients with a positive fluid balance or patients who are height, weight, and hematocrit of the patient; the reason for
receiving multiple blood components. Mitigation strategies exchange; and the number of plasma volumes to be ex-
may include transfusing at a slower rate and/or dividing blood changed.40 Multiple liters may be required to support a single
components into smaller doses.39 (See Chapter 17, “Adverse procedure. To facilitate the bedside procedure, multiple
Effects of Blood Transfusion.”) products can be pooled into a large bag that can hold up to
A procedure for the preparation of platelet aliquots for 3 L at a time.
neonates is outlined in Box 15–4.
Pooled Platelets
Pooling A single whole blood–derived platelet contains at least 5.5 ×
1010 platelets. An adult dose is considered to be 3.0 × 1011
Pooled blood components allow for multiple blood compo- platelets. To prepare an adult dose, six RDPs may be pooled
nents to be transfused at a single event. The components into one bag approved for platelet storage. Fewer RDPs may
pooled may be all the same or a combination of components be used if the individual components’ platelet yield is high
depending on the patient’s need. As a rule, multiple units of enough to support a finished product of 3.0 × 1011. Platelets
red blood cells are not typically pooled together, but other pooled in an open system must be transfused within 4 hours
product types may be pooled. The pooling facility must of pooling.
maintain traceability and trackability of each original com-
ponent to the patient who received it in the event of look- Reconstituted Whole Blood
backs or recalls. Reconstituted whole blood, which is also known as recon-
stituted red blood cells, is the only example of pooling two
Pooled Cryoprecipitate different products together. This component is produced
As previously mentioned, an adult dose of cryoprecipitate is to support neonatal exchange transfusions. Red blood cells
10 units. Cryoprecipitate can be pooled either as part of the (usually group O) and plasma (usually group AB) may be
initial manufacturing, or single frozen units can be pooled pooled together to manufacture a component that has similar
after they are thawed. Prestorage pooled units must be used qualities to whole blood. If institutional policy requires it,
within 6 hours of thawing. Products pooled after thawing red blood cells may be irradiated, then washed before adding
must be transfused within 4 hours of thawing. plasma to achieve a target hematocrit, usually 45% to 55%.
A sample may be removed from the prepared component
Pooled Plasma and tested using a spun hematocrit or automated hematol-
The total volume of plasma needed for a therapeutic plasma ogy analyzer to ensure that the finished product is within
exchange procedure depends on many factors, including the the target range.
6888_Ch15_333-354 31/10/18 9:51 AM Page 347

Chapter 15 Component Preparation 347

Frozen, Deglycerolized RBCs and the products require less delicate handling. It does, how-
ever, require a larger volume of wash solution for deglyc-
Freezing RBCs with glycerol dates back to the 1950s.41 erolization. RBCs are frozen within 6 days of collection
Frozen RBCs can be stored for up to 10 years for those pa- unless rejuvenated (see Chapter 1). RBCs can be rejuvenated
tients with rare phenotypes, for autologous use, and for the up to 3 days after expiration and then glycerolized and
military to maintain blood inventories around the world for frozen. AABB Standards7 states that RBCs must be placed in
U.S. military use. The resulting deglycerolized product is free the freezer within 4 hours of opening the system. It is advis-
of leukocytes, platelets, and plasma due to the washing able to freeze a sample of donor serum in the event addi-
process. Since all donor plasma is removed, deglycerolized, tional testing is required for donor screening.
and washed, red blood cells can be used for patients with The thawing process takes approximately 45 minutes and
paroxysmal nocturnal hemoglobinuria and IgA deficiency involves immersing units into a 30° to 37°C water bath and
with circulating anti-IgA. washing the RBCs with solutions of decreasing osmolarity
Cryoprotective agents can be categorized as penetrating (e.g., 12% NaCl, 1.6% NaCl, 0.9% NaCl, with 0.2% dex-
or nonpenetrating. A penetrating agent involves small mol- trose). An exception to this rule is a donor with sickle trait
ecules that cross the cell membrane into the cytoplasm. The in which RBCs would hemolyze upon suspension in hyper-
osmotic force of the agent prevents water from migrating tonic solutions; in this case, the cells would be washed in
outward as extracellular ice is formed, preventing intracel- 12% NaCl and then 0.9% NaCl with 0.2% dextrose, omitting
lular dehydration. Glycerol is an example of a penetrating the 1.6% solution. Automated continuous-flow instruments
agent. Nonpenetrating cryoprotective agents are made of can be utilized for washing. Once the RBCs have been de-
large molecules that do not enter the cell but instead form a glycerolized, the unit is considered an open system with an
shell around it, preventing loss of water and subsequent de- expiration date of 24 hours and is stored at 1° to 6°C.
hydration. Hydroxyethyl starch and dimethylsulfoxide, used
to freeze hematopoietic progenitor cells, are two examples Low Glycerol (20% Weight per Volume)
of nonpenetrating agents. When cryopreserving red blood In this method, the cryoprotection of the glycerol is minimal,
cell units, laboratories may either use the high-glycerol or and a very rapid, more controlled freezing procedure is
low-glycerol methods. These methods differ in the equip- required. Liquid nitrogen (N2) is routinely used for this
ment used, the temperature of storage, and the rate of freez- method. The frozen units must be stored at –120°C or below,
ing. Most blood centers practice the high-glycerol method, which is the temperature of liquid N2 vapor. Because of the
which is outlined in Table 15–3. minimal amount of protection by the glycerol, temperature
fluctuations during storage can cause RBC destruction.
High Glycerol (40% Weight per Volume)
This method increases the cryoprotective power of the glyc- Quality Control
erol, thus allowing a slow, uncontrolled freezing process The quality control procedures necessary for RBC freezing
using a mechanical freezer that provides storage at –65°C or include all of the standard procedures for monitoring
below. This particular procedure is probably the one most refrigerators, freezers, water baths, dry thaw baths, and
widely used because the equipment is fairly simple to use centrifuges. They also include procedures to ensure at

Table 15–3 Key Steps in Freezing Red Blood Cells Using High-Glycerol Concentration
Preparation Glycerolization Deglycerolization
1. Weigh RBCs 1. Place cells on a shaker and add 100 mL 1. Thaw frozen cells at 30° to 37°C in water
glycerol bath

2. Adjust to 260–400 g 0.9% NaCl 2. Stop agitation and allow cells to equili- 2. Deglycerolize cells using a continuous-flow
brate 5–30 min washer

3. Prewarm RBCs and glycerol to 25°C 3. Let partially glycerolized cells flow into 3. Apply a deglycerolize label to transfer pack;
freezing bag; slowly add glycerol ABO, Rh, WB unit numbers and expiration
date

4. Set glycerol bottles in a water bath 4. Maintain glycerolized cells at 24°–32°C 4. Dilute unit with hypertonic 12% NaCl and
for 15 min at 25°–37°C until ready to freeze (not to exceed let equilibrate for 5 min
4 hours)

5. Label the freezing bag with name of 5. Freeze at ≤65°C for up to 10 years 5. Wash with 1.6% NaCl until residual glycerol
facility, whole blood unit numbers, from date of collection is less than 1%; wash with 0.9% NaCl plus
ABO, Rh, date collected, date frozen, 0.2% dextrose; store at 1°–6°C for up to
cryoprotective agent, expiration, and 24 hours
“red blood cells frozen”
6888_Ch15_333-354 31/10/18 9:51 AM Page 348

348 PART III Transfusion Practices

least 80% RBC recovery and adequate glycerol removal (less Activated Factor VII (Factor VIIa)
than 1% residual intracellular glycerol; Box 15–5). Valeri and
colleagues froze RBCs for up to 37 years by using the high- Activated factor VII (rFVIIa) is a prohemostatic agent produced
glycerol method (40 w/v) and yielded an average RBC re- by recombinant DNA technology approved for use in patients
covery of 75%, with 1% hemolysis.42 Although researchers with hemophilia A or B who have circulating antibodies or in-
are attempting to freeze cells for more than 10 years, the FDA hibitors, in patients with congenital factor VII deficiency, and
has not yet been swayed to change the maximum storage for patients with Glanzmann thrombasthenia.44 There are
time. Studies have also shown that irradiated (25 Gy) red many published reports of rFVIIa also being used off-label in
blood cells that are subsequently frozen and deglycerolized other situations such as trauma, massive transfusion, and liver
yield similar results in RBC recovery, post-transfusion sur- transplantation, where bleeding has proved difficult to control
vival, and residual hemoglobin as nonirradiated units.43 and the patient’s life is threatened. In these other situations, it
has been most successful in controlling intracranial bleeding
Plasma Derivatives in patients with major head trauma and cerebral hematomas.28
In addition, the administration of rFVIIa has seen promising
The following products are different from blood components results in uncontrolled nonsurgical hemorrhages after implant-
because they are prepared by further manufacture of pooled ing ventricular assist devices (VADs) to treat end-stage cardiac
human source (collected by apheresis specifically for further failure.45 The disadvantage to using rFVIIa is that it has been
manufacturing) and recovered (from whole blood collections associated with an increased risk of spontaneous thrombosis
where RBC components for transfusion are needed but and thromboemboli. In addition, the product is very expensive
plasma components are not) plasma; recombinant DNA and should be used with caution and restraint.
technology; or monoclonal antibody purification. One theory for the mechanism of factor VIIa is that rFVIIa
When manufacturing plasma derivatives from source or binds to tissue factor that is released from injured tissue and
recovered plasma, the plasma is frozen when sent to the then activates factor IX and X. Factor X’s presence on the
manufacturer. The manufacturing process usually begins surface of platelets leads to the generation of thrombin. Stud-
with a separation of the cryoprecipitate from the plasma. The ies have shown that full thrombin generation occurred after
cryoprecipitate is then used to produce factor VIII concen- the addition of up to 150 nm of recombinant FVIIa.46
trate. The residual plasma is separated into various proteins
by manipulating the pH, alcohol content, and temperature Factor VIII Concentrates (FVIII)
and then is viral inactivated by any of several methods,
including heat, solvent-detergent treatment, and nanofiltra- Factor VIII concentrates are used to treat patients with hemo-
tion.28 Most derivative plasma is also further tested for philia A and have almost completely replaced cryoprecipitate
hepatitis A and parvovirus B19, the agent responsible for as the product of choice. Factor VIII concentrates may be
fifth disease. Some manufacturers require donors to be tested prepared from large volumes of pooled plasma, but more com-
for these communicable diseases as well. monly they are prepared by recombinant DNA technology.

Human Source
BOX 15–5 If prepared from pooled human plasma, the plasma must be
treated by pasteurization, solvent/detergent treatment, or
Quality Control Procedures for Deglycerolization
monoclonal purification to inactivate or eliminate viral con-
1. RBC recovery can be determined by estimating the recovered tamination. In pasteurization, stabilizers such as albumin,
RBC mass:
sucrose, or glycine are added to the factor VIII concentrate
% Recovery = wt of DRBC [g] × Hct of DRBC × 100) to prevent denaturation of the product. The product is
(pre-wt of RBC × pre-Hct of RBC) heated to 60°C for 10 hours. The stabilizers are removed,
(DRBC = deglycerolized RBCs) and the product is lyophilized. This product is safe from
HIV-1 and hepatitis transmission.
2. Glycerol must be removed to a level of less than 1% residual. The
published procedures should accomplish this. However, it is im-
In solvent/detergent treatment, ethyl ether and tri(n-
portant to perform this check on each unit before releasing it for butyl) phosphate and the detergent sodium cholate and
transfusion. Tween 80 are effective in disrupting the viral coat membrane,
• Measure the osmolarity of the unit using an osmometer. The preventing the transmission of lipid-envelope viruses like
osmolarity should be about 420 mOsm (maximum 500 mOsm). HIV and hepatitis B. The solvent and detergent are removed,
• Perform a simulated transfusion. Place one segment (approxi-
mately 3 inches) of deglycerolized cells into 7 mL of 0.7% NaCl.
and the final product is lyophilized. Combinations of TnBP
Mix, centrifuge, and check for hemolysis. (tri-n-butyl phosphate) and polysorbate 20 during the man-
• Compare with a standard hemoglobin color comparator. If the ufacture of FVIII/vWF concentrate Optivate (BPL) resulted
hue exceeds the 500 mOsm level, hemolysis is too great, and in inactivation of most enveloped viruses over a wide range
the unit is not suitable for transfusion. of conditions, including solvent/detergent concentration,
• Perform specific gravity or refractive index of supernatant from
last wash. Excess glycerol solution is present if the supernatant
protein concentration, and temperature.
has a high specific gravity/refractive index. For monoclonal purification, immunoaffinity chromatog-
raphy is used to positively select out the vWF/FVIII complex
6888_Ch15_333-354 31/10/18 9:51 AM Page 349

Chapter 15 Component Preparation 349

from the plasma pool. Briefly, a murine monoclonal antibody complex concentrates should be used with caution in
directed at the vWF/FVIII complex is bound to a solid-phase patients with liver disease due to reports of DIC and throm-
matrix. On addition of pooled plasma, the complex will bosis.29 This is most likely due to failure of the liver to pro-
attach to the monoclonal antibody. The product is in duce adequate amounts of antithrombin III and a decreased
lyophilized form and is safe from viral transmission.47 hepatic clearance of activated factors.
Recombinant factor IX (rFIX) has been commercially
Porcine Factor VIII available in Europe and in the United States since 1997. It is
This xenographic form of factor VIII is made from porcine produced in a Chinese hamster ovary cell line and is not
plasma and is beneficial for patients with hemophilia A who thought to transmit human infectious disease. In 2001, Roth
have developed inhibitors or antibodies to human factor and colleagues published a study evaluating this product in
VIII. Porcine factor VIII has been shown to provide effective the treatment of patients with hemophilia B.50 Based on this
hemostatic control for patients with intermediate FVIII study and other data accumulated by physicians using rFIX
inhibitor levels. Other studies have shown that residual for hemophilia B patients, the Committee for Proprietary
porcine vWF in the preparation of the product induces Medicinal Products (CPMP) considered the benefit–risk
platelet activation, thus providing a mechanism for enhanc- balance for rFIX for the treatment and prophylaxis of bleed-
ing hemostasis apart from the action of circulating FVIII.48 ing in previously treated patients with hemophilia B to be
favorable. However, there were concerns regarding some
Recombinant Factor VIII aspects of the study, as there was the possibility of inhibitors
The gene for FVIII was sequenced nearly 16 years ago and to rFIX and allergic reactions to this product. As a result, a
led to the production of recombinant human FVIII (rFVIII). new clinical trial will be conducted.
The first generation rFVIII products are synthesized by
introducing human FVIII gene into BHK (baby hamster Factor XIII Concentrates
kidney) cells. The rFVIII is released into culture medium and
harvested, isolated, and purified using a combination of ion- Factor XIII deficiency is a severe autosomal-recessive bleed-
exchange chromatography, gel filtration, and immunoaffinity ing disorder associated with a characteristic pattern of
chromatography. The purification and final formulation of neonatal hemorrhage and a lifelong bleeding diathesis. There
rFVIII (BHK) uses human albumin as a stabilizer. The next- are two commercially available factor XIII concentrates in
generation product is referred to as rFVIII-FS; it is formu- the United States. The human-derived factor XIII concen-
lated using sucrose as a final stabilizer instead of albumin trate temporarily replaces missing factor XIII, which corrects
and is available in three doses (250, 500, and 1,000 IU). and/or prevents bleeding. Recombinant factor XIII is a ho-
When compared with the first-generation rFVIII, rFVIII- modimer composed of two factor XIII A-subunits, indicated
FS demonstrated a predictable clinical hemostatic response for routine prevention of bleeding in patients with congenital
with an efficacy among previously treated patients and factor XIII A-subunit deficiency.
previously untreated patients with hemophilia A. In one
study with previously untreated patients, inhibitors to Immune Serum Globulin
rFVIII-FS were present in 15% of patients, which was lower
than the percentage of the first-generation product (20%).49 Immune serum globulin is a concentrate of plasma gamma
To date, no transmission of hepatitis or HIV has been re- globulins in an aqueous solution. It is prepared from pooled
ported in association with this product. The next-generation plasma by cold ethanol fractionation. Preparations are in the
native recombinant FVIII has included a solvent/detergent form of intravenous (IV) or intramuscular (IM) solutions.
step as well as a purification step to help ensure a safe and The IV preparation generally contains more IgG protein than
effective product. the IM preparation, with a half-life of 18 to 32 days.
Immune globulin preparations are indicated for patients
Factor IX Concentrates with immunodeficiency diseases (i.e., severe combined
immunodeficiency and Wiskott-Aldrich syndrome) and for
Factor IX concentrates are available in three forms: prothrom- providing passive antibody prophylaxis against hepatitis and
bin complex concentrates (PCCs), factor IX concentrates, herpes. Intravenous immunoglobulin (IVIg) is also used in
and recombinant FIX. The first contains significant levels of patients with idiopathic thrombocytopenic purpura, post-
vitamin K–dependent factors II, VII, IX, and X. PCCs are pre- transfusion purpura, HIV-related thrombocytopenia, and
pared from large volumes of pooled plasma by absorbing the neonatal alloimmune thrombocytopenia. Individuals with a
factors using barium sulfate or aluminum hydroxide. The history of IgA deficiency or anaphylactic reactions should
concentrate is then lyophilized and virally inactivated by not receive immune globulin because of the presence of trace
methods previously described (e.g., solvent/detergent). PCCs amounts of IgA.
may contain activated vitamin K–dependent factors. Whereas there are no documented cases of HIV or hepa-
Factor IX (FIX) concentrate is developed by monoclonal titis B transmission, transmission of hepatitis C has been
antibody purification and is less thrombogenic than PCCs. reported with IVIg preparations. This has led to preparations
This product contains approximately 20% to 30% of FIX and of immune globulin treated with solvent/detergent for viral
is stored in the refrigerator in lyophilized form. Prothrombin inactivation.
6888_Ch15_333-354 31/10/18 9:51 AM Page 350

350 PART III Transfusion Practices

Normal Serum Albumin (NSA) delivery. If the screening test is positive, the FMH must be
quantified using the Kleihauer-Betke test.
NSA is prepared from salvaged plasma, pooled and fraction- RhIg is also used in the event Rh-positive platelet concen-
ated by a cold alcohol process, then treated with heat inac- trates are transfused to an Rh-negative patient. A 300-mg
tivation (60°C for 10 hours), which removes the risk of dose IM (120-mg dose IV) is sufficient to protect against
hepatitis or HIV infection. It is composed of 96% albumin D-positive RBCs contained in up to 10 units of random
and 4% globulin. It is available in 25% or 5% solutions. NSA platelets or one pheresis product. (See Chapter 20, “Hemolytic
is indicated in patients who are hypovolemic and hypopro- Disease of the Fetus and Newborn.”)
teinemic and in clinical settings for shock and burn patients.
The 25% preparation is contraindicated in patients who are Synthetic Volume Expanders
dehydrated unless it is followed with crystalloid infusions
(e.g., normal saline) for volume expansion. There are two categories of the synthetic volume expanders:
crystalloids and colloids. Ringer’s lactate and normal isotonic
Plasma Protein Fraction saline compose the crystalloids; dextran and HES make up
the colloid solutions. Normal saline consists only of sodium
The preparation of plasma protein fraction (PPF) is similar and chloride ions; Ringer’s lactate consists of sodium, chlo-
to that of NSA, with fewer purification steps. PPF contains ride, potassium, calcium, and lactate ions. These solutions
83% albumin and 17% globulins. PPF is available in a are useful in burn patients because of their ability to rapidly
5% preparation; its uses parallel that of NSA. PPF, however, cross the capillary membrane and increase the plasma vol-
is contraindicated for infusion during cardiopulmonary ume. Colloids are used as volume expanders in hemorrhagic
bypass procedures. Both PPF and NSA can be stored for shock and burn patients. Dextran is prepared in a 6% and
5 years at 2° to 10°C and have not been reported to transmit 10% solution with a half-life of 6 hours. HES is available in
HIV or hepatitis. a 6% solution with an IV half-life of more than 24 hours.
Both colloids and crystalloids are free from viral transmis-
Rho(D) Immune Globulin sion. Table 15–4 provides a comparison of crystalloids and
Rh-immune globulin (RhIg) is a solution of concentrated anti- colloids.
Rho(D). It is prepared from pooled human plasma of patients
Antithrombin III Concentrates
who have been hyperimmunized and contains predominantly
IgG anti-D. RhIg has two primary uses: treatment of ITP and Antithrombin III (AT-III) concentrates, or antithrombin (AT)
prevention of Rh hemolytic disease of the newborn. as it is now called, is prepared from pooled human plasma and
The FDA has approved two doses for treatment of ITP heat-treated to prevent viral transmission. This product, pdAT,
and immunization against the D antigen: a 120-mg dose and has been approved in the United States for treating patients
a 300-mg dose.29,51 These are IV preparations that have been with hereditary AT deficiency in connection with surgical or
heat-treated. An IM preparation is available as a 50-mg dose obstetrical procedures or for those suffering from thromboem-
and a 300-mg dose. The latter is considered a full dose bolism.52 AT-III is an inhibitor of clotting factors IX, X, XI, XII,
protective against 15 mL of D-positive RBCs. and thrombin. Patients with plasma levels of AT-III less than
In preventing immunization to the D antigen during ges- 50% of normal are at risk of thrombosis.
tation, a number of scenarios with varying dosages apply.
During the first 12 weeks of pregnancy, a 50-mg dose of
RhIg is indicated for D-negative females for abortion or Table 15–4 Comparison of Crystalloid
miscarriage. After 12 weeks’ gestation, a full dose (300 mg)
and Colloid Solutions
is indicated for abortion or miscarriage in D-negative women.
The 120-mg dose is advised after 34 weeks’ gestation when Characteristic Crystalloid Colloid
amniocentesis is performed or in the event of obstetric com- Intravascular retention Poor Good
plication or following termination of pregnancy.
An antepartum dose (300 mg IM or IV) should be given Peripheral edema Common Possible
to nonimmunized D-negative females at 28 weeks’ gestation. Pulmonary edema Possible Possible
Following delivery, a postpartum blood sample is drawn from
the mother. The sample undergoes a screening test for feto- Easily excreted Yes No
maternal hemorrhage (FMH) in which D-positive RBCs from Allergic reactions Absent Rare
the newborn are detected. Additionally, the newborn’s Rh sta-
tus is determined. If the newborn is D-positive or if the Rh Cost Inexpensive Expensive
type cannot be determined on the newborn (i.e., positive Ringer’s lactate solution Albumin
direct antiglobulin test [DAT]), the mother should receive a
full dose of RhIg unless she has demonstrated previous active Examples 7.5% normal saline Dextran
immunization to the D antigen. If the screening test is negative Hydroxyethyl
for the presence of D-positive RBCs of fetal origin, then the starch
mother should receive a full dose of RhIg within 72 hours of
6888_Ch15_333-354 31/10/18 9:51 AM Page 351

Chapter 15 Component Preparation 351

A new recombinant AT (rhAT) concentrate produced


using transgenic technology has been developed on a
compassionate-use basis. It is produced by transgenic goats
expressing recombinant human AT in their milk, under the
control of the beta-casein promoter. It is purified from the
milk and concentrated. Initial studies using rhAT have indi-
cated effective support for AT-deficient patients who undergo
surgery and determined that it is a suitable alternative to
pdAT. No antibodies have been produced against rhAT in
studies to date, and adverse events, which are minimal,
include spontaneously resolving skin hyperpigmentation at
the site of drug infusion.
Table 15–2 provides a comprehensive list of blood com-
ponent characteristics.

Labeling of Components
Once the component has been made, it must be labeled in
accordance with FDA regulations. FDA recognizes two accept-
able languages for blood component labeling: ABC Codabar
and ISBT 128. In the 1970s, the Committee for Commonality
in Blood Banking Automation appointed by the American Figure 15–3. Leukocyte-reduced unit of red blood cells.
Blood Commission (ABC) published a seven-volume report
for labeling recommendations leading to adoption of ABC of collection, a sequential number, and a check character. Each
Codabar to improve and simplify the labeling of blood and its ISBT 128 donation number is unique on a worldwide basis
components. In 1985, the FDA published the Guideline for the and not repeatable for 100 years. Each blood bank or transfu-
Uniform Labeling of Blood and Blood Components supporting sion service should have its own protocol for labeling compo-
Codabar in the United States. nents. The original unit, its components, or any modifications
As time progressed and complexities of the transfusion thereof must be identified; serologic results of the unit must
medicine industry heightened, the International Society for be reviewed and the appropriate labels attached in such a way
Blood Transfusion (ISBT) working group designed a totally that is clear and readable to the naked eye. If a change must
new system based upon the bar code symbology known as be made to the unit, such as a modification of the expiration
Code 128. This allowed for multidimensional bar codes date, the handwritten change must be legible.
and nonrepetitiveness of the donor identification number The ABO/Rh is indicated by a four-digit code. The first
(DIN) in not less than 100 years. ISBT 128 is managed by the two digits describe the ABO and D type. Different codes are
International Council for Commonality in Blood Banking Au- assigned to different types of donations; an allogeneic dona-
tomation (ICCBBA). ICCBBA was established in 1994 and is tion has a different ABO/D code than does an autologous
a not-for-profit organization whose mission is to support donation of the same blood type. The second two digits are
ISBT 128 and assist in its implementation from blood estab- not used in the United States, but they may be used in other
lishments. Each blood center or transfusion service must reg- countries to communicate additional Rh, K, and MNS phe-
ister with ICCBBA when using ISBT 128 coding labels. ISBT notypes. The ABO/Rh and DIN labels are strategically placed
128 labeling has been allowed in the United States since the in line with each other because the blood type is a function
FDA recognized it as an acceptable labeling symbology in of the donor from whom the component was obtained.
2000 but has become the standard since AABB mandated its The lower left quadrant contains the component product
use beginning with the 25th edition of the Standards effective code, which is comprised of eight characters. The first char-
May 2008 and stated that facilities that had not implemented acter indicates the type of component; different characters
ISBT 128 by November 1, 2009, would be placed on condi- are used to indicate blood, tissue, and hematopoietic pro-
tional accreditation status pending successful implementation. genitor cell components. The second through fifth characters
The ISBT 128 blood component label is a 4- by 4-inch specify the type of component. The sixth character indicates
label made up of four 2- by 3-inch quadrants. The upper left donation type and should match the donation type used in
quadrant contains the donation and collection facility iden- ABO/Rh labeling; if the component has an autologous ABO/
tifiers. The upper right quadrant contains the blood type. Rh, it should also have an autologous product code. The last
The lower left quadrant contains the product description, two characters of the product code indicate aliquots and
and the lower right quadrant contains the expiration date divisions to provide the ability to track each aliquot and each
and special labels. Figure 15-3 shows a properly labeled aliquot’s aliquot to the patient who received it.
leukocyte-reduced unit of blood. The lower right quadrant contains the expiration date and
The DIN is comprised of 14 characters that contain infor- time. The expiration date and time is strategically placed in
mation relating to the country, the center of origin, the year line with the product code, since the expiration is a function
6888_Ch15_333-354 31/10/18 9:51 AM Page 352

352 PART III Transfusion Practices

of the component characteristics. The expiration barcode con- Zika virus by investigational nucleic acid test, and RBC phe-
tains 10 characters: the first three indicating the year, the next notypes. (See Figure 15-3.)
three indicating the Julian date (e.g., February 1 is the 32nd The unique identifier of the unit, the ABO and Rh type,
day of the year, therefore is represented as 032), and the last expiration date, and component labels must be checked with
four characters indicating the expiration time, with 2359 used a second person who is using a computer. If the unit is
to indicate that the component expires at midnight. shipped to a transfusion facility that applies its own unique
Also included in the lower right quadrant are barcodes for identifier for the unit, the original identifier of the collecting
special labeling indications. The types of barcodes in use in facility must not be removed. There must be a method in place
the United States include CMV seronegative, negative for for tracing the unit from its origin to its final disposition.

SUMMARY CHART
 Blood component manufacturing must be documented 12 months. Because an adult dose of cryoprecipitate is
to ensure that the individuals who performed critical usually 10 units, the individual components may be
steps in manufacturing and the equipment that was pooled at the collection facility and refrozen or at the
used is identifiable for each component made. transfusion service after thawing.
 Equipment found in a component manufacturing  Irradiated RBCs must be given a radiation dose of at
laboratory includes centrifuges, plasma expressors, least 25 Gy to the midplane of the canister, after which
scales, sterile connection devices, sealing devices, the expiration date of the product changes to 28 days
plasma freezers, refrigerators, platelet agitators, and from the time of irradiation or maintains the original
environmental chambers. These devices should be outdate, whichever comes first. The outdate of platelet
carefully selected and validated to ensure that they components is not changed by irradiation.
operate in accordance with regulatory requirements.  RBCs and platelets can be washed to remove the
 Whole blood is usually separated into components but supernatant plasma and additive solution, which is
has recently increased in popularity for transfusion in then replaced with saline.
trauma due to research presented by the U.S. military.  RBC and platelets can be divided into aliquots for
 RBCs must be prepared by a method that separates the patients who require smaller volumes to be trans-
RBCs from the plasma and results in a hematocrit level fused, such as infants and adults at risk of transfusion-
of less than or equal to 80%. Leukoreduced RBCs associated circulatory overload. Plasma aliquots are
should contain <5 × 106 residual WBCs yet retain at usually prepared and frozen within 8 hours of collec-
least 85% of the original RBC content. The outdate of tion at the donor center.
RBCs depends on the anticoagulant/preservative and  Blood components can be pooled prior to transfusion
additive solution present. to support individuals who need larger doses or spe-
 Random-donor platelets must contain at least 5.5 × 1010 cialized combinations of components.
platelets; single-donor platelets must contain at least  Human plasma may be used to manufacture a wide
3 × 1011 platelets. Leukoreduced products must con- variety of therapeutic products, including clotting
tain <8.3 × 105 or 5.0 × 106 residual WBCs, respec- factors, immune globulins, and albumin or other pro-
tively. Outdate is typically 5 days, although processes teins. Because these carry a risk of disease transmis-
are currently available to allow transfusion of platelets sion, recombinant or xenographic preparations are an
up to 7 days after collection. alternative for many products.
 FFP must be prepared within 8 hours of collection and  Blood components are most commonly labeled with
is stored at –18°C for 12 months. ISBT 128, a specialized barcode language that includes
 Cryoprecipitate is prepared from FFP and contains at definitions for the donation identification number,
least 80 units of antihemophilic factor and at least blood type, product code, expiration date, and addi-
150 mg of fibrinogen, and it is stored at –18°C for tional special labels.

2. Each unit of cryoprecipitate prepared from whole blood


Review Questions
should contain a minimum of how many units of AHF
activity?
1. Which of the following lists the correct shelf life for the
component? a. 40 IU
b. 80 IU
a.Deglycerolized RBCs—24 hours
c. 120 IU
b.RBCs (CPD)—35 days
d. 160 IU
c.Platelet concentrate—10 days
e. 180 IU
d.FFP—5 years
e.RBCs (CPDA-1)—21 days
6888_Ch15_333-354 31/10/18 9:51 AM Page 353

Chapter 15 Component Preparation 353

3. Platelet concentrates prepared by apheresis should con- 10. RBCs that have been leukoreduced must contain less
tain how many platelets? than ______ leukocytes and retain at least ______ of
a. 5.5 × 1010 original RBCs.
b. 6 × 1010 a. 8 × 106/85%
c. 3 × 1011 b. 8 × 106/90%
d. 5.5 × 1011 c. 5 × 106/85%
e. 6 × 1011 d. 5 × 106/80%

4. The required storage temperature for frozen RBCs using 11. Random-donor platelets that have been leukoreduced
the high-glycerol method is: must contain less than ______ leukocytes.
a. 4°C a. 8.3 × 105
b. ≤–20°C b. 8 × 106
c. ≤–18°C c. 5 × 106
d. ≤–120°C d. 3 × 1011
e. ≤–65°C
12. A single unit of FFP or PF24 should contain ______ mL
5. How does irradiation affect the shelf life of red blood of plasma.
cells? a. 100–150
a. Irradiation has no effect on the shelf life b. 200–400
b. The expiration date is 28 days from the date of irradi- c. 150–250
ation or the original outdate, whichever is later d. 50–150
c. The expiration date is 28 days from the date of irradi-
13. Cryoprecipitate that has been pooled in an open system
ation or the original outdate, whichever is sooner
must be transfused within ______ hours.
d. The expiration date is 25 days from the date of irradi-
ation or the original outdate, whichever is later a. 24
e. The expiration date is 25 days from the date of irradi- b. 6
ation or the original outdate, whichever is sooner c. 4
d. 8
6. Once thawed, FFP must be transfused within __________
hours unless relabeled as thawed plasma: References
a. 4 1. AABB [Internet]. Bethesda (MD): AABB; c2017 [cited 2017
b. 6 May 6]. Highlights of transfusion medicine history. Available
c. 8 from: http://www.aabb.org/tm/Pages/highlights.aspx
d. 12 2. Spinella PC, Perkins JG, Grathwohl, KW, Beekley AC,
Holcomb JB. Warm fresh whole blood is independently asso-
e. 24
ciated with improved survival for patients with combat-related
7. Quality control for nonadditive RBCs requires a maxi- traumatic injuries. J Trauma. 2009; 66(4 Suppl):S69-6.
3. Spinella PC, Perkins JG, Grathwohl KW, Repine T, Beekley AC,
mum hematocrit level of: Sebesta DJ, et al. Fresh whole blood transfusions in coalition
a. 75% military, foreign national, and enemy combatant patients
b. 80% during Operation Iraqi Freedom at a U. S. Combat Support
c. 85% Hospital. World J Surg. 2008;32:2-6.
4. Perkins JG, Cap AP, Spinella PC, Shorr AF, Beekley AC,
d. 90%
Grathwohl KW, et al. Comparison of platelet transfusion as fresh
e. 95% whole blood versus apheresis platelets for massively transfused
combat trauma patients. Transfusion. 2011;41(2):242-52.
8. AHF concentrates are used to treat: 5. Seheult J, Triulzi DJ, Alarcon LH, Sperry JL, Murdock A, Yazer
a. Thrombocytopenia MH. ABO Antibody-mediated hemolysis is not detectable fol-
b. Hemophilia A lowing cold-stored uncrossmatched whole blood transfusion
c. Hemophilia B in civilian trauma patients. Transfusion. 2016; 16A.
6. AABB, ARC, ASBP, and ABC. Circular of information for the
d. von Willebrand’s disease use of human blood and blood components. Bethesda (MD):
e. Factor XIII deficiency AABB Press; 2017.
7. AABB. Standards for blood banks and transfusion services.
9. Prothrombin complex concentrates are used to treat 31st ed. Bethesda (MD): AABB Press; 2018.
which of the following? 8. Whitaker BI, Hinkins S. The 2011 national blood collection
a. Factor IX deficiency and utilization survey report. Rockville (MD): US Department
b. Factor VIII deficiency of Health and Human Services.
9. Code of Federal Regulations. Eligibility of donors, 21 C.F.R.
c. Factor XII deficiency
Sect 640.21 (2016).
d. Factor XIII deficiency 10. Murphy S. Platelets from pooled buffy coats: an update. Trans-
e. Factor V deficiency fusion. 2005; 634-9.
6888_Ch15_333-354 31/10/18 9:51 AM Page 354

354 PART III Transfusion Practices

11. Breecher ME, Hay S. Bacterial contamination of blood compo- 32. Hall TL, Barnes A, Miller JR, Bethencourt DM, Nestor L.
nents. Clin Microbiol Rev. 2005; 195-204. Neonatal mortality following transfusion of red cells with high
12. Jacobs MR, Palavecino E, Yomtovian R. Don’t bug me: the plasma potassium levels. Transfusion. 1993; 606-9.
problem of bacterial contamination of blood components– 33. Weiskopf RB, Schnapp S, Rouine-Rapp K, Bostrom A, Toy P.
challenges and solutions. Transfusion. 2001; 1331-4. Extracellular potassium concentrations in red blood cell
13. AABB. Association Bulletin 02-08: Update on bacterial con- suspensions after irradiation and washing. Transfusion. 2005;
tamination of platelet units [Internet]. Bethesda, MD: AABB. 1295-301.
2002 [cited 2017 May7]. Available from http:www.aabb.org/ 34. Obladen M, Sachsenweger M, Stahnke M. Blood sampling
programs/publications/bulletins/DocsObsolete/AB%2002-08% in very low birth weight infants receiving different levels of
20OBSOLETE.pdf. intensive care. Eur J Pediatr. 1988; 399-404.
14. Haemonetics. eBDS—the total solution for detecting bacteria in 35. Strauss RG, Burmeister LF, Johnson K, Cress G, Cordle, D.
platelets [Internet]. Braintree, (MA): Haemonetics; 2013 [cited Feasibility and safety of AS-3 red blood cells for neonatal trans-
2017 May 7]. Available from: http://wbt.haemonetics.com/~/ fusions. J Pediatr. 2000; 136: 215-219.
media/sharepoint/consumables/whole_blood/ebds/col-copy- 36. Food and Drug Administration. Fatalities reported to FDA
000527_us_ebds-poster.pdf. following blood collection and transfusion. Final report.
15. bioMerieux. BacT/ALERT 3D: product safety [Internet]. Washington (DC): Food and Drug Administration CBER;
Durham (NC): bioMerieux; 2017 [cited 2017 May 7]. Available 2014.
from: http://www.biomerieux-usa.com/industry/bact-alert-3d- 37. Menis M, Anderson SA, Forshee RA, McKean, S, Johnson C,
product-safety. Holness L, et al. Transfusion-associated circulatory overload
16. Dumont LJ, Kleinman S, Murphy JR, Lippincott R, Schuler R, (TACO) and potential risk factors among the inpatient US
Houton J, et al. Screeing of single-donor apheresis platelets for elderly as recorded in Me dicare administrative databases
bactial contaimination: the PASSPORT study results. Transfu- during 2011. VoxSang. 2014; 144-52.
sion. 2010; 589-99. 38. Murphy EL, Kwaan N, Looney MR, Gajic O, Hubmyar RD,
17. Fenwal, Inc. FDA clears Verax Platelet PGD test as safety mea- Gropper MA, et al. Risk factors and outcomes in transfusion-
sure for platelet transfusions. Lake Zurich (IL); 22 September associated circulatory overload. Am J Med. 2013; 357.e29-.e38.
2011. 39. AABB [Internet]. Bethesda (MD): AABB; 2015 [cited 2017
18. Fenwal, Inc. Fresenius Kabi receives FDA Clearance for 7-day May 12] Transfusion-associated circulatory overload. Associa-
platelet storage using Fenwal Amicus(R) System. Lake Zurich tion Bulletin. Available from: http://www.aabb.org/programs/
(IL); 3 August 2015. publications/bulletins/Pages/ab17-01.aspx
19. Terumo BCT. FDA clears seven-day platelet storage and wire- 40. McCullough J, Chopek M. Therapeutic plasma exchange. Lab
less capability for the Trima Accel System from Terumo BCT. Med. 1981; 745-53.
Lakewood (CO); 10 August 2015. 41. Smith AU. Prevention of haemolysis during freezing and thaw-
20. Food and Drug Administration. Draft Guidance for industry. ing of red blood cells. Lancet. 1950; 910.
Bacterial risk control strategies for blood collection establis- 42. Valeri CR, Ragno G, Pivacek LE, Cassidy GP, Srey R, Hancon-
ments and transfusion services to enhance the safety and avail- sicher M, et al. An experiment with glycerol-frozen red blood
ability of platelets for transfusion. Rockville (MD): US Health cells stored at –80°C for up to 37 years. Vox Sang. 2000; 168.
and Human Services CBER; 2016. 43. Suda BA, Leitman SF, Davery RJ. Characteristics of red cells
21. Food and Drug Administration. Guidance for Industry and FDA irradiated and subsequently frozen for long-term storage.
Review Staff. Collection of Platelets by Automated Methods. Transfusion. 1993; 389-92.
Rockville (MD). US Health and Human Services CBER; 2007. 44. Goodnough LT, Levy JH. The judicious use of recombinant fac-
22. Food and Drug Administration. Guidance for industry. Pre- tor VIIa. Semin Thromb Hemost. 2016; 125-32.
storage leukocyte reduction of whole blood and blood compo- 45. Heise D, Brauer A, Quintel M. Recombinant activated factor VII
nents intended for transfusion. Rockville (MD): US Health and (Novo7(R) ) in patients with ventricular assist devices: case
Human Services CBER; 2012. report and review of current literature. J Cardiothor Surg.
23. Shapiro MJ. To filter blood or universal leukoreduction: what 2007; 47.
is the answer? Crit Care. 2004; S27. 46. Hoffman M, Monroe DM. The action of high-dose factor VIIa
24. Heddle NM, Kelton JG. Febrile nonhemolytict transfusion in a cell-based model of hemostasis. Semin Hematol. 2001:
reactions. In: Popovsky, MA, editor. Transfusion reactions. 38(suppl 12):6-9.
Bethesda (MD): American Association of Blood Banks; 1996. 47. Roberts PL, Lloyd D, Marshall PJ. Virus inactivation in a
25. Food and Drug Administration. Guidance for industry. Pre- factor VIII/vWF concentrate treated using a solvent/detergent
storage leukocyte reduction of whole blood and blood compo- procedure based on polysorbate 20. J Biol. 2009; 26-31.
nents intended for transfusion. Rockville (MD): US Health and 48. Freedman J, Mody M, Lazarus AH, Dewar L, Song S,
Human Services CBER; 2012. Blanchette VS, et al. Platelet activation and hypercoagulability
26. Cable RG, Steele WR, Melmed RS, Johnsonn B, Mast AE, Carey following treatment with porcine factor VIII (HYATE:C). Am
PM, et al. The difference between fingerstick and venous J Hematol. 2002; 192-9.
hemoglobin and hematocrit varies by sex and iron stores. 49. Suiter TM. First and next generation native rVFIII in the treat-
Transfusion. 2012; 1031-40. ment of hemophilia A. What has been achieved? Can patients
27. Scott E, Puca K, Heraly J, Gottschall J, Friedman K. Valuation be switched safely? Semin Thromb Hemost. 2002; 277-84.
and comparison of coagulation factor activity in fresh-frozen 50. Roth DA, Kessler CM, Pasi KJ, Rup B, Courter SG, Tubridy KL.
plasma and 24-hour plasma at thaw and after 120 hours of Human recombinant factor IX: safety and efficacy studies in
1 to 6 C storage. Transfusion. 2009; 1584-91. hemophilia B patients previously treated with plasma-derived
28. Fung MK, Eder AF, Spitalnik SL, Westhoff CM (eds.). Technical factor IX concentrates. Blood. 2001; 3600-6.
manual. 19th ed. Bethesda (MD): AABB; 2017. 51. George JN. Initial management of adults with idiopathic
29. King KE, editor. Blood transfusion therapy: a physician’s (immune) thrombocytopenic purpura. Blood Rev. 2002;
handbook. 11th ed. Bethesda (MD): AABB; 2014. 37-8.
30. Sandler SG, Mallory D, Malamut D, Eckrich R. IgA anaphylac- 52. Konkle BA, Bauer KA, Weinstein R, Greist A, Holmes HE,
tic transfusion reactions. Lancet. 1968; 312-5. Bonfiglio J. Use of recombinant human antithrombin in
31. Fisch C. Relation of electrolyte disturbances to cardiac arrhyth- patients with congenital antithrombin deficiency undergoing
mias. Circulation. 1973; 408-19. surgical procedures. Transfusion. 2003;43:390-4.
6888_Ch16_355-372 29/10/18 5:44 PM Page 355

Chapter 16
Transfusion Therapy
Melanie S. Kennedy, MD; Denise Harmening, PhD, MT(ASCP);
and Justin Rhees, MS MLS(ASCP) CM, SBBCM

Introduction Antithrombin and Other Concentrates Transfusion in Oncology


Blood Products Albumin Coagulation Factor Deficiencies
Whole Blood Immune Globulin General Blood Transfusion Practices
Red Blood Cells Special Considerations for Transfusion Blood Administration
Leukocyte-Reduced RBCs Leukocyte-Reduced Cellular Blood Hospital Transfusion Committee
Washed RBCs and Frozen/ Components Case Studies
Deglycerolized RBCs CMV-Negative Cellular Blood Case 16-1
Rejuvenated Red Blood Cells Components Case 16-2
Platelets and Plateletpheresis Irradiated Cellular Blood Components Case 16-3
Granulocyte Pheresis Transfusion Therapy in Special Conditions Case 16-4
Plasma Surgical Blood Order Schedule, Type, Summary Chart
Cryoprecipitate and Screen
Review Questions
Thawed Plasma, Cryoprecipitate Autologous Transfusion
References
Reduced Emergency Transfusion
Factor VIII Massive Transfusion
Factor IX Neonatal Transfusion

OBJECTIVES
1. Describe the blood products that are currently available for therapeutic use.
2. List the indications for each blood product, including the approximate volume of each product.
3. Select the appropriate blood product for patients with specific disorders.
4. State the expected incremental increase of a patient’s hematocrit level following transfusion of each unit of red blood cells
(RBCs) and platelet count following transfusion of each unit of platelets.
5. List the required procedures for preparing each blood component for transfusion.
6. Identify the groups of recipients who are at highest risk of infection from transfusion of cytomegalovirus-positive RBCs or
platelets.
7. Explain the role of irradiation in the prevention of transfusion-associated graft-versus-host disease (GVHD).
8. State the purpose of the surgical blood order schedule.
9. List the main advantages and disadvantages of autologous transfusion.
10. Identify the most important factors to consider when emergency transfusion is indicated.
11. Define massive transfusion.
12. Describe the various transfusion requirements of oncology patients.
13. Compare and contrast hemophilia A and von Willebrand disease.
14. State the respective blood products of choice for treating von Willebrand disease and hemophilia B.
15. Specify the steps involved in the proper administration of blood.

355
6888_Ch16_355-372 29/10/18 5:44 PM Page 356

356 PART III Transfusion Practices

Introduction can receive whole blood, although RBCs and plasma are
more commonly used and are equally effective clinically.
“Transfusion therapy” is a broad term that encompasses all A definite contraindication to the use of whole blood is
aspects of the transfusion of patients. Each blood compo- severe chronic anemia. Patients with chronic anemia have a
nent has specific indications for use, expected outcomes, reduced number of RBCs but have compensated by increas-
and other considerations. In addition, patients with special ing their plasma volume to restore their total blood volume.
conditions require strategies and decisions to optimize ther- Thus, these patients do not need the plasma in the whole
apy. This chapter describes the selection and use of blood blood and, in fact, may adversely respond by developing pul-
components and the strategies for transfusion in special monary edema and heart failure because of volume overload.
conditions. This is more likely to occur in patients with kidney failure
or preexisting heart failure.
Blood Products For a 70-kg (154-lb) adult, each unit of whole blood
should increase the hematocrit level 3% or hemoglobin
Blood and blood products are considered drugs because of 1 g/dL. After transfusion, the increase may not be apparent
their use in treating diseases. As with drugs, adverse effects until 48 to 72 hours when the patient’s blood volume adjusts
may occur, necessitating careful consideration of therapy. to normal. For example, a patient with a 5,000-mL blood
The transfusion of blood cells is also transplantation, in volume and 20% hematocrit level has 1,000-mL RBCs. With
that the cells must survive and function after transfusion transfusion of 500-mL whole blood containing 200-mL RBCs,
to have a therapeutic effect. The transfusion of red blood the blood volume will be 5,500 mL and will result in
cells (RBCs), the best tolerated form of transplantation, can 21.8% hematocrit. When the patient’s blood volume readjusts
cause rejection, as in a hemolytic transfusion reaction. The to 5,000 mL, the hematocrit level will be 24% (1,200 mL
rejection of platelets, as shown by refractoriness to platelet divided by 5,000 mL). The increase is greater in a smaller
transfusion, is relatively common in multiply transfused person and less in a larger one.
patients.
Transfusion therapy is used primarily to treat two condi- Red Blood Cells
tions: inadequate oxygen-carrying capacity because of
anemia or blood loss and insufficient coagulation proteins RBCs are prepared from whole blood collected into any of
or platelets to provide adequate hemostasis. Each patient re- the anticoagulant-preservative solutions approved by the
quires an individualized plan that reflects his or her changing FDA and separated from the plasma by centrifugation and
clinical condition, anticipated blood loss, capacity for com- sedimentation. RBC transfusions are indicated for increasing
pensatory mechanisms, and laboratory results. Some patients the RBC mass in patients who require increased oxygen-
do not require transfusion, even in anemia or thrombocy- carrying capacity.1,2 These patients typically have pulse rates
topenia, because their clinical conditions are stable and they greater than 100 beats per minute; have respiration rates
have little or no risk of adverse outcomes. An example is a greater than 30 breaths per minute; and may experience
patient with iron-deficiency anemia with minor symptoms. dizziness, weakness, angina (chest pain), and difficulty
Appropriate blood therapy is the transfusion of the thinking. The decreased RBC mass may be caused by
specific blood product needed by the patient. By selecting decreased bone marrow production (leukemia or aplastic
blood products, several patients can be treated with the anemia), decreased RBC survival (hemolytic anemia), or sur-
blood from one donor, giving optimal use of every blood gical or traumatic bleeding.
donation. Table 16–1 provides a summary of the blood The human body compensates for anemia by increasing
products discussed in this chapter.1,2 plasma volume, heart rate, respiratory rate, and oxygen ex-
traction from the RBCs. Normally, only about 25% of the
Whole Blood oxygen is extracted, but with increased demand at the organ
and tissue level, up to 50% of the oxygen can be extracted.
When compared with the circulating blood in the donor’s When the demand exceeds 50% of the oxygen content, the
blood vessels, the product of whole blood is diluted in a compensatory mechanisms fail, and the patient requires
proportion of eight parts circulating blood to one part transfusion.
anticoagulant. The citrate in the anticoagulant chelates ion- No set hemoglobin levels indicate a need for transfusion.
ized calcium, preventing activation of the coagulation The critical level is 6 g/dL or less. Consensus committees sug-
system. The glucose, adenine, and phosphate (if present) gest trigger values of hemoglobin of less than 7 g/dL for most
serve as substrates for RBC metabolism during storage patients and less than or equal to 8 g/dL for certain patients
(see Chapter 1, “Red Blood Cell and Platelet Preservation: with heart disease.3 Most patients can tolerate 7 g/dL, espe-
Historical Perspectives and Current Trends”). cially if on bedrest or at decreased levels of activity and given
The transfusion of whole blood is limited to a few clinical supplemental oxygen.4 In fact, healthy individuals can tolerate
conditions. Whole blood must be ABO identical with the hemoglobin levels as low as 5 g/dL with minimal effects.
recipient and should be used to replace the loss of both RBC Transfusion of RBCs is contraindicated in patients who
mass and plasma volume.1,2 Thus, rapidly bleeding patients are well compensated for the anemia. RBCs should not be
6888_Ch16_355-372 29/10/18 5:44 PM Page 357

Chapter 16 Transfusion Therapy 357

Table 16–1 Blood Components and Plasma Derivatives


Category Major Indications Mode of Action Risks Special Considerations
Whole blood Symptomatic anemia with Increases oxygen- Infectious diseases Must be ABO identical
large-volume deficit carrying capacity
Hemolytic, septic/
Increases blood volume toxic, allergic,
febrile reactions
Iron overload, TACO,
TRALI, TA-GVHD

Red blood cells: RBCs Symptomatic anemia; red Increases oxygen- Same as whole Must be ABO compatible
(adenine-saline blood cell exchange carrying capacity blood category,
added), RBC pheresis transfusion above

Red blood cells: IgA deficiency with Deglycerolization Same as RBCs Must be ABO compatible
deglycerolized, anaphylactoid/ removes plasma above
washed anaphylactic reaction proteins.
Severe allergic reactions, Risk of allergic and Hemolysis due to
rare donors, symptomatic febrile reactions incomplete deglyc-
anemia; red blood cell reduced; increases erolization can
exchange transfusion oxygen-carrying occur.
capacity

Red blood cells: Symptomatic anemia Leukocyte reduction is Same as RBCs RBCs leukocyte-reduced must
leukocyte-reduced achieved by filtration: have a residual content of
Febrile reactions Hypotensive reaction
(1) soon after collec- leukocytes <5 × 106 WBC and
due to leukocyte may occur if
tion (prestorage) or ≥85% of the original red blood
antibodies bedside leukocyte-
(2) after varying cell content.
reduction filter is
Reduction of CMV periods of storage in
used.
transmission and HLA the laboratory.
alloimmunization

Platelets/apheresis Bleeding due to thrombo- Improves hemostasis Infectious diseases One unit of platelets derived
platelets cytopenia or platelet from a whole blood collection
Apheresis platelets may Septic/toxic, allergic,
function abnormality in- usually contains ≥5.5 × 1010
be HLA (or other febrile reactions
cluding antiplatelet drugs platelets suspended in 40 to
antigen) selected.
TACO, TRALI, 70 mL of plasma. One unit of
Prevention of bleeding
TA-GVHD apheresis platelets usually
from marrow hypoplasia
contains ≥ 3 × 1011 platelets/
unit and is the therapeutic
equivalent of 4 to 6 units of
platelets.

Platelets leukocytes Same as platelets above Same as platelets Same as platelets May be prepared using an open
reduced/apheresis above above or closed system. One unit of
Prevention of febrile
platelets leukocytes platelets leukocyte-reduced
reactions, HLA
reduced should contain >5.5 × 1010
alloimmunization and
platelets and <8.3 × 105 leuko-
CMV infection
cytes. Components prepared
using an open system will expire
4 hours after preparation.

Apheresis granulocytes Neutropenia with infection Increases the level of Infectious diseases Must be ABO compatible
unresponsive to appropri- granulocytes to
Hemolytic, allergic, Contain numerous leukocytes
ate antibiotics phagocytize and kill
febrile reactions and platelets as well as 20 to
bacterial and fungal
50 mL of RBCs; the number
infections TACO, TRALI,
of granulocytes in each concen-
TA-GVHD
trate is usually >1 × 1010
granulocytes/unit.

Plasma (liquid) Initial treatment of Coagulation support Infectious diseases Must be ABO compatible
patients for life-threatening
Allergic and febrile
trauma, hemorrhages
Undergoing massive reactions, TACO,
transfusion TRALI
Continued
6888_Ch16_355-372 29/10/18 5:44 PM Page 358

358 PART III Transfusion Practices

Table 16–1 Blood Components and Plasma Derivatives—cont’d


Category Major Indications Mode of Action Risks Special Considerations
Cryoprecipitate pooled/ Hypofibrinogenemia Provides fibrinogen Infectious diseases Must contain >150 mg of
cryoprecipitated AHF (factor I), factors VIII, fibrinogen in each unit of
Factor XIII deficiency Allergic and febrile
XIII, and vWF cryoprecipitate
(Second line therapy for reactions
von Willebrand’s disease, Cryoprecipitated AHF should
hemophilia A, and uremic contain >80 IU of factor VIII in
bleeding) each unit.

Fresh-frozen plasma Clinically significant plasma Source of all coagula- Infectious diseases Must be ABO compatible
(FFP) protein deficiencies when tion proteins and
Allergic and febrile
no specific coagulation plasma proteins
reactions, TACO,
factor concentrates are
TRALI
available
TTP

PF24 Same as FFP above Source of nonlabile Same as FFP above Must be ABO compatible
(Plasma frozen plasma proteins
within 24 hours Levels of factor VIII are
after phlebotomy) significantly reduced
and levels of factor V
and other labile
plasma proteins are
variable compared
with FFP.

PF24RT24 Clinically significant Source of nonlabile Same as FFP Must be ABO compatible
(Plasma frozen deficiency of stable plasma proteins
within 24 hours after coagulation factors when
Levels of factor V, factor
phlebotomy held at no specific coagulation
VIII, and protein S are
room temperature factor concentrates are
significantly reduced,
up to 24 hours after available
and levels of other
phlebotomy) labile plasma proteins
TTP
are variable compared
with FFP.

Thawed plasma Clinically significant defi- Source of plasma Same as FFP Must be ABO compatible
ciency of stable coagula- proteins and contains
tion factors when no stable coagulation
specific coagulation factor factors (factor II,
concentrates are available fibrinogen), similar
clinically to the levels
TTP
found in FFP; other
factors are variable,
being reduced in
levels that change
over time.

Thawed plasma, cryo- TTP Plasma protein Same as FFP Must be ABO compatible
precipitate reduced replacement for
Product is deficient in factors I
plasma exchange in
(fibrinogen), VIII, XIII, as well
TTP; this product
as vWF, cryoglobulin, and
contains variable
fibronectin.
levels of albumin,
ADAMTS13, and
factors II, V, VII, IX, X,
and XI.

TACO = transfusion-associated circulatory overload; TRALI = transfusion-related acute lung injury; TA-GVHD = transfusion-associated graft-vs-host disease; CMV = cytomegalovirus;
TTP = thrombotic thrombocytopenic purpura; AHF = antihemophilic factor; ITP = immune thrombocytopenic purpura; vWF = von Willebrand factor; HLA = Human Leukocyte
Antigen; IUT = intrauterine transfusion
Modified from: Food and Drug Administration, Guidance for Industry: Circular of Information for the Use of Human Blood and Blood Products. AABB, America’s Blood Centers,
American Red Cross (ARC), and the Armed Services Blood Program (ASBP), December, 2017. Available from: https://www.fda.gov/downloads/BiologicsBloodVaccines/
GuidanceComplianceRegulatoryInformation/Guidances/Blood/UCM364593.pdf
6888_Ch16_355-372 29/10/18 5:44 PM Page 359

Chapter 16 Transfusion Therapy 359

used to treat nutritional anemia, such as iron deficiency or Table 16–2 Indications for Leukocyte-
pernicious anemia, unless the patient shows signs of decom-
Reduced RBCs and Platelets
pensation (need for increased oxygen-carrying capacity).5
RBC transfusion is not to be used to enhance general well- Accepted Controversial
being, promote wound healing, prevent infection, expand Decrease febrile nonhemolytic Decrease hospital length of stay
blood volume when oxygen-carrying capacity is adequate, transfusion reactions
or prevent future anemia.
Each unit of transfused RBCs is expected to increase Decrease alloimmunization to Decrease incidence of wound
white blood cell antigens infections postsurgery
the hemoglobin level 1 g/dL and the hematocrit level
3% in the typical 70-kg (154-lb) human, the same as whole Decrease transmission of Decrease incidence of cancer
blood. In pediatric patients, a dose of 10 to 15 mL/kg will cytomegalovirus (CMV) recurrence postsurgery
increase the hemoglobin about 2 to 3 g/dL or the hemat-
ocrit 6% to 9%.6 This may vary dependent upon the child’s
age and body mass. washed RBCs.1 The washing process removes plasma proteins,
The increase in hemoglobin and hematocrit is evident the cause of most allergic reactions. Washed RBCs can be used
more quickly than with 1 unit of whole blood, because the for the rare patient who has had moderate to severe allergic
adjustment in blood volume is less. As in the example for transfusion reactions and/or has anti-IgA or anti-haptoglobin
whole blood, the RBC volume is increased to the same antibodies.
amount, 1,200 mL, but the blood volume is increased only Freezing RBCs allows the long-term storage of rare blood
330 mL to 5,330 mL. The hematocrit level is increased donor units, autologous units, and units for special purposes,
immediately to 22.5%. such as intrauterine transfusion. Because the process needed
RBCs prepared with additive solutions such as Adsol have to deglycerolize the RBCs removes nearly all the plasma, these
greater volume than with citrate phosphate dextrose (CPD) units, although more expensive, can be used interchangeably
or citrate phosphate dextrose adenine (CPDA-1), 300 to with washed RBCs. However, the shortened outdate of washed
400 mL versus 160 to 275 mL (see Chapter 1), but the addi- or deglycerolized RBCs severely limits the use of these com-
tive solution units have less plasma. The RBC mass is the ponents. If the units were deglycerolized and washed using
same. Therefore, the hematocrit differs from 65% to 80% for an open system, then the RBCs must be transfused within
CPDA-1 RBCs to 55% to 65% for additive solution RBCs. 24 hours after thawing. If a closed system was used, then the
units may be used for up to 2 weeks after thawing. Deglyc-
Leukocyte-Reduced RBCs erolized RBCs contain 80% or more of the erythrocytes present
in the original unit of blood and have approximately the same
The average unit of leukocyte-reduced RBCs contains less expected post-transfusion survival as RBCs. The expected
than 5 × 106 leukocytes. Donor leukocytes may cause febrile hematocrit increase for washed or deglycerolized RBCs is the
nonhemolytic transfusion reactions, transfusion-associated same as that for regular RBC units.
graft-versus-host disease (TA-GVHD), and transfusion-related
immune suppression also known as transfusion-induced Rejuvenated Red Blood Cells
immunomodulation (TRIM). In addition, human leukocyte
antigens (HLA) are responsible for HLA alloimmunization. This product may be prepared from RBCs stored in CPD,
Leukocytes may harbor cytomegalovirus (CMV). CPDA-1, and AS-1 storage solutions up to 3 days after expi-
To reduce the risk of HLA alloimmunization and CMV ration. An FDA-approved solution of inosine, phosphate, and
transmission, the leukocyte content must be reduced to less adenine rejuvenates and restores 2, 3-diphosphoglycerate and
than 5 × 106, which can be achieved by using one of several ATP levels to approximately freshly drawn RBCs. These
leukocyte-reduction filters.5 After leukoreduction with these products must be washed before infusion to remove the
filters, most leukocyte counts are less than 1 × 106. In the inosine, which may have toxicity. Rejuvenated RBCs after
United States, the standard leukocyte content must be less preparation must be transfused within 24 hours or frozen
than 5 × 106. The effect of leukocyte-reduced blood on for long-term storage.
length of hospital stay and postsurgical wound infection is
controversial. However, febrile nonhemolytic transfusion Platelets and Plateletpheresis
reactions, CMV transmission, and HLA alloimmunization
are decreased by the use of leukocyte-reduced RBCs and Platelets are essential for the formation of the primary
platelets.6 It is important to note that leukoreduction does hemostatic plug and maintenance of normal hemostasis.
not reduce the risk of TA-GVHD. The indications for trans- Patients with severe thrombocytopenia (low platelet count)
fusion of leukocyte-reduced RBCs and platelets are outlined or abnormal platelet function may have petechiae, ecchy-
in Table 16–2. moses, and mucosal or spontaneous hemorrhage. The throm-
bocytopenia may be caused by decreased platelet production
Washed RBCs and Frozen/Deglycerolized RBCs (e.g., after chemotherapy for malignancy) or increased
destruction (e.g., disseminated intravascular coagulation
Patients who have severe allergic (anaphylactic) transfusion [DIC]). Massive transfusion, which is discussed later in this
reactions to ordinary units of RBCs may benefit from receiving chapter, may also cause thrombocytopenia because of the
6888_Ch16_355-372 29/10/18 5:44 PM Page 360

360 PART III Transfusion Practices

rapid consumption of platelets for hemostasis and the platelet donors can be time-consuming. HLA-matched
dilution of the platelets by resuscitation fluids and RBC platelets should be irradiated.
transfusion. A corrected count increment using a 10-minute to 1-hour
Platelet transfusions are indicated for patients who are post-transfusion platelet count can provide valuable infor-
bleeding because of thrombocytopenia or abnormally func- mation about patient response to a platelet component.1 The
tioning platelets (Box 16–1). In addition, platelets are indi- platelet count increment is corrected for differences in body
cated as prophylaxis for patients who have platelet counts size so that more reliable estimates of expected platelet in-
under 5,000 to 10,000/µL if the patient is clinically stable crement can be determined. The expected corrected platelet
with an intact vascular system and normal platelet function.7 increment is greater than 10,000/µL/ m2. One formula for
Additional guidelines were published in 2015 by the AABB corrected count increment is
as part of a clinical practice guideline and address indications
in a variety of settings.7 Absolute platelet increment/µL × body surface area (m2),
Bacterial testing is required for each platelet product. Number of platelets transfused (1011)
Plateletpheresis products and pooled platelets (from whole in which the absolute platelet increment is the post-
blood) are cultured by the blood center. However, individual transfusion platelet count minus the pretransfusion platelet
platelet products from whole blood are difficult to culture count, the body surface area (BSA) is expressed as square
because of their small volume and thus are less commonly meters, and the number of platelets transfused is 3 for
used for transfusion. plateletpheresis or pooled platelets (the number of platelets
A plateletpheresis component is prepared from one donor in each unit expressed in 1011). It should be noted that BSA
and must contain a minimum of 3 × 1011 platelets.5 One calculators are available online.
plateletpheresis unit should increase the adult patient’s For example, a patient with a 10,000/µL platelet count
platelet count to 20,000 to 60,000/µL. Each unit of platelets has a body surface area of 1.3 m2. One unit of plateletphere-
from whole blood must contain at least 5.5 × 1010 platelets5 sis is given. The 1-hour post-transfusion platelet count is
and should increase the platelet count by 5,000 to 10,000/µL 50,000/µL. Put these into the formula
in a 70-kg patient. A pool of 4 to 6 units, then, will contain
roughly 3 × 1011 platelets and should give a platelet count (50,000/µL – 10,000/µL) × 1.3.
increase similar to plateletpheresis. 3
The answer, 17,333, shows that the patient has a good
Refractory Patients
increment (greater than 10,000/µL) and is not refractory
to platelets. An answer less than 5,000/µL indicates
Advanced Concepts refractoriness.
Massive splenomegaly, high fever, sepsis, disseminated In addition to HLA- and platelet-specific antigens, ABO
intravascular coagulation (DIC), and platelet or HLA antigens are also expressed on the platelet membrane.
antibodies can cause less-than-expected platelet count Sometimes platelets are selected for transfusion without
increment and survival. The 10-minute to 1-hour post- regard to ABO; however, group O recipients may have a
transfusion platelet count increment is less affected by lower increment when given group A platelets than when
splenomegaly, high fever, and DIC than by the presence of group-identical platelets are selected.7 Group A, B, and
platelet or HLA antibodies.1 If the 10-minute increment is AB patients may also develop a positive direct antiglobu-
less than 50% of that expected on two occasions, the pa- lin test owing to passive transfer of anti-A, anti-B, or
tient is considered refractory. Positive platelet crossmatch anti-A,B when several ABO-incompatible platelet transfu-
or positive HLA antibody screen is considered evidence of sions are given.
alloimmunization. Platelet crossmatching with available in- Although platelet membranes do not express Rh anti-
ventory can speed the provision of platelets for transfusion, gens, platelet products contain small amounts of RBCs and
as HLA-typing the patient and recruiting HLA-compatible thus can immunize patients to Rh antigens. Rh-immune
globulin can be given to patients, especially girls and
women of childbearing potential, to prevent Rh sensitiza-
tion. Each 300-µg vial of Rh-immune globulin is adequate
for about 30 plateletphereses or four platelet pools of five.
BOX 16–1 Some institutions may administers RhIg to male and older
Indications for Platelet Transfusion female patients to prevent alloimmunization.
• Thrombocytopenia with bleeding or invasive procedure
• Chemotherapy for malignancy (decreased production, less than For the same reasons as for RBCs, platelet components
5,000 to 10,000/µL)
may also be leukocyte-reduced or washed. Washing platelet
• Disseminated intravascular coagulation (increased destruction,
less than 50,000/µL)
components removes some platelets and plasma proteins
• Massive transfusion (platelet dilution, less than 50,000 to
and, if using an open method, requires a 4-hour expiration
100,000/µL) time. Platelet function may also be negatively impacted due
to platelet adhesion and activation during the wash cycles.
6888_Ch16_355-372 29/10/18 5:44 PM Page 361

Chapter 16 Transfusion Therapy 361

Therefore, platelet components should be washed only to at –18°C or colder. Similar to FFP and PF24, PF24RT24
prevent severe allergic reactions or to remove alloantibodies should be infused immediately after thawing or stored
in cases of neonatal alloimmune thrombocytopenia. at 1° to 6°C. After 24 hours, the component must be dis-
carded, or if collected in a functionally closed system, may
Granulocyte Pheresis be relabeled as “thawed plasma.” Thawed plasma derived
from FFP, PF24, or PF24RT24 (prepared in a closed system)
Patients who have received intensive chemotherapy for is thawed at 30° to 37°C and stored at 1° to 6°C for up
leukemia or bone marrow transplant or both may develop to 4 days after the initial 24-hour post-thaw period. Thawed
severe neutropenia and serious bacterial or fungal infection. plasma contains stable coagulation factors (factor II, fibrino-
Without neutrophils (granulocytes), the patient may have gen), similar clinically to the levels found in FFP. Other
difficulty controlling an infection, even with appropriate factors are variable, being reduced in levels that change
antibiotic treatment. Criteria have been developed to iden- over time.
tify patients who are most likely to benefit from granulo- Plasma can be used to treat multiple coagulation deficien-
cyte transxfusions: those with fever, neutrophil counts less cies occurring in patients with liver failure, DIC, vitamin K
than 500/µL, septicemia or bacterial infection unresponsive deficiency, warfarin overdose, or massive transfusion.8,9
to antibiotics, reversible bone marrow hypoplasia, and a Sometimes, plasma is used to treat patients with single factor
reasonable chance for survival.1 Prophylactic use of gran- deficiencies, such as factor XI deficiency.
ulocyte transfusions is of doubtful value for those patients Vitamin K deficiency or warfarin overdose should be
who have neutropenia but no demonstrable infection. treated with vitamin K orally, intravenously, or intramuscu-
Newborn infants may develop overwhelming infection larly if liver function is adequate and with an adequate
with neutropenia because of their limited bone marrow re- interval (4 to 24 hours) before a major or minor hemostatic
serve for neutrophil production. In addition, neonatal neu- challenge such as surgery. Plasma is given if the patient is
trophils have impaired function. Studies show granulocyte actively bleeding or if time is not available for warfarin
transfusions to be beneficial for these patients. For an adult reversal before surgery.
or a child, the usual dose is one granulocyte pheresis product Patients with liver disease or liver failure frequently de-
daily for 4 or more days. For neonates, a portion of a granu- velop clinical coagulopathy due to impaired hepatic synthesis
locyte pheresis unit is usually given once or twice. of all coagulation factors and antithrombotic factors. Plasma
Granulocyte components usually contain greater than is the product of choice for patients with multiple-factor
1.0 × 1010 granulocytes as well as platelets and 20 to 50 mL deficiencies and hemorrhage or impending surgery. Usually 4
of erythrocytes. Because most patients receiving granulocytes to 6 units of plasma will effectively control hemostasis. Even
are immunocompromised, the products are often irradiated so, plasma may not correct coagulation tests to normal range
to prevent TA-GVHD. If granulocytes cannot be transfused because of dysfibrinogenemia. Mild hemostatic abnormalities
immediately, they may be stored at 20° to 24°C without agi- do not predict bleeding, so correction is not indicated for
tation.5 The granulocyte pheresis needs to be crossmatched minor procedures, such as liver biopsy. In addition, plasma is
because of the significant content of RBCs. The patient must not a concentrate, so volume overload may be a serious com-
be monitored for resolution of symptoms and clinical evi- plication of transfusion.
dence of efficacy. The neutrophil count will increase to Congenital coagulation factor deficiencies are rarely treated
1,000/µL or more in response to infusion of granulocyte with plasma because the dose requirement for surgical proce-
colony-stimulating factor (GCSF)–mobilized granulocyte dures and serious bleeding is so great as to cause pulmonary
pheresis. edema as a result of volume overload, even in a young indi-
vidual with a healthy cardiovascular system. Factor concen-
Plasma trates (see the “Factor VIII” and “Factor IX” sections below)
offer more effective modes of therapy. Factor XI deficiency,
Plasma includes fresh-frozen plasma (FFP), plasma frozen however, is still treated by plasma infusion, requiring 20% to
within 24 hours after phlebotomy (PF24), and thawed 30% factor XI levels for adequate hemostasis. This disease is
plasma. FFP and PF24 contain all nonlabile coagulation fac- milder than hemophilia A (factor VIII deficiency) or hemo-
tors, and FFP contains normal levels of the labile coagula- philia B (factor IX deficiency). Factor XI also has a long half-
tion factors V and VIII. PF24 contains reduced levels of life, so treatment is not needed on a daily basis.
factor VIII and protein C, while levels of factor V and other A coagulation factor unit is defined as the activity in 1 mL
labile plasma proteins are variable compared with FFP. After of pooled normal plasma, so 100% activity is 1 unit/mL or
FFP and PF24 are thawed, they should be infused immedi- 100 units/dL. About 30% activity of each of the coagulation
ately or stored at 1° to 6°C. After 24 hours, FFP and PF24 factors is required for adequate hemostasis. Thus, less than
must be discarded, or if collected in a functionally closed half of the plasma volume, or about 4 to 6 plasma units, is
system, may be relabeled as “thawed plasma.” Plasma frozen required to correct a coagulopathy such as in liver disease
within 24 hours and held at room temperature up to or DIC. With continued hemorrhage, additional doses are
24 hours after phlebotomy (PF24RT24) is prepared from usually needed if the prothrombin time (PT) is more than
apheresis collections. PF24RT24 can be held at room tem- 1.5 times normal or if the international normalized ratio is
perature for up to 24 hours after collection and then frozen greater than 1.5. Because several key clotting factors, such
6888_Ch16_355-372 29/10/18 5:44 PM Page 362

362 PART III Transfusion Practices

as factor VII, VIII, or IX, have half-lives less than 24 hours, deficiency or von Willebrand disease.9 Virus-safe factor VIII
repeated transfusions are required to control postoperative with assayed amounts of factor VIII and vWF is available.
bleeding or to maintain hemostasis. For example, factor IX
has a half-life of 18 to 24 hours, requiring daily transfusions. Thawed Plasma, Cryoprecipitate Reduced
Plasma is sometimes used as a replacement fluid during
plasma exchange (therapeutic plasmapheresis; see Chapter 18, Thawed plasma, cryoprecipitate reduced is thawed at 30°
“Apheresis”). In cases of thrombotic thrombocytopenic pur- to 37°C and maintained at 1° to 6°C for up to 4 days after
pura (TTP), plasma provides a metalloprotease (ADAMTS13) the initial 24-hour post-thaw period has elapsed. This prod-
and removes inhibitors, thus reversing the symptoms. uct contains variable levels of albumin, ADAMTS13, and
Plasma should not be used for blood volume expansion factors II, V, VII, IX, X, and XI, but is deficient in factors I
or protein replacement because safer products are available (fibrinogen),VIII, XIII, as well as vWF, cryoglobulin, and
for these purposes—serum albumin, synthetic colloids, and fibronectin.
balanced salt solutions—none of which transmit disease or
cause severe allergic reactions or transfusion-associated acute Factor VIII
lung injury.
Patients with hemophilia A or factor VIII deficiency have
Plasma should be ABO compatible with the recipient’s
spontaneous hemorrhages that are treated with recombinant
RBCs, but the Rh type can be disregarded.
or human plasma–derived factor VIII replacement.10 Plasma-
derived factor VIII is prepared from plasma obtained from
Cryoprecipitate
paid donors by plasmapheresis or from volunteer whole
Cryoprecipitate contains fibrinogen, factor VIII, factor XIII, blood donors. Factor VIII is treated by different methods,
vWF, and fibronectin. However, it is used primarily for such as pasteurization, nanofiltration, and solvent detergent,
fibrinogen replacement. The AABB requires that more than to ensure sterility for HIV, hepatitis B virus, and hepatitis C
150 mg of fibrinogen be in each unit of cryoprecipitate,5 virus (HCV).11 The recombinant human product is virus-
although quality control levels are often over 250 mg. Cry- safe.12 Recombinant human products and their indications
oprecipitated antihemophilic factor (AHF) should contain are listed in Table 16–3.12
greater than or equal to 80 IU of factor VIII in each unit. Both the plasma derived and recombinant factor VIII are
Generally, five cryoprecipitate units are pooled at the blood stored at refrigerator temperatures and are reconstituted with
center and are labeled “pooled cryoprecipitated AHF.” Each saline at the time of infusion. This ease of handling allows
of the units is rinsed with 10 to 15 mL of saline diluent self-therapy for individuals with hemophilia.
to ensure complete removal of all material. These pools The following example illustrates the calculation of a
are frozen and shipped to transfusion services where the factor VIII dose.
pools can be thawed and issued. Each pool contains 750 to A 70-kg hemophiliac patient with a hematocrit level
1,250 mg of fibrinogen. of 30% has an initial factor VIII level of 4% (4 units/dL,
Fibrinogen replacement may be required in patients with 0.04 units/mL). How many units of factor VIII concentrate
liver failure, DIC, or massive transfusion and in rare patients should be given to raise his factor VIII level to 50%?
with congenital fibrinogen deficiency. A fibrinogen plasma {desired factor VIII (units/mL) – initial factor VIII
level of about 100 mg/dL is recommended for adequate (units/mL)} × plasma volume (mL) = units of factor VIII
hemostasis with surgery or trauma. For example, a patient’s required.
fibrinogen must be increased from 30 to 100 mg/dL, or an
increment of 70 mg/dL (100 – 30 mg/dL). When used to • Blood volume = weight (kg) × 70 mL/kg.
correct hypofibrinogenemia, cryoprecipitated AHF may be • 70 kg × 70 mL/kg = 4,900 mL.
dosed at one bag per 7- to 10-kg body weight to raise plasma
fibrinogen by approximately 50 to 75 mg/dL.2
Cryoprecipitate was used as a source for fibrin sealant, Table 16–3 Recombinant Human Products
which uses cryoprecipitate as the source of fibrinogen. Product Indications
FDA-approved fibrin sealants, which have been treated to
reduce viral transmission, are preferred. Factor VIII Hemophilia A, von Willebrand’s disease
Cryoprecipitate was originally prepared as a source of Factor IX Hemophilia B
factor VIII. Each unit of cryoprecipitate must contain at least
80 units of factor VIII. However, mild or moderate factor VIII Factor VIIa Inhibitors in hemophilia A or B
deficiency (hemophilia A) is now treated with desmopressin Factor VII deficiency
acetate (1-deamino-[8-D-arginine]-vasopressin [DDAVP]) or Acquired factor VII deficiency*
factor VIII, or both, whereas severe factor VIII deficiency is • Liver disease
treated with only factor VIII. • Warfarin overdose
Cryoprecipitate was used to treat patients with von Massive hemorrhage*
Willebrand disease, a deficiency of vWF. Cryoprecipitate is
no longer considered the product of choice for factor VIII *Randomized controlled trials needed.
6888_Ch16_355-372 29/10/18 5:44 PM Page 363

Chapter 16 Transfusion Therapy 363

• Plasma volume = blood volume (mL) × (1.0 – Hct). Albumin


• 4,900 mL × (1.0 – 0.30) = 3,430 mL.
• Solution: 3,430 mL × (0.50 – 0.04) = 1,578 units. Albumin is prepared by chemical and physical fractionation of
• The assayed value on the label can be divided into the pooled plasma. Albumin is available as a 5% or a 25% solution,
number of units required to obtain the number of vials to of which 96% of the protein content is albumin. The product
be infused. is heat-treated and has proved to be virus-safe over many years
of use.
Only factor VIII products labeled as containing vWF Albumin may be used to treat patients requiring volume
should be used for patients with von Willebrand disease. replacement. Whether albumin or colloids other than crys-
talloid (i.e., saline or electrolyte) solutions are better for
Factor IX treating hypovolemia with shock is controversial. In many
Factor IX complex (prothrombin complex) is prepared from plasmapheresis procedures, albumin is used routinely as the
pooled plasma using various methods of separation and viral replacement fluid for the colloid that is removed during the
inactivation. The prothrombin complex contains factors II, VII, procedures. Albumin can also be used in the treatment of
IX, and X; however, the product is recommended for factor IX– burn patients to replace colloid pressure.
deficient patients (hemophilia B), patients with factor VII or X Albumin, with diuretics, can induce diuresis in patients
deficiency (rare), and selected patients with factor VIII in- who have low total protein because of severe liver or protein-
hibitors or reversal of warfarin overdose.10 Activated coagula- losing disease. The 25% solution brings about five times its
tion factors present in prothrombin complex may cause volume from extravascular water into the vascular space.
thrombosis, especially in patients with liver disease. Recombi- Thus, patients receiving 25% albumin need to have adequate
nant human factor IX (see Table 16–3) is effective only in the extravascular water and compensatory mechanisms to deal
management of factor IX deficiency (hemophilia B). with the expansion of the blood volume.
The dose is calculated in the same manner as that for
factor VIII concentrate, using the assayed value of factor IX Immune Globulin
on the label, with the caveat that half the dose of factor IX
Immune globulin prepared from pooled plasma is primarily
rapidly diffuses into tissues and half remains within the
IgG. Although small amounts of IgM and IgA may be present
intravascular space, so the initial dose must be doubled.
in some preparations, others are free of these contaminating
Antithrombin and Other Concentrates proteins. Products are available for intramuscular or intra-
venous administration. The intramuscular product must not
Antithrombin is a protease inhibitor with activity toward be given intravenously because severe anaphylactic reactions
thrombin. Heparin accelerates the binding and inactivation may occur. The intravenous product must be given slowly
of thrombin by antithrombin. The hereditary deficiency of to lessen the risk of reaction.
antithrombin is associated with venous thromboses, whereas Immune globulin is used for patients with congenital
the acquired deficiency is seen most frequently with DIC. hypogammaglobulinemia11 and for patients exposed to dis-
Antithrombin concentrates are licensed for use in the eases such as hepatitis A or measles. For hypogammaglobu-
United States for patients with hereditary deficiency of anti- linemia, monthly injections/infusions are usually given
thrombin. The product is pasteurized to eliminate the risk because of the 22-day half-life of IgG. The recommended
of HIV or HCV infections. Antithrombin has been shown to dose is 0.7 mL/kg intramuscularly or 100 mg/kg intra-
provide no significant clinical benefit in acquired deficiency. venously. For hepatitis A prophylaxis, 0.02 to 0.04 mL/kg
Thawed plasma is an alternative source of antithrombin. intramuscularly is recommended.
Protein C and protein S are vitamin K–dependent proteins The intravenous preparation of immune globulin is used
synthesized in the liver. Protein S functions as a cofactor increasingly in the therapy of autoimmune diseases, such as
for activated protein C, which, in turn, inactivates factors V immune thrombocytopenia11 and myasthenia gravis. Various
and VIII, thus preventing thrombus formation. Deficiency mechanisms of action have been postulated. Conceivably,
(hereditary or acquired) leads to a hypercoagulable state (i.e., the infused immune globulin blocks the reticuloendothelial
the tendency for thrombosis). Human plasma–derived pro- system or mononuclear phagocytic system.
tein C concentrates are approved for use in hereditary defi- Various hyperimmune globulins are available to treat such
ciency states. Recombinant human activated protein C has viruses as hepatitis B, varicella zoster, rabies, mumps, and
been used for DIC and sepsis. others. These are prepared from the plasma of donors who
Recombinant human activated factor VII (rFVIIa) has have high antibody titers to the specific virus causing the
been used to control bleeding episodes in hemophilia A and disease. The dose is recommended in the package insert. It
B patients with inhibitors.10,12 rFVIIa has been used in should be noted that preparations such as hepatitis B hyper-
patients with a wide variety of bleeding disorders. However, immune globulin provide only passive immunity after an
large randomized controlled trials are needed to define dose, exposure. They do not confer permanent immunity and so
indications, and adverse effects. Reports of use for liver must be accompanied by active immunization.
disease, massive transfusion, and other bleeding disorders Rh-immune globulin (RhIG) was developed to protect
have been promising.13 the Rh-negative female who is pregnant or who delivers an
6888_Ch16_355-372 29/10/18 5:44 PM Page 364

364 PART III Transfusion Practices

Rh-positive infant (see Chapter 20, “Hemolytic Disease of Leukocyte-reduced RBCs and platelets can be used to pre-
the Fetus and Newborn [HDFN]”). Much of the IgG in this vent febrile nonhemolytic transfusion reactions, to prevent
preparation is directed against the D antigen within the Rh or delay the development of HLA antibodies, and to reduce
system. Administration of this preparation allows attachment the risk of CMV transmission. Controversial effects of leuko-
of anti-D to any Rh-positive cells of the infant that have en- cyte reduction include decreased mortality and length of
tered the maternal circulation. The antibody-bound cells are hospital stay.1
subsequently removed by the mother’s macrophages, pre-
venting active immunization or sensitization. CMV-Negative Cellular Blood Components
Rh-immune globulin products, which can be adminis-
tered intravenously or intramuscularly, are approved for use CMV is carried, in a latent or infectious form, in neu-
in idiopathic thrombocytopenic purpura patients who are trophils and monocytes. Transfusion of these virus-
Rh-positive.1 The proposed mechanism of action is blockage infected cells in a cellular product such as RBCs or platelets
of the reticuloendothelial system by anti–D-coated RBCs, can transmit infection. CMV infection of the patients can
thereby reducing the destruction of autoantibody-coated be reduced by using leukocyte-reduction filters15 or by
platelets. providing CMV antibody–negative blood. CMV-negative or
For RBC transfusion accidents, the number of standard- leukocyte-reduced components are indicated for recipients
dose RhIG vials is calculated by dividing the volume of who are CMV-negative and at risk for severe sequelae of
Rh-positive packed RBCs transfused by 15 mL, the amount CMV infections.1 The risk is greatest for CMV-negative
of RBCs covered by one vial. The number of vials can be pregnant women (mainly for the benefit of the fetus),
large, so the entire dose is often divided and administered allogeneic CMV-negative bone marrow and hematopoietic
in several injections at separate sites and over 3 days. The progenitor cell transplant recipients, and premature infants
intravenous preparation may also be used. Another weighing less than 1,200 g.
approach is to perform an exchange transfusion with Rh-
negative blood and then calculate the dose based on the Irradiated Cellular Blood Components
number of Rh-positive RBCs remaining in the circulation.
Blood components are irradiated with gamma radiation
For plateletpheresis, one vial is sufficient for 30 or more
to prevent graft-versus-host disease, which requires three
products because each unit contains fewer than 0.5 mL
conditions to occur:
RBCs. The dose for leukocyte concentrates can be calcu-
lated by obtaining the hematocrit and volume of the prod- 1. Transfusion or transplantation of immunocompetent T
uct from the supplier. lymphocytes
Immune globulins may cause anaphylactic reactions 2. Histocompatibility differences between graft and recipi-
(flushing, hypotension, dyspnea, nausea, vomiting, diarrhea, ent (major or minor HLA or other histocompatibility
and back pain). Caution should be used in patients with antigens)
known IgA deficiency and previous anaphylactic reactions 3. Usually, an immunocompromised recipient16
to blood components.
Common after allogeneic bone marrow or hematopoietic
progenitor cell transplantation, GVHD is a syndrome affect-
Special Considerations for Transfusion
ing mainly skin, liver, and gut (see Chapter 19, “Cellular
Certain categories of patients require the selection of blood Therapy in the Transplant Setting”).
products that are leukocyte-reduced, CMV-negative, or irra- Transfusion-associated TA-GVHD, occurring less fre-
diated. These special products are detailed here. quently, is caused by viable T lymphocytes in cellular blood
components (e.g., RBCs and platelets). The mortality rate is
Leukocyte-Reduced Cellular Blood Components high;16 therefore, prevention is key. Prevention focuses on
irradiating cellular components before administration to
Leukocyte-reduction filters are designed to remove more significantly immunocompromised individuals. Irradiation
than 99.9% of leukocytes from RBCs and platelet products. doses can vary, with higher doses being more effective but
They can be used in the laboratory (prestorage) or be used more damaging to RBCs. However, the standard dose of
at the bedside during blood transfusion. gamma irradiation is 2,500 cGy (centigray) targeted to the
The goal is fewer than 5 × 106 leukocytes remaining central portion of the container with a minimum dose of
in the RBC or apheresed platelet unit. Prestorage filtration 1,500 cGy delivered to any part of the component. Irradia-
in the laboratory or at the time of collection, rather than at tion decreases or eliminates the mitogenic (blastogenic)
the bedside, is more reliable for leukocyte reduction. This is capacity of the transfused T cells, rendering the donor T cells
because leukocytes degranulate, fragment, or die during stor- immunoincompetent.
age, releasing their contents that could result in febrile At risk for TA-GVHD are transfusion recipients with con-
and allergic transfusion reactions. The cytokines, especially genital immunodeficiencies (severe combined immunodefi-
IL-8, that accumulate during storage have been implicated ciency, DiGeorge syndrome, Wiskott-Aldrich syndrome),
for some failures of bedside filtration to prevention febrile Hodgkin lymphoma, bone marrow transplants (allogeneic
reaction.14 or autologous), intrauterine transfusion of fetuses, exchange
6888_Ch16_355-372 29/10/18 5:44 PM Page 365

Chapter 16 Transfusion Therapy 365

transfusion of neonates, donations from blood relatives, and For a patient who is likely to require blood transfusion, the
HLA-matched platelets.1 number of crossmatched units should be no more than twice
Immunocompetent recipients have experienced TA-GVHD those usually required for that surgical procedure. Thus, the
after receiving nonirradiated directed donations primarily crossmatch-to-transfusion (C/T) ratio will be between 2:1
from first-degree relatives. The related donor is homozygous and 3:1, which has been shown to be optimal practice.
for one of the patient’s (host’s) HLA haplotypes, so the patient Some transfusion services extend this practice to non-
is incapable of rejecting the donor’s (graft’s) T lymphocytes, surgical patients, crossmatching only when the RBCs are
which then can act against the HLA antigens encoded by the requested for issue to the patient. This practice increases
patient’s other haplotype. The donor lymphocytes then reject the inventory of uncrossmatched units that can be used for
the host. The level of immunosuppression a recipient must immediate needs.
have to develop TA-GVHD is unknown, although patients
with the severe immunosuppressive conditions listed previ- Autologous Transfusion
ously are most at risk. In addition, the dose of lymphocytes
needed for TA-GVHD to occur is unknown. For this reason, Autologous (self) transfusion is the donation of blood by
prevention depends on irradiation and not on reduction of the intended recipient (Fig. 16–1). The infusion of blood
lymphocytes by filtration. from another donor is called allogeneic transfusion. The
patient’s own blood reduces the possibility of transfusion
Transfusion Therapy in Special reaction or transmission of infectious disease.
Conditions One type of autologous transfusion is the predeposit of
blood by the patient. Collected by regular blood donation
Some patients require policies and procedures that address procedure, the blood can be stored as liquid or, for longer
particular clinical situations. Patients undergoing elective storage, frozen. Patients may donate several units of blood
surgery, autologous collection and transfusion, and emer- over a period of weeks, taking iron supplements to stimulate
gency and massive transfusion are described here. Trans- erythropoiesis. Predeposit autologous donation is usually re-
fusion of neonatal, pediatric, and oncology patients and served for patients anticipating a need for transfusion, such
those with congenital coagulation deficiencies are also as for a scheduled surgery. Predeposit autologous transfusion
discussed. is expensive because about half of the donated units are not
used. In addition, patients present for surgery with lower
Surgical Blood Order Schedule, Type, and Screen hematocrits, which increases transfusion of allogeneic units
in addition to autologous units. However, patients with mul-
Most surgical procedures do not require blood transfusion. tiple RBC antibodies or antibodies to high-incidence anti-
Crossmatching for procedures with a low likelihood of trans- gens may store frozen units for use by themselves or others.
fusion increases the number of crossmatches performed, Another type of autologous transfusion, intraoperative
increases the amount of blood inventory in reserve and hemodilution, is the collection of 1 or 2 units of blood from
unavailable for transfusion if electronic crossmatch is not the patient just before a surgical procedure, replacing the
available, and contributes to the aging and possible outdating removed blood volume with crystalloid or colloid solution.
of the blood components. It is prudent to perform a type and Then, at the end of surgery, the blood units are infused into
screen prior to surgery. If the antibody screen is positive, the patient. Care must be taken to label and store the blood
antibody identification must be completed and compatible units properly and to identify the blood units with the
units found. However, if the antibody screen is negative, patient before infusion.
ABO- and Rh-type–specific blood may be released after Meticulous attention to hemostasis and salvage of shed
an immediate spin or electronic crossmatch in those rare blood has allowed surgical procedures that once required
instances when transfusion is required. Box 16–2 shows an many units of blood to be performed without the need for
example of a surgical blood order schedule. allogeneic blood (see Fig. 16–1). Several types of equipment
are available for collecting, washing, and filtering shed blood
before reinfusion. Washing of intraoperative or postoperative
BOX 16–2 salvaged blood is recommended to remove the cellular
debris, fat, and other contaminants. Heparin or citrate solu-
Surgical Blood Order Schedule tions may be used for anticoagulation of the shed blood.
Purpose: Reduce unnecessary crossmatching
Examples: Emergency Transfusion
• Type and screen
• Exploratory laparotomy Patients who are rapidly or uncontrollably bleeding may
• Cholecystectomy require immediate transfusion. Group O RBCs are selected
• Two units crossmatched for patients for whom transfusion cannot wait until their
• Cardiac bypass surgery ABO and Rh type can be determined. Group O-negative
• Total hip revision RBC units should be used if the patient is a female of child-
bearing potential. An Rh-negative male patient or an older
6888_Ch16_355-372 29/10/18 5:44 PM Page 366

366 PART III Transfusion Practices

Predeposit for Scheduled Surgery Interoperative Salvage

Interoperative Hemodilution Postoperative Salvage

Figure 16–1. Types of autologous transfusions. (Reprinted with per-


mission from Kennedy, MS (ed): Blood Transfusion Therapy: An Audio-
visual Program. AABB, Bethesda, MD, 1985.)

postmenopausal female patient can be switched from Table 16–4 Example of a Massive
Rh-negative to Rh-positive RBCs if few O-negative units
Transfusion Protocol (MTP)
are available or if massive transfusion is required. Delaying
blood transfusion in emergency situations may be more Initial Transfusion Strategy
dangerous than the small risk of transfusing incompatible Draw type and crossmatch 2 units group O RBCs
blood before the antibody screen and crossmatch are uncrossmatched
completed.
After issuing O blood or type-specific blood, the antibody Continuing
screen can be completed, and decisions can then be made Prepare massive transfusion 4 to 6 type-specific or cross-
for the selection of additional units of blood. If the patient protocol pack by transfusion matched RBCs
has been typed and screened for a surgical procedure and his service
4 plasma
or her antibody screen is negative, ABO- and Rh-type-specific
1 platelet pool or
blood can be given after an immediate spin crossmatch.
plateletpheresis
Transfusions should be reserved for those patients losing
more than 20% of their blood volume. The condition of most Monitor CBC, platelet count, Add or subtract components
patients allows determination of ABO and Rh type and se- PT/INR, PTT, fibrinogen based on lab values
lection of ABO- and Rh-type-specific blood for transfusion. Provide additional MTP packs Continue transfusion until
unneeded
Massive Transfusion

Massive transfusion is defined as the replacement of one or


more blood volumes within 24 hours, or about 10 units of A patient in critical condition and a limited supply of
blood in an adult. The strategy for treating massive hemor- type-specific blood may require a change in ABO or Rh types.
rhage has changed in recent years, as experience in the An Rh-negative male or postmenopausal female patient may
military has promoted preventive treatment to avoid coagu- be switched from Rh-negative to Rh-positive blood to avoid
lopathy. Most medical centers with high-level trauma ser- depleting the inventory of Rh-negative blood. However, an
vices have adopted a massive transfusion protocol, one of Rh-negative potentially childbearing woman should receive
which is outlined in Table 16–4. Analysis of the patient’s Rh-negative RBC products for as long as possible.
clinical status and laboratory tests is essential for guiding
appropriate transfusion therapy. The massive transfusion Neonatal Transfusion
pack can be adjusted for low hemoglobin or platelets or pro-
longed coagulation tests. For example, platelets are required Premature infants frequently require transfusion of small
if the platelet count is less than 50,000/µL, and plasma is amounts of RBCs to replace blood drawn for laboratory tests
needed if the prothrombin time (PT) ratio is greater than 1.5, and to treat the anemia of prematurity (Box 16–3). A dose
or the international normalized ratio (INR) is greater than 1.5, of 10 mL/kg will increase the hemoglobin by approximately
or the activated partial thromboplastin time (aPTT) exceeds 3 g/dL. Various methods are available for preparing small
60 seconds. Fibrinogen levels should also be monitored be- aliquots for transfusion. Small aliquots of donor blood can
cause replacement by cryoprecipitate may be indicated when be transferred from the collection bag to a satellite bag or
the fibrinogen level is less than 100 mg/dL. transfer bag, or blood can be withdrawn from the collection
6888_Ch16_355-372 29/10/18 5:44 PM Page 367

Chapter 16 Transfusion Therapy 367

In addition, some malignancies such as chronic lympho-


BOX 16–3 cytic leukemia and lymphoma are frequently complicated by
Neonatal RBC Transfusions autoimmune hemolytic anemia, increased destruction of
RBCs, and pretransfusion testing problems.
Aliquoted Units
Oncology patients with hematologic malignancies, such
• Less than 7 days old, unless infused slowly as Hodgkin’s disease and lymphoma, are at increased risk of
• O-negative or compatible with mother and infant TA-GVHD because of the chemotherapy drugs used for treat-
• CMV-negative or leukocyte-reduced ment. Therefore, these patients should receive irradiated
• Hemoglobin S–negative for hypoxic newborns cellular components.
Dose
• 10 mL/kg over 2 to 3 hours Coagulation Factor Deficiencies

Factor VIII is normally complexed with another plasma


protein, vWF. Both proteins are necessary for normal hemo-
bag or transfer bag using an injection site coupler and needle stasis. Patients with hemophilia A, or classic hemophilia,
and syringe or a sterile docking device and syringe. Red blood have factor VIII deficiency. Hemophilia A patients have
cells in CPD, CPDA-1, or additive solution can be used safely. factor VIII levels less than 50%, although clinical disease is
The aliquot must be labeled clearly with the name and generally not apparent unless the factor VIII level is less
identifying numbers of the patient and donor. The blood than 10% (normal is 50% to 150%). Individuals with a level
must be fully tested, the same as for adult transfusion. Blood less than 1% have severe and spontaneous bleeding, typically
units less than 7 days old are preferred to reduce the risk of into muscles and joints. The vWF level is usually normal in
hyperkalemia and to maximize the 2,3-diphosphoglycerate patients with hemophilia A.
levels, although in some institutions CPDA-1 RBCs 14 to Von Willebrand’s disease is defined by a deficiency of
21 days old are used. vWF. Type I von Willebrand’s disease is characterized by a
For very-low-birth-weight infants, the blood should be reduced amount of all sizes of vWF multimers and is milder
selected to be CMV-seronegative or leukocyte-reduced to than type III, in which little or no vWF is produced. Type
prevent CMV infection, which can be serious in premature IIA is distinguished by a deficiency of high-molecular-weight
infants. Indeed, pregnant women should be given CMV- multimers, whereas type IIB is identified by abnormal high-
negative or leukocyte-reduced cellular components if they molecular-weight multimers that have an increased avidity
test negative for CMV. for binding to platelets. Type I patients have the ability
Irradiation of the blood is recommended to prevent to make the full spectrum of vWF multimers but do not
possible TA-GVHD when blood is used for intrauterine produce them in normal amounts. DDAVP, a synthetic
transfusion, for an exchange transfusion (see Chapter 20, vasopressin analog, can stimulate release of the vWF from
“Hemolytic Disease of the Fetus and Newborn [HDFN]”), the vascular endothelium in type I patients. DDAVP, how-
or for transfusion of a premature (less than 1,200 g) ever, is contraindicated in type IIB von Willebrand’s disease.
neonate. Transfusions in a full-term newborn infant do not Many factor VIII products are assayed for vWF and can
require routine irradiation. be used for type III von Willebrand’s disease or in type I or
Infants who are hypoxic or acidotic should receive blood IIA disease when DDAVP treatment has failed.
tested and negative for hemoglobin S. For more detail on Hemophilia B is the congenital deficiency of factor IX.
intrauterine and exchange transfusions, see Chapter 20. Factor IX is activated by factors XIa and VIIa. The activated
factor IX (IXa), along with factor VIII, ionized calcium, and
Transfusion in Oncology phospholipid, activates factor X to Xa. Factor IX deficiency
should be treated with recombinant factor IX or prothrombin
The bone marrow of oncology patients may be suppressed complex concentrates. Factor IX concentrates are made
because of chemotherapy, radiation therapy, or infiltration virus-safe by various sterilization techniques.10
and replacement of the bone marrow with malignant cells. All coagulation factors except vWF are made in the liver.
Repeated RBC and platelet transfusions may lead to the With severe liver failure, multiple coagulation factor defi-
need for rare RBC units or HLA-matched plateletpheresis ciencies occur. In addition, some of the coagulation factors
components because of incompatibility problems. Platelet produced may be abnormal. The liver also produces many
transfusion requirements may also necessitate a change from of the thrombolytic proteins, leading to imbalance between
Rh-negative to Rh-positive products. RhIG may be given to the coagulation process and the control mechanism. Plasma,
a woman with childbearing potential to protect against im- having normal amounts of all these proteins, can be used to
munization. There is less than 0.5 mL of Rh-positive RBCs treat these patients.
present in each Rh-positive plateletpheresis component. A Vitamin K aids in the carboxylation of factors II, VII, IX,
pool of platelets may contain as much as 4 mL of RBCs. and X. With the absence of vitamin K or the use of drugs
One 300-µg dose of RhIg can neutralize the effects of up to that interfere with vitamin K metabolism, such as warfarin,
15 mL of Rh-positive cells. Thus, one dose could be used for the inactive coagulation proteins cannot be carboxylated to
30 plateletphereses or 4 platelet pools. active forms. Vitamin K administration rather than plasma
6888_Ch16_355-372 29/10/18 5:44 PM Page 368

368 PART III Transfusion Practices

transfusion is recommended to correct vitamin K deficiency


or warfarin overdose. Because several hours are required for
vitamin K effectiveness, depending on route of administra-
tion, signs of hemorrhage or impending surgery may require
transfusion of plasma.
Transfuse
Disseminated intravascular coagulation (DIC) is the < 4 hours
uncontrolled activation and consumption of coagulation
proteins, causing small thrombi within the vascular system
throughout the body. Treatment is aimed at correcting the Use a filter
cause of the DIC: sepsis, disseminated malignancy, certain
acute leukemias, obstetric complications, or shock. In some
cases, transfusion of plasma, platelets, and/or cryoprecipitate
may be required. Monitoring of the PT, aPTT, platelet count,
fibrinogen, and hemoglobin and hematocrit levels helps
Identify patient
direct the choice of the next component to be used. and blood unit Have IV in place
Platelet functional disorders may be caused by drugs, ure-
mia, or congenital abnormalities. Platelet transfusions in
these patients should be reserved for treating hemorrhage or
the impending need for adequate hemostasis (such as a
surgical procedure) to decrease development of platelet re-
fractoriness. Drugs that interfere with platelet function, such Monitor patient
periodically
as clopidogrel (Plavix), are commonly used in cardiovascular
disease and are irreversible for the life of the platelets. There-
fore, discontinuing the drug for 5 to 7 days before surgery
will minimize hemorrhage and lessen the need for massive Figure 16–2. Blood administration and patient safety. (Reprinted with permis-
transfusion. In uremia, DDAVP may be beneficial as well sion from Kennedy, MS (ed): Blood Transfusion Therapy: An Audiovisual Program.
as dialysis or RBC transfusions. DDAVP releases fresh, func- AABB, Bethesda, MD, 1985.)
tional vWF from endothelial cells. Dialysis removes by-
products of protein metabolism that degrade vWF and coat
to historical results. Another clerical check is done as blood
platelets, making both nonfunctional. RBC transfusion in-
is issued from the blood bank. The final clerical check is
creases viscosity.
performed at the patient bedside as the nurse compares the
patient armband with the blood bank tag attached to the
General Blood Transfusion Practices
component to be transfused. In combined fiscal years 2011
Although blood administration and the hospital transfusion through 2015, transfusion-related acute lung injury (TRALI)
committee are not the responsibility of the transfusion ser- caused the highest number of reported fatalities (38%),
vice, nurses, physicians, and administrators look to the followed by transfusion-associated circulatory overload
transfusion specialists to guide policies and practices. (TACO) (24%) and hemolytic transfusion reactions (HTR)
due to non-ABO (14%) and ABO (7.5%) incompatibilities.
Blood Administration Microbial contamination (10%), anaphylactic reactions
(5%), and hypotensive reactions (1%) each accounted
Blood must be administered carefully for patient safety for a relatively smaller number of reported fatalities.16
(Fig. 16–2). The positive identification of the patient, patient’s Although TRALI continues to be one of the leading causes
blood specimen, and blood unit for transfusion is essential. of transfusion-associated fatalities reported to the FDA, the
Careful identification procedures prevent transfusion-related voluntary measures taken by the transfusion community to
deaths of ABO incompatibility. The identification process reduce the risk of TRALI have coincided with a reduction
begins with positive identification of the patient—that is, in the number of TRALI deaths.17–19 Table 16-5 lists the
asking patients to state or spell their name while you read Fiscal Year (FY) 2015 transfusion fatalities reported to the
their armband. The patient identification label is compared FDA by category. (See Chapter 17, “Adverse Effects of
at the bedside to the patient’s hospital armband. This prevents Blood Transfusion.”)
a specimen tube labeled with one patient’s name being used The patient with difficult veins should have the intra-
for the collection of a specimen from another patient. The venous infusion device in place before the blood is issued
labels are applied to the specimen tubes before leaving the from the transfusion service. All blood components must
bedside to avoid labeling the wrong tube. Electronic systems be filtered because clots and cellular debris develop during
for patient and label identification are available and should storage. The AABB Standards states “Blood and blood com-
be adopted to reduce clerical errors. ponents shall be transfused through a sterile, pyrogen-free
Positive identification is carried out in the laboratory. Cler- transfusion set that has a filter designed to retain particles
ical check is performed as results are generated and compared potentially harmful to the recipient.”5 Blood components are
6888_Ch16_355-372 29/10/18 5:44 PM Page 369

Chapter 16 Transfusion Therapy 369

Table 16–5 FY15 Transfusion Fatalities Rapid transfusion, including exchange transfusion, re-
quires blood warming because the cold blood can cause
Reported to the FDA by
hypothermia in the patient, which increases the possibility
Category
of cardiac arrhythmia and hemorrhage. A patient with
Category FY 15 No. FY 15 % paroxysmal cold hemoglobinuria or with potent cold
agglutinins may also require blood warming. The blood
TRALI 12 32
warmer should have automatic temperature control with
TACO 11 30 an alarm that will sound if the blood is warmed over 42°C.
Blood units must not be warmed by immersion in a water
HTR (non-ABO) 4 11
bath or by a domestic microwave oven because uneven
Contamination (bacterial) 5 14 heating can cause damage to blood cells and denaturation
of blood proteins.
HTR (ABO) 2 5
Only isotonic (0.9%) saline should be used to dilute
Hypotensive reaction 1 3 blood components because other intravenous solutions may
damage the RBCs and cause hemolysis (dextrose solutions
Anaphylaxis 2 5
such as D5W) or initiate coagulation in the infusion set
Note: FY15 denotes an imputability of Definite/Certain, Probable/Likely, or Possible. (calcium-containing solutions such as lactated Ringer solu-
tion). In addition, some drugs may cause hemolysis if injected
through the blood infusion set.
infused slowly for the first 10 to 15 minutes while the patient After transfusion, the transfusion set may be flushed with
is observed closely for signs of a transfusion reaction. The isotonic saline. The empty blood bag can be discarded or re-
blood components should then be infused as quickly as tol- turned to the transfusion service, according to hospital policy.
erated or, at most, within 4 hours. The patient’s vital signs A copy of the blood bag tag is placed in the patient’s chart.
(pulse, respiration, blood pressure, and temperature) should
be monitored periodically during the transfusion to detect Hospital Transfusion Committee
signs of transfusion reaction (see Chapter 17, “Adverse
Effects of Blood Transfusion”). Signs and symptoms are fever The Joint Commission requires all blood transfusions to be
with back pain (acute hemolytic transfusion reaction), reviewed for appropriate use. A hospital transfusion com-
anaphylaxis, hives or pruritus (urticarial reaction), conges- mittee may serve as the peer review group for transfusions.
tive heart failure (volume overload), and fever alone (febrile Blood usage review may also be performed prospectively if
nonhemolytic transfusion reaction). A delayed hemolytic criteria are approved by the transfusion committee and the
transfusion reaction (jaundice, decreasing hematocrit level) medical staff. The blood bank director may interact and
may be diagnosed 5 to 10 days after transfusion and thus is correspond directly with the chair of individual hospital
not considered an immediate reaction. departments concerning medical staff blood component
In standard blood administration sets, the 150- to 260-µm usage patterns. Appropriate criteria for blood transfusion
filter removes gross clots and cellular debris.2 A blood have been published (Table 16–6), serving as a guide for
administration filter must be used for transfusion of all blood conducting audits.3,7,8,20,21 The results of the audits can
components. The specific product manufacturer’s package be used by the transfusion committee to recommend
insert should be reviewed for instructions pertaining to use changes in practice by the hospital staff to improve patient
of transfusion devices (e.g., filters, blood administration sets, care. The transfusion committee also reviews transfusion
and blood warmers).2 reactions to ensure that adverse reactions are unavoidable.

Table 16–6 Criteria for Transfusion Audit


All Blood Components

Review of patients with transfusion reactions of hemolysis, severe allergic signs or anaphylaxis, circulatory overload, transfusion-related acute lung
injury, or infection

RBCs1,2,6

Indications Decreased oxygen-carrying capacity or acute loss of more than 20% blood volume
or hemoglobin less than 7 g/dL or hematocrit less than 21%

Exceptions Patients with hemoglobin less than 8 g/dL and acute coronary syndrome

Outcome H/H less than 24 hours after transfusion


Surgical patients: postoperative hemoglobin less than preoperative hemoglobin
Continued
6888_Ch16_355-372 29/10/18 5:44 PM Page 370

370 PART III Transfusion Practices

Table 16–6 Criteria for Transfusion Audit—cont’d


Platelets7

Indications Platelet count less than 5,000 to 10,000/µL without hemorrhage


or hemorrhage and platelet count less than 50,000/µL
or operative procedure and platelet count less than 50,000/µL
or antiplatelet drugs

Outcome Platelet count immediately before and 10 minutes to 1 hour post-transfusion except with antiplatelet drugs (e.g., Plavix)

Plasma

Indications Patients with bleeding or invasive procedure


PTT greater than 60 seconds
or INR greater than 1.5

Outcome PT or PTT, less than 4 hours after transfusion

Cryoprecipitate

Indications Fibrinogen deficiency or factor XIII deficiency

Outcome Fibrinogen or factor XIII determination after transfusion

H/H = hemoglobin/hematocrit; PT = prothrombin time; PTT = partial thromboplastin time; INR = international normalized ratio

In addition, the transfusion committee ensures that appro- 4. If the patient is found to have von Willebrand’s dis-
priate procedures (such as for blood administration) are in ease, which blood product might be necessary?
place and are followed by hospital personnel.
The transfusion committee is most effective if the vari- Case 16-2
ous groups who order and administer blood, such as
surgeons, anesthesiologists, oncologists, and nurses are A 45-year-old woman complains of tiredness and weak-
represented on the committee. The transfusion committee ness. She appears pale. Laboratory results are as follows:
must have a mechanism for reporting activities and hemoglobin 6.2 g/dL, hematocrit 20%, MCV 75 fL,
recommendations to the medical staff and hospital admin- MCHC 28%. On further questioning, she reports exces-
istration. Optimally, the transfusion committee ensures sive menstrual bleeding, sometimes lasting for several
that the most appropriate, efficient, and safe use of the weeks.
blood supply is achieved. All adverse events related to 1. Does this patient need a transfusion? Justify your
transfusion must be reported to the transfusion service answer.
according to its local protocol. (Refer to Chapter 17 2. If the intern decides to give 1 unit of RBCs, what
“Adverse Effects of Blood Transfusion” and Chapter 26 would be the resulting hemoglobin and hematocrit
“Patient Blood Management.”) levels?

Case 16-3
CASE STUDIES
A 22-year-old woman presents with easy bruising and
Case 16-1 fatigue. A complete blood count reveals hemoglobin 9 g/dL,
hematocrit 27%, WBC 15,000/µL, and platelet count
A 55-year-old man is contemplating surgery for severe 5,000/µL. The hematologist plans to perform a bone mar-
arthritis in the right hip. row biopsy and aspiration.
1. What should be known to determine how many units 1. What blood component(s) is (are) indicated? Why?
of RBCs should be crossmatched? 2. Describe how the dose is calculated. What laboratory
2. How can it be determined if the patient can donate result is desired?
blood (autologous predeposit) for use during surgery? 3. The patient receives chemotherapy, and 2 weeks later
3. The patient has a history of bleeding after a tonsillec- the hemoglobin is 6.8 g/dL and the hematocrit 20%. The
tomy at age 7 years. What tests should be done to fur- patient complains of shortness of breath when hurrying
ther study this potential problem? to the bus stop. The physician decides to order RBC
6888_Ch16_355-372 29/10/18 5:45 PM Page 371

Chapter 16 Transfusion Therapy 371

transfusion. What dose of RBCs is indicated? How is this 2. For FFP and platelets, which blood type(s) should be
determined? selected?

Case 16-4 His type and screen is B positive with positive antibody
screen. The ED nurse calls and informs the blood bank
A 45-year-old man is admitted to the emergency depart- that the massive transfusion protocol has been started.
ment vomiting blood. His hemoglobin is 8 g/dL, platelet
3. Which components should be prepared?
count 80,000, PT/INR 18/2, fibrinogen 125 mg/dL. The
4. Which blood group and Rh type should be selected?
physician orders 4 units of RBCs, 4 units of plasma, and
5. What should be done about the positive antibody
1 pool of platelets stat.
screen?
1. Which blood group(s) should be selected for the RBC
transfusions?

SUMMARY CHART
 Transfusion therapy is used primarily to treat two con-  Plasma contains all coagulation factors and is indicated
ditions: inadequate oxygen-carrying capacity because for patients with multiple coagulation deficiencies
of anemia or blood loss and maintenance of hemostasis that occur in liver failure, DIC, vitamin K deficiency,
when the patient has insufficient coagulation proteins warfarin overdose, and massive transfusion.
or platelets.  Cryoprecipitate contains at least 80 units of factor VIII
 A unit of whole blood or RBCs in an adult should and 150 mg of fibrinogen, as well as vWF, and factor XIII.
increase the hematocrit level 3% or hemoglobin level  Factor IX is used in the treatment of persons with
1 g/dL. hemophilia B.
 RBCs are indicated for increasing the RBC mass in pa-  Immunoglobulin (IG) is used in the treatment of con-
tients who require increased oxygen-carrying capacity. genital hypogammaglobulinemia, to provide passive
 Platelet transfusions are indicated for patients who are immunity for certain infections such as hepatitis A and
bleeding because of thrombocytopenia. In addition, measles, and in certain autoimmune conditions such
platelets are indicated prophylactically for patients as immune thrombocytopenic purpura.
who have platelet counts under 5,000 to 10,000/µL.  Massive transfusion is defined as the replacement of
 Each dose of platelets should increase the platelet one or more blood volume(s) within 24 hours, or
count 20,000 to 40,000/µL in a 70-kg patient. about 10 units of blood in an adult.
 A plateletpheresis product is collected from one donor  Emergency transfusion warrants group O RBCs when
and must contain a minimum of 3 × 1011 platelets. patient type is not yet known.

3. Of the following, which blood type is selected when a


Review Questions
patient cannot wait for ABO-matched RBCs?
1. Leukocyte-reduced filters can do all of the following a. A
except: b. B
c. O
a. Reduce the risk of CMV infection
d. AB
b. Prevent or reduce the risk of HLA alloimmunization
c. Prevent febrile, nonhemolytic transfusion reactions 4. Which patient does not need an irradiated component?
d. Prevent TA-GVHD a. Bone marrow transplant recipient
2. Albumin should not be given for: b. Neonate weighing less than 1,200 g
c. Adult receiving an RBC transfusion
a. Burns
d. Adult receiving an RBC transfusion from a blood
b. Shock
relative
c. Nutrition
d. Plasmapheresis
6888_Ch16_355-372 29/10/18 5:45 PM Page 372

372 PART III Transfusion Practices

5. RBC transfusions should be given: clinical practice guidelines: Society of Thoracic Surgeons Blood
Conservation Guideline Task Force. Ann Thorac Surg. 2011;
a. Within 4 hours 91(3):944-82.
b. With lactated Ringer solution 4. Weiskopf R, Feiner J, Hopf H, Viele M, Watson J, Kramer J,
c. With dextrose and water et al. Oxygen reverses deficits of cognitive function and mem-
d. With cryoprecipitate ory and increased heart rate induced by acute severe isovolemic
anemia. Anesthesiology. 2002;96(4):871-7.
6. Which type of transplantation requires all cellular blood 5. AABB. Standards for blood banks and transfusion services.
components to be irradiated? 31st ed. Bethesda, (MD): AABB; 2018.
6. Friday J, editor. A compendium of transfusion practice guide-
a. Bone marrow
lines. 3rd ed. Washington (DC): American National Red Cross;
b. Heart 2017.
c. Liver 7. Kaufman R, Djulbegovic B, Gernsheimer T, Kleinman S,
d. Kidney Tinmouth A, Capocelli K et al. Platelet transfusion: a clinical
practice guideline from the AABB. Ann Intern Med. 2015;
7. Characteristics of deglycerolized RBCs include the 162(3):205-13.
following except: 8. Roback JD, Caldwell S, Carson J, Davenport R, Drew M, Eder A,
et al. Evidence-based practice guidelines for plasma transfu-
a. Inexpensive
sion. Transfusion. 2010;50:1227-39.
b. 24-hour expiration date after thawing 9. Nascimento B, Goodnough L, Levy J . Cryoprecipitate therapy.
c. Used for rare antigen-type donor blood Br J Anaesth. 2014;113(6):922-34.
d. Used for IgA-deficient recipient with history of severe 10. Mannucci P. Hemophilia: treatment options in the twenty-first
reaction century. J Thromb Haemost. 2003;1(7):1349-55.
11. Knezevic-Maramica I, Kruskall M. Intravenous immune
8. Select the appropriate product for a bone marrow trans- globulins: an update for clinicians. Transfusion. 2003;43(10):
plant patient with anemia: 1460-80.
12. Roberts H. Recombinant factor VIIa (Novoseven) and the safety
a. RBCs of treatment. Semin Hematol. 2001;38(4 Suppl 12):48-50.
b. Irradiated RBCs 13. Mayer S, Brun N, Begtrup K, Broderick J, Davis S, Diringer M,
c. Leukoreduced RBCs et al. Efficacy and safety of recombinant activated factor VII
d. Washed RBCs for acute intracerebral hemorrhage. N Engl J Med. 2008;
358(20):2127-37.
9. Which blood product should be selected for vitamin K 14. Sharma R, Marwaha N. Leukoreduced blood components:
deficiency? advantages and strategies for its implementation in developing
countries. Asian J Transfus Sci. 2010; 4(1):3-8.
a. Cryoprecipitate 15. Laupacis A, Brown J, Costello B, Delage G, Freedman J, Hume H,
b. Factor VIII et al. Prevention of posttransfusion CMV in the era of
c. Factor IX universal WBC reduction: a consensus statement. Transfusion.
d. Plasma 2001;41(4):560-9.
16. Cid J. Prevention of transfusion-associated graft-versus-host
10. Which fluid should be used to dilute RBCs? disease with pathogen-reduced platelets with amotosalen and
ultraviolet A light: a review. Vox Sang. 2017;18. doi: 10.1111/
a. 0.9% saline vox.12558. PMID: 28833219. [Epub ahead of print].
b. 5% dextrose and water 17. Food and Drug Administration, CBER [Internet]. Rockville
c. Immune globulin (MD): FDA [cited 2016 November 2]. Fatalities reported to
d. Lactated Ringer solution FDA following blood collection and transfusion annual sum-
mary for FY2015. Available from: http://www.fda.gov/Biologics
BloodVaccines/SafetyAvailability/ReportaProblem/Transfusion
References DonationFatalities/ UCM518148.pdf
1. King K. editor. Blood transfusion therapy: a physician’s hand- 18. Peters AL, Van Stein D, Vlaar AP. Antibody-mediated transfusion-
book. 11th ed. Bethesda (MD): AABB; 2014. related acute lung injury; from discovery to prevention.
2. Food and Drug Administration [Internet]. Rockville (MD): FDA Br J Haematol. 2015;170(5):597-614. doi: 10.1111/bjh.13459.
[cited 2017 December]. Guidance for industry: circular of 19. Popovsky M. Transfusion-related acute lung injury: three
information for the use of human blood and blood products. decades of progress but miles to go before we sleep. Transfu-
AABB, America’s Blood Centers, American Red Cross (ARC), sion. 2015;55:930-4.
and the Armed Services Blood Program (ASBP). Available from: 20. Carson J, Grossman B, Kleinman S, Tinmouth A, Marques M,
https://www.fda.gov/downloads/BiologicsBloodVaccines/Guidance Fung M, et al. Red blood cell transfusion: a clinical practice
ComplianceRegulatoryInformation/Guidances/Blood/UCM364593 guideline from the AABB. Ann Intern Med. 2012:157:49-58.
.pdf 21. Carson J, Guyatt G, Heddle N, Grossman B, Cohn C, Fung MK,
3. Ferraris VA, Brown J, Despotis G, Hammon J, Reece T, Saha S, et al. Clinical practice guidelines from the AABB: red blood cell
et.al. 2011 update to the Society of Thoracic Surgeons and the transfusion thresholds and storage. JAMA. 2016;316(19):
Society of Cardiovascular Anesthesiologists blood conservation 2025-35.
6888_Ch17_373-395 29/10/18 5:45 PM Page 373

Chapter 17
Adverse Effects of Blood Transfusion
Mark D. Pool, MD

Introduction Transfusion-Related Acute Lung Injury Adverse Metabolic Effects of


Risks of Transfusion (TRALI) Transfusion
Incidence of Adverse Reactions to Transfusion-Associated Circulatory Controversy: Immunomodulatory
Transfusion Overload (TACO) Effects of Transfusion
Transfusion-Associated Fatalities Transfusion-Associated Dyspnea (TAD) Infectious Transfusion Reactions
Recognition and Evaluation of Hypotensive Transfusion Reaction Transfusion-Transmitted Bacterial
Transfusion Reactions Febrile Nonhemolytic Transfusion Infections (TTBI)
Regulatory and Accreditation Reaction (FNHTR) Case Studies
Requirements Allergic Transfusion Reactions (ATRs) Case Study 17-1
Noninfectious Transfusion Reactions Adverse Reactions to Infusion of Case Study 17-2
Alloimmunization to RBC Plasma-Derived Products Case Study 17-3
Antigens Transfusion-Associated Graft- Summary Chart
Acute Hemolytic Transfusion Versus-Host Disease Review Questions
Reaction (AHTR) Post-Transfusion Purpura References
Delayed Hemolytic and Serologic Refractoriness to Platelet Transfusion
Transfusion Reaction (DHTR) and Alloimmunization

OBJECTIVES
1. Compare the relative risks of adverse events due to transfusion.
2. Identify the major causes of transfusion-associated fatalities.
3. Outline the steps involved in the recognition and evaluation of a transfusion reaction.
4. List the main organizations responsible for regulation and accreditation of blood banks and transfusion services and the
regulations related to transfusion reactions.
5. Define hemovigilance.
6. Classify noninfectious transfusion reactions and transfusion-transmitted bacterial infections according to definitions of the
U.S. National Healthcare Safety Network (NHSN).
7. Describe the various factors that influence the development of alloimmunization to RBC antigens.
8. Contrast between acute and delayed hemolytic transfusion reactions.
9. Explain the pathophysiological mechanisms involved in immune-mediated hemolysis and how the proinflammatory envi-
ronment is responsible for many of the signs and symptoms related to a hemolytic reaction.
10. Compare transfusion reactions in which respiratory problems are the main feature: transfusion-related acute lung injury,
transfusion-associated circulatory overload, and transfusion-associated dyspnea.
11. Discuss the factors that contribute to the pathophysiology of febrile nonhemolytic transfusion reactions.
12. Describe the range of clinical manifestations found in allergic transfusion reactions and the mechanisms implicated in the
pathophysiology of these reactions.
13. Identify patients most at risk for transfusion-associated graft-versus-host disease and explain how a definitive diagnosis is made.
14. Name the main adverse metabolic effects of transfusion and summarize the evidence for the “RBC storage lesion.”
15. Compare the clinical manifestations and microbiology between transfusion-transmitted bacterial infection due to contami-
nated RBC versus platelet products.

373
6888_Ch17_373-395 29/10/18 5:45 PM Page 374

374 PART III Transfusion Practices

Introduction Table 17–1 Incidence of Transfusion-


Related Adverse Reactions
Blood transfusion is not, and likely never will be, without
risk. Adverse events are an inevitable consequence of blood Adverse Event Incidence
transfusion in spite of the myriad processes, physical barri-
AHTR 2.0 per 100,000 transfused RBCs
ers, and software safeguards in place to ensure safe transfu-
sions. An adverse event is an unintended and deleterious DHTR 7.3 per 100,000 transfused RBCs
occurrence associated with blood component transfusion. It
DSTR 26.2 per 100,000 transfused RBCs
may occur before, during, or after a transfusion. Adverse
events include incidents and adverse reactions. An incident TRALI 1.2 per 100,000 all components transfused
is any error that could affect the quality or effectiveness of
TACO 9.1 per 100,000 all components transfused
a blood product or could have led to an adverse reaction
to a transfusion recipient. An adverse reaction is a harmful Hypotensive 6.3 per 100,000 all components transfused
effect observed in a transfusion recipient that is temporally
FNHTR 86.4 per 100,000 all components transfused
associated with a blood component transfusion.
This chapter focuses on noninfectious adverse reactions Allergic, nonsevere 104.1 per 100,000 all components transfused
and transfusion-associated bacterial infection from bacte-
Allergic, severe 7.5 per 100,000 all components transfused
rial contamination of blood products. Other transfusion-
transmitted infections are discussed in Chapter 14, while TTBI 0.56 per 100,000 all components transfused
adverse reactions associated with cellular therapy are
TA-GVHD 0.01 per 100,000 all components transfused
discussed in Chapter 19.
AHTR = acute hemolytic transfusion reaction; DHTR = delayed HTR; DSTR = delayed
Risks of Transfusion serologic transfusion reaction (DSTR); TRALI = transfusion-related acute lung injury;
TACO = transfusion-associated circulatory overload; FNHTR = febrile nonhemolytic
transfusion reaction
Incidence of Adverse Reactions to Transfusion From: Harvey AR, Basavaraju SV, Chung KW, Kuehnert MJ. Transfusion-related adverse
reactions reported to the National Healthcare Safety Network Hemovigilance
Blood transfusion is a very safe procedure in general. Indeed, Module, United States, 2010 to 2012. Transfusion. 2015;55:715.
it is safer today than at any point since blood transfusion has
been used therapeutically. Yet blood transfusion is not without
risk. It is useful to consider that blood transfusion is essen- infections (TTBIs) were the most common causes of deaths
tially a transplant of foreign cells into the recipient, and many associated with transfusion recipients. Table 17-2 shows
of the adverse events seen with transfusion are related to this transfusion fatalities associated with adverse events from FY-
fact. In addition, even noncellular plasma and plasma-derived 2011 to FY2015 that were reported to the FDA. Note that
products involve the introduction of foreign proteins into the TRALI and TACO are consistently the leading causes of
recipient that may cause a transfusion reaction. Finally, blood mortality associated with transfusion.2
transfusion also carries the risk of transfusion-transmitted
infection. Table 17–1 provides current estimates of noninfec- Recognition and Evaluation
tious risks of transfusion and transfusion-transmitted bacterial of Transfusion Reactions
infection.1
The recognition and evaluation of suspected transfusion
Transfusion-Associated Fatalities reactions involves two critical components.4 Figure 17–1
outlines the main tasks and responsibilities in investigation
In the United States, fatalities associated with transfusion or of a suspected transfusion reaction. First is the clinical recog-
blood donation are required to be reported to the U.S. Food nition by the person administering the transfusion (usually
and Drug Administration (FDA). The FDA annually issues a nurse or physician) that a suspected transfusion reaction
a summary of fatalities following transfusion and blood col- may be occurring or has occurred and an initial evaluation
lection through this mandatory reporting system.2 A team of by a physician as to the likelihood that the signs and symp-
medical officers from the FDA Center for Biologics Evalua- toms observed may be related to transfusion. Table 17–3 lists
tion and Research (CBER) investigates all reported cases. signs and symptoms that may be observed in transfusion
The 2015 report has incorporated the case definition and reactions. The first step once a transfusion reaction is sus-
likelihood of causation (imputability) criteria developed pected is to immediately stop the transfusion if the infusion
by the Centers for Disease Control and Prevention (CDC)/ is still in process. The next step is to follow a standard pro-
National Healthcare Safety Network (NHSN).3 During cedure to send appropriate specimens to the laboratory for
FY2015 (October 1, 2014, to September 30, 2015), there a transfusion reaction investigation.
were 42 fatality reports associated with transfusion recipients Second is the laboratory investigation of a transfusion
and 37 (80%) were determined to be definitely, probably, reaction. The main objective in this phase is to identify a pos-
or possibly attributed to transfusion. Transfusion-related sible hemolytic transfusion reaction. Thus, the following
acute lung injury (TRALI), transfusion-associated circulatory steps include checking for appropriate identification of
overload (TACO), and transmission-transmitted bacterial the component bag, label, paperwork, and the recipient’s
6888_Ch17_373-395 29/10/18 5:45 PM Page 375

Chapter 17 Adverse Effects of Blood Transfusion 375

Table 17–2 Transfusion-Associated Table 17–3 Signs and Symptoms of


Fatalities Reported to FDA, Transfusion Reactions
2011–2015 Fever (≥39°C or Hematuria Bronchospasm
Complication Total Number Total % ≥2°C rise) (blood in (wheezing)
urine)
HTR (ABO) 13 7.5%
Hypoxemia Hemoglobinuria Pulmonary edema
HTR (non-ABO) 24 14% (O2 sat. <90%) (hemoglobin
in urine)
TRALI 66 38%
Tachycardia Nausea or Chills/rigors
TACO 41 24% (rapid heart rate vomiting
>120/min or
Hypotension 2 1% >40/min rise)
Allergic-anaphylaxis 8 5% Tachypnea Urticaria (hives) Pain—abdominal,
(rapid breathing back, chest, infusion
TTBI 18 10%
>28/min) site, headache
TA-GVHD 1 0.5%
Hypertension Pruritis (itching) Oliguria or anuria
(rise in systolic (decreased or
TA-GVHD = transfusion-associated graft-versus-host disease BP >30 mm Hg) absent urine
U.S. Food and Drug Administration, Center for Biologics and Evaluation Research.
Fatalities reported to FDA following blood transfusion and collection annual
output)
summary for FY2015. http://www.fda.gov/downloads/BiologicsBloodVaccines/
SafetyAvailability/ReportaProblem/TransfusionDonationFatalities/UCM518148.pdf.
Hypotension Rash Disseminated
(drop in systolic intravascular
BP >30 mm Hg) coagulation (DIC)

TRANSFUSION SERVICE:
Laboratory Technologist / Physician
Responsibilities:
the blood bank supervisor or medical director to determine
if additional testing is required. Final assessment of the trans-
fusion reaction is the responsibility of the medical director
Laboratory Technologist: Transfusion Service or an appropriate designee.
• Perform primary testing on Physician:
postreaction sample • Evaluate initial transfusion
reaction workup Regulatory and Accreditation
• Report findings to the Requirements
transfusion service physician • Order additional testing as
needed
• Perform additional testing as Adverse reactions represent a significant deviation from cur-
per transfusion service • Report to patient physician rent good manufacturing practice (CGMP) and must be ad-
physician orders immediately if hemolysis, dressed according to FDA regulations and laboratory
bacterial contamination,
TRALI, or other serious accreditation requirements of AABB and the College of
adverse event related to American Pathologists (CAP) (see Chapter 27). Table 17–4
transfusion is suspected compares FDA and AABB requirements for investigation of
• Generate a final transfusion reactions. The AABB standards require that the
transfusion report, including evaluation of suspected hemolytic transfusion reactions
interpretation of the (HTRs) include the elements in Box 17–1.5 AABB standards
transfusion reaction and
recommendations for future require that blood banks and transfusion services include a
transfusions procedure for recognizing, evaluating, and reporting adverse
reactions as part of the overall blood product administration
• Notify blood center and
other outside agencies if process.5 AABB also requires these facilities to have processes
applicable for evaluating and reporting suspected hemolytic and non-
hemolytic transfusion reactions, which includes notification
Figure 17–1. Steps and responsibilities in the recognition and evaluation of a and interpretation by the medical director. Both FDA regu-
suspected transfusion reaction. IV = intravenous, TRALI = transfusion-related acute
lations and AABB standards require that a written report of
lung injury.
the investigation be made and that records of the investiga-
tion be maintained. Finally, FDA regulations require that
pretransfusion specimen; repeating ABO testing on the post- fatalities associated with transfusions be reported to the
transfusion sample; visual inspection of the pre- and post- CBER by the facility that performed compatibility testing.6
transfusion specimens for hemolysis; and performing a direct It is generally suspected in the field of transfusion medicine
antiglobulin test (DAT) on the post-transfusion specimen. that adverse reactions to transfusion are underrecognized and
Figure 17–2 shows the basic testing on the post-transfusion underreported. The concept of hemovigilance has been devel-
specimen. The results of these findings are then provided to oped in response as a way to improve the recognition and
6888_Ch17_373-395 29/10/18 5:45 PM Page 376

376 PART III Transfusion Practices

Basic Testing
Post-Transfusion Reaction Sample

Clerical Check Hemolysis Check Direct Antiglobulin Test ABO Testing


Check that the following A visual check for: • Detects the presence of • To verify patient ABO
information matches: • Free hemoglobin will red blood cells sensitized typing
• Postreaction sample manifest as a pink or in vivo
• Transfusion documentation reddish discoloration of
• Blood bag (when available) the plasma

No No Hemolysis No
Discrepancy Negative* Positive Discrepancy
Discrepancy Hemolysis present Discrepancy

Request *If suspect Notify TS Request


redraw HTR physician redraw

Notify TS Go to
Notify TS Physician Second Draw: physician secondary Discrepancy revealed
testing if
If another patient is involved Hemolysis present on Go to requested If another patient is involved
in discrepancy, take second draw secondary by TS in discrepancy, take
necessary step to prevent testing if physician necessary steps to prevent
another adverse event Notify TS physician requested another adverse event
by TS
Go to secondary testing if Go to secondary testing if physician Notify TS physician
requested by TS physician requested by TS physician
Go to secondary testing if
requested by TS physician

Figure 17–2. Basic immunohematology investigation for a suspected transfusion reaction. TS = transfusion service, HTR = hemolytic transfusion reaction.

Table 17–4 Regulatory and Accreditation Requirements Related to Transfusion Reactions


Regulation Agency
FDA AABB

Recognition, response, and action to take for transfusion reaction 4 4

Reporting transfusion reactions to the TS 4

Laboratory evaluation of transfusion reaction 4

Additional testing for investigation of transfusion reaction 4

Interpretation and reporting of transfusion reaction by TS physician 4

Participation of TS physician in resolution of mistransfusion 4 4

Mechanism to notify facility providing blood components implicated in transfusion reaction 4 4

Reporting a transfusion-associated fatality 4 4

Documentation of investigation of a transfusion reaction 4 4

Documentation of annual in-service of criteria for recognition of transfusion reactions 4

Evaluation of delayed transfusion reactions 4

Instructions for patients that post-transfusion will not be observed by medical personnel 4

TS = transfusion service
6888_Ch17_373-395 29/10/18 5:45 PM Page 377

Chapter 17 Adverse Effects of Blood Transfusion 377

alloimmunization is much higher, 30% or greater, in chron-


BOX 17–1 ically transfused patients with sickle cell disease, myelodys-
AABB Requirements for Laboratory Investigation plastic syndrome, thalassemia, or autoimmune hemolytic
of a Transfusion Reaction disease.10
Except for the D antigen, the immunological response to
1. Clerical check of the component bag, label, paperwork, and
pretransfusion patient specimen.
RBC protein antigens is not very robust. About 85% of
2. Repeat ABO testing on the post-transfusion sample.
healthy D-negative persons exposed to the D antigen will
3. Visual check of the pre- and post-transfusion specimens for
develop anti-D. However, exposure to other antigens causes
hemolysis. much lower sensitization: anti-K develops in about 10% of
4. Direct antiglobulin test (DAT) on the post-transfusion specimen. K-negative–exposed individuals, anti-E in about 7%, and
5. Quarantine additional components prepared from the same anti-c in in about 3%. Exposure to other minor blood group
donor collection. antigens appears to produce alloimmunization in less than
6. Report findings to transfusion service supervisor or medical director. 3% of antigen-negative persons.10 The risk of an individual
patient developing a non-ABO alloantibody depends upon
many factors, including the patient’s underlying diseases, the
cause of anemia, the cumulative number of transfusions, and
reporting of adverse events. Hemovigilance is “the collection the immunogenicity of the non-self RBC antigens to which
of information on the complications of transfusion, analysis the patient is exposed.11
of these data, and subsequent data-driven improvements in
transfusion practices.”4 The U.S. National Healthcare Safety
Network (NHSN) Hemovigilance Module is a voluntary Advanced Concepts
effort of the CDC to create diagnostic case criteria for adverse It has been long recognized that some individuals may be
reactions, standardize data collection, and collect data from more susceptible than others to developing RBC alloanti-
participating health-care facilities.3 The Serious Hazards of bodies. These persons, called responders, represent the
Transfusion (SHOT) program in the United Kingdom7 is majority of patients identified with alloantibodies. But
another national hemovigilance reporting system. The Inter- within patients who have been alloimmunized, studies
national Haemovigilance Network website has a list of show about a quarter of these have or develop more than
other national hemovigilance networks.8 Yet, a recent study one alloantibody. This subgroup has been called hyper-
to validate appropriate use of case definitions in the U.S. responders. There has been substantial research to identify
NHSN found that agreement on the diagnostic categories, the clinical and biological factors associated with respond-
severities, and probabilities of an association with transfu- ers. These may include genetics, the inflammatory state of
sion was found only approximately two-thirds of the time the patient, sex, and age.10,11 Antigen diversity between
and well below that in many cases. Moreover, there was donor and recipient populations may explain some alloim-
relatively poor application of the case definitions even after munization, particularly in patients with sickle cell disease.
training.9 Thus, this study suggests that the general applica- Individual difference in the HLA class II system with
bility and value of the data collected by this process remains respect to antigen presentation is another genetic variable
to be determined. that might explain responsiveness. The underlying sys-
temic inflammatory state of the recipient also influences
Noninfectious Transfusion Reactions the predisposition to alloimmunization. Patients with
underlying systemic inflammatory disorders have been
Alloimmunization to RBC Antigens shown to be more likely to develop alloimmunization.
Studies regarding the sex of the patient with regard to
Alloimmunization is the development of non-ABO anti- alloimmunization are conflicting, and there is no evidence
bodies following RBC transfusion, pregnancy, or transplan- that clearly identifies a predisposition toward or protection
tation. RBC alloimmunization can present substantial from alloimmunization for either sex. Neonates, young
challenges for blood banks and transfusion services in com- children, and the elderly appear to be poor responders.10
pleting compatibility testing in a timely manner or even The rate of alloimmunization in patients with sickle cell
obtaining antigen-negative blood. More importantly, it can disease (SCD) is much higher than the general population,
complicate patient care and increase the risk of acute and ranging from about 19% to almost 50% in different studies.
delayed hemolytic transfusion reactions. The incidence Patients with a higher cumulative number of transfusions
of alloimmunization varies markedly according to clinical have a higher rate of alloimmunization.11 Furthermore, a
setting studied and condition of the patient. Healthy blood high prevalence of alloimmunization is seen even with poli-
donors have an incidence of alloimmunization less than cies to provide blood with extended phenotype matching
0.3%, while alloimmunization in unselected hospitalized and from African American donors.12 The challenges of
patients who have had type and screen testing ranges from managing hemolytic transfusion reactions in SCD patients
about 1% to 3%. The post-transfusion alloimmunization are discussed further below.
rate of surgery patients is around 3%. The prevalence of
6888_Ch17_373-395 29/10/18 5:45 PM Page 378

378 PART III Transfusion Practices

Acute Hemolytic Transfusion Reaction (AHTR) the flanks, lower back, abdomen, or infusion site.13 Severe
and life-threatening features, although uncommon, are grave
The majority of effort expended in blood banks and transfu- because these are associated with a greatly increased risk for
sion services is directed at preventing hemolytic transfusion mortality. Hypotension occurs in about 10% of AHTRs;
reactions. An AHTR is the accelerated destruction of trans- shock occurs in a small subset of these patients. Acute
fused RBCs due to antibody-mediated incompatibility. kidney injury is usually detected biochemically by elevated
Alloantibodies in the recipient’s plasma bind to the corre- blood urea nitrogen (BUN) and creatinine but can be severe
sponding antigen on the transfused cells, which mediate enough to cause decreased or absent urine output. Although
hemolysis and removal from the circulation. HTRs are clas- free hemoglobin liberated in the plasma from hemolysis
sified as acute or delayed according to the temporal relation- is directly toxic to the kidney, shock and disseminated
ship to the transfusion. An acute HTR (AHTR) is defined as intravascular coagulation (DIC) are the main causes of
the combination of signs and symptoms associated with renal dysfunction after hemolysis. Early and aggressive fluid
hemolysis, biochemical evidence of hemolysis, and serologic resuscitation and blood pressure management are thus the
evidence of RBC incompatibility (Table 17–5) occurring dur- first steps in treating AHTRs and preventing hypotension,
ing or within 24 hours after transfusion.3 Most AHTRs occur shock, and acute kidney failure. DIC itself is another uncom-
within minutes after the start of transfusion or within a few mon but devastating complication. Note that it may be
hours after the transfusion is completed. The vast majority difficult to distinguish between DIC and other coagulation
of AHTRs is due to RBC transfusion, but they can occur with abnormalities that were preexisting conditions, a therapeutic
incompatible plasma-containing products. Note that hemol- consequence of treatment for or prevention of a clotting
ysis that is unrelated to transfusion can also occur in trans- tendency, or from the hemolytic reaction itself. Coagulation
fusion recipients by a variety of immune and nonimmune testing such as activated partial thromboplastin time
mechanisms (Table 17–6). In general, the severity of an (aPTT), prothrombin time (PT), thrombin time, D-dimer,
AHTR is related to the amount of incompatible blood trans- fibrinogen, and platelet count are essential for diagnosis and
fused. On the other hand, transfusion of an entire incompat- management.
ible unit may not be associated with any apparent deleterious AHTRs occur in several clinical situations that warrant
effect, while fatal reactions have been reported with transfu- further consideration.13 AHTRs may develop in patients who
sion of only a small amount of a unit of incompatible blood. are unable to report symptoms or in whom typical signs are
There is a wide range of clinical severity and observed absent. Infants and young children, patients who are uncon-
signs and symptoms in AHTRs. Fever is the most common scious or mentally impaired, and anesthetized patients will
symptom in AHTR and often occurs with chills or rigors. not be able to communicate symptoms. Patients in surgery
Pain is another frequent symptom and can be localized to or ICU settings are usually receiving fluids and/or drugs for

Table 17–5 Laboratory and Serologic Evidence of Hemolytic Transfusion Reaction


Laboratory Tests Confirming Hemolysis Serologic Evidence of Immune-Mediated HTR
• Decreased fibrinogen • Positive DAT for polyspecific and anti-IgG or anti-C3
• Decreased or absent haptoglobin • Positive elution test with identification of one or more alloantibodies
• Elevated bilirubin
• Elevated LDH
• Hemoglobinemia
• Hematuria or hemoglobinuria
• Spherocytes
• Inadequate rise of post-transfusion hemoglobin level or
rapid fall of hemoglobin after transfusion

LDH = lactic dehydrogenase

Table 17–6 Immune and Nonimmune Mechanisms of Hemolysis


Immune Hemolysis Nonimmune Hemolysis
• Alloimmune acute and delayed HTRs • Osmotic: incompatible fluids, improper deglycerolization
• Autoimmune hemolytic anemia • Thermal: malfunctioning blood warmer, improper storage or transport
• Cold agglutinin disease • Mechanical: small needles, malfunctioning infusion pumps, artificial heart valves
• Drug-induced hemolytic anemia • Hemoglobinopathies: sickle cell disease, HbC disease
• Paroxysmal cold hemoglobinuria • RBC membrane or enzyme disorders: hereditary spherocytosis, glucose-6-phosphate deficiency
• Paroxysmal nocturnal hemoglobinuria • Microangiopathic hemolytic anemias: thrombotic thrombocytopenic purpura (TTP), hemolytic uremic
• IVIG administration syndrome (HUS)
• Infections: Clostridial sepsis, malaria, babesiosis

HTRs = hemolytic transfusion reactions; IVIG =intravenous immune globulin


6888_Ch17_373-395 29/10/18 5:45 PM Page 379

Chapter 17 Adverse Effects of Blood Transfusion 379

blood pressure support, which may mask hypotension. or inhibitory effects. While a mixture of different IgG
Finally, HTRs can occur in patients receiving ABO mis- subclasses is usually found in the antisera of patients with
matched platelet transfusions. Platelets are typically selected alloantibodies, one or two subclasses are typically predom-
for transfusion based on shelf life and availability rather than inant. Thus, incompatible RBCs could be coated with
major or minor ABO compatibility. Major ABO incompati- antibodies that robustly or poorly activate C′ or bind to ac-
bility may result in decreased platelet increments after trans- tivating or inhibiting FcRs. In addition, there is substantial
fusion and explain occasional cases of the phenomenon of variation in the density of RBC antigens within an individ-
platelet refractoriness (see Chapter 16). Minor ABO incom- ual as well as across a population of people capable of pro-
patibility, however, may cause HTRs due to transfusion of ducing the antigen. Finally, the effector pathways that
anti-A or, rarely, anti-B in the donor plasma. The donors of amplify or diminish the inflammatory response generated
platelet units implicated in HTRs are almost always group O by antibody-mediated hemolysis show abundant genetic
and the recipients group A. The majority of implicated and functional diversity that influences the clinical appear-
platelet products are apheresis platelets, but platelet concen- ance of an HTR.
trates have also caused HTRs. Furthermore, a recent study The downstream consequences of hemolysis that can
reported that the antibody titer did not appear to predict generate a systemic inflammatory response play a critical
HTRs after minor ABO-incompatible platelet transfusion,14 role in the appearance, severity, and clinical manifestations
which suggests that other mechanisms are involved, such as of an HTR.16 After intravascular hemolysis, haptoglobin
the antibody subtype or individual characteristics of the rapidly binds free hemoglobin. Haptoglobin-hemoglobin
transfusion recipient. HTRs due to platelets are likely under- complexes are bound to CD163 on monocytes and
recognized due to the lack of awareness of this potential macrophages, and this binding initiates receptor-mediated
problem, subtle or atypical findings of an HTR in this uptake and intracellular degradation of hemoglobin.
setting,14 and attribution of worsening anemia to other Unfortunately, the circulating supply of haptoglobin is
causes. Hemolysis due to minor ABO-incompatible platelet quickly depleted in even mild hemolysis. Free hemoglobin
transfusion should be considered for any unexplained drop is observed as visibly pink to red plasma (hemoglobinemia)
in hemoglobin following platelet transfusion. and as brown to red discoloration of the urine (hemoglo-
binuria). Free hemoglobin causes multiple effects on
circulating cells and endothelial cells that trigger an inflam-
Advanced Concepts matory response. Activation of the FcR pathway is also
The pathophysiology of HTRs is more complex than just proinflammatory. It is the inflammatory response to hemol-
an alloantibody binding to a cognate antigen to produce ysis that drives the clinical features of an HTR.
hemolysis. Evidence is accruing that suggests that an HTR
is a systemic inflammatory response to antibody-mediated
hemolysis.15,16 RBC alloantibodies mediate RBC destruction Delayed Hemolytic and Serologic Transfusion
through activating the complement (C′) cascade or through Reaction (DHTR)
the Fc receptor (FcR)-mediated phagocytosis by macrophages,
primarily in the spleen. In general, AHTRs and IgM alloan- The definition of a DHTR includes a positive DAT 24 hours
tibodies are associated with intravascular hemolysis through to 28 days after transfusion with either a positive eluate or
the C′ activation pathway, whereas delayed hemolytic trans- a newly identified alloantibody in the plasma or serum and
fusion reactions and IgG alloantibodies are associated with evidence of hemolysis.3 Evidence of hemolysis includes an
extravascular hemolysis through the FcR pathway. In reality, inadequate rise in hemoglobin after transfusion, a rapid drop
both pathways are activated, albeit to varying degrees, in in hemoglobin to the pretransfusion level, the appearance
HTRs and, more importantly, both pathways activate proin- of spherocytes on peripheral blood smear examination,
flammatory events that cause many of the signs and symp- and/or biochemical evidence of hemolysis. Delayed serologic
toms seen in patients having HTRs. In addition, crosstalk transfusion reaction (DSTR) is defined as the same serologic
between the two pathways produces a complex, integrated findings as DHTR but without evidence of hemolysis.
response to antibody-mediated hemolysis.15 Most cases of DHTR appear 7–10 days post-transfusion.
A challenge to understanding the pathophysiology Some cases, especially in transfused postsurgical patients, will
of HTRs is to account for the marked variation in severity be unsuspected because postoperative anemia is attributed to
of hemolysis and in clinical findings observed in patients other causes. Furthermore, there is often incomplete labora-
who have received incompatible blood. Laboratories focus tory evaluation to substantiate the presence of hemolysis,
on identifying IgM and IgG antibodies, but human im- other than unexplained anemia.13 In these cases, the diagno-
munoglobulins are considerably more complex and diverse sis of DHTR is left as probable or possible.
than this. For example, there are four different IgG sub-
classes, and each has differing affinities for binding antigens
and FcRs and different efficiencies for activating C′. More- Advanced Concepts
over, there are six different FcRs that each have different DHTR and DSTR are a particular problem in sickle cell
affinities for different IgG subclasses and have activating disease (SCD) patients who are chronically transfused. The
6888_Ch17_373-395 29/10/18 5:45 PM Page 380

380 PART III Transfusion Practices

problem is further compounded by fragmented care in


BOX 17–2
different hospitals and outpatient settings, resulting in in-
complete records, and variation in transfusion policies for Common Causes of Acute Lung Injury in Critically
SCD patients with respect to phenotypically matched RBC Ill Patients
transfusions. SCD patients with alloimmunization have Primary Pulmonary Causes
worse survival compared to nonalloimmunized patients,17
• Pneumonia
which may reflect the proinflammatory consequences of
• Aspiration
hemolysis. The most frequent sign of DHTR in these
• Inhalation of toxic gas
patients is hemoglobinuria, which is usually associated
• Infarct
with painful crises and often acute chest syndrome.18
• Severe asthma
Mortality is 5% to 10%. In addition, multiple alloantibod-
ies and/or the development of antibodies against high- Secondary Causes Due to Systemic Diseases
frequency antigens makes obtained antigen-negative blood • Sepsis
very difficult, especially in an urgent manner.12 Molecular • Shock
genotyping in SCD patients with their first alloantibody • Trauma
should be considered to identify variant or “partial” phe- • Burns
notypes, such as with D, c, E, and U. For SCD patients with • Pancreatitis
alloimmunization, indications for RBC transfusion should • Drug overdose
be restricted.
Patients with SCD can also rarely develop hyperhemol-
ysis syndrome following transfusion. The syndrome has
alternative diagnoses to TRALI that are controversial but
also been reported in other conditions, including tha-
present in the literature, and they could be related to those
lassemia, myelofibrosis, and lymphoma. Hyperhemolysis
cases that clearly fit the diagnosis of TRALI. “Possible
is a devastating, often fatal complication of HTRs in SCD
TRALI” is designated when the above criteria are present
patients. This syndrome typically develops 7 to 10 days
but another cause of ALI is also identified, while “delayed
after transfusion and is manifested by signs and symptoms
TRALI” refers to when criteria for TRALI are present but
of intravascular hemolysis, painful vaso-occlusive crises,
the onset is 6–72 hours after transfusion.21
and severe anemia with a hemoglobin level below that if
The incidence of TRALI is difficult to precisely determine
the pretransfusion level. Notably, the DAT is often nega-
and ranges between 0.08% and 15% of patients receiving
tive and previously detected antibodies are frequently not
blood transfusion. The wide variation in the reported inci-
identified.19 Hemolysis of both patient and transfused
dence reflects variation in the definition used for TRALI, the
RBCs can be inferred from hemoglobin electrophoresis.
inclusion of possible and delayed TRALI, the time period
The pathogenesis of hyperhemolysis syndrome is not
and population studied, and how data is collected. It is
well understood but several mechanisms are likely in-
generally appreciated that TRALI is underreported and un-
volved, including bystander hemolysis from C′ activation,
derrecognized. However, it is well established that TRALI
increased macrophage destruction of RBCs, and increased
is 50–100 times more common in critically ill and surgical
programmed cell death of RBCs (called eryptosis) due
patients than in the general hospital population.22 Also, there
to increased circulating proinflammatory mediators.20
is a wide variation in disease severity ranging from a need
Transfusions should be withheld, as they tend to make
for supplemental oxygen to supportive mechanical ventila-
hemolysis worse.
tion to death. Many TRALI reactions are likely undetected
due to the complexity of critically ill and surgical patients
Transfusion-Related Acute Lung Injury (TRALI) under general anesthesia and that TRALI definitions would
exclude mild forms of the reaction.21
TRALI is a rare event associated with acute respiratory The clinical presentation is acute respiratory distress,
distress but a leading cause of mortality due to adverse including dyspnea, tachypnea, and hypoxemia. Signs and
reactions to transfusion. There is no diagnostic test for symptoms may also include fever, rigors, tachycardia, hy-
TRALI. The diagnosis of TRALI is primarily made accord- pothermia, and hypotension. Radiographic abnormalities
ing to clinical criteria that support the diagnosis and, im- may be subtler than the classical bilateral “white-out” ap-
portantly, the exclusion of other possible causes of acute pearance of acute pulmonary edema. Laboratory tests are not
lung injury. The NHSN Hemovigilance definition3 includes very helpful in the diagnosis. Transient leukopenia and
(1) the absence of acute lung injury (ALI) prior to transfu- thrombocytopenia can be present in about 25% of patients.
sion, (2) ALI during or within 6 hours after transfusion, Multiple patient risk factors for TRALI have been identified
(3) evidence of hypoxemia by blood gas or oxygen satura- (Box 17–3). Testing for anti-human neutrophil antigen
tion testing, (4) radiographic evidence of bilateral pul- (HNA) and anti-HLA antibodies can be done on donors of
monary edema, and (5) exclusion of circulatory overload implicated products once a suspected TRALI reaction is
and other causes of pulmonary edema. Box 17–2 shows identified. The differential diagnosis for acute respiratory
other common causes of acute lung injury. There are two distress in a transfused patient includes sepsis, anaphylactic
6888_Ch17_373-395 29/10/18 5:45 PM Page 381

Chapter 17 Adverse Effects of Blood Transfusion 381

It is recognized now that there are non-antibody-


BOX 17–3
mediated cases of TRALI caused by certain mediators in
Patient Risk Factors for TRALI the transfused products that activate endothelial cells in
• Mechanical ventilation the pulmonary microcirculation that induce neutrophil
• Cardiac surgery requiring cardiopulmonary bypass activation, capillary leakage, and pulmonary edema.26 It
• Patients receiving multiple transfusions, such as hematological is hypothesized that proinflammatory mediators accumu-
malignancies with intensive chemotherapy, liver failure with active late in the blood product released by RBCs and platelets
bleeding during storage or as a result of erythrocyte and platelet
• Massive transfusion aging. Mediators implicated in the process are bioactive
• Positive fluid balance lipids derived from cell membranes and soluble CD40, a
proinflammatory mediator produced by platelets. It
appears that platelet–neutrophil interactions are an im-
reactions, TRALI, transfusion-associated circulatory over- portant intermediate step in the development of non-
load, and other causes of acute lung injury. antibody-mediated TRALI. Microparticles and morpho-
Substantial evidence has implicated transfused donor logical and biochemical changes that increase adhesion of
anti-WBC antibodies to patients at risk for TRALI in the RBCs and platelets to endothelial cells may also be impor-
pathophysiology of TRALI. Investigation into the rising tant. Finally, loss of Duffy antigen for chemokines on
incidence of TRALI in the early 2000s revealed a strong as- RBCs during storage may increase cytokine availability
sociation between women donors who have been previously for neutrophils and contribute to the pathogenesis of
pregnant and TRALI reactions.23,24 Indeed, the prevalence of TRALI.26
anti-HLA antibodies increases with the number of pregnan- Neither all blood products nor all patients have an
cies. Similar studies in the UK and Canada found a similar equal risk for TRALI. The majority of blood products that
association, and these countries adopted a mitigation strat- contain anti-WBC antibodies do not cause TRALI, even
egy to minimize transfusion of high-volume plasma compo- in patients with cognate antigens. This has led to a “two-
nents from female donors. This also prompted a similar hit” or threshold model for the pathogenesis of TRALI
mitigation strategy in the United States sponsored by AABB (Fig. 17–3). In this model, the first hit involves neutrophil
and adopted by most blood centers.23,24 attraction to the pulmonary alveolar capillary network.
The second hit involves neutrophil activation, which sets
off a cascade of proinflammatory events culminating
in pulmonary capillary damage and the development of
Advanced Concepts
pulmonary edema. The threshold model allows for the
Although the incidence of TRALI has decreased after the development of TRALI in patients without necessarily un-
implementation of mitigation strategies, TRALI reactions dergoing a first hit when the stimulus involved in the sec-
continue to occur. This has led to a reconsideration of the ond hit is sufficiently strong to overcome the barriers and
pathogenesis of TRALI to propose “antibody-mediated” pathways in place to prevent activation of inflammation
and “non-antibody-mediated” forms of TRALI25,26 and a and capillary endothelial damage.25
two-hit model in the pathogenesis of TRALI.21
The majority of TRALI cases have implicated both anti-
HLA class I and anti-neutrophil antigen (anti-HNA) alloan- Transfusion-Associated Circulatory
tibodies from passive transfusion of these antibodies from Overload (TACO)
donors. Neutrophil activation occurs through direct bind-
ing of anti-HNA antibodies or through indirect binding of TACO is an adverse reaction characterized by acute respira-
anti-HLA antibodies to endothelial cells and monocytes tory distress from pulmonary edema caused by increased
and subsequent neutrophil activation. Both the titer of the intravascular volume due to excessive transfused fluid and/or
anti-WBC antibodies and the volume of plasma containing too rapid of an infusion rate and the inability of the patient
these antibodies increase the risk of TRALI.22 Although to accommodate the volume of transfused products due to
anti-HLA antibodies are the most frequently implicated impaired pulmonary, cardiac, or renal function. The NHSN
antibodies with TRALI, antibodies to HNA-1, HNA-2, and Hemovigilance definition3 specifies the presence of three
HNA-3a are associated with the most severe, even fatal, or more features of fluid overload (Table 17–7) occurring
TRALI reactions.25 Antibodies to recipient HLA or HNA within 6 hours after the transfusion. The signs and symp-
antigens must be detected in implicated donors in the toms frequently seen in TACO generally occur within
evaluation of TRALI cases to confirm the diagnosis of 2 hours after the start of transfusion but may take up to
antibody-mediated TRALI. This testing is also useful to 6 hours to be seen.
identify donors who pose a risk for TRALI to patients. TACO is the second most common cause of transfusion-
However, this testing has no value in the diagnosis of an related deaths reported to the FDA, and the incidence
acute TRALI reaction. It is available only in specialized appears to be increasing.2 Similar to TRALI, the incidence is
reference laboratories and takes several weeks to get results. most likely underestimated and ranges from 1% to 8% de-
pending on whether passive or active surveillance methods
6888_Ch17_373-395 29/10/18 5:45 PM Page 382

382 PART III Transfusion Practices

First Hit RBC FFP PLT Second Hit


(Patient Factors) (Transfusion Factors)
• Sepsis RBC
• Haematological malignancy • HLA or HNA
• Heart surgery • Bioactive lipids
• Mechanical ventilation • sCF40L
• Massive blood transfusion • Aged erythrocyte
• Chronic alcohol abuse
• Age of patient FFP
• Shock • HLA or HNA
• Acute renal failure
• Severe liver disease PLT
• Spine surgery • HLA or HNA
• Liver surgery • Bioactive lipids
• sCD40L

Figure 17–3. The two-hit model of TRALI. The first hit involves patient factors that prime neutrophils and/or pulmonary endothelial cells. The second hit is related to the
transfused product. Some factors are specific to a particular product, while others are common to all products. HLA = human leukocyte antigen, HNA = human neutrophil
antigen. From: Vlaar AP, Juffermans NP. Transfusion-related acute lung injury—a clinical review. Lancet. 2013;382:990, with permission.

Table 17–7 Features Defining Transfusion- chronic kidney disease, left ventricular heart dysfunction,
previous beta-adrenergic receptor antagonist use, isolated
Associated Circulatory
FFP (vs. RBC) transfusion, mixed product (vs. RBC only)
Overload
transfusion, and increased intraoperative fluid administra-
• Acute respiratory distress tion were associated with TACO.30 In addition, the authors
• Elevated brain natriuretic peptide (BNP) found that patients who developed TACO were more likely
• Elevated central venous pressure to require mechanical ventilation and have prolonged stays
• Evidence of left heart failure
• Evidence of positive fluid balance in the ICU and hospital as well as a significant reduction in
• Radiographic evidence of pulmonary edema survival compared to transfusion recipients who did not
develop TACO.29 Figure 17–4 illustrates a multiphasic spec-
trum of clinical appearance of TACO. This model of TACO
shows the relationship between transfusion and the body’s
are used to identify cases.27,28 The severity of the morbidity response to infusion. TACO is hypothesized to occur over a
due to TACO is also underestimated. Notably, data from the time interval and is categorized into four stages. These stages
UK Hemovigilance Network 2015 Serious Hazards of Trans- correlate with the amount of fluid (blood and nonblood
fusion (SHOT) report found that 6.6% of cases reported as products) infused to the recipient and the clinical severity
TACO were identified more than 6 hours after transfusion, observed in the patient. But it must be remembered that
and another 7.7% of cases were more than 12 hours after there is not a direct relationship between transfused volume
transfusion.29 and the severity of clinical symptoms. The ability of an indi-
TACO has been associated with the number of blood vidual patient to handle excessive fluid depends on individ-
products transfused, although this has been questioned ual variation and underlying diseases, such as anemia and
recently,27 and the infusion rate is also a contributing factor cardiac, pulmonary, or renal dysfunction.31
in some cases. However, individual patient factors, especially The measurement of brain natriuretic peptide (BNP)
the underlying cardiac capacity and fluid status, are the levels, such as NT-proBNP, may be helpful in the diagnosis,
most important contributors to the development of TACO. although it may be elevated in other conditions. A value of
A recent study showed statistically significant associations 1.5 times greater than the pretransfusion level supports the
between TACO and chronic renal failure, history of conges- diagnosis. Serial BNP level determinations following post-
tive heart failure, hemorrhagic shock, the number of blood transfusion pulmonary complications for the assessment of
products transfused, and positive fluid balance.27 These au- TACO have been recommended.30
thors also found a surprising inverse correlation with age, TACO is generally treated by stopping the transfusion
since previous studies noted an association between TACO and taking immediate measures to stabilize the patient, de-
and elderly patients. They suggest that more aggressive fluid pending on the severity of the reaction. These steps include
resuscitation in younger patients might account for this placing the patient in a more upright posture, administering
result. Another recent study focused on the perioperative supplemental oxygen, stopping infusion of other intra-
period in surgical patients and found that emergency surgery, venous fluids, and administering diuretic medications.30
6888_Ch17_373-395 29/10/18 5:45 PM Page 383

Chapter 17 Adverse Effects of Blood Transfusion 383

The Multiphasic Spectrum of TACO

Figure 17–4. The multiphasic spectrum of NP levels


clinical appearance of TACO. A multiphasic
model of TACO shows the relationship
between transfusion and the body’s response
to infusion. TACO occurs over a time interval
and is categorized into four stages. The up and BP spikes
down arrows indicate variations in presentation
between patients. The ability of an individual
patient to handle excessive fluid depends on Pulmonary
individual variation and underlying diseases, signs/Symptoms
such as anemia and cardiac, pulmonary,
or renal dysfunction. (From: Andrzejewski C,
Time Early Early Early
Casey MA, Popovsky MA. How we view and
approach transfusion-associated circulatory Stage I II III IV
overload: pathogenesis, diagnosis, manage- No TACO Mild TACO Moderate TACO Severe TACO
ment, mitigation, and prevention. Transfusion.
2013;53:3042, with permission.) (Normovolemia) Hypervolemia

It is important that clinicians and transfusionists report sus- still observed even in the era of prestorage leuko-depleted
pected TACO reactions to the transfusion service so that products.34,35
other reactions, especially acute hemolytic reactions and
TRALI, can be excluded. Febrile Nonhemolytic Transfusion Reaction
(FNHTR)
Transfusion-Associated Dyspnea (TAD)
FNHTRs are one of the most common adverse transfusion
Dyspnea, occurring alone or as the predominant symptom, reactions. Although these reactions are generally mild and
could be seen in allergic reactions, TACO, or TRALI, self-limited, they are important for a number of reasons.
or could be related to the patient’s underlying condition. They can result in discontinuation of the transfusion,
TAD is diagnosed when dyspnea occurs within 24 hours after wastage of the residual product, and delay in additional
transfusion and all other diagnoses are excluded.3 The patho- transfusions. More importantly, FNHTRs mimic more seri-
physiology is unknown. A recent detailed analysis of cases ous and severe transfusion reactions, such as, AHTR, TRALI,
designated as TAD shows most are cases of mild TACO, thus TACO, and transfusion-transmitted bacterial infection.
the validity of this entity has been called into question.32 FNHTR is defined as fever greater than 100.4°F (38°C) or a
Before any case is diagnosed as TAD, scrupulous efforts to change of at least 1.8°F (1.0°C) from the pretransfusion level
identify other entities should be made, and the diagnosis occurring during or within 4 hours after the end of the
should almost always be made with a qualification as “pos- transfusion or chills and/or rigors are present.3 An FNHTR
sible” or “probable.” is diagnosed after the patient’s underlying condition and
medications are excluded as a cause. Fever usually occurs
Hypotensive Transfusion Reaction with chills or rigors, but chills or rigors without fever can
still be diagnosed as FNHTR in spite of the absence of fever.
Hypotension is a nonspecific sign that could be seen in a Other symptoms that occur with FNHTR include headache,
variety of other transfusion reactions or as part of the pa- cold feeling, mild dyspnea, and mild nausea/vomiting. Symp-
tient’s underlying condition. This entity is diagnosed when toms typically begin toward the end of the transfusion, but
hypotension is seen alone during or within 1 hour after the 5% to 10% do not appear until 1 to 2 hours after the end of
transfusion is finished. Hypotension is defined by NHSN the transfusion.36 Despite the relatively straightforward
criteria in adults as a drop in the systolic BP of ≥30 mm Hg definition, a definitive or even possible diagnosis of FNHTR
and systolic BP ≤80 mm Hg. In children, it is defined as a is often challenging. Difficulties in making a diagnosis of
25% drop in the baseline systolic BP.3 Initial case reports im- FNHTR include complex medical conditions, multiple
plicated bedside leukoreduction filters and/or angiotensin- medications, delay between the appearance of symptoms and
converting enzyme (ACE) inhibitors in acute hypotensive the end of the transfusion, and symptoms occurring during
reaction.33 Increased bradykinin levels in stored blood or after multiple transfused products. The change in temper-
products are hypothesized to be responsible for the patho- ature is usually modest (less than 2°C/3.6°F), but when it is
physiology of these reactions, but the pathophysiology of greater than 2°C, transfusion-transmitted bacterial infection
hypotensive transfusion reactions are not understood. In must be excluded by visual inspection of the product for
addition, two recent studies show that these reactions are discoloration or clots and culture of the implicated product.
6888_Ch17_373-395 29/10/18 5:45 PM Page 384

384 PART III Transfusion Practices

The frequency of FNHTR in the general hospital population should be actively discouraged by the transfusion medicine
is about 1 in 200 units transfused. The incidence is physician evaluating such a reaction.38
much higher in hematology/oncology patients and chronic
transfusion patients, such as with sickle cell disease.36 The Allergic Transfusion Reactions (ATRs)
incidence is approximately the same for prestorage leukore-
duced RBCs and apheresis platelets but very uncommon ATRs are the most common reactions seen with platelet and
for plasma products. plasma transfusions, occurring in about 2% of transfused
Prior to widespread prestorage leukoreduction, available platelets, and are second to FNHTRs in RBC transfu-
evidence was consistent with recipient anti-WBC antibodies sions.39,40 Severe anaphylactic reactions resulting in car-
triggering release of proinflammatory cytokines as the cause diopulmonary resuscitation or death are rare. Estimates of
of most FNHTRs. Although the incidence of these reactions the frequency of severe ATRs range from 1 per 20,000 to
significantly decreased after implementation of bedside 1 per 47,000 units transfused. The terms “allergic,” “anaphy-
leukoreduction filters and then from near universal prestor- lactoid,” and “anaphylactic” are used to generally categorize
age leukoreduction of RBCs and platelets, FNHTRs still ATRs. Allergic reactions are generally mild to moderate
continue to be observed. Other observations, particularly reactions and refer to signs and symptoms limited to the skin
with platelet transfusion, also resulted in questions regarding and gastrointestinal tract. Anaphylactoid reactions are mod-
whether all of these reactions were immune mediated erately severe reactions that include oral and throat symp-
and suggested there might be other mechanisms involved toms, more severe GI symptoms, and respiratory complaints.
in the pathophysiology of these reactions.36 Laboratory Anaphylactoid reactions are severe, life-threatening reactions
studies of platelet products showed direct relationships be- in which there is profound hypotension and shock. Like
tween the number of residual WBCs in platelets, levels of FNHTRs, ATRs are important because of their frequency,
proinflammatory cytokines, and storage time. A variety of the costs involved in blood product wastage, delays in the
other leukocyte- and platelet-derived cytokines has been patient receiving adequate blood transfusion therapy, and
studied for accumulation in the liquid portion of RBCs and overlap with other serious reactions.
platelets.37 The accumulation of these proinflammatory There is a wide range of clinical severity found with
cytokines is the result of metabolically active residual WBCs ATRs, but the majority are mild skin reactions. The clinical
in the product. Platelet products, compared to RBC products, signs and symptoms are similar to those described for
are more likely to be affected by this process because of the allergic reactions from environmental or drug causes. The
differences in storage conditions. Room temperature storage most common manifestations of ATRs are mucocutaneous
and agitation have been shown to affect cytokine levels in reactions, including urticaria, pruritus, and localized or
platelet components. Patient factors are also important for generalized rash. The NHSN Hemovigilance definition of
the development of FNHTRs. A recent study of elderly adults ATRs includes only anaphylactoid and anaphylactic signs
showed that patients with preexisting conditions that cause and symptoms.3 A definite diagnosis of ATR includes
elevated proinflammatory cytokine levels appear to be more the appearance of signs and symptoms during or within
susceptible to FNHTRs.38 In addition, genetic polymor- 2 hours after the end of transfusion and the exclusion of
phisms in the cytokines themselves or their receptors are other possible drug, environmental, and dietary causes.
likely to play a role in susceptibility to reactions. It remains The clinical signs and symptoms of ATRs can be catego-
to be confirmed whether or not these results are applicable rized into mucocutaneous, gastrointestinal, respiratory,
to all patient populations. and cardiovascular (Table 17–8). The differential diagnosis
The treatment of FNHTRs is primarily antipyretics, but of ATR includes TRALI and TACO when respiratory
the primary management goal is to rule out more serious symptoms are prominent and AHTRs and transfusion-
transfusion reactions. Following a protocol-driven process transmitted bacterial infection when hypotension and/or
will ensure that AHTRs are excluded. Clinical awareness of shock are present.39
findings to evaluate for TRALI and sepsis is essential. Unfor- Mild cutaneous reactions are generally managed by
tunately, many patients with febrile reactions are treated with administration of diphenhydramine. More severe urticarial
an antihistamine and/or steroids, which has no benefit and reactions or those accompanied by oropharyngeal or upper

Table 17–8 Signs and Symptoms of Allergic Transfusion Reactions


Mucocutaneous Gastrointestinal Respiratory Cardiovascular
• Urticaria • Nausea • Throat tightness • Hypotension
• Pruritis • Vomiting • Hoarseness • Tachycardia
• Facial or generalized flushing • Abdominal pain or cramps • Stridor • Shock
• Maculopapular rash • Diarrhea • Wheezing
• Erythema and swelling around the eyes • Chest tightness
• Swelling of lips or tongue • Dyspnea
• Localized angioedema
6888_Ch17_373-395 29/10/18 5:45 PM Page 385

Chapter 17 Adverse Effects of Blood Transfusion 385

respiratory symptoms could be treated with steroids. Adverse Reactions to Infusion of Plasma-Derived
Epinephrine may also be used. Severe anaphylactoid or Products
anaphylactic reactions require prompt action to maintain
oxygen and blood pressure. Antihistamines are frequently Plasma-derived products, such as albumin, intravenous
used prior to transfusion to prevent an ATR. There is immune globulin (IVIG), and human-derived factor con-
no evidence showing the benefit from this practice, yet centrates, can be associated with adverse events other
many hospitals still have this included in standing order than viral transmission. Note that in the United States, he-
sets.39,40 mophilia patients receive recombinant factor VIII or IX.
Albumin has been associated with severe hypotensive
reactions in patients receiving ACE inhibitors (a class of
Advanced Concepts drugs used to treat hypertension). IVIG is associated
with a variety of adverse events. Most reactions are mild
IgE-mediated type I hypersensitivity reactions do not explain
to moderate allergic reactions that are transient in duration
all ATRs. It has been assumed that the pathophysiology of
and self-limited. Headache is the most commonly reported
ATRs were type I hypersensitivity reactions because of their
symptom. Fever, chills, nausea, vomiting, abdominal
similar clinical features to well-characterized reactions due
pain, diarrhea, facial flushing, urticaria, itching, muscular
to environmental or drug allergies and that ATRs were
cramps, and back pain are other common symptoms.42 The
mediated by IgE antibodies to plasma proteins. Severe ATRs
reported prevalence of adverse reaction associated with
due to plasma proteins such as IgA and haptoglobin are well
IVIG ranges widely from less than 1% to more than 80%.
described in patients with deficiencies of these proteins
For patients receiving IVIG, it is important for patients and
(see Box 17–4). There are, however, no reliable estimates of
clinicians to be aware that the onset of symptoms can be
the incidence of ATRs due to IgA or haptoglobin deficiencies
quite delayed, up to 7 days, following IVIG infusion.42
nor the number of IgA- or haptoglobin-deficient patients
who have specific alloantibodies that have not experienced
Transfusion-Associated Graft-Versus-Host Disease
an ATR.41 While exposure to a recipient to foreign donor pro-
teins could result in sensitization and ATRs upon reexposure, Transfusion-associated graft-versus-host disease (TA-GVHD)
as with IgA deficiency, this does not appear to be a common is a rare but devastating event with a very high mortality rate
mechanism. Patients with coagulation factor deficiencies or compared to other transfusion reactions as well as with
alpha-1-antitrypsin deficiency have very low rates of severe GVHD occurring in marrow/hematopoietic and solid organ
allergic reactions. On the other hand, plasma protein poly- transplantation. The NHSN definition of TA-GVHD is a
morphisms might explain some intermittent ATRs seen in clinical syndrome developing from 2 days to 6 weeks after
repeatedly transfused patients.40 transfusion characterized by the typical skin rash seen and
IgE independent mechanisms have been proposed that other forms of GVHD, diarrhea, fever, enlarged liver, elevated
implicate IgG, direct C′ activation, and platelet-derived liver enzymes, marrow aplasia, and/or pancytopenia.3 A def-
factors. A model of ATRs has been proposed in which the inite diagnosis is made by skin or (occasionally) liver, biopsy
appearance and severity of the reaction depends on the pre- showing characteristic histological features (Fig. 17–6). A
disposition of the recipient to allergic reactions and specific probable diagnosis of TA-GVHD is made when the clinical
donor or product factors (Fig. 17–5).40 criteria are met but a biopsy is negative or was not per-
formed. The most common symptoms are rash, fever, in-
creased liver enzymes, pancytopenia, and diarrhea.43 The
first symptoms appear on average 11 days from the impli-
BOX 17–4 cated transfused unit, but there is a wide range of disease
onset. Uncommonly, symptoms can occur over 6 weeks after
IgA Deficiency and ATRs
transfusion.44 Death occurs in about 90% of patients with a
IgA deficiency is one of the most common mutations in people of median of 24 days after the implicated transfusion. Mortality
European ancestry, present in about 1 in 700 individuals, and is the
is associated with older age, whole blood transfusion, local
most common primary antibody deficiency. There are marked ethnic
differences in the incidence of IgA deficiency, and it is rare in Asian reduced product, and short storage duration (fresh or less
and African groups. IgA deficiency is defined as undetectable serum than 48 hours old).44
IgA (less than 0.5 mg/dL). Most IgA-related anaphylactic reactions The rash begins as a maculopapular rash that starts over
occur in IgA-deficient persons with detectable class-specific anti-IgA the trunk and then spreads to the limbs. It is indistinguish-
antibodies. However, reactions do not occur in the majority of
able from viral and drug-induced rashes. The histological
patients, and anaphylaxis is rare. A reasonable approach to IgA-
deficient patients is to provide plasma and cellular components from features of GVHD are well described in the skin, but the
IgA-deficient donors only for IgA-deficient patients who have anti- leukocyte chimerism (the presence of donor lymphocytes and
IgA. Washed or plasma-reduced cellular components are an accept- recipient tissue) can be tested for when biopsy findings are
able alternative if IgA-deficient donor components are unavailable. equivocal.44 Analysis of microsatellite DNA polymorphisms
IgA-deficient patients without anti-IgA or history of severe ATR do
has also been used to diagnose TA-GVHD by demonstrating
not need IgA-deficient donor products or washed or plasma-reduced
cellular components. replacement of recipient WBCs by donor WBCs in peripheral
blood.45
6888_Ch17_373-395 29/10/18 5:45 PM Page 386

386 PART III Transfusion Practices

Atopic Predisposition of Recipient Necessary Plasma Mediator


• Chronic/genetic • What
– Primed basophils and mast cells – Protein (e.g., IgA, haptoglobin)
– Preformed IgE – Direct allergic agonist (e.g., CCL5,
• Subacute/acquired anaphylatoxin)
– Passively acquired IgE – Additive (e.g., ethylene oxide, plasticizer)
– Inflammatory/cytokine state of • How
underlying disease – Storage effect (e.g., platelet release)
– Modification (e.g., oxidation)
– Donor-dependent (e.g., multimeric IgE,
anti-CD36)

Allergic Transfusion Reaction

Severe
ATR
Figure 17–5. Model for pathogenesis of ATR.
ATR Severity

No ATR Isolated Recurrent ATRs are hypothesized to result from a predispo-


ATR ATR sition of the recipient to allergic reactions in
general (atopy) and the transfusion of some
Threshold for ATR plasma mediator listed. The severity of the ATR
may be related to the susceptibility of the recipi-
ent to an allergic reaction at the time of the trans-
fusion and the amount of the plasma mediator
that is infused with the product. (From: Savage
WJ, Tobian AAR, Savage JH, Wood RA, Schroeder
Plasma mediator: — — JT, Ness PM. Scratching the surface of allergic
transfusion reactions. Transfusion. 2013;53:
Allergic susceptibility: — — 1366, with permission.)

systematic review of published cases and other recent studies


challenge the notion that the immune status of the recipient
is the main determinant for susceptibility to TA-GVHD.44–46
About 75% of patients with TA-GVHD in the systematic
review and 65 out of 66 patients in another study45 were con-
sidered immunocompetent. The majority of cases of periop-
erative TA-GVHD were also in immunocompetent patients.
Most of these patients underwent cardiac surgery involving
cardiopulmonary bypass, but TA-GVHD has been associated
with a variety of other surgical procedures.46 Risk factors for
TA-GVHD include male sex, age greater than 70 years, cardio-
vascular surgery, whole blood, and fresh blood products.44
A component was implicated in only 70% of cases. The im-
plicated unit was either not specified or not identified among
several potentially responsible components in the other 30%.
The storage time of implicating components was fresh for less
Figure 17–6. Skin biopsy showing acute graft-versus-host disease. Histological
features of acute GVHD include superficial chronic inflammation at the junction be- than 10 days in 90% of cases. The closeness of the relationship
tween the epidermis and dermis, vacuolar change at the junction, single cell necrosis between the donor and recipient is another important factor
(apoptosis) of keratinocytes in the epidermis, and clusters of lymphocytes around in TA-GVHD. Donor HLA homozygosity for an HLA haplo-
individual keratinocytes (lymphocyte satellitosis). (Case provided by Dr. Vijaya type for which the recipient was heterozygous was present
Reddy, Rush University Medical Center.)
in almost 90% of cases of patients who were immunocompe-
tent.45 Donor immunologically competent CD8+ cytotoxic
The pathogenesis of TA-GVHD is not well understood, but T lymphocytes are directed against recipient cell surface histo-
it appears to be related to recipient immune status, component compatibility antigens and mediate cell destruction. Although
characteristics, and donor-recipient HLA relationships. Early the recipient is immunocompetent, donor lymphocytes are not
case reports found congenital and acquired immunodeficiency recognized as sufficiently foreign to be eliminated because of
syndromes were associated with TA-GVHD. However, a shared HLA antigens.
6888_Ch17_373-395 29/10/18 5:45 PM Page 387

Chapter 17 Adverse Effects of Blood Transfusion 387

The treatment for TA-GVHD is immunosuppressive majority of patients respond, sometimes within hours of the
medications. Hematopoietic stem cell transplantation has first infusion. Plasma exchange with FFP is the next option for
been reported to be successful, but only a small number of those patients who do not respond to IVIG.
patients have been treated so far.44 Irradiation of cellular
blood components is successful in preventing TA-GVHD.43 Refractoriness to Platelet Transfusion
Since universal irradiation of blood components began in and Alloimmunization
Japan in 2000, there have been no cases of TA-GVHD in pa-
tients supplied with blood only from JRC blood centers.45 Patients receiving multiple platelet transfusions may not
But in the United States there is considerable variation in the achieve expected platelet increments following platelet trans-
indications for irradiation of blood components between fusion and are considered refractory to platelet transfusion.
different specialty societies and institutions. A recent CAP Some of these patients do not respond to platelet transfu-
Survey showed that the most frequent indications for irradi- sions because they have become alloimmunized to HLA
ated products were blood donations from a relative and antigens or platelet-specific antigens in a manner analogous
HLA-matched platelets.47 Other common indications include to RBC alloimmunization.
neonatal exchange transfusion, intrauterine transfusion,
transfusion for preterm or low-birth-weight infants, and Adverse Metabolic Effects of Transfusion
hematopoietic stem cell transplant. Surprisingly, only 57%
The chief metabolic effects of transfusion involve citrate
of laboratories reported requiring irradiation for congenital
toxicity and hyperkalemia (increased potassium). Sodium
immunodeficiencies. In addition, a considerable number of
citrate is an anticoagulant that is a component of all RBC
laboratories required irradiation for patients generally not
preservative solutions. Citrate binds to calcium and magne-
considered to be at risk for TA-GVHD, such as healthy new-
sium to prevent activation of coagulation factors. Normally,
borns and patients with solid tumors undergoing chemother-
citrate is rapidly metabolized in the liver following transfu-
apy.47 Clearly, consensus guidelines are needed regarding
sion. However, excessive amounts of citrate can enter the
patient groups most at risk for TA-GVHD and requiring
circulation in massive transfusion or in patients with severe
irradiated cellular blood products.
liver disease. Citrate toxicity can cause hypocalcemia and
Post-Transfusion Purpura hypomagnesemia. In addition, metabolic alkalosis can de-
velop from bicarbonate produced from citrate metabolism.
Post-transfusion purpura (PTP) is a rare transfusion reaction Table 17–9 lists the major clinical effects of citrate toxicity.
in which there is a severe and sudden drop in the platelet During RBC storage, intracellular potassium slowly leaks
count, usually occurring 5 to 10 days after transfusion due from aging RBCs causing increased potassium in the super-
to alloimmunization to platelet-specific antibodies from natant. Hyperkalemia is very uncommon in massive trans-
prior transfusion or pregnancy.48 By NHSN definition, fusion in adults, but it is of special concern in neonatal
thrombocytopenia to less than 20% of the pretransfusion transfusions, especially in premature infants. The signs and
count and demonstration of alloantibodies against platelet- symptoms of increased potassium are due to neuromuscular
specific antigens are required for diagnosis.3 Usually, the im- and cardiac effects (see Table 17–9). Transfusion-associated
plicated alloantibodies are directed against human platelet hyperkalemia has been reported in infants and is associated
antigens HPA-1 or HPA-3a. It is estimated that PTP occurs with large volume transfusions and contributing comorbidi-
in about 1 in 25,000 transfused units. Over 90% of well- ties. Hyperkalemia can be prevented in large volume RBC
defined cases of PTP are women.48 transfusions to infants by issuing “fresh” RBC units (less
Thrombocytopenia is usually severe. Over 80% of patients than 7–10 days old). However, if only older units are avail-
have platelet counts less than 10,000. Patients typically have able, washed RBCs can be an acceptable alternative. Further-
purpura (slightly raised dark red–purple patches on the more, if irradiated products are required, these should also
skin), bleeding from mucous membranes (such as from the be washed or volume reduced if they are not going to be
gums or nose), GI bleeding, and hematuria. Most cases are transfused promptly after irradiation.
self-limited. The duration of thrombocytopenia in untreated
patients is about 2 weeks. Mortality has been reported to
Table 17–9 Clinical Effects of Citrate
range from 0% to 13%, mainly due to spontaneous intracra-
and Potassium Toxicity
nial hemorrhage. There are a variety of different serological
tests to detect and identify platelet alloantibodies, and geno- Citrate Toxicity: Potassium Toxicity:
typing is available to identify patients, at-risk family mem- Hypocalcemia Hyperkalemia
bers, and potential platelet antigen–negative donors.49 • Tingling of lips or fingertips • Muscular weakness
Patients are generally not responsive to either antigen • Twitching or tremors • Absent bowel sounds (‘ileus’)
untested or antigen-negative platelets, but antigen-negative • Shivering • EKG abnormalities: peaking of
platelets can be considered in patients with severe thrombocy- • Muscle contractions T waves, prolonged P-R interval
topenia and bleeding.48 However, obtaining antigen-negative (carpopedal spasms involving • Ventricular fibrillation
hands or feet) • Cardiac arrest
platelets, especially for those patients with alloantibodies other • EKG abnormalities: prolonged
than anti-HPA-1, can be very difficult. The first line of treat- QT interval
ment for PTP is intravenous immunoglobulin (IVIG), and the
6888_Ch17_373-395 29/10/18 5:45 PM Page 388

388 PART III Transfusion Practices

Controversy: Immunomodulatory Effects


Advanced Concepts of Transfusion
The storage and banking of blood is a necessary evil in order
to be able to provide blood to patients in a safe and timely Transfusion immunomodulation refers to a variety of research
manner. However, blood stored in artificial preservative findings from animal models and in vitro studies as well as
solution and under refrigerated conditions is not a normal clinical observations that suggest that allogeneic transfusion
condition. In vitro and in vivo metabolic and morphological has significant functional effects on the recipient’s immune
changes associated with RBC storage have been collectively system. Allogeneic transfusion has been associated with a
called the “RBC storage lesion” (Table 17–10).50,51 Although variety of positive and negative clinical effects, including
the details are different, a similar phenomenon is described decreased activity of autoimmune diseases, impaired wound
for platelets called the “platelet storage lesion.”52 The clin- healing, repetitive spontaneous abortions, acceleration of the
ical importance of these changes, however, in terms of trans- clinical course of HIV and CMV infections, earlier and higher
fusion outcomes and patient morbidity, is controversial. rates of tumor recurrence in colorectal cancer and other solid
Recent prospective studies have not shown any adverse tumors, and postoperative bacterial infection.59
effects between fresh and older blood in three different A number of animal studies suggest a mechanism for
patient groups (see Table 17–11).53–55 However, a recent transfusion immunomodulation after allogeneic transfusion
Cochrane review highlights that there are significant prob- is dysregulation of cellular immunity, namely, alterations in
lems with the studies to detect small, but clinically signifi- the responses of T-helper cells (Th1 and Th2 cells).59 Th1
cant, differences.56 While the evidence is indisputable that cells secrete cytokines, such as interferon-γ, tumor necrosis
there are significant measurable metabolic changes and factor, and interleukin-2 (IL-2), that are proinflammatory and
observable morphological changes in RBCs and platelets associated with macrophage activation. Th2 cells primarily
during prolonged storage, the current evidence neither secrete IL-4 and IL-5, which support humoral immune
supports nor rejects the hypothesis that there are clinically responses through B-cell differentiation. Both human and
deleterious effects due to transfusion of RBCs or platelets animal studies show that allogeneic transfusion is associated
after such storage. with increased secretion of Th2 cytokines and decreased
New technologies, especially mass spectroscopy-based secretion of Th1 cytokines. These effects are correlated with
metabolomics, are giving fresh insights into the RBC and decreased cytotoxic T cell and NK cell function and decreased
platelet storage lesions that suggests that storage time macrophage activation.59 Such effects could explain the
alone may not be a satisfactory explanation for these clinical observations seen with allogeneic transfusion.
metabolic effects. Donor and prestorage variables may There are convincing data in animal models, clinical ob-
also be important.57,58 servational studies, and randomized controlled trials that

Table 17–10 The RBC Storage Lesion: Biochemical and Morphological Changes That Occur
With Increasing Storage Time
Biochemical Changes Morphological Changes
↓ Glucose Irreversible morphological changes ⇒ ↓ deformability
↓ ATP ⇒ ↑ hemoglobin O2 affinity Formation of microvesicles
↓ 2,3-DPG ⇒ ↑ hemoglobin O2 affinity ↑ Osmotic fragility
↑ pH ⇒ ↓ hemoglobin O2 affinity
↓ S-nitrosylation of Hb ⇒ hypoxic vasodilation
Na+/K+ fluxes ⇒ loss of intracellular K+ into the supernatant
↑ Intracellular Ca2+ ⇒ activation of proteases leading to eryptosis
↓ NO ⇒ hypoxic vasodilation

ATP = adenosine triphosphate; 2,3-DPG = 2,3 diphosphoglycerate; Hb = hemoglobin; NO = nitric oxide

Table 17–11 Recent Clinical Trials Studying the Clinical Effects of Prolonged RBC Storage
Age of Red Blood Cells in Premature Infants (ARIPI)54 No difference in fresh versus old RBCs in very-low-birth-weight infants

Red Cell Storage Duration Study (RECESS)55 No difference in fresh versus old RBCs in cardiac surgery patients

Age of Blood Evaluation (ABLE)56 No difference in fresh versus old RBCs in adult ICU patients
6888_Ch17_373-395 29/10/18 5:45 PM Page 389

Chapter 17 Adverse Effects of Blood Transfusion 389

support the presence of an immunomodulatory effect for involving bacterial culture is routinely performed in any
allogeneic transfusion. However, association does imply febrile transfusion reaction.62
causation. This is a complex phenomenon, likely involving TTBI should be considered in any patient who develops
multiple mechanisms, that are still not fully understood. fever, chills, rigors, or hypotension, especially associated with
Moreover, recognition of the deleterious immunomodula- platelet transfusion. The differential diagnosis includes AHTR
tory effects of transfusion has not yet translated into signif- and FNHTR. Many patients receiving platelet transfusions also
icant changes in transfusion practices. Recent randomized have low WBC counts and are immunocompromised because
controlled trials studying the effects on clinical outcomes of their underlying disease, which makes them more suscep-
of transfusion of RBCs with shorter storage periods have tible to infection. If hypotension or respiratory symptoms are
not shown any significant clinical differences compared to more prominent, TRALI and allergic anaphylactoid reactions
older stored products.53,55,56 These studies have cast doubt should be considered in the diagnosis. In short, a high index
on the clinical significance of the RBC storage lesion. of suspicion is necessary to take the appropriate steps to
culture the implicated product and direct the clinical service
Infectious Transfusion Reactions to observe the patient for infection. Table 17–12 is a list of
triggers that should prompt an investigation for TTBI.3
Transfusion-Transmitted Bacterial The microorganisms that are implicated in TTBI differ be-
Infections (TTBI) tween contaminated RBCs and platelets chiefly due to dif-
ferences in storage conditions. Since most bacteria grow
TTBI is the most frequent infection associated with transfu- poorly or not at all in RBCs under refrigerated conditions,
sion. Despite strict donor screening, stringent collection and only those bacteria that show optimal growth under cold
storage techniques, and bacterial detection methods, bacte- conditions are pathogenic.61 The predominant bacteria as-
rial contamination of blood products remain a significant sociated with RBC contamination are gram-negative rods in
risk of transfusion, especially for platelets. The exact number the Enterobacteriaceae family. All of these organisms are ca-
or proportion of contaminated products is unknown. Al- pable of producing endotoxin and septic shock with trans-
though the incidence of TTBI has greatly decreased over the fusion. These organisms are hypothesized to end up in the
past three decades, substantial evidence suggests that most blood donation due to either transient bacteremia or asymp-
cases are underrecognized or are not reported to the trans- tomatic gastrointestinal infection in the donor at the time
fusion service or blood bank.60 of donation. In contrast, most bacteria in contaminated
The NHSN Hemovigilance definition of TTBI requires platelets are due to gram-positive cocci and are from normal
laboratory evidence of the pathogen in the recipient and skin flora introduced into the product during venipunc-
demonstration of the pathogen in at least one or more of ture.61 Staphylococcus aureus accounts for the greatest num-
the following: the transfused component, the donor at the ber of deaths due to blood product contamination reported
time of collection, an additional component prepared from to the FDA from FY2011 to 2015, all of which were platelet
the same donation, or an additional recipient of a component products.2 While these bacteria are not viable in stored re-
from the same donation.3 A definitive diagnosis of TTBI also frigerated RBCs, they grow optimally at room temperature
requires that the recipient did not have evidence of infection platelet storage conditions. Severe and fatal TTBIs are most
with the same pathogen prior to the transfusion. often associated with contaminated platelets and include
The clinical presentation of TTBI ranges from subclinical gram-positive and gram-negative bacteria.61 Table 17–13 lists
infection to sepsis and death and also varies according to
the type of product implicated in the reaction.61 RBCs tend
to be associated with septic reactions due to contamination, Table 17–12 Investigation Triggers for
most often due to gram-negative bacteria that produce Potential Transfusion-
endotoxin during storage. The infusion of endotoxin, Transmitted Infection
rather than bacteremia itself, results in a massive release of
proinflammatory mediators that produce the reaction. Clin- 1. Identification of a viral, bacterial, fungal, or parasitic infection in a
recipient within the time of possible exposure from transfusion to
ical symptoms begin during transfusion of the contami- onset of infection.
nated product and include high fever (>38.5°C/101.5°F), 2. Any laboratory testing indicating viral, bacterial, fungal, or parasitic
rigors, hypotension, tachycardia, nausea, and vomiting. contamination of the recipient unit.
Pain (back, abdominal, or infusion site) and respiratory 3. Unexpected or unexplained clinical events following transfusion
complaints can also occur. Septic shock, acute kidney fail- that could be consistent potential transfusion-transmitted infection
TTI: encephalitis, meningitis, or other central nervous system
ure, and disseminated intravascular hemolysis are common abnormality; sepsis; hemolytic anemia with or without fever; death
complications.61 In contrast, TTBI associated with platelets 4. Any infection found in a recipient for pathogens routinely screened
may occur during transfusion or be delayed. Symptoms are for in donors occurring within 6 months after transfusion if the
due to bacteremia itself as gram-positive bacteria are more index donation was negative but a donor was subsequently found
common in contaminated platelets. Many TTBIs associated to have infection or the recipient had no evidence of infection prior
to transfusion.
with platelets cause only fever and chills with nothing
else and may be attributed to an FNHTR or the patient’s From: Centers for Disease Control and Prevention. National Healthcare Safety Network
underlying illness or treatment unless active surveillance Biovigilance Component hemovigilance surveillance module. 2017 Jan; v.2.4.:21.
6888_Ch17_373-395 29/10/18 5:45 PM Page 390

390 PART III Transfusion Practices

Table 17–13 Microorganisms Implicated approach to prevent not only bacterial contamination but
other microorganisms as well.65 The INTERCEPT system
in TTBI Typically Associated
(Cerus Corporation), a photochemical treatment process
With RBCs and Platelets
using a psoralen derivative, has been approved by the FDA
RBCS Platelets for use in the United States for treatment of plasma and
platelets.
Enterobacter cloacae Staphylococcus aureus
Escherichia coli Staphylococcus epidermidis
Klebsiella oxytoca Staphylococcus lugdunensis CASE STUDIES
Klebsiella pneumonia Case Study 17-1
Pseudomonas aeruginosa
A 47-year-old male with a history of intravenous drug
Serratia marcescens
abuse presented to the emergency department with a
From: Centers for Disease Control and Prevention. National Healthcare Safety Network 1-week history of back pain, fever, shaking chills, and red
Biovigilance Component hemovigilance surveillance module. 2017 Jan;v.2.4.:21. urine.
Blood Test Results (Initial):
the most commonly documented pathogens by type of im- Hgb: 7 g/L (reference range: 14–17.4 g/dL)
plicated blood product. Total Bilirubin: 1 mg/dL (reference range: 0.2–1.2 mg/dL)
Estimates of the incidence of bacterial contamination of Urinalysis: Blood: large; microscopy: many WBCs and
blood products and TTBI have improved with hemovigilance granular casts
programs. The French National Hemovigilance Network re- Blood Typing: B-positive
ported an estimated incidence of bacterial contamination Antibody Screen: Negative
from 2000 to 2008 of 2.45 per million transfused blood prod- Immediate Spin Crossmatching: Compatible
ucts, including 24.7 per million transfused platelet concen-
trates and 0.39 per million transfused RBCs. They calculated Upon medical evaluation, the patient was diagnosed
the incidence of severe or fatal TTBIs during this time of with acute pyelonephritis and anemia. He was promptly
13.4 per million transfused platelet concentrates and started on antibiotics. Shortly after admission, the patient
5.14 per million transfused RBCs.63 The British SHOT report was transfused with 2 units of packed red blood cells. The
for 2015 reported one definite case and one possible case patient’s clinical course was significant for persistent
of TTBI and estimated the annual rate of severe TTBI to be hypotension despite hydration, requiring the addition
1.2 per million transfused blood products.64 The U.S. FDA of blood pressure support medication. Due to persistent
reported 14 fatalities due to TTBI from FY2011 to FY2015, anemia, he received 5 additional units of packed red
which is calculated to be an annual incidence rate of 1.3 per blood cells. On day 3 after admission, a new sample was
million transfused blood products.2 A recent study confirms sent to the transfusion service with a request for 2 addi-
the impression that bacterial contamination and TTBI are tional units of packed red blood cells.
underrecognized and underreported.62 This study used an Transfusion Service Evaluation:
active surveillance system to identify bacterial contamination Day 3 Sample 1
of platelet units and TTBI that would have not been identi- Blood type: O-positive
fied by passive surveillance. The authors found bacterial con- Antibody Screen: Neg
tamination in 20 out of 51,440 transfused platelet units, a Hemolysis: Yes
rate of 389 per million transfused platelets, which resulted
in five TTBIs occurring 9–24 hours after transfusion, a rate The day-3 sample revealed a blood type discrepancy.
of 97.2 per million transfused platelets. These data are alarm- The patient’s medical staff was immediately notified, and
ing and strongly suggest not only that bacterial contamina- a day-3 second sample was requested. The laboratory tech-
tion of platelets is much more frequent than was previously nologist examined both pre- and post-samples for clerical
appreciated, but that many TTBIs go undetected or possibly errors. Upon initial review, no name discrepancy or other
are categorized as FNHTR or other types of reactions. identifying information was identified among the different
Recognition of the incidence of bacterial contamination, blood bank samples and transfusion records. Both day-3
particularly with platelets, has led to a focus on various pre- samples were noticed to have “orange” appearing plasma
vention strategies. Individuals with a recent asymptomatic when compared to the admission sample. Repeat testing
bacterial infection who might still be bacteremic yet qualify was performed on the pre- and post-transfusion samples
as a donor could be identified through different donor with the following results.
screening. Improved skin disinfection and initial aliquot di- Day 3 Sample 1 Day 3 Sample 2 Admission Sample:
version can reduce skin organism contamination. CGMP for Blood type: O-positive Blood type: O-positive Blood type: B-positive
blood component processing and storage targets the intro- Antibody Screen: Neg Antibody Screen: Neg Antibody Screen: Neg
duction of bacteria during this step. Recently, there is great Hemolysis: Yes Hemolysis: Yes Hemolysis: No
interest in pathogen-reduction technologies as a proactive
6888_Ch17_373-395 29/10/18 5:45 PM Page 391

Chapter 17 Adverse Effects of Blood Transfusion 391

ABO discrepancy and hemolysis were identified. failure to recognize transfusion reaction–related signs
Transfusion service records indicated that the patient and symptoms.
had been transfused with seven B-positive packed red
1. Define acute transfusion reactions.
blood cells. No discrepancies were identified.
2. What does the orange-appearing plasma or hemoglo-
Additional testing was performed with the following
binemia indicate?
results:
3. The severity of symptoms with acute hemolytic reac-
Day 3 Sample 2 tions are most likely related to what major factor?
DAT: Positive 4. What are the key clerical checks to be performed by
(Polyspecific reagent 3+, Anti-IgG 3+, and Anti- C3d 1+) the laboratory technologist?
Eluate: ABS cell I: Neg
ABS cell II: Neg Case Study 17-2
A cells: Neg
A 62-year-old female with spinal stenosis required back
B cells: 3+
surgery. During surgery, 2 units of allogenic red blood
Transfusion Service Physician Follow-Up cells and 500 mL of salvaged blood were transfused. Once
The transfusion service physician promptly notified the the patient was in the recovery room, the anesthesiologist
patient’s physician regarding the ABO incompatibility became concerned because her urine was “more pink”
and the likelihood of an acute hemolytic transfusion than usual. The anesthesiologist notified the transfusion
reaction. The patient was evaluated for ongoing signs service physician, and a transfusion reaction workup was
and symptoms of acute hemolysis and was found to be initiated. A postreaction sample was drawn, properly
unchanged from baseline with the exception of a tem- labeled, and sent to the transfusion service with a copy of
porary decrease in urine output that was treated with the transfusion record listing the reaction. The transfused
hydration. autologous salvaged units had been discarded and there-
Additional laboratory assays were performed and fore were not available for evaluation.
results are shown below: Transfusion Service Evaluation
LDH 813 (reference range: 100–190 IU/L) The laboratory technologist examined both pre- and
Total Bilirubin 3.7 (reference range: 0.2–1.2 mg/dL) postreaction samples for clerical errors. The historic
Direct Bilirubin 0.3 (reference range: 0.1–0.4 mg/dL) patient records were checked and verified. No defects
Haptoglobin Undetectable (reference range: 30–200 mg/dL) were revealed. Additional testing was performed with the
Urinalysis Blood 3 +; many WBC, RBC, granular and red cell casts following results:
During a retrospective review of the patient’s clinical Postreaction sample:
course, an increase in body temperature, a decrease in Blood type: O-positive; no discrepancies identified
blood pressure, an increase in heart rate and respiratory Antibody Screen: Neg
rate, and shaking chills and back pain were associated DAT: Neg
with each of the transfusion episodes. These were thought Hemolysis: No hemolysis
to be related to the patient’s underlying condition and AHG Crossmatch: Both units, previously electronic crossmatched,
therefore not recognized as transfusion-related signs and compatible
symptoms. A more detailed examination of the initial Repeat ABO confirmatory testing on allogenic unit segments: O-positive
sample sent to the transfusion service the day of admis- (both units)
sion revealed that the tube’s original label had a different Transfusion Service Physician Follow-Up
name and medical record number than the “typenex
The transfusion service physician notified the anesthesi-
label” placed on top. Transfusion records of the second
ologist that the transfusion reaction showed no discrep-
name were reviewed, showing that a type and antibody
ancies or evidence of hemolysis based on the evaluation
screen had been performed but no crossmatches were
of the 2 units of allogenic blood issued by the transfusion
requested. The emergency department initiated an inves-
service and transfused during surgery. The anesthesiolo-
tigation to find the cause of the major collection error, re-
gist contacted the hospital’s cell saver charge personnel to
vealing that the patients involved in the sample collection
determine if the cause of the hemoglobinuria could have
error were in the same room when samples were drawn
been caused by the improper use of the cell saver. It was
and that the samples were labeled at the nurses’ station
revealed that the proper washing solution (0.9% NaCl)
where addressographs are kept.
was used and there was no malfunction of the cell saver.
Upon follow-up, the patient was found to be improv-
However, the surgeon had requested the use of a more po-
ing and was discharged home 10 days after reaction.
tent suction because he stated that the one that came with
Interpretation the cell saver kit was too slow. It was determined that the
This case represents an acute immune hemolytic trans- hemoglobinuria was caused by the use of an inappropriate
fusion reaction due to a major collection error with suction during blood salvage, causing hemolysis of the
6888_Ch17_373-395 29/10/18 5:45 PM Page 392

392 PART III Transfusion Practices

harvested blood that was then transfused to the patient. DAT: Neg
The patient’s urine cleared overnight and the postopera- Hemolysis: No hemolysis
tive course was unremarkable.
Three hours later, the patient returned to the emer-
Interpretation gency department with an increase in temperature and a
This case represents a nonimmune acute hemolytic trans- decrease in blood pressure. The transfusion service physi-
fusion reaction due to failure to follow the manufacturer’s cian was consulted, and further testing of the unit of
guidelines for the cell saver’s use. Communication with blood was ordered. Immediately, a Gram stain and blood
other health professionals served as the key to resolving culture were performed on the unit of blood implicated
this incident. with the reaction, and two sets of blood cultures were
drawn from the patient.
1. What is the key to resolving this case?
2. What are the key reasons for nonimmune hemolytic Gram stain unit of blood: Gram-positive cocci in clusters
transfusion reactions? Transfusion Service Physician Follow-Up
Case Study 17-3 The transfusion service physician was immediately
notified of the Gram stain results. The results were re-
A 57-year-old female bone marrow transplant (BMT) layed to the patient’s physician, who started the patient
patient with non-Hodgkin’s lymphoma required transfu- on broad-spectrum antibiotic treatment until identifi-
sions on a regular basis due to her chronic anemia. During cation and sensitivity of the cultures were complete.
one of her transfusion visits to the clinic, the patient Shortly after, the blood center was notified of the trans-
developed a 1.8°C increase in temperature with rigors. fusion service physician’s preliminary findings, and all
The transfusion was stopped two-thirds of the way available donor products were discarded. The following
through. The nursing personnel performed a clerical day, culture results revealed positive growth in both the
check by verifying the patient identification band to the patient and the unit of blood, and the organism was
transfusion record and the unit crossmatch tag to confirm identified as Staphylococcus aureus. A follow-up written
that the blood component was given to the correct notification prepared by the transfusion service physi-
patient. The patient’s physician was notified. The nurse, cian was sent to the blood center for donor evaluation.
who had monitored the patient during past transfusions, Further investigation by the blood center revealed that
expressed her concern to the physician regarding the the donor arm was not cleaned per standard operating
severity of the reaction when compared to previous reac- procedures, which led to contamination of the unit. The
tions observed. The physician requested a transfusion blood center phlebotomist was retrained on proper
reaction workup. A properly labeled postreaction sample cleansing techniques.
was drawn and sent along with the unit of blood, attached Interpretation
tubing, and solutions to the transfusion service for a
This case represents a transfusion-associated sepsis
transfusion reaction workup. The patient was treated with
(TAS) due to a failure to follow standard operating pro-
Tylenol and discharged with instructions to continue
cedures for the arm preparation prior to donation, most
monitoring her temperature.
likely resulting in contamination of the unit from the skin
Transfusion Service Evaluation flora of the donor.
The laboratory technologist examined both pre- and
1. What laboratory workup is performed to diagnose
postreaction samples, the blood bag, and the tubing for
transfusion-associated sepsis?
clerical errors. The attached solution was verified to be
2. What symptoms usually present with transfusion-
0.9% normal saline, and historic patient records were
associated sepsis?
checked and verified. No defects were revealed. Repeat
testing was performed on the postreaction sample with
the following results:
Postreaction Sample:
Blood type: O-negative
6888_Ch17_373-395 29/10/18 5:45 PM Page 393

Chapter 17 Adverse Effects of Blood Transfusion 393

SUMMARY CHART
 Allergic transfusion reactions and febrile nonhemolytic transfusion of some mediator in the transfused product
transfusion reactions are the most common adverse that provokes the reaction.
reactions, but transfusion-related acute lung injury  Stored RBCs contain citrate, an anticoagulant that can
(TRALI) and transfusion-associated circulatory over- cause hypocalcemia and can leak potassium from the
load (TACO) are the most common transfusion reac- cells into the supernatant causing hyperkalemia, espe-
tions associated with mortality. cially in neonates. In addition, RBCs and platelets
 Hemovigilance describes a process to standardize the undergo a variety of metabolic and morphological
definition of different transfusion reactions and data changes called the “storage lesion,” but the clinical sig-
collection regarding the incidence of transfusion reac- nificance of these changes is still unknown.
tions in order to improve the safety of blood transfusion.  Transfusion-associated graft-versus-host disease is a
 Acute and delayed serologic transfusion reactions can very rare, but devastating adverse effect of transfusion
be distinguished by the time of onset in relation to with a mortality rate of 90%, in which donor lympho-
transfusion, predominant clinical symptoms, and dom- cytes attack the recipient’s immune system.
inant pathophysiological mechanisms.  Transfusion-transmitted bacterial infection (TTBI) is
 Transfusion reactions in which respiratory symptoms more often associated with platelet products than
predominate include TRALI, TACO, transfusion- RBCs, and each of these products is associated with
associated dyspnea, and severe allergic reactions. a different set of microorganisms. S. aureus and gram-
However, these reactions also are associated with a positive organisms are most associated with platelets,
variety of other signs and symptoms that overlap with while gram-negative rods of the Enterobacteriaceae
other types of reactions. family are found more commonly with contamination
 The pathophysiology of many types of reactions in- RBCs.
cludes “two hits”: a predisposition of the recipient and

5. Irradiation of blood is performed to prevent:


Review Questions
a. Febrile nonhemolytic transfusion reaction
1. What component is most frequently involved with b. Delayed hemolytic transfusion reaction
transfusion-associated sepsis? c. Transfusion-associated graft-versus-host disease
d. Transfusion-associated circulatory overload
a. Plasma
b. Packed red blood cells 6. The only presenting sign most often accompanying a
c. Platelets delayed hemolytic transfusion reaction is:
d. Whole blood a. Renal failure
2. Fatal transfusion reactions are mostly caused by: b. Unexplained decrease in hemoglobin
c. Active bleeding
a. Serologic errors
d. Hives
b. Improper storage of blood
c. Clerical errors 7. Which transfusion reaction presents with fever, macu-
d. Improper handling of the product lopapular rash, watery diarrhea, abnormal liver function,
and pancytopenia?
3. Early manifestation of an acute hemolytic transfusion
reaction can be confused with: a. Transfusion-associated sepsis
b. Transfusion-related acute lung injury
a. Allergic reaction
c. Transfusion-associated graft-versus-host disease
b. Febrile nonhemolytic reaction
d. Transfusion-associated allergic reaction
c. Anaphylactic shock
d. Sepsis 8. A suspected transfusion-related death must be reported to:
4. Pain at infusion site and hypotension are observed with a. AABB
what type of reaction? b. Federal and Drug Administration (FDA)
c. College of American Pathologists (CAP)
a. Delayed hemolytic transfusion reaction
d. The Joint Commission (TJC)
b. Acute hemolytic transfusion reaction
c. Allergic reaction
d. Febrile nonhemolytic reaction
6888_Ch17_373-395 29/10/18 5:45 PM Page 394

394 PART III Transfusion Practices

9. Nonimmune hemolysis can be caused during transfu- SafetyAvailability/ReportaProblem/TransfusionDonationFatalities/


sion by: UCM518148.pdf
3. Centers for Disease Control and Prevention [Internet].
a. Use of small bore size needle Atlanta (GA). CDC [2017 Jan]. National Healthcare Safety
b. Use of an infusion pump Network Biovigilance Component hemovigilance surveillance
c. Improper use of a blood warmer module, vol. 2.4. Available from: https://www.cdc.gov/nhsn/
d. All of the above pdfs/biovigilance/bv-hv-protocol-current.pdf
4. Mazzei CA, Popovsky MA, Kopko PM. Noninfectious compli-
10. Transfusion reactions are classified according to: cations of blood transfusion. In: Fung MK. Technical manual.
Bethesda (MD): AABB Press; 2014. p. 665-96.
a. Signs or symptoms presenting during or after
5. AABB. Standards for blood banks and transfusion services,
24 hours 31st ed. Bethesda, (MD): AABB; 2018.
b. Immune or nonimmune 6. Code of Federal Regulations. Adverse reaction file, 21 C.F.R.
c. Infectious or noninfectious Sect. 606.170 (2016).
d. All of the above 7. Serious Hazards of Transfusion [Internet]. Manchester: SHOT
Office; 2017 [cited 2017 Jan 25]. Available from: http://www
11. With febrile nonhemolytic transfusion reactions: .shotuk.org
8. International Haemovigilance Network [Internet]. Manchester:
a. They are self-limited International Haemovigilance Network Secretariat; 2010 [cited
b. Fever resolves within 2–3 hours 2017 Jan 25]. Available from: http://www.ihn-org.com
c. Treatment is required 9. AuBuchon JP, Fung M, Whitaker B, Malasky J. AABB validation
d. a and b are correct study of the CDC’s National Healthcare Safety Network
e. All of the above Hemovigilance Module adverse events definitions protocol.
Transfusion. 2014;54:2077-83.
12. Absolute IgA deficiency is a classic example of a severe 10. Körmöczi GF, Mayr WR. Responder individuality in red blood
allergic reaction. A result indicating an absolute IgA cell alloimmunization. Transfus Med Hemother. 2014;41:446-51.
11. Gehrie EA, Tormey CA. The influence of clinical and biological
deficiency is: factors on transfusion-associated non-ABO alloimmunization:
a. <0.05 mg/dL responders, hyper-responders, and non-responders. Transfus
b. <0.50 mg/dL Med Hemother. 2014;41:420-9.
c. <0.50 gm/dL 12. Chou ST, Jackson T, Vege S, Smith-Whitley K, Friedman DF,
Westhoff CM. High prevalence of red blood cell alloimmuniza-
d. <5 mg/dL
tion in sickle cell disease despite transfusion from Rh-matched
13. How are mild allergic transfusion reactions with isolated minority donors. Blood. 2013;122:1062-71.
13. Davenport RD. Hemolytic transfusion reactions. In:
symptoms or hives and urticaria treated? Popovsky MA, editor. Transfusion reactions. Bethesda (MD):
a. Transfusion is stopped and transfusion reaction AABB Press; 2012. p. 1-51.
workup is initiated 14. Karafin MS, Blagg L, Tobian AA, King KE, Ness PM, Savage WJ.
b. Transfusion is stopped and antihistamines admin- ABO antibody titers are not predictive of hemolytic reactions
due to plasma incompatible platelet transfusions. Transfusion.
istrated; when symptoms improve, transfusion is 2012;52:2087-93.
restarted 15. Zimring JC, Spitalnik, SL. Pathobiology of transfusion reac-
c. Stop transfusion and prepare washed red blood cells tions. Ann Rev Pathol Mech Dis. 2015;10:83-110.
d. Continue transfusion with a slower infusion rate 16. Hendrickson JE, Eisenbarth SC, Tormey CA. Red blood cell
alloimmunization: new findings at the bench and new recom-
14. TRALI presents with the following symptoms: mendations for the bedside. Curr Opin Hematol. 2016;23:
a. Respiratory distress 543-49.
17. Nickel RS, Hendrickson JE, Fasano RM, Meyer EK, Winkler
b. Severe hypoxemia and hypotension AM, Yee MM, et al. Impact of red blood cell alloimmunization
c. Fever on sickle cell disease mortality: a case series.Transfusion. 2016;
d. All of the above 56:107-14.
18. Habibi A, Mekonto-Dessap A, Guillaud C, Michel M, Razazi K,
15. Which of the following is characteristic of iron overload? Khellaf M, et al. Am J Hematol. 2016;91:989-94.
a. Delayed, nonimmune complication occurs 19. Santos B, Portugal R, Nogueira C, Loureiro M. Hyperhemolysis
b. Chelating agents are used syndrome in patients with sickle cell anemia: a report of three
cases. Transfusion. 2015;55;1394-98.
c. Multiorgan damage may occur 20. Danaee A, Inusa B, Howard J, Robinson S. Hyperhemolysis in
d. All of the above patients with hemoglobinopathies: a single-center experience
and review of the literature. Transfus Med Rev. 2015;29:220-30.
References 21. Vlaar AP, Juffermans NP. Transfusion-related acute lung
injury—a clinical review. Lancet. 2013;382:984-94.
1. Harvey AR, Basavaraju SV, Chung KW, Kuehnert MJ. Transfusion- 22. Clifford L, Jia Q, Subramanian A, Yadav H, Wilson GA,
related adverse reactions reported to the National Healthcare Murphy SP, et al. Characterizing the epidemiology of postop-
Safety Network Hemovigilance Module, United States, 2010 to erative transfusion-related acute lung injury. Anesthesiology.
2012. Transfusion. 2015;55:709-18. 2015;122:12-20.
2. Food and Drug Administration, CBER [Internet]. Rockville 23. AABB. Association Bulletin no. 06-07. Transfusion-related
(MD): FDA. Fatalities reported to FDA following blood trans- acute lung injury. 2006 Nov 3.
fusion and collection annual summary for FY2015. Available 24. AABB. Association Bulletin no. 14-02. TRALI risk mitigation strat-
from: http://www.fda.gov/downloads/BiologicsBloodVaccines/ egy for plasma and whole blood for allogeneic transfusion. 2014.
6888_Ch17_373-395 29/10/18 5:45 PM Page 395

Chapter 17 Adverse Effects of Blood Transfusion 395

25. Peters AL, Van Stein D, Vlaar APJ. Antibody-mediated transfusion- 46. Jawa RS, Young DH, Stothart JC, Kulaylat MN, Landmark JD.
related acute lung injury; from discovery to prevention. Br J Transfusion-associated graft versus host disease in the
Haematol. 2015;270:597-14. immunocompetent patient: an ongoing problem. J Intensive
26. Peters AL, Van Hezel ME, Juffermans NP, Vlaar APJ. Pathogen- Care Med. 2015;30:123-30.
esis of non-antibody mediated transfusion-related acute lung 47. Pritchard AE, Shaz BH. Survey of irradiation practice for the
injury from bench to bedside. Blood Rev. 2015;29:51-61. prevention of transfusion-associated graft-versus-host disease.
27. Murphy EL, Kwaan N, Looney MR, Gajic O, Hubmayr RD, Arch Pathol Lab Med. 2016;140:1092-97.
Gropper MA, et al. Risk factors and outcomes in transfusion- 48. McFarland JG. Posttransfusion purpura. In: Popovsky MA,
associated circulatory overload. Am J Med. 2013;357:e29-38. editor. Transfusion reactions. Bethesda (MD): AABB Press;
28. Roubinian NH, Hendrickson JE, Triulzi DJ, Gottschall JL, 2012. p. 263-87.
Chowdhury D, Kor DJ, et al. Incidence and clinical character- 49. Hayashi T, Hirayama F. Advances in alloimmune thrombocy-
istics of transfusion-associated circulatory overload using an topenia: perspectives on current concepts of human platelet
active surveillance algorithm. Vox Sang. 2017;112:56-63. antigens, antibody detection strategies, and genotyping. Blood
29. Lucero H, Bolton-Maggs P. Transfusion associated circulatory over- Transfus. 2015;13:380-90.
load. In: Bolton-Maggs P, editor. Serious Hazards of Transfusion 50. D’Alessandro A, Kriebardis AG, Rinalducci S, Antonelou MH,
(SHOT) Steering Group. The 2014 Annual SHOT Report (2015). Hansen KC, Papassideri IS, et al. An update on red blood cell
Manchester Serious Hazards of Transfusion; 2015. p. 132-9. storage lesions, as gleaned through biochemistry and omics
30. Clifford L, Jia Q, Subramanian A, Yadav H, Schroeder DR, technologies. Transfusion. 2015;55:205-19.
Kor DJ. Risk factors and clinical outcomes associated with 51. Zimring JC. Established and theoretical factors to consider in
perioperative transfusion-associated circulatory overload. assessing the red cell storage lesion. Blood. 2015;125:2185-90.
Anesthesiology. 2017 126;409-18. 52. Rinalducci S, Zolla L. Biochemistry of storage lesions of red
31. Andrzejewski C, Casey MA, Popovsky MA. How we view and cell and platelet concentrates: a continuous fight implying
approach transfusion-associated circulatory overload: patho- oxidative/nitrosative/phosphorylative stress and signaling.
genesis, diagnosis, management, mitigation, and prevention. Transfus Apher Sci. 2015;52:262-9.
Transfusion. 2013;53:3037-47. 53. Steiner ME, Ness PM, Assmann SF, Triulzi DJ, Sloan SR,
32. Badami KG, Joliffe E, Stephens M. Transfusion-associated Delaney M, et al. Effects of red-cell storage duration on patients
dyspnea—shadow or substance? Vox Sang. 2015;109:197-200. undergoing cardiac surgery. N Engl J Med. 2015;372:1419-29.
33. Hume HA, Robillard P. Hypotensive transfusion reactions. In: 54. Fergusson DA, Hébert P, Hogan DL, LeBel L, Rouvinez-Bouali N,
Popovsky MA, editor. Transfusion reactions. Bethesda (MD): Smyth JA, et al. Effect of red blood cell transfusions on clinical
AABB Press; 2012. p. 239-61. outcomes in premature, very low birth weight infants: the ARIPI
34. Metcalf RA, Bakhtary S, Goodnough LT, Andrews J. Clinical randomized trial. JAMA. 2012;308:1443-51.
pattern in hypotensive transfusion reactions. Anesth Analg. 55. Lacroix J, Hébert PC, Fergusson DA, Tinmouth A, Cook DJ,
2016;123:268-73. Marshall JC, et al. Age of transfused blood in critically ill
35. Pagano MB, Ness PM, Chajewski OS, King KE, Wu Y, Tobian adults. N Engl J Med. 2015;372:1410-18.
AAR. Hypotensive transfusion reactions in the era of prestorage 56. Martí-Carvajal AJ, Simancas-Racines D, Peña-González BS.
leukoreduction. Transfusion. 2015;55:1668-74. Prolonged storage of packed red blood cells for blood transfu-
36. Heddle NM, Webert KE. Febrile nonhemolytic transfusion sion. Cochrane Database Syst Rev. 2015;(7):CD009330.
reactions. In: Popovsky MA, editor. Transfusion reactions. 57. Antonelou MH, Seghatchian J. Insights into the red blood cell
Bethesda (MD): AABB Press; 2012. p. 53-97. storage lesion: toward a new appreciation. Transfus Apher Sci.
37. Garraud O, Tariket S, Sut C, Haddad A, Aloui C, Chakroun T, 2016;55:292-301.
et al. Transfusion as an inflammation hit: knowns and un- 58. Nemkov T, Hansen KC, Dumont LJ, D’Alessandro A.
knowns. Front Immunol. 2016;7:1-10. Metabolomics in transfusion medicine. Transfusion. 2016;
38. Menis M, Forshee RA, Anderson SA, McKean S, Gondalia R, 56:980-993.
Warnock R, et al. Febrile non-hemolytic transfusion reactions 59. Refaai MA, Blumberg N. Transfusion immunomodulation from
occurrence and potential risk factors among the U.S. elderly a clinical perspective: an update. Expert Rev Hematol.
transfused in the inpatient setting, as recorded in Medicare 2013;6:653-63.
databases during 2011–2012. Vox Sang. 2015;108:251-61. 60. Benjamin RJ. Transfusion-related sepsis: a silent epidemic.
39. Vamvakas EC. Allergic and anaphylactic reactions. In: Blood. 2016;127:380-81.
Popovsky MA, editor. Transfusion reactions. Bethesda (MD): 61. Ramírez-Arcos S, Goldman M. Bacterial contamination. In:
AABB Press; 2012. p. 99-147. Popovsky MA, editor. Transfusion reactions. Bethesda (MD):
40. Savage WJ, Tobian AAR, Savage JH, Wood RA, Schroeder JT, AABB Press; 2012. p. 153-189.
Ness PM. Scratching the surface of allergic transfusion reac- 62. Hong H, Xiao W, Lazarus HM, Good CE, Maitta RW, Jacobs
tions. Transfusion. 2013;53:1361-71. MR. Detection of septic transfusion reactions to platelet trans-
41. Hirayama F. Current understanding of allergic transfusion re- fusions by active and passive surveillance. Blood. 2016;
actions: incidence, pathogenesis, laboratory tests, prevention 127:496-502.
and treatment. Br J Haematol. 2013;160:434-44. 63. Lafeuillade B, Ounnoughene N, Petermann R, Daurat G,
42. Callum J, Nahirniak S. Adverse effects of human-derived Huyghe G, Vo Mai MP, et al. Residual risk and retrospective
plasma derivatives. In: Popovsky MA, editor. Transfusion reac- analysis of transfusion-transmitted bacterial infection reported
tions. Bethesda (MD): AABB Press; 2012. p. 519-49. by the French National Hemovigilance Network from 2000 to
43. Jacobson CA, Anderson KC, Alyea EP. Transfusion-associated 2008. Transfusion. 2015;55:636-46.
graft-vs-host disease. In: Popovsky MA, editor. Transfusion 64. Bolton-Maggs P. Headlines: death, major morbidity, and ABO-
reactions. Bethesda (MD): AABB Press; 2012. p. 217-37. incompatible transfusions. In: Bolton-Maggs P, editor. Serious
44. Kopolovic I, Ostro J, Tsubota H, Lin Y, Cserti-Gazdewich CM, Hazards of Transfusion (SHOT) Steering Group. The 2014
Hessner HA, et al. A systematic review of transfusion-associated Annual SHOT Report (2015). SHOT Organisation. UK Blood
graft-versus-host disease. Blood. 2015;126:406-14. Services, Royal College of Pathologists Manchester, England;
45. Uchida S, Tadokoro K, Takahashi M, Yahagi H, Satake M, Juji 2015. p. 9-15.
T. Analysis of 66 patients definitive with transfusion-associated 65. Devine DV, Schubert P. Pathogen inactivation technologies—
graft-versus-host disease and the effect of universal irradiation the advent of pathogen-reduced blood components to reduce
of blood. Transfus Med. 2013;23:416-22. blood safety risk. Hematol Oncol Clin N Am. 2016;30:609-17.
6888_Ch18_396-416 29/10/18 5:40 PM Page 396

Chapter 18
Apheresis
Beth A. Hartwell, MD, MT(ASCP), SBB and Mark D. Pool, MD

Introduction Therapeutic Procedures Adverse Effects


History and Development General Considerations Citrate Toxicity
Physiology of Apheresis Plasmapheresis (Plasma Vascular Access
Anticoagulation Exchange) Vasovagal Reactions
Fluid Shifts Plateletpheresis Miscellaneous Reactions
Cellular Loss Leukapheresis Plasma Protein Interactions
Methodology Erythrocytapheresis (Red Blood Cell Fatalities
Methods of Centrifugation Exchange) Case Study
Membrane Filtration Fluid Replacement Case 18-1
Component Collection Special Procedures Summary Chart
Red Blood Cells Hematopoietic Progenitor Cells Review Questions
Plasma Immunoadsorption/Selective References
Absorption
Platelets
Photopheresis
Granulocytes

OBJECTIVES
1. Define apheresis and describe the physiology of the process.
2. Define leukapheresis, plateletpheresis, plasmapheresis, and erythrocytapheresis.
3. Compare and contrast the procedures of continuous flow centrifugation and intermittent flow centrifugation.
4. Describe the use of membrane technology for the collection of plasma.
5. List the components that can be collected using apheresis technology.
6. State the regulatory requirements for apheresis donations, including the frequency of donation.
7. Explain the rationale for the basic types of therapeutic apheresis.
8. List the indications for therapeutic apheresis, differentiating between conditions requiring plasma exchange and those
requiring cytapheresis.
9. List the different types of adsorbents and their clinical application.
10. Identify the factors that can be removed by plasmapheresis.
11. Describe the use of cytapheresis to collect hematopoietic progenitor (stem) cells.
12. Identify the possible adverse effects of apheresis.

Introduction the individual’s circulation. Through the use of sophisticated


automation, an apheresis procedure can be performed on
Apheresis (plural aphereses; from the ancient Greek aphairesis, either a blood donor or a patient. It represents a significant
“a taking away”) is a procedure in which whole blood is advance in blood component collection and the treatment of
removed from the body and passed through an apparatus specific disease states. Rather than collecting only a single
that separates out one (or more) particular blood con- unit of whole blood from a donor, apheresis allows a larger
stituent. It then returns the remainder of the constituents to volume of specific components to be collected, such as

396
6888_Ch18_396-416 29/10/18 5:40 PM Page 397

Chapter 18 Apheresis 397

platelets or red blood cells. This increases the ability to pro- Table 18–1 Types of Apheresis Procedures
duce the optimal components for patients and prevents
and Their Application
wastage.
The use of apheresis on a therapeutic basis permits the Component
removal of disease-causing or unwanted cellular or plasma Procedure Removed Application
constituents from a patient. Apheresis is also used to harvest Donor Patient
hematopoietic stem cells from the peripheral blood of donors
and patients, avoiding the need for extraction from the bone Plasmapheresis Plasma 4 4
marrow. Apheresis technology continues to evolve, and the Plateletpheresis Platelets 4 4
procedure is now commonplace in blood donor centers and
many hospitals and acute care settings.1 Leukapheresis White blood cells 4* 4**
(WBC)
History and Development Erythrocytapheresis Red blood cells (RBC) 4 4

Early developments in apheresis began some 60 years ago HPC apheresis Hematopoietic 4 4
when Harvard biochemist Dr. Edwin J. Cohn devised a large- progenitor cells
(HPC)***
scale method, based on a simple dairy centrifuge (the Cohn
centrifuge), for purifying albumin from pooled human * Typically granulocytes are the primary component collected.
plasma. This procedure, along with pasteurization, provided ** Removal of granulocytes and/or lymphocytes.
a safer therapeutic agent for resuscitating wounded soldiers *** Also called peripheral blood stem cells (PBSC).

than did using lyophilized pooled plasma, which had an


alarming risk of hepatitis transmission. The separation of blood components is based on the spe-
The device Dr. Cohn invented was ultimately not prac- cific gravity or weight of each individual blood constituent
tical for its intended purpose; however, later modifications (Fig. 18–1). If a tube of anticoagulated blood is centrifuged,
led to its use in deglycerolizing frozen red blood cells. In distinct layers develop, with the heavier red blood cells
the 1960s, A. Solomon and J.L. Fahey used centrifugation (RBCs) at the bottom and the lighter plasma portion at the
technology to separate whole blood into plasma and red top. Between these two prominent layers is a smaller layer
blood cells to perform the first reported therapeutic composed of white blood cells (WBCs) and platelets (the
plasmapheresis procedure. In 1965, collaboration between buffy coat/layer). If a pipette is placed at the appropriate
Emil J. Freireich, a physician with the National Cancer level in the test tube, any of these components can be
Institute, and George Judson, a research engineer with removed.
the IBM Corporation, led to the development of the first Apheresis technology utilizes this same principle for
continuous flow apheresis machine. In the same decade, separating blood components. Blood is removed from an in-
plasmapheresis was introduced as a means of collecting dividual (usually with a large-bore needle), mixed with an
donor plasma for fractionation. As we entered the 1970s, anticoagulant, and transported directly to the separation de-
apheresis was used to extract one cellular component and vice (typically a machine with a centrifuge bowl or belt).
return the remainder of the blood to the donor. In 1978, There it is separated into specific components. Once the
the membrane plasma separator was introduced as a components have been separated, any component can be
method for performing therapeutic plasma exchange. Since withdrawn. The remaining portions of the blood are then
these early pioneering processes were developed, many dif- mixed and returned to the donor or patient (Fig. 18–2).
ferent companies have developed apheresis technology
based on the same principles.2
Although the technology is much more advanced than in
the early years of development, an apheresis procedure per-
formed today still consists of withdrawing a small volume of
whole blood from a donor or patient and separating it into
components. One (or more) of the components is collected
and retained, and the remaining components are recombined
and returned to the individual. Apheresis can be performed
Water (91.5%)
on a donor to collect a specific blood component (donor Plasma Proteins (7%)
Other Solutes (1.5%)
apheresis), or it can be performed on a patient to remove a
particular blood component for therapeutic purposes (ther- "Buffy Coat" Lymphocytes Basophils
White Blood Cells Granulocytes Neutrophils
apeutic apheresis).3 The process of removing plasma is Platelets Monocytes Eosinophils
termed plasmapheresis. In a similar manner, platelets
(plateletpheresis), red blood cells (erythrocytapheresis), or Red Blood Cells
leukocytes (leukapheresis) can be removed (or collected)
using apheresis technology. Table 18–1 outlines the types of
apheresis procedures and their applications. Figure 18–1. Sedimented blood sample.
6888_Ch18_396-416 29/10/18 5:40 PM Page 398

398 PART III Transfusion Practices

Remaining intravascular volume. The sympathetic nervous system


Anticoagulant blood components attempts to compensate for the hypovolemia by increasing
added recombined and returned
Plasma cardiac output; this results in an increase in heart rate.
Hypovolemic reactions are not common among apheresis
Platelets donors, as regulatory restrictions limit the extracorporeal
Whole blood Lymphocytes Whole blood
volume to 10.5 mL/kg. Most modern instruments have
(vein) (vein) an extracorporeal volume well below this, except in the
Granulocytes
smallest of donors.
Erythrocytes
Hypotension may also occur during an apheresis procedure
due to a vasovagal reaction, particularly in apheresis donors.6
Blood components separated
by centrifugation and During a vasovagal reaction, the parasympathetic nervous sys-
selectively removed tem overrides the output of the sympathetic nervous system,
Figure 18–2. Principles of apheresis.
resulting in hypotension and a slowing of the heart rate. Fac-
tors that have been associated with vasovagal reactions in
apheresis donors include younger age and the sex of the donor.
Physiology of Apheresis Teenaged donors have higher vasovagal reaction rates com-
Apheresis collection of a blood component, whether for ther- pared with adults, and female donors have a higher incidence
apeutic or nontherapeutic purposes, is obviously different of these reactions than male apheresis donors.7 However, vaso-
from the routine collection of a unit of whole blood. Since vagal reaction rates are significantly lower in apheresis donors
apheresis involves removing whole blood from an individual, when compared with whole blood donors.
manipulating the removed blood, and subsequently reinfus-
ing portions of the blood, it is not surprising that the human Cellular Loss
body can be impacted in varying ways.
For both donor and patient apheresis procedures, the goal is
to intentionally remove a specific cellular component. This
Anticoagulation
would be expected to decrease circulating levels of the spe-
Citrate is used as the primary anticoagulant in apheresis pro- cific cells; however, other cellular components may be
cedures. The binding of calcium ions by citrate inhibits the affected as well. Accordingly, the FDA and the AABB have
calcium-dependent coagulation cascade; if calcium ions are requirements regulating how much time must elapse be-
not available, coagulation cannot proceed to its normal end- tween donor apheresis procedures, depending on the partic-
point. Citrate is mixed with the blood immediately as it is ular blood component being collected.
removed from the donor’s (or patient’s) vein, and effectively A platelet donor typically experiences an acute fall in
anticoagulates the blood before it enters the apheresis platelet count of 20% to 29% following apheresis donation.
machine. In normal situations, the infused citrate is not only Among females, this decrease tends to be greater. It is inter-
actively metabolized by the liver, kidneys, and muscles, but esting that the fall in platelet count does not correlate with
it is also diluted throughout the intra- and extracellular fluids the yield of the plateletpheresis procedure, as more platelets
of the body. As the citrate-calcium complex is metabolized, are collected than anticipated due to the mobilization of
the previously bound calcium ions are released back into the platelets from the spleen during apheresis collection. No
bloodstream. In addition, parathyroid hormone (PTH) is re- adverse effects of the transient decrease in platelet counts
leased in response to the decreased ionized calcium levels. have been demonstrated, and platelet counts normalize
This results in mobilization of calcium from bone,4 increased within a few days of donation.
intestinal absorption of calcium, and increased reabsorption In granulocyte collections, in addition to WBCs, platelets
of calcium by the kidneys, thereby helping to maintain ade- and significant numbers of RBCs are present in the leuka-
quate circulating calcium levels. Despite these compensatory pheresis product. Typically, a drop in hematocrit of 3% and
mechanisms, in some individuals the decrease in ionized a fall in platelet count of 22% occur after each granulocyte
calcium levels can result in symptomatic transient hypocal- donation. This fall is due to the loss of these cells in the prod-
cemia.5 This is described in more detail in the “Adverse uct and the dilutional effects of volume expansion caused by
Effects” section at the end of this chapter. the sedimenting agent, hydroxyethyl starch (HES), used dur-
ing the procedure.
Fluid Shifts Varying numbers of RBCs are lost during each donor
apheresis procedure, depending on the equipment used for
During an apheresis procedure, changes in intravascular collection and the specific component being collected. Each
volume occur secondary to removal of blood into the ex- collection facility must have procedures in place to ensure
tracorporeal circuit. During donor apheresis procedures, that the cumulative annual red blood cell loss for apheresis
the total volume of components collected may be more donors is not exceeded. This must take into account all red
than the simple whole blood donation. If additional fluid blood cell losses, including those associated with the actual
is not infused during the apheresis procedure, the donor donation of whole blood, RBCs, and other apheresis compo-
may experience hypotension due to the depletion of nents, as well as losses due to sample collection.
6888_Ch18_396-416 29/10/18 5:41 PM Page 399

Chapter 18 Apheresis 399

Methodology Inlet Port


(Whole Blood)
Outlet Port
Apheresis is performed using automated technology, and Plasma to External Bag
separation is typically performed by centrifugation. Aphere-
sis instruments in use today have a computerized control
panel, allowing the operator to select the desired component Red Cell/Buffy Coat
to be collected or removed. On-board optical sensors detect Interface
specific plasma-cell or cell-cell interfaces and divert the Red Cell
specific component to a collection bag. Currently available Mass
machines use disposable equipment, which includes sterile
single-use tubing sets, bags, and collection chambers unique
to the machine. The donor or patient remains attached to
the apheresis instrument for the duration of the procedure;
the amount of time for a particular procedure can range from
45 to 120 minutes. Collection of hematopoietic progenitor
cells takes considerably longer.
Depending on the goal of the individual apheresis proce-
dure (e.g., collecting platelets or removing plasma or RBCs),
the appropriate apheresis instrument must be selected.8 Each Figure 18–3. Cross-section of Haemonetics Centrifuge Bowl (IFC procedure).
commercially available instrument has specific performance (Courtesy of Haemonetics Corporation, Braintree, MA.)
characteristics. Some instruments are suitable only for donor
apheresis; others can be used for donor or therapeutic aphere- reinfused through the same needle). This is advantageous
sis. By manipulating certain variables on an apheresis instru- when collecting apheresis products from blood donors. If
ment, the operator can harvest plasma, platelets, WBCs, or both arms are used (double-needle procedure: one for phle-
RBCs for commercial or therapeutic purposes. The variables botomy and one for reinfusion), the amount of time for the
that are considered during an apheresis procedure include: apheresis can be reduced. The currently available systems
are versatile, portable, fully automated, and capable of
• Centrifuge speed and diameter efficient component collections (Fig. 18–4).
• Duration of dwell time of the blood in the centrifuge
• Type of solutions added, such as anticoagulants or sedi-
menting agents
• Cellular content or plasma volume of the patient or donor

Methods of Centrifugation

The most commonly used instruments employ one of two


methods of centrifugation: intermittent flow centrifugation
(IFC) and continuous flow centrifugation (CFC).

Intermittent Flow Centrifugation


In an IFC procedure, blood is processed in batches or cycles,
hence the term intermittent. Whole blood is drawn from an in-
dividual with the assistance of a pump. To keep the blood from
clotting, an anticoagulant is mixed with the blood as it is
pumped into a centrifuge bowl or chamber through the inlet
port. The bowl/chamber rotates at a fixed speed, separating the
components according to their specific gravities. A rotary seal
is used, resulting in a closed system. The RBCs, which have
greater mass, are packed against the outer rim of the bowl/
chamber, followed by the WBCs, platelets, and plasma. Once
separated, the pump is reversed and the desired component(s)
are pumped through the outlet port into a collection bag
(Fig. 18–3). The undesired components are pumped into a re-
infusion bag and returned to the individual, constituting one
cycle (or pass). The cycles are repeated until the desired quan-
tity of product is obtained (e.g., a plateletpheresis procedure
usually takes six to eight cycles to collect a therapeutic dose).
The IFC procedure can be performed as a single-needle Figure 18–4. The Haemonetics MCS Plus LN9000. (Courtesy of Haemonetics
procedure with only one venipuncture (blood is drawn and Corporation, Braintree, MA.)
6888_Ch18_396-416 29/10/18 5:41 PM Page 400

400 PART III Transfusion Practices

Continuous Flow Centrifugation


In a continuous flow centrifugation (CFC) procedure, the
processes of blood withdrawal, processing, and reinfusion are
performed simultaneously in an ongoing manner. This is in
contrast to IFC procedures, which complete a cycle before
beginning the next one. Because blood is drawn and returned
continuously during a procedure, two venipuncture sites are
necessary. Alternatively, especially with therapeutic proce-
dures, a dual-lumen central venous catheter can be used.
Blood is drawn from the phlebotomy site with the assistance
of a pump, mixed with anticoagulant, and collected in a spe-
cially designed chamber or belt, depending on the instrument.
Separation of the components is performed by centrifugation,
and the specific component is diverted and retained in a
collection bag. The remainder of the blood is reinfused to the
individual via the second venipuncture site.
Examples of machines employing this concept are the
Baxter/Fenwal CS-3000 Plus and the Amicus (Fig. 18–5), the
CaridianBCT COBE Spectra (Fig. 18–6) and Spectra Optia
(Fig. 18–7), and the Fresenius AS-104 (Fig. 18–8).

Figure 18–5. Baxter/Fenwal Amicus. (Courtesy of Baxter Healthcare Corpora-


tion, Deerfield, IL.)

A B

Figure 18–6. (A) The COBE Spectra apheresis system. (B) The separation chamber uses a unique asymmetric design to minimize contamination from RBCs and WBCs.
(Courtesy of CaridianBCT, Lakewood, CO.)
6888_Ch18_396-416 29/10/18 5:41 PM Page 401

Chapter 18 Apheresis 401

Figure 18–7. The Spectra Optia. (Courtesy of CaridianBCT, Lakewood, CO.)

The IFC and CFC machines have individual advantages


and disadvantages. The IFC equipment is usually smaller Figure 18–8. The Fresenius AS-104. (Courtesy of Fresenius USA, Walnut
Creek, CA.)
and more mobile, lending itself for use on mobile donor
collections. A single venipuncture may be used with the IFC
procedures, whereas two venipunctures are usually required (e.g., each of these apheresis machines can collect 2 units of
with the CFC procedures. However, new protocols have been RBCs; the Trima can also collect several different combinations
developed to allow some CFC machines to operate using of RBCs, plasma, or platelets during a single apheresis proce-
single-needle access (e.g., the Amicus and Spectra). Based dure). Another important improvement allows the cellular
on AABB standards, the extracorporeal blood volume (the products to be leukoreduced at the time of collection.
amount of blood out of the individual in the centrifuge
bowl/chamber and tubing) for blood donors should not Membrane Filtration
exceed 10.5 mL per kilogram of body weight at any time
during the procedure.9 The extracorporeal volume is usually
greater with the IFC than with the CFC machines. This can Advanced Concepts
be an important consideration in individuals with small Membrane filtration technology can also be used to sepa-
blood volumes, such as children and elderly individuals, rate blood components.10 Membrane separators are typi-
since the additional volume removed may leave the patient cally composed of bundles of hollow fibers or flat plate
hypovolemic during the procedure. membranes with specific pore sizes. As whole blood flows
Over the last several years, improvements in apheresis tech- over the fibers or membrane, plasma passes through the
nology have allowed IFC and CFC processes to be incorpo- pores and is collected, while the remainder of the cellular
rated into a single platform. Examples of this platform are the components is returned to the donor. This technology
Trima from CaridianBCT (Fig. 18–9) and the Baxter/Fenwal lends itself well to the collection of plasma, since pores can
Alyx. The Trima uses intermittent flow technology to draw be sized to prevent the passage of even small cellular ele-
and reinfuse blood to the donor, while the centrifuge operates ments. Filtration has several advantages over centrifuga-
with continuous flow. Improvements in both hardware tion, including the collection of a cell-free product and the
and software allow the collection of concurrent components
6888_Ch18_396-416 29/10/18 5:41 PM Page 402

402 PART III Transfusion Practices

Table 18–2 Donation Frequency


Apheresis Component Frequency of
Collected Donation*
2RBC 16 weeks

Plasma (frequent) Every 2 days (no more than


two times in 7 days)

Plasma (infrequent) Every 4 weeks (no more than


13 times/year)

Platelets, single apheresis unit Every 2 days (no more than


2 times in 7 days; no more
than 24 times in 12 months)

Platelets, double or triple Every 7 days


apheresis unit

Granulocytes Every 2 days

2RBC = double unit of red blood cells


* Donation frequency will vary, based on red blood cell loss and if more than one type
of component is collected concurrently. Donation frequency may be often if for a
specific medical reason and if it is approved by the blood bank medical director.

cumulative annual red blood cell loss does not exceed the
maximum permitted by regulations.
A qualified, licensed physician must be responsible for all
aspects of the apheresis program. Good equipment and a
well-trained, motivated staff are essential. Operators of
apheresis instruments may be medical technologists (clinical
Figure 18–9. The Trima Accel Automated Collection System. (Courtesy laboratory scientists), nurses, or technicians trained on the
of CaridianBCT, Lakewood, CO.)
job and should be friendly and outgoing. Because the aphere-
sis procedure is lengthier than whole blood donation, com-
plications may be avoided by the operator’s ability to relieve
ability to selectively remove specific plasma proteins by
the first-time donor’s boredom and anxiety. Although serious
varying the pore size. The Fenwal Autopheresis-C instru-
complications are unusual, it is essential to have another
ment combines centrifugation and membrane filtration
qualified individual immediately available to assist in case of
technology through the use of a small rotating cylindrical
emergencies.13
filter. Like other filtration technology, it is used only for
Written, informed consent must be obtained from the
plasma collection.
donor. The apheresis procedure, the testing to be performed,
and the possible risks and benefits must be explained in un-
Component Collection derstandable language.14 Collection of an apheresis blood
component does not provide specific medical benefit to
In component collection by apheresis, a healthy donor the donor; however, there may be a psychological benefit,
undergoes an automated procedure to obtain a specific blood particularly if the donor is of a rare type or is donating for a
component that will be transfused to a patient.11 In general, particular patient need. The apheresis procedure itself may
most of the requirements for whole blood donation must be more comfortable for the donor since the needle used for
be met; however, apheresis donors must meet additional venous access is typically of a larger gauge (i.e., smaller size)
requirements established by the AABB Standards for Blood than for whole blood collection. In addition, the infusion of
Banks and Transfusion Services9 and the FDA Code of Federal saline during the procedure helps reduce donor reactions
Regulations.12 Exceptions to the apheresis donation criteria due to hypovolemia. There are special risks associated with
and donation frequency may be approved by the facility apheresis donation, including side effects of the anticoagu-
medical director. lant, hypovolemia, and fainting. The FDA has provided guid-
Collection of each apheresis blood component carries ance on statements of risk that must be provided to donors
with it a different deferral period; a brief synopsis is provided of platelets, plasma, and red blood cells.15 The donor must
in Table 18–2. For example, if RBCs are not one of the com- be given the opportunity to accept or reject the apheresis
ponents collected, apheresis collections can be done more procedure.
often than would be possible for whole blood donation. There are a variety of apheresis instruments on the market
However, the RBC loss for each apheresis procedure must that can be used for component collection. Examples of in-
be closely monitored and tracked to ensure that a donor’s struments used for donor apheresis, along with the specific
6888_Ch18_396-416 29/10/18 5:41 PM Page 403

Chapter 18 Apheresis 403

blood components that can be collected with each, are listed RBC apheresis procedure is well tolerated by donors, and
in Table 18–3. Several of these instruments provide integral several collection facilities have noted a decreased incidence
leukocyte reduction filters for RBC components. of donor reactions compared with collection of whole
blood.9 Some of this may be attributed to the infusion of
Red Blood Cells saline during the procedure to replace lost volume.

RBCs collected by apheresis are typically collected as a dou- Plasma


ble unit (termed a 2RBC or double RBC procedure). Depend-
ing on the instrument used, the plasma and platelets are Collection of plasma by apheresis is termed plasmapheresis.
returned to the donor, or one or both of these components In a plasmapheresis procedure, whole blood from the donor
may be collected as a concurrent apheresis product. A is centrifuged, the plasma is diverted into a collection bag,
clinical advantage to the collection of apheresis RBCs is and the cellular components (RBCs, platelets, WBCs) are re-
reduced donor exposure for the recipient since the patient turned to the donor. This allows a larger volume of plasma
can potentially receive 2 units from the same individual. to be collected from a donor, such that each apheresis unit
An FDA guidance issued in 2001 requires the collection (“jumbo” plasma) is the volume equivalent of at least two
facility to follow specific donor selection criteria outlined whole blood–derived plasma units. In addition to direct
in each apheresis instrument manufacturer’s operator’s man- patient use, collection of apheresis plasma can serve a variety
ual.16 Donors must meet the appropriate collection criteria of purposes. It can be used to augment the inventory of
for a whole blood donation; however, many instruments fresh-frozen plasma (FFP) of a particular ABO group, espe-
have minimum gender-specific height and weight standards cially group AB. Plasma can be collected from donors with
as well. Since the volume of RBCs being collected during a high titers of antibodies directed against specific infectious
2RBC procedure is greater than it would be for a whole blood agents (hepatitis B, cytomegalovirus, varicella zoster) to
donation, the requirements for donor hematocrit are more prepare immune globulin. These preparations are used to
stringent. The hematocrit must be at least 40% regardless of provide prophylaxis against infectious organisms in exposed
gender, and the level (hemoglobin or hematocrit) must be individuals. Finally, apheresis is used commercially to collect
determined by a quantitative method; the use of copper sul- plasma for further manufacturing into such products as
fate is not acceptable.9,16 intravenous immune globulin (IVIG), hepatitis immune
If 2 units of RBCs are collected by apheresis, the donor globulin, and Rh-immune globulin.
must wait 16 weeks before providing another donation that For donor purposes, collection is divided into frequent
includes RBCs. If one RBC and one plasma and/or platelet and infrequent plasmapheresis. With infrequent plasma-
unit are collected, the donor must wait 56 days before do- pheresis, donation occurs no more than once every 4 weeks,
nating another red blood cell product. These procedures may and the donor requirements are the same as for whole
be performed on both allogeneic and autologous donors. The blood.17 With frequent, or serial, plasmapheresis, donation

Table 18–3 Examples of Instruments Used for Donor Apheresis and Component(s) Collected
Instrument 2RBC Plasma Platelets WBC Combination
Haemonetics

• MCS+ LN8150 4 RBC/P


• MCS+ LN9000 4 PLT/P
• PCS-2 4
• Cymbal 4

CaridianBCT

• COBE Spectra 4 4 PLT/P


• Trima Accel 4 4 4 RBC/PLT/P

Fenwal

• ALYX 4 RBC/P
• Amicus 4 RBC/PLT/P
• Autopheresis-C 4

Fresenius AS104 4 4 4
RBC = red blood cells; WBC = white blood cells (granulocytes); PLT = platelets; P = plasma
6888_Ch18_396-416 29/10/18 5:41 PM Page 404

404 PART III Transfusion Practices

occurs more frequently than once every 4 weeks. There must period. However, to protect the donor, if a double or triple
be at least 2 days between procedures and no more than two apheresis platelet is collected, 7 (rather than 2) days must
procedures in a 7-day period. In addition, these donors must elapse before the donor is again eligible to provide apheresis
be evaluated periodically by a physician and must undergo platelets. A donor may undergo no more than 24 platelet-
specific laboratory testing (total protein and serum protein pheresis procedures in a rolling 12-month period. Excep-
electrophoresis or measurement of immunoglobulin levels).9 tions to any of these time periods can be made in unusual
RBC loss must not be greater than 25 mL per week. FDA circumstances (HLA-matched platelets for a specific patient)
guidelines recommend 12 L (14.4 L for donors weighing but must be approved by the blood bank medical director.
more than 175 pounds) as the maximum allowable plasma Finally, the total volume of plasma collected during any one
volume donated per year.17 procedure cannot exceed 500 mL (or 600 mL if the donor
weighs 175 lb or more) or the volume of plasma cleared by
Platelets the FDA for the instrument.9,20
Apheresis platelets produced on newer instruments are typ-
Platelets obtained by an apheresis procedure provide the ically leukocyte-reduced and contain less than 5 × 106 WBCs
equivalent of six to eight units’ whole blood–derived platelets per unit, which meets the regulatory guidelines established by
(random-donor platelets). This significantly decreases the the FDA and the AABB.9,20
donor exposure for a patient. In a plateletpheresis procedure,
platelets along with a portion of plasma are removed, and Granulocytes
the remaining RBCs, WBCs, and majority of the plasma are
returned to the donor. The platelets are suspended in donor The patient population that can benefit from granulocyte trans-
plasma in a collection bag specifically designed for platelet fusions is very limited.22,23 Patients who undergo aggressive
storage. A routine plateletpheresis procedure typically takes chemotherapy may develop profound neutropenia during the
45 to 90 minutes. Like other donor apheresis procedures, course of their treatment. The decrease in neutrophils places
additional blood components may be collected concurrently, these patients at risk for acquiring bacterial and fungal infec-
including a single RBC and/or plasma product. Furthermore, tions, which may become life-threatening. Granulocyte trans-
depending on the donor’s platelet count and the apheresis fusions have been shown to be beneficial in some severely
instrument used, a high-yield product can be obtained that neutropenic patients (neutrophil count less than 500/µL)
is subsequently divided into two or three platelet products, who meet the following criteria: documented (clinically or
each containing an acceptable number of platelets.18,19 by culture) infection for 24 to 48 hours that is unresponsive
Donor selection criteria for the plateletpheresis donor are the to standard antibiotic or antifungal therapy, demonstrated
same as for whole blood donation, with two additional require- bone marrow myeloid hypoplasia, and reasonable chance of
ments.20 Prior to each plateletpheresis procedure, a sample must bone marrow recovery (i.e., the neutropenia is reversible).24,25
be collected to determine the donor’s platelet count. The regu- Granulocyte transfusions have also shown favorable results in
lations concerning donor qualification can be somewhat con- the treatment of neutropenic neonates with sepsis.26,27
fusing. The platelet count must be at least 150,000/µL in order Although granulocytes can be prepared from a whole blood
to provide an adequate platelet collection and for the donor to donation,28 the yield is insufficient for treating a pediatric or
safely undergo the collection procedure. If the donor’s platelet adult patient. Collection of granulocytes by apheresis provides
count is less than 150,000/µL, he or she is deferred from platelet a higher yield product.29 Apheresis collection requires close com-
donation until a subsequent count is at least 150,000/µL. In munication between the blood center or apheresis center, the
some collection centers, the platelet count can be measured im- blood bank physician, and the patient’s physician. Since granu-
mediately and used to qualify the donor. However, if the platelet locytes must be transfused as soon as possible after collection
count is not immediately available, then the most recent platelet for optimal therapeutic effectiveness, typically there is insuffi-
count can be used for qualification.21 If it is the initial platelet- cient time to perform all infectious disease testing on the donor.
pheresis collection for the donor, or if 4 weeks have elapsed Therefore, advance planning can allow one or more donors to
since the prior platelet donation, it is not necessary that the be prescreened prior to the actual collection procedure.
platelet count be determined prior to beginning the procedure A minimum therapeutic dose is 1 × 1010 granulocytes per
as long as it is evaluated after the platelet collection. day.24,25 The granulocyte yield is influenced by the donor’s
Plateletpheresis collections should not be performed on neutrophil count and the collection process. In general, most
potential donors taking medications that interfere with collection facilities stimulate the donor with corticosteroids
platelet function, as this would result in production of a or a colony-stimulating factor prior to the procedure and
suboptimal and therapeutically ineffective patient product. utilize a red blood cell sedimenting agent during the collec-
Antiplatelet medications have differing deferral time periods: tion process.30
48 hours for aspirin, aspirin-containing medications, and the During centrifugation of whole blood, granulocytes are
anti-inflammatory drug Feldene, and 14 days for clopidogrel found in the buffy coat between the RBC and plasma layers
(Plavix), ticlopidine (Ticlid), ticagrelor (Brilinta), vorapaxar (see Fig. 18–2). Adding the red blood cell sedimenting agent,
(Zontivity), and prasugrel (Effient).9 hydroxyethyl starch (HES), allows better separation of layers,
The interval between plateletpheresis procedures must be resulting in an improved yield with reduced RBC contamina-
at least 2 days with no more than two procedures in a 7-day tion.31,32 Since HES is added directly to the apheresis circuit,
6888_Ch18_396-416 29/10/18 5:41 PM Page 405

Chapter 18 Apheresis 405

some amount will enter the donor’s circulation during the The TA procedure is classified according to the blood compo-
collection procedure and ultimately be removed by the retic- nent removed: a cytapheresis procedure may be used to selec-
uloendothelial system. Immediate side effects of HES are due tively remove RBCs, WBCs, or platelets; a plasmapheresis
to its colloid properties, including circulatory volume expan- procedure is used to remove plasma when the pathological
sion, with headaches and peripheral edema. Residual HES has substance is found in the circulation. Therapeutic apheresis
been reported in donors up to 1 year after granulocyte aphere- has become an accepted and standard therapy for many hema-
sis; therefore, collection facilities are required to control the tologic, neurological, renal, metabolic, autoimmune, and rheu-
maximum cumulative dose of HES (or any other sedimenting matic diseases, among others.33,34
agent) given to a donor during a specified time interval.9
A number of granulocytes are normally present in the General Considerations
marginal, or noncirculating, pool. The administration of oral
Therapeutic apheresis has placed blood banks and transfu-
corticosteroids, such as prednisone or dexamethasone, can
sion services in the position of providing direct medical care
mobilize these granulocytes and significantly increase the
for a patient. This situation has necessitated a change in the
number of circulating granulocytes. The use of steroids in
perspective of the medical director and the technical staff.
donors prior to granulocyte collection may exacerbate
Clearly defined policies must delineate the responsibility of
certain medical conditions, such as diabetes, peptic ulcer, or
the blood bank and attending physicians; typically, the blood
hypertension and should be used under the guidance of the
bank physician provides TA as a consultative service. It is
blood bank physician.26
imperative that the blood bank medical director be closely
The administration of granulocyte colony-stimulating
involved with the attending physician in deciding whether
factor (GCSF), a recombinant hematopoietic growth factor,
there are clinical indications for the TA procedure requested.
to granulocyte donors has resulted in marked increases in
Issues such as who makes the decision about vascular access,
granulocyte yield. Although mild side effects, such as muscle
who orders laboratory tests to evaluate and monitor the
and skeletal pain, have been reported with the use of these
patient, and who chooses replacement fluids must be clearly
growth factors, they are usually well tolerated by donors.30
defined. The technical staff must be properly trained in the
care of very ill patients and be able to handle emergency sit-
uations. As with donor apheresis, written informed consent
Advanced Concepts must be properly obtained from the patient, outlining risks
Apheresis granulocytes contain a large number of viable and benefits. Obviously, the number and type of risks will
lymphocytes. If transfused to a severely immunocompro- vary depending on the type of TA procedure being performed
mised patient, there is a significant risk for graft-versus-host and the anticipated duration of treatment. Proper documen-
disease (GVHD). Therefore, the product should be irradi- tation of all facets of the procedure is required.35
ated prior to administration. Granulocyte function will Numerous studies performed during the last decade,
not be affected. Even with the use of sedimenting agents, many of them randomized, controlled clinical trials, have
granulocyte preparations contain a significant number of provided clinicians with sufficient data to more realistically
RBCs (the component resembles a diluted RBC rather than evaluate apheresis as a form of therapy and to define its role
a platelet concentrate). Compatibility testing is typically in the treatment of numerous disorders. Every 3 years, the
performed, since the apheresis granulocyte product contains American Society for Apheresis (ASFA) publishes guidelines
greater than 2 mL of RBCs.9 Leukocyte depletion filters for therapeutic apheresis based on the latest data from clin-
must not be used; a standard blood administration filter is ical trials, case studies, and anecdotal reports. Following a
sufficient. multidisciplinary committee review, each disease is placed
into a category for recommended guidelines.36 Each category
represents the strength of evidence for the role and efficacy
Therapeutic Procedures of TA in the management of each disease.
The indication categories for therapeutic apheresis are as
The rationale of therapeutic apheresis (TA) is based on the
follows:36
following:
• Category I. Apheresis is a first-line treatment, alone or in
• A pathogenic substance exists in the blood that con-
conjunction with other therapies.
tributes to a disease process or its symptoms.
• Category II. Apheresis is a second-line treatment, alone
• The substance can be more effectively removed by apheresis
or in conjunction with other therapies.
than by the body’s own homeostatic mechanisms.
• Category III. The optimal role for apheresis has not been
Therefore, therapeutic apheresis, like donor apheresis, in- established. Treatment should be individualized based on
volves the removal of a specific blood component, with return clinical evaluation and assessment of the anticipated risks
of the remaining blood constituents to the patient. However, and benefits.
with TA, since the component being removed is considered • Category IV. Apheresis is reported as either of no benefit
pathological (or contributing to the patient’s underlying disease or harmful in these conditions. Clinical applications
state), significantly larger volumes of blood must be processed should be undertaken only under an approved research
in order to remove as much of the offending agent as possible. protocol.
6888_Ch18_396-416 29/10/18 5:41 PM Page 406

406 PART III Transfusion Practices

These guidelines are published and periodically updated using TA to manage specific diseases. If needed, more thor-
(Table 18–4).36 Several new Category I indications in the ough reviews should be consulted.30,34,35
seventh edition of the ASFA guidelines include encephalitis
associated with antibodies against NMDA-2 receptor and Vascular Access
progressive multifocal leukoencephalopathy associated with Adequate vascular access is mandatory during TA, as larger
the drug natalizumab, a monoclonal antibody therapy for volumes of blood are processed and the duration of the pro-
relapsing multiple sclerosis.36 It should be noted that this cedure is longer than for donor apheresis. Vascular access
text is not intended to provide detailed information on can be obtained via peripheral veins, central veins, or a

Table 18–4 Indication Categories for Therapeutic Apheresis


Disease Procedure Indication Category
Renal and Metabolic Diseases

Antiglomerular basement membrane disease Plasma exchange I

ANCA-associated rapidly progressive glomerulonephritis Plasma exchange I

Hemolytic uremic syndrome (atypical) Plasma exchange I, III

Renal transplantation Plasma exchange I

Antibody-mediated rejection Plasma exchange I

Recurrent focal glomerulosclerosis Plasma exchange I

Heart transplant rejection Plasma exchange III

Photopheresis II

Acute liver failure Plasma exchange III

Familial hypercholesterolemia Selective adsorption I

Plasma exchange II

Overdose or poisoning Plasma exchange II–III

Phytanic acid storage disease Plasma exchange II

Sepsis, with multiorgan failure Plasma exchange III

Thyrotoxicosis Plasma exchange III

Autoimmune and Rheumatic Diseases

Cryoglobulinemia Plasma exchange I

Autoimmune hemolytic anemia (warm) Plasma exchange III

Rheumatoid arthritis, refractory Immunoadsorption II

Scleroderma (progressive systemic sclerosis) Plasma exchange III

Recalcitrant atopic dermatitis Plasma exchange III

Systemic lupus erythematosus, severe Plasma exchange II

Cardiac neonatal lupus syndrome Plasma exchange III

Complex regional pain syndrome Plasma exchange III

Hematologic Diseases

ABO-incompatible hematopoietic stem cell transplant Plasma exchange II

ABO-incompatible solid organ transplant

Kidney Plasma exchange II

Heart (infants) Plasma exchange II


6888_Ch18_396-416 29/10/18 5:41 PM Page 407

Chapter 18 Apheresis 407

Table 18–4 Indication Categories for Therapeutic Apheresis—cont’d


Disease Procedure Indication Category
Hematologic Diseases

Liver Plasma exchange III

Erythrocytosis/polycythemia vera Erythrocytapheresis III

Leukocytosis and thrombocytosis, symptomatic Cytapheresis I, II

Thrombotic thrombocytopenia purpura Plasma exchange I

Post-transfusion purpura Plasma exchange III


Sickle cell disease RBC exchange I–II

Hyperviscosity associated with monoclonal gammopathy Plasma exchange I

Coagulation factor inhibitors Plasma exchange IV

Immunoadsorption III

Aplastic anemia Plasma exchange III

Cutaneous T-cell lymphoma Photopheresis I

Red blood cell alloimmunization in pregnancy (if intrauterine transfusion Plasma exchange II
not available)

Malaria or babesiosis RBC exchange I, II

Erythropoietic porphyria Plasma exchange III

HELLP syndrome Plasma exchange III

HLA desensitization for HSC transplantation Plasma exchange III

Neurological Disorders

Acute inflammatory demyelinating polyneuropathy (Guillain-Barré syndrome) Plasma exchange I

Chronic inflammatory demyelinating polyradiculoneuropathy (CIDP) Plasma exchange I

Lambert-Eaton myasthenic syndrome Plasma exchange II


Multiple sclerosis

Acute CNS inflammatory disease Plasma exchange II

Chronic progressive Plasma exchange III

Myasthenia gravis Plasma exchange I

Paraneoplastic neurological syndromes Plasma exchange III

Immunoadsorption III

Paraproteinemic polyneuropathy

Due to IgG, IgA, or IgM Plasma exchange I

Due to multiple myeloma Plasma exchange III

Rasmussen’s encephalitis Plasma exchange II

N-methyl-D-aspartate receptor antibody encephalitis Plasma exchange I

Progressive multifocal leukoencephalopathy associated with natalizumab Plasma exchange I

Hashimoto’s encephalopathy Plasma exchange II

PANDAS* Plasma exchange I

*PANDAS = pediatric autoimmune neuropsychiatric disorders associated with streptococcus


Source: Szczepiorkowski Z, editor. Clinical applications of therapeutic apheresis: an evidence based approach. J Clin Apheresis. 25:81, 2010.
6888_Ch18_396-416 29/10/18 5:41 PM Page 408

408 PART III Transfusion Practices

combination of both. If only one to two TA procedures are to and replaced with sufficient physiological fluid to maintain
be performed, peripheral access can be used. In such in- the intravascular compartment. It has been postulated that
stances, two venous sites are necessary—one for removal and beneficial effects of the procedure, particularly in diseases
one for return. Therefore, the patient must have adequate that involve malfunction of the immune system, may be
veins at two sites capable of accommodating a 16- to 18-gauge attributed to the removal of factors listed in Box 18–1.48
needle. If several or frequent TA procedures are required, cen- The effectiveness of TPE is related to the volume of
tral venous access with a double-lumen catheter is desirable. plasma removed and the concentration of the pathological
Catheters designed for hemodialysis are preferable; standard substance in the blood. Apheresis is most efficient at remov-
intravenous lines or central catheters are not designed to pro- ing the substance at the beginning of the procedure and least
vide the flow rates required with apheresis. efficient at the end. A one-volume exchange removes an
amount of plasma equal to the patient’s plasma volume,
Physiological Considerations approximately 3 L for the average-size adult patient. How-
The patient’s extracorporeal blood volume (ECV) should be ever, a one-volume exchange does not reduce the unwanted
less than 15% of the total blood volume (TBV) in order to plasma component to 0% as might be expected. There are
minimize the risk of hypovolemia. Current instruments used several reasons this occurs. Physiologically, there is varying
for TA typically calculate the patient’s TBV based on height transfer of plasma proteins between the intravascular and
and weight. Calculating the patient’s plasma volume (PV) is extravascular spaces; some plasma proteins are primarily
often necessary when performing a plasma exchange in order partitioned in the extravascular compartment; and synthesis
to appropriately remove sufficient amounts of plasma during or catabolism of the protein may occur.
the TA procedure. This is easily calculated from the patient’s In addition, during the TPE procedure, replacement fluids
TBV and hematocrit. The lower the patient’s hematocrit, the are being administered to maintain the patient’s intravascular
higher the PV. volume, resulting in dilution of plasma proteins. Mathemat-
Attention must be given to the patient’s medication ical models depicting the theoretical removal of substances
schedule. Plasmapheresis in particular may dangerously from plasma by TPE show that a one-volume exchange
lower plasma levels of medications as the drug is removed should reduce the unwanted plasma component to approx-
during the procedure. Removal of a drug is based on its vas- imately 30% of its initial value. If a two-volume exchange is
cular distribution (drugs that are present primarily in the performed, the procedure becomes less efficient, reducing
intravascular space are most vulnerable), its half-life in the unwanted component from 30% to only 10%.30,49
the circulation, the timing of the drug’s administration, the Because of the diminishing effect of increased plasma re-
amount of plasma removed, and the duration of the proce- moval, it is recommended that approximately 1 to 1.5 plasma
dure. A few studies have evaluated the removal of specific volumes be exchanged per procedure.30,35
medications,37 and a clinical pharmacist can often offer As mentioned, synthesis and catabolism of the patho-
additional information on the possible extracorporeal re- logical protein and its distribution between the extravas-
moval of various drugs. cular and intravascular space are factors affecting the
The number of TA procedures performed varies with the efficiency of TPE. Disorders characterized by overproduc-
disease or disorder being treated and the individual patient. tion of a specific monoclonal or polyclonal antibody (e.g.,
For plateletpheresis and leukapheresis, one to two TA Waldenström’s macroglobulinemia, cryoglobulinemia)
procedures may be sufficient. For erythrocytapheresis (RBC can result in hyperviscosity syndromes. If the antibody is
exchange), only one TA procedure may be needed; however, IgM, TPE can be an effective therapeutic tool, since IgM
some patients benefit from an RBC exchange on a more fre- antibodies tend to be located primarily in the intravascular
quent basis. Almost all diseases treated by plasmapheresis space. However, IgG antibodies are often distributed
require several TA procedures in order to remove sufficient
amounts of the pathological substance.
A blood warmer is often used as part of the apheresis
circuit to avoid hypothermia due to the rapid infusion of BOX 18–1
room temperature fluids. Factors Removed by Therapeutic Plasmapheresis
Plasmapheresis (Plasma Exchange) • Immune complexes (e.g., systemic lupus erythematosus)
• Alloantibodies (e.g., antibody-mediated transplant rejection)
Therapeutic plasma exchange (TPE) is the removal and • Autoantibodies (e.g., Guillain-Barré syndrome, Goodpasture’s
retention of the plasma, with return of all cellular compo- syndrome)
nents to the patient. This is the most common TA procedure • Immunoglobulins causing hyperviscosity (e.g., Waldenström’s
macroglobulinemia)
performed. The purpose is to remove the agent in the
• Protein-bound toxins or drugs (e.g., Amanita mushroom
plasma, such as an antibody, toxin, or abnormal protein, that
poisoning, barbiturate poisoning)
is causing the clinical symptoms.38–43 TPE is also used to
• Lipoproteins (e.g., familial hypercholesterolemia,
replace a normal factor or substance that may be missing or hypertriglyceridemia)
deficient in the patient’s plasma.44–47 Regardless of the pur- • Phytanic acid (e.g., Refsum’s disease)
pose, a large volume of plasma must be removed during TPE
6888_Ch18_396-416 29/10/18 5:41 PM Page 409

Chapter 18 Apheresis 409

equally in the intravascular and extravascular spaces, in patients with sickle cell disease in order to decrease the
requiring a series of TPE procedures as the intravascular number of hemoglobin S–containing RBCs, thereby treating
and extravascular pools equilibrate. Because of these char- or preventing the complications (acute chest syndrome, im-
acteristics, TPE performed to remove IgG antibodies is pending stroke, unrelenting painful crisis) associated with
most effective when combined with immunosuppressive the disease.30,34,53
drugs. The therapeutic goal is to decrease the level of hemoglo-
bin S to less than 30%. This is usually accomplished with a
Plateletpheresis single red blood cell exchange procedure, requiring from
6 to 10 RBC units, depending on the patient’s age and red
Therapeutic plateletpheresis can be used to treat patients who blood cell volume. The donor RBCs selected for transfusion
have abnormally elevated platelet counts with related symp- should be ABO- and Rh-compatible, relatively fresh (less
toms.30 Thrombocytosis (at least 500,000/µL) can occur in than 10 days is preferable to allow maximum in vivo sur-
myeloproliferative disorders (essential thrombocythemia, vival), leukocyte-reduced, and negative for hemoglobin S
polycythemia vera, chronic myelogenous leukemia) or as a (donor does not have sickle cell trait). Some centers also
reactive process in response to splenectomy, infection, chronic recommend partially phenotype-matched blood for the Rh
inflammation, or malignancy. Should the platelet count reach (C, c, E, e) and K1 antigens to avoid future alloimmuniza-
levels above 1,000,000/µL, the patient is at risk for developing tion.26,35 Some patients may be placed on a long-term pro-
thrombotic or hemorrhagic complications. The preferred gram of prophylactic red blood cell exchange to decrease
method for lowering the platelet count is medication;50 how- the risk of complications associated with the disease.
ever, therapeutic apheresis may be indicated during an acute Other indications for red blood cell exchange are much
event to rapidly reduce the platelet count until pharmacolog- less common. The procedure can be considered for treatment
ical therapy takes effect. During a plateletpheresis procedure, of overwhelming malaria or Babesia infections.54–56 Both of
the platelet count will be decreased by 30% to 60%. More than these protozoa infect red blood cells, and a pronounced par-
one procedure may be required until the platelet count asitemia can occur. Red blood cell exchange has been shown
normalizes to desired levels (usually less than 600,000/µL).34 to be beneficial for treating these patients when the parasite
There are no specific guidelines as to the level to which the load is greater than 10%. A 1.5- to 2-volume red blood cell
platelet count must be reduced or a standardized procedure exchange should significantly decrease the parasite load.30,34
to reach a particular target platelet count. It is important to note that these patients are often extremely
ill and must be closely monitored during the procedure.
Leukapheresis Finally, red blood cell exchange can be used to remove in-
compatible RBCs from a patient’s circulation; for example, the
Therapeutic leukapheresis has been used to treat patients emergent transfusion of Rh-positive RBCs to an Rh-negative
with hyperleukocytosis, defined as a WBC or circulating female of child-bearing potential or an ABO-mismatched
blast count of over 100,000/µL.34 These elevated levels transfusion.35
place the patient at risk for complications associated with
leukostasis, including organ dysfunction due to the forma- Fluid Replacement
tion of microthrombi in the pulmonary and cerebral
microvasculature.51 Leukostasis is more common in patients In TA procedures, the extracorporeal circuit (tubing, collection
with acute myelogenous leukemia (AML) than with acute chamber, blood warmer) is primed with normal saline, pro-
lymphocytic leukemia (ALL).30 It is difficult to predict how viding the patient with an initial bolus of crystalloid. During
much blood volume should be processed to permit a suffi- therapeutic cytapheresis procedures, additional fluid replace-
cient reduction in WBCs or blast cells to prevent leukostasis, ment is often not necessary, since the majority of the patient’s
and WBC counts should be monitored during the procedure. plasma is being returned. Should hypovolemia become a
A single procedure should reduce the WBC count by 30% to concern, normal saline can be infused during the procedure.
60%; however, more than one procedure may be necessary In therapeutic plasmapheresis procedures, however, large
due to rapid mobilization of cells from the extravascular volumes of the patient’s plasma are retained. This fluid must
compartment. To achieve an adequate reduction in the WBC be replaced to maintain appropriate intravascular volume
count, up to 1 L of fluid may be removed, necessitating and oncotic pressure. Several options are available, and the
the use of a replacement fluid. Use of a red blood cell sedi- choice is determined by the disease being treated, the con-
menting agent, such as HES, may be of benefit during a dition of the patient, and the preference of the institution.
leukapheresis procedure.30,52 The most common replacement fluid for TPE is human
serum albumin (HSA) as a 5% solution.34,57 Although
Erythrocytapheresis (Red Blood Cell Exchange) 5% HSA can be used to replace the entire volume removed,
a crystalloid such as normal saline may be used for up to
Erythrocytapheresis, or red blood cell exchange, removes a one third of the replacement volume. The availability of
large number of RBCs from the patient and returns the pa- sufficient HSA has been challenging in recent years, leading
tient’s plasma and platelets along with compatible allogeneic to the use of pentastarch as a portion of the replacement
donor RBCs. The procedure is most commonly performed solution in addition to HSA.58,59
6888_Ch18_396-416 29/10/18 5:41 PM Page 410

410 PART III Transfusion Practices

Fresh-frozen plasma (FFP) contains all the constituents and return. Autologous HPC donors often have an existing
of the removed plasma and thus would appear to be the central venous line.
optimal replacement fluid for TPE procedures. However, There have been remarkable advances in the field of HPC
the use of FFP is not without risk, including transmission collection and transplantation over the last several years.
of infectious disease, additive effect contributing to citrate Donor or patient selection and care; laboratory testing; and
toxicity, and sensitization to plasma proteins. Therefore, the collection, storage, and infusion of HPCs are regulated
the use of FFP is usually reserved for treatment of throm- by the FDA,62 the AABB,63 and the Foundation for the Ac-
botic thrombocytopenic purpura (TTP) and related disor- creditation of Cellular Therapy (FACT).64 The enormous
ders. For patients with TTP who do not respond in a timely complexity of this practice is beyond the scope of this chap-
manner to replacement with FFP, cryoprecipitate-reduced ter; the regulations and standards as well as other relevant
plasma is an alternative.35 FFP may also be used as replace- literature should be consulted.30,35
ment during TPE on patients with a preexisting coagulopa-
thy (severe liver disease) or on those who are scheduled
to undergo an invasive procedure (such as a pretransplant Advanced Concepts
antibody reduction). Immunoadsorption/Selective Absorption

Special Procedures Therapeutic plasma exchange is used to treat a variety of


immunological disorders. In the TPE procedure, 5% HSA
Expanding on the basic principles of separation employed is typically used as replacement fluid. Although HSA is a
by apheresis, new technology has allowed the collection relatively safe fluid, there are disadvantages to its long-term
of very specialized components, as well as the removal of use on a daily or slightly less frequent basis. Most signifi-
specific plasma constituents. cant are the depletion of coagulation factors (e.g., fibrino-
gen) and reduction in immunoglobulin levels.30 This led
Hematopoietic Progenitor Cells to the development of methods to selectively remove
pathological substances, in particular antibodies.
Hematopoietic progenitor cells (HPCs), also referred to as Immunoadsorption refers to a method in which a specific
peripheral blood stem cells (PBSCs), can be collected by ligand is bound to an insoluble matrix in a column or filter.
apheresis from an autologous or allogeneic donor.30 The pro- Plasma is separated from anticoagulated whole blood by
cedure is much like donor leukapheresis, with the selective means of centrifugation or filtration, using a routine
collection of mononuclear cells, since HPCs are found in the plasmapheresis procedure. The sequestered plasma is then
upper portion of the buffy coat during centrifugation. At perfused through the column or filter, with selective re-
least one, and sometimes two to three, apheresis collections moval of the pathogenic substance and subsequent reinfu-
are usually needed to produce an acceptable “dose.” Each sion of the patient’s own plasma and cellular components.
collection procedure lasts 4 to 6 hours, since very large blood The removal is usually mediated by an antigen–antibody
volumes are processed in order to obtain the required yield. or chemical reaction. This abrogates the need for a volume-
The number and frequency of procedures are determined by replacement fluid such as 5% HSA. Figure 18–10 is a dia-
the yield of HPCs and the patient’s condition. grammatic representation of this process.
Hematopoietic growth factors, in particular GCSF, are Staphylococcal protein A is a cell wall component of
commonly used prior to the collection procedure to increase Staphylococcus aureus. It has an affinity for IgG classes 1,
the number of circulating stem cells in the peripheral circu- 2, and 4 and for IgG immune complexes. When plasma is
lation. HPCs express the cell surface glycoprotein CD34, passed through a small column containing staphylococcal
and measurement of CD34+ cells in the peripheral blood protein A, a significant amount of IgG is removed, as
prior to collection is typically performed to ensure adequate well as lesser amounts of IgM and IgA. The mechanism of
mobilization has occurred.30,60,61 In contrast to use in the action of these columns is not well established, and the
routine granulocyte donor, four to five daily injections of removal of IgG and immune complexes alone cannot
GCSF are typically required to mobilize sufficient HPCs for explain the treatment’s clinical benefit. There appears to be
collection. Therefore, it is not uncommon for individuals to a significant immunomodulatory effect of this treatment,
experience headache and musculoskeletal pain. with enhanced anti-idiotypic antibody regulation and
There are several advantages to using HPCs collected by activated cellular immune function.30,65
apheresis over traditional bone marrow collection. Anesthe- Initially licensed to treat patients with either refractory
sia is avoided, and the procedures can be performed safely idiopathic thrombocytopenic purpura (ITP)34 or rheuma-
in the outpatient setting. For the autologous HPC donor, toid arthritis,66 it was also used off-label to treat a variety
other advantages include a shorter period of cytopenia, of other disease processes.67–69 Several adverse reactions,
decreased transfusion requirements, fewer infectious com- including fever, chills, hypo- and hypertension, and allergic
plications, and decreased length of hospitalization.35 Al- reactions were reported, including death.70 In 2006, man-
though peripheral venous access is preferred (two venous ufacturing of the columns ceased in the United States, and
sites are required), a small number of allogeneic donors will they are no longer available for use.
require central venous line placement for adequate access
6888_Ch18_396-416 29/10/18 5:41 PM Page 411

Chapter 18 Apheresis 411

Reservoir

Plasma Whole Blood

Formed
Elements

Affinity Plasma Whole Blood


Column
or Filter

Figure 18–10. Perfusion of plasma over


columns or filters. (From Berkman, EM, and
Umlas, J [eds]: Therapeutic hemapheresis: a
technical workshop. American Association of Cell
Blood Banks, Washington, DC, 1980, p. 142, Separator
with permission.) Pump

Familial hypercholesterolemia is an autosomal- (cell death).30 Photopheresis has been shown to be effica-
dominant disorder associated with abnormal clearance of cious and has been approved by the FDA for the treatment
low-density lipoproteins (LDL), resulting in markedly of cutaneous T-cell lymphoma.71 It has subsequently been
elevated cholesterol levels. Both homozygotes and het- used successfully to treat acute and chronic graft-versus-host
erozygotes experience significant morbidity and mortal- disease,72 solid organ transplant rejection,73 and selected
ity from premature atherosclerotic cardiovascular disease. immunologically mediated diseases.30,74,75
TPE is only partially effective in this disorder. Several
selective removal systems have been developed; the
Kaneka Liposorber and Braun Plasmat Secura systems Adverse Effects
have both been approved for use in the United States.34
LDL-apheresis selectively removes LDL and has been as- Apheresis is accepted as a relatively safe procedure, but com-
sociated with regression of cardiovascular disease.70 Since plications do occur, especially for donors at higher risks for
the underlying disorder cannot be treated, LDL-apheresis presyncopal or syncopal reactions associated with compo-
must be performed indefinitely, typically at 2- to 3-week nent collections due to donor factors such as younger
intervals.26 age, female sex, and small total blood volume.6,7,76 Adverse
Numerous adsorptive matrices have been developed effects may be observed in therapeutic procedures as well.77
with varying clinical usefulness. These include charcoal In this case, it is sometimes difficult to evaluate whether the
for removal of bile acids, polymyxin B for removal of deleterious effects were caused by the procedure or by the
endotoxin, and cellulose acetate for removal of granulo- underlying disease. Some of the problems encountered are
cytes or monocytes, to mention a few. The majority of the listed in Box 18–2. Some of the more common reactions
adsorbent systems are not available or in clinical use in the and complications of donor and therapeutic apheresis are
United States.30 discussed here.77–80

Photopheresis Citrate Toxicity

Photopheresis utilizes leukapheresis to collect the buffy Citrate toxicity is usually observed during cytapheresis com-
coat layer from whole blood. These cells are treated with ponent collections when anticoagulated plasma is returned
8-methoxypsoralen (8-MOP), exposed to ultraviolet A at a rapid rate. Citrate is the anticoagulant used in apheresis
(UVA) light, and then reinfused into the patient. The com- and is normally metabolized quickly in the liver. If the
bination of 8-MOP and UVA irradiation results in cross- amount of citrate infused exceeds the body’s ability to me-
linking of leukocyte DNA, ultimately leading to apoptosis tabolize it, the level of ionized calcium will decrease, and the
donor may feel numbness or tingling around the mouth
6888_Ch18_396-416 29/10/18 5:41 PM Page 412

412 PART III Transfusion Practices

age, low weight, first-time donation, and inattentive collec-


BOX 18–2 tion staff.6,7
Adverse Effects of Apheresis
Miscellaneous Reactions
• Citrate toxicity
• Vascular access complications (hematoma, sepsis, phlebitis, Hypovolemia is observed more frequently with IFC instru-
neuropathy)
ments. Careful monitoring of the volume in and out is nec-
• Vasovagal reactions
essary to prevent hypovolemia and hypervolemia. Allergic
• Hypovolemia
reactions are related to the replacement fluids. This type of
• Allergic reactions
reaction is generally observed in patients in whom FFP is
• Hemolysis
being administered (such as during a TPE procedure for
• Air embolus
TTP), but it has also been reported during infusion of
• Depletion of clotting factors
albumin. Hemolysis is usually caused by a mechanical prob-
• Circulatory and respiratory distress
lem with the equipment, such as a kink in the plastic tubing.
• Transfusion-transmitted diseases
Observing the return line is critical in avoiding this problem.
• Lymphocyte loss
Air embolism and clotting factor deficiencies are not com-
• Depletion of proteins and immunoglobulins
monly observed.

Plasma Protein Interactions


(paresthesias).35 To prevent this complication, calcium sup- The concentration of most plasma substances is reduced by
plementation by mouth may also be given. Citrate toxicity 50% to 60% after one standard plasmapheresis treatment,
is also relatively common during therapeutic apheresis pro- with the rate of return to steady state concentrations varying
cedures. If unattended, the symptoms can lead to tetany and among analytes. Plasma lipids, total protein, immunoglobu-
cardiac arrhythmia. Parenteral calcium (calcium gluconate lins, and transferrin recover to steady state concentrations
or calcium chloride) may be infused intravenously while by 8 days’ postplasmapheresis, whereas ceruloplasmin con-
the patient is undergoing the therapeutic plasmapheresis centrations take longer to reach prepheresis levels.
to prevent symptoms of hypocalcemia. If FFP is used as
replacement fluid during a therapeutic plasma exchange, Fatalities
citrate toxicity is more likely to occur because of the com-
bined effects of the anticoagulant in the FFP and the citrate Rare fatalities occurring during therapeutic apheresis proce-
used in the apheresis procedure itself. dures have been reported. The majority of these have
been caused by circulatory (cardiac arrest or arrhythmia) or
Vascular Access respiratory complications (acute pulmonary edema or adult
respiratory distress syndrome). Of the cases reported, about
Though plasmapheresis is helpful in certain medical condi- half had received plasma as part or all of the replacement fluid.
tions, like any other therapy, there are potential risks and Because plasma has been associated with fatalities, its use is
complications that should be discussed with the patient be- recommended primarily in cases of TTP or hemolytic uremic
fore the procedure. Insertion of a rather large intravenous syndrome (HUS) in which there is a specific indication for its
catheter can lead to bleeding, lung puncture (depending on use. Plasma is also capable of transmitting diseases, such as
the site of catheter insertion), and if the catheter is left in too hepatitis and the human immunodeficiency viruses.
long, infection.

Vasovagal Reactions
CASE STUDY
A vasovagal episode is a feeling of malaise mediated by the
Case Study 18–1
vagus nerve. When it leads to syncope or “fainting,” it is
called vasovagal syncope, which is the most common type of A 72-year-old man presented to the emergency depart-
fainting. Another mechanism causing hypotension during ment at approximately 10:00 a.m. after waking during
apheresis procedures is the vasovagal reaction.76,78 In this re- the night with confusion and expressive aphasia. These
action, hypovolemia results in a decrease in blood pressure. symptoms initially appeared to improve and then progres-
The compensatory response for this volume depletion is to sively worsened. Apart from a recent 3-week history of
increase sympathetic nervous system output with physiolog- left shoulder pain on abduction, his previous medical
ical compensation as previously described. During a vasova- history was significant for excision of melanoma in situ
gal reaction, however, parasympathetic output that normally and multiple basal cell carcinomas, mild gastroesophageal
counteracts sympathetic output increases, resulting in a reflux, and an appendectomy 52 years earlier. He was a
slowing of heart rate and decreased vascular tone. This re- nonsmoker, did not drink alcohol, used no regular medi-
sults in hypotension. Factors that have been associated with cations, and had no known drug allergies.
vasovagal reactions in whole blood donors include younger
6888_Ch18_396-416 29/10/18 5:41 PM Page 413

Chapter 18 Apheresis 413

Physical examination findings: • Normal D-dimer and fibrinogen


• Expressive aphasia • Elevated LDH
• Cranial nerves intact • Decreased haptoglobin
• Vital signs: BP 150/90 mm Hg, heart rate (HR) 82 bpm,
Hospital Course
respiratory rate (RR) 12, temperature 101.4°F.
• All extremities moved equally, with normal strength. Overnight, the patient became increasingly agitated,
• Multiple petechiae over both lower extremities and refused oral intake, and pulled out his intravenous
abdomen. catheter. His urine output decreased significantly and he
• Computerized tomography (CT) of the brain showed became anuric. He was then transferred to the intensive
no abnormalities. care unit (ICU).
• Carotid duplex ultrasound revealed no significant
Negative Direct Antiglobulin Test (DAT)
carotid or vertebral artery disease.
• Twelve-lead electrocardiograph (ECG) was normal. Peripheral smear: decreased platelets, rare nucleated RBC,
• Chest x-ray showed normal heart size and no 2+ schistocytes.
abnormalities. Decreased ADAMTS13 level
1. What are the top five possible diagnoses for this case?
Laboratory findings during the first 48 hours:
2. What is the most likely diagnosis for this patient
• Anemia and why?
• Thrombocytopenia 3. What are the main symptoms in cases of this type?
• Normal prothrombin time (PT) and partial thrombo- 4. What is the main treatment for this disease?
plastin time (PTT)

SUMMARY CHART
 In an apheresis procedure, blood is withdrawn from a through the membrane while the cellular portion
donor or patient and separated into its components. passes over it.
One or more of the components is retained, and the  The most common anticoagulant used in apheresis is
remaining constituents are recombined and returned acid citrate dextrose.
to the individual.  Therapeutic apheresis is used to remove a pathological
 The process of removing plasma from the blood is substance, to supply an essential or missing substance
termed plasmapheresis; removing platelets is termed to alter the antigen–antibody ratio, or to remove im-
plateletpheresis or thrombocytopheresis; removing mune complexes.
RBCs is termed erythrocytapheresis; removing leuko-  The American Society for Apheresis (ASFA) has devel-
cytes is known as leukapheresis. oped categories to define the effectiveness of therapeu-
 Apheresis equipment that uses intermittent flow cen- tic apheresis in treating a particular condition or
trifugation (IFC) requires only one venipuncture, in disease. Therapeutic apheresis is most appropriate for
which the blood is drawn and reinfused through the treating category I or II disorders.
same needle. Once the desired component is separated,  In therapeutic plasmapheresis procedures, the replace-
the remaining components are reinfused to the donor, ment fluids used to maintain appropriate intravascular
and one cycle is complete. Apheresis procedures per- volume and oncotic pressure include normal saline,
formed on patients usually require many cycles to FFP, cryo-reduced plasma, and 5% human serum
reach an acceptable therapeutic endpoint. albumin.
 Continuous flow centrifugation (CFC) procedures  Complications of apheresis include vascular access
withdraw, process, and return the blood to the indi- issues, alteration of pharmacodynamics of medications,
vidual simultaneously. Two venipuncture sites are nec- citrate toxicity, fluid imbalance, allergic reactions,
essary. The process of phlebotomy, separation, and equipment malfunction (hemolysis), and infection.
reinfusion is uninterrupted. Fatalities have occurred (primarily patient rather than
 Membrane filtration technology uses membranes donor).
with specific pore sizes, allowing plasma to pass
6888_Ch18_396-416 29/10/18 5:41 PM Page 414

414 PART III Transfusion Practices

9. Peripheral blood stem cells are:


Review Questions
a. Responsible for phagocytosis of bacteria
1. The most common anticoagulant used for apheresis b. Removed during erythrocytapheresis
procedures is: c. Pluripotential hematopoietic precursors that circulate
in the peripheral blood
a.Heparin
d. Lymphocytes involved with the immune response
b.Sodium fluoride
c.Warfarin 10. Which of the following can be given to an apheresis donor
d.Citrate to increase the number of circulating granulocytes?
2. Therapeutic cytapheresis has a primary role in treatment a. DDAVP
of patients with: b. Hydroxyethyl starch (HES)
c. Immune globulin
a. Sickle cell disease and acute chest syndrome
d. G-CSF
b. Systemic lupus erythematosus to remove immune
complexes
c. Leukemia to help increase granulocyte production References
d. Myasthenia gravis to increase antibody production 1. McLeod BC. Therapeutic apheresis: history, clinical applica-
tion, and lingering uncertainties. Transfusion. 2009;50(7):
3. The minimum interval allowed between plateletpheresis 1413-25.
component collection procedures is: 2. Hester J. Apheresis: the first 30 years. Transfus Apher Sci.
a.1 day 2001;24:155-6.
b.2 days 3. Malchesky PS, Koo AP, Skibinski CI, Hadsell AT, Rybicki LA.
Apheresis technologies and clinical applications: the 2007
c.7 days international apheresis registry. Ther Apher Dial. 2009;
d.8 weeks 14(1):52-73.
4. Chen Y, Bieglmayer C, Höcker P, Dettke M. Effect of acute cit-
4. In plasma exchange, the therapeutic effectiveness is: rate load on markers of bone metabolism in healthy volunteers.
a. Greatest with the first plasma volume removed Vox Sang. 2009;97:324-9.
b. Affected by the type of replacement fluid used 5. Bell AM, Nolen JD, Knudson CM, Raife TJ. Severe citrate
c. Enhanced if the unwanted antibody is IgG rather than toxicity complicating volunteer apheresis platelet donation.
J Clin Apher. 2007;22:15-6.
IgM 6. Tomita T, Takayanagi M, Kiwada K, Mieda A, Takahashi C,
d. Independent of the use of concomitant immunosup- Hata T. Vasovagal reactions in apheresis donors. Transfusion.
pressive therapy 2002;42(12):1561-66.
7. Reiss RF, Harkin R, Lessig M, Mascari J. Rates of vaso-vagal
5. The replacement fluid indicated during plasma exchange reactions among first time teenaged whole blood, double
for TTP is: red cell, and platelet pheresis donors. Ann Clin Lab Sci.
a. Normal (0.9%) saline 2009;39(2):138-43.
8. Price TH. Centrifugal equipment for the performance of
b. Hydroxyethyl starch (HES) therapeutic hemapheresis procedures. In: MacPherson JL,
c. FFP Kaspirisin DO, editors. Therapeutic hemapheresis, vol 1. Boca
d. Albumin (human) 5% Raton (FL): CRC Press; 1985. p. 123.
9. Ooley PW, editor. Standards for blood banks and transfusion
6. The most common adverse effect of plateletpheresis col- services. 29th ed. Bethesda (MD): American Association of
lection is: Blood Banks; 2015.
a. Allergic reaction 10. Rock G, Tittley P, McCombie N. Plasma collection using an
automated membrane device. Transfusion. 1986;26:269-71.
b. Hepatitis 11. Burgstaler EA. Blood component collection by apheresis. J Clin
c. Hemolysis Apher. 2006;21:142-51.
d. Citrate effect 12. Code of Federal Regulations. Title 21 Parts CFE 606, 610.
Washington (DC): U.S. Government Printing Office; 2006.
7. Apheresis technology can be used to collect each of the 13. Balint B. Apheresis in donor and therapeutic settings: Recruit-
following components except: ments vs. possibilities—a multicenter study. Transfus Apher
a. Leukocytes Sci. 2005;33:181-9.
14. Eder AF, Benjamin RF. Safety and risks of automated collection
b. Macrophages of blood components. In: Eder AF, Goldman M, editors. Blood
c. Hematopoietic progenitor cells donor health and safety. Bethesda (MD): AABB Press; 2009.
d. Platelets pp. 103-21.
15. Rossmann SN. Talking with the donor: information, consent,
8. The anticoagulant added to blood as it is removed from a and counseling. In: Eder AF, Goldman M, editors. Blood
donor or patient during an apheresis procedure acts by: donor health and safety. Bethesda (MD): AABB Press. 2009;
a.Binding calcium ions pp 1-20.
16. Food and Drug Administration. Guidance for industry and
b.Increasing intracellular potassium FDA review staff. Technical correction: recommendations for
c.Binding to antithrombin III collecting red blood cells by automated apheresis methods.
d.Inactivating factor V Rockville (MD): FDA; 2001 Feb 13.
6888_Ch18_396-416 29/10/18 5:41 PM Page 415

Chapter 18 Apheresis 415

17. Food and Drug Administration. [1995 Mar 10]. FDA Memo- 41. Sivakumaran P, Vo AA, Villicana R, Peng A, Jordan SC,
randum. Revision of FDA memorandum of August 27, 1982. Pepkowitz SH et al. Therapeutic plasma exchange for desensi-
Requirements for infrequent plasmapheresis donors. Rockville tization prior to transplantation in ABO-incompatible renal
(MD): FDA; 1995 Mar 10. allografts. J Clin Apher. 2009;24:155-60.
18. Moog R. Feasibility and safety of triple dose platelet collection 42. Wu SG, Kuo PH, Yang PC. Successful weaning after plasma
by apheresis. J Clin Apher. 2009;24:238-40. exchange for polyneuropathy related to POEMS syndrome.
19. Picker SM, Radojska SM, Gathof BS. Prospective comparison J Clin Apher. 2009;24:170-2.
of high-dose plateletpheresis with the latest apheresis systems 43. Ezer A, Caliskan K, Parlakgumus A, Belli S, Kozanoglu I,
on the same donors. Transfusion. 2006;46:1601-08. Yildirim S. Preoperative therapeutic plasma exchange in
20. FDA Guidance for Industry and FDA Review Staff, December 17, patients with thyrotoxicosis. J Clin Apher. 2009;24:111-4.
2007, Collection of platelets by automated methods. 44. George JN, Terrell DR, Swisher KK, Vesely SK. Lessons learned
21. Alex J, Roberts B, Raife T. Postdonation platelet counts are from the Oklahoma thrombotic thrombocytopenic purpura-
safe when collecting platelets with the Trima Accel using a hemolytic uremic syndrome registry. J Clin Apher. 2008;
postdonation platelet count target of ≥50,000 platelets/µL. 23:129-37.
J Clin Apher. 2009;24:215-7. 45. Scully M. Thrombotic thrombocytopenic purpura and preg-
22. Atallah E, Schiffer CA. Granulocyte transfusion. Curr Opin nancy. ISBT Sci Series. 2007;2:226.
Hematol. 2006;13:45-49. 46. Lalmuanpuii J, Yalamanchili K, Fircanis S, Nelson JC. Hyper-
23. Price TH. Granulocyte transfusion therapy. J Clin Apher. 2006; sensitivity to plasma exchange in a patient with thrombotic
21:65-71. thrombocytopenic purpura. J Clin Apher. 2009;24:18-20.
24. King KE, editor. Blood transfusion therapy: a physician’s hand- 47. Park YA, Hay SN, King KE, Matevosyan K, Poisson J, Powers
book. 9th ed. Bethesda (MD): AABB; 2008. A, Sarode R, et al. Is it quinine TTP/HUS or quinine TMA?
25. Vamvakas EC, Pineda AA. Determinants of the efficacy of ADAMTS 13 levels and implications for therapy. J Clin Apher.
prophylactic granulocyte transfusions: a meta-analysis. J Clin 2009;24:115-19.
Apher. 1997;12:74. 48. Patten E. Pathophysiology of the immune system. In: Kilins J,
26. Roback JD, editor. Technical manual. 16th ed. Bethesda (MD): Jones JM, editors. Therapeutic apheresis. Arlington (VA):
American Association of Blood Banks; 2008. American Association of Blood Banks; 1983.
27. Sachs UJ, et al. Safety and efficacy of therapeutic early onset 49. Klein HG. Effect of plasma exchange on plasma constituents:
granulocyte transfusions in pediatric patients with neutropenia Choice of replacement solutions and kinetics of exchange.
and severe infections. Transfusion. 2006;46(11):1909-14. In: MacPherson JL, Kaspirisin DO, editors. Therapeutic hema-
28. Kikuta Ohto H, Nemoto K, Mochizuki K, Sano H, Ito M, pheresis, vol 2. Boca Raton (FL): CRC Press; 1985.
Suzuki H. Therapeutic transfusions of granulocytes collected 50. Cortelazzo S, Finazzi G, Ruggeri M, Vestri O, Galli M, Rodeghiero
by simple bag method for children with cancer and neutropenic F et al. Hydroxyurea for patients with essential thrombocythemia
infections: results of a single-centre pilot study. Vox Sang. and a high risk of thrombosis. N Engl J Med. 1995;332:1132-36.
2006;91:70-6. 51. Procu P, Cripe LD, Ng EW, Bhatia S, Danielson CM, Orazi A,
29. Leitner G, Panzer S, Reesink HW, Stiegler G, Fischer-Nielsen A, et al. Hyperleukocytic leukemias and leukostasis: a review of
Dickmeiss E, et al. Preparation of granulocyte concentrates by pathophysiology, clinical presentation and management. Leuk
apheresis. Vox Sang. 2010;98:567-75. Lymphoma. 2000;39:1-18.
30. McLeod BC, editor. Apheresis: principles and practice. 3rd ed. 52. Fenger-Eriksen C, Tønnesen E, Ingerslev J, Sørensen B. Mech-
Bethesda (MD): AABB Press; 2010. anisms of hydroxyethyl starch-induced dilutional coagulopa-
31. Bryant BJ, Yau YY, Byrne PJ, Stroncek DF, Leitman SF. Gravity thy. J Thromb Hemost. 2009;7:1099-105.
sedimentation of granulocytapheresis concentrates with hy- 53. National Heart, Lung, and Blood Institute. The management
droxyethyl starch efficiently removes red blood cells and retains of sickle cell disease. 4th ed. NIH Publication No. 02-2117.
neutrophils. Transfusion. 2010;50:1203-09. Bethesda (MD): National Institutes of Health; 2002.
32. Lee JH, Leitman SF, Klein HG. A controlled study of the efficacy 54. Spaete J, Patrozou E, Rich JD, Sweeney JD. Red cell exchange
of hetastarch and pentastarch in granulocyte collections by cen- transfusion for babesiosis in Rhode Island. J Clin Apher.
trifugal leukapheresis. Blood. 1995;86:4662-6. 2009;24:97-105.
33. Szczepiorkowski Z, editor. Clinical applications of therapeutic 55. Yarrish RL, Janas JS, Nosanchuk JS, Steigbigel RT, Nusbacher J.
apheresis: an evidence based approach. J Clin Apher. 2010; Transfusion malaria: treatment with exchange transfusion after
25:81. delayed diagnosis. Arch Intern Med. 1982;142:187-8.
34. Winter JL, editor. Clinical applications of therapeutic apheresis: 56. Cahill KM, Benach JL, Reich LM, Bilmes E, Zins JH, Siegel FP,
an evidence based approach. J Clin Apher. 2016; 31:149-162. et al. Red cell exchange: treatment of babesiosis in a splenec-
35. Areman EM, Loper K. Cellular therapy: principles, methods, tomized patient. Transfusion. 1981;21:193-8.
and regulations. 2nd ed. Bethesda (MD): AABB Press; 2016. 57. Pusey C, Dash C, Garrett M, Gascoigne E, Gesinde M,
36. Ipe TS, Pham HP, Williams LA. Critical updates in the 7th edition Gillanders K, et al. Experience of using human albumin solu-
of the American society for apheresis guidelines. J Clin Apher. tion 4.5% in 1195 therapeutic plasma exchange procedures.
Epub 2017 Jun 27. Transfusion Med. 2010;20(4):244-99.
37. Kintzel PE, Eastlund T, Calis KA. Extracorporeal removal of 58. Goss GA, Weinstein R. Pentastarch as partial replacement fluid
antimicrobials during plasmapheresis. J Clin Apher. 2003; for therapeutic plasma exchange: effect on plasma proteins,
18:67-70. adverse events during treatment, and serum ionized calcium.
38. Boga C, Kozanoglu I, Ozdogu H, Ozyurek E. Plasma exchange J Clin Apher. 1999;14:114-21.
in critically ill patients with sickle cell disease. Transfus Apher 59. Agreda-Vásquez GP, et al: Starch and albumin mixture as
Sci. 2007;37:17-22. replacement fluid in therapeutic plasma exchange is safe and
39. Hastings D, Patel B, Torloni AS, Mookadam F, Betcher J, Moss A, effective. J Clin Apher. 2008;23:163.
et al. Plasmapheresis therapy for rare but potentially fatal 60. Cabrera-Vásquez S, López-Salmorán J, et al. Starch and albumin
reaction to rituximab. J Clin Apher. 2009;24:28-31. mixture as replacement fluid in therapeutic plasma exchange
40. Gore EM, Jones BS, Marques MB. Is therapeutic plasma is safe and effective. J Clin Apher. 2008;23:163-7.
exchange indicated for patients with gemcitabine-induced 61. Itoh T, Minegishi M, Kudo Y, Saito N, Takahashi H, Suzuki A,
hemolytic uremic syndrome? J Clin Apher. 2009;24:209-14. et al. Predictive value of the original content of CD34+ cells for
6888_Ch18_396-416 29/10/18 5:41 PM Page 416

416 PART III Transfusion Practices

enrichment of hematopoietic progenitor cells from bone mar- 70. Doesch AO, Konstandin M, Celik S, Kristen A, Frankenstein L,
row harvests by the apheresis procedure. J Clin Apher. Hardt S et al. Effects of protein A immunoadsorption in
2006;21:176-80. patients with advanced chronic dilated cardiomyopathy. J Clin
62. Bishop MR, Anderson JR, Jackson JD, Bierman PJ, Reed EC, Apher. 2009;24:141-9.
Vose JM, et al. High-dose therapy and peripheral blood 71. Heustis DW, Morrison FS. Adverse effects of immune adsorp-
progenitor cell transplantation: effects of recombinant human tion with staphylococcal protein A columns. Transfus Med Rev.
granulocyte-macrophage colony-stimulating factor on the 1996;10:62-70.
autograft. Blood. 1994;83:610-6. 72. Edelson R, Berger C, Gasparro F, Jegasothy B, Heald P,
63. Food and Drug Administration. Current good tissue practice Wintroub B, et al. Treatment of cutaneous T-cell lymphoma
for manufacturers of human cellular and tissue-based products: by extracorporeal photochemotherapy—preliminary results.
inspection and enforcement; proposed rule. 21 C.F.R 1271. N Engl J Med. 1987;316:297-303.
Federal Register 66:1507, 2001. 73. Rossetti F, Dall’Amico R, Crovetti G, Messina C, Montini G,
64. Padley D, editor. Standards for cellular therapy product ser- Dini G, et al. Extracorporeal photochemotherapy for the treat-
vices. 4th ed. Bethesda (MD): AABB; 2010. ment of graft-vs-host disease. Bone Marrow Transplant.
65. Foundation for the Accreditation of Cellular Therapy [Internet]. 1996;18 Suppl 2:175-81.
Omaha [NE]: FACT-JACIE [2008]. International Standards for 74. Constanza-Nordin MR, Hubbell EA, O’Sullivan EJ, Johnson
Cellular Therapy Product Collection, Processing, and Admin- MR, Mullen GM, Heroux AL, et al. Successful treatment of
istration. 4th ed. Available from: factwebsite.org. heart transplant rejection with photopheresis. Transplantation.
66. Pineda AA. Immunoaffinity apheresis columns: clinical appli- 1992;53:808-15.
cations and therapeutic mechanisms of action. In: Sacher RA, 75. Malawista SE, Trock DH, Edelson RL. Treatment of rheumatoid
et al. Cellular and humoral immunotherapy and apheresis. arthritis by extracorporeal photochemotherapy: a pilot study.
Arlington [VA]: American Association of Blood Banks; 1991. Arthritis Rheum. 1991;34:646-54.
67. Felson DT, LaValley MP, Baldassare AR, Block JA, Caldwell JR, 76. Rook AH, Freundlich B, Jegasothy BV, Perez MI, Barr WG,
Cannon GW, et al. The Prosorba column for treatment of Jimenez S, et al. Treatment of systemic sclerosis with extracor-
refractory rheumatoid arthritis: a randomized, double-blind, poreal photochemotherapy. Arch Dermatol. 1992;128:337-46.
sham-controlled trial. Arthritis Rheum. 1999;42:2153-9. 77. Winters J. Complications of donor apheresis. J Clin Apher.
68. Mittelman A, Bertram J, Henry DH, Snyder HW Jr, 2006;21:132-41.
Messerschmidt GL, Ciavarella D, et al. Treatment of patients 78. Bramlage CP, Schröder K, Bramlage P, Ahrens K, Zapf A, Müller
with HIV thrombocytopenia and hemolytic uremic syndrome GA, et al. Predictors of complications in therapeutic plasma
with protein A (Prosorba column) immunoadsorption. Semin exchange. J Clin Apher. 2009;24:225-31.
Hematol. 1989;26 Suppl 1:15-8. 79. Yuan S, Ziman A, Smeltzer B, Lu Q, Goldfinger Det al.
69. Snyder HW Jr, Mittelman A, Oral A, Messerschmidt GL, Moderate and severe adverse events associated with apheresis
Henry DH, Korec S, Bertram JH, et al. Treatment of cancer donations: incidences and risk factors. Transfusion. 2010;
chemotherapy-associated thrombotic thrombocytopenic purpura/ 50(2):478-86.
hemolytic uremic syndrome by protein A immunoadsorption 80. Shemin D, Briggs D, Greenan M. Complications of TPE. J Clin
of plasma. Cancer. 1993 Mar 1;71:1882-92. Apher. 2007;22:270-6.
6888_Ch19_417-426 29/10/18 5:40 PM Page 417

Chapter 19
Cellular Therapy in the Hematopoietic
Transplant Setting
Scott A. Koepsell, MD, PhD and Denise M. Harmening, PhD, MT(ASCP)

Introduction Special Transfusion Requirements in HPC Case Study


HPC Transplantation Overview Transplantation Case Study 19-1
Accrediting Bodies Transfusion-Associated Graft-Versus- Summary Chart
AABB Host Disease Review Questions
FACT Transfusion-Transmitted Infectious References
Diseases
JACIE
ABO-Incompatible HPC
Clinical Uses of HPC Transplantation and Transplantation
Complications
Rh Transplant Incompatibility
Sources of HPC
Passenger Lymphocyte Syndrome
HPC Donor Considerations
Platelet Refractoriness
HPC Processing and Storage

OBJECTIVES
1. List and define the categories of hematopoietic progenitor cell (HPC) donors.
2. List five assessments of an HPC donor.
3. Name a registry for HPC unrelated donors.
4. State the chance of an HLA match between siblings.
5. Compare and contrast the collection of HPC-M and HPC-A.
6. Name the antigen that defines HPCs.
7. Identify three diseases treated with HPC transplantation.
8. List five tests performed on all HPC products.
9. State the temperature for frozen storage of HPCs.
10. Identify the cryoprotectant generally used for HPCs.
11. Contrast myeloablative with nonmyeloablative recipient conditioning.
12. Compare major, minor, and bidirectional ABO incompatibility between the HPC donor and the recipient.
13. Define graft-versus-host disease (GVHD).
14. Define acute versus chronic GVHD.
15. List three important conditions to prevent when transfusing blood products.
16. Select the correct ABO type(s) when transfusing red blood cells (RBCs) and plasma to an ABO-mismatched allogeneic
HPC transplant recipient.

417
6888_Ch19_417-426 29/10/18 5:40 PM Page 418

418 PART III Transfusion Practices

Introduction in North America in 1997. JACIE was established in 1999


and adopted the first edition of FACT Standards. Following
Cellular therapy is “the administration of products with the a joint review by FACT and JACIE, the second edition of the
intent of providing effector cells in the treatment of disease FACT Standards was published in 2002, with subsequent
or support of other therapy.”1 This broad definition can apply editions of Standards developed and approved by FACT
to applications ranging from injecting cells with reparative and JACIE. The Foundation for Accreditation of Cellular
potential into a damaged joint to engineering a person’s im- Therapy is characterized as a nonprofit organization for vol-
mune cells to kill cancer. The most common cellular therapy untary inspections and accreditation in the field of cellular
encountered in blood banking and transfusion medicine re- therapy. FACT has published standards that are described as
lates to the transplantation of hematopoietic progenitor evidence-based requirements for quality practice of cellular
cells (HPCs) as part of a bone marrow or peripheral blood therapies. Their accreditation cycle is 3 years. There are over
stem cell transplant. The transfusion service plays a vital role 200 facilities accredited by FACT in the United States.7
in supporting HPC transplantation activities, which is the
focus of this chapter. JACIE
JACIE, ISCT, and EBMT were created in 1999 by EBMT and
HPC Transplantation Overview the ISCT, respectively. Transplant centers may be accredited
under the guidance of international standards established by
Hematopoiesis is the process of formation and development JACIE. FACT collaborates with JACIE to develop and main-
of cells of the blood and immune system from a common tain standards for quality and medical laboratory practice in
hematopoietic stem cell.2 The tightly controlled process of cellular therapies. JACIE also offers online exams to anyone
hematopoiesis is complex and typically begins in the bone regarding FACT-JACIE clinical, collection, and processing
marrow where a small population of hematopoietic stem standards. JACIE has accredited 221 institutes in 25 coun-
cells become HPCs. These cells divide into multipotent pro- tries. Accreditation is on a 4-year cycle with the first trans-
genitors, which continue to divide and differentiate into the plant center accredited in 2004.8
various lineages of red blood cells, white blood cells, and
platelets.3 All of a person’s blood and immune cells are Clinical Uses of HPC Transplantation
derived from these HPCs. These hematopoietic progenitor and Complications
cells are functionally characterized by the capacity for self-
renewal and morphologically by a high nuclear-to-cytoplasm HPC transplantation can be used for a wide variety of indi-
ratio and expression of the surface protein CD34.2 There- cations that include both malignant and nonmalignant dis-
fore, CD34 is a very important marker measured in the orders. For example, in adults with lymphoproliferative or
laboratory by flow cytometry to assess the amount of HPCs plasma cell disorders or in children with some solid tumors,
in blood.4 HPCs can be collected, processed or manipulated, patients often donate their own HPCs. These cells are stored
stored, and ultimately transplanted to replace or regenerate and later transplanted back into a patient to rescue the bone
the bone marrow in the recipient. marrow function after high-dose chemotherapy and/or
radiotherapy. The use of one’s own HPCs for transplantation
Accrediting Bodies is referred to as autologous HPC transplantation. Autolo-
gous transplantation is the most common type of HPC
The accrediting bodies for transplant centers that collect, transplant performed in the United States.9
process, store, and infuse HPCs include: the AABB, The Foun- Malignant myeloproliferative processes are often treated
dation for Accreditation of Cellular Therapy (FACT) and with HPC transplantation using HPCs from another indi-
Joint Accreditation Committee of the International Society vidual, which is called allogeneic HPC transplantation.
for Cellular Therapy (ISCT) and European Group for Blood If the donor is an identical twin, the term syngeneic HPC
and Marrow Transplantation (EBMT) known as JACIE. transplantation can be used. Like autologous HPC trans-
plantation, allogeneic HPC transplantation also rescues
AABB bone marrow function after high-dose chemotherapy and/
The first standards publication for cellular therapy by the or radiotherapy. In addition to providing hematopoietic
AABB was published in 1996.5 Accreditation is renewable functions, transplanted allogeneic HPCs and immune
every 2 years, and to date there are 102 transplant facilities cells can recognize and attack residual tumor cells in the
with AABB accreditation in the United States. Internationally, recipient, resulting in a graft-versus-leukemia (GVL) or
21 countries hold AABB accreditation for cellular therapy.6 graft-versus-neoplasm (GVN) effect.10 Donors of allogeneic
HPC products have to be carefully selected, mostly through
FACT matching of human leukocyte antigen (HLA) types.
The Foundation for Accreditation for Cellular Therapy was Mismatches in HLA types between the donor and recipient
established in 1996 by the American Society for Blood and can lead to failure of the allogeneic HPC to engraft or result
Marrow Transplantation (ASBMT) and the ISCT with their in the donor cells recognizing the recipient as “foreign” and
first edition of Standards published. The inspection and inducing a potentially lethal immune attack on the recipi-
accreditation program based on these Standards was started ent, which is called graft-versus-host disease (GVHD).11
6888_Ch19_417-426 29/10/18 5:40 PM Page 419

Chapter 19 Cellular Therapy in the Hematopoietic Transplant Setting 419

GVHD is an immunologically mediated disease that con- of incompatible plasma, CD34-positive selection, or re-
tributes substantially to transplant-related morbidity and moval of T donor cells.14
mortality. The overall incidence of GVHD remains between The most common product currently used for both au-
30% and 50% and carries a high mortality rate.12 Acute tologous and allogeneic HPC transplantation is HPC-A.9
GVHD is characterized by skin rash, transaminitis, and HPC-A products are collected employing apheresis technol-
gastroenteritis and usually occurs within 100 days of ogy that uses an automated device to process blood and col-
HPC transplantation. Chronic GVHD includes dry eyes and lect peripherally circulating HPCs. Chemotherapeutic drugs,
mouth, joint contractures and reduced mobility, scleroderma- growth factors such as the granulocyte colony-stimulating
like changes in the skin, and lung damage. These changes factor (GCSF), filgrastim, and/or receptor agonists such as
contrast the transfusion-associated graft-versus-host disease, plerixafor are administered to donors to increase the number
which is discussed later. of circulating HPCs in the peripheral blood to a level high
Nonmalignant disease processes can also be treated with enough for collection. Plerixafor antagonizes the binding of
allogeneic HPC transplantation because donor HPCs can be stromal cell–derived factor-1 (SDF-1) to its cognate receptor
selected that have the missing trait. For example, HPC trans- CXCR4, resulting in rapid mobilization of HSC into the
plantation has been used to treat sickle cell disease and con- peripheral circulation; this mechanism is reversible and is
genital immune deficiencies. In these cases, matched donor synergistic with GCSF.15 The number of HPCs collected is
HPCs are selected from donors who do not have sickle cell monitored in the HPC-apheresis products by flow cytometry
disease or congenital immune deficiencies so that new RBCs assays that count the number of CD34-positive cells. Collec-
or immune cells that are made are functionally normal in the tions may continue until the number of CD34-positive cells
transplant recipient. reaches a level requested by the transplant physician, typi-
For both allogeneic and autologous HPC transplantation, cally at least 2 × 106 CD34-positive cells per kilogram weight
recipients must undergo conditioning regimens that usually of the recipient.13 The HPC-A product may be modified
consist of chemotherapy and/or radiation. Conditioning using the same techniques as for HPC-marrow. Mononuclear
may serve the purpose of destroying tumor cells as well as cells (MNC-A) may also be procured from apheresis dona-
creating a niche for donor HPCs to occupy. Without proper tions for use in donor lymphocyte infusions (DLIs), which
conditioning, infusion of donor HPCs may not engraft, is a form of adoptive immunotherapy geared toward graft-
especially in the allogeneic setting. Conditioning may be versus-leukemia effect.14
either myeloablative, meaning without intervention the There are characteristics unique to each graft source used
bone marrow and immune function would not recover with- for HPC transplantation (Table 19–1). In general, transplant
out HPC transplantation, or nonmyeloablative, which uses physicians have to weigh donor availability, potential ad-
reduced amounts of chemotherapy and/or radiation. vantages and disadvantages, and timing to determine the
optimal graft source for HPC transplantation.
Sources of HPC
HPC Donor Considerations
The three main sources of HPC products include umbilical
cord blood (HPC-cord blood or HPC-C), bone marrow For autologous HPC donors in the United States, no eligi-
(HPC-marrow or HPC-M), and peripheral blood stem bility requirements are mandated by the Food and Drug
cells collected by apheresis (HPC-apheresis or HPC-A).13
HPC-C is collected from the placenta and umbilical cord at
the time of delivery. Typically, the umbilical cord is cannu- Table 19–1 Commonly Used Allogeneic
lated after thorough cleansing, and the contents are collected HPC Products
by gravity drainage into standard collection bags containing
anticoagulant (e.g., CPD). The product collected may be HPC Product Advantages Disadvantages
washed to remove RBCs and/or plasma and frozen using 10% HPC-marrow Lower risk of Donation involves
DMSO (dimethyl sulfoxide), which is a cryoprotectant. GVHD than HPC- higher risk of
Frozen units are thawed and potentially washed before in- apheresis in the adverse events than
fusion to the patient.14 HPC recipient37 HPC-apheresis
and requires
HPC-M collections are performed under anesthesia in anesthesia18
an operating room using sterile technique, using a needle
to access the marrow space of the posterior iliac crest. HPC-apheresis Faster engraftment Higher risk of GVHD
Usually, around 5 mL of marrow is aspirated multiple times of neutrophils than HPC-marrow.28
and platelets than Better graft-versus-
at multiple sites until the desired collection volume is HPC-marrow in leukemia effect than
achieved, which could total between 100 and 2000 mL. the HPC recipient36 HPC-marrow38
The marrow is placed into a sterile container with anti-
coagulant as well as an electrolyte solution. Sterile filters HPC-cord blood Lower risk of acute Longer time until
GVHD in the HPC engraftment40
are employed to remove fat, bone particles, and other de- recipient.39 No risk
bris from the product. HPC-M may be modified or unmod- to the donor.
ified. Modifications may include RBC depletion, removal
6888_Ch19_417-426 29/10/18 5:40 PM Page 420

420 PART III Transfusion Practices

Administration (FDA). However, accrediting bodies such as in their peripheral blood, which also appears to have no
the FACT and AABB require that donors be tested for rele- long-term safety concerns but have been reported rarely to
vant infectious diseases.1,5 Testing for infectious diseases be associated with splenic rupture.16,17 There is essentially
can be useful for processing and storage facilities to evaluate no risk to either mother or infant in donation of HPC-cord
the risk of cross-contaminating other HPC products in their blood.
freezers. In general, autologous HPC donors have to be
healthy enough to tolerate donation, which is almost always HPC Processing and Storage
performed by apheresis collections as opposed to bone
marrow harvest. Processing must meet the regulations of the FDA in the United
For allogeneic HPC donors, the United States has similar States and should meet the requirements of the relevant ac-
requirements for both HPC-marrow and HPC-apheresis. crediting bodies. All processes involved in the handling of
Regulations require screening questionnaires, a physical HPC products are designed to ensure that the therapeutic
examination, review of relevant medical records, and appli- potential of HPCs is preserved. In general, HPC products are
cable infectious disease testing. Infectious disease testing handled in a way that is termed minimally manipulated,
includes screening tests for human immunodeficiency virus meaning that nothing is done to the HPC product that could
(HIV) type 1 and 2, hepatitis B virus (HBV), hepatitis C virus alter its relevant biological characteristics, which is to recon-
(HCV), human T-cell lymphotrophic virus (HTLV) type I stitute bone marrow function. Examples of minimal manipu-
and II, syphilis, and cytomegalovirus (CMV) (Table 19–2). lation include sterilely sampling the product for microbial
As regulations are updated and emerging infectious diseases contamination or cell counts, volume reduction, washing,
are encountered, required testing may change to protect re- cryopreservation, and thawing. Examples of processes that
cipients of HPCs. In addition, for HPC collections outside of exceed minimal manipulation include the addition of growth
the United States, different infectious disease testing may be factors or the selective removal of a certain cell population
omitted or added depending on disease risks. such as T cells in an HPC product. Processing and storage
Allogeneic donors must be tested for HLA antigens by steps for HPC products are often institution specific, using
DNA molecular methods.1 These include antigens HLA-A, protocols that ensure that the purity, potency, safety, and steril-
HLA-B, HLA-C, and HLA-DRB1. ity of the product are maintained.
Physically, both autologous and allogeneic donors of HPC Initial testing of the HPC product may consist of a com-
products must be healthy enough to tolerate the donation plete blood count, white blood cell (WBC) differential, CD34
procedure. Risks for donating HPC-marrow are due to gen- enumeration, and viability studies. Sampling is performed in
eral anesthesia and the invasive nature of the bone marrow a laminar flow hood with the use of a sterile connecting de-
harvest. Because significant blood loss can occur with vice or integral sampling tubes attached to the collection set.
HPC-marrow collections, most donors have a blood type and Cell counts are usually performed on automated instru-
antibody screen performed before collection in case a trans- ments with the differential performed manually to confirm
fusion is needed. HPC-apheresis collections are very safe, but mononuclear cells that contain the HPCs. The CD34 antigen
sometimes do require that donors have a central venous is used routinely to determine when a patient or donor
catheter placed if their venous access is not sufficient.1,5 should be collected by leukapheresis. Cellular therapy labs
HPC-apheresis donors also are exposed to drugs, such as measure CD34+ cells to determine the quantity of HPCs har-
filgrastim, that increase the number of CD34-positive cells vested by bone marrow, apheresis, or cord blood collection.
The HPCs in a sample can be quantified by flow cytometry
using antibody to the CD34 antigen. The CD34 antigen is
measured in autologous and allogeneic donors who are to be
Table 19–2 Viral Testing Requirements
collected by leukapheresis. The collection goal for an adult
for Cellular Therapy Products1
is 2 × 106 to 6 × 106 CD34 positive cells per kilogram of re-
HPC (M) and HPC (CB) Maternal cipient body weight (2–6 × 106/kg).13 Generally, the yield of
Agent* HPC (A) Donors Sample CD34+ cells is less for autologous donors than for allogeneic
HIV-1, 2 X X
donors, so more days of collection are required.
For allogeneic HPC transplantation, HPC-apheresis and
HBV X X HPC-marrow products can often be collected near the time of
HCV X X
transplant to avoid cryopreservation, which is optimal because
cryopreservation may result in increased cost and decreased
HTLV-I/II X X yield. In contrast, autologous HPC-apheresis products and
CMV X X
HPC-cord blood products are often collected and cryopre-
served for infusion at a later date. Cryopreservation usually
Syphilis X X involves the addition of a cryoprotectant such as DMSO to
Timing Up to 30 days before or Up to 7 days before or after
the HPC product, followed by freezing in a controlled-rate
7 days after collection collection freezer.18 Long-term storage of cryopreserved HPC products
is usually done in a liquid nitrogen freezer. RBCs are removed
*West Nile Virus and Trypanosoma cruzi may also be required by FACT. from the HPC product because the freezing process causes
6888_Ch19_417-426 29/10/18 5:40 PM Page 421

Chapter 19 Cellular Therapy in the Hematopoietic Transplant Setting 421

RBCs to lyse and release nephrotoxic hemoglobin and RBC (TA-GVHD). TA-GVHD is characterized by a skin rash, fever,
stroma. The product is transferred to freezing bags and a cry- liver enzyme elevation, pancytopenia, and diarrhea, and is
oprotectant such as DMSO is added to a final concentration usually fatal.23 Reduction of TA-GVHD risk is accomplished
of 5% to 10%.18 through irradiation of blood products or, more recently,
For cryopreserved HPC products, thawing is usually per- through pathogen-reduction techniques, both of which render
formed in a 37°C water bath with gentle kneading of the donor lymphocytes incapable of dividing by damaging DNA.24
product and careful manipulation to prevent damage to the Leukocyte reduction is not thought to prevent TA-GVHD. Due
bag and potential loss of the product.19 Occasionally, HPC to immunosuppression related to chemotherapeutic interven-
products are washed after thawing if the intended recipient tions as well as a decrease in cellular immunity post-HPC
is incompatible with contaminating red blood cells in the transplant, both autologous and allogeneic HPC transplant
HPC product or if DMSO toxicity is a concern. DMSO toxic- recipients are theoretically at an increased risk for TA-
ity is the most common complication of cryopreserved prod- GVHD. Transfusion services should have policies and pro-
uct administration. Patients may present with coughing, cedures in place to ensure both allogeneic and autologous
flushing, rash, nausea, vomiting, cardiovascular instability, HPC transplant receive irradiated or pathogen-reduced
and wheezing. If a patient is suspected of having an adverse blood products. (See Chapter 17, “Adverse Effects of Blood
reaction to DMSO (residual after thawing and washing), then Transfusion.”)
the rate of infusion should be tapered and antihistamines
administered.20 Typically, infusion of the thawed product Transfusion-Transmitted Infectious Diseases
occurs through a central venous catheter, with the circulat-
ing HPCs ultimately ending up in marrow space through in- A significant number of allogeneic HPC transplant recipients
teraction of the CD34-positive cells and the bone marrow develop complications from cytomegalovirus (CMV), which
stroma.21 Bedside leukocyte filters are never used because includes pneumonia, gastroenteritis, and retinitis.25 Because
HPCs would be trapped by the filter. Infusion of HPC prod- blood donors can be asymptomatic while having an active
ucts can often be complicated by reactions that are very sim- acute CMV infection, blood products may contain infectious
ilar to transfusion reactions caused by other blood products. viral particles. Blood donors with a remote CMV infection
can also transmit CMV via donated blood products because
Special Transfusion Requirements the virus can persist in a latent state in peripheral blood
leukocytes. To reduce the risk of transfusion-transmitted
in HPC Transplantation
CMV, transfusion services have issued blood products to
Recipients of HPC transplants can be challenging for transfu- HPC transplant recipients that are obtained from donors who
sion services because there are often profound alterations are seronegative for CMV and/or blood products that have
in their immune systems that can affect serologic reactivity been leukoreduced. Both strategies significantly reduce the
and compatibility testing as well as lead to an increase in risk of transfusion-transmitted CMV, although debate exists
susceptibility to complications of transfusion. In addition, as to the most efficacious approach at preventing disease
recipients of HPC transplants often have a period of hypopro- transmission.26 More recently, blood products that have been
liferative cytopenias that may require significant blood prod- treated with pathogen-reduction techniques have become
uct support or are complicated by issues of refractoriness to available that also are efficacious at reducing the risk of
blood product transfusion efficacy. For example, pure red cell transfusion-transmitted CMV.27
aplasia (PRCA) complicates approximately 3% to 29% of Many other existing and emerging transfusion-transmitted
major ABO-mismatched HPC transplants.22 It occurs mostly infectious diseases are of concern for HPC transplant recipi-
with major ABO mismatches. In this disorder, the reticulocyte ents (see Chapter 14, “Transfusion-Transmitted Diseases”).
count is less than 1% 60 days after hematopoietic transplant, In particular, two infectious agents not routinely screened for,
with presence of myeloid and megakaryocytic engraftment hepatitis E and Babesia microti, have both been transmitted
and absence of erythroid engraftment in the bone marrow. to HPC transplant recipients by blood products.28,29 Screen-
Numerous treatments have proven effective in resolving ing for Babesia can be accomplished with both nucleic acid
PRCA, including rituximab, bortezimib, plasma exchange, testing as well as serology, and though not licensed by the
and donor lymphocyte infusions (DLIs). The basis for PRCA FDA, some centers in endemic areas request Babesia-negative
in major ABO mismatches is recipient isoagglutinins (e.g., blood products, especially for HPC transplant recipients.30
anti-A, B) lysing developing donor A, B, or AB RBCs.
ABO-Incompatible HPC Transplantation
Transfusion-Associated Graft-Versus-Host Disease
HPCs lack ABO determinants on their cell surface. As a re-
During a transfusion of cellular blood products, such as red sult, HPCs can be transplanted into a recipient, regardless of
blood cells or platelets, any contaminating donor lymphocytes the recipient’s ABO type. When an ABO-incompatible HPC
are thought to be recognized and destroyed by the recipient’s transplant occurs, there are challenges depending on the
immune system. Rarely, transfused lymphocytes escape detec- type of mismatch, which is often categorized as major,
tion and are able to divide and harm the recipient’s tissues, re- minor, or bidirectional22 (Table 19–3). When a major mis-
sulting in transfusion-associated graft-versus-host disease match occurs, the HPC products are typically manipulated
6888_Ch19_417-426 29/10/18 5:40 PM Page 422

422 PART III Transfusion Practices

Table 19–3 ABO-Incompatible HPC Transplants40


Category of
Recipient Donor Mismatch Potential Complications
O A, B or AB Major Acute hemolysis of RBCs in HPC product.

A AB Major Delayed RBC production after transplant.


Delayed granulocyte and platelet production.
B AB Major
Pure red cell aplasia (PRCA).

A O Minor Acute hemolytic episode if the donor plasma has elevated isohemagglutinin titers.

B O Minor Delayed hemolysis of recipient RBCs due to passenger lymphocyte syndrome


where these lymphocytes produce isoagglutinins.
AB O, A, B Minor

A B Bidirectional Minor hemolysis of RBCs in HPC product.

B A Bidirectional Delayed hemolysis of recipient RBCs due to passenger lymphocyte syndrome


where these lymphocytes produce isoagglutinins.
Acute hemolytic episode if the donor plasma has elevated isohemagglutinin titers.

to reduce the total number of contaminating red blood cells HPC transplants. For HPC transplants with major mis-
(RBCs) to reduce hemolysis during product infusion.22 matches, RBCs of the recipient type or group O are usually
Recipients of majorly mismatched HPC transplants can be selected until the recipient antibodies that are incompatible
at risk for delayed red blood cell production, because recip- with HPC product are no longer detected. For HPC trans-
ient’s antibodies can lyse newly produced ABO-incompatible plants with major mismatches, plasma-containing products
RBCs (e.g., recipient is type O and donor is type A). Approx- that match the HPC donor or are group AB are usually
imately 20% to 25% of HPC transplants have major ABO selected. In HPC transplants with minor mismatches, RBCs
incompatibility.22 For minor mismatches, the plasma con- are selected that match the HPC donor or are group O, and
tent of HPC products can be reduced to avoid donor plasma plasma-containing products are selected that match the recip-
antibodies from binding to and causing destruction of recip- ient until recipient RBCs are no longer detected or serocon-
ient RBCs (e.g., recipient is type A, and donor is type O). version has taken place. For HPC transplants that have
Approximately 20% to 25% HPC transplants have a minor bidirectional incompatibility (Table 19–3) or in difficult cases,
ABO incompatibility.22 group O RBCs and AB plasma can be used until the recipient’s
In bidirectional ABO mismatches, acute hemolysis may original RBCs and antibodies are no longer detected.22
occur by ABO-incompatible antibodies in the transplant A more prudent approach to transfusion in ABO
product lysing recipient red blood cells and/or by ABO- mismatches regarding HPC transplants is to consider three
incompatible antibodies in the recipient lysing donor phases of the transplant process: (1) pretransplant (Phase I),
red blood cells (e.g., recipient is type A and donor is type B). (2) transplant (Phase II), (3) post-transplant (Phase III).13
Approximately 1% to 5% of HPC transplants have a bidirec- In a major ABO mismatch, the post-transplant phase is
tional ABO incompatibility.22 divided into demonstration of recipient antibodies or without
Recipients of ABO-incompatible HPC transplants also recipient antibodies in the reverse grouping. If the patient is
demonstrate discrepancies between forward and reverse ABO still demonstrating native isoagglutinins in reverse grouping,
typing. In general, forward typing usually switches from the the recipient should be transfused with RBCs of the recipient
recipient’s original ABO group to the HPC donor’s ABO type and platelets/fresh-frozen plasma (FFP) of the donor
group. Reverse typing usually switches from the recipient’s type.13 However, if the recipient is not demonstrating native
original type to a type that is permissive of both the recipient isoagglutinins in reverse grouping, then they should receive
and the donor’s ABO group. For example, a group A recipient RBCs, platelets, and FFP of the donor type.13
transplanted with a group B HPC product often will have a In a minor ABO mismatch, the post-transplant phase is
forward typing that transitions from A to B. Mixed field divided into recipient ABO antigens that are demonstrable
reactions in the forward ABO typing can sometimes be the or not demonstrable in forward grouping.13 If the recipient’s
only indication that the patient is an HPC transplant. There- RBCs are present in forward grouping, the patient should
fore, it is important to be able to correctly identify mixed receive RBCs of the donor type and platelets/FFP of the re-
field reactions and investigate the underlying cause. cipient type. However, if the patient is in the post-transplant
Transfusion services should have policies in place on how phase and their native RBC antigens are not present, the
to best pick blood products in the setting of ABO-incompatible patient should receive all products of the donor type.
6888_Ch19_417-426 29/10/18 5:40 PM Page 423

Chapter 19 Cellular Therapy in the Hematopoietic Transplant Setting 423

In the bidirectional ABO mismatch, the post-transplant of antibody production, with ABO or Rhesus antigens more
phase is divided into recipient antibodies and antigens present frequently encountered. Hemolysis usually occurs 9 to
or not present. If the recipient’s antigens and antibodies 16 days post-transplant.31 In addition to laboratory evidence
are present, then they should receive type O RBCs and AB of hemolysis, the direct antiglobulin test is usually positive,
platelets/FFP; if, however, they are in the post-transplant with eluates showing specificity for the implicated antigen.
phase and their antibodies and antigens are not present, then Transfusion support may include providing RBCs that are
they can receive products of the donor type. (See Table 19-4.) antigen-negative for the target of PLS.

Rh Transplant Incompatibility Platelet Refractoriness

An incompatibility can also occur with the D antigen in Candidates for and recipients of HPC transplants can have a
HPC transplants. In a major Rh mismatch, the recipient is poor response to platelet transfusions due to both immune and
Rh-negative and the donor is Rh-positive. In a minor Rh nonimmune causes.32 Immune-mediated platelet refractoriness
mismatch, the recipient is Rh-positive and the donor is is usually due to the recipient making antibodies against HLA
Rh-negative. In both cases the recipient should receive or human platelet antigens (HPA). When platelets are trans-
Rh-negative RBCs, platelets, and FFP. In a major Rh mismatch, fused that contain antigens recognized by recipient antibodies,
the recipient can be sensitized to form anti-D. In a minor Rh platelets are quickly removed from circulation, resulting in
mismatch, the donor may have preformed anti-D, necessitat- post-transfusion platelet counts that are similar to pretransfu-
ing Rh-negative products for transfusion as the donor anti-D sion levels when collected within 1 hour of transfusion.33 Non-
can be passively transferred to the recipient. Hows et al.31 immune platelet refractoriness may be due to splenomegaly,
reported three out of seven Rh D-positive patients who sepsis, fever, disseminated intravascular coagulation, medica-
received Rh D-negative grafts (minor mismatch) developed tions, and graft-versus-host disease.34
donor-derived anti-D persisting up to 1 year after transplant. Immune-mediated platelet refractoriness can be overcome
However, hemolysis was restricted to only one patient. by the transfusion service selecting donor platelets that match
the HLA type of the recipient or by selecting donor platelets
Passenger Lymphocyte Syndrome that are negative for the antibodies detected in the recipient35
(refer to Chapter 23, “The HLA System”). Alternatively, recip-
When HPC transplant recipients have RBC antigens that the ient plasma can be “crossmatched” with donor platelets, theo-
HPC donor does not have, the potential for passenger lym- retically selecting for platelets that are antigen-negative for
phocyte syndrome (PLS) exists. The development of PLS is significant recipient antibodies, with occasional success.36
thought to result from HPC donor lymphocytes that recog- Nonimmune causes of platelet refractoriness are often difficult
nize the foreign RBC antigens in the recipient, with the to overcome, requiring communication between the transfu-
subsequent manufacturing of antibodies that can result in sion service and the clinical teams caring for HPC transplant
hemolysis. Theoretically, any RBC antigen can be the target recipients regarding expectations and platelet availability.

Table 19–4 Transfusion Support for Patients Receiving ABO Incompatible HPC Transplant40
Type of Incompatibility Transplant Stage RBC FFP PLT
Major Incompatibility Phase I Recipient ABO Recipient ABO Recipient ABO

Phase II Recipient ABO Donor ABO Donor ABO

Phase III Donor ABO Donor + ABO Donor + ABO

Minor Incompatibility Phase I Donor ABO Recipient ABO RecipientABO

Phase II Donor ABO Recipient ABO Recipient ABO

Phase III Donor ABO Donor* ABO Donor* ABO

Bidirectional Incompatibility Phase I Group O Group AB Group AB

Phase II Group O Group AB Group AB

Phase III Donor ABO Donor # ABO Donor # ABO

+ = recipient is not demonstrating native isoagglutinins in reverse grouping.


* = recipient RBCs are not present in forward grouping.
# = Recipient antigens and antibodies are not present.
Please note: If Group AB platelets are not available, then consider using ABO matched to recipient blood group with the plasma volume reduced to remove unwanted antibodies.
6888_Ch19_417-426 29/10/18 5:40 PM Page 424

424 PART III Transfusion Practices

CASE STUDY The patient’s ABO testing for specific days post-
transplant is shown below.
Case Study 19-1
Day Post-transplantation Anti-A Anti-B A Cells B Cells
A 45-year-old man is diagnosed with non-Hodgkin’s lym- Day 117 0 1+ (mf) 1+ 0
phoma. He types as B, Rh-positive. His brother is found to Day 147 0 wk (mf) 2+ 0
be HLA matched but is A, Rh-positive. The brother Day 156 0 0 1+ 0
donates HPC-A, which is transplanted in the patient on Day 200 wk (mf) 0 0 0
January 25, 2010. The patient requires long-term transfu- mf = mixed field
sion support and receives 47 RBCs, 56 platelets apheresis, 3. What is the interpretation of his ABO group on
16 FFP, and 6 cryoprecipitate. day 147?
1. Which blood group(s) should be selected for the RBC 4. What is the blood group interpretation on day 200?
transfusions? On day 156?
2. For FFP and platelets, which blood type(s) should be
selected?

SUMMARY CHART
 There are many different types of cellular therapies,  For allogeneic HPC transplantation, matching of
with HPC transplantation the most commonly en- donor and recipient HLA types is important to prevent
countered by transfusion services. complications such as graft failure and graft-versus-
 HPCs are derived from hematopoietic stem cells, ex- host disease.
press CD34 antigen, and can reconstitute the function  HPC products are highly regulated and undergo testing
of the bone marrow. for safety, purity, and potency that includes infectious
 HPCs can be obtained from cord blood, bone marrow disease testing, CD34 cell counts, viability, and sterility.
aspirates, or the peripheral blood using apheresis tech-  HPCs can be frozen and stored for later use.
nology after pharmacological interventions.  HPC products can be manipulated to reduce contam-
 HPCs can be collected from patients for self-use to re- inating RBCs, plasma, or the cryoprotectant DMSO.
cover bone marrow function after chemotherapy and/  HPC transplant recipients present unique challenges to
or radiation in a process called autologous HPC the transfusion service. including transfusion-transmitted
transplantation. infectious diseases, transfusion-associated graft-versus-
 HPCs can be collected from donors and transplanted host disease, ABO compatibility issues, passenger lym-
into patients to recover bone marrow function after phocyte syndrome, and platelet refractoriness.
chemotherapy and/or radiation in a process called
allogeneic HPC transplantation.

3. Which is an advantage of an HPC transplant using um-


Review Questions
bilical cord blood as the HPC source?
1. When an HPC donor is unrelated to the recipient of an a. Recipient weight of no concern
HPC transplantation, the transplant is categorized as: b. Donor screening and testing abbreviated
c. Higher risk of GVHD
a.Allogeneic
d. No significant risk to the donor or mother
b.Autologous
c.Syngeneic 4. The recommended minimum number of CD34+ cells re-
d.Hematopoietic quired in an HPC-apheresis collection to ensure timely
engraftment is:
2. Stem cells from HPC donors may be mobilized with:
a. 2 × 102 CD34+ cells/kg
a. Plerixafor
b. 2 × 104 CD34+ cells/kg
b. Filgrastim (GCSF)
c. 2 × 106 CD34+ cells/kg
c. Chemotherapy
d. 2 × 108 CD34+ cells/kg
d. All of the above
6888_Ch19_417-426 29/10/18 5:40 PM Page 425

Chapter 19 Cellular Therapy in the Hematopoietic Transplant Setting 425

5. The cellular marker used to quantify the collection of 5. AABB. Standards for cellular therapy services. 7th ed. AABB;
HPCs using flow cytometry is: 2015.
6. AABB [Internet]: Bethesda: AABB. AABB Accredited Hematopoi-
a. CD4 etic Progenitor Cell (HPC) Facilities [2018]. www.aabb.org/sa/
b. CD33 facilities/celltherapy/pages/hpcaccrfac.aspx.
c. CD34 7. FACT [Internet]: Omaha: FACT. Why You Should Become a
d. CD59 FACT Accredited Organization? [2014]. Available from: www
.factwebsite.org/accreditation.
6. An A patient received an HPC transplant from a B donor. 8. EBMT [Internet]: JACIE Acreddited Centers [2018]. Available
What type of ABO mismatch does this represent? from: www.jacie.org/accredited-centres.
9. Pasquini MC ZX. [Internet]. Current uses and outcomes of
a. Major hematopoietic stem cell transplantation: CIBMTR Summary
b. Minor Slides. Milwaukee (WI): Center for International Blood &
c. Bidirectional Marrow Transplant Research; 2015. Available from: http://www
d. Any of the above .cibmtr.org.
10. Singh AK, McGuirk JP. Allogeneic stem cell transplantation: a
7. Which of the following terms describe an HPC trans- historical and scientific overview. Cancer Res. [Internet]. 2016
plant where donor and recipient are the same person? Nov. Available from: http://cancerres.aacrjournals.org/content/
76/22/6445.full.
a. Allogeneic 11. Cooke KR, Luznik L, Sarantopoulos S, Hakim FT, Jagasia M,
b. Autologous Fowler DH, et al. The biology of chronic graft-versus-host
c. Syngeneic disease: a task force report from the National Institutes of
d. Hematopoietic Health Consensus Development Project on Criteria for Clinical
Trials in Chronic Graft-versus-Host Disease. Biol Blood Marrow
8. Three weeks after sustaining a car accident that required Transplant. 2017 Feb;23(2):211-34. Epub 2016 Oct 13.
emergency transfusion of blood products for resuscita- 12. Wu J, Chen Y, Hageman L, Sun C, Francisco LF, Ness EC,
et al. Long-term morbidity and mortality experienced by chronic
tion, an allogeneic HPC transplant recipient developed myeloid leukemia (CML) patients after allogeneic hematopoietic
a fever, erythematous skin rash, diarrhea, and cytope- cell transplantation (HCT)– a report from BMTSS-2. Blood.
nias, which ultimately were fatal. What intervention [Internet] 2016 [cited 2018 Jan 31];128(22):823. http://
may have prevented this outcome? www.bloodjournal.org/content/128/22/823.
13. Fung MK, Eder AF, Spitalnik SL, Westhoff CM. Technical
a. The use of leukoreduced blood products
manual. 19th ed. Bethesda (MA): AABB; 2017.
b. The use of irradiated of blood products 14. AABB, America’s Blood Centers, the American Association of
c. The use of CMV-negative blood products Tissue Banks, the American Red Cross, the American Society
d. The use of washed blood products for Apheresis, the American Society for Blood and Marrow
Transplantation, the College of American Pathologists, the
9. What common cryoprotectant is added to HPC products Cord Blood Association, the Foundation for the Accreditation
for freezing? of Cellular Therapy, ICCBBA, the International Society for
Cellular Therapy, the Joint Accreditation Committee of ISCT
a. Dimethyl sulfoxide
and EBMT, the National Marrow Donor Program, and Netcord
b. Polyethylene glycol [Internet]. Bethesda (MA). AABB (2016 Jan]. Circular of In-
c. Glycerol formation for the use of cellular therapy products. Available
d. Normal saline from: http://www.aabb.org/aabbcct/coi/Documents/CT-Circular-
of-Information.pdf.
10. An O patient received an HPC transplant from a B donor. 15. Uy G, Rettig MP, Cashen AF. Plerixafor, a CXCR4 antagonist
What type of ABO mismatch does this represent? for the mobilization of hematopoietic stem cells. Exp Opin Biol
Ther. 2008;8(11):1797-1804.
a. Major
16. Miller JP, Perry EH, Price TH, Bolan CD Jr., Karanes C, Boyd TM,
b. Minor et al. Recovery and safety profiles of marrow and PBSC donors:
c. Bidirectional experience of the National Marrow Donor Program. Biol Blood
d. Any of the above Marrow Transplant. 2008;1 Suppl:29-36.
17. Pulsipher MA, Chitphakdithai P, Logan BR, Navarro WH,
References Levine JE, Miller JP, et al. Lower risk for serious adverse events
and no increased risk for cancer after PBSC vs BM donation.
1. FACT-JACIE. International standards for hematopoietic cellular Blood. 2014;123(23):3655-63.
therapy product collection, processing, and administration. 18. Lecchi L, Giovanelli S, Gagliardi B, Pezzali I, Ratti I, Marconi
6th ed. Omaha (NE): FACT-JACIE; 2015. M. An update on methods for cryopreservation and thawing of
2. Weissman IL, Shizuru JA. The origins of the identification and hemopoietic stem cells. Transfus Apher Sci. 2016;54(3):324-36.
isolation of hematopoietic stem cells, and their capability to 19. Khuu HM, Cowley H, David-Ocampo V, Carter CS, Kasten-
induce donor-specific transplantation tolerance and treat auto- Sportes C, Wayne AS, et al. Catastrophic failures of freezing
immune diseases. Blood. 2008;112(9):3543-53. bags for cellular therapy products: description, cause, and con-
3. Shizuru JA, Negrin RS, Weissman IL. Hematopoietic stem and sequences. Cytotherapy. 2002;4(6):539-49.
progenitor cells: clinical and preclinical regeneration of the 20. Nielsen JS, McNagny KM. CD34 is a key regulator of
hematolymphoid system. Annu Rev Med. 2005;56:509-38. hematopoietic stem cell trafficking to bone marrow and mast
4. Ngoma A, Saito S, Ohto H, Ikeda K, Yasuda H, Kawabata K, cell progenitor trafficking in the periphery. Microcirculation.
et al. CD34+ cell enumeration by flow cytometry: a comparison 2009;16(6):487-96.
of systems and methodologies. Arch Pathol Lab Med. 2011; 21. Shu Z, Heimfeld S, Gao D. Hematopoietic SCT with cryopre-
135(7):909-14. served grafts: adverse reactions after transplantation and
6888_Ch19_417-426 29/10/18 5:40 PM Page 426

426 PART III Transfusion Practices

cryoprotectant removal before infusion. Bone Marrow Trans- 32. Rioux-Masse B, Cohn C, Lindgren B, Pulkrabek S, McCullough
plant. 2014;49(4):469-76. J. Utilization of cross-matched or HLA-matched platelets for
22. Daniel-Johnson J, Schwartz J. How do I approach ABO- patients refractory to platelet transfusion. Transfusion. 2014;
incompatible hematopoietic progenitor cell transplantation? 54(12):3080-7.
Transfusion. 2011;51:1143-49. 33. Luban NL, McBride E, Ford JC, Gupta S. Transfusion medicine
23. Kopolovic I, Ostro J, Tsubota H, Lin Y, Cserti-Gazdewich CM, problems and solutions for the pediatric hematologist/oncologist.
Messner HA, et al. A systematic review of transfusion-associated Pediatr Blood Cancer. 2012;58(7):1106-11.
graft-versus-host disease. Blood. 2015;16;126(3):406-14. 34. Hod E, Schwartz J. Platelet transfusion refractoriness. Br
24. Pohler P, Muller M, Winkler C, Schaudien D, Sewald K, J Haematol. 2008;142(3):348-60.
Muller TH, et al. Pathogen reduction by ultraviolet C light 35. Kopko PM, Warner P, Kresie L, Pancoska C. Methods for
effectively inactivates human white blood cells in platelet the selection of platelet products for alloimmune-refractory
products. Transfusion. 2015;55(2):337-47. patients. Transfusion. 2015;55(2):235-44.
25. Boeckh M, Nichols WG, Papanicolaou G, Rubin R, Wingard JR, 36. Holtick U, Albrecht M, Chemnitz JM, Theurich S, Skoetz N,
Zaia J. Cytomegalovirus in hematopoietic stem cell transplant Scheid C, et al. Bone marrow versus peripheral blood allogeneic
recipients: current status, known challenges, and future strate- haematopoietic stem cell transplantation for haematological
gies. Biol Blood Marrow Transplant. 2003;9(9):543-58. malignancies in adults. Cochrane Database Syst Rev. 2014;
26. Vamvakas EC. Is white blood cell reduction equivalent to anti- 20(4):CD010189.
body screening in preventing transmission of cytomegalovirus 37. Byrne M, Savani BN, Mohty M, Nagler A. Peripheral blood stem
by transfusion? A review of the literature and meta-analysis. cell versus bone marrow transplantation: a perspective from
Transfus Med Rev. 2005;19(3):181-99. the Acute Leukemia Working Party of the European Society
27. Roback JD, Conlan M, Drew WL, Ljungman P, Nichols WG, for Blood and Marrow Transplantation. Exp Hematol. 2016;
Preiksaitis JK. The role of photochemical treatment with amoto- 44(7):567-73.
salen and UV-A light in the prevention of transfusion-transmitted 38. Rocha V, Wagner JE Jr., Sobocinski KA, Klein JP, Zhang MJ,
cytomegalovirus infections. Transfus Med Rev. 2006;20(1):45-56. Horowitz MM, et al. Graft-versus-host disease in children who
28. Fang DC, McCullough J. Transfusion-transmitted Babesia have received a cord-blood or bone marrow transplant from
microti. Transfus Med Rev. 2016;30(3):132-38. an HLA-identical sibling. Eurocord and International Bone
29. van der Eijk AA, Pas SD, Cornelissen JJ, de Man RA. Hepatitis Marrow Transplant Registry Working Committee on Alterna-
E virus infection in hematopoietic stem cell transplant recipi- tive Donor and Stem Cell Sources. N Engl J Med. 2000;
ents. Curr Opin Infect Dis. 2014;27(4):309-15. 342(25):1846-54.
30. Moritz ED, Winton CS, Tonnetti L, Townsend RL, Berardi VP, 39. Laughlin MJ, Eapen M, Rubinstein P, Wagner JE, Zhang MJ,
Hewins ME, et al. Screening for Babesia microti in the U.S. Champlin RE, et al. Outcomes after transplantation of cord
blood supply. N Engl J Med. 2016;375(23):2236-45. blood or bone marrow from unrelated donors in adults with
31. Hows J, Beddow K, Gordon-Smith E, Branch DR, Spruce W, leukemia. N Engl J Med. 2004;351(22):2265-75.
Sniecinski I, et al. Donor-derived red blood cell antibodies and 40. Booth GS, Gehrie EA, Bolan CD, Savani BN. Clinical Guide to
immune hemolysis after allogeneic bone marrow transplanta- ABO-Incompatible Allogeneic Stem Cell Transplantation Biol
tion. Blood. 1986;67(1):177-81. Blood Marrow Transplant. 2013; 19:1152-58.
6888_Ch20_427-440 29/10/18 5:39 PM Page 427

Chapter 20
Hemolytic Disease of the Fetus
and Newborn (HDFN)
Meghan Delaney, DO, MPH

Introduction Management of the Fetus Prevention


Etiology Fetal Ultrasound Selection of RBCs for females
Pathogenesis Invasive Monitoring—Cordocentesis Rh Immune Globulin
HDFN Caused by ABO and Amniocentesis Case Studies
HDFN Caused by RBC Intrauterine Transfusion Case 20-1
Alloimmunization Management of the Infant Case 20-2
Hemolysis, Anemia, and Erythropoiesis Cord Blood Testing Case 20-3
Bilirubin Exchange Transfusion Case 20-4
Diagnosis Simple Transfusions Summary Chart
HDFN Caused by ABO Phototherapy Review Questions
HDFN Caused by RBC Intravenous Immune Globulin References
Alloimmunization

OBJECTIVES
1. State the definition and characteristics of hemolytic disease of the fetus and newborn (HDFN).
2. Describe the role of the technologist in the diagnosis and clinical management of HDFN.
3. Compare and contrast ABO versus HDFN due to RBC alloimmunization in terms of:
• Pathogenesis
• Incidence
• Blood types of mother and infant
• Severity of disease
• Laboratory data: anemia, direct antiglobulin test (DAT), and bilirubin
• Prevention and treatment
4. Define Rh immune globulin and describe its function.
5. Identify the approach to dosing Rh immune globulin.
6. List the tests used for detecting fetomaternal hemorrhage.
7. Outline the protocol for testing maternal and cord blood in cases of suspected HDFN.
8. Describe the RBC selection requirements for intrauterine or exchange transfusions.

Introduction can be stimulated to form RBC antibodies naturally (ABO),


by previous pregnancy, or transfusion (RBC alloimmuniza-
Hemolytic disease of the fetus and newborn (HDFN) is the tion). Before the advent of Rh immune globulin (RhIG),
destruction of the red blood cells (RBCs) of a fetus and about 95% of the cases of HDFN were caused by maternal
neonate by antibodies produced by the mother. The mother antibodies directed against the Rh antigen D (RhD). The

427
6888_Ch20_427-440 29/10/18 5:39 PM Page 428

428 PART III Transfusion Practices

incidence of the disease caused by anti-D has decreased since group O individuals are most likely to form high-titered
1968 with the introduction of RhIG. Despite the decrease in IgG anti-ABO antibodies, ABO HDFN is nearly always lim-
HDFN, RhD continues to remain an important cause of in- ited to A or B infants of group O mothers with potent anti-
compatibility, although its frequency has been equaled or A,B antibodies. Epidemiologically, clinically significant ABO
surpassed by other RBC antibody specificities.1,2 HDFN occurs most frequently in group O mothers who have
The initial diagnosis of maternal RBC alloimmunization a group A infant in the white populations and group B
is serologic. After detection, the risk stratification and man- infants in the black population and appears to be more likely
agement of HDFN have been improved with advances in when these antibody titers are high (≥512).6 ABO HDFN can
technology. Ultrasonography, Doppler assessment of middle occur in the first pregnancy and in any, but not necessarily
cerebral artery peak systolic velocity, cordocentesis, fetal all, subsequent pregnancies because it does not depend on
DNA analysis from amniotic fluid or maternal plasma, and previous foreign RBC stimulation. Tetanus toxoid adminis-
intravascular intrauterine transfusion have greatly increased tration and helminth parasite infection during pregnancy
the success of accurately diagnosing and adequately treating have been linked to the production of high-titered IgG ABO
this disease. This chapter will discuss the disease process, antibodies and severe HDFN.
diagnosis, therapy, and outcomes of HDFN. The typically mild course of ABO HDFN is related to the
poor development of ABO antigens on fetal RBCs. ABO anti-
Etiology gens are not fully developed until after the first year of life.
Group A infant RBCs are serologically more similar to
Although the clinical findings in the fetus and newborn were A2 adult cells, with group A2 infant RBCs much weaker. The
noted as early as the 17th century, it was not until 1939 that weakened A antigen on fetal and neonatal RBCs is more
Levine and Stetson reported a transfusion reaction from readily demonstrable with human than with monoclonal
transfusing a husband’s blood to a postpartum woman. They anti-A reagents. As expected, group A2 infants are less likely
postulated that the mother had been immunized to the fa- to have ABO HDFN.
ther’s antigen through fetomaternal hemorrhage (FMH). The laboratory findings and clinical characteristics of the
The antigen was later identified as RhD. infant affected by ABO HDFN differ from HDFN caused by
Maternal RBC alloimmunization can be caused by previ- anti-D and other RBC alloantibodies (Table 20–1).
ous pregnancy or previous transfusion. Some authors suggest Microspherocytes and increased RBC fragility are charac-
significant impact of previous transfusion on development teristic. Like other forms of HDFN, the severity of the disease
of future HDFN.3 However, in a large international cohort of is independent of the presence of a positive DAT result or
infant-mother pairs that had a severe course of HDFN, most demonstrable anti-A, anti-B, or anti-A,B in the eluate of the
maternal alloimmunization (83%) was due to previous preg- infant’s RBCs. ABO HDFN causes a bilirubin peak at 1 to
nancy, while only 4% was due to previous transfusion and 3 days, which is later than with HDFN caused by other
14% was unable to be determined.4 antibody specificities. Phototherapy is usually sufficient for
HDFN is caused by the destruction of the fetal RBCs by slowly rising bilirubin levels. Rarely, when there are rapidly
antibodies produced by the mother. Only antibodies of the increasing bilirubin levels, IVIG or exchange transfusion
immunoglobulin G (IgG) class are actively transported with group O RBCs may be required. The serious conse-
across the placenta via Fc receptors; other immunoglobulin quences seen with other causes of HDFN, such as stillbirth,
classes, such as IgA and IgM, are not.5 Most IgG antibodies
are directed against bacterial, fungal, and viral antigens, so
the transfer of IgG from the mother to the fetus is beneficial.
In the case of HDFN, the antibodies are directed against the Table 20–1 Comparison of ABO Versus
blood group antigens on the fetal RBCs that were inherited RhD HDFN
from the father.
Characteristic ABO RhD

Pathogenesis First pregnancy can be Yes Rare


affected
HDFN Caused by ABO Disease predicted by titers No Yes

As the incidence of HDFN caused by RhD has declined, ABO Causative antibody IgG Yes (anti-A,B) Yes (anti-D, etc.)
incompatibility has become the most common cause of Bilirubin level at birth Normal range Elevated
HDFN. Statistically, mother and infant are ABO-incompatible
in one in every five pregnancies. Maternal ABO antibodies Intrauterine transfusion None Sometimes
that are IgG can cross the placenta and attach to the ABO needed
antigens of the fetal RBCs. ABO antibodies are present in Anemia at birth No Yes
the plasma of all individuals whose RBCs lack the corre-
sponding antigen (see Chapter 6 “The ABO Blood Group Phototherapy beneficial Yes Yes
System”). These antibodies, also called isohemagglutinins, Exchange transfusion needed Rare Uncommon
result from environmental stimulus in early life. Because
6888_Ch20_427-440 29/10/18 5:39 PM Page 429

Chapter 20 Hemolytic Disease of the Fetus and Newborn (HDFN) 429

hydrops fetalis, and kernicterus, are extremely rare in ABO Maternal Factors
induced HDFN. The ability of individuals to produce antibodies in response
to antigenic exposure varies, depending on complex genetic
HDFN Caused by RBC Alloimmunization factors that are not entirely defined.8 In Rh-negative individ-
uals who are transfused with 200 mL of RhD-positive RBCs,
There are several factors that affect maternal RBC alloimmu-
approximately 85% respond and form anti-D.9 Nearly all of
nization and severity of HDFN, including antigenic expo-
the nonresponders will fail to produce anti-D even with
sure, maternal immune system factors, antibody specificity,
repeated exposure to RhD-positive blood. If RhIG is not
and influence of the ABO group.
administered to an RhD-negative mother, the risk of immu-
Fetomaternal Hemorrhage nization is only about 16% after an RhD-positive pregnancy.
Immunoglobulin class and subclass of the maternal
Previous pregnancy with FMH is the leading cause of maternal
antibody affects the severity of the HDFN. Of the im-
alloimmunization. Transplacental hemorrhage of fetal RBCs
munoglobulin classes (i.e., IgG, IgM, IgA, IgE, and IgD),
into the maternal circulation occurs in most women, but it is
only IgG is transported across the placenta. The active
usually a very small amount (0.5 mL in 93% of women).7 Large-
transport of IgG begins in the second trimester and contin-
volume FMH of ≥30 mL occurs in only 3 of 1,000 women.
ues until birth. The IgG molecules are transported via the
Interventions such as amniocentesis and chorionic villus
Fc portion of the antibodies (see Chapter 3, “Fundamentals
sampling and trauma to the abdomen can increase the risk of
of Immunology”). Of the four subclasses of IgG antibody,
FMH. At delivery, the incidence is more than 50%; this is the
IgG1 and IgG3 are more efficient in RBC intravascular he-
time the placenta separates from the uterus, and fetal RBCs
molysis than are IgG2 and IgG4.10 All subclasses of IgG are
can enter the maternal circulation (Fig. 20–1).
transported across the placenta.

RBC Antibody Specificity


Hemorrhage of D-positive fetal RBCs into D-negative mother
Of all the RBC antigens, RhD is the most antigenic. The
Maternal antibody formed against paternally inherited D antigen common antigens in the Rh system (C, E, and c) are also
potent immunogens and have been associated with moder-
Next D-positive pregnancy ate to severe cases of HDFN (Table 20–2) Anti-E and anti-c
have caused severe HDFN that required intervention and
Placental passage of maternal IgG anti-D
treatment.
Maternal antibody attaches to fetal RBCs Of the non–Rh system antibodies, anti-Kell is considered
the most clinically significant in its ability to cause HDFN.
Hemolysis of fetal RBCs
Kell blood group antigens are present on immature erythroid
cells in the fetal bone marrow, so severe anemia occurs not
Immune only by destruction of circulating RBCs but also by destruc-
+ System
tion of precursors.11 Because of the impact of anti-K on fetal
RBC precursors, the anti-K titer is less predictive of severe
+ fetal anemia, and thus, all pregnant women with anti-K RBC
antibodies should be followed closely for evidence of HDFN.

Table 20–2 Antibodies Identified in


Prenatal Specimens That Can
Cause of HDFN
Common Rare Never
HDFN
Anti-D Anti-Fya Anti-Lea

Anti-D + C Anti-s Anti-Leb

Anti-D + E Anti-M Anti-I

Anti-C Anti-N Anti-IH

Anti-E Anti-S Anti-P1

Anti-c Anti-Jka

Anti-e
Figure 20–1. Pathogenesis of hemolytic disease of the fetus and newborn
caused by maternal alloimmunization and incompatibility between the fetus and Anti-K
mother.
6888_Ch20_427-440 29/10/18 5:39 PM Page 430

430 PART III Transfusion Practices

Influence of ABO Group successfully, although many suffer permanent consequences.


When the mother is ABO-incompatible with the fetus (major In a large cohort study of children treated with intrauterine
incompatibility), the incidence of detectable fetomaternal transfusion (IUT, below), those with severe hydrops were
hemorrhage is decreased. Investigators noted many years ago the most likely to have severe long-term neurological
that the incidence of D immunization is less in mothers with impairment.12
major ABO incompatibility with the fetus. This apparent pro- The process of RBC destruction continues after birth as
tection from RhD immunization is likely due to the clearing long as maternal antibody persists in the newborn infant’s
and/or hemolysis of ABO-incompatible RhD-positive fetal circulation. The rate of RBC destruction after birth de-
RBCs in the mother’s circulation before the RhD antigen can creases because no additional maternal antibody is entering
be recognized by her immune system. the infant’s circulation through the placenta. However, IgG
is distributed both extravascularly and intravascularly and
Hemolysis, Anemia, and Erythropoiesis has a half-life of 25 days, so antibody binding and hemol-
ysis of RBCs can continue for several days to weeks after
Once the mother is immunized to a RBC antigen, all sub- delivery. There are three different phases of anemia caused
sequent offspring who inherit the antigen will potentially by HDFN: early (within 7 days of birth) due to antibody-
be affected. The maternal antibody crosses the placenta and mediated hemolysis; late hemolytic anemia (2 weeks or
binds to the fetal antigen-positive cells (Fig. 20–1). Hemol- more after birth) due to continued hemolysis, the expand-
ysis occurs when maternal IgG attaches to specific antigens ing intravascular compartment, and natural decline of
of the fetal RBCs. The antibody-coated cells are removed hemoglobin levels; and late hyporegenerative anemia due
from the circulation by the macrophages of the fetal spleen. to marrow suppression as a result of transfusions and IUT,
The rate of RBC destruction depends on antibody titer and antibody destruction of RBC precursors, and deficiency of
on the number of antigenic sites on the fetal RBCs. Destruc- erythropoietin (Table 20–3).13
tion of fetal RBCs and the resulting anemia stimulate the
fetal bone marrow to produce RBCs at an accelerated Bilirubin
rate, even to the point that immature RBCs (erythroblasts)
are released into the circulation. The term erythroblastosis RBC destruction releases hemoglobin, which is metabolized
fetalis was used to describe this finding. When the bone to bilirubin in different metabolic stages. Indirect bilirubin is
marrow fails to produce enough RBCs to keep up with unconjugated and does not dissolve in water. It travels
the rate of RBC destruction, erythropoiesis outside the through the bloodstream to the liver to be conjugated and
bone marrow is increased in the hematopoietic tissues of rendered water soluble (direct bilirubin) and is excreted
the fetal spleen and liver. These organs become enlarged through the gastrointestinal tract. The fetal liver is not
(hepatosplenomegaly), resulting in portal hypertension and able to metabolize the indirect bilirubin. During pregnancy,
hepatocellular damage. the indirect bilirubin made by the fetus is transported across
Severe anemia and hypoproteinemia caused by decreased the placenta and conjugated by the maternal liver and safely
hepatic production of plasma proteins leads to the develop- excreted. After birth, the immature infant liver cannot yet
ment of high-output cardiac failure with generalized edema, metabolize bilirubin efficiently, and this leads to the accu-
effusions, and ascites, a condition known as hydrops fetalis. mulation of unconjugated bilirubin and neonatal jaundice.
In severe cases, hydrops fetalis can develop by 18 to 20 weeks’ When a newborn infant is affected by HDFN, the levels of
gestation. In the past, hydrops fetalis was almost uniformly indirect bilirubin are higher due to the pathological RBC
fatal; today, most fetuses with this condition can be treated destruction. With moderate to severe hemolysis of HDFN,

Table 20–3 Neonatal Manifestations of HDFN-Induced Anemia by Time of Onset


Early-Onset Anemia Late Hemolytic Anemia Late Hyporegenerative Anemia
Onset Within 7 days of birth ≥2 Weeks of age

Mechanism Antibody-mediated hemolysis 1. Antibody-mediated hemolysis 1. Antibody destruction of RBC precursors and
2. Natural decline of Hb levels RBCs
3. Expanding intravascular 2. Marrow suppression by IUT and transfusions
volume of growing infant 3. Erythropoietin deficiency
4. Expanding intravascular volume of growing
infant

Bilirubin Elevated Usually elevated Normal

Reticulocyte count Normal or high Normal or high Low or absent

Hb = hemoglobin; RBC = red blood cell; IUT = intrauterine transfusion


(Adapted from Rath ME, Smits-Wintjens VE, Walther FJ, Lopriore E. Hematological morbidity and management in neonates with hemolytic disease due to red cell alloimmunization.
Early Hum Dev. 2011;87(9):583-88.)
6888_Ch20_427-440 29/10/18 5:39 PM Page 431

Chapter 20 Hemolytic Disease of the Fetus and Newborn (HDFN) 431

the unconjugated, or indirect, bilirubin can reach levels toxic cases in which ABO incompatibility can be the only cause of
to the infant’s brain (generally, more than 18 to 20 mg/dL), neonatal jaundice, but the DAT result is negative, the eluate
and if left untreated can cause kernicterus or permanent of the cord RBCs may reveal ABO antibodies. The eluate can
damage to the brain. also be helpful when the mother’s blood specimen is not
available.
Diagnosis
HDFN Caused by RBC Alloimmunization
HDFN Caused by ABO
The diagnosis of HDFN due to previous RBC exposure and
Many investigators have tried to use the immunoglobulin alloimmunization requires close cooperation among the
class and titer of maternal ABO antibodies to predict ABO pregnant patient, her health-care provider, her partner, and
HDFN. These tests are laborious and at best demonstrate the the personnel of the clinical laboratory performing the test-
presence of IgG maternal antibody, but they do not correlate ing (Fig. 20–2).15 The recommended practice is to perform
well with the extent of fetal RBC destruction. Consequently, the type and antibody detection test at the first prenatal visit,
detection of ABO HDFN is best done after birth. preferably during the first trimester.16 At that time, a mater-
nal history must be taken to understand if there is a history
Postnatal Diagnosis of HDFN, the previous pregnancy outcomes, and whether
No single serologic test is diagnostic for ABO HDFN. When there is a history of prior transfusions. Previous severe dis-
a newborn develops jaundice within 12 to 48 hours after ease and poor outcome predict similar findings in the current
birth, various causes of jaundice need to be investigated. The pregnancy. Although consecutive antibody titers are useful
DAT on the cord or neonatal RBCs is the most important in assessing the extent of intrauterine fetal anemia during
diagnostic test. In all cases (100%) of ABO HDFN requiring the first affected pregnancy, antibody titers are less predictive
transfusion therapy and in 90% of those with jaundice, the in subsequent pregnancies.
DAT result has been positive.14 On the other hand, the DAT
result can be positive even in the absence of signs and symp- ABO, RhD, and Antibody Screen
toms of clinical anemia in the newborn infant. However, these The prenatal specimen must be typed for ABO and RhD. The
infants may have compensated anemia or the RBCs are not antibody detection method, or indirect antihuman globulin
being destroyed by the reticuloendothelial system. test (IAT), must be able to detect clinically significant IgG
Collecting cord blood samples on all delivered infants is alloantibodies that are reactive at 37°C and in the anti-
highly recommended. The sample should be collected by globulin phase. At least two separate reagent screening RBCs
venipuncture to avoid contamination with maternal blood that express all of the common blood group antigens (prefer-
and Wharton’s jelly (the material surrounding the blood ably homozygous) should be used. For tube testing, an
vessels) and should be anticoagulated for storage. If the antibody-enhancing medium such as polyethylene glycol
neonatal infant develops jaundice, ABO, RhD, and DAT (PEG) or low ionic strength solution (LISS) can increase
testing can be carried out and the results can be assessed. sensitivity of the assay.
When the DAT result is negative but the infant is jaundiced, Prenatal patients may produce clinically insignificant anti-
other causes of jaundice should be investigated. In the rare bodies, such as anti-Lea or anti-Leb. Therefore, immediate

Figure 20–2. Approach to paternal testing for risk stratification of


Maternal
fetus to be affected by HDFN. (US = ultrasound using Doppler middle Antibody Screen
cerebral artery velocity testing.) Positive

RBC antibody
Anti-D (non-RhD)

Genotype father Phenotype father


for RhD zygosity for zygosity

Homozygous Heterozygous Homozygous Heterozygous


DD Dd

Fetus at risk: Fetal genetic testing Fetus at risk: Fetal genetic testing
Titers and US to determine fetal Titers and US to determine fetal
inheritance inheritance
6888_Ch20_427-440 29/10/18 5:39 PM Page 432

432 PART III Transfusion Practices

spin and room temperature incubation phases can be omitted, method is the saline antiglobulin tube test, with 60-minute
and anti-IgG rather than polyspecific antiglobulin reagent is incubation at 37°C and the use of anti-IgG reagent, although
used. These steps reduce detection of IgM antibodies, which other methods have also been proposed.19,20 The RBCs used
cannot cross the placenta. Other antibody screening methods, for each titration should have the same genotype (preferably
such as solid phase or gel column, may be used. homozygous for the antigen of interest), approximately the
If the antibody screen is nonreactive, repeat testing is rec- same storage time, and the same concentration. The first
ommended before RhIG therapy in RhD-negative prenatal serum or plasma specimen should be frozen and run in par-
patients and in the third trimester if the patient has been allel with later specimens to increase accuracy since the titer
transfused or has a history of unexpected antibodies.17 test results are difficult to reproduce.21 Only a difference of
greater than 2 dilutions or a score change of more than 10 is
Antibody Identification considered a significant change in titer.
If the antibody screen is reactive, the antibody identity must The method chosen is critical for the appropriate clinical
be determined. Follow-up testing will depend on the anti- correlation. Methods using enhancing media or gel columns
body specificity. Cold reactive IgM antibodies such as anti-I, result in higher titers, as shown by comparative proficiency
anti-IH, anti-Lea, anti-Leb, and anti-P1 can be ignored. Lewis testing. Therefore, the critical titer level for these other meth-
system antibodies are rather common in pregnant women ods must be determined by the individual laboratory by re-
but have not been reported to cause HDFN. Antibodies such viewing the outcome of several pregnancies complicated by
as anti-M and anti-N can be IgM or IgG or a combination of HDFN, if this method is to be used.
both. Both anti-M and anti-N can cause mild to moderate For the recommended method, 16 is considered the crit-
HDFN, although rarely. To establish the immunoglobulin ical titer. If the initial titer is 16 or higher, a second titer
class, the serum can be treated with a sulfhydryl reagent, should be done at about 18 to 20 weeks’ gestation. A titer
such as dithiothreitol or 2-mercaptoethanol, and then reproducibly and repeatedly at ≥32 is an indication for color
retested with appropriate controls. The J-chain of IgM anti- Doppler imaging to assess middle cerebral artery peak sys-
bodies will be destroyed by this treatment; IgG antibodies tolic velocity (MCA-PSV) after 16 weeks’ gestation.22 When
will remain reactive. the titer is less than 32, it should be repeated at 4-week in-
Many Rh-negative pregnant women have weakly reactive tervals, beginning at 16 to 20 weeks’ gestation and then every
anti-D, particularly during the third trimester. Most of these 2 to 4 weeks during the third trimester. Antibody titer alone
women have received RhIG, either after an event with cannot predict severity of HDFN. In some sensitized women,
increased risk of fetomaternal hemorrhage or at 28 weeks’ the antibody titer can remain moderately high throughout
gestation (antenatal). The passively administered anti-D will pregnancy while the fetus is becoming more severely af-
be weakly reactive in testing and will remain demonstrable fected. Similarly, a previously sensitized woman can have
for 2 months or longer. This must be distinguished from consistently high antibody titer, whether pregnant or not,
active immunization. A titer higher than 4 almost always and whether the fetus is RhD-positive or RhD-negative. In
indicates active immunization; with a titer under 4, active others, the titer can rise rapidly, which portends increasing
immunization cannot be ruled out, but it is less likely. If severity of HDFN. However, antibody titers consistently
encountering a sample from a pregnant patient, using PEG below the laboratory’s critical titer throughout the pregnancy
appears to provide the least number of false positives com- reliably predict an unaffected or mild-to-moderately affected
pared to gel cards and solid phase.18 Communication with fetus, with the exception of anti-K with a K-positive fetus.
the patient’s provider to obtain a history of RhIG administra- Titration studies at time of delivery are not recommended
tion is an important confirmatory step as serological methods because they provide no clinically useful information.
alone are not able to determine the derivation of the antibody.
If the antibody specificity is determined to be clinically Paternal Phenotype and Genotype
significant and the antibody is IgG, further testing is re- A specimen of the father’s blood should be obtained and
quired. Other than anti-D, the most common and most sig- tested for the presence and zygosity (predicted copy number
nificant antibodies are anti-K, anti-E, anti-c, anti-C, and of the gene) of the corresponding blood group antigen to
anti-Fya (Table 20–2). predict fetal risk of being affected by HDFN. Fathers that are
homozygous for the blood group gene have a 100% chance
Antibody Titers of passing this gene to their offspring; heterozygous fathers
The relative concentration of all antibodies capable of cross- have a 50% chance. The mother should be counseled in pri-
ing the placenta and causing HDFN is determined by anti- vate as to the paternity of the fetus to ensure accurate pater-
body titration. The patient serum or plasma is serially nal phenotyping and genotyping.
diluted and tested against appropriate RBCs to determine the If the mother has anti-D and the father is RhD-positive,
highest dilution at which a reaction occurs.19 The method the paternal genotype determined by DNA methods is the
must include the indirect antiglobulin phase using anti-IgG only way to definitively determine how many copies of
reagent. The result is expressed as either the reciprocal of the RHD gene an RhD-positive person carries. Thus, RHD
the titration endpoint or as a titer score. zygosity genotype testing is the recommended test for
The titration must be performed exactly the same way RhD-positive fathers when the mother has anti-D antibody
each time the patient’s serum is tested. The recommended (Fig. 20–2).
6888_Ch20_427-440 29/10/18 5:39 PM Page 433

Chapter 20 Hemolytic Disease of the Fetus and Newborn (HDFN) 433

In cases of maternal antibody specificity other than anti-D,


paternal red blood cell phenotype testing of the father can
predict the chance of the fetus inheriting the implicated
blood group antigen. For example, only 9% of the population
is positive for the Kell antigen.

Fetal DNA Testing


Risk stratification of the fetus can be directly carried out by
obtaining fetal cells through amniocentesis or chorionic vil-
lous sampling (CVS) as early as 10 to 12 weeks’ gestation.
The fetal cells are grown in tissue culture, and then DNA is
extracted to determine if the fetus has genes coding for the
blood group genes, including RHD and others (c, e, C, E, K,
Fya, Fyb, Jka, Jkb, M). To avoid an invasive procedure, fetal
DNA can be isolated from maternal plasma from a peripheral
blood sample to determine RHD and KEL genotype.23,24 The
cell-free DNA (cfDNA) method has advanced fetal risk strat-
ification for mothers with RBC alloimmunization. In some
countries cfDNA methods are also used when the mother is
RhD-negative to determine if the fetus is predicted to be
RhD-positive and therefore if RhIG should be adminis-
tered.25 This may be a cost-effective approach.26
Figure 20–3. Diagram of fetal middle cerebral artery Doppler velocimetry
Management of the Fetus testing.

During pregnancy, women with known RBC alloimmuniza-


tion and/or a history of HDFN are closely monitored using product to be used for intrauterine transfusion (IUT) should
antibody titers as described above. The fetus is also closely be ready in case it is needed (see below). The fetal blood
monitored throughout the pregnancy to check for fetal well- sample can also be tested for bilirubin, ABO, Rh, DAT, and
being and fetal anemia and to determine when is the best antigen phenotype and genotype.
time to deliver. For risk stratification of fetal anemia, amniocentesis to
monitor amniotic fluid bilirubin levels has been replaced
Fetal Ultrasound with MCA-PSV.28 In the past, the concentration of bilirubin
pigment in the amniotic fluid was used to estimate the extent
At about 16 to 20 weeks’ gestation, the clinical diagnosis of of fetal hemolysis. The amniotic fluid is tested by a spec-
fetal anemia can be made using an ultrasound technique trophotometric scan optical density (∆OD) at 450 nm (the
called fetal middle cerebral artery peak systolic velocity absorbance of bilirubin). The measurement is plotted on a
(MCA-PSV) (Fig. 20–3).15 The measurement is based on the graph (Liley Curve Graph) according to gestational age. An
reduced blood viscosity at lower hematocrits and resulting increasing or unchanging ∆OD 450 nm as pregnancy pro-
in faster velocity of the blood. Readings are typically done ceeds predicts worsening of the hemolysis. High values
every 2 weeks to track the degree of fetal anemia; those indicate severe and often life-threatening hemolysis (fetal he-
that are greater than 1.5 multiples of the mean (MoM) are moglobin less than 8 g/dL) and require urgent intervention.
sensitive enough to predict significant fetal anemia in which Currently, amniocentesis may be used for obtaining fetal
intervention may be needed.15,27 amniocytes for DNA testing, as described above.

Invasive Monitoring—Cordocentesis Intrauterine Transfusion


and Amniocentesis
Intervention in the form of intrauterine transfusion becomes
When fetal anemia becomes moderate to severe as indicated necessary when one or more of the following conditions
by Doppler MoM measurements exceeding 1.5, invasive exists:
testing via cordocentesis is done to determine fetal hemat-
• MCA-PSV indicates anemia (>1.5 MoM).
ocrit.15 Using high-resolution ultrasound with color
• Fetal hydrops is noted on ultrasound examination.
Doppler enhancement of blood flow, the umbilical vein is
• Cordocentesis blood sample has hemoglobin level less
visualized at the level of the cord insertion into the placenta.
than 10 g/dL.
A spinal needle is inserted into the umbilical vein, and a
• Amniotic fluid ∆OD 450 nm results are high and/or
sample of the fetal blood is obtained. If the fetus has not
increasing.
reached an acceptable gestational age for delivery, and the
hematocrit level is less than 30%, intrauterine transfusion Intrauterine transfusion is performed by accessing the
is usually indicated. During the procedure, an RBC blood fetal umbilical vein (cordocentesis) and injecting donor
6888_Ch20_427-440 29/10/18 5:39 PM Page 434

434 PART III Transfusion Practices

RBCs directly into the vein.29 The goal of intrauterine trans- anti-B antisera than for older children and adults. In addi-
fusion is to maintain fetal hemoglobin above 10 g/dL. Once tion, infants do not have their own isohemagglutinins but
intrauterine transfusion is initiated, the procedure is typi- may have those of the mother, so reverse grouping cannot
cally repeated every 2 to 4 weeks until delivery to suppress be used to confirm the ABO group.
fetal hematopoiesis. The initial intrauterine transfusion is
rarely performed after 36 weeks’ gestation. RhD Typing
The selection of red blood cell products for intrauterine Rarely, the infant’s RBCs can be heavily antibody-bound with
transfusion is typically group O, RhD-negative (or RhD- maternal anti-D, causing a false-negative Rh type, or what
positive, depending on maternal blood group antibody), has been called blocked Rh.35 An eluate from these RBCs
leukocyte reduced, hemoglobin S negative, CMV-safe (CMV will reveal anti-D, and typing of the eluted RBCs will show
seronegative or leukocyte reduced), irradiated, and antigen- reaction with anti-D.
negative for maternal red blood cell antibody/antibodies.
The RBC unit is irradiated to prevent transfusion-associated Direct Antiglobulin Test
graft-versus-host disease. The hematocrit level of the RBCs The most important serologic test for diagnosing HDFN
should be greater than 70% because of the small volume is the DAT with anti-IgG reagent. A positive test result
transfused and the need to correct severe anemia. indicates that there is antibody coating the infant’s RBCs;
Cordocentesis, intrauterine transfusion, and amniocentesis however, the strength of the reaction does not correlate well
have several risks, including infection, premature labor, and with the severity of the HDFN. A positive test result may be
trauma to the placenta, which may cause increased antibody found in infants without clinical or other laboratory evidence
titers because of antigenic challenge to the mother through of hemolysis (e.g., mother received RhIG).
fetomaternal hemorrhage. To protect the mother from addi-
tional sensitization due to the exposure to donor RBCs through Elution
the procedure, some authors tried to prevent alloimmunization The routine preparation of an eluate of all infants with a
by using Rh C, c, E, e, and K maternally antigen-matched RBCs positive DAT result is unnecessary. Elution in cases of known
for IUT, but found that the risk of additional maternal RBC HDFN and postnatal ABO incompatibility is not needed, be-
antibodies remained.30 High-level antigen matching (Duffy, cause eluate results do not change therapy. The preparation
Kidd, Ss) of mothers with HDFN does seem to reduce the risk of an eluate may be helpful when the cause of HDFN is in
of further alloimmunization, but the RBCs become increasingly question or suspected. As noted above, the resolution of a
difficult to find and the clinical impact is not clear.31 Intrauter- case of blocked RhD typing requires an eluate.
ine transfusion alone carries a 1% to 3% chance of adverse fetal
events such as premature rupture of membranes.32 When done Exchange Transfusion
in the early second trimester, the outcomes are poor.33,34
Despite the risks of IUT, children older than 2 years who were For patients with known HDFN, close observation of bilirubin
treated with this procedure while in utero had a relatively levels and hemoglobin is warranted to determine if neonatal
low rate (4.8%) of neurocognitive impairment (cerebral palsy, exchange transfusion is needed to remove bilirubin and
severe developmental delay, bilateral deafness, blindness) so maternal antibody.13,36 When levels of bilirubin reach critical
long as they were not severely hydropic.12 levels (which depends on the infant’s gestational age), ex-
change transfusion is indicated.36 Exchange transfusion is
the use of whole blood or equivalent to replace the neonate’s
Management of the Infant
circulating blood and simultaneously remove maternal anti-
When birth occurs, the connection from the fetal to maternal bodies and bilirubin. Exchange transfusion is rarely required
circulation is severed, and the risk of hyperbilirubinemia because of advances in phototherapy and the use of IVIG.
increases because the fetal metabolic pathway to metabolize Blood product selection is similar to that of IUT; group O,
bilirubin is immature. Although many babies are affected with RhD-negative (or RhD-positive, depending on maternal
neonatal jaundice, those with HDFN-induced hemolysis are at blood group antibody), leukocyte reduced, hemoglobin S
greater risk of the bilirubin reaching very high levels and thus negative, CMV-safe (CMV seronegative or leukocyte re-
for bilirubin-induced encephalopathy. Blood product transfu- duced), irradiated, and antigen-negative for maternal red
sion can be used to treat anemia and hyperbilirubinemia. blood cell antibody/antibodies. However, because infant
whole blood is also being removed, the RBC unit is usually
Cord Blood Testing mixed with a plasma unit to create reconstituted whole
blood. Usually, RBCs units less than 7 to 10 days from
Serologic testing of the cord blood sample drawn at the time collection from the donor are selected to reduce the risk of
of birth is used to confirm HDFN and prepare for possible hyperkalemia. However, special circumstances, such as the
transfusion. need for units of the mother’s blood when high-incidence
antibodies are involved, have shown that older blood units
ABO Grouping can be safe and effective for the newborn when infused
ABO antigens are not fully developed in newborn infants; slowly. After a two-volume exchange transfusion, approxi-
their RBCs may show weaker reactions with anti-A and mately 90% of the red blood cells have been replaced and
6888_Ch20_427-440 29/10/18 5:39 PM Page 435

Chapter 20 Hemolytic Disease of the Fetus and Newborn (HDFN) 435

50% of the bilirubin has been removed. After the procedure, use K-negative RBC units for women younger than 45 to
a platelet count should be performed to monitor for iatro- 50 years of age. Other nations provide additional matching
genic thrombocytopenia. for antigens such as c and E.38 In a large international review
of women with infants with severe HDFN, a transfusion pol-
Simple Transfusions icy of prospective antigen matching for RBC transfusion did
not provide protection from HDFN. This appears to be
The infant may receive small-volume or “top-off” RBC trans- driven by the fact that 83% of maternal sensitization that
fusions to correct anemia anytime from after birth to many went on to cause severe HDFN was due to previous preg-
weeks later. The hemoglobin or hematocrit level at which a nancy and not transfusion itself.4
newborn needs an RBC transfusion has been the subject of
a number of studies, and is typically set by the neonatology Rh Immune Globulin
physician group. Infants must be carefully monitored for
clinical signs of ongoing anemia, which can be clinically sus- The risk of an RhD-negative mother becoming allosensitized
pected when the infant has poor feeding or increased sleep.13 can be reduced to from 16% to less than 0.1% by the appro-
When transfused, the RBCs selected have the same attributes priate administration of RhIG.39 At this time, there are no
as noted with IUT and exchange transfusion. Many hospitals other pharmaceutical therapies that can prevent blood group
will keep 1 unit dedicated to an infant with HDFN and draw sensitization for other blood group antigens. The mechanism
small aliquots from the parent RBC unit over time to de- of action of RhIG is uncertain. Evidence indicates it inter-
crease donor exposure over multiple transfusion episodes. feres with B-cell priming to make anti-D, although other
modes of action may occur.40
Phototherapy Because of the known risk of Rh immunization during
pregnancy, RhIG should be given to RhD-negative mothers.
Phototherapy at 460 to 490 nm is used to metabolize the The first dose is provided at 28 weeks’ gestation, as recom-
unconjugated bilirubin to isomers that are less lipophilic, mended by the American College of Obstetricians and
less toxic to the brain, and able to be excreted through Gynecologists (ACOG), since the majority of allosensitiza-
urine.37 Relatively high doses are given by using two banks tion appears to occur after this time. A second dose is given
of lights to surround the infant’s body. For infants with mild after delivery of an RhD-positive infant.41 Based on experi-
to moderate hemolysis or history of intrauterine transfusion, ments conducted many years ago, it is recommended to give
phototherapy is generally sufficient to adequately conjugate RhIG within 72 hours after delivery. Even if more than
the bilirubin and lessen the need for transfusion. 72 hours have elapsed, RhIG should still be given, as it may
be effective and is not contraindicated.
Intravenous Immune Globulin Figure 20–4 outlines a decision tree for the indications
and dose of postpartum RhIG administration. The mother
Intravenous immune globulin (IVIG) is used to treat hyper-
should be D-negative, and the infant should be D-positive
bilirubinemia of the newborn caused by HDFN. The IVIG
or D-variant. If the type of the infant is unknown (e.g., if the
competes with the mother’s antibodies for the Fc receptors
infant is stillborn), RhIG should also be administered. Anti-
on the macrophages in the infant’s spleen, reducing the
body titers are not recommended because the amount of
amount of hemolysis. IVIG appears to reduce the need for
circulating RhIG does not correlate with effectiveness of the
exchange transfusions and phototherapy, but it does not
immune suppression or with the amount of fetomaternal
affect the need for top-off transfusions.13
hemorrhage. The half-life of IgG is about 25 days, so only
about 10% of the antenatal dose will be present at 40 weeks’
Prevention
gestation. It is essential that the anti-D from antenatal RhIG
Prevention of HDFN can be divided into primary and second- present at delivery not be interpreted erroneously as active
ary measures. Because there are no international standards, rather than passive immunization.
nations differ on preventative measures for HDFN, including
Dose and Administration
dosing and dosing schedules of RhIG and the approach to
RBC transfusion. The regular-dose vial of RhIG in the United States contains
sufficient anti-D to protect against 15 mL of packed RBCs or
Selection of RBCs for Females 30 mL of whole blood. This is equal to 300 µg of the World
Health Organization (WHO) reference material. In the United
One of the tenets of transfusion medicine is to preserve RhD- Kingdom, the regular-dose vial contains about 100 µg, which
negative RBCs in the blood bank inventory for use for appears to be adequate for postpartum prophylaxis. The mi-
women of childbearing potential. This is entirely in place to crodose (United States) can be used for abortions and ectopic
reduce the risk of RhD sensitization and of HDFN affecting pregnancies before the 12th week of gestation. The total fetal
future pregnancies. blood volume is estimated to be less than 5 mL at 12 weeks.
Furthermore, in some countries, minor blood group anti- Intravenous preparations (IV) of RhIG are approved for use
gens are matched (or provided as negative) for women of in the United States. These products also contain 300 µg in
childbearing potential. For instance, a number of countries each vial and can be administered either intramuscularly or
6888_Ch20_427-440 29/10/18 5:39 PM Page 436

436 PART III Transfusion Practices

Maternal Sample ghosts. After 2,000 cells are counted, the percentage of fetal
During First Trimester cells is determined, and the volume of fetal hemorrhage is
calculated using this formula:
Number of fetal cells × Maternal blood volume =
RhD negative RhD positive Number of maternal cells
Volume of fetomaternal hemorrhage
Rhlg at 28 weeks gestation or Because one 300-µg dose covers 30 mL of whole blood,
when procedure or trauma occurs the calculated volume of fetomaternal hemorrhage is then
divided by 30 to determine the number of required vials of
Birth of infant RhIG. Because the Kleihauer-Betke is an estimate, one vial
is added to the calculated answer.16 When needed, the addi-
tional vials of RhIG should be administered within 72 hours
of delivery or as soon as possible. Although it has been a key
Cord blood Maternal postpartum
sample sample
test as described for many years, the Kleihauer-Betke test is
imprecise, and recent attention has focused on newer tech-
nologies, such as flow cytometry, to provide more accurate
Anti-D present: Rhlg not quantification of the FMH volume.42
RhD RhD positive indicated if anti-D not due to
negative or weak D previous Rhlg
Maternal Weak D
As molecular testing advances throughout the field of med-
FMH screen icine, so does the application of blood typing using molecu-
test (Rosette)
lar techniques. In certain patients, serologic reagents do not
accurately detect the RhD type. The most common genetic
backgrounds that account for this serologic typing problem
Negative Positive are called weak D phenotypes. Recently, authors have en-
couraged the use of RhD genetic testing for patients with a
weak D phenotype to provide accurate and actionable results
One vial Rhlg Quantitative FMH
(300 ug) assessment for RhD blood typing and RhIG administration.43,44 Further
scientific study is needed to elucidate the clinical significance
of different RhD genotypes in various ethnic backgrounds
Calculate Rhlg and the risk factors for RhIG failure.45
dose
Other Considerations
Figure 20–4. Decision tree for the indications and dose of RhIG as determined
by laboratory test results. If the cord or neonatal blood specimen is unavailable, RhIG is of no benefit once a person has been actively immu-
assume the fetus is D-positive. nized and has formed anti-D. However, care must be taken
to distinguish women who have been passively immunized
by antenatal administration of RhIG from those who have
intravenously. The RhIG must be injected according to the been actively immunized by exposure to Rh-positive RBCs.
product label. The IV product can also be given intramuscu- RhIG is not indicated for the mother if the infant is found
larly. The intramuscular form must be given intramuscularly to be D-negative. The blood type of fetuses in abortions, still-
only. IV injections of intramuscular preparations can cause se- births, and ectopic pregnancies usually cannot be deter-
vere anaphylactic reactions because of the anticomplementary mined; therefore, RhIG should be administered in these
activity of these products. RhIG also contains IgA and may be circumstances.
contraindicated in patients with anti-IgA and IgA deficiency
who have had anaphylactic reactions to blood products.
Massive fetomaternal hemorrhages of more than 30 mL CASE STUDIES
of whole blood occur in less than 1% of deliveries.7 These
massive hemorrhages can lead to immunization if adequate Case 20-1
RhIG is not administered. A maternal sample should be ob-
tained within 1 hour of delivery and screened using a test A 29-year-old mother is gravida 1 para 0 (G1P0). She is
such as the rosette technique for massive fetomaternal hem- 32 weeks’ pregnant and she is group O, RhD-negative. Al-
orrhage. If positive, quantitation of the hemorrhage must be though her antibody screen was negative 4 weeks ago and
done by Kleihauer-Betke or by flow cytometry assays. 6 months ago, today it is positive, and anti-D is identified
In the Kleihauer-Betke test, a maternal blood smear is with a titer of 2.
treated with acid and then stained with counterstain. Fetal 1. What is the most probable cause of anti-D in this
cells contain fetal hemoglobin (Hgb F), which is resistant to mother?
acid and will remain pink. The maternal cells will appear as
6888_Ch20_427-440 29/10/18 5:39 PM Page 437

Chapter 20 Hemolytic Disease of the Fetus and Newborn (HDFN) 437

2. How can you confirm your suspicion? 2. Is the antibody identified in the current pregnancy
3. After delivery, is this mother an Rh immune globulin clinically significant? Why?
candidate?
Case 20-4
Case 20-2
At a rural hospital a 32-year-old woman just delivered a
A 35-year-old mother is gravida 4 para 2 (G4P2) and severely anemic infant. At 6 weeks’ gestation, the mother
currently 8 weeks’ pregnant. Her first pregnancy was had been typed as A-negative, with positive antibody
uncomplicated, and she delivered a healthy infant. Her screen. The antibody was identified as anti-D, titer 256. A
pregnancy history is significant for her second child report from another hospital states that she had an in-
having a positive DAT that required no treatment and trauterine transfusion 3 weeks earlier in another state. The
her third child was stillborn. All of her pregnancies have cord blood collected at delivery is typed as A-negative. On
been with the same father. Current laboratory testing a heel-stick specimen, the infant’s hemoglobin is reported
finds her to be group O, RhD-negative with a positive as 4.3 g/dL and bilirubin as 13.9 mg/dL. Typing results of
antibody screen and anti-D identified. The antibody this specimen are as follows:
titer is negative in saline and positive at AHG 128
Anti-A Anti-B Anti-D DAT
(1:128).
0 0 0 +/–
1. What is the most likely Rh(D) type of the fetus?
1. What is the infant’s blood type? Why is the infant so
2. When should the titer be repeated?
anemic? What further testing is indicated?
3. Is there any intervention that should be done
now? Why? Further testing shows the following:
4. When should MCA-PSV be performed?
Cord Blood Heel-stick
5. Is the mother a candidate for Rh Immune Globulin
Anti-I 4+ 3+
Administration?
Kleihauer-Betke 0/1,000 23/1,000
Case 20-3 The results indicate the cord blood specimen is all
adult blood and the heel-stick specimen is nearly all adult
A 32-year-old mother is gravida 3 para 2 (G3P2) and cur-
blood.
rently 32 weeks’ pregnant. Her pregnancy history is as
follows: first pregnancy had no complications with a 2. How could that happen?
healthy infant; the second pregnancy had no complica-
Further testing is done on the heel-stick specimen in
tions, but the infant was born with a positive DAT,
which the anti-A and anti-B tubes are incubated at 4°C:
neonatal jaundice. The infant was group A, RhD-positive.
Current laboratory testing finds her to be group O, RhD- Anti-A (4°C) Anti-B (4°C) RBC Eluate
positive with a positive antibody screen and anti-Lea 0 + Anti-D
identified.
These results indicate that the infant is B-positive and
1. What is the most probable cause of the second-born has HDFN caused by anti-D.
child having a positive DAT?

SUMMARY CHART
 HDFN is the destruction of the RBCs of the fetus and group O and the newborn is group A, B, or AB; the IgG
neonate by IgG antibodies produced by the mother. antibody, anti-A,B in the mother’s circulation, crosses
 Only antibodies of the IgG class are actively trans- the placenta and attaches to the ABO-incompatible
ported across the placenta. antigens of the fetal RBCs.
 Usually, in RhD HDFN, the RhD-positive firstborn in-  Erythroblastosis fetalis describes the presence of imma-
fant of an RhD-negative mother is unaffected because ture RBCs or erythroblasts in the fetal circulation be-
the mother has not yet been immunized; in subsequent cause the splenic removal of the IgG-coated RBCs
pregnancies, fetal cells carrying the Rh antigen immu- causes anemia; the term commonly used now is HDFN.
nize the Rh-negative mother and stimulate production  Although anti-D is the most antigenic of the Rh anti-
of anti-D. bodies, anti-Kell is considered the most clinically sig-
 In ABO HDFN, the firstborn infant may be affected as nificant of the non–Rh-system antibodies in the ability
well as subsequent pregnancies in which the mother is to cause HDFN.

Continued
6888_Ch20_427-440 29/10/18 5:39 PM Page 438

438 PART III Transfusion Practices

SUMMARY CHART—cont’d
 Prenatal serologic tests for obstetric patients include by the RhD antigen on fetal cells; RhIG attaches to fetal
an ABO, Rh, and antibody screen during the first Rh-positive RBCs in maternal circulation, blocking im-
trimester of pregnancy. munization and subsequent production of anti-D.
 A cord blood workup includes tests for ABO and Rh  A Kleihauer-Betke test or flow cytometry is used to
as well as DAT; the most important serologic test for quantitate the number of fetal RhD-positive cells in
diagnosis of HDFN is the DAT with anti-IgG reagent. the mother’s circulation as a result of a fetomaternal
 RhIG administered to the mother within 72 hours hemorrhage.
following delivery is used to prevent active immunization

7. RhIG is given to RhD-negative mothers without regard


Review Questions
for fetal RhD type in all of the following conditions
except:
1. The etiology of HDFN is characterized by:
a. Ectopic pregnancy rupture
a. IgM antibody
b. Full-term delivery
b. Nearly always anti-D
c. Amniocentesis
c. Different RBC antigens between mother and father
d. Induced abortion
d. Antibody titer less than 32
8. A Kleihauer-Betke test or flow cytometry indicates 10 fetal
2. An important difference between the fetus and the
cells per 1,000 adult cells. For a woman with 5,000-mL
newborn physiology is:
blood volume, the proper dose of RhIG is:
a. Bilirubin metabolism
a. One regular-dose (300 µg) vial
b. Maternal antibody level
b. Two regular-dose vials
c. Presence of anemia
c. Three regular-dose vials
d. Size of RBCs
d. Four regular-dose vials
3. Kernicterus is caused by the effects of:
9. ABO HDFN is usually mild because:
a. Anemia
a. ABO antigens are poorly developed in the fetus
b. Unconjugated bilirubin
b. ABO antibodies prevent the disease itself
c. Antibody specificity
c. ABO antibodies readily cross the placenta
d. Antibody titer
d. ABO incompatibility is rare
4. The advantage of middle cerebral artery peak systolic
10. A woman without prenatal care delivers a healthy term
velocity Doppler (MCA-PSV) is that it is:
infant. A cord blood sample shows the infant is A-positive
a. Able to measure fetal hemoglobin and hematocrit with a positive DAT. The workup of the unexpected find-
levels ing should include:
b. Able to support antigen typing of fetal blood using
a. Anti-C3 antiglobulin test
DNA
b. ABO testing of the mother
c. Helpful for direct transfusion of fetal circulation
c. Direct antiglobulin testing of the mother’s specimen
d. Noninvasive and decreases risk of adverse events
d. ABO and Rh typing of the father
5. Blood for intrauterine transfusion (IUT) should be:
a. Irradiated, leukocyte reduced, more than 7 days old, References
HbS negative 1. van der Schoot CE, Tax GH, Rijnders RJ, de Haas M,
b. Irradiated, leukocyte reduced, less than 7 days old, Christiaens GC. Prenatal typing of Rh and Kell blood group
HbS positive system antigens: the edge of a watershed. Transfus Med Rev.
2003;17(1):31-44.
c. Irradiated, leukocyte reduced, less than 7 days old, 2. Geifman-Holtzman O, Wojtowycz M, Kosmas E, Artal R.
HbS negative Female alloimmunization with antibodies known to cause
d. Irradiated, leukocyte reduced, more than 7 days old, hemolytic disease. Obstet Gynecol. 1997;89(2):272-5.
HbS positive 3. Dajak S, Culic S, Stefanovic V, Lukacevic J. Relationship
between previous maternal transfusions and haemolytic disease
6. RhIG is indicated for: of the foetus and newborn mediated by non-RhD antibodies.
a. Mothers who have anti-D due to allosensitization Blood Transfus. 2013;11(4):528-32.
4. Delaney MWA, van de Watering L, Schonewille H, Verdoes JP,
b. Infants who are RhD-negative Emery SP, Murphy MF, et al. Blood Group Antigen Matching
c. Infants who have anti-D Influence on Gestational Outcomes (AMIGO) Study. Transfu-
d. Mothers who are RhD-negative sion. 2016;57(3):525-32.
6888_Ch20_427-440 29/10/18 5:39 PM Page 439

Chapter 20 Hemolytic Disease of the Fetus and Newborn (HDFN) 439

5. Firan M, Bawdon R, Radu C, Ober RJ, Eaken D, Antohe F, 25. Tiblad E, Taune Wikman A, Ajne G, Blanck A, Jansson Y,
et al. The MHC class I-related receptor, FcRn, plays an essential Karlsson A, et al. Targeted routine antenatal anti-D prophylaxis
role in the maternofetal transfer of gamma-globulin in humans. in the prevention of RhD immunisation–outcome of a new
Int Immunol. 2001;13(8):993-1002. antenatal screening and prevention program. PloS One. 2013;
6. Li P, Pang LH, Liang HF, Chen HY, Fan XJ. Maternal IgG 8(8):e70984.
anti-A and anti-B titer levels screening in predicting ABO 26. Teitelbaum L, Metcalfe A, Clarke G, Parboosingh JS, Wilson
hemolytic disease of the newborn: a meta-analysis. Fetal RD, Johnson JM. Costs and benefits of non-invasive fetal RhD
Pediatr Pathol. 2015;34(6):341-50. determination. Ultrasound Obstet Gynecol. 2015;45(1):84-8.
7. Sebring ES, Polesky HF. Fetomaternal hemorrhage: incidence, 27. ACOG Practice Bulletin No. 75: Management of alloimmuniza-
risk factors, time of occurrence, and clinical effects. Transfu- tion during pregnancy. Obstet and Gynecol. 2006;108(2):
sion. 1990;30(4):344-57. 457-64.
8. Higgins JM, Sloan SR. Stochastic modeling of human RBC 28. Oepkes D, Seaward PG, Vandenbussche FP, Windrim R,
alloimmunization: evidence for a distinct population of im- Kingdom J, Beyene J, et al. Doppler ultrasonography versus
munologic responders. Blood. 2008;112(6):2546-53. amniocentesis to predict fetal anemia. New Engl J Med. 2006;
9. Ayache S, Herman JH. Prevention of D sensitization after 355(2):156-64.
mismatched transfusion of blood components: toward optimal 29. Oepkes D, Adama van Scheltema P. Intrauterine fetal transfu-
use of RhIG. Transfusion. 2008;48(9):1990-9. sions in the management of fetal anemia and fetal thrombocy-
10. Singh A, Solanki A, Chaudhary R. Demonstration of IgG topenia. Semin Fetal Neonatal Med. 2007;12(6):432-8.
subclass (IgG1 and IgG3) in patients with positive direct 30. Schonewille H, Klumper FJ, van de Watering LM, Kanhai HH,
antiglobulin tests. Immunohematology. 2014;30(1):24-7. Brand A. High additional maternal red cell alloimmunization
11. Vaughan JI, Manning M, Warwick RM, Letsky EA, Murray NA, after Rhesus- and K-matched intrauterine intravascular trans-
Roberts IA. Inhibition of erythroid progenitor cells by anti-Kell fusions for hemolytic disease of the fetus. Am J Obstet Gynecol.
antibodies in fetal alloimmune anemia. N Engl J Med. 1998; 2007;196(2):143.e1-6.
338(12):798-803. 31. Schonewille H, Prinsen-Zander KJ, Reijnart M, van de Watering L,
12. Lindenburg IT, Smits-Wintjens VE, van Klink JM, Verduin E, Zwaginga JJ, Meerman RH, et al. Extended matched intrauterine
van Kamp IL, Walther FJ, et al. Long-term neurodevelopmental transfusions reduce maternal Duffy, Kidd, and S antibody for-
outcome after intrauterine transfusion for hemolytic disease of mation. Transfusion. 2015;55(12):2912-9.
the fetus/newborn: the LOTUS study. Am J Obstet Gynecol. 32. Radunovic N, Lockwood CJ, Alvarez M, Plecas D, Chitkara U,
2012;206(2):141.e1-8. Berkowitz RL. The severely anemic and hydropic isoimmune
13. Rath ME, Smits-Wintjens VE, Walther FJ, Lopriore E. Hema- fetus: changes in fetal hematocrit associated with intrauterine
tological morbidity and management in neonates with death. Obstet Gynecol. 1992;79(3):390-3.
hemolytic disease due to red cell alloimmunization. Early Hum 33. Van Kamp IL, Klumper FJ, Oepkes D, Meerman RH, Scherjon
Dev. 2011;87(9):583-8. SA, Vandenbussche FP, et al. Complications of intrauterine
14. Herschel M, Karrison T, Wen M, Caldarelli L, Baron B. Isoim- intravascular transfusion for fetal anemia due to maternal
munization is unlikely to be the cause of hemolysis in ABO- red-cell alloimmunization. Am J Obstet Gynecol. 2005;192(1):
incompatible but direct antiglobulin test-negative neonates. 171-7.
Pediatrics. 2002;110(1 Pt 1):127-30. 34. Lindenburg IT, van Kamp IL, van Zwet EW, Middeldorp JM,
15. Moise KJ Jr., Argoti PS. Management and prevention of red cell Klumper FJ, Oepkes D. Increased perinatal loss after intrauter-
alloimmunization in pregnancy: a systematic review. Obstet ine transfusion for alloimmune anaemia before 20 weeks of
Gynecol. 2012;120(5):1132-9. gestation. BJOG. 2013;120(7):847-52.
16. Fung MK, Grossman BJ, Hillyer CD, Westhoff CM, editors. 35. Sulochana PV, Rajesh A, Mathai J, Sathyabhama S. Blocked D
Technical manual. 18th ed. Bethesda (MD): AABB Press; 2014. phenomenon, a rare condition with Rh D haemolytic disease
17. Judd WJ, Scientific Section Coordinating Committee of the of newborn—a case report. Int J Lab Hematol. 2008;30(3):
AABB. Practice guidelines for prenatal and perinatal immuno- 244-7.
hematology, revisited. Transfusion. 2001;41(11):1445-52. 36. Management of hyperbilirubinemia in the newborn infant 35
18. Szkotak AJ, Lunty B, Nahirniak S, Clarke G. Interpretation of or more weeks of gestation. Pediatrics. 2004;114(1):297-316.
pretransfusion testing in obstetrical patients who have received 37. Ebbesen F, Hansen TWR, Maisels MJ. Update on photother-
antepartum Rh immunoglobulin prophylaxis. Vox Sang. apy in jaundiced neonates. Curr Pediatr Rev. 2017;13(3):
2016;110(1):51-9. 76-180.
19. Judd W, Johnson ST, Storry JR. Judd’s methods in immunohe- 38. Koelewijn JM, Vrijkotte TG, van der Schoot CE, Bonsel GJ, de
matology. 3 ed. Bethesda (MD): AABB Press; 2008. Haas M. Effect of screening for red cell antibodies, other than
20. AuBuchon JP, de Wildt-Eggen J, Dumont LJ. Reducing the anti-D, to detect hemolytic disease of the fetus and newborn:
variation in performance of antibody titrations. Arch Pathol a population study in the Netherlands. Transfusion. 2008;
Lab Med. 2008;132(7):1194-201. 48(5):941-52.
21. Bachegowda LS, Cheng YH, Long T, Shaz BH. Impact of 39. Bowman JM. Controversies in Rh prophylaxis. Who needs Rh
uniform methods on interlaboratory antibody titration vari- immune globulin and when should it be given? Am J Obstet
ability: antibody titration and uniform methods. Arch Pathol Gynecol. 1985;151(3):289-94.
Lab Med. 2017;141(1):131-8. 40. Brinc D, Lazarus AH. Mechanisms of anti-D action in the
22. Moise KJ. Fetal anemia due to non-Rhesus-D red-cell alloim- prevention of hemolytic disease of the fetus and newborn.
munization. Semin Fetal Neonatal Med. 2008;13(4):207-14. Hematology Am Soc Hematology Educ Program. 2009;2009:
23. Finning K, Martin P, Summers J, Daniels G. Fetal genotyping 185-91.
for the K (Kell) and Rh C, c, and E blood groups on cell- 41. ACOG practice bulletin. Prevention of Rh D alloimmunization.
free fetal DNA in maternal plasma. Transfusion. 2007;47(11): Number 4, May 1999 (replaces educational bulletin Number
2126-33. 147, October 1990). Clinical management guidelines for
24. Finning KM, Martin PG, Soothill PW, Avent ND. Prediction of obstetrician-gynecologists. American College of Obstetrics and
fetal D status from maternal plasma: introduction of a new non- Gynecology. Int j Gynaecol Obstet. 1999;66(1):63-70.
invasive fetal RHD genotyping service. Transfusion. 2002; 42. Sandler SG, Delaney M, Gottschall JL. Proficiency tests reveal
42(8):1079-85. the need to improve laboratory assays for fetomaternal
6888_Ch20_427-440 29/10/18 5:39 PM Page 440

440 PART III Transfusion Practices

hemorrhage for Rh immunoprophylaxis. Transfusion. 2013; patients with a serologic weak D phenotype. Transfusion.
53(9):2098-102. 2015;55(3):680-9.
43. Flegel WA. How I manage donors and patients with a weak D 45. Koelewijn JM, de Haas M, Vrijkotte TG, van der Schoot CE,
phenotype. Curr Opin Hematol. 2006;13(6):476-83. Bonsel GJ. Risk factors for RhD immunisation despite antena-
44. Sandler SG, Flegel WA, Westhoff CM, Denomme GA, Delaney M, tal and postnatal anti-D prophylaxis. BJOG. 2009;116(10):
Keller MA, et al. It’s time to phase in RHD genotyping for 1307-14.
6888_Ch21_441-475 29/10/18 5:38 PM Page 441

Chapter 21
Autoimmune Hemolytic Anemias
Denise M. Harmening, PhD, MT(ASCP), CLS(NCA); Karen Rodberg,
MBA, MT(ASCP)SBB; and Ralph E. B. Green, B. App. Sci, FAIMS, MACE

Introduction Selection of Blood for Transfusion Autoantibody Formation


Autoantibodies Treatment of WAIHA Treatment of Drug-Induced Immune
Definition Mixed-Type Autoantibodies Hemolytic Anemia
Characterization IgM Warm Autoantibodies Case Studies
Cold-Reactive Autoantibodies DAT-Negative Autoimmune Hemolytic Case 21-1
Benign Cold Autoantibodies Anemia Case 21-2
Other Cold-Reactive Autoagglutinins Drug-Induced Immune Hemolytic Anemia Case 21-3
Pathological Cold Autoagglutinins Drug-Adsorption (Hapten) Mechanism Summary Chart
Warm Autoantibodies Drug-Dependent or Immune Complex Review Questions
Clinical Findings (“Innocent Bystander”) Mechanism References
RBC Hemolysis Membrane Modification
(Nonimmunologic Protein
Serologic Characteristics and Adsorption)
Laboratory Tests Affected

OBJECTIVES
1. Define autoantibody and compare the types of immune hemolytic anemias with respect to thermal amplitude, method of
red blood cell (RBC) destruction, and the type of immunoglobulin that characteristically coats the RBCs.
2. Characterize autoantibodies that react at temperatures below 37°C and identify the common specificities of benign cold
autoagglutinins.
3. Describe problems encountered in the laboratory testing of specimens containing cold autoagglutinins.
4. Explain the techniques used to investigate serologic findings and detect underlying clinically significant alloantibodies in the
presence of cold autoantibodies.
5. Describe pathological cold autoagglutinins, including laboratory testing and treatment.
6. Differentiate between idiopathic warm autoimmune hemolytic anemia (WAIHA) and drug-induced immune hemolytic
anemia.
7. Describe the clinical and laboratory findings in WAIHA, including indicators of RBC hemolysis, difficulties in serologic testing,
and selection of blood for transfusion.
8. Explain procedures used to investigate serologic findings and detect underlying clinically significant alloantibodies in the
presence of warm autoantibodies.
9. Compare the mechanisms for drug-induced hemolysis and give examples of medications causing each type.
10. Describe the Donath-Landsteiner test and the clinical situations in which it is useful.

441
6888_Ch21_441-475 29/10/18 5:38 PM Page 442

442 PART III Transfusion Practices

Introduction the presence of autoantibody alone does not necessarily


cause decreased RBC survival.1
Immune hemolytic anemia is defined as shortened red blood Studies in animal models suggest that production of
cell (RBC) survival mediated through the immune response, antibodies against the self occurs because the immune reg-
specifically by humoral antibody. Immune hemolysis is ulatory responses fail.2,3 According to this model, under
destruction of RBCs as a result of antibody production and normal circumstances, immunoglobulins made by B lym-
is an acquired characteristic of the RBC membrane associated phocytes are regulated by T lymphocytes. Helper T cells as-
with demonstrable antibodies, as opposed to intracorpus- sist these immunocompetent B cells in making antibody
cular defects such as enzyme deficiencies and hemoglo- against foreign antigens. Another population of T lympho-
binopathies, which represent intrinsic abnormalities of the cytes, suppressor T cells, has the opposite effect on B-cell
patient’s RBCs. activity; they prevent excessive proliferation of B cells
The three broad categories of immune hemolytic ane- and overproduction of antibodies. Suppressor T cells are
mias are: thought to act through a feedback mechanism. An increas-
1. Alloimmune ing concentration of antibody activates these T cells and
2. Autoimmune suppresses further antibody production.4
3. Drug-induced In a normal individual, it is theorized that autoantibody
production is prevented through a similar mechanism. Sup-
In an alloimmune response, the patient produces anti- pressor T cells induce tolerance to self-antigens by inhibit-
bodies to foreign or non-self RBC antigens introduced into ing B-cell activity. Conversely, loss of suppressor T-cell
the circulation through transfusion, transplant, or pregnancy. function could result in autoantibody production. Support
In cases in which an alloantibody clears the antigen-positive for this concept comes from animal studies3 and patients
RBCs from circulation, intravascularly or extravascularly, a taking the drug alpha-methyldopa.5 The cause of the regu-
transient hemolytic process may result (e.g., a hemolytic latory system’s dysfunction is not understood, but microbial
transfusion reaction), sometimes causing anemia in the agents and drugs have been suggested.6 For further discus-
transfusion recipient or the affected fetus. For a discussion sion of the immune response, see Chapter 3, “Fundamentals
of alloantibody production, refer to Chapter 10, “Detection of Immunology.”
and Identification of Antibodies”; Chapter 17, “Adverse Recognition of autoantibodies in routine serologic in-
Effects of Blood Transfusion”; and Chapter 20, “Hemolytic vestigations is extremely important. Identifying an auto-
Disease of the Fetus and Newborn (HDFN).” antibody may explain decreased RBC survival in vivo to
This chapter focuses on the latter two categories of im- offer an explanation for an otherwise unexplained anemia.
mune hemolytic anemias: autoimmune hemolytic anemia In serologic investigations, the presence of an autoantibody
(AIHA) and drug-induced immune hemolytic anemia may complicate some aspects of routine testing, including
(DIIHA). An autoimmune response occurs when a patient ABO/Rh typing, antibody screening, and compatibility test-
produces antibodies against his or her own RBC antigens. A ing. If a patient’s RBCs are coated with autoantibody, the
drug-induced hemolytic anemia is the result of a patient’s patient may present with a serologic ABO discrepancy,
production of antibody to a particular drug or drug complex, a positive Rh control, or a positive DAT. If the patient’s
with ensuing damage to the patient’s RBCs. serum contains autoantibody, RBC antibody screening pro-
cedures, compatibility testing, and serum (reverse) ABO
Autoantibodies typing of the individual may require additional testing to
resolve. It is likely that the patient’s serum will be incom-
A serologist who is able to recognize and investigate both patible with all random donors. Techniques that can be
alloantibodies and autoantibodies can assist the clinician in used to resolve these difficulties are discussed in greater
both the diagnosis of immune hemolytic anemia and the detail below. Serologic and clinical problems can be found
proper selection of blood components for transfusion. separately or together.

Definition Characterization

Antibodies that are directed against the individual’s own In individuals who have not been recently transfused, the
RBCs are termed autoantibodies or autoagglutinins. Most presence of a positive DAT, a positive autocontrol, or serum
autoantibodies react with high-incidence RBC antigens. autoantibody does not necessarily confer the diagnosis of
They typically agglutinate, sensitize, or lyse RBCs of most autoimmune hemolytic anemia (AIHA). In themselves,
random donors as well as their own. RBC survival may be these findings merely indicate the presence of autoantibody.
shortened by this circulating humoral antibody. Some indi- It has been reported that approximately 0.1% of normal
viduals will produce an autoantibody that readily attaches blood donors and up to 15% of hospitalized patients have
to their own RBCs but does not cause RBC destruction. positive DATs with no evidence of hemolytic anemia.7
Approximately 1 in 1,000 healthy blood donors will have a Before the presence of an AIHA is established, it is important
positive direct antiglobulin test (DAT) but will have a nor- to verify the presence of immune-mediated RBC destruc-
mal hematocrit and be asymptomatic. In these individuals, tion. Evidence of increased RBC destruction is not always
6888_Ch21_441-475 29/10/18 5:38 PM Page 443

Chapter 21 Autoimmune Hemolytic Anemias 443

accompanied by decreased hemoglobin and hematocrit lev- It should be noted that column agglutination (gel) or
els. These may become evident only when the individual is solid-phase methodologies may have an increased sensitivity
no longer able to compensate for increased RBC destruction. in the detection of IgG-coated RBCs. Therefore, with the
In many instances, even before decreases in hemoglobin and increase in the use of methods other than conventional test
hematocrit are obvious, there will be evidence of compen- tube technique, it is possible that a greater number of indi-
sation for increased RBC destruction, including increases in viduals may be found to be DAT-positive, with or without
reticulocyte count, unconjugated (indirect) bilirubin levels, detectable evidence of RBC destruction. As mentioned
and lactate dehydrogenase (LDH) levels, and a decrease in above, the incidence of positive DATs in normal blood donor
haptoglobin. populations has been reported to be as high as 1 in 1,000 in
The individual who has immune RBC destruction may the U.S. population,1 whereas the incidence in hospitalized
experience either compensated or uncompensated anemia. patients ranges from 0.3% to 1%, using anti-IgG antiglobulin
In compensated anemia, the rate of RBC production will reagent,10,11 and as high as 15%, using polyspecific antiglob-
nearly equal the rate of RBC destruction. These individuals ulin reagent.12 Many of the latter group have only comple-
will demonstrate an increased reticulocyte count but may ment bound to the RBCs. The differences between individuals
have only a mild decrease in hemoglobin and hematocrit who are affected (i.e., those who have AIHA) and those who
levels, depending on the rate of RBC production. Those in- are unaffected by autoantibodies are not clearly understood.
dividuals with uncompensated anemia demonstrate a rate of Among the possibly significant factors are:
RBC destruction that exceeds the rate of RBC production.
• Thermal amplitude of antibody reactivity13
Hemolytic anemia is often demonstrated in a blood smear
• IgG subclass of the antibody14
by macrocytosis (evidence of a young cell population) or
• Amount of antibody bound to the RBCs14
spherocytosis (evidence of cell membrane damage). The
• Ability of the antibody to fix complement in vivo15
reticulocyte count of these individuals is generally greater
• Activity of the individual’s macrophages7
than 3%,8 and the unconjugated bilirubin levels and LDH
• Quantitative or qualitative change in band 3 and proteins
levels are also increased due to accelerated RBC destruction;
4.1 and 4.2 in the RBC membrane structure16
however, haptoglobin levels are markedly decreased due
to its role of clearing hemoglobin from the plasma. With The opposite situation also occurs; in some patients with
intravascular RBC destruction, hemoglobinemia and hemo- hemolytic anemia, autoantibodies cannot be demonstrated
globinuria may occur. by routine techniques. Some patients have more IgG than
Because there are other causes of hemolysis (e.g., hered- normal on their RBCs but less than the amount detectable
itary spherocytosis, hemoglobinopathies, and RBC enzyme by the routine antiglobulin test.17 In other cases, the patient’s
defects), AIHA must be confirmed by additional serologic RBCs are sensitized with anti-IgA or anti-IgM, not routinely
testing. Diagnostic tests in a symptomatic patient include: demonstrable using commercial antiglobulin reagents.18–21
Because polyspecific antihuman globulin (AHG) reagents are
1. Direct antiglobulin test (DAT) using polyspecific and
required by the U.S. Food and Drug Administration (FDA)
monospecific antiglobulin reagents (refer to Chapter 5,
to contain anti-IgG and anti-C3d,21,22 antibodies to other
“The Antiglobulin Test”)
immunoglobulins (e.g., IgA and IgM) are not consistently
2. Characterization of the autoantibody in the serum or elu-
present in the reagent, and cells sensitized with IgA or IgM
ate using standard antibody detection and identification
may not give a positive direct antiglobulin test.
procedures (refer to Chapter 10)
Autoantibodies can be characterized by their optimal
Based on these results and the clinical evaluation of temperature of reactivity. About 70% of the reported cases of
the patient, AIHA may be diagnosed and classified as cold AIHA are those that react best at warm temperatures (30°C
reactive, warm reactive, mixed-type, or drug-induced. The to 37°C), whereas cold-reactive (4°C to 30°C) autoagglu-
expected laboratory findings for each type are discussed in tinins account for about 18%. Drug-induced autoagglutinins
the following sections. In their book Immune Hemolytic are present in about 12% of the reported cases of AIHA.
Anemias, Petz and Garratty devote several chapters to the Characterization of autoantibodies is important because
differential diagnosis of the hemolytic anemias and another treatment of the patient and resolution of the serologic prob-
chapter to drug-induced immune hemolytic anemia. The lems will differ according to the optimal temperature of
reader is referred to their text for a complete discussion.7 reactivity and the immunoglobulin responsible; treatment of
As previously stated, some individuals may have auto- the patient will depend on the nature of the autoantibody.
antibodies in their sera and on their RBCs but display no The remainder of this chapter details the clinical and labo-
evidence of decreased RBC survival. All normal RBCs have ratory aspects of cold-reactive, warm-reactive, and drug-
a small amount of IgG and complement on their surfaces. induced autoantibodies.
Studies have shown that there may be 5 to 90 molecules of
IgG/RBC and 5 to 500 molecules of complement/RBC in the Cold-Reactive Autoantibodies
average healthy individual.7 Because conventional tube test-
ing will normally detect 100 to 500 molecules of IgG/RBC Cold-reactive autoantibodies are frequently encountered in
and 400 to 1,100 molecules of complement/RBC, these serologic testing. Most of the time, the antibodies detected
individuals are not routinely identified as DAT-positive.9 are not clinically significant but nevertheless can present
6888_Ch21_441-475 29/10/18 5:38 PM Page 444

444 PART III Transfusion Practices

challenges for the blood banker. Occasionally, cold auto- concentration and thermal amplitude of the antibody).
antibodies are clinically significant and cause immune he- Although the cold autoantibodies found in the serum of
molytic anemia. The antibody specificity may be of interest normal people do not typically interfere with testing, they
to the technologist or may be a clue to the disease process, are one of the more common causes of serologic problems;
but the important distinction between clinically significant therefore, one should be able to recognize these circum-
and benign cold autoantibodies is their thermal range. Any stances and employ methods to resolve problems associated
antibody that reacts at or near body temperature (30°C to with these antibodies. If a cold agglutinin is strong enough
37°C), regardless of specificity, is potentially clinically sig- to interfere with ABO typing, it may also interfere with indi-
nificant; this includes “cold” autoantibodies. rect antiglobulin testing even if the antibody detection
method does not intentionally include an RT incubation
Benign Cold Autoantibodies step. In these cases, troubleshooting a positive antibody
screening should include RT testing to prove the existence
When testing is performed at 4°C, the most commonly of a cold-reacting antibody.
encountered autoantibody is a benign cold agglutinin that
may be found in the serum of most normal, healthy individ- ABO Typing
uals. Normally these antibodies present no serologic problem
If an individual’s RBCs are heavily coated with cold agglu-
because routine antibody detection tests are not performed
tinins, they may directly agglutinate, causing false-positive
at this temperature. The typical cold agglutinin has a rela-
reactions with routine ABO reagents. In most cases, valid
tively low titer (less than 64 at 4°C). Occasionally, these
results can be obtained by using patient’s cells that have been
benign cold autoantibodies may agglutinate cells at room
washed once or twice with normal saline warmed to 37°C.
temperature (20°C to 24°C); however, even in this situation,
The cold autoantibody is eluted from the cells during warm
strongest reactivity is found at 4°C. Table 21–1 compares the
washing. For example, group O cells coated with cold
characteristics of benign (normal) cold autoantibodies with
autoantibody might show the following reactions before and
those of pathological cold autoantibodies, which are dis-
after warm washing:
cussed later in this chapter. Most cold agglutinins react
strongly with enzyme-treated cells; therefore, cold agglu- Anti-A Anti-B
tinins are quite likely to be detected when ficin- or papain- Serum-suspended RBCs 1+ 1+
treated cells are tested. Cold autoantibodies are IgM
immunoglobulins and therefore can activate complement in Warm-washed, saline-suspended RBCs 0 0
vitro. In serum testing, reactivity may be seen in the antiglob-
ulin phase if polyspecific antihuman serum (i.e., containing If more potent autoagglutinins are present, it may be nec-
anticomplement) is used in antiglobulin testing. Table 21–2 essary to incubate the patient’s whole blood sample at 37°C
contrasts titers and thermal amplitudes typical of benign prior to warm washing.9 In some cases, it may be inconve-
versus pathological cold autoagglutinins. nient to adhere closely to a protocol of maintaining the sam-
ple at 37°C from the point of collection; similar success has
Laboratory Tests Affected by Cold Autoagglutinins been found with warming the sample in a water bath or heat
Cold agglutinins sometimes interfere with routine serum and block to 37°C for 15 to 30 minutes and then warm washing
cell testing performed at room temperature (RT). The extent the RBCs. The warm incubation elutes the cold agglutinin
to which they cause problems depends on whether antibody from the patient’s RBCs, and the warm saline washes remove
detection tests are performed at room temperature and how it to prevent it from reattaching in vitro. In the rare situation
strongly the antibody reacts at this temperature (i.e., the in which washing with warm saline is not effective, thiol

Table 21–1 Characteristics of Normal (Benign) Cold and Pathological Cold Autoantibodies
Characteristic Normal (Benign) Pathological
Thermal range Reactive ≤20°–24°C Reactive at ≥30°C

Titer at 4°C ≤64 ≥1000

Reactivity Marginally enhanced with albumin Strongly enhanced with albumin

Common specificity Anti-I or Anti-IH Anti-I

Capable of binding complement Yes (in vitro) Yes (in vivo)


DAT: 0 – 1+ due to C3 DAT: 2 – 4+ due to C3

Clinically significant No Yes

Associated with disease No Yes. May be secondary to viral infections or M. pneumoniae


6888_Ch21_441-475 29/10/18 5:38 PM Page 445

Chapter 21 Autoimmune Hemolytic Anemias 445

Table 21–2 Benign Cold Agglutinin Compared to Pathological Cold Agglutinin: Titrations
Performed Using Normal Group O RBCs
Benign Cold Agglutinin

Tube no. 1 2 3 4 5 6 7 8 9 10 11 12 Titer

Dilution → N 2 4 8 16 32 64 128 256 512 1024 2048


Temp ↓

37°C 0 0 0 0 0 0 0 0 0 0 0 0 0

30°C 0 0 0 0 0 0 0 0 0 0 0 0 0

20°C 3+ 2+ 1+ 0 0 0 0 0 0 0 0 0 4

4°C 4+ 4+ 3+ 2+ 1+ 0 0 0 0 0 0 0 16

Pathological Cold Agglutinin

Tube No. 1 2 3 4 5 6 7 8 9 10 11 12 Titer

Dilution → N 2 4 8 16 32 64 128 256 512 1024 2048


Temp ↓

37°C 1+ ± 0 0 0 0 0 0 0 0 0 0 1

30°C 2+ 1+ 0 0 0 0 0 0 0 0 0 0 2

20°C 4+ 4+ 3+ 3+ 2+ 1+ 0 0 0 0 0 0 32

4°C 4+ 4+ 4+ 4+ 4+ 3+ 3+ 3+ 2+ 2+ 1+ 1+ >2048

reagents (e.g., dithiothreitol) can be used to disperse the and the tests with the A1, B, O and autologous cells are
autoagglutination.23 repeated with autoadsorbed serum.
See Procedure 21–1 on the textbook’s companion See Procedure 21–2 on the textbook’s companion
website. website.
Because serum ABO grouping is performed at room tem- Although it may be possible to resolve this discrepancy
perature, cold autoagglutinins frequently cause discrepancies with prewarmed testing, one must remember that not all
in the serum ABO (reverse) typing also. In the following ABO isoagglutinins are reactive at 37°C, and erroneous test
example, the cell-typing results suggest the cells are group results may be obtained; therefore, autoadsorption proce-
AB. In a group AB individual, one does not expect the serum dures are preferred.
to agglutinate either the A1 or B cells, as seen in this example.
Autologous
Although a number of explanations for this discrepancy A1 RBCs B RBCs O RBCs RBCs
exist, a cold agglutinin is the most likely cause. Group O and
autologous cells should also be tested in the investigation of Cold Auto- 0 0 0 0
a serologic ABO discrepancy. If a cold autoagglutinin is pres- adsorbed
Serum
ent, the autologous and group O cells (the negative controls)
will most likely be positive as well:
RhD Typing
Anti-A Anti-B
As in ABO cell grouping, false-positive reactions can be seen
RBCs 4+ 4+ with Rh reagents when RBCs coated with cold autoagglu-
tinins are tested. This was previously a common problem en-
Autologous
countered when typing with polyclonal high-protein anti-D
A1 RBCs B RBCs O RBCs RBCs
reagent was the routine, where the anti-D and the Rh control
Serum (or 1+ 1+ 2+ 1+ were both positive, rendering the test invalid. Today, the Rh
Plasma) reagents in common use are monoclonal or a blend of mon-
oclonal and low-protein anti-D reagents, which normally
Such a discrepancy is easily resolved if the cold-reactive yield valid results. When a monoclonal reagent is used, many
autoantibody is removed by a 4°C autoadsorption technique, manufacturers consider a negative reaction with any of the
6888_Ch21_441-475 29/10/18 5:38 PM Page 446

446 PART III Transfusion Practices

ABO reagents as a control for the D typing; however, an Rh Antibodies reactive only at room temperature are usually
control reagent is available from some manufacturers. As considered to be of no clinical significance. Benign cold
stated above, if a discrepancy exists in the ABO typing, wash- autoagglutinins do not react at 37°C, but they may inter-
ing the cells with warm saline prior to testing usually gives fere with testing at the antiglobulin phase if polyspecific
acceptable results. This also holds true when testing with antihuman reagent is used, inasmuch as they may bind
low-protein anti-D. Thiol reagents may be required when to cells at lower temperatures when the serum and
washing with warm saline is ineffective. cells are mixed together initially or during centrifugation
Cold-reactive IgM autoagglutinins can activate the com- following the 37°C incubation, and complement may
plement cascade in vitro, causing complement components be activated. Although the antibody elutes from the cell
to be bound to the RBC surface, which can lead to false- surface during the incubation or washing phases, the com-
positive reactions in the weak D (antiglobulin) test if cells plement remains bound. Polyspecific antihuman serum
from a clotted sample and polyspecific antihuman serum are will agglutinate the cells coated with complement. When
used. In this instance, the Rh control test will also be posi- enzyme-treated cells are used, reactions in all phases may
tive. The use of monospecific anti-IgG for weak D testing or be stronger. Often, omitting room temperature incubation
RBCs collected into ethylenediaminetetraacetic acid (EDTA) and the use of anti-IgG for the indirect antiglobulin test
can eliminate the problem of complement-binding cold (IAT) are all that is needed to avoid detecting cold agglu-
agglutinins in D typing. The following examples illustrate tinins by the IAT.
these results: Because most clinically significant antibodies capable of
causing accelerated RBC destruction are detected by the
Anti-D RH Control
antiglobulin test, reactions in this phase must be investi-
Indirect antiglobulin phase 1+ 1+ gated. Reactivity caused by cold autoagglutinins can mask
(poly AHG) the presence of clinically significant alloantibodies. Although
Indirect antiglobulin phase 0 0 the use of anti-IgG antiglobulin reagents will eliminate most
(anti-IgG) problems with cold autoagglutinin reactivity in the IAT
phase, it may be necessary to remove the cold-reacting au-
RBCs collected in EDTA 0 0
toantibody by cold adsorption procedures to thoroughly in-
vestigate the reactivity observed in antiglobulin testing.
Similar problems can be encountered with other antisera Rabbit erythrocyte stroma is known to be rich in I antigen
used in RBC phenotyping (e.g., anti-K, anti-S, anti-Fya) that and is commercially available and easy to use. There have
require the use of an indirect antiglobulin test. The use of been some reports that clinically significant antibodies have
anti-IgG antiglobulin reagent or a sample collected in EDTA been adsorbed using rabbit erythrocyte stroma, so caution is
is recommended when cold autoagglutinins are present. For- advised.24
tunately, most samples collected for the blood bank today Other techniques useful in differentiating between cold
are collected in EDTA. autoantibodies and alloantibodies are the prewarming tech-
nique and testing with cold autoadsorbed serum.9 Cold au-
Direct Antiglobulin Test toadsorption technique is preferred because the prewarm test
When a properly collected specimen is used (EDTA- has become quite controversial. There have been reports of
anticoagulated RBCs), the DAT of a patient with benign clinically significant alloantibodies being unintentionally
cold autoagglutinins is usually negative; however, one “prewarmed away,” either due to poor technique or because
frequently obtains a positive result using polyspecific the antibodies were partially IgM, yet this technique persists
antihuman serum if a clotted specimen is used because in practice and does have value in investigating an identified
complement can be activated in vitro. This in vitro sensi- antibody’s thermal amplitude.
tization is usually weak—less than or equal to 1+. If mono- The principle of the prewarm test is that by first warming
specific reagents are used, then these cells are reactive with the cell suspension and serum prior to mixing, avoiding
anti-C3d but not with anti-IgG. A negative control of 6% room temperature centrifugation after 37°C incubation, and
albumin or saline, used in parallel with the antiglobulin washing with saline warmed to 37°C, any reaction between
reagents, is recommended for direct antiglobulin testing, the cold autoagglutinin and RBC antigens can be prevented,
particularly when there has been difficulty obtaining valid thus avoiding complement activation. While cold-reacting
ABO or Rh typing. antibodies should not react by this method, alloantibodies
that are reactive at 37°C (i.e., potentially clinically signifi-
Antibody Detection and Identification cant) would still bind to the cells and be detectable as direct
The frequency with which cold autoagglutinins interfere agglutinins at 37°C or at the antiglobulin phase.
with detection and identification of RBC alloantibodies
See Procedure 21-3 on the textbook’s companion
depends to a large extent on the routine procedures used in
website.
patient testing. As shown in Table 21–2, cold agglutinins often
react at room temperature (RT) and are enhanced at 4°C, but An example of the results of testing a serum that contains
they are generally not detected because routine antibody a cold autoagglutinin and anti-Fya using routine antiglobulin
screening is no longer performed at these temperatures. testing with polyspecific antiglobulin and testing with the
6888_Ch21_441-475 29/10/18 5:38 PM Page 447

Chapter 21 Autoimmune Hemolytic Anemias 447

prewarmed technique is shown in Table 21–3. Reactions are enzyme-treated cells. Autologous adsorption should not be
present in routine antiglobulin tests with both Fy(a+) and performed if a patient has been recently transfused, because
Fy(a–) cells. In a prewarmed test, only the reactions ex- donor RBCs present in the patient’s circulation will also be
pected of the anti-Fya are evident. The weak reactions of the present in the sample tube and could inadvertently adsorb
cold autoagglutinin are eliminated by the prewarm tech- alloantibody. In this situation, it is best to use allogeneic
nique. The prewarm technique is simple and successful in adsorption or cold adsorption using rabbit erythrocyte stroma.
most cases; however, if the cold autoantibody is very potent,
See Procedure 21-4 on the textbook’s companion
it may still be difficult to avoid the antigen–antibody inter-
website.
action and complement activation.
Although the prewarm technique may appear to be very If anti-IgG antiglobulin reagent is used, the problems
helpful in resolving problems caused by cold autoagglu- caused by most cold agglutinins can be avoided. The use of
tinins, this technique should not be used indiscriminately. anti-IgG reagents is an attractive alternative when prewarm-
Cases have been reported in which clinically significant al- ing is not effective and there is not enough time to adsorb
loantibodies have been missed after prewarming.25 Prewarm- the serum.
ing should be used only when the reactions obtained indicate
the likely presence of a cold autoagglutinin (i.e., positive au- Compatibility Testing
tocontrol and reactivity noted below 37°C). Because there is The difficulties encountered in antibody detection and
no immediate spin reading or room temperature incubation identification tests are also found in compatibility tests,
in the prewarmed procedure, IgM immunoglobulin compo- because the most commonly encountered cold autoanti-
nents of a newly forming alloantibody may not be detected body (autoanti-I) is directed against an antigen that is
in this testing. Therefore, this testing must not be performed found on the RBCs of most random donors and on most
in patients transfused within the previous 3 months or in pa- reagent RBCs. Compatibility tests, like antibody identifica-
tients without an accurate transfusion history. A cold autoag- tion tests, can be performed using prewarmed or autoad-
glutinin is not apt to be the answer if weak reactions are sorbed serum or allogeneic adsorbed serum or using
present only in the antiglobulin phase with anti-IgG (i.e., anti-IgG antiglobulin reagent.
there is no evidence of reactivity at immediate spin or room Two other common cold autoagglutinins, anti-IH and
temperature). anti-H, distinguish between group O reagent RBCs and
When strong cold autoantibodies are present, or if one group A, B, or AB donor cells. Because these antibodies react
wishes to identify a room temperature–reactive alloantibody, with the H antigen that is only weakly expressed on the RBCs
autologous adsorption must be performed to remove the of group A, B, or AB individuals, they are not always obvious
autoantibody. Cold autologous adsorption, described in Pro- as autoantibodies when encountered. As discussed in the
cedure 21–2 on the companion website, may be performed following section on specificity, autoanti-IH and autoanti-H
if the patient has not been transfused within 3 months. In react best with group O cells; they react least with group A1
autoadsorption procedures, an aliquot of patient cells is and A1B cells, perhaps only at 4°C. Autoanti-IH and autoanti-
incubated with an equal volume of the patient’s serum at 4°C. H are found most often in the serum of group A1 and A1B
Autoantibody is adsorbed onto the cells, and alloantibody persons; therefore, the units selected for compatibility testing
remains in the serum. It may be necessary to repeat the (group A or AB) are those that give the weakest, if any,
adsorption several times if the autoantibody is of high titer. reactivity. On the other hand, group O cells (e.g., antibody
In order to enhance the adsorption process, the patient’s RBCs screening cells) give the strongest reactions.
may be treated with enzymes before adsorption to increase
the amount of autoantibody removed by the adsorption; Specificity of Cold Autoagglutinins
ideally, enzyme pretreatment should not be performed with- The specificity of the cold autoantibody is often associated
out confirming that the serum antibody is reactive with with the patient’s diagnosis. It is important to be familiar

Table 21–3 Typical Reactivity Observed With a Patient Serum Containing a Clinically
Significant Alloantibody (anti-Fya) and a Cold Autoagglutinin
Standard Antiglobulin Prewarmed Antiglobulin
Testing With Polyspecific Testing With Polyspecific Standard Antiglobulin
RBCs Tested AHG AHG Testing With Anti-IgG
Fy(a+b–) 2+ 2+ 2+
Fy(a+b+) 2+ 2+ 2+
Fy(a–b+) 1+W 0 0
Auto control 1+W 0 0
6888_Ch21_441-475 29/10/18 5:38 PM Page 448

448 PART III Transfusion Practices

with the properties and characteristics of these types of when a serum is tested with adult and cord RBCs. Benign
autoantibodies. cold autoantibodies, for example, react with adult RBCs but
not with cord RBCs. Pathological cold autoantibodies react
Anti-I, Anti-i. Most cold-reactive autoantibodies have anti-I with both adult and cord cells, but the preference for the
specificity. The I antigen is fully expressed on the RBCs of adult cells is still obvious. Alloanti-I is frequently present in
virtually all adults, whereas it is only weakly expressed on the serum of i adults.26
cord RBCs. At birth, an infant’s RBCs express the i antigen. Anti-i is a relatively uncommon autoantibody. As shown
As an infant matures, the antigen expressed is converted in Table 21–5, this antibody reacts in a manner antithetical
from the i antigen to the I antigen; the amount of I antigen to anti-I. Cord cells and i adult cells have the strongest ex-
increases until the adult levels are reached at about 2 years pression of the i antigen; adult I cells have the weakest.
of age.9 Very rarely do adult RBCs lack the I antigen; if they
do, they are termed i adults and may produce alloanti-I, Anti-H, Anti-IH. Cold agglutinins found in the sera of group A1
which is a potentially clinically significant alloantibody be- and A1B individuals (and occasionally group B) may have
cause it is typically an IgG antibody that reacts at 37°C. (See anti-H specificity. This antibody distinguishes between cells
Chapter 8 for a review of the Ii blood group system.) of various ABO groups. Group O and A2 cells react best be-
The reactivities of several examples of anti-I are given in cause they have the largest amounts of H antigen. Group A1
Table 21–4. As shown, anti-I specificity may be apparent and A1B cells have the least H antigen, so they react weakly.

Table 21–4 Typical Reactivity Observed at 4°C With Serum Containing Autoanti-I with Iadult
and icord Cells
Serum Serum Dilution Iadult Cells icord Cells
Benign Cold Neat 3+ 1+W

1:2 1+ 0

1:4 1+W 0

1:8 0 0

Pathological Cold Neat 4+ 3+

1:2 4+ 2+

1:4 4+ 1+

1:8 3+ 0

Table 21–5 Reactivity of Cold Autoagglutinins at 4°C With ABO-Compatible RBCs


RBC Phenotype Anti-I Anti-i Anti-H Anti-IH Anti-Pr*
Group O Iadult 4+ 0 to 1+ 4+ 4+ 4+
Group A1 Iadult 4+ 0 to 1+ 0 to 1+ 0 to 1+ 4+

Group A2 Iadult 4+ 0 to 1+ 2+ 2+ 4+

Bombay Oh Iadult 4+ 0 to 1+ 0 0 to 2+ 4+

Group O iadult 0 to 1+ 4+ 4+ 0 to 1+ 4+

Group O icord 0 to 1+ 4+ 4+ 0 to 1+ 4+

Group A1 iadult 0 to 1+ 4+ 0 to 1+ 0 4+

Group A2 iadult 0 to 1+ 4+ 2+ 0 to 2+ 4+

Bombay Oh iadult 0 to 1+ 4+ 0 0 4+

Group O Iadult ficin-treated 4+ 1+ to 2+ 4+ 4+ 0

*Anti-Pr is a less commonly encountered cold autoagglutinin that frequently mimics autoanti-I.
6888_Ch21_441-475 29/10/18 5:38 PM Page 449

Chapter 21 Autoimmune Hemolytic Anemias 449

Because the A1 and A1B individual’s own cells demonstrate a hemagglutinin disease (CHD) or idiopathic cold AIHA, rep-
very weak expression of the H antigen, anti-H and anti-IH resents approximately 18% of the cases of AIHA. A moderate
are actually autoantibodies that may react only with auto- chronic hemolytic anemia is produced by a cold autoantibody
logous RBCs at 4°C, but that can be easily managed by the that optimally reacts at 4°C but also reacts at temperatures
selection of type-specific RBCs. The pattern of reactivity seen between 25°C and 30°C. The antibody is usually an IgM im-
with anti-H is shown in Table 21–5. See Chapter 6, “The munoglobulin that quite efficiently activates complement.
ABO Blood Group System,” for a discussion of the ABO
system and H substance. Clinical Picture. CHD occurs predominantly in older individ-
It is very important not to confuse cold-reactive anti-H uals, with a peak incidence in those over 50 years of age.
with the anti-H found in the serum of Oh (Bombay) individ- Antibody specificity in this disorder is almost always anti-I,
uals who altogether lack the H antigen. Cold-reactive anti- less commonly anti-i, and rarely anti-Pr. It is rarely severe
H may be found in A1 or A1B individuals as an autoantibody and is usually seasonal because the winter months often pre-
even though the cells of the antibody maker (A1 or A1B) may cipitate the signs and symptoms of this chronic hemolytic
give considerably weaker reactions. The anti-H in the Oh per- anemia. Acrocyanosis of the hands, feet, ears, and nose is
son is a potent alloantibody that reacts at 4°C to 37°C with frequently the patient’s main complaint along with a sense
all cells except the rare Oh cell and is capable of causing rapid of numbness in the extremities. Changes take place when
intravascular RBC destruction. the person is exposed to the cold because the cold autoanti-
Anti-IH, another of the usually harmless cold autoagglu- body agglutinates the patient’s RBCs as they pass through the
tinins, is also found more commonly in the serum of A1 and skin capillaries, resulting in localized blood stasis. During
A1B individuals. This antibody agglutinates only RBCs that cold winter weather, the temperature of an individual’s blood
have both the I and the H antigens. As with anti-H, group O falls to as low as 28°C in the extremities, activating his or
and group A2 cells react best. The difference between these her cold autoantibody. The antibody then agglutinates the
two antibodies is that group O icord cells and group O iadult RBCs and fixes complement as the cells flow through
cells react as strongly as group O Iadult cells with anti-H but the capillaries of the skin, causing signs of acrocyanosis.
not with anti-IH (see Table 21–5). These patients may also experience hemoglobinuria, because
the complement fixation may result in intravascular hemol-
Other Cold-Reactive Autoagglutinins ysis. Figure 21–1 illustrates the relationship between ambi-
ent temperatures and LDH concentration (a reflection of the
A number of other less commonly encountered cold autoag- severity of hemolysis) over a period of 18 months.30 How-
glutinins have been described, such as anti-Pr, anti-Gd, and ever, this intravascular hemolytic episode is not associated
anti-Sdx (anti-Rx).27 Cold autoantibodies with the specificity with fever, chills, or acute renal insufficiency, any one of
of anti-M have also been described.28 Most researchers agree which is characteristic of patients with paroxysmal cold
that specificity of cold-reactive autoantibodies is primarily hemoglobinuria (PCH) or severe WAIHA.
of academic interest and usually not clinically important; Patients usually display weakness, pallor, and weight loss,
however, development of autoantibodies with specificities which are characteristic symptoms of a chronic anemia. CHD
for integral components of the RBC membrane, such as the usually remains quite stable; however, if it does progress in
glycophorins or band 3, may be precursors for developing severity, it is insidious in intensity. Physical findings such as
other autoimmune disorders such as systemic lupus erythe- hepatosplenomegaly are infrequent because of the mecha-
matosus or rheumatoid arthritis.29 nism of hemolysis. Other clinical features of CHD include
jaundice and Raynaud’s disease (symptoms of cold intoler-
Pathological Cold Autoagglutinins ance, such as pain and bluish tinge in the fingertips and toes
as a result of vasospasm). Patients with severe CHD usually
Differentiating the serologic characteristics of a benign cold live more comfortably in warmer climates.
autoagglutinin from a pathological cold agglutinin may aid
the clinician in making a diagnosis. This is very important Laboratory Findings. Laboratory findings in CHD include retic-
in terms of the treatment that is required for pathological ulocytosis and a positive DAT due to complement only. It is
cold autoantibodies as opposed to no treatment required for suggested that a simple serum-screening procedure be per-
benign cold autoantibodies. formed initially to test the ability of the patient’s serum to
agglutinate autologous saline-suspended RBCs at 18°C to
Cold Hemagglutinin Disease (Idiopathic Cold AIHA) 20°C. If this test result is positive, further steps may be taken
Most cold autoagglutinins do not cause RBC destruction, but to determine the titer and thermal amplitude of the patient’s
in some patients they can cause hemolytic anemia that varies cold autoantibody, which typically reacts as pathological cold
in severity from mild to life-threatening intravascular lysis. autoagglutinins as described in Table 21–2. If it is negative,
Cold-reactive immune hemolytic anemia may be a chronic, the diagnosis of CHD is unlikely. The peripheral smear of a
idiopathic (no identifiable cause) condition or an acute, patient with CHD may show agglutinated RBCs, polychro-
transient disorder often associated with an infectious disease masia, mild to moderate anisocytosis, and poikilocytosis
such as Mycoplasma pneumoniae pneumonia or infectious (Fig. 21–2). Autoagglutination of anticoagulated whole
mononucleosis. Cold agglutinin syndrome, also called cold blood samples is characteristic of CHD and occurs quickly
6888_Ch21_441-475 29/10/18 5:38 PM Page 450

450 PART III Transfusion Practices

500 25

450 20

Mean monthly temperature (C)


400
15
350
10
LDH (IU/L)

300

250 5

200 0
150
⫺5
100
⫺10
50

0 ⫺15
y

er

ry
t

ch

r il

ay

er

r
us

us
be

be

be

be

be

be
l

l
ar

n
Ju

Ju
Ap
ua
ob

ob
M
ar

Ju
g

g
nu
em

em

em

em

em

em
Au

Au
M
br
ct

ct
Ja
O

O
ov

ov
pt

ec

pt

ec
Fe
Se

Se
N

N
D

D
Month

LDH (IU/L) Mean monthly temperature (C)

Figure 21–1. Seasonal hemolysis in cold agglutinin disease. As the ambient temperature decreases, the amount of hemolysis (reflected in serum lactate dehydrogenase
levels) increases. During warmer months, LDH levels return to normal. (Adapted with permission from Lyckholm, LJ, and Edmond, MB: Seasonal hemolysis due to cold-
agglutinin syndrome. N Engl J Med 334:437, 1996.)

BOX 21–1

Clinical Criteria for the Diagnosis of CHD


• Clinical signs of an acquired hemolytic anemia, with a history
(which may or may not be present) of acrocyanosis and hemoglo-
binuria on exposure to cold
• Positive DAT result using polyspecific antihuman sera
• Positive DAT result using monospecific anti-C3 antisera
• Negative DAT result using monospecific anti-IgG antisera
• Presence of reactivity in the patient’s serum due to a cold
autoantibody
• Cold agglutinin titer of 1,000 or greater in saline at 4°C with visible
agglutination of anticoagulated blood at room temperature
Figure 21–2. Cold agglutinin disease (peripheral blood).
Adapted from Harmening, DM: Clinical Hematology and Fundamentals of Hemo-
stasis, 5th ed. FA Davis, Philadelphia, 2009, p 262, with permission.

as the blood cools to room temperature, causing the binding


of cold autoantibodies to the patient’s RBCs in vitro. As a re- method of choice, if possible. It is important to provide RBCs
sult of this autoagglutination, it is extremely difficult to per- compatible with any clinically significant alloantibodies.
form automated blood counts and preparation of blood Most patients tolerate transfusion of blood that is incompat-
smears with these patients’ samples. Leukocyte and platelet ible with the autoantibody. Units of i adult RBCs are
counts are usually normal. Box 21–1 lists the clinical criteria extremely rare and should be reserved for the rare i adult
for diagnosis of CHD. patient with alloanti-I.
The issue of transfusion in CHD patients is most relevant
Selection of Blood for Transfusion. Most patients with CHD do in those undergoing surgical procedures that use hypother-
not require transfusion; however, when they do, it is some- mia (lowering of the body temperature to 22°C to 30°C),
times challenging to perform the pretransfusion testing. As such as cardiac procedures. For patients with CHD, blood
previously described, potent cold autoantibodies interfere for transfusion can be warmed by an approved blood
with most routine tests. Perhaps the most difficult problem warmer, or the operative procedures can be performed with-
is detecting and identifying alloantibodies. Procedures to out subjecting the patient to hypothermia.31 Patients with
manage these problems were described earlier in this chap- benign cold autoagglutinins do not require these special
ter, but cold autoadsorption of the serum should be the arrangements.
6888_Ch21_441-475 29/10/18 5:38 PM Page 451

Chapter 21 Autoimmune Hemolytic Anemias 451

Cold Autoantibodies Related to Infection (Secondary patients develop the antibody of sufficient titer and thermal
Cold AIHA) amplitude to induce in vivo hemolysis.
CHD can also occur as a transient disorder secondary to in-
fection. Episodes of cold AIHA often occur after upper respi- Paroxysmal Cold Hemoglobinuria. Paroxysmal cold hemoglobin-
ratory infections. Approximately 50% of patients suffering uria (PCH) is the least common type of AIHA, with an inci-
from pneumonia caused by M. pneumoniae have cold agglu- dence between 1% and 2%. It is most often seen in children
tinin titers higher than 64. In the second or third week of the who have had viral illnesses such as measles, mumps, chick-
patient’s illness, CHD may occur in association with the in- enpox, infectious mononucleosis, and the ill-defined flu
fection, and a rapid onset of hemolysis is observed. Pallor syndrome. Originally, PCH was described in association
and jaundice are characteristically present. Acrocyanosis with syphilis, with an autoantibody formed in response to
and hemoglobinuria are uncommon and not consistently Treponema pallidum infection; however, with the effective
present. Usually, resolution of the episode occurs within 2 to treatment of syphilis with antibiotics, PCH is no longer com-
3 weeks because the hemolysis is self-limiting, resolving monly reported in relation to syphilis.
when the infection subsides. The offending cold autoantibody In PCH, RBC destruction is caused by a cold autoanti-
is an IgM immunoglobulin with a characteristic anti-I speci- body referred to as a biphasic autohemolysin, which binds
ficity. Very high titers of cold autoagglutinins are seen almost to the patient’s RBCs at lower temperatures and fixes
exclusively in patients with M. pneumoniae. It has been theo- complement. Hemolysis occurs when the sensitized RBCs
rized that the cold agglutinin produced in this infection is an circulate and are exposed to 37°C; the sensitized cells un-
immunologic response to the mycoplasma antigens and that dergo complement-mediated intravascular lysis. In contrast
this antibody cross-reacts with the I antigen on RBCs. to the other cold-reactive autoagglutinins, the antibody in
The antibodies produced in primary CHD and in this dis- PCH is an IgG immunoglobulin with biphasic activity. The
order secondary to M. pneumoniae both have anti-I specificity, classic antibody produced in PCH is called the Donath-
and the RBCs are sensitized with complement components. Landsteiner antibody and is an autoantibody with anti-P
If the complement cascade does not proceed to C9 (cell death specificity. Other specificities have been reported, including
by lysis), the macrophages of the reticuloendothelial system anti-i32 and anti–Pr-like.33 These antibodies are not identi-
can still clear the sensitized RBCs through their receptors for fiable using regular serologic techniques. The antibody
C3b fragments, thereby causing hemolysis. specificity is only demonstrated in the Donath-Landsteiner
test, which is the test used to confirm the diagnosis of PCH.
Treatment for CHD. Therapy for CHD is generally unnecessary. The Donath-Landsteiner test involves the collection of a
Most patients require no treatment and are instructed to fresh blood sample (collected without anticoagulant) from the
avoid the cold, keep warm, or move to a more temperate patient. Serum is collected rather than an anticoagulated sam-
climate. Patients with moderate anemia are given the same ple so that the complement is active. The sample is maintained
instructions and are urged to tolerate the symptoms rather at 37°C after collection, and the serum is then separated.
than to use drugs on a therapeutic trial basis. There is some Three sets of three test tubes, labeled A1–A2–A3, B1–B2–B3,
advantage to the use of plasma exchange in more severe and C1–C2–C3, containing the patient’s serum are incubated
cases, inasmuch as IgM antibodies have a predominantly at various temperatures with group O RBCs that express
intravascular distribution; however, response to plasma the P antigen (i.e., RBCs of common phenotype). In this test,
exchange is still variable in this patient population, and tubes 1 and 2 of each set contain 10 drops of patient’s serum,
repeated plasma exchanges are often required on a frequent and tubes 2 and 3 of each set contain 10 drops of fresh normal
basis to maintain low plasma levels of autoagglutinating serum as a complement source. One volume of 50% suspen-
antibodies. sion of washed P+ RBCs is added to each tube, and all tubes
Corticosteroids also have been used but generally have a are mixed. After mixing, the three A tubes are then placed
poor response. In some patients whose RBCs are strongly in a melting ice bath for 30 minutes and subsequently for
coated with C3, successful results have been reported with 1 hour at 37°C (biphasic incubation). The three B tubes are
corticosteroids. Some favorable responses also have been re- placed in a melting ice bath for 90 minutes. The three C tubes
ported with the alkylating drug chlorambucil. Splenectomy are kept at 37°C for 90 minutes. After the appropriate time
is generally considered ineffective. has passed, the tubes are centrifuged, and supernatant fluids
Infectious mononucleosis also may be associated with a are examined for hemolysis. Table 21–6 summarizes the re-
hemolytic anemia resulting from a cold autoantibody. Al- actions of a positive Donath-Landsteiner test.
though infrequent, it has been well documented that a high- As the name PCH implies, paroxysmal or intermittent
titered IgM anti-i with a wide thermal range plays a major episodes of hemoglobinuria occur upon exposure to cold.
role in hemolytic anemia associated with this viral infection. These acute attacks are characterized by sudden onset of
Acute illness with a sore throat and a high fever, followed fever, shaking chills, malaise, abdominal cramps, and back
by weakness, anemia, and jaundice, are characteristic fea- pain. All the signs of intravascular hemolysis are evident,
tures of infectious mononucleosis. Lymphadenopathy and along with hemoglobinemia, hemoglobinuria, and biliru-
hepatosplenomegaly are common findings. A larger percent- binemia, depending on the severity and frequency of the
age of patients with infectious mononucleosis have been attack (Fig. 21–3). This results in a severe and rapidly pro-
reported to develop anti-i, but only a small number of these gressive anemia with hemoglobin levels frequently as low
6888_Ch21_441-475 29/10/18 5:38 PM Page 452

452 PART III Transfusion Practices

Table 21–6 Donath-Landsteiner Test


Tubes 1 Tubes 2 Tubes 3
Patient Serum Patient Serum + Normal Serum† Normal Serum†
(Tubes A1, B1, C1) (Tubes A2, B2, C2) (Tubes A3, B3, C3)
Ice bath followed by 37°C + + 0
(all A tubes)

Ice bath only (all B tubes) 0 0 0

37°C only (all C tubes) 0 0 0

+ = with hemolysis
0 = without hemolysis
†Tubes with normal serum are used as control.

Note: The patient’s blood should be clotted at 37°C and the serum separated at this temperature to avoid the loss of the autoantibody by cold autoadsorption prior to testing. Fresh
normal serum should be included in the reaction medium as a source of complement as PCH patients may have low levels of serum complement. Omit the patient serum-only
tubes (A1, B1, C1) if a limited amount of blood is available (e.g., a young child).

Intravascular Hemolytic Event names and acronyms. Autoantibody has not been implicated
in PNH; a membrane defect is thought to be involved. RBC
Serum Haptoglobin
destruction in PNH is complement-mediated because of the
absence or reduced amounts of some complement regulatory
Hemoglobinuria
proteins.34

Warm Autoantibodies
Level

Hemosiderinuria
Autoantibodies that react best at 37°C are not found as often
in the random population as the almost universal cold
autoanti-I. However, many more of the true AIHAs are of the
warm type (70%) than of the cold-reactive type (18%).7 As
with the cold-reactive autoantibodies, some individuals have
Time (d) apparently harmless warm autoantibodies; however, unlike
Figure 21–3. Indicator of acute intravascular hemolysis. Within a few hours of their cold counterparts, the harmless autoantibodies are
an acute hemolytic event, free hemoglobin is cleared from plasma, and the serum serologically indistinguishable from the harmful ones. There
haptoglobin falls to undetectable levels. Hemoglobinuria ceases soon after this. If are no diagnostic tests to predetermine which autoantibodies
no further hemolysis occurs, the serum haptoglobin level recovers and methemal- will cause RBC destruction. When a warm autoantibody is
bumin disappears within several days. The urinary hemosiderin can provide more
lasting evidence of the hemolytic event. (From Hillman, RS, and Finch, CA: Red Cell
encountered, it should be characterized as such and reported
Manual, 7th ed. FA Davis, Philadelphia, 1996, p 112, with permission.) to the patient’s physician. The presence of the antibody may
alert the physician to an underlying autoimmune process.

as 4 or 5 g/dL. Polychromasia, nucleated RBCs, and poikilo- Clinical Findings


cytosis are demonstrated in the peripheral smear, findings
that are consistent with hemolytic anemia. These signs and Patients with warm autoimmune hemolytic anemia (WAIHA)
symptoms, as well as hemoglobinuria, may resolve in a few present a different problem to the blood bank than do those
hours or persist for days. Splenomegaly, hyperbilirubinemia, with cold AIHA. A significant percentage of cases suffer from
and renal insufficiency may also develop. an anemia of sufficient severity to require transfusion. The
PCH is an acute hemolytic anemia occurring almost ex- extent of anemia is variable; however, hemoglobins less than
clusively in children and young adults and almost always 7 g/dL are not uncommon. The onset of WAIHA may be
represents a transient disorder. Table 21–7 compares and insidious and may be precipitated by a variety of factors, such
contrasts PCH with CHD. as infection,35 trauma, surgery, pregnancy,36 or underlying dis-
ease. In other patients, the onset is sudden and unexplained.
Treatment for PCH. For chronic forms of PCH, protection from WAIHA may be idiopathic with no underlying disease process
cold exposure is the only useful therapy. Acute postinfection or may be secondary to a pathological disorder. Box 21–2 lists
forms of PCH are transient and usually terminate sponta- the disorders most commonly associated with WAIHA.
neously after the infection resolves. Steroids and transfusion Signs and symptoms appear when a significant anemia
may be required, depending on the severity of the attacks. has developed. Pallor, weakness, dizziness, dyspnea, jaun-
Note: Paroxysmal nocturnal hemoglobinuria (PNH) is dice, and unexplained fever are occasionally presenting com-
often confused with PCH because of the similarity of the plaints. Hemolysis is usually acute at onset and may stabilize
6888_Ch21_441-475 29/10/18 5:38 PM Page 453

Chapter 21 Autoimmune Hemolytic Anemias 453

Table 21–7 Comparison of PCH and CHD


Factors PCH CHD
Patient population Children and young adults Elderly or middle aged adults

Pathogenesis Following viral infection Idiopathic, lymphoproliferative disorder;


following M. pneumoniae infection

Clinical features Hemoglobinuria, acute attacks upon exposure to Acrocyanosis; autoagglutination of blood
cold (symptoms resolve in hours to days) at room temperature

Severity of hemolysis Acute and rapid Chronic and rarely severe

Site of hemolysis Intravascular Extravascular/intravascular

Autoantibody class IgG (anti-P specificity; biphasic hemolysin) IgM (anti-I/i) monophasic

DAT 3–4+ monospecific C3 only 3–4+ monospecific C3 only

Thermal range Moderate (<20°C) High (up to 30°C–31°C)

Titer Moderate (<64) High (>1000)

Donath-Landsteiner test Positive Negative

Treatment Supportive (disorder terminates when underlying Avoid cold


illness resolves)

Harmening DM: Clinical Hematology and Fundamentals of Hemostasis, ed 5. FA Davis, Philadelphia, 2009, p 263, with permission.

BOX 21–2

Diseases Frequently Associated With WAIHA


Hemolytic Anemia
• Reticuloendothelial neoplasms, such as chronic lymphocytic
leukemia, Hodgkin’s disease, non-Hodgkin’s lymphomas,
myelofibrosis, and myelodysplastic syndromes
• Collagen disease, such as systemic lupus erythematosus and
rheumatoid arthritis
• Infectious diseases, such as viral syndromes in childhood and
adults
• Immunologic diseases, such as hypogammaglobulinemia, dys-
globulinemia, and other immune-deficiency syndromes
• Gastrointestinal diseases, such as ulcerative colitis Figure 21–4. Autoimmune hemolytic anemia (peripheral blood).
• Carcinoma (nonovarian)
• Pregnancy
• Chronic renal failure
precursors can also react with the patient’s RBC autoantibod-
Adapted from Sokol, RJ, Booker, DJ, and Stamps, R: The pathology of autoimmune ies, reticulocytes may be destroyed as they are released from
haemolytic anaemia. J Clin Path 45:1047–1052, 1992, with permission,
the bone marrow. Therefore, reticulocytopenia at a time of
intense hemolysis is associated with a high mortality rate.
Products of hemolysis, such as bilirubin (particularly the
or may continue to accelerate at a variable rate. The periph- unconjugated indirect fraction) and urinary urobilinogen,
eral blood smear usually exhibits polychromasia and macro- are increased. In severe cases, depleted serum haptoglobin,
cytosis, reflecting reticulocytosis, or even the presence of hemoglobinemia, hemoglobinuria, and increases in LDH
nucleated RBCs, which is evidence of a hyperactive bone may be laboratory markers that help confirm the diagnosis.
marrow (Fig. 21–4). Spherocytosis and occasionally RBC
fragmentation, indicating extravascular hemolysis, may also RBC Hemolysis
be demonstrated in a blood smear from a patient with im-
mune hemolytic anemia. An uncommon manifestation of Most patients with WAIHA have both IgG and complement
WAIHA is reticulocytopenia. This may be associated with a on their RBCs (67%). A minority have either IgG only (21%)
hypoplastic marrow that is secondary to an underlying dis- or complement only (13%). Fewer still are patients with
ease state. Because antigenic determinants on erythrocyte WAIHA with IgA (2% to 3%) or IgM (less than 2%) coating
6888_Ch21_441-475 29/10/18 5:38 PM Page 454

454 PART III Transfusion Practices

their RBCs.7 The IgG subclass of coating antibody has been Serologic Characteristics and Laboratory Tests
studied in hopes of finding a correlation with severity of Affected
hemolysis, with IgG1 predominating (87%). Unexpectedly,
approximately the same percentages of patients with a posi- Because warm-reactive autoantibodies are typically IgG im-
tive DAT but no evidence of hemolysis and normal blood munoglobulins, they react best by the indirect antiglobulin
donors with a positive DAT had IgG1 on their RBCs. Nance technique. As a rule, they do not directly agglutinate saline-
and Garratty reported that, in general, the strength of the suspended RBCs after 37°C incubation. The antibodies may
DAT correlated with the presence of multiple IgG subclasses activate complement and are usually enhanced by enzyme
on the RBCs, which in turn correlated with the severity of techniques. Most of these autoantibodies react with a high-
the hemolysis.37 incidence RBC antigen, often with a general specificity
In general, IgG3 antibodies are the most destructive to within the Rh blood group system, but there are reports of
RBCs, followed by IgG1. IgG2 antibodies are less destruc- autoantibodies associated with most of the other blood
tive, and IgG4 shows little or no RBC destruction. The group systems. Identification of autoantibodies is discussed
subclasses, or isotypes, of IgG are distinguished by the later in this chapter.
number of disulfide bonds present in the hinge region of Warm autoantibodies can interfere with most routine
the molecule, accounting for their different electrophoretic blood bank tests, and they may present more of a serologic
mobility and biological properties. Refer to Chapter 3 for a dilemma than cold autoagglutinins. While most cold autoan-
complete discussion of the properties of the IgG subclasses. tibodies can be avoided if room temperature incubation is
All IgG subclasses, except IgG4, possess the ability to bind omitted, if testing is performed at 37°C, or if anti-IgG
complement via the classic pathway of activation, with antiglobulin is used, with WAIHAs, both clinically signifi-
IgG3 being more efficient than IgG1, which in turn is more cant alloantibodies and the autoantibodies themselves react
efficient than IgG2. best at the indirect antiglobulin phase; therefore, more com-
Immune RBC destruction resulting from sensitization plicated and time-consuming procedures for resolving the
with IgG antibody is primarily extravascular, taking place in problems may have to be used.
the fixed reticuloendothelial system (RES) cells of the liver
ABO Typing
and spleen. The spleen is 100 times more efficient than the
liver in removing IgG-sensitized RBCs. Macrophages are Since most warm autoantibodies are not direct agglutinins,
equipped with two important biological receptors on their ABO grouping is usually not affected. Even though the
membranes: patient’s cells may be heavily coated with antibody, the
antibodies typically do not cause spontaneous agglutina-
1. Receptors for the Fc fragments of IgG1 and IgG3 tion of RBCs at room temperature when reagent anti-A and
immunoglobulins anti-B are added. Similarly, warm autoantibodies in the
2. Receptors for the C3b fragment of complement serum usually do not directly agglutinate saline-suspended
Sensitized RBCs are phagocytized by interaction with A1 and B cells.
RES mononuclear phagocytes, depending on which protein
coats the erythrocytes. If only IgG coats the RBCs, gradual RhD Typing
phagocytosis of the erythrocytes occurs. If both IgG and False-positive Rh typing can occur when the patient’s cells
C3b coat the RBCs, then there is a rapid phagocytosis are coated with immunoglobulins. The high-protein Rh
because the C3b fragment augments the action of IgG, antisera, previously the mainstay of Rh typing, demonstrated
enhancing sequestration and phagocytosis of the coated numerous testing problems. Potentiators were added to
erythrocytes. If only C3b coats the RBCs, transient immune many high-protein Rh typing reagents that caused direct
adherence occurs. It has been estimated that more than agglutination of RBCs strongly coated with IgG. For this
100,000 molecules of the complement fragment would be reason, a negative control consisting of patient cells and the
required to induce phagocytosis; therefore, the activity of matching Rh diluent had to be tested in parallel with the
the macrophages and the severity of hemolysis via phago- D typing. The results of the D antigen typing were valid only
cytosis of sensitized RBCs depend on various factors, sum- when the control test result was negative.
marized in Box 21–3. The current reagents available for D typing are predom-
inantly monoclonal antisera containing no more than
7% protein additive. Monoclonal antisera have a low inci-
BOX 21–3 dence of false-positive test results, but false-positive results
still may occur if the RBCs are heavily coated with IgG.38
Factors Affecting Activity of Macrophages Some manufacturers offer an Rh control reagent designed
• Subclass of IgG, especially IgG1 and IgG3 to be tested in parallel with the anti-D serum. Other manu-
• Presence of complement (C3b) fragments facturers recommend that a negative test with an antiserum
• Quantity of immunoglobulin or complement of a similar protein concentration (e.g., ABO antisera) is suf-
• Number and activity of helper T cells (CD4) ficient to detect a false-positive reaction. If a patient blood
• Number and activity of suppressor T cells (CD8) type is group AB Rh-positive (i.e., all tubes in the ABO cell
typing and the D typing are positive), a separate Rh control
6888_Ch21_441-475 29/10/18 5:38 PM Page 455

Chapter 21 Autoimmune Hemolytic Anemias 455

(commercial matched diluent or 6% albumin) must be It must be confirmed that the antibody coating the patient’s
tested alongside to ensure that the ABO/Rh typing is valid. cells is an autoantibody, and limited effort should be ex-
If the diluent or 6% albumin control is positive, it may be pended determining its specificity. The main goal of addi-
necessary to pretreat the cells using EDTA/glycine acid tional testing should be to detect and identify all clinically
(EGA) or chloroquine diphosphate (CDP) to remove coat- significant alloantibodies that might be masked by the
ing IgG immunoglobulins from the RBCs to obtain a valid autoantibody. If the patient has had a previous transfusion
ABO/Rh test.39 or has been pregnant, there is an inherent risk of previous
alloimmunization.
See Procedures 21-5 or 21-10 on the textbook’s
companion website. Evaluation of Autoantibody
If the DAT of the EGA-treated or CDP-treated RBCs is neg- A positive DAT can result from RBC alloantibodies coating
ative, it is then possible to use these cells for weak D testing; recently transfused donor cells or drug-induced antibodies
however, an Rh control serum or 6% albumin should again and RBC autoantibodies. IgG immunoglobulins will most
be tested in parallel to detect false-positive reactivity. Note: likely be present on the cells in each case. It is important to
Even if some IgG remains on the RBCs after EGA or CDP differentiate the causes of a positive DAT because selection
treatment, they are usually suitable for testing with directly of RBCs for transfusion and treatment protocols differ. To
agglutinating monoclonal reagents, including anti-D, anti-C, make the distinction between these causes, one must have
anti-E, anti-c, anti-e, anti-K, anti-Jka, anti-Jkb, and others, as the patient’s medical history, including an accurate history
long as the negative control is valid. of previous transfusions and pregnancies, diagnoses, and
Another technique to detect a weak D type is the rosette medications.
test (see Chapter 20), which is commonly used in the detec- If a patient has had a recent transfusion, the possibility
tion of fetal-maternal hemorrhage. This screening test, used that alloantibodies and not autoantibodies are coating the
to detect fetal D+ cells in the circulation of the D– mother, circulating transfused donor cells must be considered. In
will also detect D+ cells in any cell population. If the patient’s most cases, by examining the DAT microscopically for
cells are D+, the rosette test will be strongly positive. Because mixed-field agglutination (which indicates a mixed-cell
the rosette test does not incorporate an antiglobulin phase, population of DAT+ and DAT– RBCs) and by determining
a patient with a positive DAT can be accurately typed for the the specificity of the antibody in the eluate, it is possible to
D antigen by this method. It is not absolutely necessary to establish alloantibodies as the cause (see Chapter 17). Be-
determine the correct weak D typing of a patient with cause warm autoantibodies are frequently associated with
WAIHA, because D– (Rh-negative) RBCs can be transfused certain diseases, such as systemic lupus erythematosus, and
if necessary. The rosette test to determine the presence of with medications, such as Aldomet, the patient’s diagnosis
D antigen should only be performed on a patient if there was and drug history are informative tools in helping establish
no recent transfusion or pregnancy. autoantibodies as the cause of the positive DAT. The speci-
ficity of the antibody compared to the patient’s RBC pheno-
DAT type is also helpful in differentiating between autoantibody
A positive DAT is expected in association with warm and alloantibody.
autoantibodies. As autoantibody is produced, it adsorbs To identify the specificity of a warm-reactive autoanti-
onto the antigen of that defined specificity present on the body, an eluate prepared from the patient’s RBCs must be
patient’s own RBCs. The RBCs may then be coated with IgG tested, in addition to the patient’s serum, with a panel of
alone (20%), IgG and complement (67%), or complement reagent RBCs.
alone (13%).7 In rare cases, the DAT may be negative or cells
See Procedure 21–6 on the textbook’s companion
may be coated only with IgA or IgM.18–20
website for instructions in preparing a digitonin
Antibody Detection and Identification acid eluate. Note: There are commercial kits also
available for the preparation of acid eluates.
The serum of a patient with warm autoantibodies may con-
tain only autoantibody or, if the patient has been previously If the patient has a well-documented history showing
transfused or pregnant, a mixture of autoantibody and that he or she has not been transfused within the past
alloantibody. When smaller amounts of autoantibody have 3 months, and there is no evidence of a mixed-cell popula-
been produced, the autoantibody may be detected only on tion in any phenotyping results, it is reasonably safe to
the patient’s cells (i.e., positive DAT), adsorbed in vivo, with assume that antibody activity in the eluate is autoantibody.
no free autoantibody detectable in the serum. If the amount But if the patient has a history of recent transfusion or preg-
of antibody produced exceeds the number of RBC antigen nancy, the serum may contain alloantibody in addition to
sites available, when the antigen-antibody equilibrium is autoantibody.
reached, serum antibody will be detected in the indirect The specificity of an autoantibody may be different in the
antiglobulin phase of testing. When warm autoantibodies are serum and in the eluate. Warm autoantibodies in the serum
present in the serum or on the patient’s cells, the extent of may show a relative anti-e specificity, reacting strongest with
further testing could be based on the patient’s history, but re- e positive RBCs and weakest with e negative RBCs while the
liable transfusion histories are notoriously difficult to obtain. eluate may show panagglutination of all RBCs tested. This
6888_Ch21_441-475 29/10/18 5:38 PM Page 456

456 PART III Transfusion Practices

apparent difference in antibody specificity may be qualitative frequently, reactivity is observed with all RBCs of normal Rh
or quantitative. The concentration of antibody removed phenotype. In order to identify the specificity of a complex
from the cells in the elution process may be greater than the Rh-like autoantibody, one must have an extensive library of
quantity of antibody in the serum. Table 21–8 illustrates an rare cells, which includes Rhnull and D– cells. Testing of these
example of such reactivity. It should also be noted that cells should allow one to categorize the antibody as anti-nl
elution procedures vary in their ability to remove coating (normal), which reacts with all cells of common/normal Rh
immunoglobulins (i.e., a heat or freeze/thaw eluate generally phenotypes but not those that are partially deleted or Rhnull;
reacts less strongly than an acid eluate or one of the chemical anti-pdl (partially deleted), which reacts with all cells except
eluates, such as dichloromethane or ether). Rhnull; or anti-dl (deleted), which reacts with all cells.40
A majority of the IgG antibodies detected in eluates pre- Fortunately, this level of antibody identification is of academic
pared from the WAIHA patient’s RBCs or in the patient’s serum interest, since few laboratories have access to these rare
have a complex Rh-like specificity, similar to those shown in RBCs for testing purposes, let alone have them available for
Table 21–9. Occasionally, an autoantibody may have what is transfusion.
termed “simple” specificity, for example, anti-e, which reacts There are numerous reports of autoantibodies with speci-
with all red cells except R2R2 (D+C–E+c+e–) cells. Much more ficities other than Rh, many of which appear to be directed

Table 21–8 Warm Autoantibody With Relative Anti-e Specificity Noted in Serum Testing, but
Broader Pattern of Reactivity Noted in Eluate
Serum Eluate
Cell Donor I.D. D C E c e RT LISS 37ºC IAT Eluate IAT Last wash IAT
1 R1R1 - 44 + + 0 0 + 0 0 3+ 4+ 03

2 R1R1 - 39 + + 0 0 + 0 0 3+ 4+ 03

3 R2R2 - 23 + 0 + + 0 0 0 ± 4+ 03

4 rr - 33 0 0 0 + + 0 0 3+ 4+ 03

5 rr - 26 0 0 0 + + 0 0 3+ 4+ 03

6 r⬘r - 4 0 + 0 + + 0 0 3+ 4+ 03

7 r⬙r - 17 0 0 + + + 0 0 3+ 4+ 03

8 R0r – 13 + 0 0 + + 0 0 3+ 4+ 03

9 R1r – 14 + + 0 + + 0 0 3+ 4+ 03

10 R1R2 - 8 + + + + + 0 0 2+ 4+ 03

11 Auto Control 0 0 3+ 4+ 3+
(untt’d BCs)

12 Auto Control 0 0 3+ 4+ 03
(EGA-tt’d )

03 = negative IAT reading with check cells added and reactive, as expected; Untt’d=untreated; EGA-tt’d=EDTA glycine acid treated

Table 21–9 Typical Serologic Reactions of Warm Autoantibodies With RBCs of Selected
Rh Phenotypes
Autoanti-e Anti-nl Anti-pdl Anti-dl
R1R1 (normal) + + + +

R2R2 (normal) 0 + + +

rr (normal) + + + +

D– – (partially deleted) 0 0 + +

Rhnull (fully deleted) 0 0 0 +

nl = normal; pdl = partially deleted; dl = fully deleted


6888_Ch21_441-475 29/10/18 5:38 PM Page 457

Chapter 21 Autoimmune Hemolytic Anemias 457

against RBC antigens of high incidence or a null phenotype. the cells to remove coating autoantibody, using either of
Among the other specificities are autoanti-U, autoanti-Wrb, the methods described above or a chemical such as ficin
autoanti-Ena, autoanti-Kpb, autoanti-Vel, and autoanti- or ZZAP (a combination of ficin and DTT).
Ge.41–43 The reader is referred to Petz and Garratty7 for a
See Procedure 21–7B on the textbook’s companion
detailed discussion of autoantibody specificity. Apparent
website. This treatment will free antigen sites for
specificities such as these can cause confusion, especially
adsorption of autoantibody.
when the patient has had a recent transfusion. Determining
the patient’s phenotype is one of the most valuable tools 3. If autoadsorption studies are not possible because the
used to classify the antibody as “auto” or “allo.” As previ- patient was recently transfused and there is evidence of
ously discussed, monoclonal antisera and commercially reticulocyte production, then it may be possible to deter-
available IgG removal agents have made this challenge eas- mine the patient’s phenotype using a reticulocyte harvest-
ier. In recently transfused patients, molecular genotyping of ing procedure. This phenotyping information can be used
the patient’s white blood cells may be helpful in predicting to select RBCs that are phenotypically similar to those of
the patient’s phenotype. Most researchers agree that it is not the patient by matching the common RBC antigens: Rh,
necessary to do extensive studies to identify the specificity Kell, Kidd, Duffy, S, and s. Allogeneic adsorptions per-
of the autoantibody, but testing of at least one example of formed using this method to select phenotypically similar
RBCs that is phenotypically similar to those of the patient donor or reagent RBCs will remove autoantibody but
is important to help distinguish between autoantibody will also adsorb an alloantibody to any high-incidence
(the phenotypically similar RBCs will be reactive) and mul- RBC antigen, if present. This technique of selecting a
tiple alloantibodies (the phenotypically similar RBCs will phenotype-similar source of RBCs for adsorption may
be nonreactive). also inadvertently adsorb alloantibody if the patient’s
Specificity of the autoantibody, if obvious, may influence RBCs express a variant antigen (e.g., partial D, variant C,
selection of blood for transfusion. Some practitioners prefer variant e, etc.)
to transfuse RBCs that are compatible with the autoantibody
See Procedure 21-11 on the textbook’s companion
if simple specificity can be assigned, such as anti-e, and if there
website.
are no preexisting circumstances that would prevent this
transfusion (e.g., one would not select Rh-positive e– RBCs 4. If it is not possible to determine the patient’s RBC phe-
for an Rh-negative patient with autoanti-e). Selecting donor notyping, differential allogeneic adsorptions may be per-
units that are compatible with the patient’s autoantibody, formed by selecting three donor units that are known to
however, cannot guarantee normal RBC survival. Unfortu- lack common RBC antigens and are of complimentary Rh
nately, the transfused cells will probably be destroyed as rap- antigen combinations. Typically, these cells include each
idly as the patient’s own cells, regardless of phenotype. of the following Rh types: R1R1 (D+C+E–c–e+), R2R2
(D+C–E+c+e–), and rr (D–C–E–c+e+). Allogeneic ad-
Detection and Identification of Alloantibodies sorptions performed using this method will remove au-
All researchers agree that detection and identification of all toantibody but will also adsorb an alloantibody to any
alloantibodies is of primary concern when one must trans- high-incidence RBC antigen, if present. Ficin or ZZAP
fuse a patient with WAIHA, especially when the patient has treatment of the allogeneic adsorbing cells is recom-
had a previous transfusion or pregnancy. When autoantibody mended because it increases antibody uptake but also al-
is found in the serum, it will typically mask any alloantibod- ters the phenotype of the adsorbing cells—for example,
ies present. In this situation, one can use several techniques: ficin will render the RBCs M–N–S– Fy(a–b–) and ZZAP
will render them K–k–s– as well. Keep in mind the cell
1. If the autoantibody demonstrates a simple specificity,
treatment used when interpreting the pattern of results
such as anti-e, test a panel of cells negative for the corre-
in the adsorbed sera.
sponding antigen (e– in this case) and positive for all
common clinically significant RBC antigens (Rh, Kell, In typical warm adsorption procedures, the patient’s
Duffy, Kidd, S, and s) to exclude or identify the presence serum and the prepared adsorbing cells are incubated
of underlying alloantibodies. at 37°C, allowing the autoantibody to bind to antigen sites
2. If the patient has not had a recent transfusion (within the on the prepared cells, leaving unadsorbed alloantibody in the
past 3 months), determine the patient’s RBC phenotype serum. The number of adsorptions needed to remove all au-
by using either monoclonal antisera, which does not toantibody depends on the amount of autoantibody present
require antiglobulin testing, or by using the patient’s in the patient’s serum and the test procedure used. Usually,
RBCs treated with an agent known to remove coating IgG the strength of reactivity in the indirect antiglobulin phase
immunoglobulins. The cells may be treated with chloro- of the antibody screen or panel is a good indicator of how
quine diphosphate (CDP) solution or EGA prior to many adsorptions will be necessary. If there is strong reac-
phenotyping. (The EGA procedure will denature all Kell tivity (3+ to 4+) of autoantibody in the serum and gel or
system antigens, among others.) If there is a sufficient polyethylene glycol (PEG) enhancement is used in testing,
quantity of patient RBCs available, prepare autologous then more than one adsorption will likely be necessary. Test-
cells to be used for autoadsorption procedures. Pretreat ing the adsorbed serum with the DAT-negative (e.g., EGA or
6888_Ch21_441-475 29/10/18 5:38 PM Page 458

458 PART III Transfusion Practices

CDP-treated) autologous RBCs or allogeneic adsorbing cells against high-incidence antigens. For example, an anti-k
used is a useful way of determining if autoantibody has been would be adsorbed onto virtually all random donor cells
adsorbed and the adsorbed serum is ready to test with addi- because the k antigen is present on the cells of more than
tional selected cells. 99% of the population (including the R1R1, R2R2, and
rr adsorption cells) unless ZZAP treatment of the adsorb-
See Procedure 21–10 on the textbook’s companion
ing cells is used. An antibody such as this would be differ-
website.
entiated from the autoantibody only if a cell negative for
Table 21–10 shows an example of a patient with a warm the high-incidence antigen happened to be present among
autoantibody demonstrable by LISS indirect antiglobulin test the cells used for adsorption. Nevertheless, allogeneic (dif-
(IAT) and alloanti-E detected in the autoadsorbed serum. ferential) adsorption technique is valuable when there is
When performing adsorption procedures, the following no alternative.
circumstances must be considered: Table 21–12 shows an example of a warm autoantibody
with underlying anti-K and anti-Jka. The allogeneic adsorp-
• Autoadsorption procedures are never recommended in
tions were performed using phenotyped donor RBCs that are
patients who have been transfused within the previous
of phenotypes complementary to each other as a “set.” Note
3 months. In vitro studies have determined that as few as
that the phenotype of the donor RBCs is unknown for P1,
10% of antigen-positive RBCs are sufficient to adsorb the
M, N, Lea, and Leb. Because antibodies to these antigens
alloantibody.44 This small amount of antigen-positive cells
would not be expected to be clinically significant unless they
may not be obvious in phenotyping.
were reactive at 37°C, they can be ignored. If interpretation
• If the patient is severely anemic, it may not be possible to
of the reactivity pattern in the adsorbed sera suggested one
obtain sufficient autologous cells for multiple adsorptions.
of these antibodies was present, the adsorbing cells could be
• Whenever an adsorption is performed, whether with
phenotyped at that point for the antigen of interest.
autologous or allogeneic cells, the serum is diluted to
When alloantibodies are detected in the serum of a patient
some extent. Some saline remains in “packed” RBCs. A
with WAIHA, it is desirable to test the patient’s untransfused
weakly reactive alloantibody could be diluted and missed
RBCs for the absence of the corresponding antigen, if not
if multiple adsorptions are performed.
already done. If no pretransfusion sample is available, un-
• If allogeneic adsorptions are performed, it is possible to
derlying antibodies must be assumed to be alloantibody in
adsorb an alloantibody to a high-incidence RBC antigen
nature.
that is present on the allogeneic cells that is not present
on the patient’s own cells.
Selection of Blood for Transfusion
When the patient has had a recent transfusion or is se-
verely anemic, cells of selected phenotypes for adsorption Many patients with WAIHA never require transfusion; they
can be used. If the patient’s phenotype is unknown and can- can be managed with medical treatment. Occasionally, how-
not be determined, adsorptions can be performed using a ever, the anemia is so severe that transfusion cannot be
trio of selected cells (R1R1, R2R2, and rr). These cells should avoided. In addition, patients who have a nonhemolytic
also lack one or more antigens for the more commonly WAIHA pose problems when blood is needed for a surgical
encountered clinically significant alloantibodies (i.e., anti-S, procedure. In these cases, after thorough serologic investi-
anti-s, anti-K, anti-Fya, anti-Fyb, anti-Jka, anti-Jkb). As shown gation, the primary concern is to ensure compatibility with
in Table 21–11, if the patient’s serum is adsorbed with cells any alloantibodies in the patient’s serum. Compatibility with
from donors of selected phenotypes and then the adsorbed the autoantibody is controversial. If the autoantibody shows
sera are tested, alloantibodies to the common antigens can a simple specificity, such as anti-e, local practice may be to
be detected. select donor units that are negative for the corresponding
Alternatively, if a pretransfusion phenotype is available antigen, when practical.
for the patient, phenotypically matched RBCs can be used As previously mentioned, this may be difficult if the
for adsorption. (See discussion of antigen typing the patient donor RBCs selected precipitated alloantibody formation
with a positive DAT in this chapter.) With the exception of that would bring bigger challenges. For example, because
possible adsorption of an antibody to a high-incidence anti- virtually all Rh-negative (D–) cells are e+, an Rh-negative
gen, adsorption with donor RBCs of a phenotype similar to (D–) patient with autoanti-e specificity should not receive
the patient’s is a useful alternative to autologous adsorption. a transfusion of Rh-positive (D+) RBCs that lack the e anti-
The patient should not form alloantibodies to RBC antigens gen.45 Yun and colleagues46 report hemoglobin increments
that he or she possesses; therefore, it is advised to focus in patients with AIHA comparable to the expected incre-
mainly on the antigens the patient lacks. For example, if the ment even when e+ units were transfused to patients with
patient’s RBCs phenotype is E–c–S–K–Fy(a–)Jk(b–) and an apparent autoanti-e. Finding compatible units for
donor RBCs are available that are E–c–K–Jk(b–), ficin treat- patients with a broad specificity warm autoagglutinin is
ment of the donor RBCs would render them also S– and virtually impossible. Even if the units are nonreactive with
Fy(a–), a phenotype similar to the patient’s. the adsorbed serum, these units will be “least incompati-
It is impossible to detect all clinically significant alloan- ble” when crossmatched with unadsorbed serum. Also, the
tibodies using allogeneic adsorptions, like those directed selection of in vitro least incompatible RBCs may not
Table 21–10 Warm Autoantibody With Underlying Alloanti-E
6888_Ch21_441-475 29/10/18 5:38 PM Page 459

Ficin Auto-ads Serum

Cell D C E c e P1 M N S s Lea Leb K k Fya Fyb JKa Jkb Other Cell RT LISS 37°C IAT IAT
R1R1 - 44 1 + + 0 0 + + + + 0 + 0 + 0 + + 0 + + Cw+ 1 0 0 3+ 03

R1R1 - 39 2 + + 0 0 + + + + 0 + 0 + 0 + + + + 0 Co(b+) 2 0 0 3+ 03

R2R2 - 23 3 + 0 + + 0 + 0 + 0 + 0 + 0 + 0 + + 0 3 0 2+ 4+ 3+

rr - 33 4 0 0 0 + + 0 + 0 + + + 0 0 + 0 + 0 + 4 0 0 3+ 03

rr - 26 5 0 0 0 + + + + + + + 0 + + + + + + 0 5 0 0 3+ 03

r⬘r - 4 6 0 + 0 + + 0 + 0 + + 0 0 + + 0 + 0 + 6 0 0 3+ 03

r⬙r - 17 7 0 0 + + + + + + + 0 0 + 0 + + 0 0 + 7 0 2+ 4+ 3+

R0r – 13 8 + 0 0 + + + + + + 0 0 0 0 + 0 0 + + V+ VS+ 8 0 0 3+ 03

R1r – 14 9 + + 0 + + + 0 + + + + 0 0 + + 0 + + 9 0 0 3+ 03

R1R2 - 8 10 + + + + + 0 + 0 + + 0 + 0 + 0 + + + Kp(a+) 10 0 2+ 4+ 3+

Auto Control 11 11 0 0 3+ 3+
(untt’d RBCs)

Auto Control 12 12 0 0 3+ 03
(EGA-tt’d)

Anti-E is also detectable at 37°C as a directly agglutinating antibody, which is not uncommon with many examples of Rh antibodies. Adsorption ×3 with ficin- ADS = adsorbed;
treated autologous RBCs removed the warm autoantibody and left alloanti-E detectable in the autoadsorbed serum. EGA-treated autologous RBCs react with
the unadsorbed serum but are nonreactive with the autoadsorbed serum. 03 = negative IAT reading with check cells added and reactive as expected.
Chapter 21 Autoimmune Hemolytic Anemias
459
6888_Ch21_441-475 29/10/18 5:38 PM Page 460

460 PART III Transfusion Practices

Table 21–11 Differential Adsorption Technique for Detecting Alloantibodies in the Serum
of a Patient With Warm-Reacting Autoantibodies
Example of Phenotype Antibody Removed Antibody Remaining
RBC of Adsorbing RBCs by Adsorption in Adsorbed Serum
R1R1 (D+C+E–c–e+) K–k+ Fy(a+b–) D,C,e,k,Fya,Jkb, S,s E,c,K,Fyb,Jka
Jk (a–b+) S+s+

R2R2 (D+C–E+c+e–) K–k+ Fy(a–b+) D,E,c,k,Fyb,Jka, s C,e,K,Fya,Jkb,S


Jk (a+b–) S–s+

rr (D–C–E–c+e+) K+k– Fy(a–b+) c,e,K,Fyb, Jka,Jkb,S D,C,E,k,Fya,s


Jk(a+b+) S+s–

translate to in vivo most compatible RBCs. The transfused Intravenous immunoglobulin (IVIG) has also been used
donor cells are likely to be destroyed as rapidly as the in patients who do not respond to prednisone therapy. Its
patient’s own RBCs. It is much more important that donor mechanism for effectiveness is not understood and appears
units be selected that are compatible with any alloantibod- to be most effective when used in conjunction with cortico-
ies identified. steroid therapy.7 The use of intravenous immune globulin in
patients unresponsive to corticosteroid treatment is contro-
Treatment of WAIHA versial and appears to have limited efficacy.49

Therapy is generally aimed at first treating the underlying dis- Splenectomy


ease, if one is present. General measures to support cardio- If steroid therapy fails, or if a patient requires large doses of
vascular function are important for patients who are severely steroids to control hemolysis, splenectomy is usually recom-
anemic. Transfusion should be avoided, if possible, because mended. The decision to perform a splenectomy requires
it may only accelerate the hemolysis; however, transfusion clinical evaluation and judgment. There are three reasons for
should not be avoided if the anemia is life-threatening. The performing a splenectomy:
volume of red blood cells transfused should be conservative,
1. Failure of steroid therapy
aiming for a relief in symptoms, not restoring a normal he-
2. Need for continuous high-dose steroid maintenance
matocrit. In many cases, even small amounts of transfused
3. Complications of steroid therapy
RBCs (even 0.5 to 1 unit of packed RBCs) are sufficient to
relieve the symptoms of the anemia.47 Forms of treatment Splenectomy results in decreased production of antibody
other than transfusion are described in the following section. and removes a potent site of RBC damage and destruction.
Patients who had a good initial response to steroids respond
Corticosteroid Administration and Use better with splenectomy than do those who failed initial
of IV Immunoglobulin steroid therapy. It has been reported that as many as 60% of
One form of therapy involves the use of corticosteroids, such patients with WAIHA benefit from splenectomy if steroid
as prednisone. Initially, high doses (100 to 200 mg) of pred- dosages greater than 15 mg per day are also used to maintain
nisone are maintained until the patient’s hematocrit level sta- remission.
bilizes. Patients who have not had transfusions seem to
respond to steroid therapy more rapidly than those who are Immunosuppressive Drugs
transfused. The use of immunosuppressive drugs is usually the last
Several mechanisms have been proposed for the action of approach for managing WAIHA. The field of organ trans-
prednisone,48 including the following: plantation has seen advances in development of immuno-
suppressive drugs, some of which have been used effectively
• Reduction of antibody synthesis
in the treatment of WAIHA that has failed other forms of
• Altered antibody activity
therapy. Azathioprine (Imuran) and cyclophosphamide are
• Alteration of macrophage receptors for IgG and C3, which
examples of immunosuppressive drugs that interfere with
reduces the clearance of antibody-coated RBCs
antibody synthesis by destroying dividing cells. Detrimental
The dosage of prednisone should be reduced when the side effects of these drugs are infection, infertility, risk
hematocrit begins to rise and the reticulocyte count drops. of birth defects, and development of malignancies. Cy-
The steroids are withdrawn slowly over 2 to 4 months. A closporin has also been used, but the risk of renal damage
beneficial response to the administration of prednisone is is high. One of the most promising drugs is Rituximab,
demonstrated in 50% to 65% of all cases of WAIHA. An a monoclonal antibody that targets antibody-producing
androgenic steroid, danazol, has also been beneficial in B lymphocytes, but it also has adverse effects, especially
prednisone-resistant cases.48 when used long-term.
Table 21–12 Warm Autoantibody With Underlying Alloanti-K and -Jka
6888_Ch21_441-475 29/10/18 5:38 PM Page 461

Unadsorbed Serum
R1R1 Allo-Adsserum
R2R2 Allo-Adsserum
r r Allo- ads Serum

D C E C e P1 M N S s Lea Leb K k Fya Fyb Jka Jkb


{
Cell RT LISS 37°C IAT IAT IAT IAT
R1R1 - 10 + + 0 0 + ? ? ? 0 + ? ? 0 + + 0 + + 03
Adsorbing
R2R2 - 18 + 0 + + 0 ? ? ? + + ? ? 0 + + + + 0 cells chosen 03
by phenotype
rr - 14 0 0 0 + + ? ? ? + 0 ? ? + + 0 + 0 + 03

R1R1 - 16 1 + + 0 0 + + + 0 + 0 + 0 0 + 0 + + 0 Set of 3 1 0 0 3+ 03 03 3+
antibody
R2R2 - 25 2 + 0 + + 0 + 0 + + + 0 + + + + + 0 + detection 2 0 0 3+ 3+ 3+ 03

rr - 9 3 0 0 0 + + 0 + + 0 + 0 + 0 + + 0 + + cells 3 0 0 3+ 03 03 2+

rr - 17 4 0 0 0 + + + + + + 0 0 + 0 + + 0 0 + 4 03 03 03
Selected RBCs
R0r – 13 5 + 0 0 + + + + + 0 + 0 0 + + 0 0 0 + to confirm 5 3+ 3+ 03
and exclude
R1r – 17 6 + + 0 + + + 0 + + + + 0 0 + + 0 + 0 common 6 03 03 3+
allo-abs
R1R2 - 12 7 + + + + + 0 + 0 + + 0 + 0 + 0 + 0 + 7 03 03 03

Auto Control 8 8 0 0 3+ NT NT NT
(untt’d RBCs)

Auto Control 9 9 0 0 3+ 03 03 03
(EGA-tt’d)

Each column in the adsorbed sera columns must be interpreted separately for purposes of antibody identification, keeping in mind the phenotype ? = phenotype of adsorbing cell is unknown;
of the adsorbing cells after ficin treatment; for example, the R1R1, R2R2, and rr adsorbing cells would all become Fy(a–b–). This means that if
anti-Fya or -Fyb were present in the adsorbed sera, they would be present in all three aliquots. 03 = negative IAT reading with check cells added and reactive as expected;
Differential adsorption ×3 with ficin-treated allogeneic RBCs removed the warm autoantibody and left alloanti-K and alloanti-Jka detectable in the NT = cell not tested.
alloadsorbed serum. EGA-treated autologous RBCs react with the unadsorbed serum but are nonreactive with the autoadsorbed serum.
Chapter 21 Autoimmune Hemolytic Anemias
461
6888_Ch21_441-475 29/10/18 5:38 PM Page 462

462 PART III Transfusion Practices

Mixed-Type Autoantibodies plasma exchange, IVIG, steroids, or transfusion, but treat-


ment is not always successful and the disease may be fatal.7
Individuals demonstrating antibody activity that appears to
have both “warm” and “cold” components are considered to DAT-Negative Autoimmune Hemolytic Anemia
have a mixed-type AIHA. Serologic testing will show autoan-
tibody components typical of both warm and cold AIHA. Although the vast majority of patients presenting with signs
The mere presence of a warm and cold autoantibody in a and symptoms of autoimmune hemolytic anemia will have
patient does not define “mixed-type” AIHA. Although the a positive DAT and other serologic evidence to support their
cold autoagglutinin in these individuals will demonstrate diagnoses, there is a minority of affected patients who appear
strongest reactivity at 4°C, it is usually reactive at 30°C or to have a negative DAT when tested with commercial
above. It is the thermal amplitude of the cold autoantibody reagents (polyspecific AHG, anti-IgG, and anti-C3). Some-
that is key to its pathogenicity. The cold agglutinin is an IgM times, the amount of IgG coating the RBCs is lower than the
hemagglutinin capable of binding complement. The warm detectable limit of commercial IgG reagents. However, if the
component is an IgG antibody; therefore, the patient will hematologist seeks a laboratory value to confirm a diagnosis,
typically demonstrate a positive DAT with both IgG and he or she may order a “super Coombs’” test. Few laboratories
complement on the RBCs. This type of AIHA is rare. have the reagents and experience to perform this test, more
Patients with mixed-type AIHA often present with accurately called a “DAT hemolytic anemia workup,” but the
extremely acute hemolysis and frequently require transfusion. laboratories that do this sort of testing have found the fol-
Because there are two separate autoantibodies present, adsorp- lowing tests to be most productive: cold saline/cold LISS
tion procedures must include both cold and warm adsorptions wash prior to DAT testing (to detect low-affinity IgG that is
to completely remove autoagglutinins. In routine adsorption lost if washing is done at RT), direct polybrene DAT, DAT
procedures, the patient’s serum or plasma may first be testing with anti-IgA and anti-IgM (noncommercial reagents
adsorbed at 4°C with one or more aliquots of selected cells standardized for RBC agglutination), and DAT by solid-phase
and then adsorbed at 37°C with additional aliquots of selected or column agglutination method. Researchers have also used
cells, using the procedures previously described; however, the flow cytometry, enzyme-linked antiglobulin assays, mono-
procedures may be performed on the same aliquot of adsorb- cyte monolayer assays (MMAs), and concentrated eluates.
ing cells if the order is reversed. Warm adsorption followed Even with these esoteric methods, immunoglobulins are
by cold adsorption in the same tube saves time and adsorbing detected on the patient’s RBCs only about half of the time.7
cells. The warm antibody will remain bound at 4°C while the Table 21–13 shows a summary contrasting the main charac-
cold antibody is secondarily adsorbed. It has been found that teristics of warm and cold autoantibodies.
corticosteroid treatment is most often effective in treating
individuals with mixed-type AIHA.50 Drug-Induced Immune Hemolytic
Anemia
IgM Warm Autoantibodies
Therapeutic drugs may have unintended consequences, in-
An unusual type of WAIHA is one associated with IgM warm cluding immune destruction of RBCs, white blood cells
autoantibodies. The serologic characteristics are often unim- (WBCs), and platelets, although the incidence is rare. He-
pressive when compared to the clinical havoc these antibod- molytic anemia, leukopenia, and thrombocytopenia can
ies wreak. Usually the patient’s RBCs are strongly coated with occur separately, but in some patients more than one cell line
complement and frequently cause difficulty in obtaining is affected. The cells may be coated with antibody, antibody
valid ABO/Rh typing and DAT because the cells sponta- and complement, or complement alone. The discussion in
neously agglutinate. This spontaneous agglutination is not this section is limited to RBC problems, but many of the
prevented by warm washing because the antibody is a warm same principles also apply to platelets and leukocytes.
agglutinin. Treatment of the RBCs with 0.01 M DTT is nec- Drug-mediated problems may come to the attention of
essary to remove enough IgM from the RBCs to allow for the blood bank technologist in one of two ways:
valid ABO/Rh and DAT results. IgM can sometimes be de-
1. A request for diagnostic testing on a patient with sus-
tected on the RBCs if suitable anti-IgM is available, but flow
pected hemolytic anemia
cytometry is the most reliable technique in detecting the
2. Unexpected results in routine testing: a positive DAT or
IgM. In antibody detection tests, there is usually no reactivity
positive autocontrol by IAT, a nonreactive eluate, a his-
of the serum at room temperature, a weak agglutinin at 37°C,
tory of recent antibiotic therapy or diagnosis which sug-
and very weak or no reactivity at the antiglobulin phase. If
gests it
enzyme-treated RBCs are tested, many examples react
strongly or are lysed by these antibodies. If antibody speci- Drugs should be suspected as a possible explanation
ficity is done, these antibodies may show a specificity for for immune hemolysis or a positive DAT when there is no
antigens on glycophorins (e.g., anti-Ena, anti-Pr, anti-Ge, other reason for the serologic and hematologic findings and
anti-Wrb).50 If the IgM warm autoantibodies detected are as- if the patient has a recent history of taking high doses of
sociated with a severe clinical anemia (Hgb less than 5 g/dL), antibiotics or other drugs associated with drug-induced
the prognosis is poor. Some patients have been treated with immune hemolytic anemia (DIIHA). Drug-induced positive
6888_Ch21_441-475 29/10/18 5:38 PM Page 463

Chapter 21 Autoimmune Hemolytic Anemias 463

Table 21–13 Comparison of Warm and Cold AIHA


Warm AIHA Cold AIHA
Optimal reaction temperature >32°C <30°C

Immunoglobulin classification IgG IgM

Complement activation May bind complement Binds complement

Site of hemolysis Usually extravascular (no cell lysis) Extravascular/intravascular (cell lysis)

Frequency 70%–75% of cases 16% of cases (PCH 1%–2%)

Specificity Frequently broad Rh specificity Ii system (PCH autoanti-P)

Adapted from Harmening, DM: Clinical Hematology and Fundamentals of Hemostasis, ed 4. FA Davis, Philadelphia, 2002, p 213, with permission.

DATs and hemolytic anemia are relatively rare (estimated Antibody


at 1 in 1 million), so other potential causes should be con- to drug
sidered and investigated first.51
Drug-induced hemolytic anemias were first reported in the Antibody to Antibody
(mainly) membrane Drug to drug and
1950s, but most of these reports were limited to association of components membrane
the onset of hemolytic anemia with the onset of drug therapy components
and resolution of the anemia when the drug was stopped.52,53
Since these events could just be coincidental, proving drug in-
ducement would have required restarting the drug therapy to
prove that hemolytic anemia would again ensue. This practice Red cell membrane
could certainly endanger the patient so was not usually done, Figure 21–5. Unifying hypothesis. The drug binds loosely or firmly to the RBC
nor is it recommended. However, this approach was used to membrane and antibody reacts with the drug itself (drug adsorption), mostly with
associate methyldopa (Aldomet) with DIIHA in the 1960s. Few the RBC membrane altered by the drug (mimics typical warm autoantibody) or with
early studies demonstrated the presence of antibody serologi- a combination of drug and RBC membrane (so-called immune complex). (From
Garratty, G: Target antigens for red-cell-bound autoantibodies. In Nance, SJ (ed):
cally, which is the approach used today.
Clinical and Basic Science Aspects of Immunohematology, American Association
In their first volume, Petz and Garratty7 reviewed the four of Blood Banks, Arlington, VA, 1991, p 55.)
classic mechanisms then proposed to account for drug-
induced problems: immune complexes, drug adsorption,
membrane modification/nonspecific protein adsorption, order to react; drug-independent antibodies are those that
and autoantibody formation. Specific drugs have often mimic warm autoantibodies serologically but where the for-
been associated with one particular mechanism, but not all mation of autoantibody was stimulated by the drug.
drugs associated with hemolytic anemia neatly fit one
model.54,55 The lines are often blurred. Other theories pos- Drug-Adsorption (Hapten) Mechanism
tulate combining aspects of these earlier proposed mecha-
nisms.40 Current thinking on these mechanisms is termed a Drugs operating through the drug-adsorption mechanism
unifying hypothesis where more than one drug-associated bind firmly to proteins, including the proteins of the RBC
antibody specificity is present. Figure 21–5 depicts the three membrane (Fig. 21–6). Presumably because of their ability to
mechanisms proposed to support the unifying hypothesis: bind to proteins, these drugs are good immunogens. Anti-
bodies to penicillin, the drug most commonly associated
1. Drug binds to the RBC membrane and antibody formed
with this mechanism, are found in about 3% of hospitalized
is directed at the drug (test methodology involves testing
patients receiving large doses of penicillin; of these, fewer than
drug-coated RBCs).
5% develop a hemolytic anemia.9 Even with the relatively high
2. Drug does not bind covalently to RBCs but rather com-
incidence of antipenicillin antibodies and the drug’s ability to
plexes with drug antibody (test methodology involves
bind to the RBC membrane, penicillin-induced positive DATs
testing in the presence of the drug).
are rare.12 The low incidence may reflect the fact that the pa-
3. Drug induces an autoimmune response, but the antibody
tient must receive massive doses (10 million units per day) of
is directed at the RBC membrane (testing cannot differen-
penicillin for the cells to be coated adequately. Also, most
tiate between drug-induced or idiopathic autoantibody).
penicillin antibodies are IgM and therefore not detected by the
Drug-related positive DATs may be additionally classified antiglobulin test. The penicillin antibody responsible for a
as drug-dependent or drug-independent. Drug-dependent positive DAT is most often IgG; less often IgM, IgA, or IgD;
antibodies are those that require the presence of the drug in and rarely binds complement.40
6888_Ch21_441-475 29/10/18 5:38 PM Page 464

464 PART III Transfusion Practices

ways. Because the complement cascade is usually not acti-


Antibody vated, cell destruction is predominantly extravascular
RBC rather than intravascular; therefore, the anemia develops
more slowly and is not life-threatening unless the cause for
the hemolysis is not recognized and the penicillin therapy
is continued. Penicillin-induced hemolysis occurs only
when the patient receives massive doses of the antibiotic,
in contrast to the small amounts of drug that are necessary
for hemolysis due to immune complexes. The patient im-
proves once the drug is withdrawn, but hemolysis contin-
ues at a decreasing rate until cells heavily coated with
penicillin are removed. The DAT may remain positive for
several weeks. Mixed-field agglutination in the DAT can be
expected because some cells are penicillin-coated while
Drug
others are not.
Figure 21–6. Drug-adsorption mechanism. Antipenicillin antibody may cross-react with ampicillin
and methicillin. Antipenicillin reacts with Keflin-treated cells
and anti-Keflin with penicillin-coated cells. Garratty suggests
Laboratory test results are consistent with this description that comparing the strength of the reactivity of the serum or
of the mechanism. Cells from patients with a positive DAT eluate (titer or score) with penicillin-coated and Keflin-
due to drug adsorption are usually coated with IgG alone; coated cells may be of value.57
however, sometimes both IgG and complement are present Other drugs that cause a positive DAT and hemolytic ane-
on the cells. The patient’s serum and eluate are nonreactive mia by this mechanism are most notably the cephalosporins.
with reagent RBCs and random donor cells. Therefore, the Distinguishing between cephalosporin-induced problems
antibody screen is negative, and crossmatches are compatible and penicillin-induced problems is technically difficult be-
in all phases. In order to demonstrate the antibody, the serum cause the drugs have antigenic determinants in common,
and eluate must be tested with penicillin-coated cells; reac- and low-titered antipenicillin is frequently present in the
tivity occurs in the antiglobulin phase.56 Because many serum of unaffected individuals. Reports of severe hemolytic
patients have low-titered antipenicillin antibodies, Petz and episodes associated with the second- and third-generation
Garratty7 emphasize that when tested with drug-coated cells, cephalosporins appear to be increasing.40,58–60 Several of
the serum antibody must be high titered and the eluate must these cases appear to involve aspects of both the immune
also contain antipenicillin antibodies before the findings are complex and the drug-adsorption mechanisms. Immune-
definitive.7 Interpretation of the confirmatory tests to demon- mediated hemolysis has occurred rapidly after administra-
strate antidrug antibody reactive by the drug-adsorption tion of only a small amount of the drug. Because these
method is outlined in Table 21–14. antibiotics are routinely used in both adult and pediatric
populations, any evidence of intravascular hemolysis should
The procedure for preparing the penicillin-coated
be noted and evaluated.
cells is given in Procedure 21–9 on the companion
Cefotetan-induced hemolysis has been the most fre-
website.
quently encountered DIIHA (greater than 50%) by Gar-
Only a small percentage of those patients with penicillin- ratty61 in the last 40 years. It causes intravascular lysis of
induced positive DATs exhibit hematologic complications. RBCs. Cefotetan is often given prophylactically for surger-
The clinical features of such a hemolytic episode differ from ies such as Caesarian sections and appendectomies and is
those of immune complex–mediated problems in several often given during surgery. Even without additional doses,

Table 21–14 Test Results Observed in the Presence of Antidrug Antibody Reactive
by the Drug-Adsorption Mechanism
Patient Serum Drug-Coated Uncoated
Test or Eluate RBCs RBCs Results
Patient serum control X — X If negative, indicates no alloantibody to RBC present.
Drug/RBC control — X — If negative, indicates drug-coated cells did not sponta-
neously agglutinate.
Patient test X X — If agglutination is present and controls are all nega-
tive, indicates the presence of antidrug antibody. If
negative, no antidrug present.

X = sample added to tube; — = no sample added to tube


6888_Ch21_441-475 29/10/18 5:38 PM Page 465

Chapter 21 Autoimmune Hemolytic Anemias 465

the patient may present with profound anemia 7 to 10 days a drug–antidrug explanation should be considered. When
after the procedure. This previously healthy patient may obtaining the medication history, it is important to realize
present with a very low hemoglobin (e.g., 4 g/dL). DIIHA that this group of patients needs to take only small doses of
is not suspected because the patient is unaware of receiving the drug to be affected. The patient recovers rapidly once the
the drug. Finding documentation of the drug may involve drug is withdrawn.
searching through the surgical notes. If a second dose is If polyspecific antihuman serum is used in DAT testing, the
given, the results can be fatal. patient’s DAT will usually be positive. If monospecific reagents
are used, the DAT may be positive due to complement alone.
Drug-Dependent or Immune Complex (“Innocent Tests with anti-IgG are usually negative, even when the anti-
Bystander”) Mechanism body is of the IgG class, because the drug–antidrug complex
is thought to elute from the cells during the washing proce-
Although the occurrence is rare, many drugs have been im- dure before the antiglobulin test.57,64 Other routine blood
plicated in causing immune-mediated problems by the so- bank tests are negative in all phases; the antibody is directed
called immune complex mechanism. This mechanism was against a drug or the drug-RBC membrane neoantigen, not a
first described in the early 1960s, and it was thought that true RBC antigen. Therefore, the antibody screening proce-
drugs operating through this mechanism combine with dures and compatibility tests are negative unless an alloanti-
plasma proteins to form immunogens.62 The antibody pro- body is also present. An eluate tested with reagent RBCs will
duced (often IgM, but IgG antibodies may also be present) also be nonreactive. A summary of typical serologic results is
recognizes determinants on the drug. If the patient ingests given in Table 21–15.
the same drug (or a drug bearing the same haptenic group) To confirm that a positive DAT is caused by a drug-
after immunization, the formation of a drug–antidrug com- antidrug reaction through the immune complex mechanism,
plex may occur. The complement cascade may be activated it must be demonstrated that the antibody in the patient’s
because of this antigen–antibody interaction. RBCs are serum reacts only if the drug is added to the test system. The
thought to be involved in this process only as “innocent by- patient’s serum is incubated with a solution of the drug in
standers.”63 The soluble drug–antidrug complex nonspecif- question and ABO-compatible (and antigen-negative if there
ically adsorbs loosely to the RBC surface. Complement, is alloantibody present) RBCs. Complement activation is the
when activated, sensitizes the cell and may proceed to lysis usual indicator of an antigen-antibody reaction; therefore,
(Fig. 21–7). one should observe for hemolysis after incubation and use
More recently, the concept of “neoantigen” formation has reagents containing anti-C3 for the antiglobulin test.
been proposed. In this model, the drug interacts in a nonco-
valent manner with a specific membrane component and A general procedure for demonstrating antibodies
forms a new antigen (neoantigen) determinant consisting of reacting by the immune complex mechanism, sug-
both drug and membrane components. This explanation is gested by Garratty,57 is given in Procedure 21–8 on
the basis of the unifying hypothesis discussed above. the companion website.
Because complement activation is involved in the im- For the test results to be interpreted correctly, adequate
mune complex, clinically affected patients frequently present controls must be performed. The patient’s serum must not
with acute intravascular hemolysis. Up to half of the patients react with the cells when saline or the diluent used to dis-
affected also have renal failure. When other causes for solve the drug is substituted for the drug solution, and the
hemoglobinemia and hemoglobinuria have been excluded, drug solution must not hemolyze the suspension of cells
nonspecifically. Examples of typical reactions with the pa-
tient’s serum and control are given in Table 21–16. An eluate
Complement from the patient’s cells is usually nonreactive even if the drug
and a source of complement are added. Very little antibody,
if any, remains on the cells after washing.
Complex
formation
In most blood banks, confirmatory testing is done only
when the patient has hematologic complications and not
when he or she simply has a history of taking the drug and
Drug
a positive DAT. Some of the drugs known to cause immune
complex–mediated problems are in frequent use, and there
are large numbers of patients with positive DATs but no ev-
RBC
idence of hemolysis. A full workup may be of only aca-
demic interest and is not required before release of RBCs
for transfusion. Treatment involves discontinuing the use
of the drug. Although hemolysis by this mechanism is rare,
the onset is usually sudden and may be characterized by
intravascular hemolysis and renal failure. Therefore, imme-
Antibody
diate cessation of the drug is essential. Steroid treatment
Figure 21–7. Immune complex mechanism. may also be given.
6888_Ch21_441-475 29/10/18 5:38 PM Page 466

466 PART III Transfusion Practices

Table 21–15 Serologic Reactions Observed With Drug-Induced Positive DATs


Mechanism Immunoglobulin on RBC Serum and Eluate React With:
Immune complex C3—occasionally IgG and IgM Cells, only if serum was incubated with the drug prior to testing. Routine
testing with reagent RBCs is negative.

Drug adsorption IgG—rarely C3 may be present Cells only if they are coated with the specific drug prior to testing. Routine
testing with reagent RBCs is negative.

Drug-independent IgG All “normal” RBCs. Routine testing with reagent RBCs is positive.

Membrane modification IgG, IgM, IgA, C3 No cells tested. The mechanism is nonimmunologic protein adsorption.

Table 21–16 Test Results Observed in the Presence of Antidrug-Antibody Reactive by the
Immune Complexing Mechanism
Test Patient Fresh Serum* Drug RBCs Results
Patient serum control X — — X If negative, indicates no alloantibody to RBC present.

Fresh serum control — X X X If negative, indicates no alloantibody to RBC or drug


present.

Drug/RBC control — — X X If negative, indicates drug solution will not cause


agglutination alone.

Patient test X X X X If agglutination is present and controls are all nega-


tive, indicates the presence of antidrug antibody. If
negative, no antidrug present.

*Fresh serum is the source of complement in this test.


X = sample added to tube; — = no sample added to tube

Membrane Modification (Nonimmunologic


Protein Adsorption)
RBC
It is hypothesized that the cephalosporins, especially
cephalothin (Keflin), both operate through the drug-adsorption
mechanism and are able to modify RBCs so that plasma proteins
(e.g., IgG, IgM, IgA, and complement) can bind to the mem-
brane (Fig. 21–8).65 Consequently, RBCs from approximately
3% of patients receiving Keflin may exhibit a positive DAT
with polyspecific and monospecific reagents. The uptake of
immunoglobulins or complement components is not the result
of a specific antigen-antibody reaction, so this mechanism is
nonimmunologic. Because antibodies with blood group speci-
ficities are not involved, tests with the patient’s serum and
eluate are negative (see Table 21–15). Numerous cases of Drug
cephalosporin-associated hemolytic anemia have been re-
ported, but as stated earlier, RBC destruction seems to have
been mediated through the drug adsorption or immune com-
plex mechanism rather than the membrane modification
mechanism. There is no treatment approach because hemolytic Proteins
anemia associated with the ingestion of these drugs has not
been described in relation to membrane modification. Figure 21–8. Nonimmunologic adsorption of proteins onto RBCs.

Autoantibody Formation drug and RBC membrane, alpha-methyldopa (Aldomet) in-


duces the production of an autoantibody that recognizes RBC
Unlike the drugs acting through the previously described antigens.66,67 Although Aldomet is used rarely to treat hyper-
mechanisms that induce production of an alloantibody tension in current practice, a review of its history is important
against a determinant on a drug or on a combination of the because it has been so well studied. The antibodies produced
6888_Ch21_441-475 29/10/18 5:38 PM Page 467

Chapter 21 Autoimmune Hemolytic Anemias 467

are serologically indistinguishable from those seen in patients are usually coated with IgG and rarely with complement
with WAIHA. Presence of the drug is not required to obtain components.
positive reactions. Positive DATs are encountered in approx- Because the serology of Aldomet-induced positive DAT/
imately 10% to 20% of patients receiving Aldomet as an an- immune hemolytic anemia is identical to that of the idiopathic
tihypertensive; however, very few (0.5% to 1%) of this group WAIHA, one cannot establish in the laboratory that Aldomet
of patients subsequently develop significant immune he- is the instigating factor; however, if the patient is receiving the
molytic anemia,7,68 and this occurs only after patients have drug, one should be highly suspicious. If Aldomet is with-
been taking Aldomet for approximately 6 months. If the drug drawn, autoantibody production will eventually stop, but it
is stopped, the hemolytic anemia gradually resolves. Other may be several months before the DAT is negative.
drugs that also cause autoantibody production are L-dopa,
mefenamic acid (Ponstel), procainamide, and diclofenac.68,69 Treatment of Drug-Induced Immune Hemolytic
The second- and third-generation cephalosporins, fludara- Anemia
bine, and cladribine, have also been associated with auto-
antibody formation. Discontinuation of the drug is the treatment of choice for
Several mechanisms by which Aldomet causes the pro- patients with a drug-induced hemolytic anemia. The pres-
duction of autoantibodies have been proposed.4,61,67 Kirt- ence of a positive DAT result does not necessarily imply that
land, Horwitz, and Mohler5 propose that methyldopa alters the drug must be discontinued if its effects are of therapeutic
the function of T-suppressor cells and suggest that this upset benefit and significant hemolysis is not present. In general,
in the immune system would allow production of antibody other drugs should be substituted, and the patient should be
against self. Worlledge and associates67 suggest that the drug observed for resolution of the anemia to confirm a drug-
alters RBC membrane components, similar to the theory of induced hemolytic process. If the patient has a positive DAT
neoantigen formation. without hemolysis, continued administration of the drug
The serologic features of this type of drug-induced prob- is optional. Generally, the prognosis for patients with drug-
lem are very different from those of the other drug-related induced hemolytic anemia is excellent, as long as the cause
immune hemolytic anemias. As shown in Table 21–17, the is recognized and the drug is stopped. Table 21–17 compares
antibody in the eluate does react with normal RBCs in the the four recognized mechanisms leading to the development
absence of the drug. Antibodies of similar specificity and of drug-related antibodies.70 Table 21–18 contrasts the anti-
reactivity may be found in the serum. The patient’s RBCs body characteristics of the various types of AIHAs.70

Table 21–17 Mechanisms Leading to Development of Drug-Related Antibodies


Prototype Frequency of
Mechanism Drugs Ig Class DAT Results Eluate Hemolysis
Immune complex Quinidine IgM or IgG Positive—often C3 only, Often negative Small doses of drugs
Phenacetin but IgG may be present. may cause acute
intravascular hemolysis
with hemoglobinemia/
hemoglobulinuria;
renal failure common.
Drug adsorption Penicillins IgG Strongly positive Often negative 3%–4% of patients
on large doses
Streptomycin
(10,000,000 units) of
Cephalosporin penicillin, which is one
of the most common
causes of immune
hemolysis, usually
extravascular.
Membrane Cephalosporins Many plasma Positive because of variety Negative No hemolysis; however,
modification proteins of serum proteins 3% of patients receiv-
ing the drug develop a
positive DAT.
Methyldopa Methyldopa IgG Strongly positive Positive—warm auto- 0.8% of patients develop
induced (Aldomet) antibody identical a hemolytic anemia
to that found in that mimics a warm
WAIHA AIHA. 15% of patients
receiving methyldopa
develop a positive DAT.

Adapted from Harmening, DM: Clinical Hematology and Fundamentals of Hemostasis, ed 4. FA Davis, Philadelphia, 2002, p 215, with permission.
6888_Ch21_441-475 29/10/18 5:38 PM Page 468

468 PART III Transfusion Practices

Table 21–18 Summary of Antibody Characteristics in AIHA


Warm-Reactive Cold-Reactive Paroxysmal Cold Drug-Related
Autoantibody Autoantibody Hemoglobinuria Autoantibody
Immunoglobulin Polyclonal IgG—occasionally Polyclonal IgM in infec- Polyclonal IgG Polyclonal IgG
characteristics IgM and IgA may be tion monoclonal kappa
present chain IgM in cold
agglutinin disease
Complement activity Variable Always Always Depends on the mecha-
nism of drug, antibody,
and RBC interaction
Thermal reactivity 20°C–37°C (optimum 37°C) 4°C–32°C, rarely to 37°C 4°C–20°C biphasic 20°C–37°C (optimum
(optimum 4°C) hemolysin 37°C)
Titer of free antibody Low to moderate(<32), High (>1,000 at 4°C) Moderate to low (<64) Depends on the mecha-
may be detectable only nism of drug, antibody,
with enzyme-treated cells and RBC interaction
Reactivity of eluate Usually panreactive Nonreactive Nonreactive Panreactive with methyl-
with antibody dopa type; nonreactive
screening cells in all other cases
Most common Anti-Rh precursor; anti- Anti-I, anti-i, anti-Pr Anti-P Anti-e–like, methyldopa
specificity common Rh; anti-LW; antidrug
anti-U
Site of RBC destruction Predominantly spleen with Predominantly liver, Intravascular Intravascular and spleen
some liver involvement rarely intravascular

Adapted from Harmening DM: Clinical Hematology and Fundamentals of Hemostasis, ed 4. FA Davis, Philadelphia, 2002, p 217, with permission.

By understanding the mechanisms by which drugs can 4. Prepare and test an eluate for RBC alloantibodies if the
cause a positive DAT or immune hemolytic anemia, it can patient has been recently transfused.
quickly be decided which laboratory tests are most likely
After evaluating this information, one can decide whether
to be informative. Many other drugs have been cited as
drugs are a possible cause of the problem and which of the
a cause for hemolytic anemia; however, the majority of
mechanisms may be involved. It is wise to also note that
these have only one reported case, and the association
many drugs cause an immunologic reaction by both drug-
between the hemolytic anemia and the drug may be
dependent and drug-independent mechanisms. When other
unclear. Before any special testing is done, proceed in the
causes (e.g., transfusion reaction) have been excluded and
following manner:
the clinical situation warrants additional testing, drug-coated
1. Obtain the patient’s medical history, including transfu- cells or solutions of the drug can be prepared for confirma-
sions, pregnancies, medications, and diagnosis. tory tests. Testing procedures for the investigation of drug-
2. Perform a DAT using RBCs collected in EDTA. Test the induced AIHA, require a library of control sera and cells and
cells with a polyspecific antihuman serum and monospe- expertise in this testing. Without appropriate controls, it is
cific reagents. possible to misinterpret test results and report either false-
3. Screen the patient’s serum for RBC alloantibodies. positive or false-negative results.

CASE STUDIES
Case 21-1

A 71-year-old man from northern Minnesota is admitted to the hospital with complaints of severe fatigue, shortness of
breath, “pounding heart,” and numbness in his fingers. He first noticed the symptoms when setting up his lake shelter for
ice fishing, as he does every year. Although he was mostly annoyed, his wife was concerned and made him go to the
doctor. An automated CBC done at the doctor’s office showed a hemoglobin value of 7.8 g/dL and hematocrit of 23.5%.
His doctor has ordered a bone marrow biopsy, a type and crossmatch for 3 units of packed red blood cells, and a direct
Coombs’ test. A sample of his blood is drawn and sent to the blood bank for the crossmatch and DAT.
6888_Ch21_441-475 29/10/18 5:39 PM Page 469

Chapter 21 Autoimmune Hemolytic Anemias 469

The type and screen results are as follows:


RBC (Forward) Typing Serum (Reverse) Typing
Anti-A Anti-B Anti-D 6% alb A1 RBCs B RBCs
Pt’s RBCs 4+ 2+ 4+ 2+ Pt’s Serum 4+ 4+
Note: The technologist also commented that the EDTA tube appeared clotted when it was first received in the blood bank.
Antibody Screening Immediate Spin LISS 37°C IAT Anti-IgG
SI 4+ 1+ 03
S II 4+ 1+ 03
S III 4+ 1+ 03
Auto Control 2+ 1+ 03
Note: 03 indicates negative IAT reading with check cells added and reactive as expected.
Direct Antiglobulin Test
Polyspecific Anti-IgG Anti-C3 6% alb
Pt’s RBCs 3+ 03 3+ 0
1. What is the patient’s ABO/Rh type?
2. How do the results of the antibody screening help explain the ABO discrepancy?
3. What immunoglobulin is coating the patient’s RBCs?
4. What is the most likely cause of this combination of results? What is the most logical next step?
The results of additional testing are below:
RBC (Forward) Typing Serum (Reverse) Typing
Anti-A Anti-B Anti-D 6% alb A1 RBCs B RBCs
Pt’s warm-washed RBCs 4+ 0 4+ 0 Pt’s cold adsorbed serum 0 4+
Antibody Screening Using Cold
Autoadsorbed Serum Immediate Spin LISS 37°C IAT Anti-IgG
SI 0 0 03
S II 0 0 03
S III 0 0 03
Auto control (with warm-washed RBCs) 0 0 03
1. What is the patient’s ABO/Rh type?
2. What sample should be used to perform crossmatching?
3. What other diagnostic testing might be useful?

Case 21-2

A 52-year-old female high school band teacher nearly fainted one afternoon while demonstrating some trombone-playing
techniques to one of her students. She went to her physician complaining of shortness of breath, mild depression, irri-
tability, and a loss of appetite (which was uncharacteristic). Her physician noted signs of anemia so had some hematology
tests done. Most notable was her hemoglobin of 5.2 g/dL, hematocrit of 14.8%, and reticulocyte count of 10.5%. He sent
blood samples to the hospital blood bank for type and crossmatch for 4 units of red blood cells for out-patient transfusion
ASAP. The patient was not thrilled at the prospect of a transfusion since she had a bad experience one time previously
when transfused for multiple fractures incurred in a motorcycle accident in her wilder days.
The blood bank tested her samples and got the following results:
RBC (Forward) Typing Serum (Reverse) Typing
Anti-A Anti-B Anti-D 6% alb A1 RBCs B RBCs
Pt’s RBCs 0 0 3+ 0 Pt’s Serum 4+ 4+
6888_Ch21_441-475 29/10/18 5:39 PM Page 470

470 PART III Transfusion Practices

Antibody Screening Immediate Spin LISS 37°C IAT Anti-IgG


SI 0 0 2+
S II 0 0 2+
S III 0 0 4+
Auto Control 0 0 3+
Unit 1 0 0 4+
Unit 2 0 0 2+
Unit 3 0 0 2+
Unit 4 0 0 4+
1. What is the patient’s ABO/Rh type?
2. What do the results of the antibody screening and crossmatches suggest?
3. What is the next logical step in investigating these preliminary results?
Answers to Case 21-2:
1. The patient is group O Rh positive.
2. Her antibody screening is positive with all three screening cells, as well as with the autologous control, at the indirect
antiglobulin phase. None of the four units crossmatched is compatible. There is some variability in the strength of the
IAT reactivity. Because the patient has no history of recent transfusion (within the last three months) the positive auto
control by IAT suggests a warm autoantibody is present. The variability of reactivity suggests alloantibody may also be
present.
3. A direct antiglobulin test should be done. Warm autoadsorption of the patient’s serum should also be done, and an
antibody identification panel tested with the adsorbed serum.
The results of additional testing are below:
Direct Antiglobulin Test
Polyspecific Anti-IgG Anti-C3 6% alb
Pt’s RBCs 3+ 3+ 1+ 0
The patient’s serum was warm autoadsorbed ×3 with ficin-treated aliquots of her RBCs. An antibody identification panel
with the autoadsorbed serum gave the following results:
Cell D C E c e P1 M N S s Lea Leb K k Fya Fyb Jka Jkb LISS IAT
1 + + 0 0 + + + + 0 + 0 + 0 + + 0 + + 2+
2 + + 0 0 + + + + 0 + 0 + + + + + + 0 3+
3 + 0 + + 0 + 0 + 0 + 0 + 0 + 0 + 0 + 03
4 0 0 0 + + 0 + 0 + + + 0 0 + 0 + 0 + 03
5 0 0 0 + + + + + + + 0 + 0 + + + + + 2+
6 0 + 0 + + 0 + 0 + + 0 0 + + 0 + 0 + 03
7 0 0 + + + + + + + 0 0 + 0 + + 0 + + 2+
8 + 0 0 + + + + + + 0 0 0 0 + 0 0 0 + 03
9 + + 0 + + + 0 + + + + 0 0 + + 0 + 0 3+
10 + + + + + 0 + 0 + + 0 + 0 + 0 + + + 2+
11 + + 0 0 + + + + 0 + 0 + 0 + + 0 0 + 03
12 0 0 0 + + + 0 + + + 0 0 0 + + + + + 2+
4. What do the results of the DAT suggest?
5. What alloantibody is identified in the warm autoadsorbed serum?
6. What is the appropriate blood selection for this patient?
7. What other treatment is she likely to receive?

Case 21-3

A 32-year-old female is admitted to the hospital in active labor at term with her first baby. After 8 hours of labor with
not much progress, there are signs of fetal distress, and she is taken for an emergency Cesarean section. The baby is soon
delivered and is healthy. After 3 days of care, mother and baby are discharged. A week later the mother returns to the
6888_Ch21_441-475 29/10/18 5:39 PM Page 471

Chapter 21 Autoimmune Hemolytic Anemias 471

emergency room of the hospital with complaints of extreme weakness, mild jaundice, and some back pain. An automated
CBC reveals a hemoglobin of 6.9g/dL and hematocrit of 21.3%. The ER physician orders 3 units for transfusion STAT.
A sample is drawn and sent to the blood bank.
The type and screen results are as follows:
RBC (Forward) Typing Serum (Reverse) Typing
Anti-A Anti-B Anti-D 6% alb A1 RBCs B RBCs
Pt’s RBCs 0 4+ 4+ 0 Pt’s Serum 4+ 0
Note: The technologist performing the testing commented that the EDTA plasma and serum were dark red, but the phle-
botomist denied having difficulty drawing the samples.
Antibody Screening Immediate Spin LISS 37°C IAT Anti-IgG
SI 0 0 03
S II 0 0 03
S III 0 0 03
Auto Control 0 0 3+
1. What is the patient’s ABO/Rh?
2. What are the results of the antibody screening?
3. Are there any atypical results worth pursuing before setting up the crossmatch?
Further testing reveals:
Direct Antiglobulin Test
Polyspecific Anti-IgG Anti-C3 6% alb
Pt’s RBCs 3+ 3+ 2+ 0
Eluate prepared from the DAT+ RBCs:
Eluate Last Wash
IAT Anti-IgG IAT Anti-IgG
SI 03 03
S II 03 03
S III 03 03
Auto Control 3+ 03
(EGA-ttd RBCs)
4. What do the results of the DAT suggest?
5. What do the results of the eluate suggest?
6. Should additional testing be done?
Cefotetan-Treated RBCs Untreated RBCs
37°C IAT 37°C IAT
Patient’s serum 1:100 4+ 4+ 0 03
Patient’s eluate 1:20 0 3+ 0 03
Last wash 1:20 0 03 0 03
Positive control 3+ 3+ 0 03
Negative control 0 03 0 03
Anti-cefotetan was identified in the patient’s serum and eluate. The patient must be warned that she should never receive
cefotetan again. Renal failure and fatality were seen in 19% of such cases in one study.8
6888_Ch21_441-475 29/10/18 5:39 PM Page 472

472 PART III Transfusion Practices

SUMMARY CHART
 Immune hemolytic anemia is defined as shortened of autoanti-P. This biphasic antibody binds to patient
RBC survival mediated through the immune response, RBCs at low temperatures and fixes complement.
specifically by humoral antibody. Hemolysis occurs when coated cells are warmed
 In alloimmune hemolytic anemia, patients produce to 37°C and complement-mediated intravascular lysis
alloantibodies to foreign RBC antigens introduced into occurs.
their circulation, most often through transfusion or  In WAIHA, most patients (67%) have both IgG and
pregnancy. complement on their RBCs, but 20% have only IgG
 Alloimmune hemolytic anemia is self-limiting; when and 13% have only complement.
the foreign RBCs are cleared from circulation, RBC  Warm-reactive autoantibodies are generally enhanced
destruction stops. by enzyme techniques and often have a broad speci-
 In autoimmune hemolytic anemia (AIHA), patients ficity within the Rh blood group system.
produce antibodies against their own RBC antigens.  When transfusing a patient with WAIHA, the primary
 In AIHA, the autoantibody is directed against the concern is detection and identification of all alloanti-
patient’s own RBCs; therefore, there is a consistent bodies that are masked by the warm autoantibody.
source of antibody and antigen present for continuous  In the immune complex drug mechanism, the soluble
RBC destruction. drug-antidrug complex nonspecifically adsorbs loosely
 In drug-induced immune hemolytic anemia, patients to the RBC surface, yielding a positive DAT with anti-C3
produce antibody to a particular drug or drug com- and sometimes with anti-IgG.
plex, with subsequent damage to RBCs.  In the drug-adsorption (hapten) mechanism, drugs
 AIHA may be classified as cold-reactive (18%), warm- such as penicillin or cefotetan bind firmly to proteins
reactive (70%), or drug-induced (12%); diagnostic of the RBC membrane. The DAT will show reactivity
tests include the DAT and characterization of the with anti-IgG and often with anti-C3.
autoantibody in the serum or eluate.  In the membrane modification drug mechanism, drugs
 In AIHA, serum antibody will not be detected until the such as the cephalosporins modify the RBCs so that
amount of antibody produced exceeds the number of plasma proteins can bind to the membrane nonim-
RBC antigen sites available on the patient’s own RBCs. munologically. The DAT will often demonstrate reac-
 The common antibody specificity in both benign and tivity with both anti-IgG and anti-C3.
pathological cold autoagglutinins is anti-I.  In the autoantibody drug mechanism, the drug (e.g.,
 In CHD, the DAT will be positive because of comple- alpha-methyldopa/Aldomet) induces production of an
ment coating of the RBCs; a cold antibody titer greater autoantibody that recognizes RBC antigens. Both the
than 1,000 may cause visible agglutination of anti- autoantibody and eluate are reactive with normal
coagulated blood at room temperature. RBCs. The DAT is reactive with anti-IgG.
 The classic antibody produced in PCH is called the
Donath-Landsteiner antibody, and it has the specificity

Review Questions 3. The blood group involved in the autoantibody specificity


in PCH is:
1. Immune hemolytic anemias may be classified in which of a. P.
the following categories? b. ABO.
a. Alloimmune c. Rh.
b. Autoimmune d. Lewis.
c. Drug-induced 4. Which of the following blood groups reacts best with an
d. All of the above anti-H or anti-IH?
2. When preparing cells for a cold autoadsorption proce- a. O
dure, it is helpful to pretreat the cells with which of the b. B
following? c. A2
a. Dithiothreitol d. A1
b. Ficin
c. Phosphate-buffered saline at pH 9
d. Bovine albumin
6888_Ch21_441-475 29/10/18 5:39 PM Page 473

Chapter 21 Autoimmune Hemolytic Anemias 473

5. With cold-reactive autoantibodies, the protein coating 13. Which of the following drugs has been associated
the patient’s cells and detected in the DAT is: with complement activation and rapid intravascular
a. C3. hemolysis?
b. IgG. a. Penicillins.
c. C4. b. Quinidine.
d. IgM. c. Alpha-methyldopa.
d. Cephalosporins.
6. Problems in routine testing caused by cold-reactive
autoantibodies can usually be resolved by all of the 14. A patient is admitted with a hemoglobin of 5.6 g/dL. Initial
following except: pretransfusion workup appears to indicate the presence of
a. Prewarming. a warm autoantibody in the serum and coating his RBCs.
b. Washing with warm saline. His transfusion history indicates that he received 6 units
c. Using anti-IgG antiglobulin serum. of RBCs 2 years ago after an automobile accident. Which
d. Testing clotted blood specimens. of the following would be most helpful in performing
antibody detection and compatibility testing procedures?
7. Pathological cold autoagglutinins differ from common
a. Adsorb the autoantibody using the patient’s enzyme-
cold autoagglutinins in:
treated cells.
a. Immunoglobulin class. b. Perform an elution and use the eluate for compatibility
b. Thermal amplitude. testing.
c. Antibody specificity. c. Crossmatch random units until compatible units are
d. DAT results on EDTA specimen. found.
8. Cold AIHA is sometimes associated with infection by: d. Collect blood from relatives who are more likely to
a. Staphylococcus aureus. be compatible.
b. Mycoplasma pneumoniae. 15. A patient who is taking Aldomet has a positive DAT. An
c. Escherichia coli. eluate prepared from his RBCs would be expected to:
d. Group A Streptococcus. a. React only with Aldomet-coated cells.
9. Many warm-reactive autoantibodies have a broad speci- b. Be neutralized by a suspension of Aldomet.
ficity within which of the following blood groups? c. React with all normal cells.
a. Kell. d. React only with Rhnull cells.
b. Duffy. 16. One method that can be used to separate a patient’s
c. Rh. RBCs from recently transfused donor RBCs is:
d. Kidd. a. Chloroquine diphosphate treatment of the RBCs.
10. Valid Rh typing can usually be obtained on a patient b. Reticulocyte harvesting.
with WAIHA using all of the following reagents or tech- c. EGA treatment.
niques except: d. Donath-Landsteiner testing.
a. Slide and modified tube anti-D. 17. Monoclonal antisera is valuable in phenotyping RBCs
b. Chloroquine-treated RBCs. with positive DATs because:
c. Rosette test. a. Both polyspecific and monospecific antihuman serum
d. Monoclonal anti-D. can be used in antiglobulin testing.
11. In pretransfusion testing for a patient with WAIHA, the b. Anti-C3 serum can be used in antiglobulin testing.
primary concern is: c. It usually does not require antiglobulin testing.
a. Treating the patient’s cells with chloroquine for reli- d. It does not require enzyme treatment of the cells prior
able antigen typing. to antiglobulin testing.
b. Adsorbing out all antibodies in the patient’s serum to 18. Autoadsorption procedures to remove either warm or
be able to provide compatible RBCs. cold autoantibodies should not be used with a recently
c. Determining the exact specificity of the autoantibody transfused patient. Recently means:
so that compatible RBCs can be found. a. 3 days.
d. Discovering any existing significant alloantibodies in b. 3 weeks.
the patient’s circulation. c. 6 weeks.
12. Penicillin given in massive doses has been associated d. 3 months.
with RBC hemolysis. Which of the classic mechanisms
is typically involved in the hemolytic process?
a. Immune complex.
b. Drug adsorption.
c. Membrane modification.
d. Autoantibody formation.
6888_Ch21_441-475 29/10/18 5:39 PM Page 474

474 PART III Transfusion Practices

References 24. Storry JR, Olsson MI, Moulds JJ. Rabbit red blood cell stroma
bind immunoglobulin M antibodies regardless of blood group
1. Allan J, Garratty G. Positive direct antiglobulin tests in normal specificity [letter]. Transfusion. 2006;46(7):1260-1.
blood donors [abstract]. Proceedings of the International 25. Judd WJ. Controversies in transfusion medicine: prewarmed
Society of Blood Transfusion, Montreal; 1980:150. tests: Con. Transfusion. 1995;35:271-5.
2. Izui S. Autoimmune hemolytic anemia. Curr Opin Immunol. 26. Chaplin H, Hunter V, Malacek A, Kilzer P, Rosche M. Clinically
1994;6(6):926-30. significant allo-anti-I in an I-negative patient with massive
3. Barthold DR, Kysela S, Steinberg AD. Decline in suppressor hemorrhage. Transfusion. 1986;26(1):57-61.
T cell function with age in female NZB mice. J Immunol. 27. Marsh WL. Aspects of cold-reactive autoantibodies. In: Bell CA,
1974;112(1):9-16. editor. A seminar on laboratory management of hemolysis.
4. Banacerraf B, Unanue ER. Textbook of immunology. Baltimore Washington (DC): American Association of Blood Banks; 1979.
(MD): Williams & Wilkins; 1979. p. 79.
5. Kirtland HH, Horwitz DA, Mohler DN. Inhibition of suppres- 28. Combs MR, O’Rourke M, Issitt P, Telen M. An auto-anti M
sor T cell function by methyldopa: a proposed cause of auto- causing hemolysis in vitro. Transfusion. 1991;31(8):756-8.
immune hemolytic anemia. N Engl J Med. 1980;302:825-32. 29. Garratty G. Target antigens for red-cell-bound autoantibodies.
6. van Loghem JJ. Concepts on the origin of autoimmune In: Nance ST, editor. Clinical and basic science aspects of im-
diseases: the possible role of viral infection in the etiology of munohematology. Arlington (VA): American Association of
idiopathic autoimmune diseases. Semin Hematol. 1965;9:17. Blood Banks; 1991. p. 33.
7. Petz LD, Garratty G. Immune hemolytic anemias. 2nd ed. 30. Lyckholm LJ, Edmond MB. Seasonal hemolysis due to cold-
Philadelphia: Churchill Livingstone; 2004. agglutinin syndrome. N Engl J Med. 1996;334(7):437.
8. Hillman RS, Finch GA. Red cell manual. 7th ed. Philadelphia: 31. Aoki A, Kay G, Zubiate P, Ruggio J, Ky J. Cardiac operation
FA Davis; 1996. without hypothermia for the patient with cold agglutinin.
9. Fung MK, Eder AF, Spitalnik SL, Westhoff CM, editors. Tech- Chest. 1993;104(5):1627-9.
nical manual. 19th ed. Bethesda, (MD): American Association 32. Shirey R, Park K, Ness P, Kickler T, Rones J, Dawson R, et al.
of Blood Banks; 2017. An anti-i biphasic hemolysin in chronic paroxysmal cold
10. Okuno T, Germino F, Newman B. Clinical significance of hemoglobinuria. Transfusion. 1986;26(1):62-64.
autologous control [abstract]. Am J Clin Pathol. 1984;16. 33. Judd WJ, Wilkinson s, Issitt P, Johnson T, Keren D, Steiner E.
11. Lau P, Haesler WE, Wurzel HA. Positive direct antiglobulin Donath-Landsteiner hemolytic anemia due to an anti-Pr-like
reaction in a patient population. Am J Clin Pathol. 1976; biphasic hemolysin. Transfusion. 1986;26(5):423-5.
65(3):368-75. 34. Reid M. Association of red blood cell membrane abnormalities
12. Judd WJ, Barnes B, Steiner E, Oberman H, Averill D, Butch S. with blood group phenotype. In Garratty G editor. Immunobi-
The evaluation of a positive direct antiglobulin test in pretrans- ology of transfusion medicine. New York: Marcel Dekker; 1994.
fusion testing. Transfusion. 1980;20(1):17-23. p. 257.
13. Garratty G, Petz LD, Hoops JK. The correlation of cold agglu- 35. Salloum E, Lundberg W. Hemolytic anemia with positive direct
tinin titrations in saline and albumin with haemolytic anaemia. antiglobulin test secondary to spontaneous cytomegalovirus
Br J Haematol. 1977;35(4):587-95. infection in healthy adults. Acta Hematol. 1994;92:39-41.
14. Engelfriet CP, Borne A, Giessen M, Beckers D, van Loghem J. 36. Benraad C, Scheerder H, Overbeeke M. Autoimmune haemolytic
Autoimmune hemolytic anemias: serological studies with anaemia during pregnancy. Eur J Obstet Gynecol Reprod Biol.
pure anti-immunoglobulin reagents. Clin Exp Immunol. 1994;55(3):209-11.
1968;3(7):605-14. 37. Nance S, Garratty G. Subclass of IgG on red cells of donors and
15. Rosse WF. Quantitative immunology of immune hemolytic patients with positive direct antiglobulin tests [abstract]. Trans-
anemia: the relationship of cell-bound antibody to hemolysis fusion. 1983;23(5):413.
and the effect of treatment. J Clin Invest. 1971;50(4):734-43. 38. Rodberg K, Tsuneta R, Garratty G. Discrepant Rh phenotyping
16. De Angelis V, De Matteis M, Cozzi M, Fiorin F, Pradella P, results when testing IgG-sensitized RBCs with monoclonal Rh
Stefan A, et al. Abnormalities of membrane protein composi- reagents [abstract]. Transfusion. 1995;35(S10):67S.
tion in patients with autoimmune haemolytic anaemia. Br J 39. Edwards JM, Moulds JJ, Judd WJ. Chloroquine dissociation of
Hematol. 1996;95(2):273-7. antigen-antibody complexes: a new technique for typing red
17. Gilliland BC, Baxter E, Evans RS. Red cell antibodies in ac- blood cells with a positive direct antiglobulin test. Transfusion
quired hemolytic anemia with negative antiglobulin serum 1982;22(1):59-61.
tests. N Engl J Med. 1971;285(5):252-6. 40. Issitt P, Anstee D. Applied blood group serology. 4th ed. Durham
18. Garratty G. Autoimmune hemolytic anemia. In: Garratty G, (NC): Montgomery Scientific Publications; 1998. p. 1021.
editor. Immunobiology of transfusion medicine. New York: 41. Win N, Kaye T, Mir N, Damain-Willems C, Chatfield C. Auto-
Marcel Dekker; 1994. p. 493. immune haemolytic anaemia in infancy with anti-Kpb speci-
19. Stratton F, et al. Acquired hemolytic anemia associated with ficity. Vox Sang. 1994;71(3):187-8.
IgA anti-e. Transfusion. 1972;12(3):157-61. 42. Becton DL, Kinney TR. An infant girl with severe autoimmune
20. Sturgeon P, Smith L, Chun H, Hurvitz C, Garratty G, hemolytic anemia: apparent anti-Vel specificity. Vox Sang.
Goldfinger D. Autoimmune hemolytic anemia associated 1986;51(2):108-111.
exclusively with IgA of Rh specificity. Transfusion. 1979; 43. Reynolds MV, Vengelen-Tyler V, Morel PA. Autoimmune
19(3):324-8. hemolytic anemia associated with auto anti-Ge. Vox Sang.
21. Hoppe PA. The role of the Bureau of Biologics in assuring 1981;41(2):61-67.
reagent reliability. In: Considerations in the selection of 44. Laine E, Leger R, Arndt P, Calhoun L, Garratty G, Petz L. In
reagents. Washington (DC): American Association of Blood vitro studies of the impact of transfusion on the detection of
Banks; 1979. p. 1. alloantibodies after autoadsorption. Transfusion. 2000;40(11):
22. Garratty G, Petz LD. An evaluation of commercial antiglobulin 1384-87.
sera with particular reference to their anticomplement proper- 45. Wilkinson SL. Serological approaches to transfusion of patients
ties. Transfusion. 1971;11(2):79. with allo- or autoantibodies. In: Nance ST editor. Immune
23. Reid ME. Autoagglutination dispersal utilizing sulfhydryl com- destruction of red blood cells. Arlington (VA): American
pounds. Transfusion. 1978;18(3):353-5. Association of Blood Banks; 1989. p. 227.
6888_Ch21_441-475 29/10/18 5:39 PM Page 475

Chapter 21 Autoimmune Hemolytic Anemias 475

46. Yun H, Cho D, Chae M, Jeon M, Sus S, Ryang D. No hemolytic 58. Eckrich RJ, Fox S, Mallory D. Cefotetan-induced immune
transfusion reactions in a patient with the apparent anti-e hemolytic anemia due to the drug-adsorption mechanism.
autoantibody following transfusion of packed red cells with Immunohematology. 1994;10(2):51-54.
CcDEe phenotype. Korean J Blood Trans.2007;18(2):116-20. 59. Bernini J, Mustafa M, Sutor L, Buchanan G. Fatal hemolysis
47. Jeffries LC. Transfusion therapy in autoimmune hemolytic ane- induced by ceftriaxone in a child with sickle cell anemia.
mia. Hematol Oncol Clin North Am. 1994;8(6):1087-1104. J Pediatr. 1995;126(5):813-5.
48. Anderson DR, Kelton JG. Mechanisms in intravascular and 60. Lascari A, Amyot K. Fatal hemolysis caused by ceftriaxone.
extravascular cell destruction. In: Nance ST, editor. Immune J Pediatr. 1995;126(5):816-7.
destruction of red blood cells. Arlington (VA): American 61. Garratty G. Drug-induced immune hemolytic anemia. Hema-
Association of Blood Banks; 1989. p. 39. tology Am Soc Hematol Educ Program. 2009;73-79.
49. Flores G, Cunningham-Rundles C, Newland A, Bussel J. Effi- 62. Shulman NR. Mechanism of blood cell destruction in individ-
cacy of intravenous immunoglobulin in the treatment of uals sensitized to foreign antigens. Trans Assoc Am Physicians.
autoimmune hemolytic anemia: results in 73 patients. Am J 1963;76:72.
Hematol. 1993;44(4):237-42. 63. Dameshek W. Autoimmunity: theoretical aspects. Ann N Y Acad
50. Garratty G, Arndt P, Domen R. Severe autoimmune hemolytic Sci. 1965;124(1):6-28.
anemia associated with IgM warm autoantibodies directed 64. Petz LD, Mueller-Eckhardt C. Drug-induced immune hemolytic
against determinants on or associated with glycophorin A. Vox anemia. Transfusion. 1992;32:2.
Sang. 1997;72(2):124-30. 65. Spath P, Garratty G, Petz LD. Studies on the immune response
51. Arndt PA, Garratty G. The changing spectrum of drug-induced to penicillin and cephalothin in humans: immunohematologic
immune hemolytic anemia. Semin Hematol. 2005;42(3):137-44. reactions to cephalothin administration. J Immunol. 1971;
52. Snapper I, Marks D, Schwarta L, et al. Hemolytic anemia sec- 107(3):854-9.
ondary to mesantoin. Ann Intern Med. 1953;39(3):619-23. 66. Carstairs K, Breckenridge A, Dollery C, Worlledge S. Incidence
53. Harris JW. Studies on the mechanism of a drug-induced of a positive direct Coombs’ test in patients on alpha-methyldopa.
hemolytic anemia. J Lab Clin Med. 1956;47:760-5. Lancet. 1966;2(7455):133-5.
54. Kerr R, Caradamone J, Dalmasso A, Kaplan M. Two mecha- 67. Worlledge S, Carstairs K, Dacie, J. Autoimmune hemolytic
nisms of erythrocyte destruction in penicillin-induced anemia associated with methyldopa therapy. Lancet. 1966;
hemolytic anemia. N Engl J Med. 1972;287(6):1322-5. 2(7455):135-9.
55. Ries C, Rosenbaum T, Garratty G, Petz L, Fudenberg H. Penicillin- 68. Petz LD. Drug-induced hemolytic anemia. Transfus Med Rev.
induced immune hemolytic anemia. JAMA. 1975;233(5):432-5. 1993;7(4):242-54.
56. Garratty G. Laboratory investigation of drug-induced immune 69. Lopez A, Linares M, Sanchez H, Blanquer A. Autoimmune
hemolytic anemia and/or positive direct antiglobulin tests. hemolytic anemia induced by diclofenac. Ann Pharmacother.
Washington (DC): American Association of Blood Banks; 1979. 1995;29(7-8):787.
57. Garratty G. Drug-induced immune hemolytic anemia. In: 70. Harmening DM. Clinical hematology and fundamentals of
Garratty G, editor. Immunobiology of transfusion medicine. hemostasis. 5th ed. Philadelphia (PA): FA Davis; 2009.
New York: Marcel Dekker; 1994. p. 523. p. 252–65.
6888_Ch22_476-496 29/10/18 5:38 PM Page 476

Chapter 22
Tissue Banking as a Part
of the Transfusion Service
Holli M. Mason, MD

Introduction Sterilization and Testing Allograft Failure


History and Overview of Tissue Implantation Reporting and Investigation
Transplantation Regulatory Overview Recalls
The Hospital Tissue Bank Federal Laws Case Studies
Tissue Bank Oversight State Laws Case Study 22-1
Acquisition of Allografts and Inventory American Association of Blood Banks Case Study 22-2
Storage American Association of Tissue Banks Summary Chart
Record-Keeping and Traceability The Joint Commission Review Questions
Quality Assurance Eye Bank Association of America References
Qualification of Vendors (EBAA)
Autologous Tissue Adverse Events
Preparation Infection
Storage Malignancy

OBJECTIVES
1. Name the principal modern organizations involved in standardization of tissue banks.
2. List examples of tissues contained within the scope of FDA CFR 1270 and 1271 and examples of tissues outside the scope of
these regulations.
3. State the minimum requirements for qualifying a tissue supplier and vendor.
4. List the minimum labeling requirements for autologous tissue and the conditions required to use autologous tissue without
registering with the FDA as a tissue-manufacturing establishment.
5. Compare and contrast the similarities and differences between the various regulatory bodies in terms of regulations for tissue
banks, and identify voluntary accrediting agencies.
6. Compare the steps taken in an adverse event investigation with those taken in a recall.

Introduction hospital administrators have evaluated their hospital structure


to determine the best place to assign this responsibility. The
The function of a hospital tissue bank is to consolidate, stan- blood bank has been a natural fit for many institutions, and
dardize, and centralize the handling of human-derived tissue. many transfusion services have taken on the additional role
The tissue bank is charged with qualifying tissue vendors, en- of tissue bank or tissue dispensing service.
suring proper handling and storage of tissues, monitoring of The Joint Commission’s standard TS.03.01.01, EP 1 does
temperature, record-keeping, and issuing procedures to ensure not require hospitals to locate the tissue bank within the
traceability. With the advent of the 2005 directive from the blood bank or require that a specific tissue bank be established;
Joint Commission to “assign responsibility for overseeing the it requires only that there is an assignation of responsibility for
tissue program throughout the organization including storage oversight.1 Some hospitals may choose to decentralize and
and issuance activity” (standard TS.03.01.01, EP 1),1 many allow surgeons to maintain their own inventories with an

476
6888_Ch22_476-496 29/10/18 5:38 PM Page 477

Chapter 22 Tissue Banking as a Part of the Transfusion Service 477

administrator who is responsible for ensuring that regulations transplantation, and non-transplant anatomical material for
and standards are followed. Validating vendor qualifications education. AATB seeks to accomplish its mission by estab-
may be handled by the purchasing department and adverse lishing and promulgating standards, administering accredi-
events investigated by the infectious diseases department. tation and certification programs, interacting with regulatory
This system may be feasible in smaller institutions but becomes agencies and legislative officials, and fostering education and
unwieldy in large institutions. The American Association of research.”5 Close cooperation is maintained with the U.S.
Tissue Banks (AATB) and the AABB Tissue Committees have Food and Drug Administration (FDA).
recommended the centralized model, stating that the ideal In 1984, the National Organ Transplant Act (NOTA) was
location for management and storage of human tissues for passed (Public Law 98.507).6 This act established a national,
transplant is in the hospital blood bank.2 centralized waiting list for organ recipients and outlawed the
The blood bank is a natural fit for a centralized model of sale or purchase of organs.
tissue banking because blood is simply tissue in liquid form. The 1990s saw an increase in the role of the FDA in tissue
Many of the standards related to tissue banking are quite banking. Beginning in 1993, the FDA required that donors
familiar to blood bankers and can be integrated into those be screened for hepatitis B, HIV, and behavioral risk factors.
already established for blood and blood products, such as In 1997, the FDA proposed a major overhaul of tissue regu-
monitored temperature control and record-keeping. Storing lations that eventually led to a more comprehensive approach
all tissues under the purview of a single department can focusing on three key areas: preventing the unwitting
simplify standardization and consistent implementation of transplant of contaminated tissues that could transmit dis-
standard operating procedures (SOPs). This chapter focuses ease, preventing improper handling that may contaminate
on the role of transfusion services in storing and issuing or damage tissue, and ensuring the demonstration of safety
tissues. It also touches upon the history of tissue transplan- and effectiveness of tissue products.
tation, the many uses of tissue products, and regulations and In 1998, the FDA proposed a new unified system for the
standards to give the reader better insight into the require- registration and listing of establishments that manufacture
ments for hospital tissue banks. For the purposes of this human cellular and tissue-based products and the listing of
chapter, the term hospital tissue bank refers to tissue man- their products. In 2001, the FDA proposed that establish-
agement services at the hospital level, generally restricted to ments manufacturing cellular and tissue-based products
receiving, storing, and issuing cells and tissues, and not to follow current good manufacturing practices (cGMP), similar
FDA-registered tissue establishments. to blood collection establishments. These include establish-
ment of standard operating procedures, quality programs,
History and Overview of Tissue record-keeping standards, and tracking procedures.7 Addi-
Transplantation tionally, the FDA required that manufacturing establish-
ments register with the agency and list their products.7
Tissue transplantation has had a long and varied history. The Joint Commission issued standards in 2005 that
Early attempts at transplantation were largely experimental included directives to assign a responsible party to oversee
and rarely successful. They were also highly controversial tissue programs within institutions for storage and issuing
from an ethical and religious standpoint. The first bone activities. Standards TS.03.01.01 and TS.03.02.01 also man-
transplant was recorded as early as 1682.3 Joints, corneas, date record-keeping for traceability and the investigation of
and skin were being transplanted by the early 20th century. adverse events.1
Organ transplant became feasible with the development of
vessel anastomosis through the work of Dr. Alexis Carrel, a The Hospital Tissue Bank
French surgeon who worked with cadaver vessels to develop
The basic function of the hospital tissue bank is to provide
the technique that was introduced in 1911.
surgeons and their patients with safe, quality tissue products
In 1949, the U.S. Navy established the first tissue bank.
while maintaining detailed records for each tissue handled
In its 50 years in existence, it created the first model for tis-
through final disposition. This includes AABB Hospital Tissue
sue harvest, processing, storage, and transplant.4 Many of
Management:8
the principles developed by the U.S. Navy Tissue Bank are
standards still followed today. The researchers here affected • Acquisition of safe and effective autografts and allografts
virtually every aspect of the process, from determining suit- • Oversight of use throughout the organization
able donors, sterilization techniques, cryopreservation, and • Qualification of suppliers
freeze-drying, to developing the immunologic principles that • Conduction of inspections
are so important in modern transplant medicine. The U.S. • Provision of temperature-controlled, monitored storage
Navy Tissue Bank was also instrumental in the formation • Record-keeping and ensuring traceability
of two major institutions in the tissue transplant field: the • Quality assurance
National Marrow Donor Program (NMDP) and the American • Selection of appropriate allografts
Association of Tissue Banks (AATB). • Investigation of adverse outcomes
The AATB was established in 1976. Its mission is “to • Administration of recalls and look-back investigations
honor donors and to save and improve lives by promoting • Compliance with regulations and standards
the safety, quality and availability of donated tissue for • Promotion of cost-effective and appropriate tissue use
6888_Ch22_476-496 29/10/18 5:38 PM Page 478

478 PART III Transfusion Practices

Common tissues found in tissue banks and used in sur- Inventory should take into account not only the “off the
gical procedures include bones, ligaments, tendons, heart shelf” utility of a particular product, but also its frequency
valves, skin, veins, cartilage, dura mater, hematopoietic stem of use and expiration date. Tissues that require specific sizes
and progenitor cells, and corneas or eyes. Bone can be ster- may be ordered in smaller quantities or only ordered in
ilized and all living tissue removed. Pieces of bone can be anticipation of specific procedures, depending on the size
further worked into spacers, wedges, screws of various sizes, of the organization and turnover of product. Tissues such as
and “chips.” These have a variety of surgical uses, including cornea have such a short shelf life that it is best to schedule
decompression of nerves in spinal surgery with donor bone patient surgeries as the tissue is available and issue the
spacers, screws to pin fractured bone together, or chips to cornea as soon after arrival as possible.9
aid in the incorporation of hip or knee replacements into the Surgeons should have access to the inventory lists and
native bone. Once implanted into the patient, bone slowly may even want to look at individual tissue prior to surgery
incorporates into the living native bone and adds strength to avoid wasting tissues that do not fit as expected for
and structure. surgeries requiring tissue of a specific size. At times, sur-
Heart valves are used for valve replacement, most com- geons may request a selection of tissues to choose from in
monly in children and women of childbearing age. The ad- the operating room when the patient’s situation is better
vantage of nonmechanical heart valves for this population understood and accurate measurements can be made.
is the avoidance of reliance on anticoagulants that are Clear guidelines specifying storage parameters should be
mandatory for recipients of mechanical valves. Drugs such based upon the tissue manufacturer’s recommendations
as Coumadin put active children in danger of serious bleed- and product insert. Storage devices may include ambient
ing and are teratogenic in pregnant women, potentially (room temperature) cabinets, refrigerators, freezers, or liquid
causing birth defects. nitrogen units. Continuous monitoring of refrigerators and
Tendons and ligaments are often used in repairs of knee freezers are required, and storage SOPs should address the
and other orthopedic injuries, particularly in the replacement steps to be taken in the event of temperature deviation
of the anterior cruciate ligament. Tendons and cartilage are from the recommended storage.10
also useful in repair of facial disfigurements. In general, the scope of the tissue bank is limited to
Skin is widely used for burn victims and often means the nonvascularized tissue. Excluded from Parts 1270 and 1271
difference between life and death. Not only are skin grafts of the FDA’s Code of Federal Regulations (CFR) are vascu-
an aesthetic improvement, but they also provide the barrier larized organs such as the liver, kidney, lung, pancreas,
to the outside world that is essential to protect the body from and heart.11 For the most part, these tissues are immedi-
pathogens. Skin grafts are also used in patients with any large ately transplanted and storage is not necessary or desirable.
loss of skin, such as pressure sores, nonhealing ulcers, auto- However, there are exceptions to this general rule. Renal
immune disorders, and surgical or traumatic loss of large transplants may be stored, if properly packaged, for several
areas of skin. days. Because kidneys are vascularized organs, and there-
Veins are commonly used in coronary artery bypass fore fall outside the general scope of the hospital tissue
grafts. They are also used frequently to bypass other vessels bank, it is not necessary to include such tissue in the tissue
that have become blocked by severe atherosclerosis. Such bank. However, organizations may opt for standardization
bypasses often make it possible for a patient to avoid limb and for the ensuring of proper storage and therefore in-
amputation. clude organs, such as kidneys, in tissue banks.1,12 The
Hematopoietic stem cells and progenitor cells are used United Network for Organ Sharing (UNOS) requires two
to repopulate bone marrow in patients who have undergone separate determinations of ABO type for both transplant
chemotherapy and irradiation to destroy tumor cells. This candidates and organ donors for some organ transplants.
type of therapy may destroy the native bone marrow as well, Major ABO incompatibility between the two can result in
which is fatal without the transplant of donor or autologous acute humoral rejection of the transplanted organ. The
stem cells following therapy. hospital blood and/or tissue bank may perform one or
Corneal transplants are used to restore sight in patients more of the blood types, depending upon how the solid organ
with corneas that have been damaged through trauma, dis- transplant program is organized. It is important to note that
ease, or birth defect. Eye tissue, or the sclera, which consists in some cases, ABO-incompatible organs are transplanted
of the “white” part of the eye, is composed of thick fibrous following a desensitization regimen, usually including
tissue. Sclera can be used as a patch for eyes that have been plasmapheresis.10
punctured or torn in a trauma.9 Sclera is also occasionally Other exclusions include autologous tissue, also known
used in dental procedures. as autograft tissue, which may also be stored in the tissue
The majority of the tissues described here have the ad- bank for convenience and standardization; blood vessels
vantage of being useful regardless of the donor’s or patient’s recovered with organs and intended for use in organ trans-
HLA type. No blood type or tissue match is required. Many plantation; minimally manipulated bone marrow; tissue
of the tissues come in standardized shapes and sizes (e.g., intended for education or nonclinical research; xenografts;
bone screws) and can be used by any patient. Other tissues blood components and blood products; and secreted or
must be ordered in advance specifically for an individual extracted products such as human milk, collagen, or cell
with a specific size, such as heart valves or whole bones. growth factors.11,12
6888_Ch22_476-496 29/10/18 5:38 PM Page 479

Chapter 22 Tissue Banking as a Part of the Transfusion Service 479

Tissue Bank Oversight He or she defines policies and procedures for the service,
ensures competency and training of the staff, manages ac-
The hospital tissue bank must have a physician-appointed quisition and inventory, and prepares reports for the tissue
medical director. In keeping with the blood bank model, the committee, including adverse event reporting. This person
AABB requires that the medical director, a licensed medical is often in the position of supervisor in the blood bank or
doctor, qualified by education, training, and experience, may be an additional person at the same level.
investigates adverse outcomes, participates in look-back
investigations, approves standard operating procedures Acquisition of Allografts and Inventory
with any deviations, and communicates with the physicians
using the end product.10,13 Because of this standard, hospital The hospital tissue bank serves as the clearinghouse for all
tissue banks within the blood bank have the advantage of tissues entering the hospital (excluding most or all vascular-
the presence of a qualified medical director. If the institution ized organs). It may be necessary or desirable to involve the
chooses to delegate this responsibility to another qualified surgical staff in decisions as to the standard inventory and
physician other than the medical director of the blood bank, day-to-day ordering; however, the tissue bank should be
this individual should participate in the development and responsible for approving tissue vendors and receiving all
review of all policies, procedures, and processes involving shipments.
the handling of tissue. He or she should also be involved in Once a shipment of tissue is received in the tissue bank,
investigating adverse events, deviations from standard oper- it should be inspected for package integrity and correct label-
ating procedures, and look-backs. An important part of the ing and reconciled with the packing invoice.13 Tissue receipt
Joint Commission requirement from 2005 is that there must records must contain, at a minimum, name and address of
be a standard operating procedure that assigns and desig- tissue supplier, description of tissue and quantity, date of
nates responsibility for the oversight of the organization’s tissue receipt, condition of tissue upon receipt, and expira-
tissue-related activities.1 tion date (AATB standard C3.000).5 Each tissue specimen
Development of a tissue committee is recommended to must have a tissue identification number (TIN). In addition,
ensure and promote best practices based on evidence-based the name of the person conducting the incoming inspection
medicine. The tissue committee may be structured in a way should be recorded on the tissue receipt record. Joint Com-
similar to the transfusion committee, including medical staff mission standards require that the individual receiving the
who are involved in the use of transplantable human tissue. shipment must verify package integrity and ensure that the
The chair may be the director of the blood bank, but the transport temperature range has been controlled and accept-
committee may prefer to choose a chair from the surgery de- able, as evidenced by residual dry or wet ice.1 The vendor
partment secondary to that physician’s knowledge regarding should be able to present evidence of validation of transport
tissue transplant and use. containers for maintenance of proper temperature range
Peer review, evidence-based clinical indications, and during transport.
audits ensuring compliance with standard operating proce- Manufacturers’ instructions for storage should be rigorously
dures should be major goals of the committee. The committee followed and the tissue entered manually or electronically into
should review operations, usage trends, shortages, wastages, an inventory list. If necessary, the storage site location should
and errors. All reported adverse outcomes should be brought be included in the inventory for ease of locating the tissue
to the tissue committee and investigated fully. The committee upon issue or inspection. Surgeons who transplant tissue
may also be involved in selecting tissue vendors and in should have access to the inventory so they are aware of
educating the hospital staff in the appropriate use of tissue. what tissues are available to them before they begin surgical
Often the tissue committee can be successfully incorporated procedures.
into the transfusion committee with committee members ful- Detailed and accurate inventory management is cost
filling both roles. saving. It will reduce wastage of products stored until expi-
Additional important roles within the hospital tissue bank ration and prevent inadvertently reordering when the
are the quality assurance coordinator and the hospital tissue product is already in inventory. Some products, such as
coordinator. The quality assurance coordinator, often known corneas, have a very short shelf life, and careful coordination
as the compliance officer, is responsible for the quality plan between the surgical staff and the tissue bank allow for order-
for the transfusion service as mandated by the AABB and ing and acquiring the appropriate tissue in time for the
College of American Pathologists (CAP).10 He or she assesses surgery, but not so far in advance that the product expires
processes, policies, and procedures to make sure they are and must be wasted. Duration of storage time will vary
in compliance with the applicable accrediting agencies. greatly between the different types of tissue. Therefore, a
Depending on the size of the organization, the job description system, preferably electronic, must be in place to anticipate
may include applying these duties to the entire laboratory or replacement of outdated tissues or management of those
to discrete parts. The quality assurance coordinator should tissues to avoid outdating.13
make recommendations to the medical director regarding To avoid waste, some tissue vendors will agree to exchange
operations. The hospital tissue coordinator is responsible for tissue that is close to outdated with similar tissue that has an
all administrative operations of the tissue dispensing service, expiration date further in the future. This is not true in all
including handling regulatory issues and hospital policies. cases and must be worked out between individual vendors
6888_Ch22_476-496 29/10/18 5:38 PM Page 480

480 PART III Transfusion Practices

and hospital tissue banks. Typically vendors who are willing function of the alarm is to allow blood and tissue bank per-
to exchange late-dated products have a network of hospital sonnel to be aware of an equipment problem before the prod-
tissue banks to which they can transfer the product for quick uct is compromised by a temperature that is too high or too
use. Wastage rate for each tissue type should be tracked and low for proper storage. Alarm systems must be set to activate
examined by the tissue committee for ways to improve the under conditions that will allow action to be taken before
process. Changes in the surgical staff or focus of the surgical products reach unacceptable conditions. Procedures should
center may result in unanticipated wastage if the same tissue be in place to instruct the blood and tissue bank personnel
inventory is kept despite changes in use. Tracking wastage on actions to take when the alarm is activated, including
and communicating with other departments using tissue can corrective action and possible relocation of products to another
keep wastage to a minimum. monitored storage device. An alarm activation must be imme-
diately investigated to find the cause of the malfunction.
Storage
Record-Keeping and Traceability
Storage conditions vary between the different tissue types.
Even the same tissue, such as bone or eye, can have different Good record keeping is essential in the hospital tissue bank
temperature and duration requirements depending upon its to demonstrate that standard operating procedures are fol-
processing and packaging.14 Therefore, it is critical to care- lowed (Box 22–1). Also important is the ability to track the
fully follow the recommendations and instructions from the tissue bidirectionally, from donor to recipient or disposal
tissue vendor or supplier for each specimen. Failure to do and back again. Each change in the status of the product—
so may result in tissue that does not function as intended including receipt into the hospital tissue bank, change in
and may interfere with the tissue’s functional and mechanical location within the tissue bank, issue, return, transplanta-
properties, making it unsuitable for transplant.14 Special tion, or disposal—must be documented concurrently with
attention should be paid to products stored at “room” or the action. The identity of the person documenting each step
“ambient” temperature. These words may vary in definition and the date should be included. Records must be accurate,
between tissue suppliers, and it is advisable to clarify the legible, written in indelible ink, and as detailed as possible
definition with the vendor. A written response to any such to provide a clear history.
question should be kept on file in the hospital tissue bank. Traceability becomes especially important during recalls.
The Joint Commission and AATB require that tissue Occasionally a tissue donor is found to have had a disease
storage shall conform to guidelines established by the dis- or disease exposure that was not apparent at the time of
tributing tissue bank5 and that organizations use standard- donation or may be suspected because one of the tissue
ized procedures to store tissues.7 The Joint Commission recipients became sick following transplant. In cases such as
further requires the hospital to continuously monitor storage this, all of the tissues that originated from that donor must
refrigerators and freezers, with records showing daily storage be investigated and tracked down to prevent transmission
temperatures.1 AABB takes a more detailed approach to of disease to other recipients. Often, transplant has already
storage. Standard 3.0 deals with equipment and specifically occurred and the recipient can be tested or treated if necessary.
includes tissue and tissue derivatives requiring identification Poor record-keeping makes it impossible to track potentially
of equipment critical to ensuring that appropriate tempera- dangerous tissues or to determine who may need treatment
tures are maintained, the same as for blood and blood com- or increased surveillance due to the transplant of a tissue
ponents.13 The blood bank is required to maintain policies,
processes, and procedures to ensure that calibration, main-
tenance, and monitoring of equipment conforms to the blood
bank or tissue bank standards.5 BOX 22–1
AABB standard 3.6 states that storage devices must have Standard Operating Procedure (SOP) Checklist
the capacity and design to ensure proper temperature is
The following SOPs should be written and included in the hospital
maintained.13 The temperature of refrigerators, freezers, and tissue bank:
platelet incubators must be monitored. Storage units using • Assignment of responsibility for the oversight of the tissue program
liquid nitrogen must have liquid nitrogen levels checked or • Validation and qualification of tissue suppliers/vendors
the temperature monitored. Tissues that can be stored at • Tissue ordering
room or ambient temperature do not require a monitored • Tissue receipt
storage device, such as a platelet incubator, as long as the • Tissue storage and temperature control
definition of room temperature does not include a narrow • Issuance and final inspection of tissue
temperature range.13 However, many tissue banks will store • Temperature alarms and emergency backup equipment
these products in a monitored device for uniformity and • Documentation of abnormalities, including instructions on actions
convenience. such as quarantine
Alarm systems have been used in blood banks for decades • Quarantine of tissues that are questionable
and are an important assurance that storage temperatures • Documentation of decisions not to use tissue
have been consistently maintained, thereby ensuring the • Disposal/disposition of expired tissue
integrity of the product being stored. The most important
6888_Ch22_476-496 29/10/18 5:38 PM Page 481

Chapter 22 Tissue Banking as a Part of the Transfusion Service 481

from a donor suspected of transmitting disease. The bidi- operating procedures and standards and regulations are being
rectional traceability is important so that if a recipient followed.
becomes ill after transplant, the tissue bank and supplier
can cooperate to trace the source of the tissue in order to Qualification of Vendors
initiate a recall.
Records must be maintained for a minimum of 10 years Tissue vendors or suppliers must meet a variety of standards.
after the expiration of the tissue product.5 If no expiration First, the tissue supplied must be safe and effective and of
date applies to a specific tissue specimen, records must be use to the physician who transplants it. The tissue must be
maintained for 10 years following issue.5 Each tissue speci- readily available in a useful time frame, and its cost must fall
men must have a unique tissue identification number (TIN), within budgetary guidelines and be within the customary
and upon receipt by the hospital tissue bank, must include price range. The surgeons who will use the tissues should be
the name and address of the tissue supplier, a description of involved in selecting the tissue vendor based on the tissue’s
the tissue and quantity received, date of receipt, condition usefulness to them. Most often, a surgeon will request a
upon receipt, and expiration date along with the name of the specific tissue from a specific vendor because of its utility. It
receiving individual. is then up to the medical director of the hospital tissue bank
When a tissue is dispensed to be used for transplant, to determine whether the vendor qualifies as a supplier of
dispensing records must be created. The dispensing record quality, safe, and effective tissues.
must include the name, address, and telephone number of Sales representatives must be compelled to seek approval
the tissue bank; the type and quantity of tissue; the unique and qualification with the hospital tissue bank prior to
TIN; the recipient’s name, medical record number, or social demonstrating any new tissue products in the operating
security number; transplantation site; date and time of release; room. It is also important to consider other physicians within
name of the ordering physician or other health professional; the organization who may also utilize tissue for patient care
name of the person dispensing the tissue; and name of the in a nonsurgical setting, such as a wound care clinic, and
person preparing the tissue if further preparation is necessary solicit their input as well.
prior to transplant.5 The dispensing records must be main- Before the qualification of a vendor can commence,
tained in a log format in the hospital tissue bank. It must the hospital tissue bank must have written guidelines and
also be included as part of the recipient’s medical record to criteria for the acceptance of a vendor or supplier.10 Some
permit tracing to the appropriate individual. Tracing forms vendors may be part of a larger entity in which different
must also be filled out and returned to the tissue supplier or establishments participated in different parts of the process
vendor for their records.5 The records should clearly specify of supplying a tissue product. For example, one establish-
the final disposition of the tissue. The tissue will have been ment may recover the tissue and pass it on to another that
implanted, meaning the tissue was implanted in the patient; processes and packages the tissue for distribution. The writ-
discarded, meaning the tissue was discarded in the operating ten policy should reflect if each of the intermediaries should
room and not implanted into the patient; or explanted, also be qualified by the hospital tissue bank. Theoretically,
meaning the tissue came into contact with the patient but qualification of the supplier directly supplying the tissue
was removed either immediately during surgery or at a should be sufficient, as that supplier must have, in turn, qual-
later date. ified its suppliers. However, some medical directors may feel
more secure in verifying this for themselves. The medical
Quality Assurance director may also take into account such things as the trans-
parency and willingness to provide information, method of
Quality is the goal of any blood or tissue bank. A quality processing and disinfecting, the involvement of the supplier’s
plan is essential for good tissue practices and excellent patient medical director, and the ability of the supplier to meet spe-
care. The goals of the quality plan are to maintain safe, func- cial needs. Table 22–1 outlines the required documentation
tional, and effective products that meet the needs of the for tissue vendor and supplier qualification.
patient and the surgeon. Preventing contamination and hav- The first step in qualifying any vendor is to verify the sup-
ing an error-free process is critical. To achieve these goals, the plier’s FDA registration. On a yearly basis, the tissue bank must
quality plan must include written procedures, validation of verify that its suppliers are registered with the FDA as required
processes, and investigation of deviations and errors. Errors by 21 CFR 1271.15 This can be done by using the FDA’s online
will occur even in the best-run tissue banks. Well-written database: www.fda.gov/cber/tissue/tissregdata.htm. In January
standard operating procedures that conform to the regula- 2001, the FDA published a rule requiring establishments
tions and are easy to follow in actual practice decrease errors. that supply human cells, tissues, and cellular and tissue-
However, analysis of deviations and errors, as well as devel- based products (HCT/Ps) to register with the agency and list
opment of corrective action to improve process and prevent their HCT/Ps.15 The final rule, 21 CFR Part 1271, became
future errors, builds quality over time. effective in April 2001 for human tissues intended for trans-
The quality plan should also verify supplier qualifica- plant that are regulated under section 361 of the Public
tions, ensure proper training of personnel, and monitor com- Health Service (PHS) Act and 21 CFR 1270.16 In March
pliance in the tissue bank. Periodic quality audits should 2001, the requirement became effective for all other HCT/Ps
be performed by the tissue service to ensure that standard except human dura mater and heart valves, which became
6888_Ch22_476-496 29/10/18 5:38 PM Page 482

482 PART III Transfusion Practices

Table 22–1 Documentation for Tissue accredited establishments on their websites, which can be
found at www.aatb.org and www.restoresight.org, respectively.
Vendor/Supplier Qualification
Finally, the Joint Commission requires that hospital tissue
Information Required/ banks ensure that tissue establishments are licensed by state
Document Location Found agencies when required by state law in the physical location
Tissue supplier’s FDA • Federal registration number of the organization.1 Not all states require licensure; in fact,
registration (Form • Physical location of the establishment most do not. As of this writing, six states require licensure:
FDA 3356)* • Reporting official’s information New York, Florida, California, Georgia, Delaware, District of
• Establishment’s function Columbia, Illinois, Louisiana, Massachusetts, Mississippi,
• Product information
• Proprietary names (www.fda.gov/ Oklahoma, Oregon, and Maryland (Table 22–2).
cber/tissue/tissregdata.htm)
Autologous Tissue
Tissue supplier’s List of accredited suppliers:
voluntary accreditation • www.aatb.org
• www.restoresight.org
The term autologous tissue refers to any tissue that is re-
moved from a patient with the intent to re-implant it at a
Tissue supplier’s state When suppliers are located in states that later date and during a different procedure from that of its
license and registration require licensure by the state removal. Strictly speaking, a bone flap that is elevated and
placed in sterile saline during a procedure and then replaced
*Required by the Joint Commission
at the end of that same procedure does not qualify as being
autologous tissue as defined in this chapter.17 In order for
the regulations and standards pertaining to autologous tissue
effective when the remaining parts of 21 CFR 1271 were to apply, that bone flap would require additional processing,
implemented in 2005.16 storage, and re-implantation at another time within the same
Establishments that meet the requirements for registra- facility.1,17 Ironically, the Joint Commission defines this as
tion with the FDA must do so within 5 days after beginning the “same procedure,” as it is done by the same surgeon in
operations.15 Registrations must be updated annually in
December in conjunction with an update of the HCT/P list
unless there is no change. Upon acceptance of the establish- Table 22–2 State Laws Governing Tissue
ment’s registration form, the FDA will assign each location a Banking29
permanent number.15 This registration number does not State Applicable Laws
imply that the establishment is in compliance with applicable
rules and regulations or is approved by the FDA. However, California License. California Health and Safety
Code § 1635.1, Chapter 4.1 and 4.2
the establishment, once registered, must be available for
public inspection by the FDA (CFR 1271).15 A yearly review Florida Certification. Florida statutes 765.542
of FDA registration of tissue vendors is recommended in
Georgia License. Georgia Comp. Rules & Regs
the first quarter of each year because of the requirement that § 290-9-8-04
the establishments update their registration each December.
A copy of the form used to register with the FDA (Form Maryland Permit. Maryland Code Ann. § 17-305
FDA 3356) should be obtained from the vendor and kept Delaware Registration. 16 Delaware Code § 2801
on file in the hospital tissue bank.15 The registration form
contains the following information: federal registration New York License. New York CLS Public Health
number, physical location of the establishment, reporting § 4364
official’s information, the establishment’s function (e.g., recov- District of Columbia License. DC Code 7-1541.03 and CDCR
ery, packaging, processing, etc.), product information, and 22-301
proprietary names. Additionally, the hospital tissue bank’s
Illinois Registration. 20 ILCS 2310/2310-330 and
medical director may request copies of any observations 77 III. Adm. Code 470.30
made during FDA inspection (Form 483), including responses
and corrective action.16 Warning letters to an establishment Louisiana Authorization. 32 L.R. § 403 and LAC
are available through the Freedom of Information Act and § 48:I.2901 and 2903
may be found on the FDA website (www.fda.gov).16 Massachusetts Licensed, accredited, or approved
Some tissue vendors will become voluntarily accredited by Dept. of Public Health. ALM GL
by a recognized organization. This is not required but may Chapter 113, § 7
weigh in the medical director’s decision to qualify a vendor Mississippi Certification. Miss. Code Ann. § 41-39-15
for his or her tissue bank. This should also be spelled out
in the written guidelines. Organizations that accredit tissue Oklahoma Permit. Oklahoma Statutes § 2209.1 and
establishments are the American Association of Tissue Banks O.A.C. § 310:505-5-1
(AATB)5 and the Eye Bank Association of America (EBAA).9 Oregon Registration. SB 341
Similar to the FDA website, these organizations list their
6888_Ch22_476-496 29/10/18 5:38 PM Page 483

Chapter 22 Tissue Banking as a Part of the Transfusion Service 483

the same facility on the same patient, despite the intervening record on file for each autologous tissue. Any deviations,
storage in the tissue bank.1 test results, adverse reactions, and the disposition should
Autologous tissue can be logistically problematic for be documented and retained in this file.
a hospital tissue bank. First, qualifying the supplier of the Within the operating room, the preparation of the autol-
tissue is changed in this circumstance. If a tissue is to be used ogous tissue falls under the regulatory standards of the Joint
within the same institution and not shipped to an off-site Commission, the AATB, and the Association of periOperative
facility under different management for any further process- Registered Nurses (AORN). AORN has developed guidelines
ing or storage, there is no requirement to register as a tissue and recommendations for autologous tissue recovery that
establishment with the FDA. It is important to note that if maintain aseptic techniques and prevent contamination.21
an autologous tissue is transferred to any other facility, such The recommended guidelines include the following:
as a different hospital because the patient was transferred,
1. Removal of tissue. The surgeon removes the tissue and
the hospital tissue bank would be required to register with
hands it off to an assistant in an aseptic manner. The
the FDA as a tissue-manufacturing establishment and would
assistant places the tissue in a sterile container. More than
be subject to FDA inspection.16 In order to remain exempt
one piece of tissue may be placed in the container if re-
from the requirement to register with the FDA, the hospital
moved from the same site at the same time. Otherwise,
tissue bank must retain the tissue until re-implantation at
separate containers should be used.
the same facility.
2. Culturing of tissue. If the procedure requires tissue
Common autologous tissues include skull bone flaps
culture, complete written instructions should be avail-
(most common), ribs, iliac crests, and endocrine tissue such
able. Culturing should include swabbing the entire tissue
as the pancreas and parathyroid.18 Occasionally, other tissues
surface, including edges. Culture swabs are labeled with
such as skin and connective tissue will require storage. The
at least two unique patient identifiers and sent to the
advantage of autologous tissue is that it affords a perfect
microbiology laboratory. Note: Culture should be done
immunological match. Additionally, the size may be ideal,
before any antibiotic is used as additive.
such as is the case for the skull bone flap that is exactly the
3. Addition of storage solution. If the tissue requires addi-
right size for the hole that must be patched. The risk of infec-
tion of storage media or antibiotics, it should be added
tious disease transmission is also diminished. However, one
after the culture step. Written protocols for each tissue
of the main disadvantages to autologous tissue is the possibil-
type must clearly establish which tissues require additives
ity of reinfection of bacteria or fungus or the reintroduction
and which additives are required. Because of the possi-
of malignant cells.19 The age and health of the donor-patient
bility of allergic reactions, hospital personnel must confirm
may impact the functionality of the tissue as well.
that the patient does not have an allergy to an additive
antibiotic. The container must be labeled with the name
Preparation of the antibiotic used.
4. Containers for storage. Containers may vary depending
The first step in the preparation for harvesting autologous
on the tissue and the institution. AORN recommends
tissue is to define and develop clearly written policies and
sterile plastic bags or sterile Nalgene jars. The Nalgene
procedures for both the surgical service and the hospital
jars are useful because they are able to withstand temper-
tissue bank.18 These policies and standard operating proce-
atures to –40°C for up to 5 years and can be used with
dures require collaboration between the two services in order
irradiation sterilization procedures.21
to create a seamless operation and to ensure full agreement
5. Wrapping the container. The sterile container is further
on all conditions that must apply. The hospital tissue com-
protected with at least two sterile outer wraps.
mittee may be involved in this process.
6. Labeling the container. The outer wrap of the container
The surgical service should include, as part of its proce-
must be labeled with two unique patient identifiers, patient
dures, a notification step to alert the tissue bank of a planned
age, date of collection, type of tissue, name and address of
procedure that will result in autologous tissue. Clear guide-
the facility, physician’s name, time of collection, and expi-
lines should be incorporated to instruct operating room per-
ration date, if applicable. It may require further biohazard
sonnel on the steps required both administratively and
labels according to FDA CFR 1271.90 and must contain a
medically. Informed consent must be obtained from the
label stating “FOR AUTOLOGOUS USE ONLY” and “NOT
donor-patient. This is best done by the surgeon treating the
EVALUATED FOR INFECTIOUS SUBSTANCES.”16
patient.
7. Tissue storage. The tissue must be placed immediately
The tissue bank should include a procedure to ascertain
into a temperature-monitored storage unit. If there is a
whether the donor-patient is being treated for bacteremia
delay, the tissue should be transported on wet ice.
or is at high risk for HIV or hepatitis.20 In general, it is not
8. Tissue processing. Tissue that is to be processed is done
advisable to use autologous tissue for re-implant if the patient
in a controlled environment.
was bacteremic at the time of harvest because of the risk of
reinfection. However, this is not true in every case, and pro- The Joint Commission also adds a requirement to “track
cedures for sterilization exist, so each case must be approved and identify materials used to prepare or process tissues
individually by the tissue bank medical director and the and instructions used for preparation”1 (Joint Commission
surgeon jointly. The tissue bank should keep a donor-patient Standard TS.03.02.01).1 Careful documentation of materials,
6888_Ch22_476-496 29/10/18 5:38 PM Page 484

484 PART III Transfusion Practices

including name, lot number, expiration date, and amount Discarding of autologous tissue should require the medical
should be made in the medical record. director’s approval. As with blood products and allogeneic
tissue, proper documentation of discard is required. Docu-
Storage mentation must clearly outline the manner and location of
disposal according to local, state, and federal regulations for
Ideally, the tissue bank will be expecting the arrival of the medical waste.16 Occasionally, a request will be made that the
autologous tissue, having been informed by the surgical staff tissue be sent to a funeral home for burial with the deceased
of the procedure ahead of time. Prior to tissue recovery, the donor-patient. This is permissible as long as transportation
surgical team and the tissue bank should confer on the pro- regulations are followed and documentation is complete.
cedure and verify that written standard operating procedures
address any individual issues that may be present for each
Sterilization and Testing
donor-patient.1 Once the tissue arrives at the tissue bank,
the tissue bank personnel must inspect the label to make Sterilization is not required and not possible with many
sure all elements are present (Box 22–2). The surgical team tissues. A medical director, in conjunction with the surgeon,
should immediately be involved to solve any discrepancies may choose a sterilization technique. For skull bone flaps, a
or unclear labeling. common and popular method of sterilization has been the
As per written standard operating procedure and recom- use of high heat with an autoclave. This has the advantage
mendation on the label, the tissue is placed in the proper of killing bacteria and even removing malignant cells; how-
storage site to ensure that the correct temperature is main- ever, the disadvantage is that the high heat denatures pro-
tained. A separate area should be reserved for autologous teins and collagen in the bone, making it less suitable for
tissues to keep them away from allogeneic tissue, as is done re-implantation. Steam has also been utilized but has the
with autologous blood. Tissue from donor-patients who are same denaturing effect. Many surgeons will opt for irradia-
known to have infectious diseases should be stored in a tion as a method to destroy bacteria. This is useful if there is
quarantine area. an irradiator on-site at the hospital and clear guidelines are
A problem common to tissue banks that store autologous worked out for exposure times and radiation amount. Note:
tissues is what to do with tissues that never get used. It is If the tissue is taken to another facility under different man-
fairly common for surgeons to decide not to use autologous agement for sterilization techniques, the hospital tissue bank
tissue after it has been harvested. This can be solved with must register as a tissue-manufacturing establishment with
good communication with the surgical staff and agreement the FDA and become subject to FDA inspection.15
of a reasonable expiration date for all tissues. If agreement Infectious disease testing is optional for autologous tissue,
cannot be reached, then an expiration date may be assigned as it is for autologous blood. Culture of autologous tissue is
by the tissue bank. Care should be taken to ensure the con- also optional and should be clearly spelled out in the oper-
tainer is suitable for the temperature and duration of storage. ating procedures as to what tissues, if any, require culture. If
The surgeon should be informed of the expiration date at the the facility opts to culture or test the tissue donor-patient for
time it is assigned. According to AATB standards, autologous infectious diseases, the same testing should be performed as
tissues should have the same shelf life as a similar allogeneic for blood donors (HBsAg, anti-HBc, anti-HCV, HCV NAT,
tissue under the same storage conditions.5 anti-HIV-1/2, HIV NAT, anti–human T-cell lymphotropic
virus (anti-HTLV) I/II, syphilis, and West Nile virus NAT).22
If any testing or culture is positive, the tissue should be re-
leased only with the medical director’s approval. The surgical
BOX 22–2 team should be notified of any positive tests, and the medical
director may approve or disapprove use based on consulta-
Requirements for Autologous Tissue Labels
tion with the transplanting surgeon. If any test is positive
• Must read “For Autologous Use Only” or if donor-patient screening indicates an increased risk for
• Must include name and address of the tissue bank infectious disease, a biohazard label is mandatory for the tis-
• Must include two unique patient identifiers sue container. If the facility opts not to test the autologous
• Must include complete description of tissue tissue, a label stating “NOT EVALUATED FOR INFECTIOUS
• Must indicate date of collection SUBSTANCES” must be affixed to the container.16
• Must include name of surgeon
• Must indicate date of expiration according to validation of packag-
ing and tissue type Implantation
• Must indicate disinfection or sterilization steps (if performed)
Issuing or dispensing autologous tissue is much the same as
• Must indicate presence and name of any additives
for allogeneic tissue. There are a few differences, however,
• Must include biohazard label if testing is positive or screening is
and the tissue bank that is located within the blood bank
positive for risk factors
can utilize familiarity with autologous blood products to con-
• Must indicate “NOT EVALUATED FOR INFECTIOUS SUBSTANCES”
unless complete donor testing has been done struct standard operating procedures that cover all of the nec-
• Must be stored at recommended storage temperatures essary requirements. As with autologous blood, the blood
bank and tissue bank personnel should be made aware of the
6888_Ch22_476-496 29/10/18 5:38 PM Page 485

Chapter 22 Tissue Banking as a Part of the Transfusion Service 485

expected surgical date for re-implantation. Prior to the sur- and regulations for each of the major organizations listed
gery date, the tissue bank must perform a thorough record here, it is recommended that a full review of each regulation
review, and all deviations and discrepancies must be resolved is done prior to developing a hospital tissue bank structure
before the tissue is needed for surgery. Suitability should be or standard operating procedures (SOP) designed for com-
documented in the donor-patient file. The issuing procedure pliance. Most agencies revise and update their standards and
should be the same as for allogeneic tissue and must include regulations as often as every 1 to 4 years. Attention should
a final visual inspection of the outer wrapping for integrity be paid to changes as they occur.
and inspection of the label to verify all required elements
are present. If any problems or deviations are discovered, the Federal Laws
tissue should be placed into quarantine until the medical
director resolves the matter in consultation with the trans- The Code of Federal Regulations, Title 21, was established
planting surgeon. for food and drugs, and part 1271 deals with human cells,
tissues, and cellular and tissue-based products (HCT/Ps).
Regulatory Overview Part 1271 is subdivided into subparts A through F:16
• A: General provisions
The number of organizations involved in tissue regulations • B: Procedures for registration and listing
and standards can be daunting to a facility intending to set up • C: Donor eligibility
a hospital tissue bank. It can be overwhelming to look at the • D: Current good tissue practices
various documents to be sure that all requirements are carried • E and F: Additional requirements, inspection, and enforce-
out and satisfied. Table 22–3 is a “crosswalk” that compares ment of establishments covered in 1271.10.
each of the major regulatory agencies, matching the categories
of regulations for easy cross-reference and comparison. Section 361 of the PHS Act refers specifically to regulations
In taking a broad overview of the regulatory agencies, it is to control communicable diseases (42 USC, section 264).23
clear that they have evolved in parallel. One may require more
than the others in one area but less in another, depending General Provisions
upon the focus of the organization. However, they overlap The purpose of part 1271 was to create a system to register and
significantly, and it is not difficult to develop an organization list establishments that manufacture HCT/Ps.16 Additionally,
that is in compliance with them all. It is important to note it is meant to establish guidelines for donor eligibility and
that compliance with AABB Standards often exceeds require- incorporate principles of good tissue practice in order to pre-
ments from other agencies, another important reason to vent the introduction, transmission, and spread of communi-
house the tissue bank within the blood bank.13 While the cable diseases through tissue transplant. An establishment
author has attempted to summarize the relevant standards is defined as a business, which may include any individual,

Table 22–3 “Crosswalk” for HCT/P-Related Standards of the AABB, AATB, EBAA,
and the Joint Commission
AABB* AATB † EBAA‡ Joint Commission§
1.0 Organization L1.000 C1.000–C1.300 TS.03.01.01
L1.100 C2.000 EP1
B2.120
B2.121

1.3 Policies, Processes, and Procedures B2.140 C3.400 TS.03.01.01


D5.000 D1.000 EPs 1–10
E1.000 E1.000
F1.000 F1.000
J1.000 G1.000
L1.000 I.1000
J.1000
K1.000
L1.000
L2.000
M1.000
Continued
6888_Ch22_476-496 29/10/18 5:38 PM Page 486

486 PART III Transfusion Practices

Table 22–3 “Crosswalk” for HCT/P-Related Standards of the AABB, AATB, EBAA,
and the Joint Commission—cont’d
AABB* AATB † EBAA‡ Joint Commission§
1.4 Emergency Preparedness E3.340 C3.200 TS.03.01.01
J1.200 EP 10
3.0 Equipment J5.000 C3.200 TS.03.01.01
3.6 Storage Devices for Blood, Components, Reagents, D6.300 I1.000 EPs 8–10
Tissue and Derivatives
E3.100
3.7 Alarm Systems
E3.330
J5.600

4.0 Supplier and Customer Issues F1.200 C3.500 TS.03.01.01


C3.510 EPs 2, 3,5, 6, 7
4.1 Supplier Qualification B1.500 G1.100
J1.000
4.3 Incoming Receipt, Inspection, and Testing E1.000
5.0 Process Control C1.100 G1.000 TS.03.02.01
5.1.6 Identification and Traceability E1.700 G1.100 EPs 1–4, 7
5.1.6.1 Process or Procedure Steps E2.200 E1.000–E1.300
5.1.6.2 Traceability J1.200 D1.200
5.1.8 Handling, Storage, and Transportation K1.100 J1.000
L3.100 I1.000
L1.100
L2.000
K1.000–K1.200
M1.500
5.10 Final Inspection F1.000
Transfusion Service Related Activities F4.000
5.11 Samples and Requests H1.000
5.18 Final Inspection Before Issue
5.20 Tissue Preparation J1.200
6.0 Documents and Records C1.000 D1.100–D1.120 TS.03.02.01
Reference Standard 6.2D C3.000 K1.100 EPs 1–6
Retention of Tissue Records M1.00-M1.500
C3.400
10.0 Facilities and Safety J3.000 C3.600 TS.03.03.01
C3.700 EPs 1–5
10.3 Discard of Blood, Components, Tissue, and Derivatives L3.300

Horizontal alignment represents only general relationships among standards. This table represents only selected highlights of AABB HCT/OP-related standards. See Chapters 9 through
12 for more detail on the above standards and organizations; see Frizzo14 for a fuller treatment of all relevant AABB standards and those of the AATB, the College of American
Pathologists, and the Joint Commission.
*American Association of Blood Banks. Standards for blood banks and transfusion services. 30th ed. Bethesda (MD): AABB; 2016.
†Maye T, Malone P, Brubaker S, editors. Standards for tissue banking, 14th ed. McLean (VA): American Association of Tissue Banks; 2016.
‡Eye Bank Association of America. Medical standards. Washington, (DC): EBAA; 2015.
§The Joint Commission, Comprehensive accreditation manual for hospitals. TS.03.01.01–TS.03.03.01. Oakbrook Terrace (IL): Joint Commission; 2017.

AABB = American Association of Blood Banks; AATB = American Association of Tissue Banks; EBAA = Eye Bank Association of America; HCT/P = human cells, tissues, and cellular and
tissue-based products
Updated with permission from Eisenbrey, AB, and Eastlund, T (eds): Hospital Tissue Management: A Practitioner’s Handbook, 1st ed. AABB, Bethesda, 2008.
6888_Ch22_476-496 29/10/18 5:38 PM Page 487

Chapter 22 Tissue Banking as a Part of the Transfusion Service 487

partnership, corporation, or association, under single manage- HCT/Ps to another facility for use on a patient. It does not
ment, in a single location that engages in the manufacture include the return of tissue to a supplier.
of HCT/Ps as well as facilities that contract manufacturing
services. Procedures for Registration and Listing
Human cells, tissues, or cellular tissue–based products are Establishments that meet the requirements for registration
defined as articles containing or consisting of human cells with the FDA must do so within 5 days after beginning oper-
or tissues intended for transplantation or otherwise trans- ations using Form FDA 3356.15 Registrations must be updated
ferred into a human recipient. Some examples include bone, annually in December in conjunction with an update of the
ligament, skin, hematopoietic stem cells derived from periph- HCT/P list, unless there is no change. If a change in the HCT/P
eral and cord blood, manipulated autologous cells, epithelial list occurs between December and June, the change must be
cells on a synthetic matrix, semen, and other reproductive updated in June.15
tissue.7 This list is by no means exhaustive and represents a In addition to the annual renewal and notifications of
small number of examples of HCT/Ps. change in HCT/P list within 6 months, any change in loca-
HCT/Ps that are regulated under section 361 of the PHS tion or ownership of the establishment must be amended on
Act meet the following criteria: The tissue is minimally ma- the registration within 5 days of the change.15 Upon accept-
nipulated; it is intended for and labeled for homologous use ance of the establishment’s registration form, the FDA will
only; it is not combined with another vehicle except for assign each location a permanent number.15 This registration
water, crystalloids, and sterilizing and preserving agents, number does not imply that the establishment is in compli-
unless such additives compromise the clinical safety of the ance with applicable rules and regulations or is approved by
HCT/P; and the HCT/P does not have a systemic effect and the FDA. However, the establishment, once registered, must
is not dependent upon the metabolic activity of living cells be available for public inspection by the FDA.16
for its primary function.6 Alternatively, the HCT/P may have
a systemic effect or may depend on the metabolic activity of Donor Eligibility
living cells for primary function if it is for autologous use, Establishments that manufacture HCT/Ps must develop and
allogeneic use in a first- or second-degree blood relative, or maintain procedures for all steps in testing, screening, donor
reproductive use. eligibility, and all other requirements.16 The procedures are
Tissues that are not considered HCT/Ps include vascular- subject to review and approval of a responsible individual.
ized human organs for transplantation; whole blood or blood Donor eligibility is determined by screening relevant medical
components or blood derivative products; secreted or ex- records to ensure that the donor is free from risk factors and
tracted human products (except for semen), such as milk, col- clinical evidence of relevant communicable diseases, that there
lagen, and cell factors; minimally manipulated bone marrow are no communicable disease risks associated with xenotrans-
for homologous use and not combined with another article, plantation, and that results of donor testing for relevant disease
except for additives for preservation and storage; ancillary agents are negative or nonreactive. Relevant communicable dis-
products used in the manufacture of HCT/P; cells, tissue, and eases include HIV, types 1 and 2; hepatitis B virus; hepatitis C
organs derived from animals; in vitro diagnostic products; and virus; human transmissible spongiform encephalopathy;
blood vessels recovered with an organ intended for transplant Creutzfeldt-Jakob disease; and Treponema pallidum.24 For
and labeled “For use in organ transplant only.”5 Although leukocyte-rich tissue, there should be additional testing for
these items are not HCT/Ps, they may be found as part of a human T-cell lymphotropic virus, types 1 and 2. Cells and
hospital tissue bank at the discretion of the hospital adminis- tissue that are reproductive in nature must also be tested for
tration and for the sake of uniformity in handling biological Chlamydia trachomatis and Neisseria gonorrhea.24
items for human transplant, transfer, or infusion. Relevant medical records that must be reviewed include
There are a few exceptions to who must comply with the a current donor medical history interview; a current report
requirements of part 1271. HCT/Ps used solely for nonclin- of the physical assessment of a cadaveric donor or a physical
ical or educational purposes are exempt.16 Entities that examination of a living donor, if available; laboratory test
merely receive, carry, and deliver HCT/Ps and entities that results in addition to those outlined above; medical records;
do not recover, screen, test, process, label, package, or distrib- coroner or autopsy reports; and records from any source
ute but only receive and store tissues for use on patients only pertaining to risk factors for communicable diseases.
in that facility are also exempt.16 The latter type of institution Once the donor is deemed eligible, records pertaining to
is the usual model for hospital tissue banks. Thus, the require- the donor eligibility must accompany the tissue at all times.
ments set forth in 21 CFR 1271 are not regulations that hos- The required documentation includes a distinct identifica-
pital tissue banks that only store and issue HCT/Ps must tion code that links the HCT/P to the donor. Except in the
meet. However, it is important that the tissue banker be cases of autologous tissues or HCT/Ps donated directly to a
aware of the requirements of its suppliers, how the tissues first- or second-degree relative, the identification code
are handled, and what tests are routinely run. Hospital tissue should not include the donor’s name, social security number,
banks that participate in harvesting, processing, packaging, or medical record number. A statement that the donor has
or distribution of tissues must comply with the Code of been determined “eligible” or “ineligible,” along with a
Federal Regulations (21 CFR 1271) and become registered summary of the records used to make this determination, is
establishments.15 Distribution is defined as the transfer of required. In addition, a statement that disease testing was
6888_Ch22_476-496 29/10/18 5:38 PM Page 488

488 PART III Transfusion Practices

performed by a Clinical Laboratory Improvements Amend- tissue banks be licensed as clinical laboratories. New York
ments (CLIA)-certified laboratory or equivalent, a list of tests and Florida have adopted extensive technical regulations.
performed with results, and the name and address of the Other states require permits, certification, or authorization.
establishment determining eligibility are required. If readers reside in any of these 13 states, it is recommended
Donor eligibility determination is not required in some they study the laws of that state to ensure compliance with
circumstances. Examples of this include cells and tissues for those laws. The Joint Commission requires that tissue banks
autologous use, reproductive cells or tissue donated by a in states that have additional laws pertaining to tissue must
sexually intimate partner of the recipient, or cryopreserved be in compliance.1
cells for reproductive use (when possible, measures should
be taken to test the semen and oocyte donors before transfer American Association of Blood Banks
of an embryo). In these cases, the tissue must be clearly
labeled “FOR AUTOLOGOUS USE ONLY” or, for allogeneic The AABB has been involved in setting standards for tissue
tissue, “NOT EVALUATED FOR INFECTIOUS SUBSTANCES” banking for almost two decades. In 1993, in its 15th edition
and “WARNING: Advise recipient of communicable disease of the Standards for Blood Banks and Transfusion Services
risks.”16 (Standards), a section was added to address tissue storage
and issue. Since 1947, it has been involved in blood bank-
Current Good Manufacturing Practice ing, another form of human tissue transplant, which makes
Subpart D sets forth current good tissue practice (CGTP) AABB-accredited hospital blood banks the recognized
requirements.16 These requirements are intended to prevent and logical leaders in the field of tissue banking. In the
the introduction, transmission, or spread of communicable 1990s, the AABB revised its quality plan to reflect what the
diseases by HCT/Ps by ensuring that they do not contain organization considers to be its ten quality system essen-
disease agents, are not contaminated, and do not become tials (QSEs):
contaminated during manufacturing. The CGTP requirements 1. Organization
govern all steps in the manufacture of HCT/Ps, specifically 2. Resources
donor eligibility, screening, and testing. Core CGTP require- 3. Equipment
ments cover facility control, environment control, equipment, 4. Supplier and customer issues
supplies and reagents, recovery, processing, labeling, storage, 5. Process control
receipt, distribution, and donor eligibility.16 Contracts 6. Documents and records
between tissue suppliers are addressed here. Establishments 7. Deviations, nonconformances, and adverse events
that recover and process tissue must be in compliance with 8. Internal and external assessments
subparts C and D as they apply to the operations performed 9. Process improvement through corrective and preventa-
at that establishment.16 If another establishment performs tive action
some of the operations, such as sterilization, that establish- 10. Facilities and safety
ment is also required to conform to the regulations for the
operations performed at that facility. Before entering into a Human tissues for transplantation are treated in a manner
contract, the primary establishment is responsible for veri- almost identical to blood by the AABB Standards.13 In gen-
fying that the second establishment is in compliance. If at eral, hospital tissue banks that operate within hospital blood
any time the primary establishment becomes aware of non- banks can assume compliance because the same standards
compliance at the secondary establishment, the contract or apply to blood and tissue equally, unless a standard explicitly
agreement must be terminated. excludes tissue. Table 22–4 lists AABB standards that specif-
Also part of CGTP is the establishment and maintenance ically mention tissue.
of a quality program. Functions of the quality program must
include having appropriate procedures relating to core CGTP American Association of Tissue Banks
requirements and procedures for receiving, evaluating, and
The AATB was founded in 1976 by a group of scientists and
documenting core CGTP requirements; ensuring that appro-
doctors from the U.S. Navy Tissue Bank.4 These individuals
priate corrective actions relating to core CGTP requirements
recognized that the scope of tissue banking should be widened
are done, including reaudits of deficiencies and verification
to include the entire nation. It is the only national tissue bank
that corrective actions are effective; ensuring proper training
organization and accredits over 100 tissue suppliers nation-
and education of personnel; establishing appropriate moni-
wide. AATB’s Standards was first published in 1984. The Stan-
toring systems; and investigating and documenting devia-
dards, now in its 14th edition, is a comprehensive and detailed
tions and trends.16
guide to tissue banking.5 This guide has been used interna-
State Laws tionally as a framework for other national tissue banking
organizations. At least six states (including California, Georgia,
Only a few states have enacted laws regarding the transplant and Maryland) require that tissue-manufacturing establish-
and manufacture of human tissue products (see Table 22–2). ments be accredited by the AATB. In 1988, the organization
Only three of these states (New York, District of Columbia, initiated a certification program, Certified Tissue Banking
and California) have comprehensive laws regarding licensing Specialist (CTBS), for individual workers in the tissue banking
of tissue banks. Georgia and Maryland require only that industry. It currently certifies over 4,000 people.
6888_Ch22_476-496 29/10/18 5:38 PM Page 489

Chapter 22 Tissue Banking as a Part of the Transfusion Service 489

Table 22–4 AABB Standards Pertaining to Tissue


Standard Purpose
1.0 Organization Clearly defined structure

1.3 Policies, Processes, and Procedures Quality SOPs to ensure the requirements of the Standards are satisfied

1.4 Emergency Preparedness SOPs for internal or external emergencies

3.0 Equipment Calibration, maintenance, and monitoring of critical equipment

3.6 Storage Devices Capacity, temperature monitoring

3.7 Alarm Systems Set to alarm before tissue is compromised, investigate

4.0 Supplier and Customer Issues Supplier qualification, testing in AABB or equivalent accredited laboratory, registered
with FDA

4.3 Incoming Receipt, Inspection, and Testing Inspection, testing as necessary, label verification

5.0 Process Control SOPs to ensure quality of tissue, derivatives, and services

5.1 Identification, Traceability, and Inspection Documentation of each step and identification of person who performed it, traceability,
no more than two donation identifications per container, handling, storage, and
transportation

5.11.1 Requests Requests from authorized health professionals for tissue and derivatives with sufficient
information to uniquely identify the patient

5.20 Preparation of Tissue Procedures to ensure that preparation steps are performed before dispensing tissue in
accordance with the source facility’s SOPs

5.21 Preparation of Derivatives Procedures to ensure that preparation steps are performed before dispensing deriva-
tives in accordance with the source facility’s SOPs

5.22 Final Inspection Before Use Inspection of tissue and derivatives at the time of use

5.24 Issue of Tissue and Derivatives Verification of documents, quantity, inspection, intended recipient, expiration, and
date/time of issue

5.26 Reissue of Tissue and Derivatives Conditions that must be met to accept tissues or derivatives into inventory after return
to the tissue bank

5.29 Medical Record Documentation Documentation of type of tissue/derivative, identifier, quantity, expiration date, date of
use, and adverse events

6.0 Documents and Records SOPs for documents to be identified, reviewed, approved, and archived

6.2.3 Record system to trace any tissue from source to final disposition

6.2D Retention of Tissue Records Minimum retention time of tissue records

6.2E Retention of Derivative Records Minimum retention time of derivative records

7.0 Deviations, Nonconformances, and Adverse Events SOPs to capture, assess, investigate, and monitor deviations, responsibility for review,
prevention of issue, quarantine, and notification

7.1.4 Released Nonconforming Tissue Determination of effect, notification of customer if necessary, records created

7.2 Fatality Reporting Fatalities related to blood reported to outside agencies as required

7.4.4 Adverse Events Related to Tissue SOP for investigating and reporting adverse events

7.4.5.1 Transmissible Diseases Investigation of events by the medical director and reporting of implicated tissue or
derivative to the source facility

9.1 Corrective Action Process for corrective action of deviations, nonconformances, and complaints

10.0 Facilities and Safety Safe environmental conditions

10.3 Discarded Tissue Minimize potential for human exposure to infectious agents

(Data from: Standards for Blood Banks and Transfusion Services, 30th ed. AABB, Bethesda, 2016.)
6888_Ch22_476-496 29/10/18 5:38 PM Page 490

490 PART III Transfusion Practices

In the 1990s, the AATB expanded its scope from tissue- evaluating health care organizations and inspiring them to
manufacturing establishments to include hospital tissue banks. excel in providing safe and effective care of the highest qual-
This era also saw the beginnings of a partnership and collabo- ity and value.”25 Its history reaches back to 1910, when
ration between the AABB and the AATB. In 1993, a new section Ernest Codman, MD, proposed the “end result system of
appeared in AATB’s Standards entitled “Medical Facility Tissue hospital standardization.”26 He felt that if every hospitalized
Storage and Issuance.” Subsequently, several more sections patient could be tracked long enough to determine effective-
that apply to the hospital tissue bank were added, including ness of treatment, medicine would benefit. The benefit
“Records Management,” “Release and Transfer of Tissues,” would come primarily in determining for whom treatment
“General Operations,” and “Quality Assurance and Quality was not effective, determining why, and changing practices
Control Programs.” Following AABB’s change to the 10 QSEs, for future patients. In 1917, a one-page list of minimum stan-
the AATB Standards were reorganized in the same format. dards for hospitals was published, and the following year,
In widening its scope to include the hospital tissue bank, the organization began inspections based on these minimum
the AATB’s intention was to provide greater safety and trace- standards.26 Of the 692 hospitals surveyed that year, only 89
ability to final disposition, whether implanted into a patient or met the minimum standards.26
disposed. By 1950, the original document grew significantly, and there
The section in the AATB Standards that is most applicable were as many as 3,200 accredited hospitals.26 In 1951, the
to the hospital tissue bank is the L1.000 series for tissue American College of Physicians (ACP), the American Hospital
dispensing services.5 Standards that apply to autologous Association (AHA), the American Medical Association (AMA),
tissue can be found throughout. See Table 22–5 for a list of and the Canadian Medical Association (CMA) joined with the
standards relevant to the hospital tissue bank. A comparison original American College of Surgeons (ACS) to create the Joint
of Tables 22–4 and 22–5 shows the similarity between the Commission on Accreditation of Hospitals (JCAH).26
Standards for Blood Banks and Tissue Services and the Standards In 1965, Congress passed the Social Security Amendments
for Tissue Banking as they each pertain to tissue banking. with the provision that hospitals accredited by JCAH had
deemed status with the Medicare Conditions of Participation
The Joint Commission for Hospitals and could, therefore, participate in Medicare
and Medicaid programs.26 In 1978, JCAH joined forces with
The Joint Commission is a nonprofit accrediting agency that the College of American Pathologists (CAP) to further eval-
accredits and certifies over 17,000 health care organizations in uate laboratories. JCAH adopted the name Joint Commission
the United States. Its mission is “to continuously improve health on Accreditation of Healthcare Organizations (JCAHO) in
care for the public, in collaboration with other stakeholders, by 1987 to reflect its expanding scope of activities.26

Table 22–5 AATB Standards Pertaining to Hospital Tissue Banks


Standard Purpose
L1.000 General, Director SOPs addressing receipt, storage, final disposition, traceability, supervision by a qualified medical professional
L2.000 Storage Storage in conformance to guidelines
L2.200 Equipment Maintenance, calibration, QC
L2.300 Labeling Tissue shall not be relabeled; labels shall not be altered
L3.100 Dispensing Physician’s order required, all associated written material is to be available to the end user
L3.200 Release to Intermediary All original written materials, label, and tissue identification number must accompany documentation of transfer
to another facility
L3.300 Tissue Disposal Minimize hazard to staff or environment, comply with applicable laws and regulations
L3.400 Return of Tissue Cryopreserved reproductive tissue may not be redistributed for use, except as required by local or state regulations
L4.000 Records Concurrently record all steps for traceability, maintain records for 10 years after expiration
L4.100 Tissue Receipt Records Each specimen must have a unique tissue identification number, minimum requirements for receipt records
L4.2 Dispensing Records Minimum requirements for disposition records, maintained in log format
L5.000 Adverse Outcomes Potential or suspect adverse outcomes, directly or indirectly related to tissue reported to tissue processor,
investigated, and documented
L6.000 Recalls SOPs for the recall of tissue

(Data from: Maye, T, Malone, P, and Brubaker, S (eds): Standards for Tissue Banking, 14th ed. American Association of Tissue Banks, McLean, VA, 2016.)
6888_Ch22_476-496 29/10/18 5:38 PM Page 491

Chapter 22 Tissue Banking as a Part of the Transfusion Service 491

Effective July 1, 2005, JCAHO adopted the tissue storage disposition, or expiration of tissue (whichever is
and issuance standards for the ambulatory care, office- latest).
based surgery, critical access hospitals, and hospital accred- EP 7: Compliance with completion and return of tissue usage
itation programs that store and/or issue human tissue. information cards requested by the source facility must
These were in the form of standards PC.17.10, PC.17.20, occur.
and PC17.30 (now renamed: TS.03.01.01, TS.03.02.01, and
Hospital Standard TS.03.03.01 concerns the process of
TS.03.03.01). In 2007, JCAHO officially shortened its name
investigating adverse reactions to tissue or donor infections.
to “Joint Commission.”26
This standard is in five parts:
Hospital Standard TS.03.01.01 addresses the use of stan-
dardized procedures to receive, store, and issue tissues. This EP 1: Have SOPs in place to investigate recipient adverse
standard is broken down into eleven parts, or elements of events related to tissue use.
performance (EP):26 EP 2: Investigates tissue adverse events, including disease
transmission or other complications.
EP 1: The organization assigns responsibility for overseeing
EP 3: Reports the infection or adverse event to the tissue
the tissue program throughout the hospital.
supplier.
EP 2: The organization develops and maintains standardized
EP 4: Sequesters tissue whose integrity may have been
written procedures for the acquisition, receipt, storage,
compromised.
and issuance of tissues.
EP 5: Identifies and informs tissue recipients of infection risk
EP 3: The organization confirms that tissue suppliers are reg-
when donors are subsequently found to have infectious
istered with the U.S. Food and Drug Administration
agents known to be transmitted through tissue.
(FDA) as a tissue establishment and maintain a state
license when required. A review of the tissue standards from the Joint Commis-
EP 4: The organization coordinates its acquisition, receipt, stor- sion reveals striking similarity to regulations from AABB,
age, and issuance of tissues throughout the organization. AATB, and FDA. The most notable addition is the require-
EP 5: The organization follows the tissue suppliers’ or man- ment for source facilities to comply with applicable state laws
ufacturers’ written directions for transporting, handling, for registration and accreditation.
storing, and using tissue.
EP 6: The organization documents the receipt of all tissues. Eye Bank Association of America (EBAA)
EP 7: The organization verifies that package integrity is met
The EBAA accredits facilities that provide eye tissue for
and transport temperature range was controlled.
surgery, education, or research.9 It is a nonprofit organization
EP 8: The organization maintains daily records to demon-
that also certifies eye bank technicians. The organization
strate that tissues requiring a controlled environment are
was established in 1961 by the American Academy of Oph-
stored at the required temperatures.
thalmology’s Committee on Eye Banks.9 It is the oldest national
EP 9: The organization continuously monitors the tempera-
tissue transplant organization in the United States, and its
ture of refrigerators, freezers, nitrogen tanks, and other
mission is to “restore sight through the promotion and ad-
storage equipment used to store tissues.
vancement of eye banking.” In addition to accreditation and
EP 10: Storage equipment used to store tissues at a controlled
certification, EBAA also develops uniform medical standards
temperature will have functional alarms and an emergency
and procedures for its member banks to maintain proficiency
backup plan.
in eye tissue recovery, storage, preservation, and transplan-
EP 11: The organization complies with state and/or federal
tation.9 These medical standards are reviewed and approved
regulations when it acts as a tissue supplier.
semiannually by the American Academy of Ophthalmology.
Hospital Standard TS.03.02.01 is concerned with record- The EBAA requires facilities providing tissue to be regis-
keeping, particularly in the bidirectional traceability from tered with the FDA.9 The source facility may voluntarily be
donor to final disposition. There are seven parts to this accredited by EBAA; however, the Joint Commission does
standard: not require this as a condition for hospital tissue banks to
receive tissue from an establishment. The hospital tissue
EP 1: Records must permit tracing of any tissue.
bank medical director may select suppliers of ocular tissue
EP 2: Records must track and identify material used to pre-
from EBAA-accredited facilities as an added level of safety
pare or process tissue.
and security.9
EP 3: Records must identify who accepted the tissue, who
prepared the tissue, and the date and time of preparation. Adverse Events
EP 4: Documentation must be included in the recipient’s
clinical record of tissue use, including unique identifier An adverse event is a negative change, unexpected outcome,
of tissue. or reaction to the tissue implant. Adverse events can be due
EP 5: Records must be maintained for at least 10 years to infectious disease, failure of the allograft to function as
following expiration. expected, or transmission of malignancy. It is important to dis-
EP 6: Tissue records must be retained for a minimum of cover and report adverse events primarily for the protection of
10 years beyond the date of distribution, transplantation, other patients who may receive tissue from the same donor.2,7
6888_Ch22_476-496 29/10/18 5:38 PM Page 492

492 PART III Transfusion Practices

Infection invade neurological tissue and cause death. Incubation times


vary, and patients have shown symptoms as early as 6 months
Transmission of infectious disease through allograft trans- following transplant and as late as 16 years.
plantation is relatively rare. Many of the recorded cases of Another possibility for introduction of infectious agents
disease transmission occurred in years prior to the current, is through perimortem blood transfusions. The author is
comprehensive standards and improved donor testing. Infec- currently investigating several donors whose blood was
tions are most often transmitted by tissues that are fresh transfused to a trauma victim who died and subsequently
or cryopreserved. These methods of storage allow some became a tissue donor. A recipient has contracted par-
infectious entities to survive and cause disease in the new vovirus B19. Appropriate investigation of the tissue donor
host. Fortunately, the most common type of tissue used, has been unrevealing. Because blood is not routinely tested
freeze-dried bone, has not been seen to transmit disease read- for parvovirus B19, the donors of the units transfused at
ily. Often the biggest cause of disease transmission is human death are being investigated. Fortunately, transfusion-
error. Inadequate donor screening, inadequate sterilization, transmitted disease is rare.
and other system failures allow potentially contaminated
tissue to remain in the system. For these reasons, the above Malignancy
regulations have been developed.
Similar to blood donation, the first step in ensuring safe Malignancy transmitted through tissue transplant is ex-
tissue is to search the donor population for individuals tremely rare. Two cases of carcinoma transmitted through
with low risk of disease. This can be a challenging task for corneal transplant have been recorded.19 Almost all other
manufacturing facilities. Medical records may be incomplete, known cases of malignancy transmission have been through
and if family of a deceased donor is available for interview, organ transplant and include a number of adenocarcinomas,
they may not be aware of all the details that may be important. melanoma, sarcoma, lymphoma, glioblastoma, and choriocar-
It may also be unclear what caused the donor’s death. A cinoma.19 Tissues, such as bone, tendon, and ligament, have
misdiagnosis of a fatal infection could result in that infection few viable cells present at transplant and undergo several
passing on to a tissue recipient. This was the case for a rural sterilization steps. Heart valves and vascular tissue rarely har-
man who died with “chest numbness and back pain” in bor malignancies and are therefore less likely to transmit it.
1979, reported by Houff and colleagues.27 His corneas were Of note, there has never been a recorded incident of transmis-
transplanted into an individual who died 6 weeks later, fol- sion of malignancy through blood transfusion. Both tissue
lowing development of headaches, eye pain, and respiratory and blood donors are excluded from donation on the basis of
failure. Studies on both donor and recipient revealed a rabies malignancy known to be present at time of donation.
infection in both individuals. Had the clinical history of the
deceased been more accurate, the transplant and subsequent Allograft Failure
second death could have been avoided. Infections may lie The allograft may fail to function as expected or intended.
dormant for years. There may be only limited mention in In many cases, this may be because of the patient’s underly-
medical records, or none at all for a virus that is dormant ing condition. However, it is important to investigate other
with an undetectable antibody titer. Antibody titers may causes not related to the patient. Failure of the tissue manu-
also be unreliable and in the “window period” shortly after facturer to faithfully adhere to standard operating procedures
initial infection. For these reasons, RNA testing for HIV and can cause changes that may affect the stability and durability
hepatitis C has become standard practice. of the tissue product. Discovering the source of tissue failure,
Bacterial and fungal infections transmitted by tissue either intrinsic to the donor or because of an irregularity in
transplant are getting more attention in recent years due to manufacturing, will lead to recall of other products that may
increased awareness and regulatory oversight. It is likely have potential to harm other patients.
that many adverse events due to fungal and bacterial con-
tamination went undetected, usually with the assumption Reporting and Investigation
that the recipient acquired the infection de novo. Investiga-
tion into the source of these types of adverse events have Reporting and investigating suspected adverse outcomes is
shown that the organism was often not part of the pre- important for many reasons. This area of medicine has recently
mortem condition in the donor but was introduced during received a great deal of attention with efforts to create a
the surgical harvest of the tissue or the subsequent process- national biovigilance network. Biovigilance, as a structured,
ing. Many tissue products undergo minimal processing region-wide entity, has been in practice for many years in
without sterilization steps. These include heart valves, ten- Europe. The backbone of a quality program is the ability to
dons, ligaments, and cartilage. The tissue bank medical di- capture errors and unexpected outcomes and then investi-
rector should be certain to make transplanting surgeons gate them to determine, as much as possible, the root cause.
aware of bacterial and fungal risks. This information is of little value if it is not shared with
Dura mater has been known to transmit Creutzfeldt- everyone involved in the process so that meaningful change
Jakob disease (CJD) in more than 100 cases of transplant.24 can take place. In the individual case, it is crucial to alert
CJD is caused by a prion, which is neither virus, bacteria, the tissue supplier of a suspected disease transmission or
nor fungus but rather is a self-replicating protein known to contamination so that the supplier may track down all other
6888_Ch22_476-496 29/10/18 5:38 PM Page 493

Chapter 22 Tissue Banking as a Part of the Transfusion Service 493

tissue harvested from the same donor to prevent other adverse steps may be taken to quickly sequester any other tissue from
events. Often, tissue tracked from the same donor may have that donor that may exist at different facilities.
already been transplanted. In that case, it is equally impor- The medical director must then confirm the complaint
tant to know of possible contamination so that the patient and begin to determine what other causes may be responsi-
may be appropriately tested or treated. ble. He or she should look into the possibility of nosocomial
The most basic, and arguably the most important, step in infection, other potential routes of infection, and risk factors
the reporting process is recognizing an adverse event by the independent of tissue transplant and should examine patient
clinical staff. Postoperative infections are not uncommon, records for any irregularities associated with tissue transplant
and a true adverse reaction may be dismissed as a routine procedures. If warranted, corrective action should be imple-
postoperative complication. Some adverse reactions may mented depending on the result of the investigation. The
take weeks, months, or even years to express themselves, tissue bank must cooperate fully with the tissue supplier’s
such as hepatitis C infection or CJD. The medical director investigation.
of the tissue bank should communicate regularly with the Other ancillary investigation should include testing of any
surgeons, especially on this topic. It may be helpful to sched- unused tissue from the same donor and investigation into
ule grand rounds or other educational talks to ensure that the postoperative course of other patients who received tis-
all surgical staff is aware of the risks and what to look for. sue from the donor. The results of the investigation must be
Some clinical staff may be falsely assured by sterile packaging reported to all parties involved, including the transplanting
or disinfecting steps in processing. While contamination is surgeon and the tissue supplier, as well as the hospital risk
rare, it is more common in tissues with viable cells, such as management department and the tissue committee. Although
valves and vessels. Disinfection steps may not be effective not required, the tissue bank is encouraged to report the
for every organism. Additionally, steps may be missed due findings to the Joint Commission. If the event is severe or
to human error. fatal, the Joint Commission recognizes this as a sentinel
Contamination by bacteria or fungus should be suspected event requiring root cause analysis and corrective action.
based on the type of organism cultured, especially if the Although not required, the event may be reported to the
infection is deep and not a superficial wound infection. FDA through MedWatch. Reporting to the state is necessary
Unusual organisms for the wound or tissue should also be for those cases involving reportable disease. A final report
suspect. Although the time frame of onset can be factored should be placed in the file created for the case, and another
into the overall decision to report the infection as a suspected copy should be kept in the tissue vendor’s qualification file.
adverse event, it is less reliable, as it may vary greatly depend- It is important to remember to keep the patient informed
ing on the organism. Once the surgeon has sufficient infor- as well. The Code of Medical Ethics and the Joint Commis-
mation to suspect that the transplant is the reason for the sion are clear that the patient has a right to know when
infection, he or she should immediately report the suspected errors or unanticipated outcomes occur, particularly if the
event to the tissue bank. adverse event is severe or considered a sentinel event. It
Suspicion of viral or prion transmission by tissue is best if the treating surgeon conveys this information
should similarly be reported as soon as the physician be- because a relationship and level of trust already exists. If
comes aware. Obvious other causes of infection should be another physician must inform the patient, care should be
sought at the time of diagnosis. For example, a patient newly taken not to alienate the patient and treating physician, and
diagnosed with hepatitis C at some point following tissue the treating physician should be fully informed beforehand.
transplant should be questioned about IV drugs, tattoos, nee- When informing a patient of an error or allograft-related
dle sticks, and household contacts as possible sources of infection, it should be done compassionately, factually,
infection. and without blame. Expressions of concern and regret are
The surgeon may also report the suspected disease trans- appropriate, and there should be a plan for how to proceed.
mission to the FDA through the MedWatch program An offer of assistance in further treatment as appropriate
(www.fda.gov/medwatch). However, it is important that the should be made as well.
hospital tissue bank is not bypassed in the process, as an
important function of the medical director is to report to the Recalls
tissue supplier and investigate the case.
In the tissue bank, there are several important steps to the Recalls are almost always voluntary and may be initiated at vir-
investigation process. First, every complaint or suspicion tually any step in the process from the donor to the recipient.
should be taken seriously, even if there is a low chance that The recall is initiated after an error is discovered, even if it is
the event is related to tissue transplant. A file should be months to years after the tissue has been processed. An irreg-
opened on each case, and all documents generated in the ularity may be discovered in an FDA inspection that initiates
investigation should be kept together in the file. Immediately the recall, or a problem may be reported by workers or doctors
following a complaint, the personnel in the tissue bank who discover it in the course of using or handling the tissue.
should look for and quarantine any tissue from the same When a problem is discovered in the course of an investigation,
donor, pending outcome of the investigation. The medical the FDA may compel the establishment to initiate a recall.
director must be notified as soon as possible. The tissue There are three classes of FDA recalls (Box 22–3). Class I
supplier must also be notified as soon as possible so that is the most severe and serious. These recalls are reserved for
6888_Ch22_476-496 29/10/18 5:38 PM Page 494

494 PART III Transfusion Practices

patient. If the surgeon cannot be reached or does not wish


BOX 22–3 to notify the patient, it becomes the tissue bank’s responsi-
FDA Recall Classes bility, and reasonable attempts to reach the patient must be
made. Reasonable attempts to notify should occur in less
• Class I recall: Potential to cause serious injury or death
than 12 weeks of the recall. The explanation to the patient
• Class II recall: Potential to cause temporary or less serious health
problems should include information regarding the reason for the
• Class III recall: Unlikely to cause harm but violates FDA regulations recall, possible effects to him or her, and the opportunity to
or labeling make a decision regarding further testing or counseling with
a list of testing locations.

products that may cause serious harm, health problems, or


death if used. Class II is intermediate and is for products that CASE STUDIES
may cause temporary health problems or less serious injury.
Class III is likely the most common type of recall and per- Case 22-1
tains to products in which a manufacturing irregularity or A new orthopedic surgeon has just been hired at the hos-
violation in labeling is discovered, but the product is unlikely pital. He has a great deal of experience using a particular
to cause harm if used. type of processed bone that can be acquired from only one
Typically, the manufacturing establishment where the tissue vendor. He requests that the hospital tissue bank
tissue was originally processed notifies its customers of begin ordering this product for his upcoming scheduled
recalls. The FDA also disseminates information on recalls of surgeries. When the approved vendor list is consulted,
all types. These recalls can be found on the FDA website the vendor is not on the list.
through MedWatch, the FDA’s safety information and adverse
event reporting tool. Hospital tissue banks may also subscribe 1. What do you tell the surgeon who is expecting the
to a national recall reporting service such as the National tissue next week?
Recall Alert Center, although that is not required. 2. What is the minimum requirement to qualify the new
Just as there are steps in reporting adverse events, noti- vendor?
fication of a recall has several important steps. Upon receiv-
Case 22-2
ing notification of a recall, the hospital tissue bank must
verify that the product is or was in inventory, determine the A neurosurgeon calls the hospital tissue bank in advance
disposition of any and all tissue included in the recall, and of a planned craniotomy. He explains that he will not
quarantine any unused tissue. All tissues that have been be able to replace the bone flap at the completion of the
transplanted must be tracked to the recipient. If any tissues surgery planned for the next day, but within 1 to 2 weeks,
have been sold, transferred, or returned, the receiving facility he would like to close the head wound with the patient’s
must be notified of the recall. Just as in the adverse event own bone. He further states that it is possible that once
investigation, a file should be created for any investigation stabilized, the patient may be moved to another facility
into the disposition of recalled tissue and maintained for for the second procedure. He requests that the autologous
10 years. tissue be stored in the tissue bank and inquires as to the
Regulations on notification of the recipient have been procedure for transferring the tissue with the patient.
addressed by the Joint Commission and AABB. Both require
that recipients of tissue from donors who are subsequently 1. What are the minimum requirements for labeling
found to be positive for HIV, human T-cell lymphotropic autologous tissue?
virus (HTLV) type I or II, viral hepatitis, or other known 2. Under what condition could the hospital tissue bank
infectious agents transmissible by tissue must be notified. transfer the tissue?
This would include making reasonable attempts to notify the 3. Give two examples of procedural steps that should be
implanting physician and request that he or she notify the discussed prior to the surgery.

SUMMARY CHART
 In 2005, the Joint Commission ruled that hospitals  The FDA requires that any tissue-manufacturing es-
must assign responsibility for overseeing the tissue tablishment must register yearly with the FDA and
program throughout the organization, including stor- submit to inspection.
age and issuance activity (TS.03.01.01).  Hospital tissue banks that receive, store, and issue
 AABB and AATB recommend a centralized model of tissue do not need to register with the FDA unless their
tissue banking, and AABB states that the ideal location function also involves procurement, processing, or
for the tissue bank is in the hospital blood bank. distribution.
6888_Ch22_476-496 29/10/18 5:38 PM Page 495

Chapter 22 Tissue Banking as a Part of the Transfusion Service 495

SUMMARY CHART—cont’d
 Transfer of tissue to another facility is considered  Records must be maintained a minimum of 10 years
distribution. beyond the date of distribution, transplantation,
 Vascularized organs, such as liver, kidney, lung, pancreas, disposition, or expiration of tissue (whichever is lat-
and heart are excluded from FDA CFR 1270 and 1271. est). If required by state and/or federal laws, organi-
 The tissue bank should have a physician-appointed zations may have to retain tissue records longer than
medical director in line with the AABB’s model for 10 years.
blood bank.  Each tissue product must have a unique tissue identi-
 A tissue committee should be established that is fication number (TIN).
similar to, or part of, the transfusion committee.  The hospital tissue bank must verify any supplier and
 Tissue delivered to the tissue bank must be inspected vendor’s FDA registration on a yearly basis.
for package integrity and labeling and reconciled with  The Joint Commission requires that hospital tissue
packaging invoice, and must receive verification that banks ensure that suppliers are licensed by state agen-
temperature conditions have been maintained appro- cies as the laws apply for the state in which the vendor
priate to the tissue delivered. or tissue bank is located.
 The hospital tissue bank must maintain continual  Autologous tissue, used within the same facility with
monitoring of storage refrigerators and freezers. no intervening shipments off-site for further process-
 Record-keeping must detail each step or change in sta- ing, may be stored and issued with no requirement
tus of the tissue with notation of the person who per- to register with the FDA as a tissue-manufacturing
formed it and the date to allow for bidirectional tracking establishment.
from donor to final disposition.

5. The American Association of Tissue Banks (AATB) is:


Review Questions
a. A mandatory accrediting agency for all tissue banks
1. Implant records must be kept for what duration? b. A voluntary accrediting agency for tissue-manufacturing
establishments
a. Ten years after the tissue has been harvested
c. An historic name for the U.S. Navy Tissue Bank
b. Indefinitely
d. A subdivision within the AABB
c. For a reasonable time to ensure that the recipient is not
still alive when records are destroyed 6. Transmission of malignancy in tissue:
d. Ten years following the disposition or expiration of the a. Is most likely to occur with the use of bone
tissue b. Is relatively common (1/10,000 cases)
2. FDA CFR 1270 and 1271 include all of the following c. Is more likely to occur in whole organ transplant
tissues except: d. Has never been reported in cornea transplant
a. Cancellous bone chips 7. The medical director for the tissue bank can be:
b. Blood vessels associated with vascular organs for a. Any individual appointed by the hospital medical
transplant director
c. Cornea b. The lead supervisor in the blood bank
d. Heart valve c. The head nurse/transplant coordinator from surgical
3. Hospital tissue banks must register with the FDA if: nursing
d. A qualified physician involved in tissue transplant or
a. Tissue for transplant is stored
blood banking
b. Autologous tissue is stored and issued
c. Tissue is transferred to another facility 8. Notification of a recipient of tissue that has been recalled
d. The tissue bank is located outside the blood bank because of possible contamination:
4. The Joint Commission requires all of the following except: a. Should be conducted by the tissue bank director only
b. Should be conducted by the patient’s transplanting
a. Hospital tissue banks must ensure that suppliers are
surgeon
complying with applicable state laws
c. The patient does not need to be told unless an infec-
b. Tissue-manufacturing establishments must register
tion develops
with the FDA
d. The informed consent covers this contingency and no
c. Hospitals must assign responsibility for overseeing the
further notification is necessary
tissue program throughout the organization
d. Hospital tissue banks must verify supplier’s registration
with the FDA yearly
6888_Ch22_476-496 29/10/18 5:38 PM Page 496

496 PART III Transfusion Practices

9. Tissue receipt records must include all of the following 13. AABB. Standards for blood banks and transfusion services.
except: 31st edition. Bethesda (MD): AABB; 2018.
14. Frizzo W. Tissue management self-assessment tool for transfu-
a. Unique tissue identification number sion services and hospitals. Bethesda: AABB; 2008.
b. Name and address of tissue supplier 15. Code of Federal Regulations: Tissue Establishment Registra-
c. Expiration date tion. Title 21 CFR Part 1270 and 1271 [Internet]. [Cited 2017,
d. Tissue supplier’s FDA registration number May 12]. Available from: www.fda.gov/BiologicsBloodVaccines/
GuidanceComplianceRegulatoryInformation/Establishment
10. Records that must be reviewed to determine donor Registration/TissueEstablishmentRegistration/default.htm.
eligibility by the tissue manufacturer include: 16. Code of Federal Regulations: Current Good Tissue Practice
for Human Cell, Tissue, and Cellular and Tissue-Based Prod-
a. Donor family history uct Establishments; Inspection and Enforcement. Title 21
b. Records from any source pertaining to risk factors for CFR Parts 16, 1270, and 1271; Final Rule. Federal Register.
communicable diseases 69:68612; 2004.
c. Interview of next-of-kin 17. Shulman IA, forum editor and moderator [Internet]. California
Blood Bank Society e-Network Forum [2001, cited 2017
d. Consent to harvest tissue
12 May]. Should Blood Banks Store Autologous Bone Flaps
in Their Freezers? Available from: http://www.cbbsweb.org/
References forums/permalink.asp?id=1341769.
18. Shulman IA, forum editor and moderator [Internet]. California
1. Joint Commission. Tissue storage and issuance standards:
Blood Bank Society e-Network Forum [2006, cited 2017 May 12].
TS.03.01.01. TS.03.02.01, and TS.03.03.01. Oakbrook Terrace
Management and Ensuring Compliance of “Tissue Banking/
(IL): Joint Commission; 2017.
Tissue Dispensing Services.” Available from: http://www
2. Eastlund T, Eisenbrey AB, editors. Guidelines for managing
.cbbsweb.org/forums/permalink.asp?id=1341217.
tissue allografts in hospitals. Bethesda: AABB; 2006.
19. Gandhi MJ, Strong DM, editors. Donor derived malignancy
3. New York Organ Donor Network [Internet]. Troy [NY]: The
following transplantation: a review. Cell Tissue Bank. 2007;8:
Network [cited 2017, May 12]. The network organ and tissue
267-86.
transplant history. Available from: www.donatelifeny.org/all-
20. Southeast Tissue Alliance: Donor Alliance Donation Process.
about-transplantation/tissue-transplant-history/.
2012. Available at http://www.donoralliance.org/why-donate/
4. Strong DM. The U.S. Navy Tissue Bank: 50 years on the cutting
donation-process/. Accessed February 22, 2017.
edge. Cell Tissue Bank. 2000;1:9-16.
21. Bashaw MA. Implementation: autologous tissue management.
5. Osborne JC, Norman KG, Maye T, Malone P, Brubaker S, editors.
AORN J. 2015;102(3):270-83.
Standards for tissue banking. 14th ed. McLean (VA): American
22. Centers for Disease Control and Prevention. West Nile virus
Association of Tissue Banks; 2016.
infections in organ transplant recipients—New York and
6. The National Organ Transplant Act. P.L. 98-507. October 19,
Pennsylvania, August–September, 2005. MMWR Morb Mortal
1984 [cited 2017 May 12]. Available from: www.history.nih.
Wkly Rep. 2005;54:1021-3.
gov/research/downloads/PL98-507.pdf.
23. United States Code: Regulations to Control Communicable
7. Food and Drug Administration [Internet]. Rockville (MD):
Diseases. Section 361 Public Health Service Act, 42 USC § 264(a).
FDA [cited 2017 May 12] Guidance for industry: MedWatch
Washington, DC. US Government Printing Office, 2006.
Form FDA 3500A: Mandatory reporting of adverse reactions
24. Eastlund T, Strong DM. Infectious disease transmission through
related to human cells, tissues, and cellular and tissue-based
tissue transplantation. In: Phillips GO, editor. Vol. 7, Advances
products (HCT/Ps), 2009. Available from: www.fda.gov/Safety/
in tissue banking. Singapore: World Scientific Publishing; 2003.
MedWatch/HowToReport/ucm085568.htm.
pp. 51–131.
8. Eisenbrey AB, Eastlund T, editors. Hospital tissue management:
25. Joint Commission [Internet]. Oakbrook Terrace [IL]: Joint
a practitioner’s handbook. 1st ed. Bethesda: AABB; 2008.
Commission [cited 2017 May 12]. About The Joint Commission.
9. Eye Bank Association of America [Internet]. Washington, DC:
Available from: www.jointcommission.org/about_us/about_
EBAA [cited 2017 May 12]. Medical standards. Available from:
the_joint_commission_main.aspx.
http://restoresight.org/what-we-do/publications/medical-
26. Joint Commission [Internet]. Oakbrook Terrace [IL]: Joint
standards-procedures-manual/.
Commission [cited 2017 May 12]. History of The Joint Com-
10. Trainor LD, Reik RA. Human allografts and the hospital trans-
mission. Available from: www.jointcommission.org/about_us/
fusion service. In: Fung M, Grossman B, Hillyer C, Westhoff C,
history.aspx. Accessed May 12, 2017.
editors. Technical manual. 18th ed. Bethesda: AABB; 2014.
27. Houff SA, Burton RC, Wilson RW, Henson TE, London WT,
pp. 773-84.
Baer GM, et al. Human-to-human transmission of rabies virus
11. Code of Federal Regulations: Title 21 CFR Part 1270 and 1271
by corneal transplant. N Engl J Med. 1979;300:603-4.
[Internet]. Washington, DC: US Government Printing Office;
28. American Association of Blood Banks. Standards for blood
2017 (revised annually) [cited 2017 May 12]. Available from:
banks and transfusion services. 30th ed. Bethesda: AABB; 2016.
www.accessdata.fda.gov/scripts/cdrh/cfdocs/cfCFR/CFRSearch
29. American Association of Tissue Banks [Internet]. McLean
.cfm?CFRPart=1271.
[VA]: The Association [2017 May 11]. State Requirements for
12. Hillyer CD, Josephson CD. Tissue oversight in hospitals: the role
Tissue Bank Licensure, Registration or Certification. Available
of the transfusion services (editorial). Transfusion. 47:185-7;
from: http://www.aatb.org.
2007.
6888_Ch23_497-514 29/10/18 11:35 AM Page 497

Leukocyte Antigens and


Relationship Testing Part IV

Chapter 23
The HLA System
Donna L. Phelan, BA, CHS(ABHI), MT(HEW) and Gerald P. Morris, MD, PhD

Introduction Advanced Concepts: Clinical Significance Heart and Lung Transplantation


HLA Function of the HLA System Liver Transplantation
HLA Nomenclature Paternity Pancreas and Islet Cell Transplantation
HLA Genetics Disease Association United Network for Organ Sharing
Antibodies Against HLA Platelet Transfusion Summary Chart
Techniques of Histocompatibility Testing Transfusion-Related Acute Lung Injury Review Questions
HLA Genotyping Hematopoietic Stem Cell References
HLA Antibody Detection Techniques Transplantation
Microlymphocytotoxicity Kidney Transplantation

OBJECTIVES
1. Define the abbreviations HLA, MLR, SSO, SSP, SBT, NGS, TRALI, and MHC.
2. List the characteristics of HLA genes.
3. Describe the current nomenclature for HLA genes.
4. Define the terms haplotype and linkage disequilibrium.
5. Describe the difference between HLA phenotype and HLA genotype.
6. List the characteristics, importance, and clinical relevance of HLA class I and class II proteins.
7. Describe the techniques used for HLA genotyping.
8. Describe the techniques for HLA antibody detection.
9. Describe crossmatch techniques to evaluate tissue compatibility.
10. Describe the role of HLA testing in paternity testing, disease association, platelet transfusion, TRALI, and transplantation.
11. Explain the role of virtual crossmatches with single-antigen antibody testing.

Introduction It is necessary to evaluate the HLA antigen composition in


prospective donor-recipient pairs before organ transplantation
Human leukocyte antigen (HLA) testing is a specialized and in candidates for platelet therapy refractory to random
branch or division of immunology for human histocompati- donor platelets. Even more important is the evaluation and
bility testing. HLA testing supports a number of clinical spe- identification of HLA antibodies in the serum of recipients be-
cialties in transplantation, transfusion, and immunogenetics. fore transplantation and transfusion, which can predict clinical

497
6888_Ch23_497-514 29/10/18 11:35 AM Page 498

498 PART IV Leukocyte Antigens and Relationship Testing

rejection or refractoriness to transfusion. Evidence for human Class I (HLA-A, HLA-B, and HLA-C) molecules consist
leukocyte blood groups was first advanced in 1954 by Jean of an α chain with a molecular weight of 45,000 daltons that
Dausset,1 with the observation that patients whose sera con- associates noncovalently with β2-microglobulin, a smaller
tained leukoagglutinins had received a larger number of blood 12,000-dalton nonpolymorphic protein.3 The heavy chain
transfusions than other patients. He determined that these folds into three domains and is inserted through the cell
agglutinins were not autoantibodies as had been thought pre- membrane via a hydrophobic sequence. Class II (HLA-DR,
viously but rather were alloantibodies produced by the infusion HLA-DQ, and HLA-DP) molecules consist of two similar-sized
of cells bearing alloantigens not present in the recipient. Dausset chains of a molecular weight of 33,000 (α) and 28,000 (β)
determined that patients with alloantibodies segregated into daltons associated noncovalently throughout their extracel-
two groups based upon their reactivity; antibodies from group lular portions.4 In these molecules, both chains are inserted
1 specifically recognized antigens on group 2. Through this through the membrane via hydrophobic regions. The extra-
method, Dausset identified HLA-A2, the first known HLA mol- cellular portions of these chains fold into two domains.
ecule.2 HLA antigen testing is also used in disease correlation, Though there are differences in specific structures, the
relationship testing, and anthropologic studies. peptide-binding groove in class I molecules is formed by the
This chapter is intended to serve as an introduction to the α1 and α2 domains of the α chain while in class II molecules
basic concepts of HLA and clinical applications of HLA test- the peptide binding-groove is formed by α1 and β1 domains
ing. It is written for the reader with some training in the bio- of the α and β chains, respectively; the function of class I and
medical sciences and familiarity with general immunology class II HLA molecules is generally similar. Both class I and
concepts. The emphasis of the chapter is on the principles class II molecules fold to form two α helices with a β-pleated
and concepts of HLA structure and function, HLA proce- sheet floor that creates a pocket that holds short linear
dures, and the clinical application of HLA testing to im- peptides for presentation to T cells.
munogenetics (e.g., paternity and disease association), While both class I and class II molecules function to
transfusion practices, and transplantation. Although HLA present antigens to T cells, there are important differences.
testing is a technologically complex area, this chapter pro- Class I molecules present antigens to cytotoxic CD8+ T
vides a general overview of the topic. lymphocytes, while class II molecules present antigens to
CD4+ T lymphocytes.5 The specificity of this interaction is
HLA Function determined by interactions of the CD4 and CD8 corecep-
tors with the class II β chain and class I α chain, respec-
HLA molecules are the human orthologs of the major histocom- tively.6 Class I molecules are expressed ubiquitously and
patibility complex (MHC), a group of molecules with important are present on all nucleated cells as well as platelets,
function in regulating the activity of the immune system. HLA whereas class II molecules have a much more restricted dis-
proteins are expressed on the surface of cells and function pri- tribution. They are found predominantly on specialized
marily to present antigens to T lymphocytes. There are two types antigen-presenting cells such as B lymphocytes, macrophages,
of HLA molecules, class I and class II HLA (Fig. 23–1). monocytes, and dendritic cells, but can be expressed
by most cells including T cells and endothelial cells under
Human Leukocyte Antigen (HLA) inflammatory conditions.7
T lymphocyte The specific peptide antigens that can be presented to
T lymphocytes by HLA molecules are determined by the spe-
CD8 TCR CD4 cific amino acids that form the peptide-binding pocket.8
Antigen There is significant diversity in HLA, with multiple genes en-
coding HLA molecules (polygeny), numerous protein-
 2m   HLA chain
coding variations (alleles) for each HLA gene present in the
population (polymorphism), and simultaneous expression
A Class I Class II
of both chromosomal copies of each HLA gene (codominant
expression). This diversity is an essential component of HLA
Class I Class II function, as it promotes presentation of a wide array of anti-
 Chain gens to T cells. However, it is this diversity that often presents
 Chain a clinical challenge in transplantation and transfusion.

HLA Nomenclature
 Chain The extensive polymorphism of the HLA genes requires a
Antigen binding
B pocket systematic approach to nomenclature. The antigenic speci-
ficities, defined by serologic (antibody) reactivity, are des-
Figure 23–1. HLA molecules function to present antigen to T lymphocytes. A. ignated by numbers following the locus symbol (e.g.,
Diagram of interactions of class I HLA with CD8+ T cells and class II HLA with CD4+
T cells. B. Illustration of crystal structures of the antigen-binding pockets of class I
HLA-A1, HLA-A2, HLA-B5, HLA-B7, and so on). Table 23–1
HLA (PDB ID: 1HLA) made from the α chain and class II HLA (PDB ID: 2SEB) lists the current specificities of the HLA system recognized
formed from the α and β chains. by the World Health Organization Nomenclature Committee
6888_Ch23_497-514 29/10/18 11:35 AM Page 499

Chapter 23 The HLA System 499

Table 23–1 Recognized HLA Serologic Specificities


A B C DR DQ DP
A1 B5 B50 (21) Cw1 DR1 DQ1 DPw1

A2 B7 B51 (5) Cw2 DR103 DQ2 DPw2

A203 B703 B5102 Cw3 DR2 DQ3 DPw3

A210 B8 B5103 Cw4 DR3 DQ4 DPw4

A3 B12 B52 (5) Cw5 DR4 DQ5 (1) DPw5

A9 B13 B53 Cw6 DR5 DQ6 (1) DPw6

A10 B14 B54 (22) Cw7 DR6 DQ7 (3)

A11 B15 B55 (22) Cw8 DR7 DQ8 (3)

A19 B16 B56 (22) Cw9 (w3) DR8 DQ9 (3)

A23 (9) B17 B57 (17) Cw10 (w3) DR9

A24 (9) B18 B58 (17) DR10

A2403 B21 B59 DR11 (5)

A25 (10) B22 B60 (40) DR12 (5)

A26 (10) B27 B61 (40) DR13 (6)

A28 B2708 B62 (15) DR14 (6)

A29 (19) B35 B63 (15) DR1403

A30 (19) B37 B64 (14) DR1404

A31 (19) B38 (16) B65 (14) DR15 (2)

A32 (19) B39 (16) B67 DR16 (2)

A33 (19) B3901 B70 DR17 (3)

A34 (10) B3902 B71 (70) DR18 (3)

A36 B40 B72 (70) DR51

A43 B4005 B73 DR52

A66 (10) B41 B75 (15) DR53

A68 (28) B42 B76 (15)

A69 (28) B44 (12) B77 (15)

A74 (19) B45 (12) B78

A80 B46 B81

B47 B82

B48 Bw4

B49 (21) Bw6

Numbers in parentheses refer to broad serologic cross-reactive group.

for Factors of the HLA System.9 Several HLA alleles demon- the Bw4 and Bw6 epitopes, where the w is present to distin-
strate serologic cross reactivity due to structural similarity. guish them as significant serologic epitopes found on multi-
For example, HLA-A9, HLA-A23, and HLA-A24 all react with ple HLA-A and HLA-B alleles, rather than independent HLA
antibodies specific for HLA-A9, but HLA-A23 and HLA-A24 specificities (Table 23–2).
can be “split” from A9 by unique reactivity with antibodies Serologic reactivity was the first method for differentiating
specific for each. Two exceptions to HLA nomenclature are HLA alleles, but it is not sufficient for differentiation of all
6888_Ch23_497-514 29/10/18 11:35 AM Page 500

500 PART IV Leukocyte Antigens and Relationship Testing

Table 23–2 Bw4 and Bw6 Associated 5. The second numeric field defines the specific allele
protein. This typically describes serologically equivalent
Specificities
proteins that have differential ability to stimulate T-
Bw4 B5, B5102, B5103, B13, B17, B27, B37, B38 (16), B44 lymphocyte responses. However, some alleles within the
(12), B47, B49(21), B51(5), B52 (5), B53, B57 (17), same allele group have different serologic reactivity. This
B58(17), B59, B63(15), B77 (15) and A9, A23(9),
A24(9), A2403, A25 (10), A32 (19) is the source of serologic split antigens (i.e., the serologic
DQ3 group contains all of the alleles in the DQB1*03 allele
Bw6 B7, B703, B8, B14, B18, B22, B2708, B35, B39(16), group, but DQB1*03:01 encodes DQ7 serologic reactivity,
B3901, B3902, B40, B4005, B41, B42, B45(12), B46, DQB1*03:02 encodes DQ8 serologic reactivity, and
B48, B50 (21), B54 (22), B55 (22), B56 (22), B60(40),
B61(40), B62 (15), B64 (14), B65 (14), B67, B70, B71 DQB1*03:03 encodes DQ9 serologic reactivity).
(70), B72 (70), B73, B75 (15), B76 (15), B78, B81, B82 6. The third and fourth numeric fields convey other, less
clinically relevant but scientifically important informa-
tion. The third field identifies alleles that differ only by
synonymous nucleotide substitutions within the coding
biologically relevant HLA alleles. The 165 recognized sero- sequence. The fourth field identifies alleles that only
logic specificities represent only 1% of the different HLA differ in non-protein-coding regions such as introns or
allelic variant proteins identified.9 These “serologically silent” untranslated regions. Some of these polymorphisms can
variants are predominantly the result of amino acid differ- affect gene expression by creating low-expression or null
ences in the peptide-binding pocket, which are inaccessible (nonexpressed) variants. Clinically relevant effects are de-
to antibodies, but critically influence T-lymphocyte responses noted by the addition of a capital letter at the end of the
through differential presentation of peptide antigens.10 This fourth field (N for null, L for low expression).
complexity and the clinical impact of HLA genetic variation
have necessitated development of the following nomenclature HLA Genetics
for HLA genes (Fig. 23–2):
The HLA genetic region is an approximately 4-MB region of
1. HLA-prefix designates the MHC gene complex. the short arm of chromosome 6 that contains approximately
2. Capital letters indicate a specific gene. Class I genes are 250 genes.11 The region is also referred to as the major his-
a single capital letter (A, B, C). All class II genes are pre- tocompatibility complex (MHC), as the classical MHC or
fixed by the letter D and followed by a second capital let- HLA genes are the principal determinants of transplantation
ter (DR, DQ, DP). Genes coding for the specific class II α compatibility. The HLA locus is grouped into three regions,
and β proteins are identified by capital A or B along with the class I, class II, and class III regions (Fig. 23–3). The class
a number to indicate the specific gene (i.e., DRA1, DRB1, I region contains the genes for HLA-A, -B, and -C. It also en-
DRB2, etc.). codes for additional nonclassical HLA, including HLA-E, -F,
3. Alleles for each of the genes are identified by a four-field and -G. The class II region contains genes for the α and β
series of two- to three-digit numbers separated by a colon. chains of HLA-DR, -DP, and -DQ. DP molecules are the prod-
An asterisk is placed between the gene name and the nu- uct of DPA1 and DPB1 genes, and DQ molecules are the
merical identifier for the allele to differentiate between product of DQA1 and DQB1 genes. HLA-DR molecules are
the gene nomenclature and HLA serologic reactivity. more complex, as all use an α chain encoded by DRA1 but
4. The allele group is designated by the first numeric field. can use β proteins coded by DRB1 (the classic DR specifici-
This frequently correlates with the serologic group reac- ties, which are the most abundantly expressed), DRB3 (DR52
tivity (i.e., HLA-A*02 alleles demonstrate A2 serologic molecules), DRB4 (DR53), and DRB5 (DR51). DPA2, DPB2,
group reactivity). DRB2, DRB6, DRB7, DRB8, and DRB9 are pseudogenes that
do not encode for expressed proteins. The class III region
encodes structurally and functionally diverse molecules, in-
HLA gene nomenclature cluding complement factors, 21-hydroxylase, and tumor
HLA-A*01:01:01:02N necrosis factor.
Protein
expression The physical linkage of the HLA genes on chromosome
Noncoding 6 results in all of the genes on a single chromosome typically
DNA sequence
Gene Allele Allele Synonymous HLA locus chromosome 6p
group DNA polymorphism
R /5
D 3/4
D 1
B1

D 1

A1
D 1
1

B
B
PB
PA

Q
Q

R
R

Low-resolution genotype and serology


D

C
B

HLA-A*02 A2
DP DQ
High-resolution genotype DR DR51/52/53 B C A
HLA-DRB1*04:01
Figure 23–3. HLA genes encoding the HLA proteins are located together on
Figure 23–2. Nomenclature for HLA genes. the short arm of chromosome 6.
6888_Ch23_497-514 29/10/18 11:35 AM Page 501

Chapter 23 The HLA System 501

being inherited together. The entire set of HLA-A, -B, -C, would be expected based on their individual gene frequen-
-DR, -DQ, and -DP genes located on one chromosome is cies. Linkage disequilibrium can be useful in evaluating HLA
called a haplotype. Genetic crossovers and recombination in haplotypes, providing important clues to assure correct
the HLA region are uncommon,12 and thus a complete set of genotype assessment. Examples of useful linkage disequilib-
alleles located on a chromosome is usually inherited by chil- rium are shown in Tables 23–3 and 23–4.
dren as a unit (haplotype). Figure 23–4 illustrates the segre-
gation of HLA haplotypes in a family. The two haplotypes of Antibodies Against HLA
the father are labeled a and b, and the mother’s haplotypes
are c and d. Each child inherits two haplotypes, one from The diversity of HLA genes provides a significant source of
each parent. Four possible haplotypes—ac, ad, bc, and bd— phenotypic diversity between different individuals, suffi-
can be found in the offspring. Thus, 25% of the offspring will cient for stimulating strong immune responses. Some of the
have identical HLA haplotypes, 50% will share one HLA hap- differences between HLA molecules are capable of stimulat-
lotype, and 25% will share no HLA haplotypes. An important ing humoral immune responses resulting in the formation
corollary is that a parent and child can share only one hap- of antibodies against allogeneic HLA molecules (alloanti-
lotype, making an identical match between the two unlikely. bodies). Antibodies against allogeneic HLA molecules are
It should also be apparent that uncles, grandparents, and an important barrier to successful transplantation because
cousins are very unlikely to have identical haplotypes with they can cause rejection of transplanted organs13 or destruc-
any given child. These are important factors when looking tion of transfused cells.14
for a well-matched organ or blood donor. Unlike antibodies to the AB blood group antigens, anti-
An event that infrequently complicates HLA typing inter- bodies against allogeneic HLA are not naturally present. In-
pretation and haplotype determination is crossover, or re- stead, they arise as a consequence of a sensitizing event or
combination of HLA genes between haplotypes. During exposure to allogeneic HLA.15 The most common cause of
meiosis, complementary exchange of genetic material be- sensitization against allogeneic HLA is pregnancy, with esti-
tween chromosomes can occur. The farther apart two loci mates of 20% to 35% of women developing anti-HLA anti-
are on a given chromosome, the more likely it is that genetic bodies after pregnancy. The second most common source of
exchanges will take place. For example, recombination be- sensitization against HLA is through transfusion. The degree
tween HLA-A and HLA-DPB1 genes occurs commonly, of risk depends on the blood product transfused, ranging
whereas recombination between HLA-DQB1 and HLA-DRB1 from leukoreduced PRBCs having an approximate risk of
is a rare event. Crossing over has the effect of rearranging 10% to 20% of patients developing antibodies against allo-
the genes on the chromosome to produce new haplotypes in geneic HLA to single donor platelets carrying a 50% risk.
the general population. The highest risk for developing anti-HLA antibodies comes
The conservation of HLA haplotypes results in predictable from transplantation, where as many as approximately 70%
associations between alleles of HLA genes. Linkage disequi- of patients develop anti-HLA antibodies within 5 years of
librium refers to associations between HLA gene alleles that transplant. Antibodies against HLA are similar to antibodies
occur more frequently than would be expected by random produced in response to other immunizing events. The ma-
assortment in the population. For example, if HLA-A*01 and jority of HLA alloantibodies are IgG subclasses capable of me-
HLA-B*08 gene frequencies are 0.16 and 0.1, respectively, in diating typical immune functions such as activation of
a population, the expected occurrence of an HLA haplotype complement and opsonization that lead to antibody-mediated
bearing both A1 and B8 should be 0.16 × 0.1 × 100, or 1.6%. rejection (AMR).16
In certain Caucasian populations, however, the actual occur- Antibodies to HLA molecules can be divided into two
rence of this haplotype is as high as 8%, far in excess of the groups, those that detect a single HLA protein (“private”
expected frequency. This is similar to the Rh system, where antibodies binding to an epitope unique to a specific HLA
blood groups C, D, and e are found together more often than allele) and those that detect more than one HLA protein.

Father Mother
ab cd
A*01 A*30 A*02 A*30 Recombination during meiosis
B*08 B*13 B*58 B*42 generated “crossover” haplotype
A*02 A*30
DRB1*03 DRB1*07 DRB1*11 DRB1*03
B*58 B*42

DRB1*11 DRB1*03

Typical haplotype inheritance

ac ad bc bd a c/d
A*01 A*02 A*01 A*30 A*30 A*02 A*30 A*30 A*01 A*02
B*08 B*58 B*08 B*42 B*13 B*58 B*13 B*42 B*08 B*58
DRB1*03 DRB1*11 DRB1*03 DRB1*03 DRB1*07 DRB1*11 DRB1*07 DRB1*03 DRB1*03 DRB1*03

Figure 23–4. Inheritance of HLA haplotypes.


6888_Ch23_497-514 29/10/18 11:35 AM Page 502

502 PART IV Leukocyte Antigens and Relationship Testing

Table 23–3 HLA Alleles Frequently Table 23–4 Common HLA-DRB1–DRB3/4/5


Associated Through Haplotype Linkage
Disequilibrium DRB1 DRB3/4/5
Ethnic Background HLA-A, HLA-B, HLA-DR *01 None
European Caucasian A*01:01, B*08:01, DRB1*03:01
*03 DRB3
A*03:01, B*07:02, DRB1*15:01
*04 DRB4
A*02:01, B*44:02, DRB1*04:01
*07 DRB4
A*02:01, B*07:02, DRB1*15:01
*08 None
A*29:02, B*44:03, DRB1*07:01
*09 DRB4
African American A*30:01, B*42:01, DRB1*03:02
*10 None
A*01:01, B*08:01, DRB1*03:01
*11 DRB3
A*68:01, B*58:02, DRB1*12:01
*12 DRB3
A*68:02, B*15:10, DRB1*03:01
*13 DRB3
A*33:03, B*53:01, DRB1*08:04
*14 DRB3
Asian Pacific Islander A*33:03, B*58:01, DRB1*03:01
*15 DRB5
A*02:07, B*46:01, DRB1*09:01
*16 DRB5
A*33:03, B*44:03, DRB1*07:01

A*30:01, B*13:02, DRB1*07:01


A2 CREG
A*33:03, B*58:01, DRB1*13:02 A2
A68 A69
Hispanic A*29:02, B*44:03, DRB1*07:01 A23 A24 A2403
B57 B58
A*01:01, B*08:01, DRB1*03:01 A1 CREG
A25 A26
A32 A33 A34
A1 A3
A*03:01, B*07:02, DRB1*15:01 A43 A66
A11 A29 A30
A74
A*30:02, B*18:01, DRB1*03:01 A31 A36
A10 CREG
A80
A*33:01, B*14:02, DRB1*01:02 B5 CREG
B35 B46 B51 B52
B53 B62 B63 B71 B72
B73 B75 B76 B77
B78
Antibodies that are reactive against multiple HLA molecules B49 B12
B8 B18 B50
may be cross-reactive (binding similar epitopes) or may rec- CREG
CREG
ognize shared or “public” epitopes shared by more than one B37 B44
B38 B39 B13 B41
HLA gene product (i.e., Bw4 or Bw6).17–19 Cross reactivity B64 B8 B45
B59 B47 B60
is a phenomenon in which an antiserum directed against B65 B61
B67 B7
one HLA antigenic determinant reacts with other HLA anti- B27 B42 B48
genic determinants as well.20,21 Cross-reactive antigens B54 B55 B56
share important structural elements (and epitopes) with one B81 B82
another but retain unique, specific elements. The majority B7 CREG
of cross-reactive alloantibodies detect HLA specificities of
allelic molecules coded by the same locus. On the basis of Figure 23–5. HLA serologic cross-reactive groups (CREGs).
these reactions, most specificities can be grouped into major
cross-reactive groups (CREGs) (Fig. 23–5). For example, Recently, CREGs have been defined molecularly using
the HLA-A locus antigens A2, A23, A24, and A28 share a amino acid residues. Antibody-reactive epitopes have been
common epitope and therefore make up the A2 CREG, or predicted from analysis of DNA-encoded protein sequence
A2C. HLA-A28 also shares a different epitope with A26, and validated through analysis of cross-reactive sera.26–32
A33, and A34, defining the A28 CREG. There are also at Publicly available resources, such as HLAMatchmaker,26,27
least three cross-reactions between the HLA-A and HLA-B provide tools for prediction of cross-reactive serologic epi-
loci: HLA-A23, HLA-A24, HLA-A25, and HLA-A32 with topes. CREGs and epitope prediction are used predomi-
HLA-Bw420–23; HLA-A11 with HLA-Bw620; and HLA-A2 nantly in defining antibodies present in recipient antisera
with HLA-B17.24,25 and predicting antigens to avoid in potential organ donors.32
6888_Ch23_497-514 29/10/18 11:35 AM Page 503

Chapter 23 The HLA System 503

Techniques of Histocompatibility oligonucleotide probes specific for allele-defining DNA se-


Testing quences that are labeled with enzymatic or fluorophore
probes.35 SSO probe hybridization is performed under strin-
The principles used in histocompatibility testing are concep- gent conditions that ensure that only probes that accurately
tually similar to those used for red blood cell (RBC) testing. complement the target sequence hybridize and produce a
Donors and recipients are typed for HLA genotype/phenotype, positive signal. Current techniques utilize probes bound to
recipient serum is screened for the presence of HLA antibodies fluorophore-barcoded solid-phase substrates, which enable
using immunoassays, and compatibility is directly determined extensive multiplexing of probes to an amplicon, increasing
by crossmatching of donor cells and recipient sera. the ability of the assay to identify allele-specific sequences.

HLA Genotyping Sequence-Specific Primers


HLA antigens were historically identified using sera with In SSP approaches, oligonucleotide primers for PCR ampli-
known anti-HLA antibodies, similar to the methods per- fication are designed to target specific DNA sequences that
formed for RBC phenotyping. However, the diversity of HLA identify specific HLA alleles.36 PCR amplifications using SSP
genes has necessitated utilizing DNA-based methods for ac- are performed under stringent conditions that ensure that
curate typing. The level of specificity of HLA typing depends amplicons are only generated when the SSP sequence pre-
upon the clinical application. Hematopoietic stem cell trans- cisely complements the target sequence. HLA genotyping is
plantation requires high-resolution allele-level (two-field) determined by analysis of the presence or absence of ampli-
HLA genotyping.33 This enables accurate HLA matching of fication products from multiple SSP PCR reactions. Typically,
donor and recipient to minimize graft-versus-host disease, a amplicon detection is performed by gel electrophoresis.
pathological response of donor T lymphocytes to recipient However, quantitative PCR (qPCR) methods can also be
tissues. In solid organ transplantation and transfusion, the used to detect amplification products from multiplexed SSP
primary concern is for antibodies against allogeneic HLA reactions.
that could mediate rejection. Thus, the concern is to identify
potentially serologically reactive antigens.34 However, given DNA Sequence-Based Typing
the considerable diversity of HLA, this is most commonly Direct nucleotide sequencing of HLA genes is utilized for
accomplished by DNA-based genotyping and prediction of high-resolution typing and is required in the definition of new
phenotype. Most commonly, the first field (allele group) of alleles. SBT is performed by two methods in the clinical lab-
the allele conveys the serologic specificity, although there are oratory, Sanger-based DNA sequencing and next-generation
several well-documented cases where additional allele-level DNA sequencing (NGS). Sanger-based typing utilizes terminal-
(second field) information is required to predict the presence end incorporation of labeled nucleotides during PCR reac-
of common serologic antigens (Table 23–1). The three com- tions.37 Since Sanger-based methods are limited to sequencing
mon HLA typing assays employed in the clinical laboratory 800 to 1,000 base pair sequences, the most polymorphic re-
are sequence-specific oligonucleotides (SSO), sequence- gions of the HLA genes are targeted: exons 2, 3, and 4 of class
specific primers (SSP), and DNA sequence-based typing I genes, and exon 2 of the class II β chain genes. Fluorescently
(SBT) (Fig. 23–6). tagged DNA amplification products are sorted by capillary
electrophoresis, and terminal nucleotide fluorescence is
Sequence-Specific Oligonucleotides measured. This generates a sequence of fluorophore signals
SSO typing involves PCR amplification of HLA gene regions that corresponds to the nucleotide incorporated at each po-
using primers to conserve DNA sequences flanking the sition. Heterozygous positions are identified by mixed fluo-
region. Amplified DNA is denatured and hybridized with rescence signals at a position. The resulting sequences of

Sequence-specific Sequence-specific DNA sequence-based


oligonucleotides (SSO) primer PCR (SSP) typing (SBT)
PCR x’ Stringent PCR PCR
A*01:01 x x x
amplification A*01 conditions amplification
A*02:01 y y
of all alleles x’ + enables only of all alleles
y complementary
primers
y’ to anneal and
x A*02 amplify target AGTCGTTAC DNA sequence
A*01 Probes with of amplicons
y’ + determined
x’ + complementary (’) AGTAGTTAC
x y Alleles detected
A*02 DNA sequences by presence or Alleles identified
y’ + detect known z’
y x absence of by comparison
polymorphisms A*03
A*03 defining amplicons in with HLA
z’ – primer-specific HLA-A*01:01 sequence
– specific alleles
z’ y reactions HLA-A*02:01 database

Figure 23–6. Methods of HLA genotyping.


6888_Ch23_497-514 29/10/18 11:35 AM Page 504

504 PART IV Leukocyte Antigens and Relationship Testing

nucleotides are compared to known DNA sequences of HLA not caused by the presence of antibodies in general, but
alleles to find a match, identifying the HLA allele.38,39 A ro- rather by the presence of donor-specific anti-HLA antibodies
bust database of HLA allele DNA sequences, the Immuno (DSA)—those anti-HLA antibodies that recognize donor
Polymorphism Database-International Immunogenetics In- HLA molecules on transplanted tissues. DSA are specifically
formation System (IPD-IMGT)9 is curated to ensure accurate associated with accelerated graft rejection and with poor
information is available for clinical use. response to platelet transfusion. Thus, the clinical manage-
Sanger-based SBT enables unbiased evaluation of HLA ment of patients, pre- and post-transplant, includes screen-
genotypes but with several significant limitations. Sanger- ing for and determining the specificity of anti-HLA class I
based SBT is typically limited to focused analysis of specific and II antibodies that may be present.
exons rather than complete gene sequences. More impor- Anti-HLA antibody detection and identification involves
tantly, Sanger-based SBT is unable to resolve the complemen- serologic and cellular procedures, similar to blood bank pro-
tarity (phase) of polymorphic nucleotide sequences from cedures. Crossmatch testing, which directly evaluates the
two alleles in the same amplicon sequence. This means that presence of antibodies in serum reacting against donor
a mixed nucleotide (i.e., A or G) at one position in the DNA leukocytes, is performed by microlymphocytotoxicity and
sequence cannot be linked to a mixed nucleotide (i.e., G or T) flow cytometry techniques.49 Analysis of anti-HLA anti-
at a second position in the sequence. Therefore a sequence bodies is performed using cytotoxicity and flow cytometry
that is A/G C T G/T C by Sanger SBT could represent methods against cell panels as well as recombinant HLA
ACTGC, ACTTC, GCTGC, or GCTTC. As the number of molecules. Taken together, these assays enable confident
polymorphic nucleotides increases to n for a given gene identification and definition of antigenic specificity of anti-
being examined, the number of potential DNA sequences be- HLA antibodies.
comes 2n. Many of these possible sequences can be deter-
mined to be less probable by their absence in the reference Microlymphocytotoxicity
database, although this still provides significant ambiguity
in HLA genotyping results. The complement-dependent cytotoxicity (CDC) method
The primary limitations in Sanger-based SBT have been utilizes the biological function of antibodies to activate com-
overcome by newer DNA sequencing technologies. Next- plement and disrupt cell membrane integrity. Serum being
generation sequencing (NGS) refers to DNA sequencing tested for the presence of anti-HLA antibodies is incubated
methods in which multiple sequencing reactions are per- with leukocytes from either a potential donor (or reference
formed in parallel, which greatly improves the throughput sample with known HLA phenotype) in the presence of ex-
and accuracy of DNA sequencing.40 Clinical NGS methods ogenous complement and a fluorescent dye (Fig. 23–7).50 In
rely on “sequence by synthesis” approaches, in which the the absence of antibodies binding the cells, the dye will be
incorporation of specific nucleotides to the terminal end of excluded from the cells. If antibodies against donor HLA
DNA strands is detected by emission of ions or photons molecules are present, they will bind the leukocytes and ac-
or fluorophore incorporation. Nucleotide sequences for tivate the classical complement cascade, resulting in forma-
millions of 50 to 500 bp clonal DNA strands are recorded tion of the complement membrane attack complex that
independently and assembled in silico into much larger forms holes in the cell membrane allowing the dye to enter
contiguous stretches of DNA sequence by aligning homol- the cell. The reaction is graded as the percentage of cells that
ogous regions. This approach can provide DNA sequences have taken the dye using titered sera. When performing CDC
covering complete HLA gene sequences. The ability to in- crossmatch to evaluate compatibility between a patient and
terrogate full exon, intron, and noncoding sequences en- potential organ donor, a reaction causing uptake of dye in
ables improved accuracy in genotyping. Additionally, since
the DNA sequence was determined by sequencing individ-
ual strands of DNA, NGS methods can determine the com- Complement-Dependent Lymphocytotoxicity (CDC)
plementarity of heterozygous nucleotides and accurately Assays
attribute them to different alleles, eliminating a significant AMOS Antiglobulin (AHG)
CDC augmented
source of ambiguity in HLA genotyping. Early applications modified
Stage 1
of NGS methods for HLA genotyping have occurred in a Antigen-antibody Cells + Serum
research setting, although NGS use in clinical laboratories interaction
30 min.
is increasing.41–45 WASH
Stage 2 x1 WASH x 3
Complement- Complement x2 add AHG
HLA Antibody Detection Techniques mediated or 2 min.
cell injury x3
60 min.
Antibodies to HLA alloantigens may cause significant com-
Stage 2
plications in transplantation13,46,47 or transfusion.14 Anti- Visualize Vital Stain
HLA antibody in donor plasma to recipient leukocytes has cell membrane
injury
been associated with severe pulmonary infiltrates and respi-
ratory distress following transfusion, known as transfusion- Figure 23–7. Complement-dependent microlymphocytotoxicity assay with two
related acute lung injury (TRALI).48 These pathologies are variations: three-wash Amos and antiglobulin.
6888_Ch23_497-514 29/10/18 11:35 AM Page 505

Chapter 23 The HLA System 505

greater than 40% of cells is typically considered a positive lymphocytes have the advantage of storage and rapid prepa-
reaction. Serum can also be tested against panels of cells as ration, facilitating rapid testing. However, the process of
a screening method to detect anti-HLA antibodies. In this freezing renders lymphocytes more fragile and susceptible
case, assay results are interpreted as the percentage of cellular to cell lysis, resulting in false positives. An additional prob-
phenotypes that elicited a positive reaction among all lem is the limited sensitivity of CDC methods. Significant
phenotypes tested. This reactivity is reported as the panel- data indicate that anti-HLA antibodies present at low titers
reactive antibody (PRA) percentage. Provided the cell panel may not be identifiable, even by enhanced CDC methods,
tested is representative of the potential donor population, but can still negatively affect transplant outcomes.
the PRA provides an estimate of the likelihood of the patient
being incompatible with a random donor. Flow Cytometry
The CDC method has been used to predict transplant The flow cytometric crossmatch directly detects antibody
compatibility since the late 1960s and is the historic refer- binding to leukocytes.52 Serum is incubated with donor
ence standard for detection of anti-HLA antibodies that are leukocytes, washed, and bound antibody is detected using
expected to cause antibody-mediated rejection (AMR). How- fluorophore-labeled anti-immunoglobulin (Fig. 23–9). Assay
ever, the standard CDC has limited analytical sensitivity, results are determined by comparing the fluorescence of cells
which has promoted implementation of technical modifica- incubated with patient serum with that of cells incubated
tions.51 Increasing the incubation time can enhance the abil- with negative control serum lacking anti-HLA antibodies.
ity to detect low-titer antibodies. The Amos-modified The degree of antibody binding is reported as the mean chan-
technique adds a wash step after the initial serum-cell incu- nel shift (mcs) in fluorescence intensity attributable to pa-
bation. This wash step removes anti-complement activity. tient serum. T cells expressing class I HLA and B cells
Addition of dithiothreitol (DTT) can be used to disrupt expressing both class I and class II HLA can be differentiated
disulfide bonds of IgM molecules, which may have nonspe- by labeling cells with anti-CD3 and anti-CD19, respectively.
cific low-affinity high-avidity binding causing false-positive Flow cytometry–based cellular crossmatch demonstrates im-
reactions. Sensitivity of the CDC test can be greatly enhanced proved sensitivity for detection of anti-HLA antibodies ca-
by adding an antihuman globulin (AHG) reagent following pable of mediating rejection compared to CDC methods.
serum and cell incubation.51 Use of AHG is similar to the ad- However, care must be taken to avoid false-positive reactions
dition of Coombs reagent in testing for antibodies to RBC from nonspecific antibody binding. In particular, B cells ex-
antigens. To distinguish between antibodies against class I press receptors for the Fc portion of antibodies that can
and class II HLA molecules, separated or antibody-labeled cause significant background or nonspecific antibody bind-
B-lymphocyte suspensions must be used. ing.53 To address this, Fc receptors can be enzymatically re-
Lymphocytotoxicity testing has several disadvantages. moved using pronase, a mix of proteolytic enzymes derived
First, it requires having cells available for testing. In the case from the bacteria Streptomyces griseus. Pronase significantly
of direct crossmatch testing against a potential donor, it is reduces nonspecific antibody binding, but can also cleave
necessary to collect leukocytes and perform cellular testing HLA molecules from the cell surface, an action that could
in a timely manner to enable transplantation. For anti-HLA theoretically cause false-negative crossmatch reactions.54 The
antibody screening using CDC methods, it is necessary to improved analytical sensitivity of flow cytometry as well as
maintain reliable and consistent antigen panels that repre- a relative technical simplicity and a decreased effect of cell
sent a broad range of HLA antigens. The viability of T and B
lymphocytes is essential for an accurate assessment of the
presence of antibody. In most cases, donor lymphocytes for
100
crossmatch testing are isolated from peripheral blood by den-
sity gradient centrifugation (Fig. 23–8). However, CDC test-
ing can also be performed using frozen lymphocytes. Frozen

50
Diluted
whole Centrifuge Plasma
blood

Lymphocytes
and monocytes
Ficoll-
Hypaque

Red blood cells 0 0


10 101 102 103 104
and granulocytes
Figure 23–9. Flow cytometry histogram illustrating the shift in fluorescence light
Figure 23–8. Lymphocyte separation. intensity emitted by a cell population coated with fluorescein-labeled antibody.
6888_Ch23_497-514 29/10/18 11:35 AM Page 506

506 PART IV Leukocyte Antigens and Relationship Testing

viability makes flow cytometry–based crossmatch a robust population frequencies of HLA against which the serum has
platform for crossmatch testing. antibodies (calculated PRA [cPRA]).59 The United Network
Flow cytometry can also be used to screen for the pres- for Organ Sharing (UNOS) utilizes an algorithm based on
ence of HLA antibodies using cell panels, similar to the population HLA frequency to calculate cPRAs for patients
approach described for micro lymphocytotoxicity assays. waiting for organ transplantation.60
Because flow cytometry evaluates antibody binding directly, The most significant application of SAB assays is to define
cell lysates or HLA molecules purified from cell lysates the antigenic specificity of anti-HLA antibodies in a patient
(phenotype panels) can be used to improve throughput serum. Unlike phenotype panels, SAB enables identification
and specificity.55,56 of specific antibodies recognizing donor HLA molecules
(donor-specific antibodies [DSA]). Whereas PRA gives the
Multiplex Single-Antigen Bead Immunoassay (SAB) likelihood of compatibility with a random donor from the
The extensive polymorphism of HLA molecules, combined population, the presence or absence of DSA relates to the com-
with codominant expression of multiple genes and alleles, patibility with a given donor with a known HLA type. Detec-
would require testing a prohibitively large number of cell tion of DSA by SAB, either before or after transplantation, is a
phenotypes to accurately identify anti-HLA antibody speci- strong predictor of antibody-mediated rejection.61–63
ficities using cell-based methods. Initial approaches at facil- The MFI is generally related to the amount of anti-HLA
itating multiplexed high-throughput analysis utilized cell antibody present, although there is not a direct correlation
lysates and ELISA methods.55,56 This increased throughput, between MFI and titer.64,65 The quantitative accuracy of SAB
but was still limited by codominant expression of multiple is significantly limited by standard issues affecting immune
HLA molecules, precluding accurate identification of anti- assays, such as a limited linear range and saturation or pro-
HLA antibody specificities. Development of multiplex im- zone effects,64,65 as well as issues more specifically related to
munoassays using recombinant individual HLA proteins, or HLA testing, such as interference by complement compo-
single-antigen bead (SAB) assays, has overcome this limi- nents,66–68 variable amounts of recombinant HLA on the bead
tation.57,58 The SAB assay tests patient sera against a panel surface, and a high degree of intra-assay variability.58,69 SAB
of recombinant soluble HLA antigens, each bound to spe- demonstrates significantly increased analytical sensitivity
cific microbeads (Fig. 23–10). Each microbead is labeled compared to earlier ELISA-based tests or cellular crossmatch
with a distinct set of fluorophores, enabling direct evalua- and is capable of detecting anti-HLA antibodies that are not
tion of antibody-binding specificity. Anti-HLA antibody detected by other methods. The clinical significance of anti-
binding is detected by labeling with PE-conjugated anti-Ig HLA antibodies only detectable by SAB is currently a subject
and measured on a specialized flow cytometer. Positive re- of debate. Thus, established guidelines suggest defining a
actions are described as the mean fluorescence intensity minimum MFI cutoff value that correlates with cellular cross-
(MFI) of positive beads. match assays to provide clinical relevance to SAB results.70
Similar to anti-HLA antibody testing using cell panels or The question of clinical significance of antibodies identi-
phenotype beads, SAB can evaluate the overall degree of al- fied by SAB has led to the development of modified SAB as-
losensitization. Since SAB uses individual recombinant HLA says designed to evaluate potential for activation of serum
molecules rather than panels of HLA from representative complement and thus cause antibody-mediated rejection.
cells, serum reactivity by SAB is calculated as the sum of the These modified SAB assays add exogenous complement after

Phenotype Panel Antibody Testing Single Antigen Bead Antibody Testing


Donor cell phenotype Detection
anti-IgG
A1, A2
antibody
B8, B44
Cw5, Cw7

A1, A11
B8, B35
Cw4, Cw7 Detection Anti-HLA antibody
anti-IgG from patient serum
A2, A24 antibody
B54, B72 A1 A2 A11 A24 A30 A33
Cw1, Cw2
Recombinant HLA
on individual beads
A30, A33
B13, B58 Anti-HLA antibody
Cw3, Cw6 from patient serum Result = Antibodies against A1, A2, and A24 detected
cPRA = 74%
Result = 3/4 cells tested positive, PRA = 75%

Figure 23–10. Phenotype panel and single-antigen bead antibody testing.


6888_Ch23_497-514 29/10/18 11:35 AM Page 507

Chapter 23 The HLA System 507

incubation of the beads with patient serum and subsequently accomplish this, the laboratory uses several techniques to
measure the antibody-induced binding of complement pro- accurately identify the polymorphic genetic markers in a pa-
teins with fluorophore-labeled detection antibodies against ternity trio. Combinations of various genetic markers are
complement proteins such as C1q, C3d, or C4d.71,72 These used, including RBC markers and enzymes, serum proteins,
complement-modified SAB assays have excellent positive pre- HLA typing, and DNA testing.77 Previously, the most com-
dictive power to identify DSA-mediating allograft rejection. mon combination of testing included RBC markers together
Additionally, this approach eliminates interference from en- with HLA typing. Owing to increased costs of HLA typing
dogenous complement bound to anti-HLA antibodies that reagents and the significantly high power of exclusion when
can cause false-negative SAB results. However, complement- testing multiple DNA loci, DNA methodologies are used
modified SAB assays have significantly decreased analytical increasingly more than HLA typing.
sensitivity compared to unmodified SAB assays.65,73 This de-
creased sensitivity is relevant, as anti-HLA antibodies detected Disease Association
by unmodified SAB but not detected by complement-modified
SAB are capable of mediating rejection. It has been determined that HLA antigens are associated with
disease susceptibility to a greater extent than any other known
Virtual Crossmatch genetic marker.11 Many genetic markers have been suspected
Since the first description of anti-HLA antibodies detected to be associated with disease, the most extensively studied
by CDC as a defining risk factor for graft rejection, improve- being blood groups, enzymes, and serum proteins. To date,
ments in the detection of DSA have nearly eliminated hyper- over 500 diseases have been characterized to have a genetic
acute rejection. Serologic crossmatch remains the gold risk factor associated with HLA; a list of the most significant
standard assay for determining immunologic compatibility, HLA and disease associations is given in Table 23–5.
although the labor-intensive nature, complex interpretation, The exact cause for the association of HLA to disease is
and limited donor and recipient samples preclude perform- unclear. In some cases, it has been documented to result
ing crossmatching for all potential donor and recipient pairs. from presentation of unique pathogenic epitopes to T cells
The strategy of virtual crossmatching, or selecting donor- by classical HLA molecules.78,79 In other cases, susceptibil-
recipient pairs based upon knowledge of donor HLA type ity may be related to altered immunologic responsiveness
and recipient alloantibody profile, has led to improved organ affected by the other immune-associated genes in the HLA
allocation, directing shared organs to centers with the most complex that are in linkage disequilibrium with the classical
likely compatible matches.74–76 HLA genes. It is also probable that the diseases in question
Virtual crossmatching is not infallible, however, as it is may be a result of both multiple gene interactions and
critically limited by the information used to determine com-
patibility: accurate evaluation of immunologic compatibility
requires full knowledge for donor HLA type and recent Table 23–5 Significant HLA Disease
alloantibody profile of the recipient. Knowledge of the
Associations
donor organ HLA type is often limited to HLA-A, HLA-B,
and HLA-DR antigens, while multiplex alloantibody testing Disease HLA
is most commonly limited to anti-HLA antibodies, ignoring Carbamazepine-induced B*15:02
the contribution of allotypic antibodies against MICA and Stevens-Johnson syndrome
other alloantigens. This reinforces the necessity of serologic
Ankylosing spondylitis B*27:01, B*27:04, B*27:05
or flow crossmatch with donor cells being performed prior
to any solid organ transplantation to minimize the risk of Reiter’s syndrome B*27
hyperacute rejection.
Anterior uveitis B*27
Advanced Concepts: Clinical Behçet’s disease B*51
Significance of the HLA System
Abacavir hypersensitivity B*57:01
The identification of HLA antigens was driven by their Rheumatoid arthritis DRB1*04
potential clinical application to transplantation. The HLA
system is still of primary clinical importance in transplanta- Type 1 diabetes DRB1*03:01–DQB1*02:01
tion, but it has recently become of great interest to individ- DRB1*04–DQB1*03
uals in the field of human genetics and to investigators of
disease associations. Multiple sclerosis DRB1*15:01

Narcolepsy DQB1*06:02
Paternity
Celiac disease DQA1*05:05, DQA1*05:01–
Relationship testing involves analyzing genetic markers from DQB1*02:01
the mother, child, and alleged father to determine whether DQA1*03:01–DQB1*03:02
the tested man could be the biological father of a child. To
6888_Ch23_497-514 29/10/18 11:35 AM Page 508

508 PART IV Leukocyte Antigens and Relationship Testing

environmental factors. Most commonly, HLA genotyping is with TRALI.48 The current model of TRALI suggests that
used to evaluate the likelihood of a disease process. For ex- these allotypic antibodies activate neutrophils in the lung,
ample, in cases of enteropathy, the patient is genotyped for which release cytokine and chemokine mediators that cause
DQB1; the presence of the pathologically associated pulmonary edema.
DQB1*02:01 or DQB1*03:02 alleles suggest that celiac dis- Suspected cases of TRALI are typically evaluated by ana-
ease may be a potential explanation. Conversely, since lyzing the donor blood product for anti-HLA and anti-HNA
nearly all (>98%) cases of celiac disease are associated with antibodies. However, the diagnosis of TRALI is primarily on
those DQB1 alleles, their absence in a patient would make clinical symptoms correlated with a recent transfusion event.
a diagnosis of celiac disease unlikely and direct the clinician While laboratory evaluation is not required to make a diag-
to investigate other possible causes. nosis of TRALI or begin treatment, it is important for con-
firmation, particularly to evaluate the potential risks of
Platelet Transfusion transfusing blood products from donors associated with
TRALI. Demonstration of anti-HLA or anti-HNA antibodies
Transfusion of platelets is a useful treatment modality for pa- will likely not affect treatment decisions regarding a patient
tients with coagulopathy due to excessive bleeding and with symptoms of TRALI, but they are important to deter-
platelet consumption or hematopoietic defects in platelet mine whether the donor associated with the suspected
production. Platelets express HLA class I molecules on the causative blood product should be removed from the donor
cell surface, presenting both a potential source for immu- pool. (Refer to Chapter 17, “Adverse Effects of Blood Trans-
nization against allogeneic HLA and a target for destruction fusion,” for more information on TRALI.)
by alloantibodies in the recipient.14,80 Alloimmunization to
HLA can result in refractoriness, or failure to achieve a clin- Hematopoietic Stem Cell Transplantation
ically relevant increase in platelet count 1 hour after platelet
transfusion, to platelet transfusions. Considering the highly Hematopoietic stem cell transplantation (HSCT), often
polymorphic nature of the HLA system, it is impossible to referred to as bone marrow transplantation, refers to the
obtain sufficient numbers of HLA-typed donors to provide transplantation of multipotent hematopoietic stem cells
HLA-matched platelets for all alloimmunized patients. How- (HSCs). Under normal conditions, HSCs in the bone mar-
ever, selection of donors that do not express HLA molecules row continuously generate red blood cells, leukocytes, and
against which a recipient has anti-HLA antibodies can effec- platelets. However, under certain conditions, including
tively provide hemostasis for refractory patients.80 HLA hematologic malignancies, congenital and acquired aplasias,
matching is not an absolute guarantee for platelet survival, and after intense radiation and chemotherapy, the bone mar-
as poor transfusion results are sometimes obtained despite a row’s hematopoietic capability is compromised. In these in-
perfect HLA match. In these cases sensitization to non-HLA stances it is beneficial to transplant HSCs, either collected
antigens, such as human platelet-specific antigens, may be from the patient prior to bone marrow ablation (autologous
the cause of platelet refractoriness. HSCT) or from a healthy donor (allogeneic HSCT) to recon-
stitute normal hematopoietic function.82 Allogeneic HSCT
Transfusion-Related Acute Lung Injury enables reconstitution with fully functioning, healthy HSCs,
replacing those compromised by malignancy or metabolic
TRALI is the clinical syndrome of acute lung injury (ALI) defects, with demonstrated benefit in immune-mediated
that develops with a clear temporal relationship to transfu- antitumor function (graft-versus-leukemia [GVL] disease).
sion in patients with or without alternate risk factors for However, it has adverse consequences, most notably the de-
ALI.48 TRALI manifests as fever, hypoxemia, and pulmonary velopment of graft-versus-host disease (GVHD). GVHD is
edema occurring acutely (within 6 hours) following trans- directly related to the degree of alloantigenic mismatch,
fusion of blood components. The phenomenon of respiratory with the classical HLA genes being the most essential deter-
distress following transfusion has been observed for 60 years, minants of compatiblity.10
although TRALI has only been recognized as a distinct clin- Current guidelines suggest that the optimal HSCT donor
ical entity since 1985. TRALI is the most severe adverse out- should match the recipient at the allele-level (high-
come from transfusion, with estimates of prevalence ranging resolution or two-field) genotype for HLA-A, -B, -C, -DRB1,
from 0.02% to 0.16% of patients receiving transfusions. and -DQB1.33 The search for a potential HSC donor often
Treatment is supportive, with supplemental oxygen and res- begins by testing the patient’s siblings, which due to linkage
piratory support if necessary. Most cases resolve within of the HLA genes in the HLA locus, have a 25% chance of
96 hours, although a mortality rate of 5% to 10% is reported. inheriting the same maternal and paternal haplotypes as
TRALI is strongly associated with the presence of allotypic the patient and thus being a match. However, only about
antibodies present in donor blood products. Antibodies 33% of patients have an HLA-matched sibling available for
against HLA class I and class II molecules have been found in HSC donation.33 This necessitates searching for unrelated
50% to 89% of products associated with TRALI.48,81 Addition- individuals who might be a suitable HSC donor. In re-
ally, antibodies to HNA-1a, HNA-1b, HNA-2a, and HNA-3a, sponse to this, the National Marrow Donor Program
alloantigens found specifically on neutrophils, have been (NMDP) was formed with cosponsorship of the AABB,
described in as many as 72% of blood products associated American Red Cross, and the Council of Community Blood
6888_Ch23_497-514 29/10/18 11:35 AM Page 509

Chapter 23 The HLA System 509

Centers to establish a searchable registry of informed, (2) immunosuppressive therapy. Immunosuppressive agents
willing, and medically suitable HLA-typed volunteers for such as azathioprine, prednisone, thymoglobulin, cyclosporine,
potential HSC donation. The NMDP currently operates the and tacrolimus are used to diminish the destructive immuno-
Be The Match registry of volunteer donors and umbilical logic responses to the graft. These agents are nonselective and
cord blood units in the United States. It is the world’s carry risks of serious side effects, especially life-threatening in-
largest hematopoietic cell registry, listing more than fection. To reduce exposure to immunosuppressive agents, as
13.5 million individuals and 225,000 umbilical cord blood well as to avoid situations where immunosuppression cannot
units to facilitate over 74,000 matches between patients prevent rejection, an effort is made to minimize graft foreignness
needing HSCT and potential HSC donors.83 (Refer to Chap- through matching of donor and recipient alloantigens.
ter 19, “Cellular Therapy in the Transplant Center,” for Antigen disparities that most influence graft rejection in-
more information on HSCT) clude the ABO blood group antigens and the HLA antigens.
Because of the continuous difficulty of finding well- Traditionally, ABO blood group compatibility between donor
matched donors for HSCT, HSCs from umbilical cord and recipient has been considered an absolute requirement
blood are an accepted alternative to HSCT when HLA mis- for successful kidney transplantation. The barrier presented
matches between donor and recipient are unavoidable.33 by ABO blood groups is due to the ubiquitous expression of
Umbilical cord blood contains reduced numbers of mature ABO antigens on the cell surface and the universal presence
T lymphocytes, which reduces the risk for GVHD and fa- of isoagglutinins against nonself ABO antigens. However,
cilitates transplantation across HLA-mismatch barriers. strategies to reduce isoagglutinin titers by immunosuppres-
However, umbilical cord blood typically is limited by total sion and plasma exchange therapy can enable successful
cells available, with insufficient HSCs to effectively recon- transplantation across ABO blood group barriers.88
stitute hematopoietic function from a single unit. Thus, HLA compatibility in solid organ transplantation can be
umbilical cord blood products from multiple donors (most considered in two ways: (1) the degree of matching between
commonly two) must be used for successful HSCT.84 Due the donor and recipient and (2) the avoidance of donors with
to the fact that it can take a long time to identify an unre- antigens to which the recipient has preexisting anti-HLA
lated donor via a registry and the inherent problem of ob- antibodies (DSA). As mentioned above, matching the donor
taining enough progenitor cells from umbilical cord blood, and recipient for HLA, particularly HLA-A, -B, and -DR,
a third source of stem cells for allogeneic HSCT is partially results in the best outcomes with the lowest incidence of im-
HLA-mismatched or haploidentical related donors.85 Be- mune rejection.60 However, due to the considerable diversity
cause any patient shares exactly one HLA haplotype with of HLA in the population, less than 10% of patients are able
each biological parent or child and there is a 50% chance to get a kidney allograft matched for HLA-A, -B, and -DR. In
that a sibling will share one HLA haplotype, an eligible most cases, donor selection is made to avoid incompatibility
HLA-haploidentical donor can be identified rapidly in between preexisting anti-HLA antibodies and donor HLA
nearly all cases. However, the HLA mismatches in hap- antigens. While immunosuppressive therapy is effective in
loidentical HSCT impart an increased risk of graft rejection inhibiting the activation of T and B lymphocytes to initiate
and severe GVHD, requiring either extensive manipulation new alloimmune responses, it has limited ability to block the
of the graft by removal of T lymphocytes or aggressive effects of preexisting alloantibodies to mediate AMR.
post-transplant immunosuppression.86 Cellular crossmatch testing is the gold standard in evalu-
The choice between these three graft sources, unrelated ating immunologic compatibility in solid organ transplanta-
matched HSCs, umbilical cord blood, and HLA-haploidentical, tion.50,52 This enables direct assessment of the presence of
and the intensity of conditioning must balance other consid- antibodies in recipient serum that can bind donor cells.
erations. These include the patient’s age, weight, medical con- However, the development of advanced approaches such as
dition, the biological characteristics of the cancer being treated, SAB to identify anti-HLA antibodies combined with improve-
and the relative quality and compatibility of the graft.33 ments in HLA genotyping has enabled prediction of cellular
crossmatch results—a process referred to as the virtual cross-
Kidney Transplantation match.74–76 The virtual crossmatch is not infallible, with im-
portant limitations due to interpretive considerations in
Transplantation of kidneys, heart, lungs, liver, or pancreas is antibody testing and limited data for evaluating non-HLA
a curative therapy for organ failure. Kidney transplantation antibodies. However, the virtual crossmatch can be invalu-
is preferred over dialysis in treating patients with chronic able for situations in which the cellular crossmatch provides
renal failure because it is more cost-effective and it usually equivocal results, such as the presence of autoantibodies. In
returns patients to a state of relatively normal health.87 most cases, these strategies of cellular and virtual crossmatch
Over 16,000 kidney transplants are performed in the United are used in combination, with virtual crossmatch serving
States each year.60 The best graft survival rates occur when to facilitate organ sharing between centers and cellular
kidneys are obtained from HLA-identical, ABO-compatible crossmatch providing a high degree of confidence to ensure
siblings, but such donors are rarely available. In kidney immunologic compatibility.
transplantation, as well as other solid organ transplants, two Histocompatibility testing is also important after kidney
general strategies are used to minimize immunologic rejec- transplantation. Improvements in donor-recipient match-
tion of the allograft: (1) reduction of graft “foreignness” and ing and immunosuppressive regimens have reduced the
6888_Ch23_497-514 29/10/18 11:35 AM Page 510

510 PART IV Leukocyte Antigens and Relationship Testing

incidence of kidney allograft rejection within the first year potentially significant clinical consequences.93 While it is
after transplant to less than 15%.60 However, immunologic not currently standard practice to perform anti-HLA anti-
rejection remains the primary cause of long-term graft loss. body testing and crossmatching for liver transplants, recent
Rejection can be either cellular, predominantly driven by data suggest that identification of DSA can be useful in
T lymphocytes, or humoral, driven primarily by antibodies, diagnosis of post-transplant graft dysfunction and can help
or a mixture of both.87 For antibody-mediated rejection, a direct treatment.94
principle component of the diagnosis is the identification of
anti-HLA DSA in the recipient. The presence of anti-HLA Pancreas and Islet Cell Transplantation
antibodies in general is not predictive of rejection, but rather
only the antibodies against HLA molecules expressed by The primary indication for pancreas transplantation is dia-
the donor organ.61–63 Thus, it is essential to consider donor betes. The majority of pancreas transplants performed are
HLA type information when interpreting post-transplant simultaneous pancreas and kidney transplants (81%),
anti-HLA antibody testing. with pancreas following kidney (12%) and pancreas alone
(5%). HLA matching has a major effect on graft survival,
Heart and Lung Transplantation particularly in the pancreas after kidney and pancreas alone
transplants.94 Because of increased risks of myocardial com-
Transplantation of heart and/or lung is performed with the plications with pancreas transplantation, islet cell transplan-
same goal as in kidney transplantation—to replace a failing tation has been used to restore pancreas function.
organ and restore normal function.89,90 In general, heart and
lung transplantation follow the same general principles as United Network for Organ Sharing
with kidney transplant: attempting to reduce foreignness,
avoidance of donors with antigens to which the recipient has In an effort to ensure equitable and useful allocation of do-
preexisting alloantibodies, and use of immunosuppressive nated organs, the UNOS was established in 1986. This or-
therapeutics to inhibit development of immunologic rejec- ganization received the federal contract to operate the
tion. However, due to the relative scarcity of heart and lung national Organ Procurement and Transplantation Network
donors, as well as more stringent requirements to avoid tis- (OPTN) and to develop a scientific and medically sound
sue damage from cold ischemia, the histocompatibility test- organ allocation system. The OPTN is charged with devel-
ing requirements are somewhat different. Determination of oping policies that maximize use of organs donated for trans-
immunologic compatibility and the decision to proceed with plantation, ensuring quality of care for transplant patients,
transplantation is often made using pretransplant anti-HLA and addressing medical and ethical issues related to organ
antibody testing and virtual crossmatch strategies, with cel- transplantation in the United States.
lular crossmatch performed perioperatively to identify On December 14, 2014, a modified Kidney Allocation
unexpected immunologic challenges.91 Even though the de- System (KAS) was implemented by UNOS due to inconsis-
cision to transplant is made using the virtual crossmatch, the tencies in the makeup of HLA-typed target cell panels used
cellular crossmatch still provides essential information as it across the country. One of its major goals was to prioritize
can identify DSA that require immediate therapeutic Deceased Donor (DD) allocation to candidates with calcu-
intervention to avoid rejection. lated PRAs (cPRAs) of 100% by awarding 202.1 points,
which would catapult these patients to the top of every
Liver Transplantation match run. This meant that many kidneys procured and
placed locally would now be shipped for transplant into pa-
Liver transplantation is a successful therapeutic modality tients with cPRA values of 99% and 100%. That such a rev-
for more than 7,000 patients with end-stage liver disease olutionary change in the national organ allocation policy
each year.92 Immunologic factors in recipient/donor match- could even be considered is testament to the entire trans-
ing for liver transplantation and recipient presensitization plant community that recognized a particular group of
have largely been ignored in the past because of the liver’s patients (the very highly sensitized) were selectively disad-
unique abilities to regenerate in response to tissue damage. vantaged with regard to receiving a transplant and to ad-
However, it is becoming increasingly evident that antibody- vances in histocompatibility testing that allowed more
mediated rejection does occur in liver transplantation, with reliable predictions of donor-recipient compatibility.
6888_Ch23_497-514 29/10/18 11:35 AM Page 511

Chapter 23 The HLA System 511

SUMMARY CHART
 HLA molecules are the molecules responsible for pre-  The diversity of HLA molecules results in robust stim-
senting antigens to T lymphocytes. There are two ulation of the immune system by allogeneic cells.
types: class I (HLA-A, -B, and -C), which presents anti- Alloimmune responses can be mediated by T lympho-
gens to CD8+ T cells, and class II (HLA-DR, -DQ, and cytes as well as B lymphocytes and the antibodies they
-DP), which presents antigens to CD4+ T cells. produce.
 The genes encoding HLA molecules are clustered on  Techniques of histocompatibility testing include HLA
the short arm of chromosome 6 in the HLA region. Class genotyping, HLA antibody detection and identifica-
I HLAs are encoded by highly polymorphic HLA-A, -B, tion, and crossmatching. Improvements in HLA geno-
and -C genes. Class II genes are encoded by the highly typing and antibody identification have enabled
polymorphic HLA-DRB1, -DQB1, and -DPB1 genes, integration of these data to perform “virtual” cross-
which encode a β chain that pairs with the less poly- matches to predict immunologic compatibility.
morphic α chains encoded by HLA-DRA1, -DQA1, and  Matching of HLA genes between donor and recipient
-DPA1, respectively. is essential for hematopoietic stem cell transplantation.
 The HLA haplotype represents the association of the It is beneficial, but not essential, for solid organ trans-
HLA alleles on a single chromosome. Inheritance of plantation.
the HLA genes typically occurs through inheritance of  Anti-HLA antibodies can mediate immunologic de-
haplotypes. struction of transfused platelets, transplant HSCs, and
 HLA genes are extremely diverse, with thousands of transplanted organs. Pre-transplant testing via cellular
protein-coding genetic variants for each of the genes. and virtual crossmatching is essential to avoid im-
A standardized nomenclature exists to describe this munologically incompatible transplantation and avoid
variation. HLA genes are commonly referred to by their antibody-mediated rejection.
allele group (low-resolution) and specific allele (high-
resolution) typings.

Review Questions 5. Why is HLA matching not feasible in cardiac transplan-


tation?
1. The HLA genes are located on which chromosome? a. No HLAs are present on cardiac cells
a. 2 b. No donors ever have HLA antibodies
b. 4 c. Total ischemic time is too long
c. 6 d. Total ischemic time is too short
d. 8 6. DR52 molecules are the product of which alleles?
2. The majority of HLA antibodies belong to what im- a. DRA and DRB1
munoglobulin class? b. DRA and DRB3
a. IgD c. DRA and DRB4
b. IgE d. DRA and DRB5
c. IgG 7. What is the molecular technique that detects undefined
d. IgM alleles?
3. What is the test of choice for HLA antigen testing? a. Restriction fragment length polymorphism
a. Agglutination b. Sequence-specific primer typing
b. Molecular c. Sequence-specific oligonucleotide typing
c. Cytotoxicity d. Direct nucleotide sequencing
d. ELISA 8. What represents the association of the alleles on the two
4. Of the following diseases, which one has the highest rel- C6 chromosomes as determined by family studies?
ative risk in association with an HLA antigen? a.Haplotype
a. Ankylosing spondylitis b.Genotype
b. Juvenile diabetes c.Phenotype
c. Narcolepsy d.Xenotype
d. Rheumatoid arthritis
6888_Ch23_497-514 29/10/18 11:35 AM Page 512

512 PART IV Leukocyte Antigens and Relationship Testing

9. Which type of HSCT can be performed within a rela- 16. Honger G, Hopfer H, Arnold ML, Spriewald BM, Schaub S,
tively short period of time? Patrizia A. Pretransplant IgG subclasses of donor-specific
human leukocyte antigen antibodies and development of
a. Matched unrelated antibody-mediated rejection. Transplantation. 2011;91:41-7.
b. Cord blood 17. Legrand L, Dausset J. The complexity of the HLA gene prod-
c. Autologous uct: possible evidence for a “public” determinant common
d. HLA haploidentical to the first and second HLA series. Transplantation. 1975;
19:177-80.
10. The SAB describes the amount of bound antibody on 18. Rodey G, Fullet T. Preliminary report of the American Society
each bead as: for Histocompatibility and Immunogenetics phase I workshop
on HLA class I public epitopes. Transpl Proc. 1987;19:872-3.
a. MCS 19. Rodey GE, Fuller TC. Public epitopes and the antigenic struc-
b. MFI ture of HLA molecules. Crit Rev Immunol. 1987;7:229-67.
c. CDC 20. Belvedere M, Mattiuz PL, Curtoni ES. An antibody cross-reacting
d. AHG with LA and four antigens of the HL-A system. Immunogenetics.
1975:1:538-48.
21. Scalamogne M, Mercuriali F, Pizzi C, Sirchia G. Crossreactivity
between the first and second segregant series of the HLA
References system. Tissue Antigens. 1976;7:125-7.
22. Kostyu DD, Cresswell P, Amos DB. A public HLA antigen
1. Dausset J. Leukoagglutinins: leukoagglutinins and blood associated with HLA-A9, Aw32, and Bw4. Immunogenetics.
transfusion. J Vox Sang. 1954;4:190-8. 1990;10:433-42.
2. Dausset J. Iso-leuco-anticorps. Acta Haematol. 1958;20:156. 23. Muller C, Ziegler A, Muller G, Schunter F, Wernet P. Monoclonal
3. Bjorkman PJ, Saper MA, Samraoui B, Bennett WS, Strominger antibody (Tu 48) defining alloantigenic class I determinants
JL, Wiley DC. Structure of the HLA class I histocompatibility specific for HLA-Bw4 and HLA-Aw23, -Aw24 as well as -Aw32.
antigen, HLA-A2. Nature. 1987;329:506-12. Human Immunol. 1982;5:269-81.
4. Brown JH, Jardetzky TS, Gorga JC, Stern LJ, Urban RG, 24. McMichael AJ, Parham P, Rust N, Brodsky F. A monoclonal
Strominger JL, et al. Three-dimensional structure of the human antibody that recognizes an antigenic determinant shared by
class II histocompatibility antigen HLA-DR1. Nature. 1993; HLA-A2 and B17. Hum Immunol. 1980;1:121-9.
364:33-9. 25. Claas F, Castelli-Visser R, Schreuder I, van Rood J. Alloantibodies
5. Meuer SC, Schlossman SF, Reinherz EL. Clonal analysis of to an antigenic determinant shared by HLA-A2 and B17. Tissue
human cytotoxic T lymphocytes: T4+ and T8+ effector T cells Antigens. 1982;19:388-91.
recognize products of different major histocompatibility com- 26. Duquesnoy RJ. HLAMatchmaker: a molecularly based algo-
plex regions. Proc Nat Acad Sci USA. 1982;79:4395-9. rithm for histocompatibility determination. I. Description of
6. Li Y, Yin Y, Maurizza RA. Structural and biophysical insights the algorithm. Human Immunol. 2002;63:339-52.
into the role of CD4 and CD8 in T cell activation. Front 27. Duquesnoy RJ, Marrari M. HLAMatchmaker: a molecularly
Immunol. 2013;4:206. based algorithm for histocompatibility determination. II.
7. Steimle V, Siegrist CA, Mottet A, Lisowska-Grospierre B, Verification of the algorithm and determination of the relative
Mach B. Regulation of MHC class II expression by interferon- immunogenicity of amino acid triplet-defined epitopes. Human
gamma mediated b the transactivator gene CIITA. Science. Immunol. 2002;63:353-63.
1994;265:106-9. 28. El-Awar N, Cook D, Terasaki PI. HLA class I epitopes: A and B
8. Falk K, Rotzschke O, Stevanovic S, Jung G, Rammensee HG. loci. Clin Transpl. 2006:79-94.
Allele-specific motifs revealed by sequencing of self-peptides 29. Akaza T, El-Awar N, Nguyen A, Kitawaki J, Terasaki PI. HLA
eluted from MHC molecules. Nature. 1991;351:290-6. class I epitopes: C-locus. Clin Tranpl. 2006:95-102.
9. Robinson J, Halliwell JA, Hayhurst JH, Flicek P, Parham P, 30. Cai J, Kohanof S, Terasaki PI. HLA-DR antibody epitopes. Clin
Marsh SGE. The IPD and IMGT/HLA database: allele variant Tranpl. 2006:103-14.
databases. Nucl Acid Res. 2015;43:D423-31. 31. Deng CT, Cai J, Tarsitani C, El-Awar N, Lachmann N, Ozawa
10. Flomenberg N, Baxter-Lowe L, Confer D, Fernandez-Vina M, M. HLA class II DQ epitopes. Clin Transpl. 2006:115-22.
Filipovich A, Horowitz M, et al. Impact of HLA class I and 32. Kramer CSM, Roelen DL, Heidt S, Claas FHJ. Defining the im-
class II high-resolution matching on outcomes of unrelated munogenicity and antigenicity of HLA epitopes is crucial
donor bone marrow transplantation: HLA-C mismatching for optimal epitope matching in clinical renal transplantation.
is associated with a strong adverse effect on transplantation Tissue Antigens. 2017;1-12.
outcome. Blood. 2004;104:1923-30. 33. Howard CA, Fernandex-Vina M, Appelbaum FR, Confer DL,
11. Shiina T, Hosomichi K, Inoko H, Kulski JK. The HLA genomic Devine SM, Horowitz MM, et al. Recommendations for donor
loci map: expression, interaction, diversity and disease. J Human human leukocyte antigen assessment and matching for allo-
Genet. 2009;54:15-39. geneic stem cell transplantation: consensus opinion of the
12. Huttley GA, Smith MW, Carrington M, O’Brien SJ. A scan for Blood and Marrow Transplant Clinical Trials Network (BMT
linkage disequilibrium across the human genome. Genetics. CTN). Biol Blood Marrow Transplant. 2015;21:4-7.
1999;152:1711-22. 34. Organ Procurement and Transplantation Network [Internet].
13. Kissmeyer-Nielson, F,Olsen S, Petersen VP, Fjeldborg O Hyper- Richmond (VA): OPTN [2017]. OPTN policies: Policy 4: His-
acute rejection of kidney allografts associated with preexisting tocompatibility. Available from: https://optn.transplant.hrsa
humoral antibodies against donor cells. Lancet. 1966;1:662. .gov/media/1200/optn_policies.pdf.
14. Yankee RA, Grumet FC, Rogentine GN. Platelet transfusion 35. Wordsworth BP, Allsopp CE, Young RP, Bell JI. HLA-DR typing
therapy—the selection of compatible platelet donors for refrac- using DNA amplification by the polymerase chain reaction and
tory patients by lymphocyte HLA-A typing. N Engl J Med. sequential hybridization to sequence-specific oligonucleotide
1969;281:1208-12. probes. Immunogenetics. 1990;32:413-8.
15. Scornik JC, Meier-Kriesche HU. Blood transfusions in organ 36. Olerup O, Zetterquist H. HLA-DR typing by PCR amplification
transplant patients: mechanisms of sensitization and implica- with sequence-specific primers (PCR-SSP) in 2 hours: an
tions for prevention. Am J Transplant. 2011;11:1785-91. alternative to serological DR typing in clinical practice including
6888_Ch23_497-514 29/10/18 11:35 AM Page 513

Chapter 23 The HLA System 513

donor-recipient matching in cadaveric transplantation. Tissue human leukocyte antigen antibody specificities. Transplantation.
Antigens. 39:225-35. 2003;75:43-9.
37. Sanger F, Nicklen S, Coulson A. DNA sequencing with chain ter- 58. El-Awar N, Lee J, Terasaki PI. HLA antibody identification with
minating inhibitors. Proc Natl Acad Sci USA. 1977;74:5463-7. single antigen beads compared to conventional methods.
38. Santamaria P, Boyce-Jacino M, Lindstrom A, Barbosa J, Faras Human Immunol. 2005;66:989-97.
A, Rich S. HLA class II “typing”: direct sequencing of DRB, 59. Cecka JM, Kucheryavaya AY, Reinsmoen NL, Leffell MS. Cal-
DQB, and DQA genes. Human Immunol. 1992;33:69-81. culated PRA: initial results show benefits for sensitized patients
39. Santamaria P, Lindstrom AL, Boyce-Jacino MT, Myster SH, and a reduction in positive crossmatches. Am J Transplant.
Barbosa JJ, Faras AJ, et al. HLA class I sequence-based typing. 2001;11:719-24.
Human Immunol. 1993;37:39-50. 60. Hart A, Smith JM, Skeans MA, Gustafson SK, Stewart DE,
40. Metzker ML. Sequencing technologies–the next generation. Cherikh WS, et al. OPTN/SRTR 2015 annual data report:
Nat Rev Genet. 2010;11:31-46. kidney. Am J Transplant. 2017;17S1:21-116.
41. Lind C, Ferriola D, Mackiewicz K, Heron S, Rogers M, Slavich L, 61. Patel AM, Pancoska C, Mulgaonkar S, Weng FL. Renal trans-
et al. Next-generation sequencing: the solution for high- plantation in patients with pre-transplant donor-specific
resolution, unambiguous human leukocyte antigen typing. antibodies and negative flow cytometry crossmatches. Am J
Human Immunol. 2010;71:1033-42. Transplant. 2007;7:2371-7.
42. Holcomb CL, Hoglund B, Anderson MW, Blake LA, Bohme I, 62. Amico P, Honger G, Mayr M, Steiger J, Hopfer H, Schaub S.
Egholm M, et al. A multi-site study using high-resolution HLA Clinical relevance of pretransplant donor-specific HLA anti-
genotyping by next generation sequencing. Tissue Antigens. bodies detected by single-antigen flow-beads. Transplantation.
2011;77:206-17. 2009;87:1681-8.
43. Wang C, Krishnakumar S, Wilhelmy J, Babrzadeh F, Stepanyan 63. Dunn TB, Noreen H, Gillingham K, Maurer D, Ozturk D,
L, Su LF, et al. High-throughput, high-fidelity HLA genotyping Pruett TL, et al. Revisiting traditional risk factors for rejection
with deep sequencing. Proc Natl Acad Sci USA. 2012;109: and graft loss after kidney transplantation. Am J Transplant.
8676-81. 2011;11:2132-43.
44. Hosomichi K, Jinam TA, Mitsunaga S, Nakaoka H, Inoue I. 64. Liu C, Wetter L, Pang S, Phelan DL, Mohanakumar T, Morris
Phase-defined complete sequencing of the HLA genes by next- GP. Cutoff values and data handling for solid-phase testing for
generation sequencing. BMC Genomics. 2013;14:355. antibodies to HLA: effects on listing unacceptable antigens for
45. Lange V, Bohme I, Hofmann J, Lang K, Sauter J, Schone B, thoracic organ transplantation. Human Immunol. 2012;73:
et al. Cost-efficient high-throughput HLA typing by MiSeq am- 597-604.
plicon sequencing. BMC Genomics. 2014;15:63. 65. Tambur AR, Herrera ND, Haarberg KM, Cusick MF, Gordon
46. Smith JD, Danskine AJ, Laylor RM, Rose ML, Yacoub MH. The RA, Leventhal JR, et al. Assessing antibody strength: compari-
effect of panel reactive antibodies and the donor specific cross- son of MFI, C1q, and titer information. Am J Transplant.
match on graft survival after heart and heart-lung transplanta- 2015;15:2421-30.
tion. Transplant Immunol. 1993;1:60-5. 66. Zachary AA, Scholander JT, Houp JA, Leffell MS. Using real
47. Gammie JS, Pham SM, Colson YL, Kawai A, Keenan RJ, Weyant data for a virtual crossmatch. Human Immunol. 2009;70:
RJ, et al. Influence of panel-reactive antibody on survival and 574-9.
rejection after lung transplantation. J Heart Lung Transplant. 67. Guidicelli G, Anies G, Bachelet T, Dubois V, Moreau JF, Merville P,
1997;16:408-15. et al. The complement interference phenomenon as a course
48. Popovski MA, Moore SB. Diagnostic and pathogenic consider- for sharp fluctuations of serum anti-HLA antibody strength in
ations in transfusion-related acute lung injury. Transfusion. kidney transplant patients. Transpl Immunol. 2013;29:17-21.
1985;25:573-7. 68. Schwaiger E, Wahrmann M, Bond G, Eskandary F, Bohmig GA.
49. Mittal KK, Mickey MR, Singal DP, Terasaki PI. Serotyping for Complement component C3 activation: the leading cause of
homotransplantations. 18. Refinement of microdroplet lym- the prozone phenomenon affecting HLA antibody detection on
phocytotoxicity test. Transplantation. 1968;6:913-27. single-antigen beads. Transplantation. 2014;97:1279-85.
50. Patel R, Terasaki PI. Significance of the positive crossmatch test 69. Reed EF, Rao P, Zhang Z, Gebel H, Bray RA, Guleria I, et al.
in kidney transplantation. N Engl J Med. 1969;280:735-9. Comprehensive assessment and standardization of solid phase
51. Fuller TC, Phelan D, Gebel HM, Rodey GE. Antigenic speci- multiplex-bead arrays for the detection of antibodies to HLA.
ficity of antibody reactive in the antiglobulin-augmented lym- Am J Transplant. 2013;13:1859-70.
phocytotoxicity test. Transplantation. 1982;34:24-9. 70. Tait BD, Susal C, Gebel HM, Nickerson PW, Zachary AA,
52. Iwaki Y, Lau M, Cook DJ, Takemoto S, Terasaki PI. Cross- Claas FH, et al. Consensus guidelines on the testing and clinical
matching with B and T cells and flow cytometry. Clin Transpl. management issues associated with HLA and non-HLA anti-
1986;277-84. bodies in transplantation. Transplantation. 2103;95:19-47.
53. Bearden CM, Agarwal A, Book BK, Sinder RA, Gebel HM, 71. Wahrmann M, Exner M, Haidbauer B, Schillinger M, Regele M,
Bray RA, et al. Pronase treatment facilitates alloantibody flow Kormoczi G, et al. [C4d]FlowPRA screening—a specific assay
cytometric and cytotoxic crossmatching in the presence of for selective detection of complement-activating anti-HLA
rituximab. Human Immunol. 2004;65:803-9. alloantibodies. Human Immunol. 2005;66:526-34.
54. Hetrick SJ, Schillinger KP, Zachary AA, Jackson AM. Impact 72. Chen G, Sequeira F, Tyan DB. Novel C1q assay reveals a clini-
of pronase on flow cytometric crossmatch outcome. Human cally relevant subset of human leukocyte antigen antibodies in-
Immunol. 2011;72:330-6. dependent of immunoglobulin G strength on single antigen
55. Pei R, Wang G, Tarsitani C, Rojo S, Chen T, Takemura S, Liu A, beads. Human Immunol. 2011;72:849-58.
et al. Simultaneous HLA class I and class II antibodies 73. Claisse G, Absi L, Cognasse F, Alamartine E, Mariat C, Malliard
screening with flow cytometry. Human Immunol. 1998;59: N. Relationship between mean fluorescence intensity and
313-22. C1q/C3d-fixing capacities of anti-HLA antibodies. Human
56. Appel JZ 3rd, Hartwig MG, Cantu E 3rd, Palmer SM, Reinsmoen Immunol. 2017;78:336-41.
NL, Davis RD. Role of flow cytometry to define unacceptable 74. Bray RA, Nolen JD, Larsen C, Pearson T, Newell KA, Kokko
HLA antigens in lung transplant recipients with HLA-specific KA, et al. Transplanting the highly sensitized patient: the
antibodies. Transplantation. 2006;81:1049-57. Emory algorithm. Am J Transplant. 2006;6:2307-15.
57. Pei R, Lee JH, Shih NJ, Chen M, Terasaki PI. Single human leuko- 75. Vaidya S, Partlow D, Susskind B, Noor M, Barnes T, Gugliuzza K.
cyte antigen flow cytometry beads for accurate identification of Prediction of crossmatch outcome of highly sensitized patients
6888_Ch23_497-514 29/10/18 11:35 AM Page 514

514 PART IV Leukocyte Antigens and Relationship Testing

by single and/or multiple antigen bead Luminex assay. Trans- transplantation for hematologic malignancies. Biol Blood
plantation. 2006;82:1524-8. Marrow Transplant. 2017;23:882-96.
76. Bielmann D, Honger G, Lutz D, Mihatsch MJ, Steiger J, Schaub S. 85. Zuckerman T, Rowe JM. Alternative donor transplantation in
Pretransplant risk assessment in renal allograft recipients using acute myeloid leukemia: which source and when? Curr Opin
virtual crossmatching. Am J Transplant. 2007;7:626-32. Hematol. 2007;14:152-61.
77. Polesky HF. Impact of molecular (DNA) testing on determi- 86. Dey BR, Spitzer TR. Current status of haploidentical stem cell
nation of parentage. Arch Pathol Lab Med. 1999;123: transplantation. Br J Hematol. 2006;135:423-37.
1060-2. 87. Nankivell BJ, Alexander SI. Rejection of the kidney allograft.
78. Hovhannisyan Z, Weiss A, Martin A, Wiesner M, Tollefsen S, 2010;363:1451-62.
Yoshida K, et al. The role of HLA-DQ8 beta57 polymorphism 88. Okumi M, Toki D, Nozaki T, Shimizu T, Shirakawa H,
in the anti-gluten T-cell response in coeliac disease. Nature. Omoto K, et al. ABO-incompatible living kidney transplant:
2008;456:534-8. evolution of outcomes and immunosuppressive management.
79. Illing PT, Vivian JP, Dudek NL, Kostenko L, Chen Z, Bharadwaj Am J Transplant. 2016;16:886-96.
M, et al. Immune self-reactivity triggered by drug-modified 89. Colvin M, Smith JM, Skeans MA, Edwards LB, Uccellini K,
HLA-peptide repertoire. Nature. 2012;486:554-8. Snyder JJ, et al. OPTN/SRTR 2015 annual data report: heart.
80. Duquesnoy RJ, Filip DJ, Rodey GE, Rimm AA, Aster RH. Suc- Am J Transplant. 2017;17S1:286-356.
cessful transfusion of platelets “mismatched” for HLA antigens 90. Valapour M, Skeans MA, Smith JM, Edwards LB, Cherikh WS,
to alloimmunized thrombocytopenic patients. Am J Hematol. Uccellini K, et al. OPTN/SRTR 2015 annual data report: lung.
1977;2:219-26. Am J Transplant. 2017;17S1:357-424.
81. Eder AF, Herron R, Strupp A, Dy B, Notari EP, Chambers LA, 91. Zangwill SD, Ellis TM, Zlotocha J, Jaquiss RD, Tweddell JS,
et al. Transfusion-related acute lung injury surveillance Mussatto KA, et al. The virtual crossmatch—a screening tool
(2003–2005) and the potential impact of the selective use for sensitized pediatric heart transplant recipients. Pediatr
of plasma from male donors in the American Red Cross. Trans- Transplant. 2006;10:38-41.
fusion. 2007;47:599-607. 92. Kim WR, Lake JR, Smith JM, Skeans MA, Schladt DP,
82. Gooley TA, Chien JW, Pergam SA, Hingorani S, Sorror ML, Edwards EB, et al. OPTN/SRTR 2015 annual data report: liver.
Boeckh M, et al. Reduced mortality after allogeneic hematopoi- Am J Transplant. 2017;17S1:174-251.
etic-cell transplantation. N Engl J Med. 2010;363:2091-101. 93. Demetris AJ, Bellamy C, Hubscher SG, O’Leary J, Randhawa PS,
83. D’Souza A, Zhu X. Current uses and outcomes of hematopoietic Feng S, et al. 2016 comprehensive update of the Banff working
cell transplantation (HCT): CIBMTR Summary Slides. Milwau- group on liver allograft pathology: introduction of antibody-
kee (WI): Center for International Blood & Marrow Transplant mediated rejection. Am J Transplant. 2016;16:2816-35.
Research; 2016. Available from: http://www.cibmtr.org. 94. Kandaswamy R, Stock PG, Gustafson SK, Skeans MA, Curry
84. Barker JN, Kurtzberg J, Ballen K, Boo M, Brunstein C, MA, Prentice MA, et al. OPTN/SRTR 2015 annual data report:
Cutler C, et al. Optimal practices in unrelated donor cord blood pancreas. Am J Transplant. 2017;17S1:117-73.
6888_Ch24_515-530 29/10/18 11:34 AM Page 515

Chapter 24
Relationship Testing
Robert W. Allen, PhD*

Introduction RFLP Analysis Nonclassic Situations


History STR Analysis Advantages and Disadvantages of Genetic
Goals of Testing Single Nucleotide Polymorphisms Systems
Criteria for Selection of Genetic Systems Terminology and the Interpretation of Social and Legal Issues
Types of Genetic Systems and Technology Results Accreditation and Quality Issues
Used Exclusion Criteria for Classical Systems Case Studies
Classical Systems: Blood Groups Exclusion Criteria for DNA Systems Case 24-1
Classical Systems: Enzymes and Inclusionary Calculations Case 24-2
Proteins Paternity or Relationship Index Summary Chart
Classical Systems: HLA Probability of Relatedness or Review Questions
DNA Polymorphisms: Repetitive Parentage References
Elements in Genomic DNA Probability of Exclusion Bibliography

OBJECTIVES
1. State the goals of parentage testing.
2. List the criteria used to select a genetic system for parentage testing.
3. Briefly describe the testing technologies used for the different types of genetic systems.
4. Outline the advantages and disadvantages of the different types of genetic systems.
5. Define and give examples of false direct and indirect exclusions.
6. List at least two causes of false exclusions produced using serologic testing and one cause of false exclusion with DNA typing.
7. Define relatedness index, posterior probability of relatedness, and power of exclusion.
8. List organizations involved and resources available in quality improvement for parentage testing laboratories.

Introduction and force them to provide financially and emotionally for


their children. However, the field of parentage testing, or
The frequency with which parentage (or relationship) testing more appropriately now, relatedness testing, has expanded
is performed in the United States has risen to the point that over the past 20 years to include criminal investigations,
in 2010 the AABB Annual Report Summary for Testing listed immigration, and the identification of human remains from
approximately 400,000 tests performed that year (you can disasters, war, or genocide.
explore the relationship testing field on the AABB website in Relationship or parentage testing refers to the testing of
the relationship testing area at aabb.org). The growth of re- genetic markers that are inherited to confirm or refute an al-
lationship testing in the United States has its roots in the leged biological relationship. (Both terms will be used inter-
rapid rise in illegitimacy during the 1960s and 1970s and the changeably throughout this chapter.) Relationship testing has
government’s efforts to identify primarily delinquent fathers relevance for the physician or blood banker in being able to
visualize in real life the somewhat abstract concepts learned
in genetics and perhaps practiced with fruit flies in an under-
*Acknowledgment: Some of the content in this chapter was carried over from earlier
editions and was written by the coauthor for those earlier editions, Chantal R.
graduate laboratory. Genetic disease, transplantation, and
Harrison MD (ret.). even transfusion of specialized blood products may call for

515
6888_Ch24_515-530 29/10/18 11:34 AM Page 516

516 PART IV Leukocyte Antigens and Relationship Testing

basic knowledge and ability in predicting the passage of ge- All these genetic systems were expressed on or in different
netic traits from one generation to the next. Parentage testing cell types (red blood cells [RBCs]or leukocytes) or in serum
is most commonly applied to determine whether a male is (some polymorphic proteins), but they had one important aspect
the biological father of a child, but it can also be applied to in common: the nature of the polymorphisms detected flowed
determining maternity and other questioned kinships. from polymorphisms residing in the genes encoding them. This
group of genetic systems is often termed the classic systems.
History Technological advances developing over the late 1980s and
early 1990s laid the foundation for a revolution in relatedness
The processes currently used to resolve cases of questioned testing, heralded by the description by Jeffreys4 in 1985, and
parentage can be traced back to the statistical analysis pub- soon thereafter by Nakamura et al.,5 of a new type of poly-
lished by Bernstein1 in 1924 demonstrating that the ABO morphism in human DNA: hypervariable minisatellites often
blood group frequency distribution in Austria was most com- referred to as variable number tandem repeats (VNTR).
patible with a three-allele theory. ABO blood group test re- DNA polymorphisms could be revealed using technologies
sults were first used to establish nonpaternity in Vienna in that directly assess the variability of DNA sequence, and over
1926. As other blood groups were described, and their the past 20 or more years they have become the norm.
inheritance established, they were used with the ABO system In addition, there has been a rapid evolution of DNA poly-
for paternity studies. The field of relationship testing ad- morphism testing technologies used for relationship testing
vanced further in 1955 when Smithies2 described the charge and forensic analysis. Early on, polymorphic VNTR systems
polymorphism of haptoglobin, which could be revealed were revealed by treating extracted DNA with restriction en-
through gel electrophoresis; this opened the door to testing zymes, followed by Southern blotting and hybridization to
a new type of genetic polymorphism: electrophoretic vari- labeled VNTR probe(s). This technology, referred to as re-
ability of enzymes and proteins. The field advanced even fur- striction fragment length polymorphism (RFLP) testing,
ther in 1972 when the extreme polymorphism of the human dominated the field for several years. Figure 24–1 is a
leukocyte antigen (HLA) system was demonstrated.3 schematic representation of RFLP analysis of genomic DNA.

Tandem array Figure 24–1. Schematic representation of RFLP


Chromosome homologues. analysis from genomic DNA. Chromosomal DNA ex-
Restriction sites denoted by tracted from a biological specimen (i.e., blood, buccal
vertical arrowheads. swab, or some other tissue) is digested with restriction
Tandem array endonuclease (vertical arrowheads). One pair of homol-
ogous chromosomes is shown that contains a VNTR.
Among the thousands of restriction fragments produced
are the two shown in the figure that harbor the tandemly
repeated sequence (five repeats in one chromosome
Restriction enzyme digestion, agarose and eight repeats in the other). The restriction digest is
gel electrophoresis. Hybridization to electrophoresed in an agarose gel, and the fragments
DNA probe. are transferred to a nylon membrane using Southern
blotting. The membrane is hybridized to a labeled DNA
probe targeting the tandem array, and the specific VNTR
alleles are visualized through autoradiography (if the
probe is radioactively labeled) or through another visu-
alization method (colorimetric or chemiluminescence).
6888_Ch24_515-530 29/10/18 11:34 AM Page 517

Chapter 24 Relationship Testing 517

RFLP testing ceded precedence to polymerase chain re-


action (PCR) technology in the 1990s. Originally, PCR was BOX 24–1
often used to detect polymorphisms in functional genes har- Genetic Markers Tested Serologically in Cases
boring point mutations, termed single nucleotide polymor- of Questioned Parentage
phisms (SNPs), but PCR was soon also used to analyze
RBC Antigens
VNTR-type loci in which the repeated unit was small (i.e.,
four to five nucleotides). Such loci are termed short tandem • ABO
repeat or small tandem repeat (STR) loci. This technology • Rh
represents that which is used by virtually all relationship • MNSs
testing laboratories today. • Kell
• Duffy
Goals of Testing • Kidd
RBC Enzymes and Serum Proteins
The ultimate goal of relationship testing is to confirm or re-
• PGM1
fute a specific biological relationship between two or more
• ACP
individuals whose relationship is in question (usually a child
• ESD
and a father or a mother). When applied to the usual trio
• Hp
(mother, child, and alleged father), the goals are to exclude
• Gc
all falsely accused men and to provide compelling inclusion-
ary evidence if a man is not excluded. Immunoglobulin Allotypes
• Gm
Criteria for Selection of Genetic • Km
Systems • Am
HLA
The following factors are considered when selecting genetic
systems to use for relationship testing: • A and B locus

1. The system should be polymorphic—that is, it should have


multiple alleles, ideally in Hardy-Weinberg equilibrium.
list has been expanded to include a total of 20 STR loci.
2. The inheritance pattern should be well established and
Box 24–2 compares the original CODIS loci to the expanded
follow Mendel’s rules.
sytems. Figure 24–2 shows the chromosomal locations of the
3. The mutation rate should be known and should be low.
most common STR loci in current use. Table 24-1 lists the
4. Possible genotypes should be readily deducible from the
chromosomal locations for each STR locus.
phenotype.
5. Testing methodology should be reliable, reproducible,
and available in more than one laboratory.
BOX 24–2
6. Markers tested should be stable and not affected by envi-
ronmental factors, age, disease, reagents, or methodology STR Systems Included in CODIS
employed. Expanded CODIS STR
7. Databases of allele frequencies should be available for all Original CODIS Systems Systems
ethnic groups that may be tested. D3S1358 D3S1358
8. Allelic distributions should provide a high probability of D5S818 D5S818
excluding an alleged father. D7S820 D7S820
9. Each genetic system should be known to be genetically D8S1179 D8S1179
independent (e.g., no linkage disequilibrium) of all other D13S317 D13S317
D16S539 D16S539
genetic systems used by the laboratory in the test battery. D18S51 D18S51
D21S11 D21S11
Types of Genetic Systems CSF1P0 CSF1P0
and Technology Used FGA FGA
TH01 TH01
Box 24–1 lists the most common classic genetic systems that TPOX TPOX
vWA vWA
were widely used in the past. In the last few years, there has Amelogenin Amelogenin
been an almost complete migration of laboratories to rely on – D1S1656
DNA polymorphism testing using STR systems and PCR – D2S441
technology. Although literally thousands of STR loci exist in – D2S1338
human DNA, the FBI officially recognized 13 STR loci plus – D10S1248
– D12S391
amelogenin (for sex determination) as the “core” loci to be – D19S433
used to create and query the database of convicted offender – D22S1045
DNA profiles (CODIS). Effective January 2017, the CODIS
6888_Ch24_515-530 29/10/18 11:34 AM Page 518

518 PART IV Leukocyte Antigens and Relationship Testing

13 CODIS Core STR Loci experience accumulated. Phenotype determination is based


with Chromosomal Positions on standard RBC agglutination techniques with regulated
reagents and, with some extra built-in quality control steps,
TPOX is indistinguishable from testing performed daily in most
blood banks or transfusion services. This is why, prior to the
D3S1358 introduction of DNA testing methods, the majority of labo-
ratories performing relationship testing were associated with
TH01
D8S1179 a blood bank or a transfusion service. In performing serologic
D5S818 VWA
testing, duplicate testing of each specimen was required, and
FGA D7S820 it was also recommended that testing should be done by two
CSF1PO
individuals using two different sources of antisera. Such test
1 2 3 4 5 6 7 8 9 10 11 12 redundancy ensured the accuracy of test results normally
traceable to interpretive errors and antiserum reagent vari-
AMEL ability. A review of a recent participant summary from the
College of American Pathology, who administers one of the
D13S317 oldest proficiency tests for relationship laboratories, revealed
D16S539 D18S51 D21S11 AMEL
there are no participating laboratories that still utilize red
13 14 15 16 17 18 19 20 21 22 X Y blood cell serology in their testing programs.

Figure 24–2. Chromosomal locations of the STR loci that comprise the original Classical Systems: Enzymes and Proteins
CODIS STR loci. (Courtesy of The National Institute of Standards and Technology
[NIST]). Testing of RBC enzymes and serum proteins is also rarely, if
ever, performed. Polymorphisms in amino acid sequence
leading to electrical charge differences in allelic forms of red
Table 24–1 Chromosomal Locations
blood cell enzymes and serum proteins were a common test
of STR Loci methodology in the 1970s and 1980s. The different allelic
Locus Chromosomal Location forms of an enzyme or protein system were separated based
upon net charge differences using electrophoresis, followed
D2S1338 2q35
by staining. Specialized electrophoretic procedures such as
D3S1358 3p21.31 isoelectric focusing were able to subtype some enzyme and
protein systems, expanding the number of alleles for a
D5S818 5q23.2
marker and, hence, the discriminatory power of the system.
D7S820 7q21.11 Distinguishing enzyme and protein phenotypes was based
upon the number and respective position of protein bands
D8S1179 8q24.13
observed in an electrophoresis gel and through comparison
D13S317 13q31.1 against control specimens whose phenotype was known.
Furthermore, interpretation of the enzyme or protein phe-
D16S539 16q24.1
notype was to be made twice independently, ideally by two
D18S51 18q21.33 different observers.
Because rare variants exist in most systems, laboratories
D19S433 19q12
were encouraged to maintain a file of variants to permit iden-
D21S11 21q21.1 tification of rare phenotypes when they were encountered.
CSF1PO 5q33.1
Classical Systems: HLA
FGA 4q28
The HLA gene complex represents perhaps the most poly-
HUMTHO1 11p15.5 morphic genetic system in the human genome. As discussed
TPOX 2p25.3 in Chapter 23, “The HLA System,” it comprises multiple
linked loci expressed as a number of major and minor poly-
VWA 12p13.31 morphic antigens expressed on leukocytes. In relationship
testing with classic serologic detection of HLA antigens (i.e.,
not detected through molecular methods), the antigens
Classical Systems: Blood Groups expressed by the A and the B loci were primarily targeted by
the serologic testing performed.
Six blood group antigen systems were routinely used in The usual method for revealing the HLA phenotype is
parentage tests in the 1970s and 1980s: ABO, Rh, MNSs, Kell, termed microlymphocytotoxicity. HLA phenotyping depends
Duffy, and Kidd. These systems have been in use for a very on the evaluation of the reactions of live lymphocytes with
long time, and potential pitfalls in interpretation associated a panel of cytotoxic antibodies of known specificity in the
with their use are well known from the vast, worldwide presence of complement. Testing is performed in 60 or
6888_Ch24_515-530 29/10/18 11:34 AM Page 519

Chapter 24 Relationship Testing 519

72 microwell trays preloaded with reagent antisera. It was electrophoresis, Southern blotting, or hybridization of mem-
recommended that a sufficient number of antisera be used brane blots with a labeled DNA probe. Restriction enzymes
so that all HLA-A and HLA-B specificities recognized by recognize specific DNA sequences of four to six bases and
the 1980 HLA Nomenclature Committee of the World cut the double-stranded DNA at that site. These enzymes
Health Organization were reliably identified. Additional have been isolated from bacteria and are named after the
antigens could be tested if appropriate antisera were avail- bacteria from which they were isolated (e.g., EcoRI from
able. The larger the number of antigens that can be de- Escherichia coli RY13; HaeIII from Haemophilus aegyptius).
fined, the more powerful the system becomes—that is, the VNTR phenotypes are defined by the length of the DNA
better it can exclude alleged fathers. Because of the large fragments in base pairs or kilobase pairs (kb), estimated
number of existing antigens, no single tray of anti-HLA through comparison of restriction fragments from samples
antisera can identify all relevant antigens. This is especially to a sizing ladder consisting of DNA fragments of known
true when performing testing with specific racial groups size. Some variability in migration may occur from lane to
inasmuch as some antigens occur more frequently in spe- lane within a gel, and because the resolution of agarose gel
cific racial groups such as African Americans or Asians. electrophoresis is limited, it is recommended that the DNA
When dealing with individuals from these backgrounds, of each alleged parent with each child be mixed and elec-
antisera trays specific for these groups should be selected trophoresed together in a single lane to maximize resolution
when appropriate. and to normalize potential electrophoretic anomalies. Ulti-
Antisera are of human origin (alloantisera) or are mono- mately, the goal of the process is to confirm or refute sharing
clonal (created in the laboratory), and monospecific sera are of alleles between alleged parent and child. Figure 24–3
generally not available for a large number of antigens. HLA illustrates this process.
typing used for parentage, like HLA typing used in support Although RFLP analysis is a suitable technique to reveal
of transplantation or transfusion, requires that each antigen single nucleotide substitution–type polymorphisms (i.e.,
be defined by at least two different operationally monospe- SNPs), it was VNTR-type polymorphisms that were the target
cific sera, by one monospecific and two multispecific sera,
or by three multispecific sera. Phenotypes must be verified
by reading two independent trays or tray sets. Each tray or
tray set must be read independently.
The identification of HLA phenotypes requires a certain
amount of expertise, requiring knowledge of cross reactivity
between antigens and splits of broad reactivity (e.g., A9 split-
ting into A23 and A24) and familiarity with the unexpected
reactivity of the antisera used (false-negative or extra reac-
tions). Because of the variability of HLA antisera, it is rec-
ommended that all individuals in a parentage case be tested
in the same laboratory, ideally with the same reagents.

DNA Polymorphisms: Repetitive Elements


in Genomic DNA

Although single nucleotide polymorphisms are the most nu-


merous in the genome, polymorphisms due to the variable
repetition of a short sequence of nucleotides (termed a
“variable number of tandem repeats”) exhibit the highest
collective degree of polymorphism in the human population.
Visualization of VNTR polymorphisms is possible using
restriction enzymes coupled with Southern blotting and
probe hybridization. VNTR polymorphisms can also be
visualized through PCR amplification of STR loci using PCR
primers that hybridize to genomic DNA outside the repeti-
tive array and are labeled with a fluorescent dye. DNA typing
of VNTR loci has become the principal methodology for
evaluating cases of questioned relatedness. Figure 24–3. RFLP analysis in two paternity cases. DNA extracted from blood
samples of an alleged father (AF), child (C), and mother (M) in two paternity cases
RFLP Analysis were subjected to RFLP analysis using a radioactively labeled DNA probe directed
against the D16S85 locus. Profiles visualized in case 1 show the AF to be included
RFLP (restriction fragment length polymorphism) analysis because he shares the smaller of his two alleles with the child, and this is the obli-
gate gene in the child’s profile. In contrast, in case 2 the AF and the child do not
refers to the visualization of polymorphisms in DNA by share any alleles; this is evidence of nonpaternity for this tested man. The lanes
restriction enzyme digestion of genomic DNA, normally marked “X” represent mixtures of DNA from the AF and the C to maximize resolution
extracted from a blood sample followed by agarose gel of the RFLP process.
6888_Ch24_515-530 29/10/18 11:34 AM Page 520

520 PART IV Leukocyte Antigens and Relationship Testing

of RFLP analysis by parentage laboratories because of or probability of exclusion) may differ between two labora-
their high discriminatory power characteristics. As shown in tories for the same locus with the same restriction enzyme,
Table 24–2, the fragment size of VNTR alleles depends on the even if their band sizes are identical.
number of repeats they contain and the restriction enzyme VNTR loci are very polymorphic and thus are very power-
utilized. This is an important concept: The phenotype as rep- ful for excluding alleged fathers in paternity tests and in pro-
resented by band size estimates in kilobase pairs of the same viding compelling inclusionary evidence in true biological
individual at the same locus will differ between two labora- relationships. However, the mutation rate for adding or delet-
tories if each uses a different restriction enzyme. This is why, ing repeats from the tandem array can be up to 2 per 1,000,
when reporting RFLP results, it is mandatory to identify not which is small but not negligible.6 In the past 15 years, labo-
only the locus and probe tested but also the restriction en- ratories have stopped using RFLP methods, moving instead
zyme used. In the United States, the most common restriction to technologies that involve PCR amplification of small tan-
enzymes used were HaeIII and PstI, whereas in Europe, HinfI dem repeat–type polymorphisms (i.e., STRs).
was the more common.
An important limitation of RFLP analysis of VNTR poly- STR Analysis
morphisms is that allele identification is based on an esti-
mate of the size of a restriction fragment that generally The transition of relationship testing laboratories from serol-
consists of thousands of nucleotides. As in any quantitative ogy and HLA to RFLP analysis was short lived, largely be-
measurement, there is variability. Variability in repeated size cause of the characterization of VNTR loci consisting of
estimates for restriction fragments produced from the same tetranucleotide repeats that could be amplified using PCR.
sample was termed sigma (σ). Another source of variability Figure 24–4 graphically describes PCR amplification of STR
relates to the resolving capabilities of the electrophoresis sys- loci using fluorescent detection using capillary electrophoresis
tem used to separate restriction fragments of similar size. (CE). The figure shows STR amplification products separated
This uncertainty was given the name delta (∆). Resolution in one of four capillaries in the array that are labeled with
may vary greatly among laboratories, depending on the either red, yellow, blue, or green fluorescent dye (depending
methodology utilized, especially the technical conditions on the particular STR locus). (Note: Because of publishing
used during electrophoresis. Because of these two sources of cost considerations, the electropherograms are shown mono-
variability, the allele frequency distribution of VNTR alleles color.) Electropherograms are displayed as fluorescent bands
detected using RFLP analysis is a size continuum as opposed (left panel) or as a histogram (right panel), the latter of
to the discrete distribution of allele frequencies as in the clas- which is the more common method for STR profile presen-
sic genetic systems. For example, in most laboratories, a tation. The banding results are correlated with the histogram
DNA fragment measured at 1.60 kb cannot be considered by capillary number.
different from a fragment measured at 1.57 kb or 1.63 kb, DNA typing of STR loci eliminated many of the shortcom-
even though differing numbers of repeats would exist in a ings associated with RFLP analysis. The amount of DNA re-
group of alleles of this size. Through the use of validation, quired for testing dropped about 1,000-fold, making it
each laboratory must identify the limit of resolution for possible to use buccal swabs as a source of DNA instead of
which two closely spaced alleles cannot be distinguished blood samples, which were often difficult to collect from
from one another. The approach taken to incorporate this newborns. The reduced quantity of DNA needed also allows
variability in analyzing casework was to create a size bin with for direct DNA testing from prenatal samples of amniotic
properties defined by the results of validation in identifying fluid or chorionic villi (CVS).
values for sigma and delta. This range of matching sizes, or A second advantage of STR analysis is the increased reso-
bin, was also used to declare a match in RFLP profiles and lution of alleles associated with the electrophoresis systems
then to determine the allele frequency of an allele important used: normally, polyacrylamide gel electrophoresis (PAGE)
in a case. The allele frequency used in the statistical calcula- or capillary electrophoresis (CE), both of which can reliably
tions is actually the frequency of all the alleles falling into distinguish two DNA fragments differing by a single base pair.
the bin for the allele size measured. Since the bin size varies With the enhanced resolution of the electrophoresis systems
with the technical capabilities of the laboratory, the final sta- came the ability to designate discreet STR alleles, naming
tistical result (i.e., likelihood ratio, probability of parentage, each based upon the number of repeats of the tetranucleotide

Table 24–2 DNA-RFLP Results in a Paternity Case at the D2S44 Locus


Restriction Enzyme Used for Digestion
Individual Tested Hae II (kb estimate) HinfI (kb estimate) PstI (kb estimate)

Mother 3.43, 4.25 4.55, 5.36 12.68, 13.53

Child 2.91, 3.43 4.03, 4.55 12.16, 12.68

Alleged father 2.91, 3.13 12.16, 12.45 12.16, 12.46


6888_Ch24_515-530 29/10/18 11:34 AM Page 521

Chapter 24 Relationship Testing 521

Chromosome homologues,
PCR primer binding sites denoted by arrows

PCR with fluorescent primers, capillary electrophoresis with


fluorescent STR alleles.
Capillary 1

7000 7127
5000 5127
3000 3127
1000 1127

3000 4000 5000 6000 7000


Intensity vs Scan Number
Capillary 2

3418
3000

1418
1000

3000 4000 5000 6000 7000


Intensity vs Scan Number
Capillary 3

3000 2836

1000 836

–1000
3000 4000 5000 6000 7000
Intensity vs Scan Number
Capillary 4

5000 4836
3000 2836
1000 836
–1000
3000 4000 5000 6000 7000
Intensity vs Scan Number

Figure 24–4. PCR amplification of STR loci using fluorescent detection. Genomic DNA recovered from a biological sample is added to a multiplex PCR reaction containing
fluorescently labeled primers for multiple STR loci. Following amplification, PCR products are separated by size using polyacrylamide gel or, in this case, capillary electrophoresis.
The gel or capillary is then irradiated with a laser, causing the STR alleles to emit light at several wavelengths depending on the particular fluorescent dye coupled to the
different primers. In this figure, STR alleles are displayed in one of two formats—horizontal bands in a gel track much like traditional RFLP profiles or in the newer histogram
format that is the norm among STR typing laboratories that use capillary electrophoresis platforms to separate STR alleles. STR alleles, whether displayed as bands or peaks,
are displayed monocolor. Size of the PCR amplicons and the color emitted serve to identify the particular genetic marker visualized and the particular allele present in a
sample.

or pentanucleotide contained within the tandem array. Elim- (i.e., the core loci) for a nationwide databank of crime scene
inating the use of bins in the matching process and in deter- DNA evidence material and reference samples from con-
mining allele frequencies also facilitated the general victed felons (see Box 24–2). As was stated earlier, as of
acceptance of STR typing in court. January 2017, the acceptable list of CODIS loci expanded to
The third advantage of the technology is that multiplexing 20 (see Box 24–2), in part because of the availability of com-
of multiple STR loci during PCR amplification allows simul- mercial kits that multiplex 25 different STR loci in a single
taneous testing of several loci with fast allele identification PCR reaction.
by capillary electrophoresis. The steps of STR analysis are Readily available commercial kits provide convenient
also easily amenable to the use of automation and hence, multiplex amplification for loci selected to match the CODIS
faster turnaround times. STR core loci plus additional STR markers to further en-
The panels of STR loci currently in use consist mostly of hance the discriminatory power of the multiplex.
repeats with core sequences of four bases (tetranucleotide A group of STR loci located on the Y chromosome has
repeats), although a few loci based on core sequences of five also become available and has successfully delineated the
bases (pentanucleotides) have recently been introduced. A male lineage within family groups. Y-STR analysis can be es-
group of 13 loci are common to all commercially available pecially useful for establishing or refuting a claimed rela-
multiplex PCR kits in conjunction with amelogenin that tionship within the male lineage of a pedigree. However,
identifies the sex of the sample donor and nothing else. This caution must be used in reporting results since all males
group has been selected by the FBI as the core testing panel within the lineage are equally likely to be the parent,
6888_Ch24_515-530 29/10/18 11:34 AM Page 522

522 PART IV Leukocyte Antigens and Relationship Testing

brother, grandfather, uncle, and so on, of a male child whose Terminology and the Interpretation
paternity is in question. of Results
Phenotypes for all STR loci should be reported following
the recommendations of the International Society for Foren- In interpreting the testing results on a classic trio (mother,
sic Genetics: alleles are identified by the repeat number. child, and alleged father), it is always assumed that the
Repeat variants that do not consist of a complete 4 or 5 base- mother is the biological mother (unless the questioned re-
pair repeat element are identified by the repeat number lationship is one of maternity). This assumption sets the
followed by a period and the number of additional bases stage for the analysis of results in that it is expected
(e.g., a common allele of TH01 is 9.3, which means that it that the mother and child will share at least one allele for
has nine tetranucleotide repeats and three additional bases). every genetic marker tested. Therefore, the remaining
allele in the child’s profile is of paternal origin and can be
Single Nucleotide Polymorphisms compared with the profile of the alleged father (AF). If
the AF lacks the paternally inherited allele in the child, it
In addition to polymorphism contained within nuclear DNA, is evidence of nonpaternity. If he is not excluded, a statis-
mitochondrial DNA (mtDNA) exhibits single nucleotide tical analysis is performed to determine his relative prob-
polymorphisms (SNPs), which can be useful for kinship ability of being the biological father of the child as
testing. To reveal these SNPs, noncoding hypervariable seg- compared with a random man unrelated to him or to the
ments (the displacement loop, also known as hypervariable mother.
regions I and II, or HVI and HVII) of mtDNA are amplified There is terminology associated with relationship testing
by PCR and then sequenced. The sequence is then compared that deserves some discussion. Two terms that ultimately
with a published reference mtDNA sequence (known as the define the overall result of the testing are exclusion and in-
Cambridge sequence). Because mitochondria are passed on clusion. Although the definitions of these terms may seem
with the cytoplasm of the oocyte and not by spermatozoa, obvious, the criteria by which these terms are used to com-
mitochondrial DNA is passed from mother to child. There- municate results have evolved along with the technologies
fore, mtDNA typing is especially useful when analyzing a used for testing. For example, when serology was widely
questioned maternal lineage. used for testing, a single inconsistent test result between
Mitochondrial DNA appears to be stable for extremely alleged parent and child could be sufficient to exclude
long periods (thousands of years), probably because the parentage. For example, a type AB child could not be the off-
mtDNA genome is small and there are 500 to 1,000 copies spring of a type O alleged parent.
present in most cells in the body. These genetic systems have
been applied to anthropological studies aimed at solving Exclusion Criteria for Classical Systems
human evolution questions. Preserved mitochondrial DNA
has also been isolated from mummies and fossils.7 This tech- In classical systems, inconsistencies have been customarily
nique is very useful in demonstrating maternal lineage in referred to as exclusions, direct or indirect, with the rule
long-distance relatives such as grandmother to grandchild, that a single direct exclusion was enough to exclude the
and it was applied to the authentication of the remains of the alleged father as the biological father of the child, while at
family of Tsar Nicholas II of Russia, who were assassinated least two indirect exclusions were necessary to form a final
during the Bolshevik uprising.8 conclusion of exclusion. A direct exclusion exists when
Single nucleotide polymorphisms in nuclear DNA are also either the child possesses a marker that is not present in
being explored for parentage testing. Given the extensive re- either the mother or the alleged father (e.g., the child is
source of GenBank, some laboratories have developed panels K-positive while both the mother and the alleged father are
of multiple hundreds of SNP loci that are coamplified in one K-negative) or when the alleged father demonstrates heterozy-
or a limited number of reactions, and then the SNP pheno- gosity for two different alleles in the same system and the child
type of the sample can be determined by sequencing or using does not demonstrate either (e.g., an AB alleged father with
DNA arrays. Although individually an SNP exhibits low dis- an O child).
criminatory power, collectively these SNP panels provide a On the other hand, an indirect exclusion, also referred to
very high discriminatory power in cases of questioned relat- as apparent opposite homozygosity, exists when the child
edness. In one special application of the technology, several demonstrates only one marker (i.e., appears to be homozy-
laboratories offer prenatal parentage testing using a sample gous for the marker) and the alleged father demonstrates a
of maternal blood. When DNA is extracted from the sample, different single marker (i.e., appears to be homozygous for
it will consist of a mixture of mostly maternal DNA, but a different marker). As this situation requires the assumption
there will also be a detectable amount of fetal DNA in the of homozygosity, it is considered indirect evidence. The
sample. If the lab examines hundreds of SNP loci, those in transmission of an alternate allele that is either silent or
which the mother and true father differ in genotype can be not routinely tested for can lead to this apparent opposite
detected in the maternal sample, designated as fetal poly- homozygosity in a true biological parent/child pair. Such
morphisms, and compared against the constellation of SNP alleles exist at a low but significant frequency in most of the
genotypes of an alleged father. classical systems.
6888_Ch24_515-530 29/10/18 11:34 AM Page 523

Chapter 24 Relationship Testing 523

Exclusion Criteria for DNA Systems replication “slips and jumps” as it moves down a chromo-
some. If the complex slips and remains in one spot on the
One consequence of the technology shift to DNA-related chromosome while replicating, a new repeat may be added to
methods was the discovery of the rather high mutation rate the daughter strand. If the complex jumps over a repeat due
characteristic of the VNTR systems.6,8 Such mutation rates are to a loopout of the DNA template or for some other reason, a
sufficiently high to cause a laboratory to be wary of single repeat may be lost from the daughter chromosome. The net
DNA mismatches between alleged parent and child as com- effect of either occurrence will be the production of a gamete
pelling evidence of exclusion. Mutations associated with harboring a genetic marker distinct from the parent. If this
VNTR loci are generally the result of DNA replication errors gamete is involved in conception, the offspring will differ from
that occur during meiosis and alter the number of repeats in the parent in genotype. When an inconsistency in the inheri-
the tandem array. Changes in the number of repeats is usually tance pattern is detected in a DNA system, the term exclusion
small (i.e., 1% to 3% of allele size for VNTR loci detected with is not used; instead, the terms mismatch or inconsistency are
RFLP methods and single repeat changes for STR loci), and used to indicate that the alleles between the child and the
repeats can be added to or subtracted from daughter alleles alleged father do not match. The term exclusion is reserved for
with approximate equal frequency.6,8 There is a confirmed the final interpretation of the entire set of genetic systems
difference in mutation rate in the male versus female lineages, tested. A minimum of two mismatches is required before an
with males exhibiting a 10- to 20-fold higher rate than females opinion of nonpaternity (or nonmaternity) is rendered, and
for some loci, possibly due to an increased number of cell with the increased number of STR loci coamplified with
divisions that occur during spermatogenesis (Table 24–3). multiplex kits currently in widespread use, many labs require
Although the mechanism underlying alterations in repeat a minimum of three mismatches before a conclusion of exclu-
number resulting from mutation in VNTR loci is not certain, sion is rendered. One element of such multiple mismatches
current evidence does not support unequal recombination be- that must be considered is the degree to which the obligate
tween sister chromosomes during meiosis as the mechanism. alleles are mismatched. If two mismatches exist in a case and
Rather, it appears the replicative complex responsible for DNA both involve single repeat differences, then it is much more
likely the case involves a double mutation as opposed to a sec-
ond case in which one or two mismatches involving more
Table 24–3 Summary of Mutation Rates than a single repeat are seen. Mutations involving the gain or
for STR Loci loss of more than one repeat are about 10-fold less common
than mutations involving the gain or loss of a single repeat.
Paternal Maternal
Locus Lineage Lineage Inclusionary Calculations
CSF1PO 0.002021 0.000283
Three types of statistical evaluations can be reported when
D13S317 0.001743 0.000436 the alleged father is not excluded: the parentage index (PI),
D16S539 0.001127 0.000481 the probability of paternity (PP), and the probability of ex-
clusion (PE). The parentage index is now more often referred
D18S51 0.00253 0.000748 to as the relationship index (RI) as genetic testing capabilities
D19S453 0.000745 0.000596 available today allow for testing of questioned relationships
beyond alleged parent and child. Questioned sibships, half-
D21S11 0.001709 0.001295 sibships, aunt/uncle, grandparent, etc. are increasingly sub-
D2S1338 0.001526 0.000245 jected to STR typing to try and establish or refute claimed
familial relationships. The relationship index produced in all
D3S1358 0.001691 0.000211 cases compares two mutually exclusive hypotheses in the
D5S818 0.001742 0.0003 form of a ratio. Generally, the hypothesis located in the
numerator is a statement of probability that the claimed rela-
D7S820 0.001348 0.000073 tionship is true (i.e., in a paternity case, the alleged father is
D8S1179 0.002031 0.000333 the true father), whereas the denominator of the ratio reflects
the probability (in a paternity case) that an unrelated, random
FGA 0.003713 0.000522 man is the true father of the child. In a case of questioned
PENTA D 0.000259 <0.000253 paternity, the standard calculations are not valid if a brother,
father, uncle, or other close biological relative of the tested
PENTA E 0.00026 <0.000253 man is also a possible biological father of the child.
THO1 0.00007 0.000043 An important aspect of the calculations is the need to have
accurate allele frequencies for each genetic system tested,
TPOX 0.00013 0.000081 estimated from databases drawn from an adequate sample
VWA 0.003258 0.000494 population of the appropriate ethnic group. When referring
to a sample size for a database, the term adequate will depend
6888_Ch24_515-530 29/10/18 11:34 AM Page 524

524 PART IV Leukocyte Antigens and Relationship Testing

largely on the degree of polymorphism exhibited by the ge- probability of producing a child genetically identical to the
netic marker in question. For example, for hypervariable tested child through a mating of the known parent with the
VNTR systems tested using RFLP analysis in the 1980s and alleged parent. The denominator mathematically states the
1990s, databases consisting of multiple hundreds or even probability of producing the child through a mating of
thousands of individuals were generally deemed adequate for the known parent with a randomly selected, unrelated indi-
accurate probability calculations. In contrast, databases cre- vidual in the same ethnic population. The RI is calculated
ated for STR loci may consist of only 200 to 300 individuals one genetic system at a time and is sometimes referred to as
because there are far fewer alleles possible per locus. the system index (SI). If all the genetic systems tested are in-
dependent from one another, the overall final RI is the prod-
Paternity or Relationship Index uct of all the system indices multiplied together. When the
RI is greater than 100, the evidence for parentage is consid-
The relationship index (RI), compares two mutually exclusive ered very strong. Figure 24–5 shows STR typing in a pater-
hypotheses that are expressed as a likelihood ratio. In a parent- nity case involving two alleged fathers, and Table 24–4
age case, the numerator generally states the mathematical outlines the testing results for the case. For the D3S1358

Alleged father #1

Alleged father #2

Child

Mother

Figure 24–5. STR typing in a paternity case involving two alleged fathers. DNA was extracted from buccal swabs from two alleged fathers (1 and 2), a child, and a
mother and subjected to STR analysis using the Identifiler STR typing kit (Applied Biosystems, Foster City, CA). Products were separated and analyzed using capillary elec-
trophoresis on an ABI310 Genetic Analyzer (Applied Biosystems). Shown in the figure are the results from those loci labeled with the green fluorescent dye (shown as red
here) known as JOE consisting of 5 of the 15 STR loci coamplified with the Identifiler kit. Shown under each peak in the histogram are labels containing the allele identification
(labeled “al”), the size of the amplicon (labeled “sz”), and the intensity of the fluorescence (labeled “ht”). The results of the analysis are shown in Table 24–4.
6888_Ch24_515-530 29/10/18 11:34 AM Page 525

Chapter 24 Relationship Testing 525

Table 24–4 System Indices Produced in a Paternity Case With Two Alleged Fathers
System Mother Child Alleged Father No. 1 PI (AF No. 1) Alleged Father No. 2 PI (AF No.2)
D3S1358 15, 18 16, 18 16, 17 2.1598 16 4.32

TH01 7, 9 7, 9 6, 7 1.4819 7, 9 2.96

D13S317 11, 12 12 12, 13 1.6197 8, 11 0

D16S539 9, 12 9 9, 12 4.8077 9, 12 4.8

D2S1338 18, 20 17, 20 17, 18 2.8835 19, 23 0

system, the mother and child share the 18 allele, thus the and will pass it with a frequency of 0.5. The calculation
obligatory gene (paternal allele in the child) is allele 16. thus becomes:
Alleged father number 1 (AF1) is heterozygous for this allele;
SI = [(0.5 M passing 12)(0.5 AF passing 12)]/
thus, his chance of passing it on to an offspring is 50%
[(0.5 M passing 12)(0.309 RM passing 12)]
(i.e., 0.5). Alleged father number 2 (AF2) is homozygous for
SI = 1.62
allele 16 and thus will pass it to 100% of his offspring. The
population frequency for allele 16 is 0.232 (which also rep- The overall RI for AF1 and AF2 is the product of the in-
resents the chance the random man [RM] will pass 16 to the dividual system indices for the five STR systems shown in
child). Therefore, the system index calculation for AF1 takes Figure 24–5 and Table 24–4. For AF2, since two of those sys-
the following form: tems have SI values of 0, the combined relatedness index is
0 and the man is excluded. For AF1, the individual system
SI = [(0.5 M passing 18)(0.5 AF passing 16)]/
indices are multiplied to produce a combined paternity index
[(0.5 M passing 18)(0.232 RM passing 16)]
of 71.74. This value means that AF1 is about 72 times more
SI = 0.5/0.232 = 2.16.
likely than is a random man to produce the single sperm that
Therefore, based upon this test result alone, AF1 is contains all the genetic information this child inherited from
2.16 times more likely to be the father of the child than is a his or her biological father.
random man of the same ethnic background. Interestingly,
AF2 is twice as likely to be the father of the child as is AF1, Probability of Paternity or Relatedness
because he is homozygous in phenotype at the D3S1358
locus. However, he is excluded as the child’s father at the The posterior probability of parentage or relatedness (PP)
D13S317 and D2S1338 loci because he lacks the obligate is a restatement of the weight of the evidence for relatedness,
alleles seen in the child. This is one reason why it is impor- expressed in percentage form. To calculate the PP, the RI
tant for a parentage testing laboratory to test multiple genetic is considered in light of other known facts about the case.
markers in a trio so that random coincidences are not overly In other words, multiple probabilities both for and against
emphasized in developing an opinion of inclusion or exclu- relatedness can be combined into a single result according
sion about a case. to probability theory developed by Bayes. The Bayes theorem
In the second system listed, the mother and child are phe- allows multiple types of evidence for and against a hypoth-
notypically identical, so it is uncertain which allele, 7 or 9, esis to be combined into a final statement of probability. In
is the obligatory gene. AF1 is heterozygous for allele 7 and the case of parentage testing,
thus is included as a possible father. However, the SI calcu-
PP = ([(RI)(prior for)]/[(RI)(prior for) + prior against].
lation must consider both allele 7 and allele 9 as possible
obligatory genes and takes the following form: In order to calculate the PP, a value for a prior probability
favoring a hypothesis must be chosen and included in the
SI = [(0.5 M passing 9)(0.5 AF passing 7) +
formula. In relatedness testing, the value for the prior essen-
(0.5 M passing 7)(0 AF passing 9)]/
tially reflects the probability of the questioned relatedness
[(0.5 M passing 7)(0.0.165 RM passing 9) +
being true before the testing was performed. In the case of
(0.5 M passing 9)(0.172 RM passing 7)]
a parentage test, the prior probability reflects the evidence
SI = 1.48
favoring parentage of a child before any genetic testing is
Again, AF2 has twice the likelihood of being the child’s performed. Typically, relationship testing laboratories assign
father, in this case because his phenotype contains both a neutral prior of 0.5 (i.e., 50%), neither favoring nor refut-
possible obligate alleles seen in the child (i.e., he can trans- ing the alleged parent/child relationship. However, based
mit either allele 7 or 9 to his offspring). In the third system, upon available information, a judge can change the prior to
the child is homozygous and inherits one copy of allele 12 something other than 0.5. For example, if an alleged father
from the mother. AF1 is heterozygous for the obligate allele has a sperm count 1% of normal, that could reduce his
6888_Ch24_515-530 29/10/18 11:34 AM Page 526

526 PART IV Leukocyte Antigens and Relationship Testing

fertility and a judge could reduce the prior probability to discriminatory power of the STR test batteries available to lab-
1% or at least reduce it from 50%. Such a reduction would oratories is such that a known parent or other known relative
necessitate a higher RI value to get the same final PP value, is not as important in terms of enhancing the discriminatory
which may require additional testing be performed. When power of the test as when laboratories depended upon blood
the prior is 0.5, the PP formula is reduced to: group serology and HLA. In addition, a significant portion of
the testing performed in the United States nowadays is in sup-
PP = RI/(RI + 1).
port of immigration in which applicants regularly originate
Currently, laboratories target a PP of 99% or greater as the from conflict areas of the world in which known family mem-
threshold of proof of parentage for an alleged parent or re- bers may not be available. Calculations in parentage cases in
latedness for other familial situations. At the 99% level in a which a known parent is lacking follow the typical procedure
parentage test, the burden of proof shifts from the known with the exception that a “random known parent” is substi-
parent to prove the alleged parent is a true parent to the tuted into the RI equation in place of the 0.5 or 1.0 values
alleged parent to prove they are not a true parent. Some reflecting heterozygosity or homozygosity for the known par-
consumers of relationship testing services have minimum ent phenotype. Instead, the transmission frequency for an
acceptable probabilities higher than 99%. For example, the allele possibly shared between the known parent and the child
U.S. Citizenship and Immigration Services sets the minimum becomes the population frequency for that allele being trans-
acceptable probability at 99.5% for applicants claiming to be mitted from the random known parent.9 When a “random
related to a citizen or permanent resident who is sponsoring known parent” must be included in the calculation, either al-
their application for immigration into the United States. lele in a heterozygous child could have come from the known
parent, and their respective transmission probabilities are set
Probability of Exclusion to equal the population frequency for each.10
Occasionally, even the alleged parent is not available for
The probability of exclusion (PE) is the probability of exclud- testing, and a reconstruction of his or her possible genetic
ing one who is falsely accused of being related to someone in makeup is attempted by testing all available and known par-
a given way. The PE reflects the probability of excluding one ents, siblings, and other undisputed children of the alleged
who is alleged to have a relationship, and this statistic is prob- father. Other biological relationships, such as siblings, may
ably most meaningful in a case of disputed parentage. The be in question in adoption, immigration, or inheritance cases.
probability of exclusion is entirely dependent on the pheno-
types of the known parent and the child and does not require Advantages and Disadvantages
knowledge of the alleged parent’s phenotype. In a paternity of Genetic Systems
test, calculation of PE relies on first calculating the proportion
of men who would not be excluded (representing all homozy- The field of family relatedness testing has moved entirely to
gous and heterozygous phenotypes including allele 16); this STR testing platforms as the technology of choice for several
number is called random men not excluded (RMNE). PE is thus important reasons. The first reason has to do with discrimi-
calculated by 1 – RMNE. If p is the sum of all frequencies of natory power. Red blood cell antigen systems represent the
the possible paternal alleles, then RMNE = p(2 – p) for each genetic markers with the least discriminatory power, with
genetic system. The overall RMNE for all the genetic systems MNSs and Rh representing classic markers with the greatest
tested is the product of the RMNE values for each system, and PE, at 31% and 27%, respectively. Remaining red blood cell
the overall probability of exclusion is 1 – overall RMNE. In antigen systems and enzyme and protein markers have typical
the case shown in Table 24–4, the overall RMNE is: individual PE values of less than 20%. HLA-A and HLA-B
antigens have a high PE value (~87% together), but HLA typ-
0.410 × 0.560 × 0.197 × 0.316 = 0.0075, ing is subject to changes in the availability of antisera present
and the combined probability of exclusion is therefore in commercially available plates. Still, the classic serologic
0.9925. The probability of exclusion represents the propor- test platforms have the advantage of the vast and diverse ex-
tion of untested men who would have been excluded by the perience accumulated over the decades, during which they
extent of testing performed (99.25% in this case). Histori- have been in place in many countries.
cally, PE has been used in some jurisdictions in resolving DNA polymorphisms visualized using RFLP analysis are
paternity disputes, but RI and PP are more meaningful extremely powerful genetic systems with average powers of
methods of considering relatedness cases because both exclusion almost always above 70% and sometimes reaching
approaches consider the actual phenotype of the questioned above 90% for a single genetic system. The high PE value as-
relative whereas PE does not. sociated with VNTR loci analyzed using RFLP means that
fewer loci are needed to conclusively resolve cases of disputed
Nonclassic Situations parentage. Thus, testing only four different loci by DNA-RFLP
ensures very high levels of PP for nonexcluded men.
Historically, the majority of parentage testing cases consisted STR polymorphisms generally exhibit 5 to 10 alleles per
of the classic trio of mother, child, and alleged father. How- locus. Thus, STR loci do not exhibit the extreme degree of
ever, cases of disputed parentage that deviate from the classic polymorphism characteristic of RFLP systems and so the
trio are common these days for several reasons: first, the number of STR systems needed to achieve a comparable level
6888_Ch24_515-530 29/10/18 11:34 AM Page 527

Chapter 24 Relationship Testing 527

of PE is increased. PE values for a typical STR system average support agencies account for much of the activity of some re-
about 50% to 60%. Even though the individual PE value for lationship testing laboratories. The majority of cases outside
an STR system is less than for a system detected with RFLP, the child support enforcement agencies also have a legal im-
multiplex STR typing kits with as many as 24 different loci plication, such as custody in divorce cases, inheritance rights,
simultaneously amplified in a single PCR reaction are readily immigration, and more recently, the identification of human
available commercially today. The discriminatory power of remains in natural disasters, cases of murder, or genocide.
such multiplex kits even exceeds the typical collection of Occasionally, cases also have criminal implications relating
four to six RFLP systems. to incest or statutory rape or even searching the CODIS con-
Another advantage of testing DNA systems over the classic victed felon database for relatives who may have also com-
serology systems is the flexibility in sample type usable for test- mitted unsolved crimes (see the case of Lonnie David
ing. Any tissue or body fluid from which sufficient DNA can Franklin Jr.).12 STR testing is also widely used in other areas
be extracted for analysis will suffice as a sample for testing. Pe- of medicine, including monitoring engraftment in patients
ripheral blood, buccal smears, amniotic fluid, chorionic villi, who received a bone marrow transplant, assessing the purity
and tissue biopsies can all serve as a sample. Once extracted, of presumed fetal DNA obtained from amniocentesis or CVS
DNA can be kept frozen for an indefinite period. It should be specimens from pregnant mothers and tested for genetic dis-
recalled that the amount of DNA required for RFLP analysis in ease that may actually represent a mixture of maternal and
a parentage test is about 1,000-fold higher than that required fetal DNA, and the identification of pathology/histology spec-
for STR analysis (i.e., micrograms versus nanograms). STR imens thought to perhaps be mislabeled or contaminated
analysis in particular also further broadened the scope of sam- with someone else’s tissue during processing.
ple types amenable to testing. Because of the low-input DNA Regardless of the application for the technology, every
template requirements, bone, teeth, even hair bearing a follicle step in the collection, storage, processing, and testing of the
were suitable as a source of DNA for testing, and this opened samples must be carefully documented as to time of occur-
the door to apply parentage testing concepts to the identifica- rence and person performing the task. An unbroken chain
tion of human remains and even in criminal investigations. of custody must be maintained. The individuals tested must
The movement away from serology and RFLP analysis be carefully verified, and procedures must be in place to en-
and to STR analysis was also motivated by the automation sure that unauthorized persons do not have access to the
possible for high-volume laboratories working with an STR samples or test results. Without careful attention to these as-
analysis platform. Serology and RFLP analysis are labor- pects, the results may not be valid in court. If called to testify
intensive methodologies while STR analysis can be largely on the validity and significance of the testing results, the lab-
automated in terms of liquid-handling robots being able oratory director must be able to re-create from the docu-
to set up PCR reactions and the genetic analyzer being auto- mented evidence every step in the handling of the case.
mated for electrophoresis and data analysis.
Finally, there is the consideration of data analysis and Accreditation and Quality Issues
presentation to the consumer (i.e., the court, the client, or
their representative). In the case of DNA testing, RFLP analy- Approximately 30 years ago, the American Association of
sis suffers from a lack of precision and ideal reproducibility Blood Banks (AABB) took the lead in promoting the establish-
when results from different laboratories are compared. This ment of standardization and quality in the field of relatedness
limitation does not affect STR analysis because of the high testing and created a standing committee on parentage testing.
precision associated with the capillary electrophoresis used This committee developed educational events such as work-
to resolve STR alleles. Different alleles can be identified as shops and also developed publications concerned with parent-
discreet entities and identified using a nomenclature based age testing. With the participation of the American Medical
upon the number of repeats in the tandem array associated Association, the American Bar Association, and the Office of
with each allele. With the transition to STR methods, labo- Child Support Enforcement, the Committee on Parentage
ratories can reliably obtain the DNA profile results that are Testing organized an international conference in 1982 in Air-
identical to those produced by other laboratories. lie, Virginia, wherein a group of international experts estab-
lished a consensus approach for the interpretation of
Social and Legal Issues inclusionary evidence. An accreditation program for parentage
testing laboratories was implemented, and the first edition of
In 2010 (the most recent data available from the AABB), almost Standards for Parentage Testing Laboratories was published in
400,000 cases of relationship testing were performed in the 1990. A complementary Accreditation Requirements Manual
United States.11 Whereas traditionally the majority of parentage followed in 1991. The College of American Pathologists, with
cases were done to establish paternity for children with the aim joint sponsorship by the AABB, initiated a proficiency testing
of obtaining child support, an increasing number of tests are program for parentage testing laboratories in 1993.
performed now in support of applications for immigration. Continuing involvement by professional organizations in
Child support enforcement agencies in all states actively the promotion of quality improvement and in the provision of
pursue the identification of the biological father of children opportunities for continuing education in the field of relation-
who are eligible for Aid to Families with Dependent Children ship testing is essential to maintaining a high level of accuracy
for the purpose of seeking child support. Contracts with child and consistency in ascertaining biological relationships.
6888_Ch24_515-530 29/10/18 11:34 AM Page 528

528 PART IV Leukocyte Antigens and Relationship Testing

CASE STUDIES 1. The fourth genetic system tested (D2S44/HaeIII)


represents a(n):
Case 24-1 a. RBC antigen system tested by agglutination.
b. RBC enzyme system.
An African American trio consisting of a mother, child, c. DNA polymorphism tested by RFLP.
and alleged father is tested to establish paternity. The fol- d. STR-DNA polymorphism tested by PCR.
lowing results are obtained: 2. The fifth genetic system tested (D3S1358) represents
Genetic System Mother Child Alleged Father a(n):
a. RBC antigen system tested by agglutination.
ABO O B A
b. RBC enzyme system.
D3S1358 15, 18 16, 18 16, 17
c. DNA polymorphism tested by RFLP.
THO1 7, 9 7, 9 6, 7
d. STR-DNA polymorphism tested by PCR.
D13S317 11, 12 12 8, 11
3. Among the following statements relating to the inter-
D16S539 9, 12 9 9, 12
pretation of these results, choose the one that is true:
D2S1338 18, 20 17, 20 17, 18
a. The alleged father is excluded due to a direct ex-
1. Which of the genetic systems tested excludes the AF? clusion at D3S1358.
a. THO1 and D16S539 b. The alleged father may be excluded due to an
b. ABO and D13S317 inconsistency at D3S1358.
c. ABO and THO1 c. A mutation may have occurred at D3S1358.
d. No system excludes the AF. d. The alleged father is excluded due to a direct
2. Given the test results in question 1, if this was the only exclusion in the ABO system.
testing performed, would you exclude the AF based upon
currently accepted standards for parentage testing?
a. Yes
b. No

Case 24-2

A white trio consisting of a mother, child, and alleged


father is tested to establish paternity. The following results
are obtained:
Genetic System Mother Child Alleged Father
ABO A O B
MNSs MNs MNs NSs
D10S28/HaeIII 3.34, 2.98 3.34, 4.52 1.95, 4.52
D2S44/HaeIII 3.42, 4.24 2.90, 3.42 2.90, 3.11
D3S1358 15, 16 15, 17 14, 18
FGA 21, 22 22, 25 20, 25
TH01 6, 9.3 9, 9.3 7, 9

SUMMARY CHART
 Relationship testing refers to the testing of genetic expressed by the A and B loci are considered for rela-
markers that are inherited to determine the presence tionship testing with non-DNA-based typing.
or absence of a biological relationship.  RFLP refers to polymorphisms of the DNA that can be
 The ultimate goal of relationship testing is to confirm a detected using restriction enzymes, gel electrophoresis
specific biological relationship with the individual in ques- and blotting, and DNA probe hybridization; polymor-
tion, usually a father or a mother or sometimes a sibling. phisms detected in relationship testing relate to the
 The blood group systems used most often in paternity presence of VNTR.
testing are ABO, Rh, MNSs, Kell, Duffy, and Kidd.  In PCR, the DNA fragment of interest is amplified in
 The HLA complex represents the most polymorphic amount thousands of times, and the resulting product
genetic system in the human genome; only antigens is normally identified directly through fluorescence
6888_Ch24_515-530 29/10/18 11:35 AM Page 529

Chapter 24 Relationship Testing 529

SUMMARY CHART—cont’d
resulting from fluorescently labeled primers incorpo- demonstrates two markers and the child has neither
rated in the PCR reaction. one of them.
 STRs have repeats that are 4 to 5 bp long.  An indirect exclusion occurs when a single marker is
 STR present on the Y chromosome can verify only detected in the child and a different single marker is
male lineage. detected in the alleged father.
 Mitochondrial DNA can verify only maternal lineage.  False direct exclusions can occur as the result of mu-
 The term mismatch is used when the bands between tations that are significant enough to alter the final
the child and the alleged father do not match; a mini- product, lack of precursor substance, suppressor activ-
mum of two mismatches is required before an opinion ity at a locus unlinked to the one tested, or chimeric
of nonpaternity (or nonmaternity) is rendered. state of one of the tested individuals.
 A direct exclusion occurs when a marker is detected  False indirect exclusions occur secondary to the pres-
in the child and is absent in the mother and the ence of silent alleles (e.g., Fy).
alleged father or when the alleged father’s phenotype

2. Smithies O. Zone electrophoresis in starch gels: group varia-


Review Questions tions in the serum proteins of normal human adults. Biochem
J. 1955;61:629.
1. Among the combinations of attributes described below, 3. Dausset J, Colombani J, editors. Histocompatibility testing
select the one that would not be suitable for a genetic 1972. Proceedings of the 5th International Histocompatibility
Workshop and Conference 1972; Evian les-Bains, France.
system used in parentage testing analysis. Munksgaard, Copenhagen: Williams & Wilkins; 1973.
a. The system has multiple alleles in Hardy-Weinberg 4. Jeffreys AJ, Wilson B, Thein SL. Hypervariable “minisatellite”
equilibrium regions in human DNA. Nature. 1985;314:67.
b. The system has a high mutation rate 5. Nakamura Y, Leppert M, O’Connell P, Wolff R, Holm T, Culver,
et.al. Variable number of tandem repeat (VNTR) markers for
c. Databases of allele frequencies are available for all
human gene mapping. Science. 1987;235:1616-22.
ethnic groups tested by the laboratory 6. Brinkman B, Klintschar M, Neuhuber F, Hühne J, Rolf B.,
d. All systems selected are genetically independent from Mutation rate in human microsatellites: influence of the struc-
each other ture and length of the tandem repeat. Am J Hum Genet. 1998;
62:1408.
2. In which of the following genetic systems is the allele 7. Krings M, Stone A, Schmitz RW, Krainitzki H, Stoneking M,
frequency distribution continuous (not discrete)? Pääbo S., Neanderthal DNA sequences and the origin of mod-
ern humans. Cell. 1997;90:19.
a. DNA polymorphisms by RFLP 8. Ivanov P, Wadhams MJ, Roby RK, Holland MM, Weedn VW,
b. DNA polymorphisms by PCR Parsons TJ., Mitochondrial DNA sequence heteroplasmy in
c. RBC antigens the grand duke of Russia: Giorgig Romanov establishes the
d. RBC enzymes authenticity of remains of Tsar Nicholas II. Nat Genet.
1996;12:417.
3. A false direct exclusion in RBC antigen genetic systems 9. Edwards M, Allen RW. Characteristics of mutations at the
can be caused by: D5S818 locus studied using a tightly linked marker. Transfu-
sion. 2004;44:83-90.
a.A silent allele 10. Brenner CH. A note on paternity computation in cases lacking
b.A lack of precursor substance a mother. Transfusion. 1993;33:51.
c.An alternate untested allele 11. Annual Report Summary for Testing in 2010, www.aabb.org/
d.Weak reagents sa/facilities/Documents/rtannrpt10.pdf
12. Gerber MA. The controversial DNA search that helped nab the
4. Among the following organizations, which one offers an ‘Grim Sleeper’ is winning over skeptics. LA Times. Oct. 25,
accreditation program for parentage testing laboratories? 2016 https://www.latimes.com/local/lanow/la-me-familial-dna-
20161023-snap-story.html
a. AABB
b. ASCP
c. FDA Bibliography
d. HCFA AABB. Guidance for standards for parentage testing laboratories.
5th ed. Bethesda (MD): American Association of Blood Banks;
References 2002.
AABB. Standards for parentage testing laboratories. 5th ed. Bethesda
1. Bernstein F. Ergebnisse einer biostatischen zusammenfassenden (MD): American Association of Blood Banks; 2001.
Betrachtung über die erblichen Blutstrukturen des Menschen. AABB. Annual report summary for relationship testing laboratories
Klin Wschr, 1924;3:1495. [Internet]: Bethesda (MD): American Association of Blood Banks;
6888_Ch24_515-530 29/10/18 11:35 AM Page 530

530 PART IV Leukocyte Antigens and Relationship Testing

2008. Available from: www.aabb.org/sa/facilities/Documents/ Edwards M, Allen RW. Characteristics of mutations at the D5S818
rtannrpt08.pdf. locus studied using a tightly linked marker. Transfusion.
Allen RW, Wallhermfechtel M, Miller WV. The application of re- 2004;44:83–90.
striction fragment length polymorphism mapping to parentage Polesky HF. Parentage testing: use of DNA polymorphisms and
testing. Transfusion. 1990;30:552. other genetic systems. In: Henry JB editor. Clinical diagnosis and
Brinkmann B, Klintschar M, Neuhuber F, Huhne J, Rolf B. Muta- management by laboratory methods. 20th ed. Philadelphia: WB
tion rate in human microsatellites: influence of the structure Saunders; 2001. pp. 1390-1401.
and length of the tandem repeat. Am J Hum Genet. 1998; Walker RH. Molecular biology in paternity testing. 1992;Lab Med.
62:1408-15. 23:752.
Committee on DNA Forensic Science. The evaluation of forensic
DNA evidence: an update. Washington (DC): National Academic
Press; 1996. pp. 53-4.
6888_Ch25_531-551 29/10/18 11:34 AM Page 531

Quality Management and


Compliance Part V

Chapter 25
Quality Management in the Blood Bank
Laurie Gillard, MS, MLS(ASCP)CM SBB, Theresa Bostock, MS, MT(ASCP)SBB
and Lucia M. Berte, MA, MT(ASCP)SBB, DLM; CQA(ASQ)CMQ/OE

Introduction Personnel LEAN


Compliance Versus Quality Management Purchasing and Inventory A Quality Management System for
The Foundation of Quality Equipment Medical Laboratories
Quality Control Process Management The Cost of Quality
Risk Assessment (RA) Documents and Records Conformance Costs
Quality Control Plan (QCP) Information Management Importance of a Facility Quality
Quality Assessment Nonconformance Management Management System
Quality Assurance Assessments Case Studies
Quality Management System Continual Improvement Case 25-1
Quality Management System Essentials Process Improvement and the Quality Case 25-2
Organization Management System Summary Chart
Customer Focus Corrective Action and Preventive Action Review Questions
Facilities and Safety Define, Measure, Analyze, Improve, Control References

OBJECTIVES
1. Explain the differences between compliance and quality management.
2. List the three key components of quality.
3. Explain the purpose of Centers for Medicare and Medicaid Services creating an individualized quality control plan (IQCP).
4. Describe the framework of a quality system for a blood bank and a medical laboratory.
5. List the 12 quality management system (QMS) essentials and the blood bank operations to which they are applied.
6. Describe a process using a flowchart.
7. Explain the role of validation in introducing a new process.
8. Identify five blood bank process controls.
9. Describe the differences between a form and a record.
10. Explain the importance of document control for procedures and forms.
11. State the differences between remedial and corrective action.
12. Describe the role of auditing in a QMS.
13. Describe the process steps of define, measure, analyze, improve, control (DMAIC) methodology.
14. List and describe four types of quality costs.
15. Explain how the principles of the QMS of a blood bank can be applied to the medical laboratory.

531
6888_Ch25_531-551 29/10/18 11:34 AM Page 532

532 PART V Quality Management and Compliance

Introduction regulatory and accreditation inspections only find deviations


and deficiencies after they have occurred. Facilities need to
One of the definitions of quality states that it is the degree to design work processes in a way that prevents deficiencies and
which a product or service meets requirements. Blood banks errors from occurring in the first place.
must provide quality to their customers in many forms, Quality management (QM) is actively and continuously
including: practiced by blood bank leaders, managers, and staff
• Safe, satisfying donation experiences for blood donors. throughout all blood bank operations. With QM, the blood
• Accurately labeled and tested blood components provided bank is always ready for an inspection because it validates its
to transfusion services. processes, monitors process performance, and identifies
• Timely, accurate transfusion services provided to physi- where the problems are. The root cause is then determined
cians and other health-care personnel. and actions are taken to correct the situation. The correction
• Safe and efficacious blood transfusions to patients. process includes identification, resolution, and documenta-
tion as well as communicating to laboratory personnel, when
Blood centers, hospital blood banks, and transfusion ser- appropriate, to reduce or eliminate recurrence of the problem.
vices need to embrace the following quality philosophy to Quality is not something we do in addition to our job, it is
ensure a high degree of safe blood donation and transfusion built into our job.
practices to regulatory agencies, accrediting agencies, blood
donors, physicians, patients, and patients’ families: The Foundation of Quality
Quality, safety, and effectiveness are built into a product; qual-
The fundamental components of quality management are
ity cannot be inspected or tested into a product. Each step in
the process must be controlled to meet quality standards.1
quality control (QC), quality assurance (QA), and the quality
management system (QMS). Figure 25–1 demonstrates the
The method for bringing this quality philosophy into all structure of a quality management system (QMS) and the
operations involves each facility developing a foundation of internationally accepted definitions.8
quality: quality control (QC), quality assurance (QA), and
quality management system (QMS). When the key compo- Quality Control
nents are assembled, and the facility’s management and staff
are actively involved in monitoring and maintaining the All blood bank medical laboratory scientists and technicians
QMS, quality management (QM) has been achieved. are familiar with routine blood bank QC procedures, such
as daily testing the reactivity of blood typing reagents. Man-
dated equipment functions checks include calibrating sero-
Compliance Versus Quality
logic centrifuges; timer checks; and monitoring temperatures
Management
Blood bank compliance with federal regulations and accred-
itation standards is required by the following organizations:
• United States Food and Drug Administration (FDA)2,3 Quality
• The Joint Commission4,5 Management
• College of American Pathologists (CAP)6
• AABB (formerly known as American Association of Blood Activities of the management function
that determine the quality policy
Banks)7
Compliance programs evaluate how effectively the facil- Organizational structure,
ity meets regulatory and accreditation requirements by de- Quality procedures, processes,
tecting errors, deficiencies, and deviations. Compliance System and resources needed to
implement quality management
inspections (also called surveys or assessments) measure the
state of the facility’s program with respect to the applicable Planned, systematic activities
requirements and are usually conducted every 2 years. The Quality implemented within the quality
Assurance system to provide confidence
purpose of compliance is to ensure the organization is meet- that requirements for quality
ing applicable requirements. Once the survey inspection has will be fulfilled
been performed, whether or not deviations or deficiencies
were identified, the organization must have policies and Quality Operational techniques and
activities used to fulfill the
processes in place to support and maintain their compliance Control requirements for quality
practices.
In the event subsequent inspections reveal the same de-
Figure 25–1. The structure of a quality management. (Definitions from Interna-
viations and deficiencies, it is often because the facility’s cur- tional Organization for Standardization: ISO 9000:2005 Quality management
rent QC and QA programs did not identify and resolve the systems—Fundamentals and vocabulary. International Organization for Standardiza-
fundamental quality problems. It is important to remember tion, Geneva, 2015.9)
6888_Ch25_531-551 29/10/18 11:34 AM Page 533

Chapter 25 Quality Management in the Blood Bank 533

of refrigerators, freezers, and thawing devices. Requirements for testing QC. The medical director is responsible for re-
for the type and frequency of QC are determined by regula- view, approval, and signing the QCP before patient testing
tions and accreditation standards, manufacturers’ package and reporting of results commences.
inserts, and state and local requirements. Regular perform-
ance of QC reveals when a test method is not working as Quality Assessment
expected.
On January 1, 2016, the Centers for Medicare and Medi- The quality assessment requires monitoring for effectiveness
caid Services (CMS) released an alternative Clinical Labo- of the IQCP as well as for potential changes that could
ratory Improvement Amendments (CLIA) quality control introduce risk. Quality assessment monitors could include
option, called an individualized quality control plan reviews of proficiency testing records and QC results, turn-
(IQCP).9-12 The frequency of testing quality control mate- around time reports, personnel competency records, and cor-
rials is determined by the manufacturer’s instructions (the rective action reports. Good monitors identify problems or
minimum required QC per the manufacturer) and by the demonstrate that the IQCP is fulfilling its function.
individual laboratory’s risk assessment. IQCP applies to In transfusion services and blood banks, the manufacturer
the entire testing process, which includes preanalytic, an- already requires daily QC testing for testing reagents and
alytic, and postanalytic processes for all nonwaived testing. methods. However, an IQCP also applies to testing that does
IQCP allows each laboratory to customize its QC program, not occur daily, such as with test kits and antibody identifi-
depending upon its unique testing. The three parts of cation panels. Adoption of IQCP is voluntary; however, lab-
IQCP require a risk assessment, a quality control plan oratories may find they can reduce the amount of QC
(QCP), and quality assessment. performed, and ultimately implementing an IQCP can be a
cost savings measure.
Risk Assessment (RA)
Quality Assurance
In the risk assessment, five areas must be evaluated for po-
tential errors (examples of risk factors are provided but are Quality assurance is a set of planned actions that ensure that
not all inclusive): systems and elements that influence the quality of the prod-
• Specimen uct or service are working as expected, individually and
a. Patient preparation collectively.8 Quality assurance looks beyond the perform-
b. Specimen collection ance of a test method or piece of equipment; it addresses
c. Specimen labeling how well an entire process (i.e., a sequence of activities) is
• Test system functioning. This is particularly important in processes that
a. Failure of system checks go across functional or departmental lines. For example,
b. Detection of clots, hemolysis, lipemia a blood center could monitor the number of times and rea-
c. Transmission of data to LIS sons why a set of whole blood units transported from the
• Reagent collection site to the component processing site did not
a. Shipping/receiving arrive in time or was not in an acceptable condition to make
b. Storage conditions blood components. In the transfusion service, it is impor-
c. Preparation tant to monitor the source, the number of times, and the
• Environment reason why specimens collected for compatibility testing do
a. Temperature not meet predetermined acceptance criteria. Box 25–1 lists
b. Airflow/ventilation common QC and QA activities and indicators practiced by
c. Humidity most blood banks.13 Box 25–2 gives an example of IQCP in
• Testing personnel the blood bank.
a. Training
b. Competency Quality Management System
c. Staffing
A quality guideline published by the FDA1 sets the meaning,
Quality Control Plan (QCP) emphasis, and organization of quality activities for blood
banks. Accrediting agencies such as the Joint Commission
CLIA requires all laboratories to have QC procedures to and AABB have established their quality requirements to be
monitor the accuracy and precision of patient testing. more comprehensive and more coordinated than QC and
Once the risk assessment has been completed and the data QA. A QMS provides a framework for applying quality prin-
analyzed, the next step is to establish the QC plan (QCP). ciples and practices uniformly across all blood bank opera-
The QCP must include the number and type of QC testing tions, starting with donor selection and proceeding through
materials, the frequency of testing, and the criteria for ac- transfusion outcomes.
ceptable QC results. At the very minimum, the QCP must In its Standards for Blood Banks and Transfusion Services,7
be at least as stringent as the manufacturer’s instructions the AABB defined quality system essentials (QSEs) for
6888_Ch25_531-551 29/10/18 11:34 AM Page 534

534 PART V Quality Management and Compliance

demonstrates that the QMS essentials are the foundation that


BOX 25–1 supports blood bank operations in the path of workflow for
Common Blood Bank QC Activities and QA Indicators both donor centers and transfusion services.
QC Activities QA Indicators
Quality Management System
Donor Collection • Number of donor forms Essentials
• Microhematocrit instrument with incomplete or incorrect
controls information The QMS essentials are in a logical order that can be ex-
• Hemoglobin instrument • Number and types of plained as follows for any new or changed product or service.
controls unusable units and blood
components The blood center or transfusion service organization develops
• Apheresis equipment function its mission, vision, values, goals, and objectives around the
check • Number of blood typing
discrepancies in donors and products and services it plans to offer. The facility should
• Blood-weighing scales
accuracy patients have a customer focus by determining its customers’ needs
• Number of and reasons for and expectations and designing its processes and procedures
Blood Components invalid tests to meet those needs and all regulatory and accreditation re-
• Red blood cell hematocrit • Number of and reasons for quirements. The blood center or transfusion service should
• Cryoprecipitated labeling check failures have the physical facilities to support the products and ser-
antihemophilic factor • Number and source of vices offered and ensure that safety requirements are met. The
• Platelet counts in platelet improper and incomplete
requests for blood facility determines the qualifications needed for each job and
units
components hires qualified personnel who are then trained and expected
• Residual leukocyte counts in
leukocyte-reduced • Number and location of to maintain competence in their assigned duties.
components patients without proper Before products and services can be provided to customers,
• Bacterial contamination of identification at time of equipment and materials need to be purchased and main-
platelet units specimen collection or tained in inventory. Equipment must be managed according
transfusion
Reagent Controls to manufacturer, regulatory, and accreditation requirements.
• Number of, source of, and
reasons for unacceptable Before any work commences, the facility needs to design, doc-
• Copper sulfate
specimens ument, and validate work processes with appropriate manage-
• Reagent red blood cells
• Number of times wrong ment to ensure their correct performance. Documents provide
• Reagent antisera
component or ABO was instructions for how the work generates records of work per-
• Test kits for infectious disease selected for crossmatch
testing formance. Patient and donor information is managed in a way
or use to ensure that requirements for confidentiality and informa-
Laboratory Equipment Function • Number and type of tion integrity are met.8 The process of capturing information
Checks transfusion complications
on nonconformances is managed to identify recurring process
• Heating instruments • Number of and reasons for
turnaround time failures problems that can affect patient safety, laboratory credibility,
• Water baths and the operating budget. Internal and external assessments
• Thawing devices for blood measure and monitor the facility’s performance to identify op-
components
portunities for improvement. The facility should constantly
• pH meters
strive for continual improvement.
• Cell counters
• Centrifuges, refrigerated and
serologic Organization
• Cell washers
The organization’s type and size determine the configuration
• Blood irradiators
of the blood bank’s QMS. In hospitals, there is usually an
• Refrigerators
organization-wide quality department that prioritizes and
• Freezers
coordinates quality projects, approves resources, and receives
• Platelet incubators
reports and information from all hospital departments. The
• Blood warmers
hospital-based blood bank or transfusion service must par-
• Shipping containers
ticipate in blood bank quality-related activities, laboratory-
wide quality initiatives, and the hospital’s continual
improvement program. The blood bank should state in writ-
ing its policies, goals, and objectives for each of the QMS
blood collection and transfusion service facilities and the
essentials and relate them to the bigger laboratory and hos-
blood bank operations to which they are applied. Box 25–3
pital quality goals. There should be an organizational chart
lists the QSEs and blood bank operations on which the AABB
showing the following:
assesses blood banks and transfusion services in its accredi-
tation program. • Relationships among blood bank personnel by job title
The next sections of this chapter describe the blood bank’s • The blood bank’s link to the laboratory
role and responsibilities in fulfilling QMS essentials, which • The blood bank’s link to the hospital
extend far beyond historic QC and QA practices. Figure 25–2 • How the blood bank links to the hospital’s quality function
6888_Ch25_531-551 29/10/18 11:34 AM Page 535

Chapter 25 Quality Management in the Blood Bank 535

BOX 25–2

Individual Quality Control Plan (IQCP)10–13


How do you create an individual quality control plan (IQCP) in a Severity of harm to patient
transfusion service or blood bank? The first step is to review the • Negligible (temporary discomfort)
frequency of QC performance, determined by the manufacturer’s • Minor (temporary injury; not requiring medical intervention)
instructions and by establishing the laboratory’s individual risk assess-
ment for the specific test system. This example discusses a blood • Serious (impairment requiring medical intervention)
bank’s implementation of an IQCP for antibody identification reagent • Critical (life threatening consequences)
red cell panels. Step 2
The three steps involved in creating an individual quality control
plan (IQCP) include: A quality control plan (QCP) should be prepared after completion
1. Risk assessment: What can go wrong? of the risk assessment. Determining the frequency of QC requires
supporting documentation, including the number, type, and frequency
2. Quality control plan: What procedures are already in place or that of testing control materials. Note that the QCP must be reviewed,
you plan to implement to reduce harm? approved, and signed by the laboratory director and cannot be
3. Quality assessment: How will you monitor the effectiveness of delegated.
your plan?
Step 3
Step 1
Quality assessment (QA) will measure the effectiveness of the IQCP.
The risk assessment (RA) for an antibody identification (ABID) The IQCP needs to be monitored for changes and possible new risks.
reagent red cell panel would include looking at the test system: the Items to consider for monitoring effectiveness of a QCP may
specimen, personnel, environment, reagents, and respective reagent include, but are not limited to:
QC. Next determine whether the current practices are sufficient to • QC reviews
detect the sources of error or failure in your test system through
the review of previous variances, reagent QC problems/failures, • PT performance reviews
proficiency test failures, etc. Finally determine the frequency of • Chart reviews
occurrence of each current or newly identified source of harm and • Specimen rejection logs
its possible severity. • Turnaround time reports
A. Historical quality review • Complaint reports
• Validations of test system, automation for antibody identification,
software upgrades SUMMARY
• Inspection of reagents and panel for hemolysis or signs of The key points to remember for an IQCP include the following:
contamination • Always follow the manufacturer’s directions. Your IQCP may result in
• Daily QC: Daily quality control of screening cells is performed performing more QC than the manufacturer requires, but you cannot
• Proficiency results for antibody identification do less than required. For example, when the package insert says
to perform QC on ABO typing reagents daily, you cannot decide to
• Correlation of the results of antibody screen, antigen phenotype, perform QC weekly based on your IQCP risk assessment.
and patient’s previous history
• To determine whether you need to do an IQCP in the blood bank,
B. Determination of risk level = Frequency × Severity evaluate every reagent, method, or kit for which you don’t perform
Frequency of occurrence: QC at least daily.
• Unlikely (once every 2–3 years) Please note: Inspectors seek to confirm that an annual review has
• Occasional (once a year) been performed on the IQCP and to determine whether any risks have
• Probable (once a month) changed (such as adding a new reagent manufacturer). The medical
director must review and sign the IQCP annually.
• Frequent (once a week)

A freestanding blood center must develop and manage the the entity or person who receives and must be satisfied with
entirety of its QMS. There may be a quality council repre- a product or service.9 For blood centers the customers are
sented by top-level management from the various depart- the donors, who want a safe and satisfying donation experi-
ments. The council develops the QMS policies, goals, and ence, and the transfusion services served by the blood center,
objectives and its strategies for QMS implementation; pro- that want properly tested and labeled blood components of
vides resource support; prioritizes identified improvement the appropriate types on demand. For transfusion services,
projects; and provides support for cross-functional process customers are the physicians, who want the blood transfu-
improvements. There may also be a quality steering commit- sions they order to occur in a timely manner, and the nurses,
tee composed of senior managers and department staff who who want the correctly issued blood components in a timely
operationalize the quality strategies and implement process manner for administration to patients. For all types of facil-
improvements. ities, internal customers are the personnel.
The important issue about having a customer focus is to
Customer Focus understand the customer’s needs and to design the work
processes and procedures to meet those needs. In addition
Although both blood centers and transfusion services work to fulfilling customer needs, the processes and procedures
on behalf of patients, the real customer of these facilities is must also meet regulatory and accreditation requirements.
6888_Ch25_531-551 29/10/18 11:34 AM Page 536

536 PART V Quality Management and Compliance

addition, any blood bank that performs irradiation of blood


BOX 25–3 components must also have a radiation safety program and
QMS Essentials and Blood Bank Operations document appropriate training.
A freestanding blood center must develop its own facility
QMS Essentials7 • Preparation of components
management program. All regulations and accreditation re-
• Organization • Testing of donor blood
quirements for emergency preparedness, chemical hygiene,
• Resources • Final labeling
infection control, and radiation safety training and documen-
• Equipment • Final inspection before distri-
bution tation also apply.14
• Suppler and customer issues
• Process control • Patient samples and requests
• Serologic confirmation of
Personnel
• Documents and records
donor blood ABO/D
• Deviations, nonconformances, Quality begins and ends with people. The blood bank
and adverse events • Pretransfusion testing of pa-
tient blood management has to work with the human resource depart-
• Assessments: Internal and ment of the blood center or hospital to define qualifica-
external • Selection of compatible blood
and components for transfusion tions for all blood bank jobs and to write job descriptions
• Process improvement through that include educational qualifications, experience, and
corrective and preventive action • Crossmatch
• Special considerations for federal, state, and local licensing requirements, where ap-
• Facilities and safety
neonates plicable, so that qualified persons can be hired. Box 25–4
Blood Bank Operations7 • Selection in special circum- lists the major types of training that personnel need to re-
• Donor qualification stances ceive once they are hired. This training extends signifi-
• Autologous donor qualification • Issue for transfusion cantly beyond the task specifics of a particular job.
• Apheresis donor qualification • Blood administration Periodic evaluation and documentation of the continuing
• Blood collection • Rh-immune globulin competence of personnel to perform their assigned job
• Cytapheresis collection functions and tasks is also required. Competence assess-
ment must include:
1. Direct observation of patient test performance.
2. Monitoring the recording and reporting of test results.
Facilities and Safety 3. Review of test results or worksheets, QC records, profi-
ciency testing results, and preventive maintenance
In hospitals, the Joint Commission mandates an environ-
records.
mental control program that addresses all significant
4. Direct observation of performance of instrument mainte-
environmental issues for facility management and mainte-
nance and function checks.
nance, such as temperature control, electrical safety, fire pro-
5. Assessment of test performance through testing previ-
tection, and so forth.5 The Joint Commission also requires
ously analyzed specimens or external proficiency testing
that hospital laboratories have training programs for all
samples.
laboratory personnel on emergency preparedness, chemical
6. Assessment of problem-solving skills.
hygiene, and infection control.5 Therefore, hospital-based
blood banks and transfusion services are already participat- Evaluating and documenting the performance of testing
ing in the facility’s management and safety training. In personnel must occur at least semiannually during the

A Quality Management System for the Blood Bank


Blood Bank Path of Workflow

Component Collections, Preparation, Testing, Labeling, Distribution, Compatibility Testing,* Issue, Administration

Quality System Essentials: The Building Blocks

The Blood Bank The Work The Performance

Organization Process Management Nonconformance Mgmt


Customer Focus Documents & Records Assessments
Facilities & Safety Information Management Continual Improvement
Personnel
Purchasing & Inventory
Equipment

*Links with Laboratory Path of Workflow, Figure 25-9.

Figure 25–2. A quality system for the blood bank, showing the relationship of quality management system essentials to blood bank operations.
6888_Ch25_531-551 29/10/18 11:34 AM Page 537

Chapter 25 Quality Management in the Blood Bank 537

to have effective processes for managing inventories of


BOX 25–4 reagents, supplies, and blood components.
Training for New Employees Blood centers need to have specified processes for select-
ing vendors of equipment, supplies, and services, and for en-
Orientation
tering into and amending agreements. In addition, blood
• Organization centers must also have processes for receipt, inspection, and
• Department testing (where required) of incoming critical materials (such
• Section as blood bags and infectious disease testing kits), blood com-
Quality Training ponents (such as leukocyte-reduced platelets), and blood
• Current good manufacturing practice (cGMP) products (such as albumin, clotting factor concentrates, and
• Quality management systems (QMS) immune globulins).
Computer Training
Equipment
• Facility information system (hospital, blood center)
• Department information system (e.g., laboratory) Before putting equipment into operation, the blood bank
Safety Training needs a process to ensure that installation, operational, and
performance qualifications for the piece of equipment have
• Emergency preparedness
been met. Installation qualification demonstrates that the
• Accident reporting
equipment is correctly installed; operational qualification
• Chemical hygiene program
ensures the equipment operates as intended; and perform-
• Hazardous waste disposal program
ance qualification evaluates the personnel, procedures,
• Infection control (including universal precautions, bioterrorism)
and supplies in the facility work environment.1 Schedules
• Radiation safety, where applicable
for calibration, preventive maintenance, RPM and timer
Work Processes and Procedures Training checks, and monitoring of temperature-regulated equip-
• Work processes performed on the job ment are required, with frequencies determined by regula-
• Procedures performed tions, accreditation requirements, manufacturers’ written
Compliance Training instructions, usage, testing volume, and equipment relia-
bility. Defective equipment and instruments must be iden-
• Medicare necessity requirements
tified and repaired when necessary. Records must be kept
• Fraud and abuse reporting
of all installation, calibration, maintenance, servicing, and
• Reporting quality and safety concerns
repair activities in conformance with federal and state reg-
ulatory requirements.16

Process Management

first year the person tests patient specimens and annually A process can be defined as a set of interrelated resources
thereafter.15 and activities that transforms inputs into outputs. Process
A competency plan should be developed that includes a control is a set of activities that ensures a given work
schedule of the activities to be completed. The plan should process will keep operating in a state that is continuously
also indicate what the criteria are for acceptable performance able to meet process goals without compromising the
and what remediation actions would be needed should the process itself. Total process control is the evaluation of
person require additional competency assessment. The out- process performance, comparison of actual performance to
comes of the annual competency testing should be analyzed a goal, and action taken on any significant difference.
to identify staff learning needs. Process control is a means to build quality, safety, and effec-
tiveness into the product or service from the beginning. It
Purchasing and Inventory is important to understand and document the sequence of
activities in a process, develop written procedures for the
In hospital-based blood banks and transfusion services, the individual activities, validate the entire process to ensure it
hospital’s purchasing department is often responsible for works as expected before actual use, measure process pa-
vendor contracts and purchasing decisions. Blood bank per- rameters to see they stay in control, and understand when
sonnel may or may not have control over the specific ven- and why the process has variations.
dors with which the organization has agreements to purchase The FDA has published its requirements for process con-
blood components, reagents and kits for testing, equipment, trol as the current Good Manufacturing Practices (cGMP) that
and other important supplies and materials. At a minimum, are cited in the Code of Federal Regulations.2,3 The cGMP
hospital-based blood banks and transfusion services need to requires that facilities design their processes and procedures
have a process by which incoming blood components and to ensure that blood components are manufactured consis-
critical supplies are inspected and tested (where required). tently to meet the quality standards appropriate for their
In addition, blood banks and transfusion services need intended use.
6888_Ch25_531-551 29/10/18 11:34 AM Page 538

538 PART V Quality Management and Compliance

Flowcharts involves ordering the test, collecting an appropriate speci-


Figure 25–3 is a sequential flow of the elements of total men, delivering it to the laboratory, performing the test, and
process control. An effective tool for understanding any reporting the results. Different people order the test, collect
process is the flowchart. Flowcharts graphically represent the specimen, perform the ABO/Rh test, and deliver the
the sequence of activities in a process and show how the in- results. For this process, there should be specific written
puts are converted into outputs. Flowcharts help to develop procedures for ordering tests manually or in the computer
a common understanding of a process. Mapping a facility’s system, collecting and labeling the blood specimen, deliv-
current work processes reveals bottlenecks, missing actions, ering the specimen to the laboratory, performing and
decision points, dead ends, and choices that can lead to recording the ABO/Rh testing, and reporting the results. In
errors, delays, and unnecessary work. Mapping a new a written procedure, the instructions are presented in step-
process facilitates understanding of where human and other wise order.
resources will be needed for successful accomplishment.
Flowcharts can be created on paper or with commercially Validation
available software programs. Figure 25–4 is a sample flow- To ensure that new processes will work as intended, they
chart for the activities in the pretransfusion testing process. must be validated before being put into use. Validation
It illustrates different decision points and test method tests all activities in a new process to ensure that the
choices. process will work in the live environment. For example,
when a new test for a transfusion-transmitted disease is
Standard Operating Procedures added to those performed on donated blood units, the new
A work process involves one or more persons who per- testing process—with its associated instruments, test kits,
form a sequence of activities over a period of time. A computer functions, and procedures—must be validated
process flowchart visually describes the sequential activi- in each blood bank that will perform the new test. The
ties to provide an overview of how the process occurs. The validation ensures that the new test will perform as
boxes in the flowchart represent activities performed by expected with the blood bank instrumentation, written
one person. procedures, personnel, and computer systems. The culmi-
Standard operating procedures provide instructions for nation of this activity is a validated process and set of new
each activity in the larger process. For example, the process procedures on which all personnel who will perform the
to provide a physician with a patient’s ABO and Rh type new test must be trained. Training must be documented
and personnel must demonstrate competence in the new
processes and procedures before these processes and
procedures are implemented in the live environment.
Total Process Control
Process Controls
Process is flowcharted
It is essential to monitor processes to ensure they are
performing as required, to correct process problems before
they affect output, and to improve processes to meet
Procedures are identified
changing needs and technology. Routine process controls
include

Procedures are drafted • QC of test methods and reagents,


• Review of work, QC, and equipment records, and
• Capture of nonconformances when the process did not
perform as expected.
Process is validated
These routine process controls monitor whether a process is
functioning as needed.
Procedures are finalized Proficiency testing is another example of a process con-
trol. In proficiency testing, the methods and procedures of
one laboratory are compared with those of other laborato-
Training is performed ries for the ability to get the same result on a set of un-
known specimens. Regulations require that all laboratories
participate in proficiency testing for diagnostic laboratory
Initial competence is assessed testing. Blood bank proficiency test challenges include
serologic testing for blood types, detecting and identifying
unexpected antibodies, checking compatibility of cross-
Process performance is monitored matched blood, testing for diseases transmitted through
blood transfusion, and checking for contamination of pre-
Figure 25–3. The sequential activities in total process control. pared blood components.
6888_Ch25_531-551 29/10/18 11:34 AM Page 539

Chapter 25 Quality Management in the Blood Bank 539

Sample arrives

Sample and request are accessioned

Sample and request are evaluated

Red blood cell request is evaluated

Patient history is checked

Sample is processed

Automated
MANUAL TESTING AUTOMATED TESTING testing
process

ABO/D is done ANTIBODY SCREEN IS DONE

Or

Is there an Tube IAT screen Gel screen Solid phase screen


ABO or D
discrepancy?
Yes

ABO discrepancy is
resolved
and/or No
D discrepancy is
resolved Is the Yes
antibody screen and/or
positive?
Antibody Send
No identification out
process process

Are
red blood cell units
needed now? Yes

Crossmatch
No process
Sample and request are archived

Figure 25–4. An example of a flowchart for the pretransfusion testing process. (Adapted from Berte, LM (ed): Transfusion Service Manual of SOPs, Training Guides and
Competence Assessment Tools, 2nd ed. American Association of Blood Banks, Bethesda, MD, 2007.)

Other process controls include manual and automated Documents and Records
actions to prevent the occurrence of errors. One common
process control in serologic testing is the use of green- Documents are approved information contained in a
colored antiglobulin serum to ensure that the antiglobulin written or electronic format. Documents define the QMS
serum was indeed added at the antiglobulin phase of testing. for external inspectors and internal staff. Examples of doc-
Another common process control is the addition of IgG- uments include written policies, process flowcharts, pro-
coated reagent red blood cells after the antiglobulin phase cedures and instructions, forms, manufacturers’ package
reading to ensure that the antiglobulin serum was working. inserts, computer software and instrument operator man-
Computer process controls include automatic comparison uals, and copies of regulations and accreditation require-
of current blood type interpretation with the previous com- ments. Records capture the results or outcomes of
puter records on the same donor or patient to prevent ABO performing procedures and testing on written forms or
errors and warning signals when ABO-incompatible units are electronic media, such as manual worksheets, instrument
issued for transfusion. printouts, tags, or labels. Both documents and records
6888_Ch25_531-551 29/10/18 11:34 AM Page 540

540 PART V Quality Management and Compliance

must be controlled to provide evidence that regulations control system includes instructions for creating documents
and accreditation requirements are being met. in approved formats, assigning document identification and
version designation, approving new and revised documents,
Document Control preparing a master document list, and maintaining docu-
A structured document control system links a facility’s poli- ment history files. This change control process usually re-
cies, processes, and procedures and ensures that only the lat- quires the completion and routing of a form that contains
est approved copies of documents are available for use. A information about the reason for a new document or a
typical document control system is structured as a pyramid, change to an existing one. Other important change informa-
as shown in Figure 25–5. The following example best illus- tion includes when the change was requested; who requested
trates how a blood bank should link its policies, processes, the change; what other documents, if any, are affected by this
and procedures. change; and dates for approval, training, and in-use.
The blood bank should have a written policy document A master copy of the new, approved version is added to
stating that the blood bank maintains a process and rele- the master document history file, which contains copies of
vant procedures for correcting erroneous entries or results all previous versions of that same document. The hard copies
on a paper record or in the computer. A second document within this master file capture the original signatures and
(such as a process flowchart) should describe the se- provide a paper backup when electronic files cannot be ac-
quence of activities in identifying the need for a correc- cessed. The master document list is updated with the new
tion, obtaining any necessary approvals for the changed version number. When a revised version of a document is
information, making the correction in the computer or on ready for release, the distribution process must be controlled
paper (or both), and notifying all appropriate parties of to ensure that copies of the obsolete document (e.g., proce-
the correction. Procedure documents instruct personnel dures in a working manual or a form) are replaced with the
how to record the need for a correction, how to obtain any new version and the obsolete copies are destroyed. Personnel
necessary approvals for making the change, how to prop- should not keep and refer to copies of procedures and forms
erly record a change to an entry on a paper record, how hidden in lockers, drawers, and personal files. Use of unap-
to properly record a change to an entry in a computer proved, outdated documents could lead to errors or omis-
record, and how to notify appropriate parties of the sions that could cause harm to patients.
change and document the notification. The process and
procedures should be written in a way that ensures that Records Management
all regulatory and accreditation requirements are met. Forms are specially designed documents—either paper or
Recording the actions taken throughout the process pro- electronic—on which are recorded the results or outcomes
vides a tracking record that the requirements were met. of performing a given procedure. Forms are subject to the
The blood bank needs a system to control identification, document control activities described in the previous para-
approval, revision, and archiving of its policies, process graph. The document identification system should link the
descriptions, procedures, and related forms. The document form to its respective procedure. When a form (either paper
or electronic) is filled out, it becomes a record. Regulations
and accreditation requirements mandate the review of
Quality Management System Documentation
records by authorized personnel and specify the type of
records and length of time they are stored for possible future
reference. State, local, and facility requirements for record
retention periods may also apply.15

Information Management
Policy
(What will Whereas documents provide information about what to do,
be done)
records tell what happened, who did it, and when it was
done. Information management activities include using and
manipulating the donor or patient information and test re-
Process sults in the facility’s paper and electronic information sys-
(How it happens) tems. For example, maintaining the confidentiality of
privileged donor and patient information is mandated by
government regulations,16 and the facility must ensure its
processes preserve that confidentiality. The facility must also
Procedure ensure the integrity of any data or information sent inside
(How to do it) or outside the facility, whether the medium used is paper or
electronic. For example, the facility should verify that infor-
mation faxed or sent across instrument and computer inter-
Figure 25–5. A simple structure for organizing QMS documents. faces is identical to the original information.
6888_Ch25_531-551 29/10/18 11:34 AM Page 541

Chapter 25 Quality Management in the Blood Bank 541

Nonconformance Management information about events that occurred across the entire path
of workflow for blood collection and transfusion service activ-
FDA regulations require that blood banks report any error ities whether or not a patient was involved.
or accident in the manufacture of blood components that All personnel need to participate in nonconformance re-
may affect the safety, purity, potency, identity, or effectiveness porting. It is essential that staff not perceive nonconformance
of the component or that compromises the safety of the reporting as a tool for disciplinary action. Instead, all staff
blood donor or recipient. Each blood bank must have a must understand that nonconformances represent blood
process for detecting, reporting, evaluating, and correcting bank processes that do not work as they should, so knowl-
deviations and any nonconformance with its own proce- edge of these problems provides opportunities for improve-
dures, products, or services. Nonconformance management ment. Nonconformances are usually identified by staff in the
is one name for such a process. Other commonly used names course of routine activities or by supervisors during review
include occurrence, incident, or variation management. of records. Complaints received in the blood bank are also
Table 25–1 provides definitions of terms used to further clas- considered nonconformances. The facility needs to capture
sify occurrences of blood bank nonconformances. information about all nonconformances, including those
identified before blood components are distributed or issued.
Nonconformance Reporting
Personnel should be trained to recognize and report such
Information about events involving the blood bank that deviate events. When reporting, the person who discovered the non-
from accepted policy, process, or procedure need to be captured conformance describes the who, where, and when of the
and acted upon. Hospitals usually have a risk management pro- nonconformance and then briefly describes what happened
gram in place; this program type typically captures information and what he or she did at the time to remediate the problem.
about events involving patient and visitor safety that could A standard report form can be used to capture information
result in financial loss to the facility. An internal blood bank on each nonconformance (Fig. 25–6).
nonconformance management system captures and analyzes
Investigation and Corrective Action
Supervisors and those trained in quality should enter the
Table 25–1 Common Classifications nonconformances into a spreadsheet or database so that the
of Various Types of resolution process can be tracked. The nonconformances are
Nonconformances reviewed, investigated, and further classified as errors, acci-
dents, complaints, or another definition.
Nonconformance The immediate action, also known as remedial action,
Type Definition does not uncover the real cause of the nonconformance,
Accident Nonconformance generally not attrib- which can be determined only through investigation. Inves-
utable to a person’s mistake, such as tigation of complaints or errors provides an opportunity to
a power outage or an aged instru- identify underlying factors that contributed to the noncon-
ment’s malfunction
formance. Process-improvement tools are used to identify
Adverse reaction Complications that occurred to the these contributing factors and to determine the best way to
donor during or after the donation remove them through implementing corrective action.
process or to the recipient of trans- Often, corrective action involves making changes in the
fused blood components
process. All personnel performing the process must be
Complaint Expression of dissatisfaction from inter- informed of the changes and retrained when necessary.
nal customers (physicians, employ- Sometimes the corrective action involves retraining only
ees) or external customers (donors, specific individuals who may not have been adequately
patients)
trained or who have been making unapproved deviations
Discrepancy Difference or inconsistency in the out- from the established processes or procedures.
comes of a process, procedure, or The nonconformance reporting process must be clearly
test results defined so that information is tracked and acted upon and
Error Nonconformance attributable to a feedback is provided. The person responsible for the quality
human or system problem, such as a function in the blood bank reviews the nonconformance re-
problem from failure to follow estab- ports, assigns an accession number, and forwards the report
lished procedure or a part of a to the individuals who will be involved in the investigation.
process that did not work as expected
When an identified error or accident needs to be reported to
Postdonation The receipt of information (call or in- the FDA, the corresponding process is initiated.
formation letter) from a donor with An effective nonconformance management program will
additional details regarding his or her trend the nonconformances by category. Trend analysis is
donation, such as subsequent illness
or neglecting to mention an illness or
important for identifying problematic processes and provides
medication significant support when a process needs to be changed.
Facility staff must make a conscious decision not to simply
6888_Ch25_531-551 29/10/18 11:34 AM Page 542

542 PART V Quality Management and Compliance

NONCONFORMANCE REPORT FORM

Database number: __________________


ORIGINATOR: Reported by: ____________________________
Date and time occurred: _______________________________ Date and time discovered: _____________
Date and time reported: _______________________________

Explain the error / complaint / incident / nonconformance. Attach copies of any relevant records, as needed.

How was the error / complaint / incident / nonconformance discovered?

Describe what you did to take care of the immediate problem (remedial action):

INVESTIGATOR: Name: ________________________________


Findings of investigation: Attach copies of any related paperwork, as needed.

Will remedial action be effective in eliminating future nonconformances of this same type?
Circle one: Yes No

If no, what further action should be taken?

QUALITY DEPARTMENT: Name:________________________________ Date: _____________________

Is this nonconformance FDA reportable? Circle one: Yes No


If yes, initiate FDA-CBER report – see procedure.

What further action needs to be taken?

System, process, procedure, form change needed to reduce or eliminate recurrence:

DATABASE ADMINISTRATOR
Entered into database by: ______________________________________ Date: _____________________

Figure 25–6. An example of a nonconformance report form.

respond with remedial actions but rather to use nonconfor- operations need to be assessed. Compliance inspection and
mance information for removing the underlying root causes other checklists4–7 can be used as assessment tools; however,
of the problem and to make improvements that truly con- these assess the adequacy of only the listed items. The qual-
tribute to the safety and efficacy of transfusion medicine. ity indicators (QI) monitored by hospital-based blood banks
and transfusion services as part of the laboratory QA pro-
Assessments gram are also helpful but do not usually cover all important
aspects of each operation.
Assessments measure the state of a facility’s quality program Common transfusion service quality indicators include
with respect to the applicable requirements at a single point the percentage of specimens received in the blood bank lab-
in time. Internal and external assessments are used for mea- oratory not acceptable for testing and the number of times
suring and monitoring performance and identifying oppor- the transfusion service met its established turnaround time
tunities for improvement. for emergency release of uncrossmatched blood to the emer-
gency department (see Table 25–1 for additional examples
Internal Assessments of quality indicators).
Blood banks need to have processes in place to continuously A very effective assessment tool is the internal audit. Un-
monitor the effectiveness of its QMS. (See Fig. 25–2 for a re- like compliance inspections, audits review a specific facility
view of the QSEs supporting the blood bank path of work- process and determine—by examining documents and
flow.) Both the quality essentials and the facility’s specific records, interviews, and observations—whether the facility is
6888_Ch25_531-551 29/10/18 11:34 AM Page 543

Chapter 25 Quality Management in the Blood Bank 543

meeting the applicable requirements. For example, in a retro- and performs the testing using its routine processes, proce-
spective audit, also referred to as a tracer audit, an auditor dures, and personnel. Results are compared to those of the
could randomly select a red blood cell component unit num- provider and the other laboratories, and the facility gets a re-
ber and track through each activity involved in how the com- port of its performance. Successful performance on profi-
ponent was received, tested, issued, and transfused. The ciency testing challenges is a requirement for laboratory
training and competence assessment records of each person licensure and accreditation.15 The second type of external as-
involved in handling the component are reviewed, as are the sessment is the external inspection, performed by regulatory
QC records for the reagents, and the equipment records for and accreditation organizations for the purposes of obtaining
the storage refrigerator, centrifuges, and other instruments and maintaining the facility’s license or accreditation. Exter-
used in compatibility testing for that unit. The performance nal assessments are periodically conducted by the FDA, the
on the proficiency test most recent to the unit testing is re- Joint Commission, the CAP, and the AABB to determine the
viewed. Copies of procedures and forms used at the worksta- facility’s compliance with the respective requirements.2–7,15
tions for all testing and QC are examined to determine
whether they are the most current version, according to the Continual Improvement
master list. Samples of records are reviewed for inclusion of
all required information, interpretations, and required super- Data gathered from quality management activities provides
visory reviews. information regarding how well laboratory processes are
For blood collection operations, a donor name or number functioning so that action can be taken to improve any prob-
could be randomly selected and the same process repeated lematic processes.
for the donation record, computer files, all related proce-
dures, training and competence records, component produc- Identifying Opportunities for Improvement
tion records, serologic and infectious disease testing records, Opportunities for improvement for both blood collection fa-
and related QC, labeling, storage, and shipping records. cilities and transfusion services can be identified from several
When prospective audits are conducted, the auditor main sources:
watches the staff performing activities in the selected
process. The auditor may ask questions of staff members • The nonconformance trending process points to opera-
such as, “How were you trained to perform this procedure tional processes that are not functioning as well as
and when?” or “Where in the procedure are the instructions intended
for what you just did?” • Customer feedback such as complaints, solicited feedback,
Audits should be conducted by personnel who have or suggestions from external customers the facility serves
been trained to perform audits and to identify system prob- and from internal customers (employees)
lems. Auditors should not audit the procedures they per- • Information derived from monitoring quality indicators
form in their jobs. In a hospital-based blood bank or of operations, particularly when it is benchmarked against
transfusion service, there may be insufficient personnel to peer groups in other institutions
have a separate quality function, and the supervisor or se- • Internal audit feedback, whereby objective evidence col-
nior personnel or staff from another laboratory area may lected by the auditor should support the facility’s under-
have to perform some auditing activities. A freestanding standing of why corrective action is needed and should be
blood center should have sufficient personnel to designate taken
a quality officer and to separate the quality function from • Feedback from external compliance inspections (AABB,
routine operations. CAP, or the Joint Commission) or reports from other
The auditor presents his or her findings to the appropriate departments in the hospital’s organization-wide quality
management and operations personnel at the closing meet- committee function, such as nursing or emergency depart-
ing on a form similar to that in Figure 25–7. The auditor may ment problems with the blood bank
request corrective action for each finding. A process should
be in place for management personnel to evaluate and review Process Improvement and the Quality
the audit to ensure that corrective actions were implemented. Management System
Follow-up audits may be necessary to ensure that the cor-
rective action was successful in removing the causes of the The quality system essentials (QSEs) provide a systematic
findings. Facilities must prepare an annual summary of their approach to achieving quality management in the blood
audit findings and the corrective actions taken. bank as well as for the entire medical laboratory. Two com-
ponents of a vigorous QMS are corrective action and preven-
tive action (CAPA) and continuous improvement, which are
External Assessments
both driven by data.
There are two types of external assessments—proficiency
testing and external inspections. Proficiency testing is a Corrective Action and Preventive Action
means to demonstrate that the facility’s testing processes pro-
vide results comparable to those of other facilities with the Whereas corrective action is defined as the action taken to
same instruments and methods. In proficiency testing, the identify and eliminate the cause of a nonconformance, pre-
facility receives samples for testing from a designated provider ventive action is defined as the action taken to reduce or
6888_Ch25_531-551 29/10/18 11:34 AM Page 544

544 PART V Quality Management and Compliance

QUALITY ASSURANCE INTERNAL AUDIT

Area/Function Assessed

Subject Area: ________________________________________________ Date: _____________________

Key Positive Findings:

Key Opportunity Areas:

Recommendations for Improvements:

Auditors: ____________________________________________________ Date: _____________________

Response: Planned Actions and Completion Dates

Area Mgmt.: _________________________________________________ Date: _____________________

Approved by: ________________________________________________ Date: _____________________

Figure 25–7. An example of an internal audit form.

eliminate the potential for a nonconformance to occur at all. the result of an internal audit or external assessment. A tool
Corrective action is often the immediate response to an often used for problem identification is the Pareto chart,
undesirable event and is a reactive approach to the problem. which graphically depicts data in the form of a bar chart
This method of responding may alleviate the problem; how- where the data being plotted are arranged in descending
ever, it doesn’t solve the problem and prevent its recurrence. order. Several Pareto charts may be used to stratify and cat-
This type of response is considered remedial and is often a egorize data and provide a focus for problem prioritization.
temporary solution. Once the problem to solve has been selected, the next step
Identifying and removing the root cause of a nonconfor- is defining the scope of the improvement project. The scope
mance is the only way to effectively reduce or eliminate its documents specific goals, also referred to as deliverables, and
recurrence. who will be involved. Scope also includes a timeline, pre-
senting a common understanding of what should be in-
Define, Measure, Analyze, Improve, Control cluded in or excluded from a project, and how long the
improvement project will take to complete.
Different approaches are available to implement continual The Measure phase involves gathering data about
improvement. The Six Sigma methodology of define, mea- process problems and failures. It is important to understand
sure, analyze, improve, control (DMAIC) takes an identified the current state of the process. This can be achieved
problem and through the use of tools and techniques is able through the use of a process map and by performing a fail-
to arrive at a sustainable solution17 (Fig. 25–8). ure modes and effects analysis (FMEA). Different types of
The Define phase begins with identifying a problem that process maps include the SIPOC diagram (Suppliers, Inputs,
may be tied to a customer complaint or a regulatory issue as Process, Outputs, Customers), a value stream map (VSM),
6888_Ch25_531-551 29/10/18 11:34 AM Page 545

Chapter 25 Quality Management in the Blood Bank 545

Define
Determine product/service
specifications
Develop the scope
Assemble process
improvement team

Control Measure
Create a control plan Create process map of current state
Monitor impact of process Perform failure mode and effects
improvements analysis (FMEA)
Celebrate success Determine how well the process
meets customer expectations
Assess the measurement system to
ensure data are accurate
Improve
Utilize knowledge and
model the ideal process
using either design of Analyze
experiment or an Implement action items identified by
Figure 25–8. DMAIC cycle. A common quality improve- observational study the process improvement team
ment process.

and a flowchart. The important consideration regarding LEAN


process maps is that all the information is captured about
the process. In performing the FMEA, failures in the process Using LEAN in process improvement helps identify non-
activities can be identified and prioritized according to their value-added activities that are considered waste.18,19 LEAN
frequency of occurrence and the level of failure detectability. has defined seven areas of waste:
Numbers are assigned on a scale of severity of the failure
Overproduction—preparing extra reports, taking extra tubes
mode resulting in a risk priority number (RPN), which
of blood during phlebotomy
quantifies the risk for each activity. Through the use of the
Correction (defects)—incorrect results, repeat testing
RPN, the process activities that have the most serious con-
Inventory—supplies ordered in bulk, transactions not
sequences can be prioritized for improvement.
processed
The Analyze phase begins with understanding the cause
Motion—extra steps in data entry, unnecessary movement
and effect relationships in the process. The actions identified
of personnel
at the end of the Measure phase are tested, and the resulting
Overprocessing—communications, reports, e-mails that con-
data are collected and further analyzed using statistical tools.
tain more information than necessary
Maintaining good documentation and gaining greater knowl-
Conveyance—distance travelled, moving paper around
edge about what factors are significant and those that are not
Waiting—delayed work due to lack of communication from
are critical to this phase.
another person or group
The Improve phase can begin with a design of experiment
(DOE) or an observational study. The DOE tool mathemat- Some LEAN projects may require only one person to com-
ically models the ideal process with the actions tested and plete it. When a team is necessary for a larger project, the
identified as significant in the Analyze phase. However, when members should include a project champion, a subject mat-
dealing with the human condition, the DOE may not apply.18 ter expert, and individuals who perform the work. It is also
Depending upon the situation, an observational study may suggested to include one member from outside the area of
be the appropriate action. improvement so this individual can add objectivity to the
The Control phase is the final phase in the DMAIC processes being discussed. The ideal team size is fewer than
methodology. This phase ensures that the implemented eight people.
changes are sustained. Creating an effective control plan ver- One frequently used LEAN activity is referred to as per-
ifies that the improved process is being followed and also forming a 5S:
helps determine whether additional actions are necessary to
achieve the goal. Ownership of the control phase is crucial 1. Sort—eliminate all unnecessary items, keeping only what
to sustaining the improvement gains. Identifying an individ- is important.
ual(s) who will be responsible for data collection and analy- 2. Set in order—arrange things in order of frequency of use
sis and follow-up actions when the improvement results and quickest to locate.
don’t meet the targeted goals is very important. The key to 3. Shine—clean the work area.
sustaining improvement is evaluating data, the frequency of 4. Standardize—arrange work stations the same so all per-
which is defined in the control plan that will demonstrate sons doing the same job can easily find items.
that the process can be repeated and reproduced.15 5. Sustain—maintain and control the improvement.
6888_Ch25_531-551 29/10/18 11:34 AM Page 546

546 PART V Quality Management and Compliance

LEAN is a very user-friendly process-improvement tool. costs.23 In the blood bank, transfusion service, and medical
It engages the work force and encourages staff members to laboratory, prevention costs include validation activities,
identify and participate in improving the workplace. preventive maintenance, work process training, and quality
management activities such as improvement teams and qual-
A Quality Management System for Medical ity system training.
Laboratories Appraisal costs are the costs of implementing processes
and controls to appraise or evaluate the blood bank, trans-
The path of workflow, whether in the blood bank or any of fusion service, or medical laboratory’s performance. There
the clinical laboratories is essentially the same. The QSEs are internal and external appraisal costs. Internal evaluation
supporting the path of workflow are universal. costs include those for calibration materials and reagents,
A review of the International Organization for Standard- equipment calibration, quality control materials and
ization quality standards demonstrates that blood bank and reagents, any interim inspections of blood products or
laboratory QSEs are included in the international stan- records, and the internal audit program. External evaluation
dards.20,21 Therefore, the discussion in the section on QSEs costs include those for proficiency testing and periodic li-
applies equally to hospital laboratories. It is not only possible censure or accreditation inspections. The budget contains
but also highly desirable to expand the blood bank’s QMS funds for these activities because they are mandated by reg-
building efforts so that the entire laboratory benefits from ulatory and accreditation agencies. Appraisal activities help
improved organization, coordination, and effectiveness of its ensure that problems are caught and corrected so as to min-
many processes.22 (See Fig. 25–9 for a review of the QSEs imize any negative impact on customers and patients.
supporting the medical laboratory path of workflow.) Nonconformance costs, which include internal and ex-
ternal failure costs, are considered the costs of poor quality
The Cost of Quality (COPQ). These nonconformance costs are associated with
failure to meet customer requirements and regulatory and
Although it may sound like a great amount of effort and accreditation requirements.15 Internal failures are those
expense to implement a QMS in a blood bank, transfusion caught and corrected before they adversely affect customers
service, or medical laboratory, the costs involved are signifi- or patients. Examples of internal failures include discarded
cantly lower than the expenses the facility experiences when donated blood units, samples unacceptable for compatibil-
there are major and minor nonconformances that require cor- ity testing that need recollection, retesting when controls
rection23. Expenses are multiplied when the same noncon- do not give acceptable results, instrument failures, and
formance recurs and needs to be resolved again. A basic downtime.
understanding of the two types of quality costs is vital to com- External failure costs are expensive and hard to measure
prehending the value of quality management to the customers because they include both actual and intangible costs in-
and patients served by the facility. curred to correct a nonconformance that has reached or ad-
versely affected the customer. Examples of external failure
Conformance Costs costs include customer complaint resolution, misdiagnoses,
recalls, and lawsuits. The cost to correct these and other
The cost of conformance, also known as the cost of good errors that are preventable, and serious in their conse-
quality (COGQ), is described as prevention and appraisal quences for patients, greatly erodes the operating budget

A Quality Management System for the Laboratory


Laboratory Path of Workflow
Preanalytic Analytic Postanalytic

Test Order, Sample Collection, Transport, Receipt and Process, Testing, Reporting, Sample Archiving

Quality System Essentials: The Building Blocks

The Blood Bank The Work The Performance

Organization Process Management Nonconformance Mgmt


Customer Focus Documents & Records Assessments
Facilities & Safety Information Management Continual Improvement
Personnel
Purchasing & Inventory
Equipment

Figure 25–9. A QMS for the medical laboratory.


6888_Ch25_531-551 29/10/18 11:34 AM Page 547

Chapter 25 Quality Management in the Blood Bank 547

and customer confidence in the facility’s quality, safety, and that health-care providers such as hospitals and blood centers
credibility. are involved in organization-wide quality improvement pro-
grams that ensure the safety of donors and patients. Only
Importance of a Facility Quality those organizations demonstrating measurable quality im-
Management System provements are approved for agreements for products and
services. Consumers of blood center, hospital, and transfu-
In order to survive the changes facing the nation’s health-care sion services accept no less than total quality. Organizations
industry, consumers of health-care services want evidence that provide less will not survive.

CASE STUDIES
Case 25-1

Example of an FMEA
The hospital administration of an academic medical center, while working with nursing and the transfusion service, iden-
tified that specimen collection was a critical success factor in blood administration and patient safety. The team, which
included members from the hospital inpatient units and the transfusion service, met weekly over a period of 4 months
to analyze the blood administration process from the initial order to the completion of transfusion. The team decided to
prepare a failure modes and effects analysis (FMEA) and collected background information using flowcharts of the process
to determine the failure modes and assign risk priority numbers (RPN). The highest RPNs were used to finalize recom-
mendations for risk reduction.
The team charted the basic blood administration processes:
1. Specimen collection
2. Specimen transport
3. Specimen receipt and testing
4. Blood product preparation and labeling
5. Blood product issue and transport
6. Transfusion of blood product
Each identified process was further divided into individual activities. Each activity was described, and the following
ratings were determined:
• Occurrence. How often does the failure occur?
• Severity. How severe is the effect of the failure?
• Detectability. What is the likelihood of detecting the failure?
For each process activity, the occurrence, severity, and detectability were ranked and graded using the following criteria:
Occurrence Rating
Rank Description

1 Remote probability (never happens)


2–5 Low probability (happens once/year)
6–7 Moderate probability (happens once/month)
8–9 High probability (happens daily)
9–10 Very high probability (happens multiple times a day)

Severity Rating
Rank Description

1 Minor (no real effect on service, customer doesn’t notice)


2–3 Low (causes a slight customer annoyance)
4–6 Moderate (causes some customer dissatisfaction, causes unscheduled repairs or damage)
7–8 High (high degree of customer dissatisfaction, disruption to subsequent process or services)
9–10 Very high (affects patient/visitor/ staff safety and involves noncompliance with government regulations)
6888_Ch25_531-551 29/10/18 11:34 AM Page 548

548 PART V Quality Management and Compliance

Detectability Rating
Rank Description

1 Very high (Controls will almost certainly detect the failure; the defects are obvious.)
2–5 High (Controls have a good chance of detecting the failure.)
6–8 Moderate (Controls may detect the failure.)
9 Low (Controls will most likely not detect the failure.)
10 Very low (Controls will almost certainly not detect the failure; defect is latent, and item is not
checked or able to be checked.)
For a potential failure mode, the risk priority number reflects how frequently the event occurs, how serious the effects
are, and what controls are in place to prevent or notice it before it happens.

For the specimen collection process, the team identified 13 steps and the risk priority number for each step.
Step Number Step Description Total *RPN

1 Nurse verifies order. 177


2 Patient care technician notified 46
to collect specimen.
3 Patient care technician obtains 19
laboratory label for type and screen.
4 Patient care technician obtains blood 53
bank transfusion requisition and red band.
5 Patient care technician obtains 55
phlebotomy supplies.
6 Patient care technician goes to 250
patient room.
7 Patient care technician identifies patient 400
by name and medical record number.
8 Patient care technician collects blood sample. 248
9 Patient care technician assembles red arm band, 295
labels tube, and transfusion requisition in the
presence of the patient.
10 Patient care technician puts the red band on patient. 153
11 Patient care technician signs transfusion requisition. 15
12 Patient care technician returns transfusion 500
requisition to nurse.
13 Nurse completes transfusion requisition. 9

*RPN = Occurrence ranking × Severity ranking × Detectability ranking

FMEA Action Items


Number Process Step Recommendation

1 Patient care technician identifies patient (step 7) Nurse checks blood at the patient bed-
side per Patient Care Policy. Imple-
ment an electronic bar coding system
for specimen collection.
2 Patient care technician returns transfusion requisition Increase auditing of transfusions on
to nurse (step 12) nursing units as deterrent to the pre-
transfusion “desk check.”

1. Based on the results of the RPN, which steps pose the highest risk to the patient?
2. How could step 12, “Patient care technician returns transfusion requisition to nurse,” result in such a high RPN?
3. Compare and contrast root cause analysis (RCA) with a failure modes and effects analysis (FMEA).
6888_Ch25_531-551 29/10/18 11:34 AM Page 549

Chapter 25 Quality Management in the Blood Bank 549

Case 25-2

LEAN
During a mock accreditation inspection, a tracer audit was performed for an urgent blood transfusion. The audit revealed
a delay between the time the red blood cells were ordered and the time the red blood cells were available for transfusion.
To determine whether this event was isolated, an internal audit was performed in the transfusion service. This audit
demonstrated that turnaround time (TAT) for stat red blood cell orders was inconsistent with the required TAT stated in
the laboratory policy. The acceptable stat TAT for red blood cells was stated as within 60 minutes (excluding complicated
samples requiring additional antibody investigation). Data analysis revealed only 45% of the stat crossmatches met the
60-minute requirement.
The Laboratory Quality Team was chartered and included a laboratory administrator as the project champion; technol-
ogists from the transfusion service first, second, and third shifts; and a technologist from another area in the clinical lab-
oratory to provide an outside perspective. The team’s goal was to use LEAN and Six Sigma principles to guide the TAT
improvement project. The LEAN tools19 used would focus on eliminating waste, which includes waiting, defects and re-
works, moving materials or people, and adding additional steps to get the process right. The Six Sigma tools would reduce
any variation in the crossmatch process.
A flowchart outlining the process activities to crossmatch red blood cells demonstrated there was process variation be-
tween personnel and the different shifts. A spaghetti diagram was created next, which visually represents how much walk-
ing is required to perform a crossmatch and identifies redundancies in the work flow. The diagram visually illustrates
where opportunities exist to expedite the process.
The LEAN ideas to improve the TAT in the blood bank included:
• Flowchart the work process.
• Standardize the work flow through all shifts.
• Create a spaghetti diagram to show unnecessary movement (walking) and areas of congestion.
• Determine a process family to create areas to localize specific job duties (to reduce work space congestion, reduce walking).
• Use a point of use storage (POUS) to store materials where they will be used.
• Use a pull system so that additional red blood cells may be irradiated and available on demand.
• Provide visual cues to the technologists to signal when to reorder supplies or irradiate units.
Changes that were needed included defining the new standardized work process and writing the procedures. New work
assignments and improved handoff were developed, which decreased wait time and standardized the process. Relocating
the specimen centrifuge closer to the pneumatic tube system and the label printer closer to the blood refrigerator decreased
walking and motion. Replacing equipment to facilitate efficiency (e.g., a high-speed centrifuge and new automated tech-
nology) were supported by the laboratory administrator (the project champion). Finally, visual cues were identified and
implemented for inventory replenishment.
The average TAT improved from 124 minutes to 52 minutes, and the standard deviation (variation) was reduced from
44 minutes to 14 minutes. It was not a single change, but many changes over a period of time that resulted in improving
this process.
1. How do you define value for your transfusion service or blood bank?
2. Who are your customers (list both internal and external)?
3. Does your laboratory use visual cues? List examples.
6888_Ch25_531-551 29/10/18 11:34 AM Page 550

550 PART V Quality Management and Compliance

SUMMARY CHART
 Blood bank compliance with federal regulations and able to meet process goals without compromising the
accreditation requirements is mandated by the FDA, process itself.
the CMS (as CLIA), the Joint Commission, the AABB,  cGMP requires that facilities design their processes and
and the CAP. procedures to ensure that blood components are man-
 Compliance inspections measure the state of the facil- ufactured consistently to meet the quality standards
ity’s processes with respect to the applicable require- appropriate for their intended use.
ments at a single point in time and are usually  Process validation challenges all activities in a new
conducted every 2 years. process before implementation to provide a high degree
 Quality control procedures in blood banking include of assurance that the process will work as intended.
daily testing of the reactivity of blood typing reagents  Routine QC procedures, review of records, and capture
and positive and negative controls in infectious disease of nonconformances when the process did not perform
testing. Equipment function checks include calibration as expected are routine process control measures that
of serologic centrifuges, timer checks, and temperature monitor whether a process is functioning as needed.
monitoring of refrigerators, freezers, and thawing  Nonconformance management is a name for processes
devices. that detect, report, evaluate, and correct events in
 IQCP is an individualized quality control plan that blood bank operations that do not meet the facility’s
meets CLIA’s QC requirements and is only applicable or other requirements.
to nonwaived testing.  An internal audit reviews a specific facility process and
 Quality assurance is a set of planned actions to provide determines—by examining documents and records, in-
confidence that processes and activities that influence terviews, and observations—whether the facility is
the quality of the product or service are working as ex- meeting applicable requirements and its own policies,
pected, individually and collectively. processes, and procedures.
 A quality management system provides a framework  Quality system essentials provide a systematic ap-
for uniformly applying quality principles and practices proach to creating and sustaining a quality manage-
across all blood bank operations, starting with donor ment system in the blood bank, transfusion service,
selection and proceeding through transfusion out- and the entire clinical laboratory.
comes.  LEAN is a process improvement methodology that
 Process control is a set of activities that ensures a given helps to identify non-value-added activities that are
process will keep operating in a state that is continuously considered waste.

Review Questions 4. Internal and external failure costs are:


a. Readily identifiable in facility reports
1. A compliance program: b. Controlled through prevention and appraisal
c. Built into the facility’s operating budget
a. Evaluates how effectively the facility meets regulatory
d. Part of prevention and appraisal
requirements
b. Always identifies quality problems 5. Which one statement below is correct?
c. Is part of quality control a. A process describes how to perform a task
d. Is an evaluation of efficiency b. A procedure simply states what the facility will do
2. The quality system essentials are applied to: c. A procedure informs the reader how to perform a task
d. A policy can be flowcharted
a. The blood bank’s management staff
b. Blood bank quality control activities 6. A blank form is a:
c. Blood component manufacturing a. Record
d. The blood bank’s path of workflow b. Procedure
3. cGMP refers to: c. Flowchart
d. Document
a. Regulations pertaining to laboratory safety
b. Validation of testing
c. Nonconformance reporting
d. Manufacturing blood components
6888_Ch25_531-551 29/10/18 11:34 AM Page 551

Chapter 25 Quality Management in the Blood Bank 551

7. An example of a remedial action is: 8. Department of Health and Human Services. Code of Federal
Regulations, Title 45, Parts 160 and 164. Washington (DC):
a. Applying the problem-solving process U.S. Government Printing Office; revised annually.
b. Starting a process improvement team 9. International Organization for Standardization. ISO 9000:2015
c. Resolving the immediate problem Quality management systems—fundamentals and vocabulary.
d. Performing an internal audit Geneva: International Organization for Standardization; 2015.
10. Center for Medicare and Medicaid Services [Internet]. Wash-
8. The DMAIC methodology is used for: ington (DC): CMS [cited: September 1, 2017]. CLIA brochure
a. Problem resolution 11: CLIA individualized quality control plan introduction.
Available from: https://www.cms.gov/Regulations-and-Guidance/
b. Process control Legislation/CLIA/Downloads/CLIAbrochure11.pdf
c. Validation 11. Center for Medicare and Medicaid Services [Internet]. Washing-
d. Auditing ton (DC): CMS [cited: September 1, 2017]. CLIA brochure 12:
CLIA individualized quality control plan considerations when de-
9. ___________________________ is a set of planned ciding to develop an IQCP. Available from: https://www.cms.gov/
actions that ensure that systems and elements that in- Regulations-and-Guidance/Legislation/CLIA/Downloads/
fluence the quality of service are working as expected. CLIAbrochure12.pdf
12. Center for Medicare and Medicaid Services [Internet]. Washington
a. Quality control
(DC): CMS [cited: September 1, 2017]. CLIA brochure 3: Individ-
b. Quality assurance ualized quality control plan. What is an IQCP? Available from:
c. Quality indicator https://www.cms.gov/Regulations-and-Guidance/Legislation/
d. Quality management system CLIA/Downloads/CLIAbrochure13.pdf
13. Centers for Medicare and Medicaid Services [Internet]. Wash-
10. The Centers for Medicare and Medicaid Services (CMS) ington (DC): CMS [cited: September 1, 2017]. Developing an
developed an alternative quality control option, an IQCP: a step by step guide. Available from: https://www.cms.gov/
individualized quality control plan (IQCP). How is Regulations-and-Guidance/Legislation/CLIA/Downloads/IQCP-
Workbook.pdf
the minimum frequency of running quality controls 14. Fung, MK, Eder AF, Spitalnik SL, Westhoff, CM, editors.
determined? Technical manual, 19th ed. Bethesda (MD): AABB; 2017.
a. Through risk assessment 15. Centers for Medicare and Medicaid Services. Code of Federal
b. By the quality control plan Regulations, Title 42, Part 493.1 to end. Washington (DC): U.S.
Government Printing Office; revised annually.
c. After quality assessment
16. Centers for Medicare and Medicaid Services. Code of Federal
d. By the manufacturer Regulations, Title 42, Parts 430 to end. Washington (DC): U.S.
Government Printing Office; revised annually.
References 17. Shankar R. Process improvement using six sigma: a DMAIC
guide. Milwaukee (WI): American Society for Quality, Quality
1. Food and Drug Administration, Center for Biologics Evaluation Press; 2009.
and Research. Guideline on general principles of process vali- 18. Walters LM, editor. S3: simple six sigma for blood banking,
dation. Rockville, (MD): Food and Drug Administration; 2011. transfusion, and cellular therapy. Bethesda (MD): AABB Press;
2. Food and Drug Administration, Department of Health and 2007.
Human Services. Code of Federal Regulations, Title 21, Parts 19. Dennis P. Lean production simplified. 3rd ed. Plain language
200–299. Washington (DC): U.S. Government Printing Office; guide to the world’s most powerful production system. New
revised annually. York: Productivity Press; 2015.
3. Food and Drug Administration, Department of Health and 20. International Organization for Standardization. ISO 9001:2015
Human Services. Code of Federal Regulations, Title 21, Parts Quality management systems—requirements. Geneva: Inter-
600–799. Washington (DC): U.S. Government Printing Office; national Organization for Standardization; 2015.
revised annually. 21. International Organization for Standardization. ISO 15189:2012
4. The Joint Commission. Accreditation survey activity guide for Medical laboratories—particular requirements for quality and
health care organizations. Oakbrook Terrace (IL): Joint Com- competence. Geneva: International Organization for Standard-
mission Resources; 2017. ization; 2012.
5. The Joint Commission. Comprehensive accreditation manual 22. Clinical and Laboratory Standards Institute. A quality manage-
for laboratories and point-of-care testing. Oakbrook Terrace (IL): ment system model for laboratory services; approved guideline,
Joint Commission Resources; 2017. QMS01. Wayne (PA): Clinical and Laboratory Standards Insti-
6. College of American Pathologists. Inspection checklists for lab- tute; 2018.
oratory accreditation. Northfield (IL); College of American 23. Wood DC, editor. Principles of quality costs: Financial mea-
Pathologists; 2018. sures for strategic implementation of quality management.
7. Standards for blood banks and transfusion services. 31st ed. 4th ed. Milwaukee (WI): American Society for Quality Press;
Bethesda, (MD): AABB; 2018. 2013.
6888_Ch26_552-573 29/10/18 5:08 PM Page 552

Chapter 26
Patient Blood Management
Julie L. Cruz, MD, FACP and Steven F. Gregurek, MD

Introduction Concurrent Review Minimize Iatrogenic Blood Loss


Key Players in a Patient Blood Prospective Review Predictive Modeling
Management Program Feedback Education
The Transfusion Safety Officer: A Key Intervention Strategies Continuous Improvement
Resource Other Opportunities for Review Metrics
The Transfusion Committee Patient-Focused Care Ongoing Monitoring and Improvement
Transfusion Guidelines Correction of Anemia Case Studies
Blood Order Sets Autologous Blood/Red Blood Cell Case Study 26-1
Decision Support and Computerized Order Recovery Techniques Case Study 26-2
Entry Optimize Hemostasis Summary Chart
Utilization Auditing and Review Viscoelastic Blood Testing Review Questions
Review Criteria Continuous Noninvasive Hemoglobin References
Retrospective Review Monitoring

OBJECTIVES
1. Describe the purpose and goals of a patient blood management program (PBM).
2. List and describe PBM tools, and identify the goals they address when appropriately utilized.
3. Describe the role of the transfusion safety officer (TSO), and identify key functions the TSO performs in PBM.
4. Define transfusion guidelines. Describe how they are created and explain their importance in PBM. Demonstrate the potential
utility of leveraging computerized physician order entry (CPOE) and decision support algorithms in PBM.
5. Discuss strategies for the process of blood utilization auditing and review.
6. Identify the differences among the prospective, concurrent, and retrospective blood utilization reviews.
7. List intervention strategies and describe how they may be employed with the various blood utilization review models.
8. Discuss patient-focused care strategies in the context of PBM and the three pillars of patient blood management.

Introduction committee. Providers such as physicians, nurses, perfusion-


ists, and technologists in areas such as surgery, anesthesia,
The terms patient blood management (PBM) and blood and hematology, as well as risk and quality personnel, par-
utilization management (BUM) are often used interchange- ticipate in these programs. Because of the potential for con-
ably, though the scope and details of their definitions vary siderable cost savings, these programs often obtain high
across the literature. Both terms encompass the concept of visibility with administrative and executive professionals.
active health-care provider programs intended to limit and Because of this, it is important to carefully define the expec-
prevent inappropriate or unnecessary transfusion, while tations and limitations. In short, PBM/BUM is a multidisci-
promoting strategies to reduce the overall transfusion re- plinary systems-based process that encompasses all
quirements of patients. These programs aim to improve disciplines and activities related to blood transfusion to
transfusion safety, usually while decreasing blood costs. achieve the following goals: (1) enhanced safety, (2) quality
They are not limited to the purview of the blood bank improvement, (3) patient-focused care, (4) resource conser-
and its medical director, nor even the facility’s transfusion vation, and (5) waste reduction/cost control.1

552
6888_Ch26_552-573 29/10/18 5:08 PM Page 553

Chapter 26 Patient Blood Management 553

PBM as a descriptor also arose in the literature and con- administered by skilled personnel can result in patient harm.
tinues to be used in a more narrow sense, specifically in the For example, despite intense mitigation strategies that have
context of management of surgical patients through the elec- reduced the risk of transfusion-related acute lung injury
tive preoperative period through discharge. The focus is to (TRALI), residual risk remains.2 In developing a program, it
avoid, or at least minimize, the patient’s need for transfusion is necessary to build a quality model that can qualify this risk.
through anticipatory management and employment of spe- In the model presented in this chapter, this risk can be
cial techniques. These strategies have been expanded beyond described as “peril.”
elective surgeries and will be further discussed later. Traditional methods to mitigate peril focus on interven-
Many tools are available from the above-mentioned disci- tions designed to increase the safety of the individual blood
plines to structure a successful PBM program. Those, which component, such as donor deferral questions, infectious dis-
we will discuss further, are listed in Table 26–1. Although the ease testing, the crossmatch, and the second label check.
tools may be primarily paired with a particular goal, they Quality assurance processes ensure that safe, pure, and po-
overlap, and when properly utilized, deliver benefits across tent components are manufactured in the blood center and
several, if not all, of the elucidated goals. It must be remem- released for transfusion by the blood bank. A safe component
bered that the goal of a successful PBM/BUM program is to is one that minimizes harm. A pure component is one with-
improve patient safety through the evidence-based optimiza- out contamination from chemicals or infectious agents.
tion of blood transfusion. Every hospital is unique in its med- Finally, potency refers to the capability of the transfusion to
ical culture, localized standard of care, and treatment focus. produce the desired outcome for an approved indication.
Therefore, each hospital will require a unique plan and set of Blood is a drug, and manufactured components must be col-
tools to obtain desired utilization goals. The concepts pre- lected, processed, and stored in such a way that the “unit”
sented in this chapter provide a framework for designing and contains the expected amount of the desired biologic capable
optimizing a PBM/BUM program. Only after meeting with of delivering the desired bioreactivity. In other words, a po-
key hospital personnel can a course of action be determined. tent blood component works. The transfusion service
In some cases, only one or a few tools will be required. For mantra is “the right blood, to the right patient, at the right
example, a well-designed and comprehensive anemia clinic time.”3 The “right patient” in this context refers to identity
for elective surgery patients may be the best use of resources. (e.g., John A. Smith in bed A vs. John J. Smith in bed B). In
In other cases, a more comprehensive strategy may enhance contrast, the utilization program ensures the patient actually
results. The bottom line is that an effective program requires requires each transfusion. Figure 26–1 demonstrates the par-
careful planning and should not be just a haphazard checklist adigm shift, illustrating that greater risk reduction can be
of every known blood utilization technique. Enhanced safety achieved by considering both individual component safety
is a primary goal of PBM. All transfusions pose an inherent measures and utilization optimization strategies.
baseline risk of adverse events. Even optimally selected, fully To receive benefits from transfusion, the patient must have
tested, appropriately stored and matched components pro- a deficiency or pathological lesion that can be remedied by
duced under good manufacturing practices and properly functioning stored components. This process can be perfected

Table 26–1 PBM Tools and the Goals They Address


Tool PBM Goal(s) Addressed
Transfusion safety officer Safety, quality improvement, patient-focused care, resource conservation, waste
reduction/cost control

Transfusion guidelines Safety, quality improvement, patient-focused care, resource conservation, waste
reduction/cost control

Maximum surgical blood order schedule (MSBOS) Safety, quality improvement, waste reduction/cost control

Blood order sets Safety, quality improvement

Decision support algorithms/computerized physician Safety, quality improvement, patient-focused care, resource conservation, waste
order entry reduction/cost control

Utilization auditing and review Safety, quality improvement

Utilization metrics and benchmarking continuous monitoring Safety, quality improvement, resource conservation, waste reduction/cost control
and feedback

Preoperative anemia clinics Patient-focused care, resource conservation, safety

Surgical/anesthesia blood-sparing or salvage techniques Patient-focused care, resource conservation

Education Safety, quality improvement, patient-focused care, resource conservation, waste


reduction/cost control
6888_Ch26_552-573 29/10/18 5:08 PM Page 554

554 PART V Quality Management and Compliance

Strategies focused on improving patient safety by promot-


Risk Benefit ing good utilization practice can also promote the reduction
or elimination of waste. Considering waste in the context of
LEAN is a useful concept, although any systems-based eval-
ent uation strategy may be employed. LEAN is a systematic ap-
Compon tion
k m it ig a proach to identifying actions within a process that create
ris
Risk Benefit value, then optimizing the alignment of these actions to de-
liver to customers exactly what is desired, in the quantity de-
sired, precisely when it is required. It is based upon an
nt
approach first developed by the Toyota Corporation follow-
Patie n ing World War II to better manage operations, including cus-
le c t io
se
tomer relations, supply chain management, development,
nt
pone and production. Central to accomplishing this goal is the
Com igation
m it
risk elimination of waste (also described by the Japanese term
Risk Benefit
muda). Waste is any action that does not create value as
Figure 26–1. Traditional transfusion service risk mitigation strategies are focused defined by the customer. DOWNTIME is a useful acronym
on increasing the safety of the individual component, yet inherent residual risk re- utilized in LEAN for identifying types of waste: defects,
mains. Patient blood management confers additional safety by incorporating con- overproduction, waiting, non-value-added processing, trans-
sideration of patient-based factors to ensure transfusion is appropriate and optimally portation, inventory excess, motion, employee/people waste.5
selected, hence a greater safety benefit is achieved.
Table 26–2 provides transfusion medicine examples of waste
identified when applying this concept.
by ensuring optimal timing and administering the optimal Although some transfusion services may consider the or-
dose for correction without exposing the patient to increased dering clinician to be the customer, the patient is the true
transfusion risk. For example, several coagulation factors in customer, and value is ultimately determined by the thera-
plasma have short half-lives once transfused. If transfused to peutic benefit (or harm) the patient receives from blood
correct a coagulopathy too soon before a planned surgery or component transfusion. The system strives for “just in time”
invasive procedure, the effectiveness in correcting the coag- delivery as determined by customer need (also known as
ulopathy could be diminished. In the worst case, such as “pull”). Laboratorians are most familiar with this concept as
transfusion of the plasma the night before the planned pro- “turnaround time.” Improper BUM practices decrease value
cedure, the transfusion provides no benefit, but exposes the and drive the result toward peril and waste. Thus, there are
patient to a risk of adverse events such as fluid overload, not only human losses in terms of the patient’s health and
allergic reaction, or TRALI. This is an example of inappropri- well-being but also financial, labor, and resource losses. For
ate timing. In this case, “the right patient” refers to his or her example, the transfusion of a blood component to a healthy
suitability to receive the requested transfusion, taking into person creates no value but increases peril and waste. The
account both individual clinical factors and established Cruz-Gregurek model, shown in Figure 26–2, demonstrates
guidelines. In this context, PBM is the complement to the this concept. Optimal value is the penultimate goal and is
crossmatch. The crossmatch assesses the safety of a particular accomplished by developing standardized methods targeted
component for the patient, while utilization management to reduce waste and peril. As noted in the figure, some inap-
ensures that the patient is appropriate for the transfusion. propriate practices create both waste and peril. These prac-
Although cost savings are often emphasized, the safety of tices represent high-yield opportunities for intervention. In
blood transfusion is the single most important aspect of addition to safety and financial benefits, such interventions
effective BUM. According to Szczepiorkowski,4 “the decision are expected to reduce variability in practice and provide bet-
to transfuse or not to transfuse is one of the more complex ter stewardship through optimization of a limited resource.
decisions made by medical practitioners.” Despite the layers Those interested in applying LEAN concepts are referred to
of safety designed to reduce risk associated with blood com- the literature, and a number of professional organizations
ponents, inherent risk remains; thus, zero risk is equivalent and consultancies offer resources.6
to no transfusion. Even as a relatively new field, PBM and blood utilization
Appropriate BUM also recognizes the risk of undertrans- are in the 10 most important technological advances and in-
fusion (i.e., failure to transfuse when benefits outweigh risk) novations in the last 50 years of blood banking.7 PBM works
and inappropriate or suboptimal component selection. From because it improves patient safety, increases quality, and de-
this perspective, the safest transfusion is the one that is given creases blood costs. In fact, the number of blood transfusions
appropriately. Although many transfusion risks are prevent- in the United States decreased nearly 5% between 2011 and
able, rare errors account for a significant number of fatal 2013.8 When extended across multiple hospitals or hospital
transfusion-related events. Fortunately, rigorous attention to systems, PBM programs have been shown to lead to consid-
procedure and good manufacturing practice has decreased erable improvements in patient safety and cost reduction
the incidence of these untoward events. However, it is im- with significant decrease in length of stay and mortality. Im-
portant to recognize that despite our best efforts, total elim- provements have also been demonstrated with decreased
ination of all transfusion risk (peril) is impossible. emergency room readmissions, decreased rates of infection,
6888_Ch26_552-573 29/10/18 5:08 PM Page 555

Chapter 26 Patient Blood Management 555

Table 26–2 Types of Waste in Transfusion Medicine


Action: Blood Component Is: Waste and Potential Drivers
Ordered inappropriately and transfused: • Available units diverted from patients with greater need
• Wasted inventory/misallocated resource
• Suboptimal component selected • Overproduction
• Transfusion not indicated • Non-value-added processing—treatment of transfusion reactions
• Improper timing
Drivers: Facility culture, lack of provider knowledge, prior physician training,
provider practicing “defensive medicine”

Appropriate transfusion ordered with inappropriate “stat” • Employee waste: staff diverted to perform unnecessary tasks
order prepared and transfused • Non-value-added processing
• Wasted motion (stat activities)
• Transport waste (stat transport)
• Increases waiting for other orders
Drivers: Not trusting timely delivery if ordered as routine, poor communication
between clinical service and laboratory

Ordered, prepared, and not transfused: • Employee waste: staff diverted to perform unnecessary tasks
• Wasted inventory
• “Type and Crossmatch” ordered when “Type and Screen” • Transportation
appropriate • Overproduction
• Thawed FFP expires unused
• Unused washed units allowed to expire on floor Drivers: Not trusting timely delivery if ordered as needed, lack of provider
• Unused issued components improperly returned knowledge, facility culture, prior physician training, provider practicing “defen-
• Failure to discontinue standing order for HLA-matched sive medicine,” lack of nursing staff knowledge, poor communication between
platelets clinical staff and laboratory

Unnecessary component modification • Non-value-added processing


• Employee waste: staff diverted to perform unnecessary tasks
• Irradiation, washing
Drivers: Facility culture, lack of provider knowledge, prior physician training,
provider practicing “defensive medicine,” lack of robust process to identify
appropriate patients

VALUE

• Overtransfusion • Overtransfusion
• Suboptimal component • Undertransfusion
• Expired after issue • Suboptimal component
• Units prepared/not used • Improper timing

Figure 26–2. The Cruz-Gregurek model of optimization. Inappro-


priate utilization practices result in both waste and peril. Measures to
improve utilization drive transfusion practice toward optimal value,
ultimately defined as the patient’s clinical response to safe and effective
transfusion. Interventions addressing some inappropriate practices will
reduce both waste and peril and are therefore high yield. WASTE PERIL

and decreased rates of strokes. From a blood utilization of Blood Management (SABM), founded in 2001.10 These or-
standpoint, aggressive PBM can decrease transfusions across ganizations and others provide education and resources, as
all product lines by up to 40%.9 well as high visibility to hospital leadership and others regard-
PBM has become a focus point and mainstay of initiatives ing the potential benefits of a well-run PBM program. In ad-
and curricula for professional organizations whose interests dition, the blood supplier often has contacts or resources to
relate to blood and blood transfusion, such as the AABB and blood utilization literature and expertise, and professional
a relatively new organization, the Society for the Advancement consultancies are also available.
6888_Ch26_552-573 29/10/18 5:08 PM Page 556

556 PART V Quality Management and Compliance

Key Players in a Patient Blood training, validation, and maintenance of records. As a part
Management Program of the blood bank, procedures and records are often the focus
of scrutiny and inspection. The utilization program should
A well-designed PBM program involves the coordination of be conducted with the same level of rigor as other functions
a number of individuals with critical expertise to design, im- within the blood bank. Each facility will have staffing needs
plement, maintain, and perform continuous improvement determined and based on its particular goals. For most pro-
activities. Current publications and local and national organ- grams, the inclusion of a team member from information
izations provide resources for implementation of blood man- technology will be important, at least in the design stages.
agement and utilization programs and can be very useful for Data mining and analysis will be critical in assessing the cur-
planning teams unfamiliar with this process.11,12 Various in- rent state and designing reports for continuous monitoring.
dividuals across disciplines may fill the roles of planners, ap- A great deal of information can be gleaned and processed by
provers, implementers, and reviewers. Table 26–3 illustrates well-constructed queries to the Laboratory Information Sys-
this concept, elucidating roles and examples of individuals tem (LIS), electronic medical record, billing system, and
or groups who may fulfill them. An individual may fill more other databases. Often these systems do not readily interface;
than one role, and as the program evolves and needs change, in-house design of well-tailored validated programs may pro-
the designees serving various functions may change as well. vide the best solution. However, before any such design is
The transfusion service medical director is responsible for undertaken, there must be agreement and clear communi-
maintaining guidelines and approving blood bank proce- cation of the precise data and metrics that will be desired.
dures. The medical director is also ultimately responsible for Redesign and rework of such programs is resource intensive.
the safety and efficacy of transfused blood components. This Additional waste is generated if the ultimate result does not
individual has the authority to appoint appropriate surro- serve the purpose for which it was intended. It must be
gates to these duties, including screening procedures, deny- remembered to focus on the desired outcomes.
ing product in prospective programs, and providing feedback
regarding inappropriate transfusion in retrospective audits. The Transfusion Safety Officer: A Key Resource
Blood utilization champions are the physicians, practition-
ers, and nurses who order and administer blood compo- Despite efforts to ensure patient safety in the blood bank,
nents. They are useful in bridging the gap between the blood there is substantial literature that indicates that the majority
bank and clinical services and can be recruited through of medical errors, particularly those in ABO incompatible
newsletters, e-mails, or directly at hospital lectures and pre- transfusion, occur outside the blood bank. The clinical ser-
sentations. Dedicated blood management personnel are vice, rather than the blood bank, was implicated 89% of the
essential to the success of the utilization program, and the time in a study of 15,134 errors during transfusion with high
duties and assignments should be described in detail in a risk of causing substantial patient harm over a five-year
standard operating procedure. This includes the proper period. The great majority of these errors was attributed to
phlebotomy or nursing.13 To address this issue and bridge
the laboratory and clinical realms, the transfusion safety
Table 26–3 Key Players in a Utilization officer (TSO), a unique career role, was first developed in
Management Program European and Canadian blood banks. Whether from a clinical
Role Examples (usually nursing or physician’s assistant) or laboratory
(medical technologist) background, the TSO must develop
Planners • Medical director of blood bank
• Transfusion committee
knowledge in all aspects of transfusion but be particularly ca-
• Supervisor of blood bank pable in interactions with the various patient health-care
• Transfusion safety officer teams.14 Essentially, the TSO coordinates and/or participates
• Delegated clinical leadership in most functions within the PBM program. Examples in-
• Quality assurance clude education (development, coordination, and delivery);
• Information technology
transfusion record audit and blood utilization review; metric
Approvers • Executive staff for physicians tracking and reporting; and development of guidelines, poli-
Policy approval, resource • Physician groups with high cies, and procedures. The individual must also be well versed
provision, program support, blood utilization
• Nursing leadership
in transfusion reactions and safe blood administration. TSOs
and awareness
• Hospital administration liaise with nursing staff to ensure appropriate pretransfusion,
intratransfusion, and post-transfusion procedure perform-
Implementers • Coordinator (transfusion safety
officer)
ance and care, as well as with clinicians to help drive behavior
Day-to-day operations toward evidence-based practice. This role requires excellent
• Evaluators (auditors, controller,
screeners) communication skills, clinical knowledge, and ability to
• Educators (consultant, lecturer) manipulate database software. This allows the TSO to per-
• Physician ambassadors form audits, educate physicians and nurses, and assist with
• Nursing ambassadors
• Suppliers (blood bank staff fol-
the development of other transfusion safety programs.14 The
lowing utilization procedure) TSO should also be capable of preparing and presenting ed-
ucational materials or reports to varied audiences, including
6888_Ch26_552-573 29/10/18 5:08 PM Page 557

Chapter 26 Patient Blood Management 557

bedside nurses and other line staff, the facility’s transfusion Transfusion Guidelines
committee, physicians, laboratorians, and the hospital’s med-
ical executive committee. Most importantly, the transfusion A concise, evidence-based set of transfusion guidelines that
safety officer must be comfortable in the role of physician ed- are known, understood, adopted, and embraced system-wide
ucation, especially in relation to the audit process. In parti- are foundational to blood management. Transfusion guide-
cular, because of the role in physician education, the lines are systematically developed statements relevant to
transfusion safety officer must be skilled in interpersonal transfusion therapy intended to assist clinicians and their
communication, and be comfortable in interacting with the patients in making decisions related to transfusion. Transfu-
wide variety of personalities that can occur in a sometimes sion guidelines are just that—guidelines. They are not in-
stressful and demanding hospital environment. tended to be prescriptive, nor prohibitive when hemotherapy
The role of the transfusion safety officer cannot be is indicated. Hemotherapy is treatment with blood transfu-
overemphasized, and this is well supported by the literature. sion. They are optimally founded on appropriately powered,
In fact, the transfusion safety officer has effectively been de- high-quality, randomized clinical trials, and many are vetted
ployed in all aspects of PBM, patient safety, and blood bank and summarized in guideline statements by professional
quality. In addition, the transfusion safety officer can coor- organizations such as the AABB, the British Committee for
dinate the transfusion committee and provide expertise for Standards in Haematology, the American Society of Clinical
other hospital committees and meetings. Even in hospitals Oncology, the Society of Thoracic Surgeons, and others. In
where PBM has had difficulty in getting a foothold and recent years, a number of well-done randomized clinical
demonstrating realization of blood cost savings, the addition trials have provided high-quality evidence for the basis of
of a full-time transfusion safety officer has been shown to professional practice guidelines related to transfusion of the
significantly decrease blood utilization. This results in pro- various blood components. One example is the AABB Clin-
moting adherence to transfusion guidelines through an ac- ical Practice Guidelines for Red Blood Cell (RBC) Transfu-
tive role in education of physicians that were identified as sion Thresholds and Storage.16
noncompliant during audits. Case reports have demon- A well-written facility transfusion guideline will contain
strated blood cost savings approaching $500,000, which the indications for transfusion and thresholds used in screen-
more than pays for itself in terms of a full-time salary for a ing and auditing of transfusion records.17 If the existing
transfusion safety officer.15 guidelines are not current, then current evidence-based med-
icine should be reviewed, and the guidelines amended to re-
The Transfusion Committee flect best practices. Since the study of the indications and
dosing for transfusion has been rapidly evolving over the last
The transfusion committee may be a separate entity or a part two decades, changes are occurring frequently. Evidence-
of a facility’s larger quality-assurance or laboratory utilization based guidelines should be carefully scrutinized and in-
committees. It is part of the requirement to provide a mecha- cluded only if they fit within the scope of the hospital’s
nism for continuous improvement and quality assurance to practice and patient population. In cases where the evidence
ensure a facility’s hemotherapy practices are compliant and is limited, adoption of expert consensus panels should be
subject to identification of areas for improvement. In addition considered in conjunction with local practices. When devel-
to the transfusion service medical director, key blood bank oping a specific transfusion guidelines policy for a hospital,
medical technologists such as the general supervisor, and the it is important to use a multidisciplinary approach. Repre-
facility’s TSO (if applicable), the committee should be made sentatives from specialties with high blood utilization should
up of representatives from services that routinely transfuse be involved, including cardiac surgery, hematology, intensive
blood. This often includes clinicians from anesthesiology, sur- care, and emergency services. In addition to national practice
gery, emergency medicine, intensive care, obstetrics, neonatal guidelines, the practice standards within the community
care, pediatrics, and internal medicine. Nursing staff from key must be considered. Transfusion guidelines may be concise
areas should also be represented, as well as quality assurance. and one-page, or large and detailed documents. This is often
A representative from the institution’s blood supplier often determined by the particular hospital culture and level of ac-
participates as well. The committee should be chaired by a ademic versus practical interest. Guidelines will ideally be
clinician who is able to facilitate enumeration, approval and made available as an electronic document, and the judicious
achievement of the committee’s goals, as well as champion the inclusion of hyperlinks to definitions, articles, or other
work of the committee to the medical executive leadership source material and resources may enhance the utility of the
and facility peers. The committee should meet at least quar- document. Most national guidelines include one or more
terly to perform routine work related to utilization review but hematology laboratory values as a “trigger” or “threshold.”
may need to meet more frequently or in subcommittees to ad- Although the term “trigger” has been commonly used,
dress specific projects or issues.12 The transfusion committee “threshold” is preferable. The former implies action should
may also serve as a facility’s PBM committee, or there may be be taken (i.e., transfusion ordered) while the latter more
a separate dedicated group. Some facilities utilize a separate broadly implies a value at which one could consider action.
committee for the patient-focused-care aspect of PBM de- These thresholds can be useful in informing the develop-
scribed later, which then liaises with the transfusion commit- ment of a transfusion guideline policy. As an example, the
tee for larger context. AABB guidelines for RBC transfusion indicate that most
6888_Ch26_552-573 29/10/18 5:08 PM Page 558

558 PART V Quality Management and Compliance

patients may benefit from transfusion at the 7 g/dL hemoglo- other component-specific information. For example, a rec-
bin threshold, and some patients (those with heart condi- ommendation for single-unit transfusion of RBCs for the
tions, perioperative, etc.) may benefit from transfusion at the nonbleeding patient, with reassessment prior to ordering
higher threshold of 8 g/dL hemoglobin.16 Similarly, the AABB additional units, serves the purpose of providing needed he-
recommends that most adult patients may benefit from motherapy without transfusing more blood than is necessary.
platelet transfusion at the 10,000 platelets/µL threshold, and It is important to emphasize general guidelines that affect
some patients (perioperative) may benefit from platelet trans- the hospital’s patient population and will form the basis for
fusion at the higher threshold of 50,000 platelets/µL.18 These most physicians’ and surgeons’ clinical decision making. An
upper and lower thresholds can be a convenient starting point example of content for a simple one-page sample transfusion
during physician transfusion education. guideline is shown in Table 26–4.
In addition to thresholds, guidelines typically contain in- As mentioned above, physician groups with high utiliza-
dications, contraindications, dose recommendations, and tion rates should also be offered the opportunity to review

Table 26–4 Sample Transfusion Guidelines


Component Type Typical
Adult Dose Sample Indications For Transfusion
(These are sample indications and may change as new evidence develops. Actual indications must
be approved by transfusion committee and be supported by current evidence.)

Packed Red Blood Cells Prophylactic Hemoglobin less than 7.0 g/dL threshold is appropri-
1 Unit typically raises hemoglobin 1 g/dL. ate for hospitalized, hemodynamically stable patients,
Typical therapeutic endpoint is hemoglobin including critical care patients
between 7.0 and 9.0 g/dL.
Hemoglobin less than 8.0 g/dL threshold is recom-
mended for patients undergoing orthopedic/cardiac
surgery or with preexisting cardiac disease

Bleeding or Symptomatic Profuse bleeding or symptomatic anemia


Platelets Prophylactic Platelet count less than 10,000/µL threshold is
1 Apheresis Unit typically raises platelet appropriate for hospitalized adult patients
count by 20,000 to 60,000/µL.
Minor Invasive Procedures Platelet count less than 20,000/µL threshold is ap-
propriate for patients undergoing minor invasive pro-
cedures such as central venous catheter placement

Major Surgery or Bleeding Platelet count less than 50,000/µL threshold is ap-
OR propriate for perioperative patients and patients un-
Invasive Procedures With Increased dergoing invasive procedures with increased bleeding
Bleeding Risk risk such as lumbar puncture or biopsy of solid
organs such as liver, lung, or kidney

Neuraxial Surgery/Bleeding Platelet count less than 80,000–100,000/µL and


bleeding risk or surgery of brain, spinal cord,
inner/middle ear, eye

Plasma Major Surgery or Bleeding INR greater than 2.0 threshold is recommended for
2–6 Units depending on severity. OR perioperative patients with excessive microvascular
Minor Invasive Procedures bleeding (coagulopathy) in the absence of heparin
and in patients undergoing minor invasive proce-
dures such as inferior vena cava filter, peripherally
inserted central catheter placement, thoracentesis,
or paracentesis

Neuraxial Surgery or Bleeding INR greater than 1.5 threshold is recommended for
OR patients with neuraxial bleeding/surgery involving the
Procedures With Increased Bleeding brain, spinal cord, inner/middle ear, or eye OR in pa-
Risk tients undergoing invasive procedures with increased
bleeding risk such as lumbar puncture or biopsy of
solid organs such as liver, lung, or kidney

Emergent Warfarin Reversal Emergent warfarin reversal before Vitamin K effect

Cryoprecipitate Major Surgery or Bleeding Fibrinogen less than 80–100 mg/dL and excessive
5–10 Units typically raise fibrinogen 50 to bleeding (higher thresholds may be indicated for
75 mg/dL obstetric bleeding)
6888_Ch26_552-573 29/10/18 5:08 PM Page 559

Chapter 26 Patient Blood Management 559

and comment upon the proposed guidelines. Emphasis policy. The order (or set) contains blood component and
should be placed on the hazards of transfusion, therapeutic component-related orders that constitute the desired thera-
indications, and conservative transfusion practices. It is crit- peutic intervention for a specific clinical circumstance. This
ical to obtain input from clinical staff in support of guidelines process may be paper or electronic. Such tools should be
to improve compliance by ensuring they are broad and flex- simple, intuitive to use, and not cluttered or overwhelming.
ible enough to meet the majority of clinical situations. If They must also be readily available, whether electronic or
guidelines are too restrictive or ignore significant clinical lit- paper-based. If electronic, careful consideration should be
erature consensus opinions or guidelines in favor of more given to the design of “drop down” menus to avoid inadver-
stringent recommendations from the transfusion medicine tent selection of the wrong choice. An example of an elec-
literature, compliance will be poor. Published evidence- tronic blood order is shown in Figure 26–3. It is important
based guidelines and consensus opinions forming the basis to keep in mind that blood order sets are not foolproof meth-
of the guidelines should be referenced. These references are ods for preventing inappropriate orders. Order sets that are
sometimes omitted at the hospital level but should still be too complex, difficult to access, or too restrictive will result
available on the hospital intranet or within the blood bank. in the development of “workarounds” to bypass the screen-
Often, separate transfusion guidelines are developed for ing process. In one study, those who had employed this tactic
adult and pediatric patients due to the significant difference reported they were directed to do so by a more superior
in the two populations. Appropriate physician documenta- physician. Such “solutions” on the part of clinical staff im-
tion is critical when transfusion orders appear to deviate pede flow in the blood bank, increase waste, and inhibit the
from established guidelines. ability to provide timely service.19
Although the development of the hospital transfusion Generally, any blood utilization program will use a screen-
guideline document is extremely useful in improving blood ing tool such as the blood order set. The TSO is ideally suited
utilization, the document is ineffective without a successful to engage stakeholders and assist in creation and adoption.
education and promotion campaign to gain acceptance Care should be taken to address issues such as massive and
among the physician leadership and eventually the entire emergent transfusion. It may be necessary to address special
physician staff. In particular, difficulties within the hospital populations such as neonates and pediatric patients with
environment, poor communication, and resistance to develop separate specifically tailored order sets. Blood should not be
new initiatives have been shown to be significant obstacles denied simply because a specific clinical indication was not
to transfusion guideline adherence.17 The hospital medical, included among the choices on the blood order form. These
nursing, and administrative leadership can be useful in dis- rare instances require consultation with the blood bank
semination of the transfusion guidelines. Continuing educa- physician before issuing or denying a product and may pro-
tion and committee meetings are other useful avenues. vide opportunities for continuous improvement of the form
through updated versions. Based on the level of comfort and
Blood Order Sets experience with blood bank staff, the standard operating pro-
cedures should address the types of criteria required to con-
Blood order sets are a standardized method to clarify indi- tact the medical director. One issue that can occur with
cations and aid guideline compliance during physician order blood order sets (either inadvertently or as a “work around”)
entry. They contain standard, predefined orders that have is when the indication does not appear to match the patient.
been developed according to the approved guidelines and For example, a 52-year-old patient admitted for a knee

Figure 26–3. An example of electronic blood order set options


Component Type Packed Red Blood Cells
for packed red blood cells. Expanded order sets may also include
premedication and administration options. They may also be tied
Priority Routine
to decision support algorithms and coupled with interruptive alerts
(Choose from dropdown) STAT
for orders that do not meet facility guidelines.

Dose 1 Unit
(Choose from dropdown) 2 or more units (complete Special Instructions section)

Indication HGB less than 7


(Choose from dropdown) Cardiac disease and HGB less than 8
Surgery and HGB less than 8
Symptomatic anemia
Profuse bleeding
Other (complete Special Instructions section)

Special Instructions
(free text, may require
medical director
consultation)
6888_Ch26_552-573 29/10/18 5:08 PM Page 560

560 PART V Quality Management and Compliance

replacement has an order for transfusion of two units of transfusion. A retrospective and representative sample of
RBCs, and the indication selected is “extracorporeal mem- records from transfused patients should be reviewed to iden-
brane oxygenation (ECMO).” Auditing (discussed later) tify existing issues and patterns of misuse. The review should
should include a plan to discover and address these types of compare the indications used for actual transfusions against
orders. the transfusion guidelines. Specific areas and physician prac-
tice groups should be ranked by total utilization and appro-
Decision Support and Computerized priateness. Next, the members of the PBM committee who
Order Entry are considering the available resources and the desired out-
comes should determine specific goals. Program complexity
The advancement of technology with the electronic medical should be determined, including type, frequency, and scope
record and the laboratory information system has allowed of audit. Special attention should be given to each type of
for the creation of decision support algorithms during com- blood component. As well as sharing common risk factors,
puterized physician order entry (CPOE). Decision support packed red blood cells, platelet concentrates, plasma prod-
usually consists of a series of additional questions or warn- ucts, and cryoprecipitate have individualized risk with differ-
ings that appear during blood order entry, and alert the or- ing probabilities of generating specific adverse reactions. In
dering practitioner of a potentially unnecessary transfusion addition, cost and wastage concerns vary according to com-
based on existing laboratory values. In some instances, the ponent. Will all components be reviewed equally? Will there
decision support algorithms may be guided by the ordering be disproportionate representation of more frequently or-
physician’s chosen transfusion indication. Interruptive alerts dered components? Consideration must also be given to
provide information about facility transfusion guidelines and which patient populations will be audited, such as pediatric,
policy and how the selected order deviates from that prac- neonatal, high-complexity surgery, hematology-oncology, and
tice. The provider can then choose to cancel the order, alter so on. Particular populations may require tailored auditing
the order, or proceed; although, “hard stops” (inability to and intervention methods. Particular attention should be
proceed) are also possible. The novel approach to automat- placed on improving patient safety. Therefore, appropriate
ing some aspects of PBM are most useful in cases where medical oversight and review must be incorporated into each
transfusion decisions are primarily guided by a particular development stage as the program is designed.
laboratory threshold (such as transfusing stable, nonbleeding
patients using a trigger of 7 g/dL). However, these ap- Review Criteria
proaches lose effectiveness in more complex or critical situ-
ations.20 The decision-support algorithms must be carefully After the transfusion guidelines are updated and the scope
designed and validated and should not hinder or delay the of the program is defined, criteria should be generated for
ordering of blood in emergency situations. An additional use in the audit process. If the transfusion service is small
benefit of decision-support algorithms is the ability to auto- and sufficient personnel are available, a 100% manual review
matically generate prospective blood utilization reports, as of records could be considered. However, as the number of
discussed below. In conjunction with an existing blood transfusions increases, this method can quickly exhaust
utilization review process, decision-support algorithms available resources. Screening criteria can be selected to flag
successfully decreased blood orders by 12% to 25% at a large cases with a high probability of inappropriate use and dese-
multihospital system.21 Additionally, although there is a lect those with low probability. This maximizes the efficiency
paucity of prospective clinical trials, decision-support algo- of the review process. Algorithms can be developed that in-
rithms appear to be safe and may improve quality of care.22 clude laboratory data and information from blood order sets.
Unfortunately, without an aggressive prospective blood Screening criteria that are too specific will fail to identify
utilization program within the blood bank, decision-support many inappropriate transfusions, while overly sensitive
algorithms are not always effective in stand-alone situations. criteria will result in detailed review of many appropriate
Some studies have shown that decision support is not useful transfusions, making the program inefficient. Screening
in changing the ordering practices and behavior of physi- thresholds using fixed laboratory values or excluding certain
cians who do not wish to follow guidelines.23 These methods conditions or indications may be used to focus the audit. Al-
using computerized order entry may also not be effective in gorithms must be incorporated into the standard operating
reducing unnecessary two-unit orders without a primary procedure, and should be validated for both effectiveness and
focus on traditional provider educational strategies.24 to make sure that no delays or patient harm occurs. During
design, consideration should be given to potential automa-
Utilization Auditing and Review tion and incorporation into CPOE to enable the possibility
of automated review, discussed later.
Facilities are required to have blood utilization review pro- Blood utilization review attempts to optimize transfusion
grams that ensure ongoing monitoring and appropriate inter- utilization by minimizing inappropriate transfusion. Most
vention related to their transfusion practices. The AABB, The programs emphasize overutilization of blood products; only
Joint Commission, and The College of American Pathologists a few institutions monitor for underutilization of blood. This
all require these processes. Once guidelines are written and is because studies that have addressed underutilization have
approved, the next step is to assess the scope of inappropriate failed to show significant practice deviations. However, with
6888_Ch26_552-573 29/10/18 5:08 PM Page 561

Chapter 26 Patient Blood Management 561

continued efforts to lower transfusion thresholds and reduce facilities experience limitations in electronic systems and data-
overutilization, it is conceivable that the risk may increase.25 base compatibilities, and perform a hybrid-type review that is
This may be especially true if educational efforts to improve partially automated and partially manual.
utilization are strongly focused on potential transfusion- Although there is still an absence of multicenter prospec-
associated adverse events. At an academic center with an ac- tive studies that compare blood utilization outcomes among
tive PBM program, a review of medical records revealed no the various education and intervention strategies, the near
evidence of transfusion underutilization in a study of 93 se- future is promising. This new field of implementation sci-
verely anemic or thrombocytopenic patients who were not ence may provide clues into the best intervention strategies
transfused. However, the time required to examine the med- in improving blood utilization. Implementation science stud-
ical record and difficulty of discerning the justification of ies the appropriateness and effectiveness of educational and
withholding transfusion, poses a considerable obstacle to behavioral strategies in promoting change in clinical practice
screening for undertransfusion. It is suggested that monitor- and the adoption of evidence-based medicine into actual
ing processes should be included in the blood utilization clinical practice. As applied to transfusion medicine, there
audit process.25 is over a decade’s worth of evidence that most intervention
Blood utilization review is comprised of three broad cat- strategies within the context of PBM are useful in improving
egories of audits: retrospective, concurrent, and prospec- both transfusion safety and blood utilization. More research
tive. These strategies have direct corollaries from managed is needed, but there is currently no front-runner strategy that
care.26 As in managed care, each category has specific advan- appears more useful than the others. There is also no defin-
tages and requires a different labor investment. Two subtypes itive advantage of a multipronged intervention approach ver-
of prospective review—targeted and discontinuous—are sus the rigorous application of just one strategy. In some
particularly useful when there are resource constraints. ways, this is intuitive, since the purpose of intervention
The feature common to all methods is the requirement for strategies is to educate physicians; once this is accom-
comparison of transfusion records from individual patients plished, no further training is needed except for reinforce-
to criteria developed from standardized guidelines. Each ment.28 In fact, this is supported anecdotally when a
transfused component is assessed for medical necessity, hospital with a successful retrospective intervention strategy
adherence to indication guidelines, component dosing, and failed to show additional improvement in utilization using
timing of administration. It is then categorized as either aggressive prospective review.29 Just as individualized med-
appropriate or inappropriate. Results of the audit are often icine is patient specific, it may turn out that each hospital
submitted for further review and possible intervention strate- may require a certain intervention strategy, or combination
gies. Some methods are better suited for particular targeted- of strategies, based on factors such as hospital culture, pres-
end goals. ence of a formal continuing education program, hospital-
Reviews can be manual, semiautomated, or automated. based versus private-practice relationships, and hospital
Manual reviews are often time-consuming, involve detailed system policies. Of course, this is speculative, and we
medical record review of the entire selected audit population, eagerly await additional evidence. No matter which type of
and require the involvement of clinical health-care profes- review is performed, the goal is to educate clinicians and
sionals such as nurses or physicians or medical residents. raise their awareness of current guidelines, thereby improv-
Semiautomated reviews include computerized screening cri- ing future transfusion decisions.27
teria that flag particular records for manual review. A screen-
ing process may add efficiency to the process by removing Retrospective Review
records for review that are below a predetermined screening
threshold and by flagging the remaining records for review.27 Retrospective blood utilization review can be performed
It is important to note that the screening threshold is differ- after the transfusion event has taken place. It can occur when
ent from the review criteria threshold. Screening thresholds convenient without interrupting other staff duties. Detailed
must be carefully selected to prevent missing inappropriate chart reviews can be performed by the TSO, nursing, or
transfusions, yet not so strict as to compromise efficiency physician staff to determine appropriateness. Large numbers
and speed. Consideration of the screening thresholds should of transfusions can be reviewed and compared to previous
emphasize the attainment of utilization target goals but be data if medical records are readily available. Hence, this
in balance with the available resources. They are usually method is often used to generate trending reports on a large
delineated in terms of a specific algorithm. scale, including utilization within the hospital, particular
Automated review is carried out completely by algorithm physician practice group, or diagnosis and billing code.
and can be very quick and expeditious. Such reviews lack the In retrospective reviews, the feedback communication
flexibility of a manual system that employs the use of clinical usually consists of letters, focused educational efforts, or
judgment, but can be an excellent method to display trend- both. Retrospective review with weekly physician feedback
ing. Many times the screening process or automated review has been associated with improved mortality, adherence to a
can be performed entirely electronically.18 The automated restrictive transfusion strategy, and improved single-unit
screening process is particularly effective when a transfusion transfusions.30 The major disadvantage of this methodology
indication must be linked to a decision support function is that the inappropriate transfusion has occurred and is
when computerized physician order entry is available. Many therefore not prevented, merely identified later.1 Another
6888_Ch26_552-573 29/10/18 5:08 PM Page 562

562 PART V Quality Management and Compliance

drawback is that feedback and education of the clinician oc- the blood bank.27 Possible outcomes include approval of the
curs remotely from the event, and indications for transfusion order after additional information is provided, consultation
in the medical record are usually vague or suboptimal, de- and agreement upon a more appropriate component, or a
creasing the likelihood that the clinician will remember the more informed decision resulting in cancellation of the
specifics of the transfusion decision.31 When a retrospec- transfusion order.
tively audited transfusion record is flagged, it is necessary to Criteria for prospective blood utilization review are usu-
rule out that the transfusion was not given for unexpected ally derived from the use of triggers and thresholds that are
or emergent bleeding. In particular, it can be difficult to as- equal to or more liberal than the hospital’s transfusion guide-
certain if a particular transfusion was inappropriate if the lines. These thresholds usually involve hemoglobin, platelet
medical record does not unambiguously document the indi- count, or coagulation testing for RBCs, platelets, or plasma,
cation for transfusion. respectively. Examples of possible prospective audit thresh-
olds include the ordering of two or more units or a pretrans-
Concurrent Review fusion hemoglobin of more than 7 g/dL.
If the prospective review process is used, the correspond-
Managed-care audit models define concurrent review as a ing blood bank standard operating procedure (SOP) should
type of retrospective utilization review that occurs shortly address four issues:
after delivery of service, usually during the hospital stay.26
1. The type of screening method must be incorporated into
Concurrent review is a type of retrospective review that oc-
the procedure, including required computer data entry or
curs shortly after the transfusion while case details are still
algorithm. Validation of the screening algorithm should
fresh in the mind of the ordering practitioner. It provides the
be specifically addressed to prevent errors.
blood bank with the opportunity to obtain further informa-
2. There must be specific instructions for handling flagged
tion to facilitate review.12 In blood utilization models, it al-
blood orders detailing the circumstances under which
lows review of the relevant guidelines with the ordering
products are issued, denied, or the physician consulted.
physician to encourage future compliance. This type of re-
3. There must be a process that describes what action to
view process is more labor-intensive than retrospective re-
take in unusual circumstances or if the physician evalu-
views because of the additional requirement to contact the
ator cannot be reached.
ordering physician. It can, however, be an important one-
4. An efficient method of recording details of the event, in-
on-one education engagement role for the TSO. Like the ret-
cluding the pertinent discussion, disposition of the com-
rospective audit, interventions occur after transfusion and
ponent order, and the name of the approving physician
focus on preventing future occurrence.
evaluator, is needed.

Prospective Review This process should not cause unnecessary delay in issu-
ing components for patients in urgent need. However, the
A prospective blood utilization review is the most complex majority of transfusions are not performed urgently, and
method and combines review with real-time intervention. If there is ample time for consultation to make the best thera-
adopted on a national scale, blood cost savings have been es- peutic choice for the patient. A well-written SOP with ade-
timated to approach $130 million.27 This type of audit quate training time is essential to ensure a robust, consistent
methodology uses manual or semiautomated methods and process. It is important to estimate the time requirements for
requires real-time evaluation of component requests. If this process. The process should not delay the turn-around
prospective review is performed, the blood bank staff will be time to dispensation of blood components. A complex
intimately involved in the audit process. Blood bank person- prospective review process requiring detailed medical record
nel, typically the staff performing crossmatch and compati- review for all blood orders may add a considerable workload
bility testing, are responsible for correctly screening and potentially require increased staffing. Other obstacles to
transfusion orders in a timely manner that does not delay performing prospective reviews are the considerable time
preparation of the component. The clinician’s order is re- and resources required and the potential for conflict between
viewed in real time in light of available patient laboratory the transfusion service and clinician.12
data and clinical information. It is compared to evidence-
based criteria and the facility’s established transfusion guide- Targeted Prospective Review
lines. The component is issued as ordered if transfusion is A targeted prospective review is a subtype of prospective
indicated, within guidelines, and the component is optimally review used during a drug utilization review to pinpoint a
selected. If the order appears to deviate from facility- high-priority, expensive, or potentially hazardous medica-
approved transfusion guidelines, or if the requested compo- tion.32 When this concept is applied to the blood bank, the
nent is suboptimal, blood bank personnel can communicate targeted prospective review involves ongoing prospective
with the clinician to obtain more information or offer guid- monitoring of one or a few specifically selected products or
ance and education. Mechanisms must be in place for noti- indications. These should be chosen from areas where inter-
fying the on-call physician evaluator, and prospective review vention and behavioral modifications are expected to have
is often effective in hospitals with an active graduate medical high-yield results. For example, some hospitals have high
education program with medical residents rotating through usage of platelet products on their cardiac surgery units. A
6888_Ch26_552-573 29/10/18 5:08 PM Page 563

Chapter 26 Patient Blood Management 563

targeted prospective review would specifically focus on cardiac surgeons at their institution on adherence to single-
platelet orders for cardiac surgery patients. This type of re- unit RBC transfusion orders and the agreed-upon hemoglo-
view is also useful to concentrate consultation and commu- bin threshold of 8 g/dL.30 Although improvement was noted,
nication between the blood bank medical director and the when those same metrics were presented with individual
surgeon or physician specialists targeted. There is some evi- physician-specific behavior identified, significantly greater
dence that suggests that prospective review techniques may adherence was noted. In fact, the rate of single-unit transfu-
be useful in directing blood conversation strategies, improv- sions improved to greater than 90%. This study demonstrates
ing consistency of transfusion-ordering practices, and low- leveraging peer disclosure in the regular feedback of metrics
ering bleeding risk.33 This process is most useful when to ordering physicians may have a greater impact on driving
resources are limited and specific inappropriate utilization physician behavior, especially where education has produced
“targets” are readily identifiable. However, evidence suggests limited results.30
that targeted approaches may not be optimal for promoting
culture change and that utilization strategies are best ap- Intervention Strategies
proached using a hospital-wide model.34 Therefore, this type
of review process likely has greatest utility in jump-starting Each particular hospital and situation may require different
a new PBM/BUM program, or in quickly intervening when interventions or perhaps a combination of strategies to mit-
there is an unexpected increase of a particular blood com- igate inappropriate practices, and in some cases, interven-
ponent type that appears to be localized to a particular tions that work at one facility may not work at another.24
medical specialty or physician group. Interventions may be categorized as educational, punitive,
or confrontational. They may be employed individually or
Discontinuous Prospective Review collectively, depending on the culture of the institution. Un-
A discontinuous prospective review is a process that can be fortunately, there have been no large-scale studies completed
employed by facilities with limited transfusion medicine re- to compare the effectiveness of different intervention strate-
sources, or by facilities with only occasional deviations from gies. However, there are some studies that are ongoing.28 De-
transfusion utilization benchmarks. It is a method to con- spite this, as discussed below, most intervention strategies
serve resources when intervention that is more aggressive is are useful in improving utilization outcomes.
not expected to yield a significantly better response. Typi- Educational strategies may require a hospital-wide focus
cally, it is combined with retrospective or concurrent review if inappropriate utilization occurs in multiple departments
strategies and is selectively triggered when significant devi- or in groupings that are not easily categorized. Lectures,
ations are identified. It may be limited to a particular product grand rounds, and job aids are all useful resources. If greater
type, DRG (diagnosis related group—a categorization system inappropriate usage is found among specific physician
utilized for medical billing, based on diagnosis) clinical ser- practice groups or in patients with a particular disease,
vice, or individual clinician. In fact, a multi-institutional classification-focused educational interventions are useful.
study revealed that fewer than 30 diagnostic categories Small group lectures or discussions (“lunch and learn”) and
accounted for more than 75% of the cost of transfusion.35 phone conversations are excellent vehicles to relay current
Once improvement has been demonstrated and a steady state practice guidelines.
achieved, intense review may be discontinued until signifi- Punitive strategies involve reporting aberrant ordering
cant deviations are again identified. The risk in employing practices to certain peer groups or hospital administrative
this strategy is that during periods of “relaxation,” appropri- groups relevant to the ordering physician. These groups
ate monitoring does not occur, and the review process is not must agree with the mission of the blood utilization program
reactivated in a timely manner. and endorse the metrics and guidelines or such reports will
be ignored. Transfusion utilization may be included in
Feedback annual “physician report cards.” Practitioners may then be
required to complete annual training that emphasizes
Although the preceding strategies focus on correcting aber- evidence-based practice and the facility guidelines criteria.
rant ordering behavior, positive feedback should occur as Confrontational methods involve preventing or obstruct-
well. The development of physician champions has been ing certain physician orders. With institutional approval,
successful in improving quality in other areas of health system-wide constraints can be placed on transfusion orders.
care.36 Physicians who practice consistently within estab- Restrictions prevent the release of specific blood components
lished guidelines should be recognized, and where possible, for indications that are blatantly ineffective or better reme-
asked to share their practice philosophy with peers. These died by other lower risk or more cost-effective strategies. An
individuals may make excellent “utilization ambassadors,” example would be restricting all “fresh” blood to certain age
assisting in shifting the institutional culture toward the de- populations. Gatekeeping is another confrontational method
sired goal. As practicing clinical peer physicians, they will that requires approval of any transfusion order that does not
also have an additional level of credibility with other clini- meet predefined criteria. Decision support logic added to
cians versus the communications with laboratory personnel blood order sets is an example of a gatekeeping role that has
or transfusion service medical directors. Beaty and colleagues been shown to reduce inappropriate blood utilization.20,23 This
utilized weekly service-specific feedback reports to the intervention can also be used to deny release of components
6888_Ch26_552-573 29/10/18 5:08 PM Page 564

564 PART V Quality Management and Compliance

for transfusion order requests when indications are clearly in- practice, and adverse event monitoring and reporting. Mon-
appropriate. If product release is denied or delayed, it must fol- itoring and remediating issues with, or improving upon met-
low specific direction from the transfusion medicine physician rics and processes such as these, are required to meet
or must follow strict adherence to explicit instructions for standards and/or regulatory requirements. An added benefit
denial within the standard procedures. is the ability to discover how some of these may drive facility
If planned interventions include component denial or ad- culture toward inappropriate or wasteful practices. This type
ministrative actions, then it is of great importance to have of review can inform creation of new tools, such as a maxi-
the transfusion committee endorsement before implement- mum surgical blood order schedule (MSBOS). The MSBOS
ing such actions. Consideration should be given to methods is a list of suggested preoperative blood orders for various
of creating official hospital policy and seeking administrative types of common surgeries in an institution. It is based on
support. In addition, consensus and support should be ob- expected blood use for a particular procedure and makes a
tained from physician practice groups that frequently order corresponding recommendation for transfusion-related
components or have expected utilization issues. Groups with ordering. Therefore, no blood-related order or testing may be
poor utilization should be notified and educated before any indicated, a type and screen only may be recommended, or a
stricter policies are enforced. Mechanisms must be included specific number of units may be suggested for type and cross-
for bypassing any potential for delay in a truly emergent sit- match. In this way the MSBOS has the potential to decrease
uation such as during massive hemorrhage. Prior to imple- delays and waste by decreasing excessive orders while assist-
mentation, all practitioners should be notified of the process ing appropriate proactive component preparation.37 Of
for ordering outside the criteria. There should also be a course, the blood bank must continue to have a mechanism
process to mitigate potential conflicts and prevent delays or in place to address unexpected emergency bleeding.
perceived delays with ordering practitioners.
Intervention strategy styles should be paired with an ap- Patient-Focused Care
propriate audit type, since not all are complementary to each
other. Figure 26–4 shows a schematic of the interactions be- Most PBM programs concentrate heavily on RBC transfu-
tween blood utilization review and intervention strategies and sion. Red blood cells usually have the largest budget impact
the potential impact on the ordering clinician. It is important on hospitals and are also associated with unique risks such
to remember that even in facilities where there are significant as life-threatening acute hemolysis that occur even at the
improvement opportunities, many of the transfusions are ap- start of single-unit transfusions. By far the most common ap-
propriate and will have a therapeutic effect. The program proach to navigate the complexities of a hospital-wide PBM
should not be structured in such a way as to limit the capa- strategy is by the use of the so-called three pillars of patient
bilities of the primary clinician, especially in emergencies. blood management. This is a physiological approach that re-
quires a thorough understanding of transfusion require-
Other Opportunities for Review ments. In its most current form, the first pillar is the
optimization of erythropoiesis, the second pillar is the min-
In addition to audit and review of the appropriateness of imization of blood loss and bleeding, and the third pillar is
transfusions ordered by clinicians, there are other opportu- to harness and optimize the physiological reserve of anemia.
nities for audit, review, and process improvement. These In the surgical setting, each of these pillars must be assessed
should also be tracked, trended, and acted upon as indicators and evaluated differently if the patient is awaiting surgery, in
of quality and safety. Examples include: appropriate prepara- surgery, or postoperative. With the pillar approach, at each
tion of blood components, turn-around time from order to operative phase, interventions from each pillar are enacted.
component issue and then to transfusion, improper specimen For example, the preoperative patient should be treated for
labeling, patient identification, transfusion-administration the underlying anemia (pillar 1), bleeding risk should be

Clinician autonomy Blood bank autonomy

Educational Punitive Confrontational


Group Retrospective
• Multimedia • Training activity • Restrictions
intervention strategies
• Lectures/training • Remediation • Gatekeeping

• Correspondence • Admin reporting • Coercion


Individual Prospective
• Conversation • Peer reporting • Denial
intervention strategies

Figure 26–4. Common intervention strategies in patient blood management. Strategies on the left encourage clinician autonomy while those on the right encourage
blood bank autonomy. Strategies near the top of the figure are designed for large groups, and those near the bottom are designed for individuals.
6888_Ch26_552-573 29/10/18 5:08 PM Page 565

Chapter 26 Patient Blood Management 565

identified and managed (second pillar), and patient’s physi- until anemia has been addressed should be the rule rather
ological reserve for anemia tolerance should be assessed and than the exception. The development of an anemia clinic for
optimized (third pillar). Similarly, such concepts are applied many elective surgery patients with iron deficiency anemia
with each pillar intraoperatively as well as postoperatively.38 can reduce RBC transfusion needs by over 50%. Also, patient
The involvement of these pillars at each step requires a outcomes are improved and hospital stays are actually
multidisciplinary approach that should begin prior to hos- shorter in patients managed by presurgical anemia clinics.42
pitalization. This includes programs such as presurgery ane-
mia clinics, development of appropriate order sets prior to Autologous Blood/Red Blood Cell Recovery
surgery, procedures for anesthesia and nursing personnel, Techniques
and appropriate training postoperatively. This method re-
quires practitioners have a working knowledge of evidence- In the past, presurgical autologous blood donation (PAD)
based transfusion requirements and a concept of the was a popular modality among patients and surgeons. As
therapeutic and treatment potential from RBC transfusion. such, it was intended to address both the patient’s concerns
The TSO should assist in the development and review of regarding transfusion-transmitted infections (TTI) such as
patient-focused programs and can provide evidence-based HIV, HCV and HBV, as well as concern surrounding the ad-
literature and training. As originally conceptualized, these equacy and availability of the volunteer donor blood supply.
pillars were designed for surgical patients. In particular, de- The PAD process allows the patient to donate his/her blood
finitive strategies can be used to reduce the need for trans- for his/her own use prior to surgery, which is then processed
fusion in the preoperative, operative, and postoperative and stored to be available for the peri- or postoperative pe-
setting. However, these principles appear to apply to all pa- riod. Unfortunately, nearly 14% to 50% of autologous blood
tients receiving transfusions, including medicine and oncol- collected presurgically is not used (wasted).43 Patients who
ogy patients.39 donate autologous blood are at higher risk of anemia at time
of surgery, since they usually have not had adequate time for
Correction of Anemia erythropoiesis to replace what has been withdrawn. For ex-
ample, there are some institutions that advise anemic pa-
The most common cause of anemia and decreased RBC mass tients to donate autologous blood within two weeks of their
is iron deficiency. Unfortunately, correction of iron status elective surgery.43 Since an increasing body of evidence high-
prior to surgery continues to be a missed opportunity to im- lights the importance of correcting anemia prior to surgery,
prove patient outcomes, despite the known risks of compli- it is counterproductive to encourage patients to perform PAD
cations from preoperative anemia associated with transfusion unless one of the following occurs: 1) the patient has a rare
after surgery.40 Iron deficiency occurs when the stored iron blood type or 2) has developed alloantibodies to high-
in the body is depleted and the bone marrow is unable to frequency antigens or 3) combinations of multiple antigens
produce sufficient hemoglobin within red blood cells. A that require very rare phenotype-matched products. Patient
decrease in stored iron can occur when there is a long-term religious or other strongly held convictions may also indicate
deficiency of iron in the diet, a disease state that limits the a need for PAD. However, for the majority of patients it is
ability to absorb iron; a bleeding or hemolysis state where generally not indicated; and for some, may not accomplish
iron in the form of hemoglobin is lost from the body; or the desired effect. In fact, a study of adult spine surgery pa-
other conditions such as certain medications and pregnancy. tients showed that autologous blood donation was associated
Excessive blood donation has even been linked to iron defi- with higher perioperative transfusion rates without any pro-
ciency. Iron deficiency anemia is treated through the admin- tective effect from exposure to allogeneic blood.44 Patients
istration of iron compounds either orally or intravenously. should be reassured in the current era of nucleic acid testing
Oral preparations are limited by slow absorption into the (NAT) that blood products are significantly safer with respect
bloodstream, and several months may be required to rebuild to bloodborne disease. Robust availability in recent years has
iron stores. For quicker response, one dose of intravenous also allayed most concerns related to “blood on the shelf” at
iron is sufficient to rebuild iron stores, and changes in he- the time of surgery. PAD has been particularly entrenched in
moglobin can often be seen within one week and remove the the literature and practice of orthopedics, and engaging these
necessity of oral iron therapy after surgery or chemother- surgeons to discuss the practice along with alternatives and
apy.41 Anemia due to causes other than iron deficiency may its appropriate limited use, represents an opportunity for the
be treated with other anemia-correcting medications like PBM program and a common role for the TSO. Discussions
iron formulations, vitamin B12, or folate, collectively known should include the potential use of alternatives that are as
as hematinics, as indicated.12 Erythropoietin, a drug that effective as autologous blood. For example, Kasparek et al.45
stimulates RBC production in the bone marrow, should be recently reported the use of topical tranexamic acid during
used with caution after careful consideration of benefit total hip arthroplasty as an equivalent strategy to targeted
versus risk of drug-related adverse events. Protocols requir- preoperative blood donation. In the modern context, modal-
ing hematinics with high expense or cost profiles should also ities other than PAD are available in select situations that
be approved by the hematology/oncology and pharmacy provide the opportunity to use autologous blood periopera-
leadership. When possible, surgery should be delayed to tively to avoid or minimize RBC transfusion. For example,
allow for correction of anemia. For elective surgery, delay patients with an appropriate preoperative hemoglobin, acute
6888_Ch26_552-573 29/10/18 5:08 PM Page 566

566 PART V Quality Management and Compliance

normovolemic hemodilution (ANH) may be utilized. ANH transfusion decisions during cardiac surgery. Most clinical
is a technique whereby a predetermined number of units of trials associated with VT have been with cardiac surgery.
the patient’s whole blood are collected just prior to surgery However, this testing methodology has also been used to
with simultaneous replacement of a colloid or crystalloid guide transfusion strategies in other specialty areas including
fluid. Thus, the patient’s remaining blood volume is diluted, trauma, obstetrics, liver transplantation, and hemophilia.
creating a relative anemia. The result is fewer RBCs are lost Various algorithms are often employed to standardize the
to bleeding during surgery, and tissue oxygenation may be interpretation of results, and guide responses based on them
enhanced due to the decreased blood viscosity and cardiac such as transfusion of platelets, fresh-frozen plasma (FFP),
compensation.12 The patient’s blood is then returned once or use of antifibrinolytics. Whole blood coagulation is as-
blood loss is controlled. sessed through each stage of clot formation, determining clot
RBC recovery techniques include autologous blood sal- kinetics and clot strength. In addition, viscoelastic testing
vage devices, as well as the recovery and reinfusion of blood also provides information regarding abnormal fibrinolysis.
shed from surgical wounds and drains. The use of autolo- These tests work by measuring physical parameters that es-
gous blood salvage devices, “cell savers,” involves the col- timate the current strength of a forming clot plotted against
lection of autologous patient blood that has bled into the time.48 In this way the processes of clot initiation, propaga-
sterile surgical site and reinfusion of this blood after simple tion, stabilization, and dissolution can be evaluated. These
processing. Centrifugation and washing removes platelets, reported parameters allow the distinction of the impact of
plasma, RBC stroma, and anticoagulant; red blood cells in fibrinogen versus platelets on overall clot strength. A variety
normal saline are returned to the patient. However, it is im- of activators and inhibitors can also be used in the assess-
portant to remember care must be taken when alternatives ment. Oxygenation or hemoglobin concentration are not
to transfusion are considered. PBM strategies should be measured, so while VT is useful in guiding therapeutic deci-
evidence-based and may only apply to certain patient popu- sions related to FFP, cryoprecipitate, and platelets, clinical
lations and situations. For example, for patients with higher judgment in conjunction with point-of-care hemoglobin or
preoperative hemoglobin, this technique may not be useful other similar tests are often employed to guide RBC transfu-
and can increase costs.46,47 sion. Operational deployment of VT varies. The testing may
Another RBC recovery technique is the postoperative re- be performed in the blood bank and interpreted by the trans-
covery and reinfusion of blood recovered from surgical fusion service medical director who assists in real-time blood
wounds or drains. This practice is generally limited to car- product consultation. Others perform the testing in the lab-
diac and orthopedic surgery. Recovered blood may be rein- oratory, with monitors to the operating suite for interpreta-
fused with or without washing, but a minimum of 200 mL tion of results by a treating clinician, for example the
is required. Devices are available for washing and concen- anesthesiologist. In some facilities the testing and interpre-
trating the shed blood. tation is performed entirely in the operating suite. In any
case, the key is a process that provides rapid testing and in-
Optimize Hemostasis terpretation to best facilitate optimal hemostasis and rapid
control of bleeding.
Every effort should be made to minimize perioperative blood A recent meta-analysis of 15 clinical trials that involved
loss by optimizing hemostasis. Careful examination of the pa- 8,737 cardiac surgery patients showed that viscoelastic test-
tient’s medications and any herbal or vitamin supplements that ing was ineffective at reducing mortality. In addition, VT was
could affect clotting function should occur before surgery. If not associated with improved patient outcomes in all cases,
possible, these should be discontinued at appropriate intervals except for acute kidney injury. The same meta-analysis con-
prior to surgery to ensure they do not facilitate bleeding. To cluded the clinical trials did demonstrate a decreased use of
reduce surgical bleeding, drugs such as desmopressin, RBC and platelet components associated with the use of VT.
ε-aminocaproic acid, tranexamic acid, and recombinant factor However, these findings may have been heavily influenced
VIIa have been utilized. The use of topical tranexamic acid in by bias arising from the difficulty in effectively blinding vis-
orthopedic surgery has been previously mentioned. As with coelastic testing results during an active cardiac surgery.49
any pharmaceutical, careful consideration of side effects and Despite these findings, viscoelastic testing does correlate to
adverse events, evidence for efficacy, and relative cost, should a decreased transfusion exposure in cardiac patients. How-
occur in the context of patient-specific circumstances before ever, the costs associated with this testing should be weighed
the decision is made to administer it. Other methods to facil- against alternative point-of-care coagulation algorithms be-
itate optimal function of platelets and coagulation factors in- fore adoption.50
clude maintaining patient normothermia and ensuring proper
metabolic homeostasis during surgery.38 Continuous Noninvasive Hemoglobin Monitoring

Viscoelastic Blood Testing An evolving technology in acute anemia management in the


ICU and surgical suite is continuous noninvasive hemoglo-
Viscoelastic testing (VT) includes laboratory equipment such bin measurement. These devices measure hemoglobin indi-
as rotational thromboelastometry (ROTEM) and thromboe- rectly through a mechanism similar to pulse oximetry. It has
lastography (TEG). These tests are most often performed as been suggested that these devices may be useful for observ-
point-of-care or near real-time tests in the laboratory to guide ing sudden downward trends in hemoglobin and serve as an
6888_Ch26_552-573 29/10/18 5:08 PM Page 567

Chapter 26 Patient Blood Management 567

early warning of the need for RBC transfusion. Unfortu- Predictive Modeling
nately, there has been discrepancy between hemoglobin
measurements from these noninvasive devices and labora- Predictive modeling is a type of personalized medicine ap-
tory hemoglobin measurements. Because of this, the use of plied to the blood bank. With predictive modeling, various
this device with PBM is controversial and is often limited to patient demographic and laboratory parameters including
research.51 Nevertheless, developing the capability to moni- disease staging, hematology, chemistry, and coagulation
tor hemoglobin without direct sampling and the associated parameters, are used to develop a personal score to predict
blood lost from phlebotomy, holds promise for rapid care individual expected or maximum transfusion requirements.
with less iatrogenic blood loss. Predictive modeling incorporates many of the PBM tools de-
scribed, but it is still an active area for research and validation.
Minimize Iatrogenic Blood Loss This technique was successfully demonstrated in predicting
the need for massive transfusion in liver transplant patients
Iatrogenic blood loss, the removal of blood from the patient through the use of viscoelastic coagulation findings and cir-
for laboratory testing, can be a significant contributor to ane- rhosis staging.54 As this technology develops, the blood bank
mia in the hospitalized patient. Protocols or standing orders and TSO should be actively involved in the development of
for periodic or daily laboratory testing can lead to significant blood utilization metrics related to the program.
cumulative blood loss, especially if multiple tubes are needed
due to different anticoagulant requirements. Some studies Education
have shown an average blood loss of approximately 30 mL
per day within the first 2 days of admission, and a typical Education is a key component of a PBM program, and must
loss of 16 to 30 mL per day for longer admissions.52 Over occur regularly in all areas where blood is utilized and at all
the course of the patient’s hospital stay, this phlebotomy loss levels of transfusion care (i.e., transfusing nurses, physician
can tip the scale to render the patient anemic and require extenders, physicians, etc.) in both the inpatient and outpa-
transfusion. One study showed intensive care blood losses tient settings. Physician training in transfusion medicine is
averaging 435 mL, with some patients losing 781 mL of variable and often limited to behavior learned “on the floor.”
blood from phlebotomy alone.53 Pressure from specialists or more senior medical staff may
Several strategies should be employed to decrease unnec- influence transfusion-related decisions made by medical res-
essary diagnostic phlebotomy. First, in combination with idents. Physician educational activities can be focused on a
quality and compliance requirements to eliminate the order- single practitioner or group. A direct phone call may be made
ing of unnecessary laboratory testing, standing or protocol- to a physician who appears to order inappropriately. Another
driven orders for daily testing in stable patients should be example is a small group discussion with a physician group
eliminated where appropriate. Protocols, procedures, and about current guidelines for transfusion, with focus on par-
policies should be examined for “hidden” test triggers. Edu- ticular components or clinical situations as appropriate. For-
cation regarding appropriate test-order practices (timing, mal lectures at grand rounds are also excellent for relaying
volume requirements, expected value of information) will transfusion guidelines to the community. Resident education
help to guide caregiver decisions. Redesigning test order is an ideal way to instill proper transfusion practice. If edu-
forms and leveraging CPOE may also be useful. cational efforts are burdensome on available resources and
When laboratory tests are necessary, there are several op- time, consideration should be given to participation in hos-
portunities for the laboratory to engage in decreasing the pital grand rounds or sponsorship of invited expert speakers.
overall volume withdrawn; thereby, lessening the impact of Brief prepackaged computer-based training modules can be
the phlebotomy on the patient. When appropriate, point-of- included annually to reinforce guidelines. Sharing the evi-
care devices are useful because they require smaller amounts dence that forms the basis for the facility’s transfusion guide-
of blood. The use of smaller tubes at the time of phlebotomy lines is key to understanding and buy-in. In some cases, it
also decreases blood loss without rendering insufficient vol- may mean convincing providers that new evidence has
ume for required tests. When possible, the laboratory may usurped traditional teaching. Perhaps the best example of
consider shared specimens for multiple assays to decrease this is two-unit RBC transfusions: providers were tradition-
the need for additional phlebotomy tubes to be drawn. ally taught that a “dose” was two units, and single-unit trans-
Once again, the TSO is an ideal resource to lead a multi- fusions were discouraged as either unnecessary or
disciplinary project such as reducing iatrogenic blood loss. ineffective. In 2014, the AABB joined the American Board of
The TSO can liaise with the laboratory and the clinical Internal Medicine Foundation’s “Choosing Wisely” initiative
teams to ensure bidirectional information flow and cooper- in partnership with Consumer Reports to foster discussion
ation. It is important that the laboratory understand the di- to avoid unnecessary and/or wasteful medical tests, proce-
agnostic and monitoring needs of the physicians and clinical dures, and treatments. At that time, AABB released its list
staff. At the same time, the clinical staff needs to understand “five things physicians and patients should question.” Those
the technical requirements for the laboratory to perform five things are (1) don’t transfuse more units of blood than
timely and reliable testing. Education by the TSO and em- absolutely necessary; (2) don’t transfuse RBCs for iron defi-
phasis on the important contribution to the overall PBM ciency without hemodynamic instability; (3) don’t routinely
strategy and patient care, are important to ensure adoption use blood products to reverse warfarin; (4) don’t perform se-
and compliance. rial blood counts on clinically stable patients; and (5) don’t
6888_Ch26_552-573 29/10/18 5:08 PM Page 568

568 PART V Quality Management and Compliance

transfuse O negative blood except to O negative patients and success of a blood utilization management program.6 The key
in emergencies for women of child-bearing potential with concept is that the program is not a static deployment of a com-
unknown blood group.55 The first recommendation also calls pleted product. Instead, ongoing monitoring of the program
for restrictive transfusion thresholds and single-unit RBC and its feedback loops are required. This accurately measures
transfusions. Leveraging campaigns such as this can enhance program performance and outcomes and enables identification
educational materials and efforts. Podlasek et al.56 reported of opportunities for improving the system and further moni-
a system-wide campaign at their health system focusing on toring. This monitoring is measured through carefully selected
encouraging single-unit transfusion, entitled “Why give two metrics that concisely measure performance objectives and give
when one will do?”, using hospital newsletters and computer hints and direction for program changes and improvements.
screensavers. They observed a decrease in the percentage This system with its metrics must also take into account new
of two-unit orders from 68% to 31% in the three hospitals and evolving external drivers and information as well. Without
with the highest rate of double unit orders following the an ongoing monitoring and improvement process with appro-
campaign. Approaches such as this, combined with tradi- priate metrics, desired outcomes in blood utilization may only
tional lecture, publication sharing, observational learning, be realized in the short term. Blood usage may then return to
or computer-based learning can augment and reinforce the previous state of inappropriate or overuse.59
evidence-based practice and drive behavior.
Nursing education should include both the whys and the Metrics
hows. In other words, attention should be paid to educating
nurses regarding transfusion-related adverse events, indica- There are no standardized metrics for blood utilization, de-
tions for transfusion, and the evidence-base for the facility’s spite numerous attempts to define them in the literature.
transfusion guidelines, including standardization, consis- Blood utilization metrics can be generally broken up into
tency, and detail-orientation in procedure performance. three categories and are usually measured on a monthly, an-
Many of the educational points listed above for physicians nual, or quarterly basis. First, there are metrics associated
will be appropriate for nurses as well. However, they will also with overall blood use or cost—these are quantity metrics.
require their own specialized, focused training since they Hospitals with little change in patient population can use the
play a key role at the interface between transfusion and pa- total blood cost or actual number of products. To correct for
tient and can also often be influencers when transfusion is changes in size, the cost or number of transfusions can be
being considered. reported per patient day, adjusted patient day, or number of
Sufficient lead time must be allocated for training and discharges. If a robust and helpful information technology
competency of technical staff, including determination of service is available, the use of DRG case mix indexes (a well-
limitations and documentation of responsibilities. The known metric of patient disease severity with hospital ad-
blood bank is the last stop before blood components are ministrators) have been shown to have a high correlation
released. Therefore, the blood bank staff should be thor- with blood transfusion rates and is an interesting area for
oughly versed with blood utilization policies and proce- metric development.60 The second type of metric is a quality
dures, including a thorough understanding of adverse metric, a measurement of inappropriate or unnecessary
reactions to transfusion and indications for transfusion. In transfusion. Examples of quality metrics include the propor-
particular, emphasis should be placed on urgent procedures tion of single-unit to dual-unit transfusions in nonbleeding
such as emergency release, massive transfusion protocols, patients and the average pretransfusion hemoglobin. In some
and attention to detail. Awareness of special transfusion cases, the number of transfusions administered above an ar-
needs for certain patient populations such as obstetric, pe- bitrary laboratory value that is less restrictive than the trans-
diatric, or oncology patients is also important.57 The exist- fusion guideline can provide a useful metric of the
ing blood bank quality report should include metrics from proportion of noncompliant transfusions. For example, a
the PBM and blood utilization programs, and compliance typical red blood cell indication for a stable patient would
and performance details for blood bank staff should be in- be a hemoglobin threshold of 7 g/dL. A useful metric would
cluded when prospective auditing or screening is per- be the number or proportion of transfusions administered
formed. Deviations from procedure and unexpected above 8 g/dL. This value should decrease as utilization im-
outcomes should be handled in a manner similar to routine proves.61 The third type of metric is targeted to particular
blood bank deviations. In particular, the blood bank staff objectives, populations, or physician specialty and is essen-
should be able to reference the transfusion guidelines and tially a measure of compliance or effectiveness. Particular
determine if a transfusion order is within guidelines, is an physician subspecialties’ transfusion rates can be compared
emergency need, or if further review is needed through ei- against particular patient diagnoses. Number of prospective
ther a blood utilization review process or approval by the audits performed by residents or blood bank staff can be use-
medical director or designee.27,58 ful to track policy compliance when prospective review is
utilized. The effectiveness of presurgery anemia clinics can
Continuous Improvement be assessed through the measurement of the proportion of
surgery patients admitted with anemia. The effectiveness of
Continuous improvement (also known as kaizen) through computerized order entry decision support algorithms can
LEAN or another quality process, is required for long-term be measured by determining the proportion of orders that
6888_Ch26_552-573 29/10/18 5:08 PM Page 569

Chapter 26 Patient Blood Management 569

were not compliant with a particular trigger. In the case of of an adversarial relationship. This forum also allows for the
RBCs, a possible metric is the number of orders submitted review of flagged occurrences of misuse and determination
for stable ICU patients with hemoglobin 7 g/dL or greater.9,21 of appropriate actions. However, in the philosophy of con-
Any metrics that are chosen should be meaningful within tinuous improvement, it is critical that metrics are shared
the system, readily evaluable, and widely disseminated. This across the entire system, and reporting is not confined to the
will allow for individuals, cross-functional continuous im- transfusion committee or hospital administration. This rec-
provement teams, and departments at all levels of the system ognizes all participants as stakeholders and opens the door
to continuously monitor their own subprocess performance for improvement suggestions to be generated by personnel
and progress toward improvement targets. It will also enable at all levels across the entire process. Organizational newslet-
teams to benchmark among themselves and against other ters or intranet home pages are ideal forums for this and for
components of the program. Many of these may already be disseminating evidence-based literature. To maintain support
trended and tracked by the blood bank or quality assurance for the transformation and build the foundation for a con-
personnel, such as crossmatch-to-transfusion ratio and num- tinuous improvement culture, it is absolutely critical that
ber of mislabeled or unlabeled specimens. However, instead management maintains personal two-way communication
of automatically including existing metrics, one should con- with every team. Likewise, insightful feedback should be
sider their potential impact across the entire system. Con- provided to teams and individuals when their recommenda-
sider not only what is measured and reported, but also how. tions cannot be adopted. Without such personal interaction,
For example, the number of mislabeled or unlabeled speci- employees will perceive that they are not truly empowered
mens in a particular time period is often tracked and re- and participation will decline.
ported to the transfusion committee and to individual
nursing units. Yet might this information have additional im-
pact if reported not just as the number of “defects” or op- CASE STUDIES
portunities for sentinel events, but also communicated as
extension of turnaround time? Case 26-1
Waste begets waste within a system, and in this example
the interval between the blood order and the availability of You are the blood bank supervisor at a medium-sized hos-
the component for transfusion is significantly prolonged pital. Administration has been impressed by reported
while a subsequent properly labeled specimen is obtained cost-reduction realized in other facilities with active blood
and delivered to the blood bank. Such delays may contribute utilization and management programs. You have been di-
to the perception that blood bank turnaround time is too rected to begin such a program in your facility.
long and may be partially responsible for the overproduction 1. What will you tell administration about cost reduction
issues previously noted in Table 26–2. Would selecting met- as a singular goal for a blood utilization and manage-
rics that both include the absolute numbers of such speci- ment program?
mens and quantify the delay in the system caused by them 2. What will be your first steps?
(in minutes or hours) increase the ability to drive behavior? 3. Who will be included on the blood utilization man-
Would it better demonstrate that as waste is removed, flow agement team?
improves, thus decreasing strain on blood bank resources 4. How will you establish appropriate utilization criteria?
and resulting in more timely provision of appropriate units? 5. What type(s) of review might you consider?
These types of questions must be considered, and ultimately
the metrics chosen will reflect the individual institution’s Case 26-2
goals and priorities, and inform future action.
You are the transfusion safety officer of a large, growing
tertiary-care hospital facility. Your facility’s blood utiliza-
Ongoing Monitoring and Improvement
tion management program consists of monthly retrospec-
Monitoring and continuous improvement may include most tive analysis of blood usage by diagnosis and procedure
of the same players involved in the planning stages. The codes. Your results are reported at the quarterly transfu-
medical director must be aware of trending patterns and an- sion committee, where review determines whether place-
ticipate changes to the screening criteria. A significant re- ment of a letter in the physician’s file is warranted. You
duction in usage may require changes in staffing and notice an increasing trend in both plasma usage and
ordering components from vendors. An increase in inappro- plasma wastage. You must report your findings and analy-
priate utilization or a failure to improve may require reallo- sis at the next transfusion committee meeting.
cation of resources and more intense auditing procedures. 1. How will you assess the source of the problem?
Additional personnel resources for chart review and clinician 2. How will you assess the decrease in value as a result
interaction may be required. of the problem?
The transfusion committee is an ideal forum for the re- 3. What strategies should be used to develop possible im-
view and assessment of the program’s success. Because of its provements?
multidisciplinary nature, feedback can be shared between 4. How will the corrective action be monitored?
the blood bank and clinicians, preventing the development
6888_Ch26_552-573 29/10/18 5:08 PM Page 570

570 PART V Quality Management and Compliance

SUMMARY CHART
 Enhanced safety, quality improvement, patient-focused employed to promote uniformity and complete docu-
care, resource conservation and waste reduction/cost mentation.
control are goals of a well-designed PBM program. The  Meaningful metrics are chosen to reflect the progress
program should be multidisciplinary, tailored to the of the system toward facility-defined goals. Trending
needs of the institution, rely upon evidence-based cri- and tracking these metrics allows monitoring of pro-
teria, and employ mechanisms for continuous moni- gram effectiveness and indicates opportunities for con-
toring and improvement. LEAN or another quality tinuous improvement.
process can be utilized to organize the system and pro-  Blood utilization review consists of retrospective, con-
vide consistency and value. current, and prospective methods. Based on current
 Blood utilization management is a process designed to and future needs, a facility may choose a single method
ensure that blood transfusions are administered in ac- or may use them in combination.
cordance with evidence-based guidelines and prede-  When review processes identify deviations, interven-
termined criteria. Effective program design and good tions are triggered. Like review processes, there are
compliance ensure that patients receive transfusions multiple interventions, and selected types are deter-
only when necessary and medically appropriate. mined based on institutional culture and needs. How-
 PBM in the context of patient-focused care seeks to ever, not all review processes and interventions are
avoid or minimize the need for transfusion through an- mutually compatible.
ticipatory management. Various strategies and inter-  Key personnel in PBM include the transfusion service
ventions that can be categorized to the “three pillars medical director, blood bank supervisory personnel,
of patient blood management” (pillar 1: optimization and representatives from nursing and the facility’s
of erythropoiesis; pillar 2: minimization of blood loss practicing physicians—especially areas of routine
and bleeding; pillar 3: harness and optimize physio- transfusion. Transfusion safety officers can play a key
logical reserve of anemia) are utilized in combination role in a successful PBM program. A representative
according to patient-specific plans. from information technology may also prove invalu-
 Standardization is achieved through thoughtfully able to the team.
prepared transfusion guidelines and carefully selected  Support and approval by hospital administration is
evidence-based criteria. Blood order sets may also be crucial to the survival and success of the program.

3. Optimal value as it relates to blood utilization is best


Review Questions
obtained by which of the following:
1. Which type of review does not require direct discussion a. Development of standardized cost metric for blood
between the ordering clinician and transfusion service utilization
personnel? b. Reduction in variabilities in transfusion ordering prac-
tice from evidence-based standards
a. Discontinuous prospective
c. Consistent reporting of outcome metrics such as
b. Targeted prospective
decreased sepsis or emergency room admissions
c. Concurrent
d. Decreasing peril and waste by encouraging bloodless
d. Retrospective
surgery and conversion to lower volume phlebotomy
e. Prospective
tubes
2. Which of the following is the most important first step in e. Use of LEAN practices to improve value and prevent
developing a comprehensive PBM/BUM program? waste by considering the ordering clinician as a customer
a. Identification of a qualified transfusion safety officer 4. To receive benefit from a transfusion the patient must
with excellent blood banking bench skills have:
b. Determination of patient populations within the hos-
a. A hemoglobin level less than 8 g/dL
pital with the highest blood utilization
b. An invasive procedure planned
c. Creation of a multidisciplinary transfusion committee
c. A pathological lesion or deficiency that can be reme-
to determine the category of blood utilization review
died by functioning stored components
d. Meet with key physician and nursing leadership to
d. A blood order signed by the attending physician
facilitate the creation of a hospital-wide transfusion
e. Understood the risks, benefits and alternatives, and
guideline
given informed consent
e. Determine the estimated cost savings through the im-
plementation of an anemia clinic for elective surgical
patients
6888_Ch26_552-573 29/10/18 5:08 PM Page 571

Chapter 26 Patient Blood Management 571

5. Which of the following statements best describes the most 9. Metrics should be:
important reason for creating a PBM/BUM program? a. Chosen to indicate progress during the process of
a. PBM/BUM decreases blood bank exposure to risk man- continuous improvement
agement and litigation by promoting optimal docu- b. Tracked and disseminated only to members of the
mentation of transfusion indication and expected blood utilization management team
outcome within the electronic medical record c. The same for all institutions
b. PBM/BUM reduces patient exposure to transfusion as- d. Selected only by the transfusion service leadership
sociated acute lung injury (TRALI) through physician e. Only qualitative in nature, since medical decision
education and by prospectively encouraging mitigation making is a complex process
strategies from the blood supplier
10. Which statement is most accurate regarding continuous
c. PBM/BUM improves patient safety by promoting
improvement as it relates to PBM/BUM?
evidence-based transfusion, transfusion avoidance
strategies, and prevention of inappropriate transfusion a. Blood utilization metrics should be periodically con-
d. PBM/BUM are laboratory and hospital regulatory re- verted to national standardized metrics
quirements that are essential for ensuring a hospital b. An example of an ideal PBM metric is the ratio of the
culture that promotes patient safety through the peri- facility cost of one unit of RBCs divided by the re-
odic direct observation and assessment of transfusion gional red blood cell cost
administration by nursing staff c. Continuous improvement should be performed by a
e. PBM/BUM provides significant and substantial direct different set of players than were used in the planning
and indirect cost savings to both patients and blood process
banks by reducing the number of unnecessary or in- d. Outcomes during prospective review should be lim-
appropriate transfusions ited to the transfusion committee
e. Feedback loops and appropriate metrics are required
6. Which of the following is an example of targeted prospec- to achieve long-term improvements
tive review as it related to blood utilization?
a. Continuing education on the proactive use of iron to References
correct anemia in presurgical patients 1. Savage W. Implementing a blood utilization program to opti-
b. Viscoelastic testing to assess real-time platelet need for mize transfusion practice. ASH Education Program Book. 2015;
cardiac surgery patients (1):444-7.
2. Shaz BH. Bye-bye TRALI: by understanding and innovation.
c. Hematologist directed erythropoietin clinic for anemic
Blood. 2014; 123(22):3374-6.
cancer patients prior to chemotherapy 3. Schlenke P, Spahn DR. Patient blood management: it is time to
d. Decision support pop-up restricted to routine orders streamline targeted therapy options. Transfusion Medicine and
for two or more units of RBCs Hemotherapy. 2015; 42(2):72-3.
e. Report of average RBC usage per patient for a targeted 4. Szczepiorkowski ZM, Dunbar NM. Transfusion guidelines:
when to transfuse. ASH Education Program Book. 2013;
procedure or physician group
2013(1):638-44.
7. Which of the following pairs best describes an interven- 5. Womack JP, Jones DT. Lean thinking: banish waste and create
wealth in your corporation. New York: Free Press; 2003.
tion strategy that is best paired with a utilization review
6. Womack JP, Jones DT. Lean solutions: how companies and cus-
category? tomers can create value and wealth together. New York: Free
a. Minimize unnecessary phlebotomy loss, discontinuous Press; 2005.
prospective review 7. Goodnough LT, Shander A. Patient blood management. Anes-
thesiology. 2012; 116(6):1367-76.
b. Periodic physician feedback, retrospective review
8. Chung KW, Basavaraju SV, Mu Y, van Santen KL, Haass KA,
c. Annual continuing education for medical residents, Henry R, Berger J, et al. Declining blood collection and utiliza-
prospective review tion in the United States. Transfusion. 2016; 56(9):2184-92.
d. Predictive modeling, retrospective review 9. Leahy MF, Hofmann A, Towler S, Trentino KM, Burrows SA,
e. Automatic cancellation of surgical blood orders, con- Swain SG, et al. Improved outcomes and reduced costs associ-
ated with a health system–wide patient blood management
current review
program: a retrospective observational study in four major
8. The PBM program planning team should include: adult tertiary care hospitals. Transfusion. 2017 2 Feb. doi:
10.1111/trf.14006.
a. Nursing representatives 10. sabm.org [Internet]. Society for the Advancement of Blood
b. Representatives from the facility’s practicing clinicians Management [cited 2017 Jun 1]. Available from: https://www
c. The blood bank medical director .sabm.org.
d. A laboratorian who is knowledgeable in blood bank 11. AuBuchon JP, Puca K, Saxena S, Shulman IA, Waters JH. Get-
ting started in patient blood management. Bethesda (MA):
policies and procedures
AABB; 2011.
e. All of the above 12. Becker J, Shaz B. Guidelines for patient blood management and
blood utilization. AABB; 2011.
13. Maskens C, Downie H, Wendt A, Lima A, Merkley L, Lin Y,
et al. Hospital-based transfusion error tracking from 2005 to
6888_Ch26_552-573 29/10/18 5:08 PM Page 572

572 PART V Quality Management and Compliance

2010: identifying the key errors threatening patient transfusion avoidance strategies and reduce bleeding in patients undergo-
safety. Transfusion. 2014;54(1):66-73. ing percutaneous coronary intervention: prospective cohort
14. Dunbar NM, Szczepiorkowski ZM. How do we utilize a trans- study before and after implementation of personalized bleeding
fusion safety officer? Transfusion. 2015;55(9):2064-8. risks. BMJ. 2015;350:h1302.
15. Levine RL, Walczyszyn BA, Zacharia G. The transfusion safety 34. Oliver JC, Griffin RL, Hannon T, Marques MB. The success
officer: an effective tool in patient blood management. Blood. of our patient blood management program depended on an
2015;126(23):4742. institution-wide change in transfusion practices. Transfusion.
16. Carson JL, Guyatt G, Heddle NM, Grossman BJ, Cohn CS, 2014;54(10 pt 2):2617-24.
Fung MK, et al. Clinical practice guidelines from the AABB: 35. Beguin C, Closon MC, Vandekerckhove P, Baele P. Concentra-
red blood cell transfusion thresholds and storage. JAMA. tion of transfusion resources on a few pathologies and a few
2016;316(19):2025-35. patients: Analysis of the comprehensive in-hospital patient
17. Hannon T, Gross I. Transfusion guidelines: development and database. Transfusion. 2007;47(2):217-27.
impact on patient blood management. 2nd ed. In: Saxena S, 36. Hare GM, Freedman J, Mazer CD. Review article: risks of ane-
editor. The transfusion committee: putting patient safety first. mia and related management strategies: can perioperative
2nd ed. Bethesda (MD): AABB Press; 2013. blood management improve patient safety? Can J Anesth.
18. Kaufman RM, Djulbegovic B, Gernsheimer T, Kleinman S, 2013;60(2):168-75.
Tinmouth AT, Capocelli KE, et al. Platelet transfusion: a clinical 37. Frank SM, Oleyar MJ, Ness PM, Tobian AA. Reducing unnec-
practice guideline from the AABB. Ann Intern Med. 2015; essary preoperative blood orders and costs by implementing
162(3):205-13. an updated institution-specific maximum surgical blood order
19. Wood EM, Yazer MH, Murphy MF. Blood transfusion in hospi- schedule and a remote electronic blood release system. Anes-
tals. Practical Transfusion Medicine. 2013;233-53. thesiology. 2014;121(3):501-9.
20. Goodnough LT, Shah N. Is there a “magic” hemoglobin num- 38. Hofmann A, Farmer S, Shander A. Five drivers shifting the para-
ber? Clinical decision support promoting restrictive blood digm from product-focused transfusion practice to patient blood
transfusion practices. Am J Hematol. 2015;90(10):927-33. management. The Oncologist. 2011 Aug 1;16(suppl 3):3-11.
21. Butler CE, Noel S, Hibbs SP, Miles D, Staves J, Mohaghegh P, 39. Gross I, Trentino KM, Andreescu A, Pierson R, Maietta RA,
et al. Implementation of a clinical decision support system Farmer S. Impact of a patient blood management program and
improves compliance with restrictive transfusion policies an outpatient anemia management protocol on red cell trans-
in hematology patients. Transfusion. 2015;55(8):1964-71. fusions in oncology inpatients and outpatients. Oncologist.
22. Goodnough LT, Maggio P, Hadhazy E, Shieh L, Hernandez- 2016;21(3):327-32.
Boussard T, Khari P, et al. Restrictive blood transfusion prac- 40. Lasocki S, Krauspe R, von Heymann C, Mezzacasa A, Chainey S,
tices are associated with improved patient outcomes. Spahn DR. PREPARE: the prevalence of perioperative anaemia
Transfusion. 2014;54:2753-9. and need for patient blood management in elective orthopaedic
23. Friedberg RC. Utility of alert-based CDSS in CPOE to improve surgery: a multicentre, observational study. Eur J Anaesthesiol.
compliance with plasma transfusion guidelines. J Pathol Inform. 2015;32(3):160-7.
2013;5(1):1-6. 41. Camaschella C. Iron-deficiency anemia. N Engl J Med.
24. Patel VM, Rains AW, Clark CT. Effectiveness of provider edu- 2015;372(19):1832-43.
cation followed by computerized provider order entry alerts in 42. Froessler B, Palm P, Weber I, Hodyl NA, Singh R, Murphy EM.
reducing inappropriate red blood cell transfusion. J Blood The important role for intravenous iron in perioperative patient
Transfus. 2016;2016:2859720. doi: 10.1155/2016/2859720. blood management in major abdominal surgery: a randomized
25. Hibbs S, Miles D, Staves J, Murphy MF. Is undertransfusion a controlled trial. Ann Surg. 2016;264(1):41-6.
problem in modern clinical practice? Transfusion. 2015; 43. Monsef JB, Figgie MP, Mayman D, Boettner F. Targeted pre-
55(4):906-10. operative autologous blood donation: a prospective study of
26. Abbey DC. Cost-based, charge-based, and contractual payment two thousand and three hundred and fifty total hip arthroplas-
systems. Boca Raton (FL): CRC Press; 2012; p. 106. ties. Int Orthop. 2014;38(8):1591-5.
27. Haldiman L, Zia H, Singh G. Improving appropriateness of 44. Kelly MP, Zebala LP, Kim HJ, Sciubba DM, Smith JS, Shaffrey CI,
blood utilization through prospective review of requests for et al. Effectiveness of preoperative autologous blood donation
blood products: the role of pathology residents as consultants. for protection against allogeneic blood exposure in adult spinal
Lab Med. 2014;45(3):264-71. deformity surgeries: a propensity-matched cohort analysis.
28. Lorencatto F, Stanworth SJ, Gould NJ. Bridging the research to J Neurosurg Spine. 2016;24(1):124-30.
practice gap in transfusion: the need for a multidisciplinary 45. Kasparek MF, Faschingbauer M, Waldstein W, Boettner CS,
and evidence-based approach. Transfusion. 2014;54(10pt2): Boettner F. Topical tranexamic acid is equivalent to targeted
2588-92. preoperative autologous blood donation in total hip arthro-
29. Zuckerberg GS, Scott AV, Wasey JO, Wick EC, Pawlik TM, Ness plasty. J Arthroplasty. 2017;32(4):1176-9.
PM, et al. Efficacy of education followed by computerized 46. So-Osman C, Nelissen RG, Koopman-van Gemert AW,
provider order entry with clinician decision support to reduce Kluyver E, Pöll RG, Onstenk R, et al. Patient blood management
red blood cell utilization. Transfusion. 2015;55(7):1628-36. in elective total hip-and knee-replacement surgery (part 1).
30. Beaty CA, Haggerty KA, Moser MG, George TJ, Robinson CW, A randomized controlled trial on erythropoietin and blood
Arnaoutakis GJ, et al. Disclosure of physician-specific behavior salvage as transfusion alternatives using a restrictive transfu-
improves blood utilization protocol adherence in cardiac sur- sion policy in erythropoietin-eligible patients. Anesthesiology.
gery. Ann Thorac Surg. 2013;96(6):2168-74. 2014;120(4):839-51.
31. Madrigal E, Prajapati S, Avadhani V, Annen K, Friedman MT. 47. So-Osman C, Nelissen RG, Koopman-van Gemert AW,
Adequacy of physician documentation and correlation with as- Kluyver E, Pöll RG, Onstenk R, et al. patient blood management
sessment of transfusion appropriateness: a follow-up study in in elective total hip-and knee-replacement surgery (part 2). A
the setting of prospective audits and patient blood manage- randomized controlled trial on blood salvage as transfusion al-
ment. Transfusion. 2016 Nov 1. doi.org/10.1111/trf.13917. ternative using a restrictive transfusion policy in patients with
32. Navarro, R: Managed care pharmacy practice. 2nd ed. Burling- a preoperative hemoglobin above 13 g/dl. Anesthesiology.
ton (MA): Jones and Bartlett Learning; 2009. 2014;120(4):852-60.
33. Spertus JA, Decker C, Gialde E, Jones PG, McNulty EJ, 48. Whiting D, DiNardo JA. TEG and ROTEM: technology and
Bach R, et al. Precision medicine to improve use of bleeding clinical applications. Am J Hematol. 2014;89(2):228-32.
6888_Ch26_552-573 29/10/18 5:08 PM Page 573

Chapter 26 Patient Blood Management 573

49. Serraino GF, Murphy GJ. Routine use of viscoelastic blood tests 55. Callum JL, Waters JH, Shaz BH, Sloan SR, Murphy MF. The
for diagnosis and treatment of coagulopathic bleeding in cardiac AABB recommendations for the Choosing Wisely campaign of
surgery: updated systematic review and meta-analysis. Br J the American Board of Internal Medicine. Transfusion. 2014;
Anaesth [Internet]. doi: 10.1093/bja/aex100. Epub 2017 May 4. 54(9):2344-52.
50. Wikkelsø A, Wetterslev J, Møller AM, Afshari A. Thromboe- 56. Podlasek SJ, Thakkar RN, Rotello LC, Fleury TA, Demski RJ,
lastography (TEG) or thromboelastometry (ROTEM) to mon- Ness PM, et al. Implementing a “Why give 2 when 1 will do?”
itor haemostatic treatment versus usual care in adults or Choosing Wisely campaign. Transfusion. 2016;56(9):2164.
children with bleeding. Cochrane Database Syst Rev. 57. Engelbrecht S, Wood EM, Cole-Sinclair MF. Clinical transfu-
2016;(8):CD007871. sion practice update: haemovigilance, complications, patient
51. Barker SJ, Shander A, Ramsay MA. Continuous noninvasive blood management and national standards. Med J Aust.
hemoglobin monitoring: a measured response to a critical re- 2013;199(6):397-401.
view. Anesth Analg. 2016;122(2):565. 58. Tavares MM, Diquattro PJ, Sweeney JD. Reduction in red blood
52. Salisbury AC, Reid KJ, Alexander KP, Masoudi FA, Lai SM, cell transfusion associated with engagement of the ordering
Chan PS, et al. Diagnostic blood loss from phlebotomy and physician. Transfusion. 2014;54:2625-30.
hospital-acquired anemia during acute myocardial infarction. 59. Mazzocato P, Stenfors-Hayes T, von Thiele Schwarz U, Hasson H,
Arch Int Med. 2011;171(18):1646-53. Nyström ME. Kaizen practice in healthcare: a qualitative analy-
53. Branco BC, Inaba K, Doughty R, Brooks J, Barmparas G, sis of hospital employees’ suggestions for improvement. BMJ
Shulman I, et al. The increasing burden of phlebotomy in the Open. 2016;6(7):e012256.
development of anaemia and need for blood transfusion 60. Stonemetz JL, Allen PX, Wasey J, Rivers RJ, Ness PM, Frank SM.
amongst trauma patients. Injury. 2012;43(1):78-83. Development of a risk adjusted blood utilization metric. Trans-
54. Pustavoitau A, Lesley M, Ariyo P, Latif A, Villamayor AJ, Frank SM, fusion. 2014;54:2716-23.
et al. Predictive modeling of massive transfusion requirements 61. Abelow A, Gafter-Gvili A, Tadmor B, Lahav M, Shepshelovich
during liver transplantation and its potential to reduce utiliza- D. Educational interventions encouraging appropriate use of
tion of blood bank resources. Anesth Analg. 2017;124(5): blood transfusions. Vox Sang. 2017;112(2):150-5.
1644-52.
6888_Ch27_574-589 29/10/18 5:05 PM Page 574

Chapter 27
Transfusion Safety and Federal Regulatory
Requirements
Judy Ellen Ciaraldi, BS, MT(ASCP)SBB, CQA(ASQ)
and Richard H. McBride, Jr., MS, MT(ASCP)SBB

Introduction Overview of the FDA’s Inspection Process Short Supply Arrangements


History of Biologics Regulation Inspection Authorities Biological Product Deviation Reporting
Public Health Service Act Inspection Procedures Product Recalls
Federal Food, Drug, and Cosmetic Act Enforcement Actions Fatality Reporting
Food and Drug Administration Advisory Actions: Warning Letter Medical Devices
Regulatory Process Administrative Actions: Suspension, Summary Chart
Manufacturers, Manufacturing, and Revocation Review Questions
Biological Products Judicial Actions: Seizure, Injunction, References
Statutes and Laws Prosecution
Regulations Current Good Manufacturing Practice
Guidance Documents Regulations
Center for Biologics Evaluation and Quality Control Unit
Research Quality Reviews
Registration and Licensure Additional Requirements of Biological
Registration Product Manufacturing
Licensure Alternative Procedures (“Variances”)
Unregistered Transfusion Services Contract Manufacturing

OBJECTIVES
1. Describe why laws governing the regulation of biological products were enacted.
2. Define a biological product manufacturer.
3. Describe the regulatory process.
4. List the requirements for and responsibilities of being registered with the U.S. Food and Drug Administration (FDA).
5. List the requirements for and responsibilities of FDA licensure.
6. Describe the FDA’s inspectional authority of biological product manufacturers.
7. Distinguish between licensed and unlicensed manufacturers and between interstate and intrastate commerce.
8. Describe the role of the U.S. Food and Drug Administration’s (FDA’s) current good manufacturing practice (CGMP) in biological
product manufacturing.
9. List the possible enforcement actions.
10. List the FDA’s five layers of safety for protecting the blood supply.

574
6888_Ch27_574-589 29/10/18 5:05 PM Page 575

Chapter 27 Transfusion Safety and Federal Regulatory Requirements 575

Introduction History of Biologics Regulation


The Department of Health and Human Services (DHHS) Public Health Service Act
is the U.S. government’s principal health agency for protect-
ing the health of all Americans. The Centers for Disease Con- The regulation of biological products in the United States
trol and Prevention (CDC), the Centers for Medicare and began when Congress passed the Biologics Control Act of
Medicaid Services (CMS), the National Institutes of Health 1902 (also known as the Virus-Toxin Law).2
(NIH), and the Food and Drug Administration (FDA) are In 1901, there was a serious epidemic of diphtheria that
the primary agencies of the DHHS with responsibilities resulted in a great demand for the diphtheria antitoxin. At
directly related to blood. DHHS is part of the Executive that time, there were no requirements for safety testing. A
Branch of the U.S. government. horse from which serum was used to make the diphtheria
In addition to regulating other products such as food, antitoxin had contracted tetanus, and injection of persons
veterinary medicine, and tobacco, the FDA promulgates and with the serum from the horse resulted in tetanus infection
enforces regulations to ensure the safety and efficacy of bio- in persons who received the antitoxin. The Biologics Control
logical products, drugs, and devices, which include human Act of 1902 was passed following the deaths of 13 children
blood and blood components, blood collection supplies and who received injections of the diphtheria antitoxin contam-
instruments, blood establishment computer software (BECS), inated with tetanus.3
blood bank reagents, and tests for relevant transfusion- The Biologics Control Act required biological products to
transmitted infections (RTTI). Regulations for biological be manufactured in a manner that ensured the safety, purity,
products promulgated under the Public Health Service (PHS) and potency of the product. The act included provisions for
Act and the Federal Food, Drug, and Cosmetic (FD&C) Act licensing products, suspending or revoking the license
are found in Parts 210 to 211, 600 to 680, and 800 to 820 of for violations, labeling requirements, and authorizing entry
Title 21, Code of Federal Regulations (CFR).1 These regula- of federal officials into manufacturing facilities to conduct
tions set forth the regulatory requirements under the inspections.
statutes, including adherence to current good manufacturing The Biologics Control Act was incorporated into the Public
practice (CGMP) requirements. In addition, the regulations Health Service (PHS) Act in 1944 as Section 351. The new
address licensing of biological products and establishment PHS Act required a biologics license to be in effect before a
registration, as well as product-specific standards for blood biological product is entered into interstate commerce.4 A
and blood components. biologics license application can be approved only after the
This chapter describes the history of biologics regulations, manufacturer demonstrates that both the product and the
the FDA’s regulatory process and requirements, inspection manufacturing facility meet standards to ensure the contin-
and enforcement activities, and the CGMP requirements for ued safety, purity, and potency of the products.
blood and blood components. The appropriate citations for The 1902 Act did not address the regulation of blood and
specific regulations are provided next to the topic. Common blood components. In 1970, the definition of a biological
abbreviations used here and throughout the chapter are product within the PHS Act was expanded to include blood
listed in Box 27–1. and blood components and derivatives.

Federal Food, Drug, and Cosmetic Act


BOX 27–1
The Federal Food, Drug, and Cosmetic (FD&C) Act was
Commonly Used Abbreviations passed in 1938 to further define the FDA’s regulatory author-
BECS Blood Establishment Computer Software
ity. This Act, which has been amended many times, requires
BLA Biologics License Application drug manufacturers to demonstrate that a drug is safe and
BPDR Biological product deviation report effective before marketing it. In addition, it authorizes facility
DHHS Department of Health and Human Services inspections of drug manufacturers and requires drug manu-
CBER Center for Biologics Evaluation and Research facturers to register. The FD&C Act prohibits the introduc-
CFR Code of Federal Regulations
CGMP Current good manufacturing practice(s)
tion or delivery for introduction into interstate commerce
CLIA Clinical Laboratory Improvement Act (1988) any food, drug, device, or cosmetic that is adulterated or mis-
CMS Center for Medicare and Medicaid Services branded.5 A drug or device is misbranded if its labeling is
CSO Consumer Safety Officer false or misleading. A drug is adulterated if the methods used
FDA Food and Drug Administration in its manufacture do not conform with CGMP to ensure the
FD&C Act Federal Food, Drug, and Cosmetic Act
FR Federal Register
drug is safe, and has the identity, strength, quality, and purity
FY 2008 Fiscal Year 2008 (October 1, 2007, to September characteristics that it is represented to possess.
31, 2008) The FD&C Act has been amended many times to incor-
OBRR Office of Blood Research and Review porate new provisions. For example, in 1976 the FD&C Act
OCBQ Office of Compliance and Biologics Quality was amended to strengthen the FDA’s authority to regulate
ORA Office of Regulatory Affairs
PHS Act Public Health Service Act
medical devices. In 1992, the Prescription Drug User Fee
RTTI Relevant transfusion-transmitted infections Act (PDUFA) was passed to expand the FDA’s capability to
hire reviewers using industry application and supplement
6888_Ch27_574-589 29/10/18 5:05 PM Page 576

576 PART V Quality Management and Compliance

submission processing fees in exchange for the FDA’s agree- therapeutic biological products, and the Center for Biologics
ment to complete drug reviews within specified time frames. Evaluation and Research (CBER), which regulates biological
Device user fees were first enacted in 2002. In 1997, the Food and related products, including blood, vaccines, allergenics,
and Drug Administration Modernization Act (FDAMA) was tissues, cellular and gene therapies, and related devices. The
enacted to implement many changes, including those to bring Office of Blood Research and Review (OBRR) and the Office
the regulation of biological products in closer harmony with of Compliance and Biologics Quality (OCBQ) in CBER are
the regulations for drugs. Some of the provisions in FDAMA responsible for overseeing the regulation and enforcement
include eliminating the need for individual establishment and of biologics regulations for blood and blood components.
product licenses, streamlining the approval process for man- The Office of Regulatory Affairs (ORA) is responsible for
ufacturing changes, and incorporating risk-based regulation performing FDA postmarket inspections of blood and blood
of medical devices. In 2007, the FDA Amendments Act component manufacturing facilities and other surveillance
(FDAAA) renewed the Prescription Drug and Device User activities.
Fee programs, increased the public transparency of current The FDA has identified five overlapping layers of safety
regulatory actions, and provided the FDA with numerous that work together to prevent an unsuitable blood compo-
new authorities to assess and act on safety issues that may nent from being collected or transfused to a patient.7 Blood
only be recognized after a drug is marketed. The Food and and blood component manufacturers must ensure they
Drug Administration Safety and Innovation Act (FDASIA) have processes in place to perform and monitor each of
was passed in 2012. It expands the FDA’s authorities and these procedures:
strengthens the agency’s ability to safeguard and advance pub-
1. Donor screening: Donors are informed about potential
lic health by giving the FDA the authority to collect user fees
risks to blood safety and are required to answer questions
from industry to fund reviews of innovator drugs, medical
about factors that may affect the safety of their blood. For
devices, generic drugs, and biosimilar biological products;
example, donors with a history of nonprescription injec-
promoting innovation to speed patient access to safe and ef-
tion drug use are deferred.
fective products; increasing stakeholder involvement in FDA
2. Blood testing: After donation, each unit of donated blood
processes; and enhancing the safety of the drug supply chain.
undergoes a series of tests for RTTIs, including HIV, HBV,
The 21st Century Cures Act was signed into law in 2016. It
and HCV.
is designed to help accelerate medical product development
3. Donor deferral records: Blood establishments must keep
and bring new innovations and advances to patients who
a current record of deferred donors and use it to make
need them faster and more efficiently.
sure they do not collect blood from anyone ineligible to
donate.
Food and Drug Administration
4. Quarantine: Donated blood must be quarantined until it
The FDA is the oldest comprehensive consumer protection is tested and shown to be free of RTTIs.
agency in the U.S. federal government. The FDA began as 5. Problems and deficiencies: Blood establishments must in-
the Division of Chemistry (later renamed the Bureau of vestigate manufacturing problems, correct all deficien-
Chemistry) in the U.S. Department of Agriculture. The cies, and notify the FDA when product deviations from
Bureau of Chemistry’s name changed to the Food, Drug, and CGMPs have occurred resulting in the distribution of
Insecticide Administration in 1927. That name was short- unsuitable products.
ened to FDA in 1930. Originally, the Laboratory of Hygiene In addition, plasma derivative products undergo pathogen
of the Marine Hospital Service was charged with enforcing inactivation/reduction procedures and pathogen reduction
the Biologics Control Act. In 1930, this laboratory was re- procedures are available for certain blood components.
named the National Institutes of Health (NIH). In 1937, the
Division of Biologics Control (later renamed as the Bureau Regulatory Process
of Biologics) was formed within the NIH to regulate biolog-
ical products. In 1968 the FDA was transferred to the Public Manufacturers, Manufacturing, and Biological
Health Services within what was then known as the Depart- Products
ment of Health, Education, and Welfare. The responsibility
for regulation of biological products was transferred from Biological product manufacturers are defined as “any legal
NIH to the FDA in 1972. In May 1980, education functions person or entity engaged in the manufacture of biological
were removed from the department and it became known as products subject to licensure under the PHS Act.” The term
the Department of Health and Human Services.2,6 “manufacturer” also includes any legal person or entity who
Since 1972, the FDA has overseen the safety, purity, and is an applicant for a license where the applicant assumes re-
potency of the U.S. blood supply by ensuring that blood and sponsibility for compliance with applicable product and es-
blood component manufacturers conduct their operations tablishment standards (21 CFR 600.3(t)). Manufacturing of
and manufacture their products in accordance with appli- blood and blood products means the collection, preparation,
cable laws and regulations, including CGMP. Biological processing, or compatibility testing by chemical, physical,
products are regulated in two FDA centers: the Center for biological or other procedures of any blood product that
Drugs Evaluation and Research (CDER), which regulates meets the definition of a drug as defined in section 201(g)
6888_Ch27_574-589 29/10/18 5:05 PM Page 577

Chapter 27 Transfusion Safety and Federal Regulatory Requirements 577

of the Act. It includes the manipulation, sampling, testing, products are safe and effective (for drug products) or safe,
or control procedures applied to the final product or to any pure, and potent (for biological products) before they are
part of the process and to the packaging, labeling, repackag- marketed in interstate commerce in the United States. They
ing, or otherwise changing the container, wrapper, or label- also require that all biological product, drug, and device
ing of any blood product package in furtherance of the manufacturers follow the applicable CGMP regulations and
distribution of the blood product from the original place of label their products appropriately. The FD&C and PHS Acts
manufacture to the person who makes final delivery or sale grant the FDA the authority to oversee and enforce the re-
to the ultimate consumer (21 CFR 607.3(d)). quirements described in the acts, including inspecting the
Biological products are defined as “any virus, therapeutic manufacturing facilities and imposing penalties for viola-
serum, toxin, antitoxin, vaccine, blood, blood component or de- tions when appropriate. Over the years, there have been sev-
rivative, allergenic product, protein (except any chemically syn- eral amendments to the Acts to further define the agency’s
thesized polypeptide), or analogous product or arsphenamine regulatory authority.
or derivative of arsphenamine (or any other trivalent organic
arsenic compound), applicable to the prevention, treatment, or Regulations
cure of diseases or condition of human beings” (42 USC 262(i)).
Biological products, including blood and blood components, are Agencies promulgate regulations to establish a specific stan-
unique in that they may be considered as biological products dard or requirement by manufacturers set by the federal gov-
under the PHS Act and as drugs or devices (depending on their ernment under the statutory authority granted by Congress
mode of action) under the FD&C Act. This means that both the (21 CFR 10.90). Regulations are an interpretation of the
CGMP regulations for finished pharmaceuticals (21 CFR 210 statutes and are legally binding on both the manufacturing
and 211) and the CGMP regulations for blood and blood industry and the government agencies charged with enforcing
components (21 CFR 606) apply to blood products for transfu- the laws. The regulations define specific minimum standards
sion. Table 27–1 provides a comparison of biological products that manufacturers must meet. They often do not provide
and drugs. specific procedures on how to meet the standards. There may
There are a variety of tools used by the FDA to commu- be many acceptable ways to achieve a required objective, and
nicate and enforce the regulatory requirements, agency rec- a requirement that is too specific may become outdated.
ommendations, and product standards. These include Proposed and final regulations are published in the Federal
regulations and guidance documents. Register (FR), an official publication of the federal govern-
ment, under the authority of the Administrative Procedure
Statutes and Laws Act. Regulations usually require notice and an opportunity
for the public to comment before they are finalized. FDA reg-
Statutes are the acts passed by Congress and signed into law ulations are codified in Title 21 of the CFR. The regulations
by the President to establish the FDA and to grant the agency applicable to blood and blood components are found in sub-
authority to fulfill its mission. Statutes such as the PHS Act chapter F, Parts 600 to 680, and subchapter C, Parts 210 to
and the FD&C Act require biological product, drug, and de- 211, of Title 21 of the CFR. Major regulations pertaining to
vice manufacturers to demonstrate to the FDA that their blood and blood components are listed in Box 27–2.

Table 27–1 Comparison of Biological Products and Drugs


Biological Products (Excludes
Medical Devices Regulated as
Biological Products) Drugs
Definition Medical products typically derived from living Finished dosage form (pill, solution, cream, etc.) that
sources that are used to prevent, treat, or contains an active pharmacological ingredient and is
cure diseases or conditions of human beings used to prevent, treat, or cure diseases or injuries in
humans

Examples • Blood and blood components • Over-the-counter medications


• Tissues, cellular therapies • Prescription medications
• Vaccines, antitoxins • Chemotherapies
• Plasma derivatives (e.g., albumin, clotting
factors, intravenous immune globulin)

Regulatory authority • Public Health Service Act • Federal Food, Drug, and Cosmetic Act
• Federal Food, Drug, and Cosmetic Act

CGMP regulations 21 CFR Parts 210 and 211 and 606 21 CFR Parts 210 and 211

Regulatory center in the FDA Center for Biologics Evaluation and Research Center for Drugs Evaluation and Research (CDER)
(CBER)
6888_Ch27_574-589 29/10/18 5:05 PM Page 578

578 PART V Quality Management and Compliance

• Are announced in the Federal Register through a Notice of


BOX 27–2 Availability
Major Regulations Pertaining to Blood and Blood • Are typically issued in draft format for public comment be-
Components1 fore implementation, although the FDA will issue a final
guidance without an initial draft when the agency finds that
Title 21 CFR, Chapter I, Subchapter C, Parts
prior public participation is not feasible or appropriate
210 Current good manufacturing practice in manufacturing • Are issued as a final guidance after the FDA has reviewed
drugs: general
the public comments
211 Current good manufacturing practice for finished
pharmaceuticals
Level 2 Guidance Documents (21 CFR 10.115(c)(2))
Title 21 CFR, Chapter I, Subchapter F, Parts
• Set forth existing practices or minor changes in interpre-
600 Biological products: general tation or policy
601 Licensing
606 Current good manufacturing practice for blood and blood • Include all guidance documents that are not classified as
components Level 1
607 Establishment registration and product listing for manufac- • Do not require a Notice of Availability in the Federal Register
turers of blood and blood components • Are immediately implemented, unless the FDA indicates
610 General biological product standards otherwise
630 Requirements for blood and blood components intended
for transfusion or for further manufacturing • Invite public comment
640 Additional standards for blood and blood products The public may comment on any guidance document
after final issuance and the FDA will determine if a revised
guidance document should be issued. Guidance documents
and memoranda applicable to blood and blood components
Guidance Documents can be found on the CBER website at www.fda.gov/
BiologicsBloodVaccines/GuidanceComplianceRegulatory
Guidance documents describe the agency’s current interpre- Information/Guidances/Blood/default.htm and at www.fda
tation of or policy on a regulatory issue. The recommenda- .gov/BiologicsBloodVaccines/GuidanceComplianceRegulatory
tions in FDA’s guidance documents are not legally binding Information/OtherRecommendationsforManufacturers/
on the manufacturing industry or the FDA unless specific MemorandumtoBloodEstablishments/default.htm.
regulatory or statutory requirements are cited. However, the
recommendations in these documents often become the in- Center for Biologics Evaluation and Research
dustry standard of practice. In addition, the FDA employees
generally act consistently with the recommendations set CBER regulates allergenic products (e.g., allergenic extracts),
forth in agency guidance documents, although they may de- cellular and gene therapy products, human tissue and tissue-
part from them with appropriate justification and super- based products (e.g., bone, skin, corneas), vaccines, (e.g.,
visory concurrence (21 CFR 10.115(d)(3)). hepatitis B vaccine, influenza virus vaccine), and blood and
Guidance documents (called memoranda up to 1996) are blood components.
issued in order to: Blood and blood components intended for transfusion
(e.g., Whole Blood, Red Blood Cells, Platelets, plasma prod-
• Describe the agency’s current interpretation of, or policy
ucts, Cryoprecipitated Antihemophilic Factor (AHF)) or for
on a regulatory issue.
further manufacturing (e.g., Source Plasma, Source Leuko-
• Establish principles, practices, procedures, or standards.
cytes) are regulated by the Office of Blood Research and Re-
• Assist the manufacturing industry and government agen-
view (OBRR). Examples of other biological products
cies by clarifying the regulations.
regulated by OBRR include:
• Explain how the manufacturing industry may comply
with the regulations. • Medical devices used for blood component collection and
• Provide application review and compliance approaches manufacturing, such as blood bags (with or without anti-
coagulants and preservative solutions), blood bank
Guidance documents are developed and issued under the
reagents, RTTI tests for screening blood donors, BECS,
Good Guidance Practices (GGP) regulations (21 CFR
apheresis collection devices, leukocyte-reduction filters,
10.115). GGP regulations allow for two types of guidance
pathogen reduction devices, and blood warmers, and
documents.
• Biological products used to treat and prevent diseases,
Level I Guidance Documents (21 CFR 10.115(c)(1)). such as plasma volume expanders, and hemoglobin-based
oxygen-carrying solutions.
• Set forth initial interpretations of statutory or regulatory
requirements Questions about the regulation of blood and blood com-
• Set forth changes in interpretation or policy that are of ponents should be sent to CBER’s Office of Communications,
more than a minor nature Outreach and Development, FDA, 10903 New Hampshire
• Can include complex scientific issues Avenue, Building 71, Room 3103, Silver Spring, MD 20993-
• May cover highly controversial issues 0002 (ocod@fda.hhs.gov).
6888_Ch27_574-589 29/10/18 5:05 PM Page 579

Chapter 27 Transfusion Safety and Federal Regulatory Requirements 579

Registration and Licensure inspected by FDA investigators. Registered blood establish-


ments that do not hold an approved BLA may only distribute
Registration their products in intrastate commerce.
The regulations exempt some blood establishments from
The FD&C Act requires manufacturers to register their registration (21 CFR 607.65). The exemption applies, but is
establishments with the FDA and list the products they man- not limited, to:
ufacture. Regulations for blood establishment registration
are found in the CFR (21 CFR 607). The following blood • Transfusion services that neither collect nor process blood
establishments are required to register with the FDA (21 CFR components and that only perform compatibility testing
607.20): (crossmatching) and transfusion, pool Platelets and Cry-
oprecipitated AHF units immediately before transfusion,
• Collection centers or issue bedside leukocyte-reduction filters with blood
• Community blood banks components, and are either Clinical Laboratories Improve-
• Component preparation facilities ment Amendments (CLIA) certified or have met equiva-
• Hospital blood banks lent requirements as determined by CMS;
• Plasmapheresis centers • Carriers that transport blood products; and
• Product testing laboratories • Brokers who do not take possession, manipulate, or rela-
• Storage and distribution centers bel the product.
• Brokers who take possession and manipulate and/or rela-
bel the product Table 27–2 summarizes the registration of blood establish-
ments. Additional information about registration can be found
Blood establishments must register within 5 days after be- on the CBER website at www.fda.gov/BiologicsBloodVaccines/
ginning their manufacturing operations or within 5 days GuidanceComplianceRegulatoryInformation/Establishment
after the submission of a biologics license application (BLA) Registration/BloodEstablishmentRegistration/default.htm.
(21 CFR 607.21). Blood establishment registration and prod-
uct listing must be submitted electronically through the Licensure
Blood Establishment Registration and Product Listing
System (21 CFR 607.22(a)) (www.fda.gov/BiologicsBlood The PHS Act requires a biologics license to be in effect if a man-
Vaccines/GuidanceComplianceRegulatoryInformation/ ufacturer wants to distribute biological products in interstate
EstablishmentRegistration/BloodEstablishmentRegistration/ commerce. A biologics license will be issued only after the
default.htm). manufacturer demonstrates that the product is safe, pure, and
The manufacturer must list all products and manufactur- potent and that the facility where the product is manufactured
ing processes performed at the establishment and register meets applicable requirements to ensure the biological product
each manufacturing facility. Registered establishments are continues to be safe, pure, and potent (21 CFR 601.2(d)).
required to update their registration each year (21 CFR Unlike most biologics manufacturers who typically have
607.21). In addition, all registered establishments are re- one BLA approval per product, blood establishments manu-
sponsible for complying with the FDA regulations, including facture multiple licensed products under one license and one
CGMP and applicable product standards, and are routinely BLA approval.

Table 27–2 Summary of the Registration of Blood Establishments


The Following Facilities Are Required The Following Facilities Are Exempt From
to Register: Registration:
Collection centers

Community blood banks

Component preparation facilities

Hospital blood banks Transfusion services that do not routinely collect blood; only perform cross-
matching and transfusion, pool Platelets and Cryoprecipitated AHF units
immediately before transfusion, issue bedside leukocyte-reduction filters,
and are either CLIA certified or have met equivalent requirements as
determined by CMS

Plasmapheresis centers

Product testing laboratories

Storage and distribution centers Carriers (only transport product)

Brokers (take possession and manipulate and/or relabel product) Brokers (do not take possession, manipulate, or relabel product)
6888_Ch27_574-589 29/10/18 5:05 PM Page 580

580 PART V Quality Management and Compliance

The review of a BLA involves two steps: evaluating the The 30-day wait is waived for some moderate changes that
submitted application documents (e.g., standard operating are reported as a Supplement-Changes Being Effected
procedures (SOPs), forms, labels, product quality control) (CBE) (21 CFR 601.12(c)(5)). For submissions reporting
and conducting a prelicense inspection of the manufacturing these moderate changes, the product may be distributed
facility (21 CFR 601.2). The applicant submits the necessary before the FDA has approved the change; therefore imple-
application forms and information about their operations to mentation of the change and distribution of the product
CBER (21 CFR 601.2).8 Consumer Safety Officers (CSOs) in made using the change are performed at the manufac-
OBRR evaluate the application and supporting documents. turer’s own risk. The FDA will review the submission, and
The CSOs will inform the applicant if additional information if deficiencies are identified, will notify the manufacturer
is needed to complete the evaluation of the documents. As to submit the necessary information. In some cases, the
part of the application review, CSOs and ORA investigators FDA may require the manufacturer to stop distribution
will conduct a prelicense inspection of the manufacturing until any identified deficiencies have been corrected.
facility. The FDA investigators will observe the manufactur- • Minor changes are reported to the FDA in an annual report
ing processes to determine if the product is prepared accord- (21 CFR 601.12(d)). Minor changes do not need to be ap-
ing to the regulations, CGMP, product standards, and the proved by the FDA before the product made using the minor
manufacturing description provided in the application. The change is distributed into interstate commerce. The FDA
applicant should respond to any inspectional observations will review the annual report to determine if the changes
noted during the inspection. have been reported in the proper category. The FDA will no-
After the applicant has addressed all deficiencies noted tify the manufacturer if the annual report contains changes
during the evaluation of the submitted application docu- that should be submitted as a supplement for FDA approval
ments and the facility inspection, the FDA will make a deci- and may require the manufacturer to stop distribution until
sion regarding licensure. Once the manufacturer is licensed, the supplement is reviewed and approved.
the license number must appear on the label of the products
approved in the application and the licensed products may Unregistered Transfusion Services
be distributed into interstate commerce. Licensed manufac-
The regulations exempt some blood establishments from reg-
turers are responsible for complying with all applicable FDA
istering with the FDA because they do not routinely collect
regulations, including CGMP and applicable product stan-
or process blood components and are either CLIA certified
dards, and are inspected by the FDA regularly.
or have met equivalent requirements as determined by CMS
Licensed manufacturers are required to inform the FDA
(21 CFR 607.65). The majority of unregistered blood estab-
about any intended change in manufacturing from previously
lishments operate as transfusion services that perform only
approved procedures (21 CFR 601.12). Originally, when a
compatibility testing (crossmatching) and transfusion, pool
manufacturer submitted a change to the FDA, the manufac-
Platelets and Cryoprecipitated AHF units immediately before
turer had to wait for CBER to approve the change before the
transfusion, and issue bedside leukocyte-reduction filters.
products could be distributed into interstate commerce. In
Even though they are not registered or licensed, these man-
1997, the CBER issued a new regulation to reduce the report-
ufacturers must still comply with the applicable FDA regu-
ing burden by the manufacturing industry.9 The regulations
lations, including CGMP and applicable product standards.
in the CFR were revised to be consistent with similar require-
There are certain instances when the FDA will inspect these
ments in FDAMA. Title 21 CFR 601.12 describes how
blood establishments; these are typically “for-cause” inves-
licensed applicants must report their manufacturing changes
tigations, such as investigating a transfusion-related fatality.
to the FDA. Manufacturing changes are divided into the fol-
In 1983, the FDA entered into an agreement with the
lowing three categories as determined by the potential of the
Health Care Financing Administration (HCFA), now called
change to adversely affect the safety, purity, and potency of
the Centers for Medicare and Medicaid Services (CMS).11
the product. CBER has published guidance to help blood es-
The memorandum of understanding (MOU) between the
tablishments determine the appropriate reporting category:10
FDA and CMS consolidated within CMS the responsibility
• Major changes (based on associated risk) are reported as for inspecting and surveying unregistered transfusion ser-
a Prior Approval Supplement (PAS) (21 CFR 601.12(b)). vices in order to minimize duplication of effort and to reduce
These submissions must be approved before the applicant the burden on the affected blood establishments. Transfusion
can distribute the product made using the change into in- services that engage in the compatibility testing and trans-
terstate commerce. fusion of blood and blood components, and do not routinely
• Moderate changes (based on associated risk) are reported collect or process blood components, are now covered by
as a Supplement-Changes Being Effected in 30 Days CMS. CMS, state survey agencies (including those in CLIA-
(CBE30) (21 CFR 601.12(c)). These submissions must be exempt states), and accreditation organizations (such as the
sent to the FDA for review and approval. The applicant College of American Pathologists (CAP) and AABB) conduct
may distribute the product made using the change in in- routine biennial surveys of transfusion services on behalf of
terstate commerce 30 days after the FDA has received the the FDA. The FDA does not routinely survey transfusion
submission, unless the FDA has informed the applicant services, although the FDA may inspect any transfusion ser-
not to distribute because of deficiencies in the submission. vice at any time.
6888_Ch27_574-589 29/10/18 5:05 PM Page 581

Chapter 27 Transfusion Safety and Federal Regulatory Requirements 581

Table 27–3 summarizes the regulatory oversight of blood inspections are conducted according to the policies and pro-
and blood component manufacturing facilities. cedures described in the ORA Investigations Operations
Manual (IOM)12 and in written compliance programs and
Overview of the FDA’s Inspection compliance policy guides. The compliance programs provide
Process instructions on how to conduct the inspection and specify
the process for recommending appropriate regulatory ac-
Inspection Authorities tions.13 The compliance policy guides inform investigators
and compliance officers about FDA policies and interpreta-
The statutory provisions granting the FDA the authority to tions on specific regulatory issues.14 These documents can
enter biological product manufacturing facilities for the pur- be found on the ORA website at www.fda.gov/aboutfda/
poses of conducting inspections are found in both the PHS centersoffices/officeofglobalregulatoryoperationsandpolicy/
and FD&C Acts. The FDA conducts prelicense and preap- ora/default.htm.
proval inspections, routine CGMP inspections, and for-cause Each FDA inspection is conducted using a risk-based sys-
investigations. tems approach by covering some or all of the following man-
FDA investigators responsible for conducting inspections ufacturing systems in a blood establishment operation to
receive intensive training in the technical and regulatory ensure compliance with applicable regulations and standards:13
aspects of the products and manufacturing facilities they in-
spect. The investigators for blood-related manufacturing are • Quality Assurance System: The system for managing qual-
specialized into two teams: (1) Team Biologics, which is re- ity within a manufacturing establishment and assuring
sponsible for inspecting plasma derivative, reagent, vaccine, overall compliance with CGMP and internal written stan-
and allergenics manufacturers, and (2) the Biologics Cadre, dard operating procedures (SOPs) and specifications
who inspect blood, plasma, and tissue establishments. This • Donor Eligibility System: The system that protects donor
reorganization was designed to facilitate inspections with safety as well as recipient safety
improved consistency and appropriate citations. • Product Testing System: The system that includes properly
testing products collected for transfusion or for further
Inspection Procedures manufacture for evidence of RTTIs consistent with 21 CFR
610.40, blood grouping and typing (21 CFR 640.5), and
As part of the biologics license application process, CBER crossmatching blood for transfusion by direct testing or
and ORA investigators conduct prelicense and preapproval electronically (21 CFR 606.151)
inspections of manufacturing facilities that have submitted • Product Collection, Component Preparation, and Labeling
a BLA or a supplement to an approved BLA. Following ap- System: The system that covers operations from collection
proval of the BLA, ORA investigators who are trained as of the blood product through component preparation and
members of the Biologics Cadre will inspect the manufac- labeling
turing facility. The Biologics Cadre also inspects unlicensed, • Quarantine/Inventory Management System: The system
registered blood establishments that only distribute blood that manages product quarantine, storage, and distribu-
and blood components in intrastate commerce. All FDA tion, and prevents release of unsuitable products

Table 27–3 Summary of the Regulatory Oversight of Blood and Blood Component
Manufacturing Facilities
Manufacturers That
Manufacturers That Do Not Perform
Engage in Interstate Manufacturers That Manufacturing Steps
and Intrastate Engage in Intrastate That Require
Commerce Commerce Only Registration
Requirements for registration Must be registered Must be registered Exempt from registration
Requirements for licensure Must hold an approved biolog- Do not need to be licensed Do not need to be licensed
ics license application
Applicable CGMP regulations 21 CFR 210, 211, 606 21 CFR 210, 211, 606 21 CFR 210, 211, 606
include:

Inspections Conducted by FDA: CBER Conducted by FDA: ORA Covered under CMS (CMS,
(prelicense and preapproval) investigators state survey agencies, accred-
and ORA investigators ited organizations); FDA
may conduct “for-cause”
inspections
Regulatory oversight FDA FDA CMS and FDA
6888_Ch27_574-589 29/10/18 5:05 PM Page 582

582 PART V Quality Management and Compliance

FDA investigators discuss their observations with the corrective actions and must not engage in interstate com-
manufacturer as the inspection progresses. At the end of merce of the product while the license is suspended.
the inspection, investigators list significant observations on A manufacturer’s license can be revoked for several rea-
Form FDA-483, Inspectional Observations. Investigators sons: the FDA’s inability to gain access to the facility to con-
present Form FDA-483 to the manufacturer and listen to duct a meaningful inspection, discovery of significant CGMP
the proposed corrective actions. Investigators may also dis- deficiencies, or a determination is made that the product
cuss less serious observations with the manufacturer to point is not safe or effective for its intended use or is misbranded
out areas that could potentially cause significant problems. (21 CFR 601.5). The FDA may also revoke a license when it
Both significant and less serious observations, as well as observes that the deficiencies have been ongoing and have
any discussions with the manufacturer, are included in the not been corrected after numerous inspections and prior no-
Establishment Inspection Report (EIR) prepared by the tice. The FDA may revoke a license or call for a voluntary
FDA investigators.12 Most inspections result in voluntary revocation if the manufacturer no longer makes the product,
compliance by the manufacturer to take corrective action, and therefore the FDA cannot conduct a meaningful inspec-
but inspections with numerous significant observations may tion or evaluation. Revocation will prohibit the manufacturer
trigger advisory, administrative, or legal actions, such as from distributing the product into interstate commerce. Ad-
warning letters, license suspensions or revocations, injunc- ministrative actions, such as suspensions and revocations,
tions, seizures, and prosecutions. are listed on the FDA’s website at https://www.fda.gov/
BiologicsBloodVaccines/GuidanceComplianceRegulatory
Enforcement Actions Information/ComplianceActivities/AdministrativeActions
Biologics/default.htm.
When a biological product manufacturer violates any of the
laws or regulations the FDA enforces, the manufacturer is
Judicial Actions: Seizure, Injunction, Prosecution
often given an opportunity for voluntary correction before
the FDA pursues enforcement actions. There are three types Seizure is an action taken to condemn violative products
of FDA actions: advisory, administrative, and judicial.15 and remove them from distribution. The FDA and the U.S.
Marshals Service take possession of the product typically
Advisory Actions: Warning Letter under a court order. The court usually gives the owner of the
seized product 30 days to decide on a course of action. If the
Advisory actions alert manufacturers that they have violated
owner does not communicate proposed actions, the FDA
FDA regulations and provide an opportunity for corrective
will dispose of the product. The owner may contest the
action before further compliance action is taken. Warning
charges and litigate in court to have the seized products re-
Letters are one example of an advisory action. The FDA will
turned. The owner is required to provide a monetary deposit
issue a Warning Letter to a biological product manufacturer
(e.g., a bond) to ensure the court’s orders will be carried out.
for violations of regulatory significance. A Warning Letter is
An injunction is a civil process initiated to stop or prevent
a written communication from the FDA to the manufacturer
violation of the law, such as to halt the flow of violative prod-
notifying them that their product, practice, or other activity
ucts in interstate commerce and to give the manufacturer an
is in violation of the law. The Warning Letter serves as a prior
opportunity to correct the conditions that caused the viola-
notice that the FDA may decide to take further action, and
tion to occur. The FDA may seek to obtain an injunction
it offers the manufacturer an opportunity to correct the de-
when a health hazard has been identified or the manufac-
ficiencies listed in the letter. The FDA will determine if cor-
turer has a history of violations and the evidence suggests
rective actions have been implemented during the next
that serious violations will continue.
inspection of the manufacturing facility. Warning Letters
An injunction may be used whether or not the manufac-
may have significant financial and legal ramifications for a
turer holds a biologics license. If there is no license to sus-
company. These letters are available on the FDA’s website at
pend or revoke, an injunction may be the only way for the
www.fda.gov/ICECI/EnforcementActions/WarningLetters/
FDA to halt the flow of violative products in interstate com-
default.htm.
merce and to correct the conditions that caused the violation
Administrative Actions: Suspension, Revocation to occur. If a manufacturer violates the terms of the injunc-
tion, a court may impose fines or even hold the company or
Administrative actions of license suspension or license revo- specific corporate officers in contempt, and the FDA may
cation apply only to licensed manufacturers. These actions are take further enforcement actions.
more formal than advisory actions, such as issuing warning A prosecution is a criminal action directed against a man-
letters, and the agency usually must follow certain proce- ufacturer or responsible individual(s) or both. The FDA may
dures, such as providing notice and an opportunity for a hear- consider referring a matter to the Department of Justice for
ing. In addition, an affected company can seek a judicial prosecution when fraud, health hazards, or continuing sig-
review of the FDA’s administrative decisions. The FDA nificant violations are encountered. The FD&C Act allows
may suspend a manufacturer’s license if a danger to health individuals or corporations for whom prosecution is being
exists (21 CFR 601.6). The FDA can administer this action considered to be offered an opportunity to explain to the
immediately, and the manufacturer must take appropriate agency why the prosecution is not appropriate. Prosecution
6888_Ch27_574-589 29/10/18 5:05 PM Page 583

Chapter 27 Transfusion Safety and Federal Regulatory Requirements 583

will proceed without a hearing if the violations are fraudulent In 1963, the FDA published the first drug CGMP regula-
or the responsible individuals are likely to flee. Table 27–4 tions after observing that unsuitable products were being re-
summarizes the enforcement actions. leased to the public.16 The FDA believed that this was due
to an increase in number and complexity of the tests being
Current Good Manufacturing Practice performed and untrained or undertrained personnel deviat-
Regulations ing from established standards. During inspections, the FDA
observed that there were often no controls in place to mon-
Under the FD&C Act, a drug is adulterated if the methods itor the whole manufacturing process. The CGMP regula-
used in, or the facilities or controls used for, its manufacture, tions were published to provide direction for this control and
processing, packing, or holding do not conform to or are not include the elements of quality assurance, quality control,
operated or administered in conformity with current good and process validation. The CGMP regulations for blood and
manufacturing practice. Drugs must meet the requirements blood components were published in 1975.17 Blood and
of the FD&C Act as to safety and must have the identity and blood component manufacturers must follow both the
strength and meet the quality and purity characteristics that CGMP regulations in 21 CFR 210 and 211 and the more spe-
it purports or is represented to possess. cific CGMP regulations for blood and blood components in
CGMP regulations for finished pharmaceuticals are found 21 CFR 606. The more specific requirements in 21 CFR 606
in 21 CFR 210 and 211, and CGMP regulations specific for supplement and do not supersede the more general CGMP
blood and blood components are found in 21 CFR 606. The requirements set forth in 21 CFR 210 and 211.
CGMP regulations describe the minimum current good man-
ufacturing practices and require all manufacturing facilities Quality Control Unit
to implement and follow these requirements. The FD&C Act
similarly provides that a medical device is adulterated if the The CGMP regulations require each manufacturer to designate
methods used in, or the facilities or controls used for, its a quality control unit that has the responsibility and authority
manufacture, packaging, storage, or installation are not in to approve or reject all components, containers, closures, la-
conformity with device CGMP regulations, which the FDA beling, and drug products. Although the regulations identify
has issued in the Quality System Regulations found in 21 this unit as a “quality control unit” (21 CFR 211.22), many
CFR 820. manufacturers identify it as a quality assurance unit.

Table 27–4 Summary of Enforcement Actions15


Advisory actions Warning Letter • Notifies manufacturer that their product, practice, or other activity is in violation of the law
• Serves as prior notice that the FDA may take further action
• May be issued to both licensed and unlicensed manufacturers

Administrative actions Suspension • Applies to licensed manufacturers only


• May occur if reasonable grounds for revocation and a danger to health exist
• Provides immediate withdrawal of the authorization to ship a biological product in interstate
commerce
• Manufacturer cannot engage in interstate commerce until deviations are corrected and sus-
pension is lifted
• FDA can take this action quickly

Revocation • Applies to licensed manufacturers only


• FDA may take this action if agency staff cannot gain access to facility to inspect or product is
not safe or effective or is misbranded
• Manufacturer cannot engage in interstate commerce after license is revoked

Judicial actions Seizure • Court order needed


• FDA may seek seizure to condemn violative products and remove from distribution
• FDA and U.S. Marshals Service take possession and may dispose of product

Injunction • Civil process to stop or prevent violation of the law (e.g., to stop producing a product and to
comply with particular regulations)
• Manufacturer can contest injunction in court
• Court may impose penalties
• Applies to both licensed and unlicensed manufacturers

Prosecution • FDA will refer cases to Department of Justice for possible prosecution
• Applies to both licensed and unlicensed manufacturers
• FDA will seek to prosecute for criminal conduct, including fraud or practices that lead to unsafe
products or a pattern of violations
6888_Ch27_574-589 29/10/18 5:05 PM Page 584

584 PART V Quality Management and Compliance

Under 21 CFR 211.22, the quality control unit has the re- Additional Requirements of Biological
sponsibility and authority, at a minimum, to: Product Manufacturing
• Approve and reject components, containers (e.g., collec-
All biological product manufacturers must comply with all
tion bags), supplies, and drug products.
applicable biologics regulations in the CFR. This section lists
• Review production records for accuracy and completeness.
some of the additional elements of biologics regulation.
• Investigate errors and deviations.
• Review and approve standard operating procedures.
Alternative Procedures (“Variances”)
The responsibilities and procedures applicable to the
quality control unit must be in writing. The CGMP regula- The regulations allow for blood and blood component man-
tions do not prescribe how each manufacturer should de- ufacturers to perform procedures that vary from what is re-
velop its quality control unit or program but provide quired in some sections of the CFR with the approval of
minimum requirements that must be adhered to when de- CBER’s Director (21 CFR 640.120). Specifically, manufactur-
veloping the program. This is to allow flexibility so that each ers may request from CBER a variance to any requirement
manufacturer can develop a program that will work best in in subchapter F of the CFR (Parts 600 to 680). This provi-
its own manufacturing environment. The FDA has pub- sion provides flexibility to accommodate changes in tech-
lished a guidance document to assist blood and blood com- nology and unanticipated circumstances that may warrant a
ponent manufacturers in developing a quality assurance departure from regulations.20
program.18 The FDA generally recommends that the quality The alternative procedures addressed by 21 CFR 640.120
control functions be separate from the manufacturing oper- are also known as “variances” and fall into two categories:
ations because both have their own goals that may some- an alternative procedure that is incorporated into blood es-
times conflict. tablishment SOPs and an exception that is a singular event
related to a specific emergency situation. Manufacturers
Quality Reviews wishing to implement a procedure that varies from the reg-
ulations in these specific sections of the CFR must submit a
The CGMP regulations require manufacturers to retain man- written request to CBER. A request for an exception or al-
ufacturing and blood collection records, such as donor se- ternate procedure from a licensed blood establishment must
lection records (21 CFR 211.180 and 606.160). At least be submitted as a PAS supplement and contain sufficient in-
annually, the data from written records and quality standards formation to show that the safety, purity, or potency of the
of each product must be reviewed to determine the need for product manufactured at a variance from the regulations will
changes in product specifications or manufacturing or con- not be adversely affected (640.120(a)(1)). Manufacturers
trol procedures (21 CFR 211.180(e)). The FDA’s Guideline cannot implement an alternate procedure or exception until
for Quality Assurance in Blood Establishments18 identifies they have received approval from CBER.
the systems in a blood and blood component manufacturing Manufacturers may make an oral request for an exception
operation and the critical control points that should be re- if there are difficult circumstances and a written request is
viewed. The quality control unit or other assigned individu- not feasible. The manufacturers must follow up the oral re-
als are responsible for the quality activities, including the quest by submitting a written request within 5 working days
annual reviews. (21 CFR 640.120(a)(2)). The FDA will follow up with a
The review should identify trends that could lead to the written approval (21 CFR 640.120(c)).
release of an unsuitable product. The quality control unit Examples of alternative procedures and exceptions that
must investigate any unexplained discrepancy or the failure the FDA has approved are posted on the CBER website at
of a lot or unit to meet any of its specifications (21 CFR www.fda.gov/BiologicsBloodVaccines/BloodBloodProducts/
606.100(c)). The FDA expects the outcome of such investi- RegulationoftheBloodSupply/ExceptionsandAlternative
gations will be the development and implementation of cor- Procedures/default.htm.
rective and preventive actions. Implementing corrective and The CBER Director may issue a notice of an alternative
preventive actions could result in changes to product speci- procedure or exception regarding blood, blood components,
fications or manufacturing and control procedures. The or blood products if a variance is necessary to assure that
quality control unit must monitor these changes to ensure these products will be available in a specified location or lo-
they adequately address the error and will result in the man- cations in response to an urgent and immediate public health
ufacture of a quality product. The review, investigation, and need for blood, blood components, or blood products or to
corrective actions must be documented. The quality control provide for appropriate donor screening and testing (21 CFR
unit should have the authority to stop production if it deter- 640.120(b)).
mines the quality of the product is being adversely affected.
The quality control unit should share the results of the re- Contract Manufacturing
view with the management of the manufacturing operations
in order to affect the necessary changes to correct and pre- Under the current regulations, one manufacturer may em-
vent ongoing problems. Records of the reviews may be ploy the services of a contract manufacturer to perform some
requested and reviewed during an FDA inspection.19 or all of the product-manufacturing steps.21 The contractor
6888_Ch27_574-589 29/10/18 5:05 PM Page 585

Chapter 27 Transfusion Safety and Federal Regulatory Requirements 585

is not under the direct control of the original biological prod- CBER as soon as possible but no later than 45 calendar days
uct manufacturer, but the original manufacturer must ensure from the date the event was discovered. Manufacturers may
that the manufacturing steps performed by the contractor submit a hard copy report or report the BPDR electronically
conform with FDA regulations. Contract manufacturers at www.fda.gov/BiologicsBloodVaccines/SafetyAvailability/
must be registered with the FDA because they are performing ReportaProblem/BiologicalProductDeviations/default.htm.
a manufacturing step on a regulated product (21 CFR For additional information on BPDRs, contact CBER at
607.20(a)). In addition, the contractor must follow CGMP www.accessdata.fda.gov/scripts/email/cber/bpdrcontact.cfm
and all applicable regulatory product standards. Contractors or refer to the BPDR website above.
should notify the original manufacturer of any changes in In fiscal year (FY) 2015, CBER received 45,773 BPDRs
their operations because these changes could affect the from blood and blood component manufacturers.23 The ma-
safety, purity, or potency of the product. The original manu- jority of the reports (71.5%) involved postdonation informa-
facturer’s quality control unit is responsible for approving or tion (Table 27–5), which is information received from
rejecting drug products manufactured, processed, packed, donors following the collection of the product. On occasion,
or held under contract by another company (21 CFR the products from these donors are still at the blood estab-
211.22). Examples of contract manufacturing include an lishment, but often they have already been distributed. The
outside testing laboratory performing RTTI testing on dona- manufacturer should take appropriate action after reviewing
tions and a blood center irradiating blood components for a the impact of the donor’s information on product safety. The
hospital transfusion service. manufacturer may need to identify each affected product col-
lected from a donor who is subsequently determined to be
Short Supply Arrangements unsuitable and recall these products.

A short supply arrangement is an example of a cooperative Product Recalls


manufacturing arrangement. Short supply was introduced in
1948, and the provisions governing short supply are found The FDA may order the recall of a biological product that pre-
in the CFR (21 CFR 601.22).21 The short supply regulations sents an imminent or substantial hazard to the public health
allow a licensed biological product manufacturer to arrange or a medical device if there is a reasonable probability that a
for the partial manufacture of a biological product at an un- device intended for human use would cause serious, adverse
licensed facility under controlled conditions when the prod- health consequences or death. However, the FDA rarely has
uct is considered in short supply. Specifically, it allows an to resort to these authorities and instead relies on the volun-
unlicensed product, such as recovered plasma, to be shipped tary recall policy contained in 21 CFR Part 7. Voluntary prod-
in interstate commerce and used to manufacture a licensed uct recalls are the manufacturer’s removal or correction of a
final product. Short supply arrangements are most typically marketed product that FDA considers to be in violation of
established when a blood or blood component manufacturer the laws (21 CFR 7.3 and 7.40). Voluntary recalls can be ini-
wants to ship recovered plasma. The short supply arrange- tiated by the manufacturer or requested by the FDA if the
ment must be in writing and is between the blood establish- product represents a risk of illness or injury or if there is gross
ment and the licensed final product manufacturer. The
written arrangement specifies the necessary manufacturing
procedures and labeling. Table 27–5 Biological Product Deviations
Brokers frequently act as intermediaries between suppli- Reported to the CBER in FY
ers and licensed final product manufacturers. Selling recov- 201523
ered plasma to a broker does not relieve the supplier of the
Manufacturing System % of Total (45,773)
responsibility of obtaining a short supply arrangement. In
addition, brokers who take physical possession of recovered Postdonation information 71.5
plasma must register with the FDA. Quality control and distribution 9.7

Biological Product Deviation Reporting Miscellaneous 8.0


Labeling 3.6
The regulations require all biological product manufacturers,
including licensed and unlicensed manufacturers of blood and Donor screening 2.9
blood components and transfusion services, to report to the
Collection 2.0
FDA any event that resulted in the distribution of an unsuit-
able biological product. Specifically, a manufacturer of blood Routine testing (ABO, Rh, 1.7
and blood components must report to the FDA deviations antibody screen)
from CGMP or unexpected events in a manufacturing opera- Component preparation 0.4
tion that may adversely affect the safety, purity, or potency
of the product that was distributed and is no longer under Donor deferral 0.1
the manufacturer’s control (21 CFR 606.171).22 The biolog- Viral (RTTI) testing 0.0
ical product deviation report (BPDR) must be submitted to
6888_Ch27_574-589 29/10/18 5:05 PM Page 586

586 PART V Quality Management and Compliance

consumer deception, the firm has not initiated a recall, and transfusion-related fatalities26 (Table 27–6). Additional in-
agency action is necessary to protect the public health. formation on reporting fatalities can be found on the CBER
The manufacturer is responsible for developing a recall website at www.fda.gov/BiologicsBloodVaccines/Safety
strategy and conducting the recall. The manufacturer is also Availability/ReportaProblem/TransfusionDonationFatalities/
responsible for contacting customers with information iden- default.htm.
tifying the affected products and prescribing appropriate ac-
tions to take (21 CFR 7.49). The FDA will monitor the recall, Medical Devices
including assessing the manufacturer’s efforts to notify the
appropriate parties. A recall is classified as completed or CBER regulates medical devices used in the collection, pro-
closed when all reasonable efforts have been made to remove cessing, testing, manufacturing, and administration of blood,
or to correct the product. The FDA may seize the product if blood components, human cells, tissues, and cellular- and
it determines that the recall is ineffective. tissue-based products (HCT/Ps).27 Certain products meeting
Recalls of blood and blood components are different from the definition of a medical device that are used in blood col-
most drug product recalls because the blood or blood com- lection or processing are subject to license under Section 351
ponent may already have been transfused. In these cases, of the PHS Act. Other medical devices used in blood collec-
there can be no physical recall or return of the unit. When tion or processing are regulated under the device authorities
blood or a blood component has already been transfused, a of the FD&C Act. Licensed biological products that meet the
recall is really a notification that the product failed to meet definition of a device are reviewed in the same manner as
standards for safety, purity, and potency. other licensed biological products. Examples of such licensed
Recalls are accompanied by a health hazard evaluation of biological products are blood grouping reagents and tests
the recalled product by FDA medical staff and scientists to used to screen donations for RTTIs. Examples of devices used
determine if the product caused or could cause harm to a re- in blood collection or processing regulated by CBER under
cipient (21 CFR 7.41). The recall will be classified as a class the FD&C Act include pathogen reduction devices, blood
I, II, or III recall based on the potential for a product to cause warmers, BECS, leukocyte-reduction filters, and apheresis
serious health problems as outlined below: collection instruments. Manufacturers of licensed biological
products that are devices must register with the FDA and
• Class I: Situation in which there is a reasonable probability list their products under 21 CFR 607, and manufacturers of
that the use of, or exposure to, a violative product will FDA-approved or -cleared biological devices must register
cause serious adverse health consequences or death. with the FDA and list their products under 21 CFR 807. Both
• Class II: Situation in which there is a remote possibility that types of manufacturers must follow device CGMP, known as
use of, or exposure to a violative product may cause tem- the Quality Systems Regulations (21 CFR 820). Additional
porary or medically reversible adverse health consequences. information about biological devices can be found on
• Class III: Situation in which use of, or exposure to, a violative the CBER website at www.fda.gov/BiologicsBloodVaccines/
product is not likely to cause adverse health consequences. DevelopmentApprovalProcess/510kProcess/default.htm.
In FY 2016, CBER classified 575 recalls.24 Most of the recalls Additional information about the FDA and regulatory is-
involved blood and blood components that were either not sues related to blood and blood components can be found at
likely to cause an adverse reaction or would at most cause only www.fda.gov/BiologicsBloodVaccines/default.htm.
temporary health problems. Additional information about bi-
ological product recalls can be found on the CBER website at
www.fda.gov/BiologicsBloodVaccines/SafetyAvailability/
Table 27–6 Transfusion-Related Fatalities
Recalls/default.htm. Reported to the CBER in FY
201526
Fatality Reporting Complication % of Total (37)
Blood and blood component manufacturers must notify Transfusion-related acute lung injury 32
CBER of any transfusion-related fatalities or donation-related (TRALI)
deaths as soon as possible. The regulations require the blood Transfusion-associated circulatory overload 30
and blood component manufacturer to submit a written re- (TACO)
port to CBER within 7 days after the fatality (21 CFR
Microbial contamination 14
606.170(b)).25 The 7-day written report should describe all
information related to the fatality. Collection facilities are re- Hemolytic transfusion reaction (non-ABO) 11
quired to report donation-related deaths, and the facility that
Hemolytic transfusion reaction (ABO) 5
performed the compatibility tests must report transfusion-
related fatalities. Anaphylaxis 5
FDA investigators may visit the reporting facility to follow
Hypotensive reaction 3
up on the fatality reports. In FY 2015, transfusion-related
acute lung injury (TRALI) and transfusion-associated circu- Note: The FY15 transfusion-related fatalities denotes an imputability of definite/certain,
latory overload (TACO) were the most common causes of probable/likely, or possible.
6888_Ch27_574-589 29/10/18 5:05 PM Page 587

Chapter 27 Transfusion Safety and Federal Regulatory Requirements 587

SUMMARY CHART
 The FDA promulgates and enforces regulations to en- community blood banks, component preparation fa-
sure the safety and efficacy of biological products, cilities, hospital blood banks, plasmapheresis centers,
drugs, and devices, which include blood and blood product testing laboratories, storage and distribution
components, blood collection supplies and instru- centers, and brokers who take possession and manip-
ments, blood grouping reagents, and donor screening ulate and/or relabel the product.
tests for RTTIs.  The PHS Act requires an approved biologics license to
 A drug or device is misbranded if its labeling is false be in effect before biological products are distributed
or misleading. in interstate commerce.
 A drug is considered adulterated if the methods used  The FDA may suspend or revoke a biologics license if
to manufacture it do not conform with CGMP to en- there are continued violations that result in harm to a
sure that the drug is safe and has the identity, strength, blood or plasma donor or blood or blood component
quality, and purity characteristics that it is represented recipient.
to possess.  CGMP is the minimum current good manufacturing
 The ORA is responsible for performing routine FDA practice for methods to be used in, and the facilities or
inspections and other surveillance activities. controls to be used for, the manufacture, processing,
 The FDA’s five overlapping layers of safety of blood and packing, or holding of a drug to ensure that the drug
blood components include determining donor eligibil- meets the requirements of the FD&C Act as to safety
ity, testing the donation for certain RTTIs, checking and has the identity and strength and meets the quality
donor deferral registries, quarantining unsuitable and purity characteristics that it purports or is repre-
products, and investigating and correcting manufac- sented to possess.
turing problems.  CBER must approve alternative procedures or excep-
 Biological products are defined as any virus, therapeu- tions from the regulations before the products made at
tic serum, toxin, antitoxin, vaccine, blood, blood com- a variance from the regulations are distributed.
ponent or derivative, allergenic product, protein  Deviations in biological product manufacturing that
(except any chemically synthesized polypeptide), or affect the safety, purity, or potency of the products
analogous product or arsphenamine or derivative of must be reported to the FDA if the affected product
arsphenamine (or other trivalent organic arsenic com- was distributed.
pound) applicable to the prevention, treatment, or cure  A product recall is the removal or correction of a mar-
of diseases or conditions of human beings. keted product that the FDA considers to be in violation
 Statutes are the acts passed by Congress and signed by of the laws.
the president into law to establish the FDA and to  When a complication of blood collection or transfu-
grant the agency authority to fulfill its mission. The sion is confirmed to be fatal, the manufacturer must
Acts enforced by the FDA that are most relevant to notify CBER as soon as possible and provide a written
blood regulation are the FD&C Act and the PHS Act. report within 7 days of the fatality.
 Blood establishments required to register with the
FDA under the FD&C Act include collection centers,

3. What was the important tragedy that led to the regulation


Review Questions
of biological products?
1. Which of the following is responsible for overseeing the a. Three patients contracted hepatitis C following trans-
safety of the nation’s blood supply? fusion
b. A child died following transfusion of hemolyzed red
a. Joint Commission on Accreditation of Healthcare
blood cells
Organizations
c. A group O patient received group A blood
b. Food and Drug Administration
d. Thirteen children died after receiving diphtheria anti-
c. College of American Pathologists (CAP)
toxin contaminated with tetanus
d. Occupational Safety and Health Administration
4. What is required to ship blood and blood components
2. Where are the regulations for blood and blood compo-
across state lines (interstate)?
nents published?
a. AABB accreditation
a. The AABB Technical Manual
b. State license
b. CAP inspection checklist
c. CMS certification
c. The Code of Federal Regulations
d. Approved biologics license application
d. State Inspectional Guidance Documents
6888_Ch27_574-589 29/10/18 5:05 PM Page 588

588 PART V Quality Management and Compliance

5. Which of the following government organizations in- 2. Commemorating 100 years of biologics regulation. Science and
spect blood and blood component manufacturers? the regulation of biological products from a rich history to a
challenging future. Center for Biologics Evaluation and Re-
a. CBER search, FDA, September 2002. Available from www.fda.gov/
b. ORA AboutFDA/History/FOrgsHistory/HistoryofFDAsCentersand
c. CMS Offices/ucm2017807.htm
d. All of the above 3. The Coroner’s Verdict in the St. Louis Tetanus Cases. 1901 New
York Medical Journal 74; Special Article: Fatal results from
6. Which of the following is true about CGMP? diphtheria antitoxin. Minor comments: Tetanus from antidiph-
theria serum. JAMA. 1901;37:1255, 1260.
a. CGMP is the minimum current practice for methods
4. Public Health Service Act. Regulation of Biological Products.
and facilities used to manufacture a drug to ensure Title 42 U.S. Code, Chapter 6A, Part F, Sect. 262 (42USC262).
that it is safe, pure, and potent Available from https://www.gpo.gov/fdsys/granule/USCODE-
b. The FDA will approve a biologics license application 2010-title42/USCODE-2010-title42-chap6A-subchapII-partF-
if the manufacturer does not have a quality control subpart1-sec262
5. Federal Food, Drug and Cosmetic Act. Drugs and Devices.
plan Chapter V, Subchapter A. Available from: www.fda.gov/
c. The quality control unit must perform all the quality RegulatoryInformation/LawsEnforcedbyFDA/FederalFood
functions DrugandCosmeticActFDCAct/default.htm
d. Blood and blood components do not have to be in 6. Food and Drug Administration [Internet]. History of the FDA.
compliance with the drug CGMP regulations Available from: www.fda.gov/aboutfda/history/default.htm
7. Food and Drug Administration [Internet]. Keeping blood
7. A donor calls the blood bank and informs them that transfusions safe: FDA’s multi-layered protections for donated
within a year prior to his donation, he had intimate con- blood. Available from: www.fda.gov/BiologicsBloodVaccines/
SafetyAvailability/BloodSafety/ucm095522.htm.
tact with a person diagnosed with HIV. Which of the fol- 8. Food and Drug Administration [Internet]. Guidance for Indus-
lowing actions is not required by the FDA? try: For the submission of chemistry, manufacturing and con-
a. Identify and quarantine all blood and blood compo- trols and establishment description information for human
nents produced from the blood supplied by the blood and blood components intended for transfusion or for
further manufacture and for the completion of the Form FDA
donor 356h “Application to Market a New Drug, Biologic or an
b. Report the biological product deviation to CBER if Antibiotic Drug for Human Use.” May 1999. Available from:
the product has been distributed www.fda.gov/BiologicsBloodVaccines/GuidanceCompliance
c. Enter the donor in a record so that he can be identi- RegulatoryInformation/Guidances/Blood/ucm077087.htm.
fied and his product not be distributed while he is 9. Changes to an Approved Application. Federal Register. 62 FR
39901, July 24, 1997.
deferred 10. Food and Drug Administration [Internet]. Guidance for indus-
d. Notify the AABB try; Changes to an approved application: biological products:
human blood and blood components intended for transfusion
8. A patient dies following transfusion of ABO-incompatible or for further manufacture. November 2014. Available
blood. To whom should this event be reported? from: www.fda.gov/downloads/BiologicsBloodVaccines/
a. The Center for Biologics Evaluation and Research GuidanceComplianceRegulatoryInformation/Guidances/Blood/
b. Center for Medicare and Medicaid Services UCM354668.pdf
11. Food and Drug Administration [Internet]. Memorandum of
c. The AABB central office
understanding between the Health Care Financing Adminis-
d. The Occupational Safety and Health Administration tration and the Food and Drug Administration. FDA 225-80-
4000. June 6, 1983. Available from: www.fda.gov/AboutFDA/
9. Which federal agency has the responsibility to routinely PartnershipsCollaborations/MemorandaofUnderstandingMOUs/
inspect an unregistered transfusion service that does not DomesticMOUs/ucm116313.htm.
collect blood? 12. Food and Drug Administration [Internet]. FDA Office Of Reg-
a. Food and Drug Administration ulatory Affairs. Investigations Operations Manual. 2017. Avail-
able from: www.fda.gov/ICECI/Inspections/IOM/default.htm.
b. Centers for Medicare and Medicaid Services
13. Food and Drug Administration [Internet]. FDA Office of Reg-
c. Occupational Safety and Health Administration ulatory Affairs.. FDA Compliance Program Guidance Manual—
d. State health department 7342.001: Inspection of licensed and unlicensed blood banks,
brokers, reference laboratories, and contractors. Implementa-
10. Which of the following is not one of the FDA layers of tion date: June 1, 2016. Available from: www.fda.gov/Biologics
safety? BloodVaccines/GuidanceComplianceRegulatoryInformation/
a. Donor screening ComplianceActivities/Enforcement/CompliancePrograms/
default.htm.
b. Biologics License Application
14. Food and Drug Administration [Internet]. FDA Office of Reg-
c. Investigation of manufacturing problems ulatory Affairs. Inspections, Compliance, Enforcement, and
d. Testing for relevant transfusion-transmitted infections Criminal Investigations. Chapter 2. Biologics. FDA Compliance
Policy Guides. Available from: www.fda.gov/ICECI/Compliance
Manuals/CompliancePolicyGuidanceManual/ucm116336.htm.
References 15. Food and Drug Administration [Internet]. FDA Office of Reg-
1. Code of Federal Regulations, Title 21, FDA.: U.S. Government ulatory Affairs. FDA Regulatory Procedures Manual. February
Printing Office, Washington, DC, April 1, 2017. Available from 2014. Available from: www.fda.gov/ICECI/ComplianceManuals/
www.gpoaccess.gov/CFR/ RegulatoryProceduresManual/default.htm.
6888_Ch27_574-589 29/10/18 5:05 PM Page 589

Chapter 27 Transfusion Safety and Federal Regulatory Requirements 589

16. Drugs: current good manufacturing practice in manufacture, Establishments. October 2006. Available from: www.fda.gov/
processing, packing, or holding. Federal Register. 28 FR 6385, BiologicsBloodVaccines/GuidanceComplianceRegulatory
June 20, 1963. Information/Guidances/Blood/ucm073455.htm.
17. Human blood and blood products: collection, processing and 23. Food and Drug Administration [Internet]. Biological product
storage. Federal Register. 40 FR 53531, November 18, 1975. and HCT/P deviation reports – annual summary for fiscal year
18. Food and Drug Administration [Internet]. FDA Guideline for 2015. Available from: www.fda.gov/BiologicsBloodVaccines/
Quality Assurance in Blood Establishments. July 11, 1995. SafetyAvailability/ReportaProblem/BiologicalProductDeviations/
Available from: www.fda.gov/BiologicsBloodVaccines/Guidance ucm129757.htm.
ComplianceRegulatoryInformation/Guidances/Blood/default.htm. 24. Food and Drug Administration [Internet]. Enforcement activ-
19. Food and Drug Administration [Internet]. FDA Office of ity. Available from: www.fda.gov/iceci/enforcementactions/
Regulatory Affairs. Inspections, Compliance, Enforcement, ucm247813.htm.
and Criminal Investigations. FDA compliance policy guide, 25. Food and Drug Administration [Internet]. Guidance for Indus-
Sec. 130.300, FDA access to results of quality assurance pro- try. Notifying FDA of Fatalities Related to Blood Collection or
gram audits and inspections. June 2, 2007. Available from: www Transfusion. September 2003. Available from: www.fda.gov/
.fda.gov/ICECI/ComplianceManuals/CompliancePolicyGuidance downloads/BiologicsBloodVaccines/GuidanceCompliance
Manual/ucm073841.htm. RegulatoryInformation/Guidances/Blood/UCM062897.pdf
20. Blood and blood products: amendment to allow for alternative 26. Food and Drug Administration [Internet]. Fatalities reported
procedures; removal of a labeling requirement. Federal Register. to FDA following blood collection and transfusion: annual
55 FR 10420, March 21, 1990. summary for fiscal year 2015. Available from: www.fda.gov/
21. Food and Drug Administration [Internet]. Guidance for Industry: BiologicsBloodVaccines/SafetyAvailability/ReportaProblem/
Cooperative Manufacturing Arrangements for Licensed Biologics. TransfusionDonationFatalities/default.htm
November 2008. Available from: www.fda.gov/downloads/ 27. Food and Drug Administration [Internet]. CBER’s annual
biologicsbloodvaccines/guidancecomplianceregulatoryinformation/ report for fiscal year 2016. Available from:www.fda.gov/
guidances/general/ucm069908.pdf AboutFDA/CentersOffices/OfficeofMedicalProductsand
22. Food and Drug Administration [Internet]. Guidance for Indus- Tobacco/CBER/ucm535743.htm.
try. Biological Product Deviation Reporting for Blood and Plasma
6888_Ch28_590-606 29/10/18 5:07 PM Page 590

Chapter 28
Laboratory Information Systems
in the Blood Bank
Ann Tiehen, MT(ASCP)SBB, Phyllis Straff, MS(ISM), MT(ASCP),
and Gale Suwe, BS, MT(ASCP)SBB

Introduction Transfusing Facility Summary Chart


Types of Information Systems General System Applications Review Questions
System Components Regulatory and Accreditation Requirements References
Hardware System Management
Software Standard Operating Procedures
People Validation of Software
Blood Bank Software Applications Virtual Blood Bank
Donation Facility Future Trends

OBJECTIVES
1. Explain the purpose of an information system in a blood bank.
2. Correlate the hardware components of a blood bank information system with their functions.
3. Describe a blood bank information system hardware configuration.
4. Describe the functions of the software components of a blood bank information system.
5. List the responsibilities of operating and maintaining a blood bank information system.
6. Identify regulatory and accrediting agency requirements pertaining to blood bank information systems.
7. Apply the functions of blood bank software applications to blood bank processes.
8. Explain the purpose of a truth table.
9. Construct a truth table for a routine blood bank test.
10. Identify the standard operating procedures (SOPs) needed to manage a blood bank information system.
11. Describe and justify the kinds of testing that should be included in the validation of a blood bank information system.
12. Interpret the results of a software validation test.
13. Identify the times at which validation testing must be performed.
14. Define control function and differentiate between process control and decision support.
15. Evaluate a validation test plan and identify its component parts.

Introduction computerized information systems to assist in the handling


of this important information.
Information processing plays a vital role in blood banking Regulatory and standard-setting agencies have put in-
practice. The proper management of information related to creasing pressure on blood banks to store information in a
blood donors, blood components, and patients receiving safe manner and retrieve it in a timely fashion. It must be
transfusions is crucial to ensuring the safety and traceability possible to track a blood component from the time of its
of blood products. Blood bank personnel rely heavily on donation, through all of the processing steps, and to the

590
6888_Ch28_590-606 29/10/18 5:07 PM Page 591

Chapter 28 Laboratory Information Systems in the Blood Bank 591

patient who receives it. It is also essential to be able to per- System Components
form that trace in reverse order, from the recipient back to
the donor. A computer system includes three major components: hard-
Before computerized information systems were available, ware, software, and people. Hardware components are the
all blood bank data were stored on paper, or “hard-copy,” physical pieces of equipment. Software is a set of instructions
records requiring labor-intensive and sometimes error-prone written in computer language that tells the computer how to
procedures to retrieve it. As blood bank information systems operate and manipulate the data. The people interface with the
have become more sophisticated, they have made recovering hardware to enter the data that are manipulated by the software.
the information much more efficient and have offered the in- Just as there are many different kinds of people who enter data,
dustry a great variety of process controls to reduce errors. so there are many different kinds of hardware and software.
Several acronyms unique to the specialty of information sys-
tems will be used throughout this chapter and are listed in Hardware
Table 28–1.
Most of the hardware components of a computer system are
Types of Information Systems readily identifiable because they can be seen and touched,
although some are hidden inside a case or a central system
Computerized blood bank information systems are available unit. Each piece of hardware performs a specific function in
in many different configurations, but they comprise three the handling of information. The three main functions per-
general categories: formed by hardware components are processing, input and
output, and storage.
1. A highly complex system like that found in a community Hardware components include a central system unit,
blood center sometimes referred to as the box, and several different pe-
2. A small part of a complete clinical laboratory informa- ripheral devices that send or receive information through the
tion system (LIS) system unit. Peripheral devices include display terminals,
3. A stand-alone system, usually found in a hospital blood keyboards, bar code readers, scanners and wands, pointing
bank or transfusion service devices such as mice, printers, and modems. The hardware
The functionality of these systems varies from category components and the way in which they are connected to
to category and from vendor to vendor within each category. each other is the system configuration. Figure 28–1 illus-
Some systems may act merely as record keepers, whereas trates one such configuration.
others have the ability to use the information in such a way
as to prevent errors in donor acceptance, blood component Processing Hardware
release, or patient transfusion. For example, a basic system The central hardware component of a computer system is
used in a hospital transfusion service may simply allow the the central processing unit (CPU), which is an electric
entry of an ABO group and Rh type in a patient’s record with circuit or silicon “chip” that processes information. The
subsequent generation of a charge for the test. A more com- CPU, also called a processor, is the core of the machine. It
plex system in a hospital blood bank can record the donation controls the interpretation and execution of instructions
of an autologous unit in the donor room and then make the
transfusion service technologist aware of its presence when
User terminals
the patient is admitted for a type-and-screen test at the time
of surgery.

Printers Barcode
Table 28–1 Common Information System reader

Acronyms
Tape drives
Acronym Definition Disk drives
CPU Central processing unit
HIS Hospital information system Central processing unit
CPU Modem
LIS Laboratory information system Interface Interface
PC Personal computer
RAM Random access memory
ROM Read-only memory Testing
instruments HIS/LIS
SOP Standard operating procedure
Figure 28–1. Example of a blood bank information hardware configuration.
6888_Ch28_590-606 29/10/18 5:07 PM Page 592

592 PART V Quality Management and Compliance

provided by the software. Other pieces of processing hard- bar coding of blood products has been accepted worldwide
ware that exist in the system unit are the ROM and RAM to expedite the accurate input, transfer, and use of blood
chips. Read-only memory (ROM) contains the “start-up” in- products across the globe.1 Scanning the bar codes with
structions for the computer. Random access memory (RAM) an optical or laser device allows efficient entry of the blood
is an array of chips in which data are temporarily entered component data.
while they are being processed. When a computer is turned The most common output device is the monitor, which
on, these chips interact with each other and with the oper- displays information as it is typed on the keyboard. Taken
ating system software to prepare the computer to accept data together, the monitor and the keyboard make up the user
and instructions from its users. terminal. The monitor may also display historical informa-
tion at the user’s request.
Input and Output Devices Another commonly used output device is the printer,
Blood bank systems do not usually utilize voice recognition, which provides hard-copy output on paper. Printers can pro-
thus commands and data must be entered in a format that the duce labels, donor registration records, compatibility tags,
computer can understand. This action requires tools that are management reports, patient chart reports, and donor corre-
connected to the system’s CPU, such as keyboards, pointing spondence. The kind of printer chosen for any of these appli-
devices, bar code readers, and testing instruments. Keyboards cations depends on the quality of print desired. For example,
are used to type instructions that tell the computer what to a bar code label requires a high-resolution printing that bar
do or to enter data such as donor demographic information code readers can accurately interpret. Management reports,
or patient test results. Many systems also accept barcoded in- which are usually used internally, can be of lower quality print.
formation from sources such as blood component labels or An example of a combined input and output device is a
test tubes. Blood component labels contain many pieces of modem. Modems were widely used in the past to allow com-
barcoded information, including the unit number, blood type, puter systems to communicate with each other via network
product type, facility identification number, and expiration or telephone lines. Today’s network technology utilizes stan-
date. Entry of this information by a bar code reader is much dardized TCP/IP Internet communication protocol for com-
more accurate than when done by manual entry methods. munication and data transfer over wired and wireless
Figure 28–2 shows a blood component label with this bar- networks such as the Internet, intranet (within a facility), or
coded information using ISBT 128 bar coding. This uniform VPNs (virtual private networks). Blood banks use this tech-
nology to connect remote facilities to the main computer sys-
tem so that all sites have access to the system’s databases.
This communication technology can also be found in blood
centers with several collection or testing locations or in a
system of hospital affiliates. Technological advances in com-
puter and networking capabilities create new opportunities
to help blood banks and transfusion services respond to the
demands of modern medicine. For example, Verlicchi et al
described the use of a computer system to implement elec-
tronic remote blood issue methods in remote sites for trans-
fusion support of surgical procedures.2 The technical support
staff of software vendors use secured network connections
(or could use modems) to investigate and solve system prob-
lems and to transfer files to the customer.

Information Storage Hardware


All computer systems have hardware that allows long-term
storage of data. This is sometimes referred to as memory but
should not be confused with RAM, which is only temporary
and active when the system is turned on. Long-term memory
is data saved on a medium from which it can later be re-
trieved, such as a hard disk. Most systems contain a hard
disk controlled by a hard drive. The disk is made of metal
coated with a magnetic film and contains software applica-
tions (programs) and user-entered data. The hard disk is
often contained in the main system unit, but it may also be
a peripheral device. The information on the disk can be ac-
cessed by entering commands, usually with a keyboard con-
nected to a display terminal. Other information storage
hardware may be used for archiving old information that has
Figure 28–2. Bar codes on unit label. been removed from the hard disk.
6888_Ch28_590-606 29/10/18 5:07 PM Page 593

Chapter 28 Laboratory Information Systems in the Blood Bank 593

Software These files, which look different in each blood bank, define
the terminology that the blood bank uses. When a new blood
Software tells the computer what to do with all of the infor- bank information system is installed, the static files must be
mation it has received. Minimally, every computer system defined before the system can be put into use. One of the
has two kinds of software—operating system software and most important functions of the static files is to provide a
application software. Some systems may also use interface “dictionary” of coded terms that the information system can
software, which allows the system to communicate with use to sort and organize the tremendous amount of data en-
other computer systems. tered into it. For example, two static files, one containing
codes for physicians and the other codes for blood products,
Application Software can allow the system to sort information regarding the
An application is software that has been designed to perform crossmatch-to-transfusion (C/T) ratio of each physician who
specific tasks. Personal computers (PCs) can be equipped with ordered blood within a particular time period.
application programs such as word processing, spreadsheets, Figure 28–3 shows how the parts of a database relate to
and databases. In a blood bank information system, the appli- each other. The database itself can be considered a file cabinet
cation software allows users to perform tasks that are specific containing folders or files that contain specific records. This
to blood bank operations. In a donor setting, some of these illustration is of a dynamic file containing patient records. A
tasks might include entering donor demographic information static file illustrated in this way—for example, for blood prod-
and test results, confirming blood component labeling, and uct codes—would list a separate file for each blood product
generating donor recruitment lists. In a transfusion service, code used in the facility. The associated records would include
the computer may help with tasks such as searching for a such information as the official name of the blood component,
blood component of a particular ABO group, entering blood its maximum allowable shelf life, and whether it contains red
component modification information, and issuing blood for blood cells (RBCs) and thus requires crossmatching.
transfusion. These tasks are distinctly connected to blood
banking and would be difficult, if not impossible, to perform Operating System Software
in an application not specifically designed for blood bank use. The tasks performed by the operating system are fairly invisi-
One of the most important functions performed by blood ble to users because this software works in the background. It
bank applications is maintaining a database of donors, blood is a set of instructions that controls the computer’s hardware,
components, and transfusion recipients. A database is an or- manipulates the application software, and coordinates the flow
ganized set of information divided into files and then further of data to and from disks and memory. When new data are en-
subdivided into records. Files exist in one of two ways: they tered into one of the application programs, it is the operating
may be static or dynamic. The dynamic files contain records system that places those data on the disk for storage; when a
related to a specific donor, patient, or blood component. request for those data is made through the application program,
These records are frequently changed as updated information the operating system retrieves them and sends them to the ap-
is added to them. For example, the status of a blood compo- plication software for display on a monitor or printed report.
nent can go from “quarantined” to “transfused,” with many
intermediate statuses during its shelf life. Interface Software
The static files contain information that is updated infre- Frequently, different information systems must be allowed
quently, such as the list of blood products used in the facility. to share data with each other to take full advantage of their

Figure 28–3. Relationship between data- Record


base components from general database files Record
to individual patient record items. Patient number
items
File
Patient name
Patient 12345
Database Admit date
Patient 12346
Patient file Doctor
Patient 12347
Unit file ABO type
Patient 12348
Test result file Rh type
Patient 12349
Doctor file
Etc.
Product code file

Antigen code file

Comment code file

Etc.
6888_Ch28_590-606 29/10/18 5:07 PM Page 594

594 PART V Quality Management and Compliance

functionality. Because different systems communicate in dif- entire system, but there should also be a designated blood
ferent computer languages, they need an interpreter that will bank system manager. In blood banks equipped with a
allow data to flow between the two systems in a controlled stand-alone blood bank computer, designated system man-
manner. The interpreter is another set of software called an agers may also have other supervisory, technical, or quality
interface. Interface software may be used to allow data to assurance duties.
flow between a hospital information system (HIS) and the Whatever the duties of the system manager, he or she also
blood bank system or between the LIS and the blood bank. oversees the maintenance of the system’s hardware and soft-
For example, an interface between the HIS and the blood ware. Some specific duties include adding or deleting items
bank information system can allow patient demographic in- from the static database files, assigning access codes to new
formation to flow from the HIS to the blood bank system and users, and implementing software upgrades from the vendor.
can allow test results and blood component information to In addition, the system manager will be required to investi-
flow from the blood bank system to the HIS, where they can gate problems encountered by the users and to report them
be displayed on terminals in patient care areas. to the system vendor.

People Blood Bank Software Applications


The human components of a blood bank information system The kinds of data that must be managed by a particular blood
are the users and at least one person designated as the system bank information system depend on the types of services pro-
manager. Users have access to the technical applications vided by the blood bank. Most community blood centers focus
needed to perform daily blood bank operations. System man- on donations and distribution of the donated blood compo-
agers require access to a wider range of applications, includ- nents to customer hospitals. In the hospital transfusion ser-
ing system maintenance functions. vice, the primary concern is the transfusion of patients.
However, a community blood center or a hospital blood bank
Users
with both a transfusion service laboratory and a donation
Entry of commands and data into the system is performed center must address issues of donor and patient management.
by the users. The success with which a blood bank computer Application programs exist for every task, from scheduling
system can be used depends not only on its ease of use but donors to assigning a transfused status to a blood component.
also on the training provided to its users. Most systems are Many of the typical blood bank information system applica-
equipped with both a “live” or production database and a tions currently available are discussed in this section.
“test” database. The static files in both databases are identi-
cal, but their dynamic files contain different information. Donation Facility
The production database contains real information, whereas
the test database consists of fictitious records. The test data- The applications utilized at a donor center enable the users
base offers the user an opportunity to practice the applica- to manage the flow of donors and blood products from donor
tions of the system (and to make mistakes) without recruitment to distribution of the final products to hospital
corrupting the database containing actual donor and patient customers.
information. Users should be trained in every application
they will be expected to use. Donor Management
In addition, user security levels should be assigned that The system should allow the capture of donor demographic
grant the user access to information necessary for perform- information necessary for definitive donor identification, tele-
ing their jobs. The security levels will vary depending on the phone contact for recruitment, and notification in the event
nature of the job performed. For example, a donor technol- that abnormal laboratory testing results are obtained. In ad-
ogist may have the job of screening donors and collecting dition, the system should have a means for preventing a do-
blood, so functions such as donor screening, donor status, nation from a deferred individual or from one attempting to
assigning unique donor identification numbers, etc. should donate before the required waiting period between donations.
be easily accessible to those staff. Laboratory technologists
would have access to a different combination of functions Donor Registration. When a potential donor registers at a do-
which support performing tests, entering/retrieving results, nation facility, several pieces of demographic data must be
quarantining unsuitable components and so forth. provided, such as name, date of birth, phone number, and
mailing address. When this information is entered into the
System Managers donor database, the system can search for a previous dona-
The configuration and location of the information system tion record from the same donor. If none is found, a new
will determine the identity and quantity of people perform- donor record is created. If there is a record of a previous do-
ing this task. In a large community blood center, there may nation, the system can review the donor’s eligibility status.
be an entire staff dedicated to managing the system. In a This would include calculating the length of time elapsed
hospital blood bank or transfusion service that uses the since the last donation and examining the record for a de-
blood bank module of a complete clinical LIS, the labora- ferral because of medical history or disease testing results. If
tory system manager is likely to have responsibility for the the donor is ineligible because of an inadequate amount of
6888_Ch28_590-606 29/10/18 5:07 PM Page 595

Chapter 28 Laboratory Information Systems in the Blood Bank 595

time since the last donation or a previous deferral, the system into the system with the unique donation identification num-
can alert the registrar and prevent an unsuitable donation ber assigned at the time of donation, new blood product codes,
from occurring. time of preparation, and expiration dates. The system could
also capture critical production information such as equipment
Donation Data. Information regarding the donation event must used, production staff names, times of production, and other
be captured in the system. This data entry can include such required elements of traceability.
important information as the unique identification number
applied to the collection container, the type of donation Laboratory Testing. Samples of the donor’s blood that were col-
made (e.g., whole blood or apheresis), the collection time, lected at the time of donation and labeled with the same
and the occurrence of a donor reaction. If the donation is in- unique donation identification number are tested for ABO,
tended for a specific recipient, as in the case of an autologous Rh, atypical antibodies, and markers of transfusion-
or designated donation, data regarding the intended recipient transmitted diseases such as hepatitis and HIV. The results
can be entered. Comprehensive systems, such as those found of all these tests are entered into the system so that they are
in large donor centers utilize handheld devices to capture associated with the unique donation identification number
this information, but a smaller hospital-based donor center and with the donor. In larger community blood centers, the
may continue to use paper records on which information is results of testing performed on automated instruments can
manually recorded. If manual capture of information is used, be sent directly from the instrument to the system via an in-
then the donation data must be entered into the computer strument interface.
system as soon as possible after the donation is complete.
Quarantine and Release. Donated blood components must not
Donor Suitability. Most large blood centers use computers to be labeled and released until all requirements for screening
capture donor interview questions and the results of donor and testing are met. Computer systems must include safe-
health history screening tests (i.e., blood pressure, hemat- guards to prevent unprocessed or unsuitable components
ocrit, oral temperature). In many cases donors are given a from being released. Once the blood is collected, the system
handheld device to allow them the privacy to answer the should give the components a status to reflect that the re-
donor interview questions and obtain consent for the dona- quired processing is not complete. Some systems assign a sta-
tion. Additional manual evaluation, such as performing a tus of “unprocessed” to reflect that test results have not been
blood pressure or hemoglobin screening, can be performed completed or some other requirement has not been met, and
by the donor technologist and recorded into the device. De- the software must prevent those units from being labeled and
ferring conditions can be captured in real-time and added to released. A status of “quarantine” would be applied if the unit
the donor database. If the donor interview is performed man- was found unsuitable for reasons such as initial positive viral
ually, then the information can be entered into the system marker test results or errors in component production that
when completed. require further review. If further testing or review reveals that
the blood is suitable, then the status can be changed to allow
Donor Recruitment. The capture of donor demographic infor- the blood to be labeled and released. Computer systems
mation at registration allows the collecting facility to recruit should be selected that will foster quarantine/release func-
the donor from a system-generated list of eligible donors. tions and block those units deemed unsuitable for use.
Mailing labels, lists of telephone numbers, or e-mail ad-
dresses can be generated for recruitment efforts. Once labo- Label Application and Verification. After completion of component
ratory testing is associated with the donor’s record, lists of production and laboratory testing, the blood products are
donors meeting special needs can be printed. Such lists may labeled with all required elements including the ABO/Rh
include donors with a specific ABO group, other RBC anti- type and expiration date. This is a crucial step and must be
gen type, or cytomegalovirus (CMV) seronegativity. As with controlled stringently so that no unsuitable blood products
any system that captures identifying information, donor are released into the blood supply. After the label has been
records must be maintained in a confidential fashion and ac- applied, the unique donation identification number and
cess granted only to those individuals that need the infor- ABO/Rh type bar codes on the blood component label can
mation to perform their job tasks. be scanned into the system with a bar code reader. This al-
lows the system to perform a final check on donor suitability
Blood Component Management and blood type. If the component “passes” this verification,
Aspects of blood component management include component it can be placed in the available inventory. Alternatively,
preparation, laboratory testing, label application and verifica- some systems generate on demand labels as each individual
tion, and inventory management and product shipping. component’s identification number is scanned.

Component Production. After a whole blood unit has been col- Inventory Management and Product Shipping. The collection facil-
lected, it is usually delivered to a component-processing labo- ity staff members who are responsible for product distribu-
ratory where it is separated into different components, tion to customer hospitals must have access to the entire
including RBCs, fresh-frozen plasma, and platelets, and labeled inventory of available blood products. As transfusion ser-
appropriately. Data on the new components created are entered vices place requests for quantities and ABO/Rh types of blood
6888_Ch28_590-606 29/10/18 5:07 PM Page 596

596 PART V Quality Management and Compliance

components, the distribution staff can monitor and control components that have undergone special testing, such as
distribution to optimize blood use within the community. tests for antibodies to CMV or specific RBC antigens.
For example, blood products nearing their expiration date
can be sent to active transfusion services where there is a Component Status Tracking. After blood components are received
high probability of transfusion before expiration. As prod- in the transfusion service, they are assigned various statuses,
ucts are shipped to transfusing facilities, their status is up- ending with a final disposition of “transfused” or “discarded.”
dated in the system, along with the identification of the Each blood component record in the information system
facility to which they were shipped. It is critical that all blood should contain a complete status history. Figure 28–4 illus-
components are issued through the computer system before trates the various statuses attributed to blood components
shipping so that safeguards are in place to prevent unsuitable throughout their shelf life.
components from being released. It is also critical that the
Patient Management
inventory management system accurately reflect the cus-
tomers to where each blood component is shipped in the Blood bank information systems enhance patient safety by
event that there is a recall situation. providing users with the infrastructure to manage functions
such as patient identification, communication of orders and
Transfusing Facility results with a hospital information system (HIS), patient test-
ing, and blood component reservation and issue.
The applications utilized at a transfusing facility enable the
user to maintain accurate histories of patient and donor unit Patient Identification and Specimen Collection. A blood bank infor-
testing and ensure the release of compatible blood products mation system used by a facility issuing blood for transfusion
for transfusion. should have the capability of capturing patient demographic
information such as name and unique facility identification
Blood Component Management number. Recent developments in technology have made it
Aspects of component management unique to the transfu- possible to obtain this information from a barcoded patient
sion service include product receipt and entry, inventory identification wristband at the time of specimen collection.
management, blood component modification, and compo- Other essential data that the system should catalog include
nent status tracking. previous ABO/Rh type, transfusion history, previously iden-
tified clinically significant antibodies, and transfusion
Product Receipt and Entry. When the products are received at the instructions such as the need for irradiated or leukocyte-
transfusing facility, they are entered into the blood bank in- reduced cellular components. Specimen collection date and
formation system, where the entire inventory of blood com- time, as well as the identification of the individual who col-
ponents is maintained. If ABO/Rh confirmation is required, lected the sample must be captured to comply with AABB
as in the case of components containing RBCs, a status of standards.3
quarantine will be assigned by the system until confirmation
testing is completed. Components not requiring ABO/Rh
confirmation testing may be assigned an “available” status Unit received
from blood center
on entry into the system. Unit donated
Unit
Inventory Management. A computerized inventory makes it easy retyped
for blood bank staff members to monitor inventory levels so Unit discarded
that levels do not drop below predefined minimums. Most Quarantine Available or
Unit shipped to
systems sort the inventory by product code, ABO/Rh, and tested another facility
expiration date, and output it to a display monitor or printed
report. When a selection list of blood components is indexed
Crossmatched
in this way, the products that are closer to the end of their
shelf lives can be chosen for patients with a high likelihood
of transfusion.
Reserved
Blood Component Modification. Many components require mod- for patient
ification to meet the special transfusion needs of a particular
patient. Modifications include irradiation, leukocyte reduc-
tion, aliquoting (dividing), washing, and pooling. As these Issued
Returned
physical modifications are performed in the blood bank, the
steps associated with them are captured by the information
system, either by changing the product name or by adding
a special attribute to the component data. Some of these Transfused
steps may also require shortening the component’s shelf life,
and the system can calculate and apply the new expiration Figure 28–4. Various statuses of a blood component in a blood bank informa-
date and time, if needed. Attributes may also be added to tion system.
6888_Ch28_590-606 29/10/18 5:07 PM Page 597

Chapter 28 Laboratory Information Systems in the Blood Bank 597

Handheld devices are available that utilize bar code readers ABO Truth Table
to scan patient ID wristbands and generate on-demand Working Behind the Scenes
labels at the patient’s bedside. While not required by regula-
Anti-A Anti-B A1 cells B cells Interpretation
tory and accrediting agencies, these devices can improve pos-
itive patient identification and reduce errors in collection and O O + + O
labeling of specimens
+ O O + A
Order Entry. On receipt of a specimen for patient testing, the
system can retrieve previous test results and alert the tech-
nologist to the patient’s special transfusion requirements. For O + + O B
example, the record for a patient with a history of a clinically
+ + O O AB
significant antibody can alert the technologist to the need
for specific antigen-negative RBC components. Physicians’
orders for blood components can also be received from the
HIS through an interface or can be entered manually.

Patient Testing. Computer systems allow direct entry of test


results, thus replacing paper worksheets. If appropriate
“truth tables” have been set up in the system, the entered
test results can be compared with those of the truth tables
for accuracy. Truth tables define the combination of results
considered valid for a particular test. This can prevent the ABO Group Test Entry Function
release of invalid results such as nonmatching forward and NAME: Doe, Jane MEDICAL RECORD: 123456789
reverse types. Table 28–2 shows an example of a truth table.
Each row represents one combination of individual test re-
Anti-A Anti-B A1 cells B cells Interpretation Verification
sults and an interpretation, which the system will allow in
+ O O + A YES
its ABO test entry function.
In the case of the ABO test, there are only four valid result
combinations, and the system warns the user if any other
combination is entered. When the results are entered cor-
rectly and match one of the valid combinations in the truth
table, the results are accepted and verified. Other tests, such
as RhD and antibody screen, may also have to meet their
own truth table requirements. Figure 28–5 illustrates what
a user would see on the display monitor when a valid ABO
group is entered. Invisible to most users is the truth table,
which in this figure is highlighted, where the valid combi- Figure 28–5. Display monitor showing entry of a valid ABO group.
nation of results has been found to indicate the match with
the valid ABO results entered and displayed on the monitor.
Truth tables make their presence known when an invalid
combination of results is entered.
Figure 28–6 illustrates what a user might see when invalid
ABO results are entered. In this example, an incorrect result
has been entered for the reaction of the patient’s plasma or ABO Group Test Entry Function
serum with group B reagent RBCs. When the technologist NAME: Doe, Jane MEDICAL RECORD: 123456789
attempts to verify the results, the computer responds with a
warning message, indicating that it was not able to find a
Anti-A Anti-B A1 cells B cells Interpretation Verification
match in the truth table for this particular combination of
+ O O O

Table 28–2 ABO Truth Table


Anti-A Anti-B A1 Cells B Cells Interpretation ***ABO Group Does Not Match Truth Tables!***

0 0 + + O

+ 0 0 + A

0 + + 0 B

+ + 0 0 AB
Figure 28–6. Display monitor showing entry of an invalid ABO group.
6888_Ch28_590-606 29/10/18 5:07 PM Page 598

598 PART V Quality Management and Compliance

results. Truth tables must be validated with rigorous testing being issued. Verification of unit inspection before issue can
to verify that they are consistently accurate and functional.4 also be recorded in the system. When a product is issued,
Computer validation is discussed later in this chapter. the system updates its status in the inventory.
The most critical test performed in the blood bank is the
patient ABO/Rh type, and it is required that previous test re- General System Applications
sults be compared with current results. When this compari-
son, performed by the system, reveals a discrepancy, a In addition to the requirements specific to a collection or
warning alerts the technologist to the possibility of a misla- transfusion facility, there are general applications that are
beled specimen or incorrect test results. found in all blood bank information systems. These func-
tions assist with system security, quality assurance, and man-
Blood Component Reservation. The system can aid in selecting agement assistance.
blood components that will satisfy special transfusion needs
and that are compatible with the patient’s ABO/Rh. Selections Security
may be made by manual entry or bar code entry or from an It is critical that only authorized users have access to the in-
indexed selection list. If autologous or designated blood formation contained in a blood bank computer system.6 Ac-
components are available for a patient, the system can alert cess is obtained through the use of assigned user codes and
the user. As blood components are selected and either cross- passwords, and it is the users’ responsibility to keep their
matched or reserved for a patient, the system links those passwords confidential. A second level of security restricts
products to the patient record and prints compatibility tags. some users to some of the system’s functions. This is impor-
Computer crossmatch, also known as electronic cross- tant for two reasons: donor/patient confidentiality and pro-
match, allows electronic verification of recipient and donor tection of data from accidental or unauthorized destruction
compatibility and dispenses with the serologic crossmatch or modification. Sensitive information, such as test results
test. Both the Food and Drug Administration (FDA) and the for markers of transfusion-transmitted disease, should be
AABB publish requirements for implementing systems for available only to the medical director and selected supervi-
computer crossmatch 3,5 A patient is eligible for the com- sory personnel. The system manager has access to most or
puter crossmatch when his or her records indicate the fol- all of the functions, including those that allow modification
lowing criteria have been met: (1) there is no current or past of the static database files. Technical staff members can ac-
history of clinically significant antibodies, and (2) there are cess those functions that are specific to their jobs, such as
at least two concordant ABO grouping test results. test result entry, labeling, or component production. Clerical
In addition, the system must contain the donation iden- staff members may be restricted to inquiry functions that
tification number, component name, component ABO group allow them to answer questions from patients or physicians
and Rh type, the interpretation of the component ABO con- but not to enter or modify patient data. In addition to pro-
firmatory test, and recipient information, including ABO viding system security, user codes capture the identity of
group and Rh type. The system must process this informa- each person performing each step in the information system.
tion to alert the user when there are discrepancies between
donor unit labeling and blood group confirmatory test inter- Quality Assurance
pretation and when ABO incompatibilities exist between the Examples of quality assurance activities performed by a
recipient and donor unit. Finally, the system must require blood bank information system include control functions,
verification of correct entry of data before blood components the generation of corrected and amended results, and tools
are released. These stringent requirements for a computer that can be used to monitor blood product utilization.
crossmatch must be in place to prevent the issue of ABO-
incompatible components. Control Functions. One of the most useful features that a blood
bank information system can offer is assistance to users in
Result Reporting. Information regarding patient test results and ensuring safe transfusion. When blood bank personnel de-
any products linked to the patient should be accessible to pend on this assistance in making decisions at critical points
the patient’s caregivers. This may be in the form of printed in the selection of donors and release of blood products for
reports or monitor displays. The status of linked products transfusion, the system is said to be exerting “control func-
should be updatable—that is, as the products go from cross- tions.” Control functions can be placed in two general cate-
matched or reserved to issued to transfused or returned, the gories: process control and decision support. Process control
status should be reported in real time. functions are decisions made by the system without human
intervention. Decision support control functions occur when
Blood Component Issue. When blood components are requested the system displays information to the user, who then de-
for transfusion, the system can assist in issuing them appro- cides on the next course of action. Table 28–3 shows some
priately. For example, if both an autologous RBC and an al- examples of control functions.
logeneic RBC have been reserved for the same patient, it is
essential that the autologous unit be issued first. Many Corrected or Amended Results. Sometimes it is necessary to cor-
systems alert the issuer if he or she attempts to issue the rect or amend incorrect or incomplete information that was
allogeneic unit first. In addition, the system can warn if a entered into the system. This need arises when errors are
special transfusion need is not being met by the product made in testing or when testing is performed on a mislabeled
6888_Ch28_590-606 29/10/18 5:07 PM Page 599

Chapter 28 Laboratory Information Systems in the Blood Bank 599

Table 28–3 Examples of Control Functions


Application Process Control Decision Support
Donor management Preventing registration of permanently Warning of current ABO/Rh not matching
deferred donor previous results
Blood component date management: collection Preventing unit release if testing Calculation of component expiration
facility unacceptable
Blood component management: transfusing Preventing entry of donation identification Warning when entering an expiration date
facility number that already exists in the system that exceeds the possible shelf life of a
blood component
Patient management Preventing issue of outdated unit Warning of product not meeting special
transfusion needs

specimen so that results are associated with the wrong pa- reporting capabilities are designed into the system, they can
tient. When corrections must be made, both the original er- provide valuable information to assist in management deci-
roneous results and the corrected results must be clearly sions. Reports of the number of donors drawn and deferred
specified as such on monitor displays and printed reports. or blood products transfused and expired can help evaluate
Figure 28–7 illustrates an example of a corrected result. This productivity and future resource needs. In addition, patient
method of error correction alerts caregivers who may have or hospital billing can be done more accurately and consis-
made clinical decisions based on the incorrect results. The tently than with manual systems. Many computer systems
information system should make it impossible to simply have built-in mechanisms to provide data reports that assist
delete the erroneous result and substitute a corrected result. management in assessing transfusion practices, errors, and
inefficiency. Evaluating operations for reasons such as QA or
Blood Product Utilization Review. Blood banks and transfusion LEAN practices can be made much easier if the computer
services are required by several accrediting agencies to mon- system can provide reports with the pertinent data.8
itor the utilization of blood products within the facility. This
includes both blood ordering and transfusing practices of Regulatory and Accreditation
physicians. An information system can be of tremendous Requirements
help in sorting and reporting utilization data as well as mak-
ing work processes more efficient.7 Crossmatch to Transfu- As computer systems have assumed greater significance in
sion(C:T) ratios can be reported for individual physicians blood banking, regulatory oversight has also increased. In the
and for services such as surgery and obstetrics. late 1980s, the FDA recognized that unsuitable blood compo-
nents were being released into the blood supply because of in-
Management Assistance adequately controlled and validated computer systems. In 1988
Enormous amounts of different kinds of data can be entered and 1989, the FDA issued memoranda to registered blood
into the blood bank information system. When appropriate establishments to provide guidance on the use of computer
systems. In 1994 and 1995, memoranda were sent to blood es-
tablishment computer software manufacturers, advising them
that the FDA intended to regulate blood bank software as a
medical device under the Safe Medical Devices Act of 1990.
Any Hospital In 2002, the FDA issued an update of the 1997 draft ver-
Any City, State, Zip
sion of “General Principles of Software Validation; Final
Patient Result Report
Guidance for Industry and FDA Staff” which outlined a val-
NAME: Doe, Jane MEDICAL RECORD: 123456789 idation program in compliance with current good manufac-
turing practices.9 These regulatory initiatives have had
DATE TESTED: 01/01/XX
SAMPLE #: 12345 significant impact on the availability and implementation of
ABO: A blood bank information systems. Both software vendors and
RH: Negative INVALID RESULTS
users must follow prescribed guidelines in the introduction
DATE TESTED: 01/01/XX of new software. Vendors must perform thorough validation
SAMPLE #: 12345
ABO: A testing before making software available for purchase, and
RH: Positive CORRECTED RESULTS users must document validation procedures on site before
using such a system. A list of FDA 510(k) cleared Blood Es-
tablishment Computer Software (BECS) is available at
fda.gov.10 Blood banks and transfusion services should care-
fully evaluate the available BECS and select the system that
Figure 28–7. Display monitor showing patient report with corrected result. is best suited to the facility’s particular needs.
6888_Ch28_590-606 29/10/18 5:07 PM Page 600

600 PART V Quality Management and Compliance

Voluntary accreditation agencies, such as the Joint Com- sometimes inexplicable, failures of software or hardware can
mission, the American Association of Blood Banks (AABB), occur. Worse, a natural disaster such as a fire or flood can
and the College of American Pathologists (CAP), also have destroy parts or all of an information system. Software and
standards or inspection checklist items that emphasize the databases should be routinely copied to some storage
responsibilities of operating an information system. A blood medium, such as a backup hard drive. The copies can then
bank must have documented evidence of validation of its be used to restore any corrupted or lost information. The fre-
system as well as written procedures for all aspects of system quency with which this backup routine is performed depends
management so as to comply with regulatory and accredita- on each blood bank’s data volume and the recommendations
tion requirements. of the software vendor. The copies should be stored in a safe
location separate from the information system so that they
System Management will not be affected by disastrous events such as a fire or flood.
There should also be a procedure to identify and restore any
Many responsibilities are associated with the operation and information that was not included in the backup copies. This
maintenance of a computerized blood bank information sys- can be done by manually reentering data that was input be-
tem. The bulk of these responsibilities occur when a system tween the time of the most recent backup and the disaster.
is first installed as new SOPs are written, the system is vali-
dated, and personnel are trained. After a system is estab- Computer Downtime
lished, routine maintenance procedures are performed to With any computer system, there will be times when the sys-
ensure the ongoing operation of the system. tem is not available to users. These disruptions in availability
are commonly referred to as downtime. Sometimes the
Standard Operating Procedures downtime is planned, as when the system must undergo
Written standard operating procedures (SOPs) for every maintenance procedures, backup, or software enhancement.
blood bank operation are required by all regulatory and ac- Other downtimes are unplanned and usually unpleasant be-
creditation agencies, and computer operations are no excep- cause the system is experiencing some difficulty such as a
tion. The computer-related tasks associated with each of the power outage. In either case, there must be a written proce-
blood bank’s technical operations can be incorporated into dure that will allow blood bank operations to continue. The
each technical procedure or can be addressed in a separate procedure must address the need for retrieving historical pa-
section of the procedures manual. In addition, there are sev- tient and donor data and for recording new data obtained
eral computer-specific SOPs that must also be available. The during the downtime.
subjects of these are included in the following discussions. Transfusion service staff must be able to compare current
patient test results with previous records before blood com-
Archiving Data ponents are issued. Donor center staff must have access to
As the system’s databases grow in size, the hard disk becomes permanently deferred donor files. This essential information
crowded with this stored information. As a result, the com- may be in the form of paper records or may be available to
puter’s response time slows, and the hard disk is no longer personnel on a PC where the information has been previously
able to store new data. When this happens, additional hard downloaded from the blood bank information system. New
disks may be added to the system, or old data may be re- data generated during downtime—such as test results, blood
moved from the hard disk and placed in archival storage. Re- donations, and distribution or issuance of blood products—
moving old data frees up space on the hard disk so that new can be recorded on hard-copy worksheets or forms and then
data can be placed onto it. Old data may be archived to long- “backloaded” into the computer when it becomes functional.
term storage media such as microfiche and flash or external
Hardware Maintenance
hard drives. A procedure for periodically assessing hard disk
space should be instituted so that data can be archived in an Information system hardware, like all pieces of equipment,
orderly fashion before the hard disk becomes full. Procedures requires periodic cleaning, lubrication, and replacement of
for archiving must also be written and should adhere to ap- parts to ensure continued operation. Maintenance proce-
plicable regulatory and standard-setting agency requirements dures or system upgrades may require that the system be
for data retention. For example, AABB standards require that “taken down” or turned off for a period. For that reason, reg-
each donor’s ABO group and Rh type be retained for a min- ular maintenance procedures should be scheduled and
imum of 10 years, and patient records of adverse reactions posted so that blood bank staff members can prepare for
to transfusion must be retained for 5 years.3 Written proce- downtime. When possible, maintenance should be sched-
dures for retrieving archived data must ensure that records uled to minimize interruption of service, such as during
can be accessed within a reasonable period to maintain usual periods of low activity.
patient care.
Security Maintenance
Backup of Software Programs and Data A procedure for adding users to a system must be estab-
Blood banks have come to depend heavily on their informa- lished, including the basis on which security levels will be
tion systems, but systems are not infallible. Unexpected, and assigned. For example, the list of functions or applications
6888_Ch28_590-606 29/10/18 5:07 PM Page 601

Chapter 28 Laboratory Information Systems in the Blood Bank 601

to which a user will have access can be created for each job performed on an existing system, the testing should be done
description. It is also important to have a method for delet- in the test database, where it will not affect the production
ing the access codes of users who leave the facility. An ad- data. As with any validation, documentation must be retained
ditional level of security can be obtained by requiring users for all activities performed and the results, including unex-
to change their passwords periodically; many security ap- pected result or failures with appropriate corrective action.9
plications can be programmed to require this at specified
intervals. Risk Assessment
Proper validation of the system requires careful planning.
Tracking and Correcting Errors Risk assessment is a required analysis tool that focuses vali-
Error management, required by regulatory and accrediting dation efforts on system functions or settings that present
agencies, has become an integral part of blood bank opera- the greatest potential to harm the patient, donor, or business
tions. It includes detecting, documenting, and investigating in the event of a failure. Vendor descriptions of control func-
errors; implementing corrective actions; and reporting to ap- tions and on-site system configuration settings are reviewed
propriate agencies. Errors in the operation of a blood bank and assigned a risk category (such as high, medium, or low)
information system may be caused by inadequate training, by the system manager or validation team. A validation test
incomplete or cumbersome written procedures, or system plan is then prepared, ensuring that the highest risk areas re-
problems.11 In any case, tracking and categorizing errors is ceive an appropriate degree of validation.
essential if corrective actions are to be taken. Identified in-
formation system problems should be reported to the soft- Test Plan
ware vendor so that other users can be notified and remedies A test plan describes the series of exercises that will be used
can be included in a future software revision. to validate the blood bank software. Each system function
that the blood bank will use must be included in the test
Personnel Training plan. A test plan should be created for each function or op-
The most elegant and user-friendly information system can eration that the computer will perform. Risk analysis will de-
be a disaster if staff members are inadequately trained in its termine the extent of testing required. Test plans should
use. Training programs should address every function and include the following items:
procedure that each user will be expected to perform. Flow- • Control functions
charts and checklists are useful training tools. Training mod- • Data entry methods (keyboard, bar code reader, radio fre-
ules can be designed that present users with the situations quency identification (RFID), instrument interface, LIS,
that they will encounter in their work. The competence of or HIS interface)
each staff person must be documented before allowing rou- • Specific test cases
tine use of the computer. Competence assessment methods • Documentation methods
can include direct observation, review of system-generated • Acceptance criteria (expected outputs)
records, and written examinations. Once initial competence • Result review
has been demonstrated, periodic assessment must be done • Corrective action (if necessary)
to document the continued competence of blood bank staff • Acceptance
to use the information system appropriately. As previously
discussed, it is most beneficial to allow users access to a test Most of these items will be different in each of the system’s
database for training purposes. If one is not available, test applications, so a separate test plan for each application may
data can be used in the production database. be necessary. Control functions should be defined by the
vendor, but data entry methods will be selected by the user;
Validation of Software some applications may have multiple data entry methods.
Specific test cases are discussed below. Screen printouts,
Software validation is the establishment of documented evi- written logs, or printed reports can be used to document the
dence that provides a high degree of assurance that the sys- testing. After the testing is completed, the results should be
tem will consistently function as expected. It is the compared with acceptance criteria to determine acceptability.
responsibility of the software vendor to validate, to the ex- If unacceptable results are obtained, they should be investi-
tent possible, the functionality of a software product before gated, and corrective action should be implemented. When
it is marketed to blood banks. However, software vendors corrective action is necessary, testing should be repeated
cannot simulate the conditions in which each system will be until it is found acceptable.
used. Differences in environment, hardware, databases,
SOPs, and people require that each blood bank perform on- Test Cases
site validation testing to prove that the system will perform Validation test cases include the specific steps to be followed
appropriately under those unique conditions.9,12 by those performing the testing. Test cases should assess the
Validation testing must be performed before a new system system under normal operating conditions but must also
can be implemented and whenever new software, databases, offer challenges to the system. The goal is to make sure that
or hardware is added to the system. When validation is to be the system repeatedly performs intended functions and does
6888_Ch28_590-606 29/10/18 5:07 PM Page 602

602 PART V Quality Management and Compliance

not perform unintended functions. The types of testing that to patients at remote facilities.13 Virtual blood bank is an idea
should be performed are: that began with the computer crossmatch, namely that vali-
dated computer systems with appropriate truth tables and
• Normal testing
algorithms can be used to automate the selection of suitable
• Boundary testing
blood components.
• Invalid test cases
Patient specimens and segments from red blood cell
• Special test cases
(RBC)–containing products are sent from the satellite facility
• Stress testing
to the main central blood bank laboratory. RBC and other
Normal testing uses typical blood bank inputs to produce blood components can be stored at the remote facility in
normal, or routine, outputs. Boundary testing involves forc- monitored temperature-controlled storage devices. Patient
ing the system to evaluate data that are slightly below or specimens can be fully tested and the results entered into the
slightly above valid ranges. This kind of testing might be blood bank laboratory information system (BBLIS). If a sero-
used for disease test results. Invalid test cases assess the sys- logic crossmatch is required, then the RBC segment can be
tem’s ability to recognize and reject incorrect inputs. Exam- used for that purpose and the results entered into the BBLIS.
ples of invalid inputs include entering Q for an ABO If the patient is found to meet the criteria for electronic
interpretation or entering a blood product code that has not crossmatch, then the electronic crossmatch can also be per-
been defined in the static database file. Special test cases are formed at the central blood bank. The selection of appropri-
those that make the system react to unusual inputs. A special ate blood components can be made at the central blood bank
case could be designed to see how the system responds when for the patients and blood component inventory at the re-
more than one person attempts to add or edit special trans- mote satellite facility. Properly trained staff can then issue
fusion instructions in the same patient’s record. Stress test- the blood components as they are needed through the BBLIS.
ing involves pushing the system to its physical limits. This Figure 28–8 illustrates an example of a virtual blood bank
might be accomplished by allowing large volumes of data to using a blood vending machine.13 A blood vending machine
be entered into the system via all available input devices. usually consists of a temperature-controlled blood storage
Not every type of test case may be appropriate for each and dispensing refrigerator with a computer-controlled lock
function to be tested. For example, boundary test cases will and a computer terminal that allows authorized personnel
probably not be applicable in a donor registration function. to log in and retrieve units assigned to a particular patient.14
Another kind of validation testing is parallel testing. This in- As with any other BBLIS, the blood vending machine must
volves running two systems in parallel and comparing the out- be fully validated, the staff properly trained, security levels
puts of both. For a blood bank switching from a manual to a assigned and access to the device restricted to authorized
computerized system, every procedure would be performed personnel.
manually and in the computer. The virtual blood bank is a viable solution to improving
Validating a blood bank information system is a very turn-around-time on providing blood components as well as
lengthy and labor-intensive process, but it provides great a way to address staff shortages for trained and competent
benefits if undertaken in a thorough manner. Extensive test- blood bank technologists. In terms of blood utilization, the
ing will identify potential problems in the way that the blood literature is not consistent in demonstrating that there is
bank intends to use the system. Sometimes workarounds improved blood utilization after implementing electronic de-
have to be created, but when it is done as part of validation cision support systems (DSS). Adams et al and Bennet-
testing, staff members can be properly trained before going Guerrero et al demonstrated that blood transfusions decrease
“live” on the new system or software. The creation and per- after implementing DSS.14,15 However, studies performed by
formance of comprehensive and detailed validation test cases Collins et al and Hibbs et al did not show a significant overall
require allocation of significant resources to the project, but change in blood usage.16,17
the end result—a well-validated blood bank information
system—is worth the resource costs. Future Trends

Virtual Blood Bank As technology continues to advance, the design and function
of blood banks and transfusion services will change. Imple-
The advent of wireless technology, including radio frequency menting changes that increase the decision-making respon-
identification (RFID) is changing the way that large institu- sibility of BBLIS will require additional validation and
tions with remote satellite facilities utilize blood bank com- safeguards built into the systems. Staff training and compe-
puter systems. Concepts such as the virtual blood bank tency must also advance and keep pace with technology to
capitalize on the benefits of electronically linked blood bank maintain the high level of safety and efficiency in providing
information systems that allow technologists in a central suitable blood components to patients with a minimum of
location to select and assign appropriate blood components adverse effects.
6888_Ch28_590-606 29/10/18 5:07 PM Page 603

Chapter 28 Laboratory Information Systems in the Blood Bank 603

Central Blood Bank Remote Point of Care

Temperature-monitored
blood transport system

Hematology LIS

CBP, PT/APTT

BBLIS
BBLIS Tx record BBLIS
workstation Tx reaction workstation
Tx requirement Blood vending
Blood storage
refrigerator machine
CMS
(Clinical
Management
Telepathology System)
analyzer

Porter route
Information flow

Figure 28–8. Example of a virtual blood bank schematic. BBLIS = Blood bank laboratory information system; CBP = complete blood picture; PT = prothrombin time;
APTT = activated partial thromboplastin time; Tx = transfusion. (From: Wong, Kit Fai: Virtual blood bank. Journal of Pathology Informatics 2:6 Published online 2011 Jan 24
doi: 10.4103/2153-3539.76155. Open-access article distributed under the terms of the Creative Commons Attribution License.)

SUMMARY CHART
 Blood bank information systems assist in the manage- deleting items from the static database files, assigning
ment of data and can allow tracing of a blood compo- access codes to new users, implementing software up-
nent through all processing steps from donation grades from the vendor, and investigating and report-
through transfusion (final disposition). ing problems encountered by the users.
 A computer system is composed of three main compo-  A blood bank information system can have numerous
nents: hardware, software, and people. specific applications related to the management of
 Hardware components perform functions related to donors, patients, and blood components.
input and output, processing, and storage of data.  User codes and passwords prevent unauthorized use
 Software tells the computer what to do with the infor- of the system and provide a means for capturing
mation it has received. the identity of each person who performs a task on the
 Application software allows users to perform tasks that information system.
are specific to blood bank operations.  Control functions assist in making decisions at critical
 Donor, patient, and blood component information is points in the selection of donors and release of blood
maintained in databases, which are divided into files components for transfusion.
and further subdivided into individual records.  When results must be corrected or amended, they
 Static database files define the terminology that the must be clearly designated as such on printed reports
blood bank will use and provide a dictionary of coded and monitor displays used by patient caregivers.
terms that the system can use to sort data.  A blood bank must have documented evidence of
 Operating system software controls the hardware, mani- validation of its system as well as written procedures
pulates the application software, and coordinates the flow for all aspects of system management in order to com-
of information between the disks and memory. ply with regulatory and accreditation requirements.
 The system manager oversees the maintenance of the  Blood bank SOPs must address computer tasks related
system’s hardware and software, including adding or to blood bank technical duties and must address

Continued
6888_Ch28_590-606 29/10/18 5:07 PM Page 604

604 PART V Quality Management and Compliance

SUMMARY CHART—cont’d
computer-specific procedures such as computer down- under the unique combination of hardware, databases,
time, backup of software programs and data, system environment, SOPs, and people at the site.
maintenance, security, error management, and person-  Advances in technology and BBLIS foster implemen-
nel training. tation of Virtual Blood, Blood Vending machines and
 On-site software validation must provide documented streamlined procedures with increased reliance on de-
evidence that provides a high degree of assurance that cision support systems (DSS) which require additional
the system will function consistently as expected validation and maintenance.

7. Preventing the issue of an incompatible blood compo-


Review Questions
nent is an example of:
1. Components of an information system consist of all of a. Inventory management
the following except: b. Utilization review
c. System security
a. Hardware
d. Control function
b. Software
c. Validation 8. Information is stored in a collection of many different
d. People files called the:
2. To be in compliance with regulatory and accreditation a. Database
agency requirements for blood bank information systems, b. Configuration
blood banks must maintain SOPs for all of the following c. Hardware
except: d. Disk drive
a. Vendor validation testing 9. Application software communicates with this type of
b. Computer downtime software to retrieve data from the system disks:
c. System maintenance a. Interface
d. Personnel training b. Operating system
3. A validation test case that assesses the system’s ability to c. Security
recognize an erroneous input is called: d. Program
a. Normal 10. Validation testing for software should consider all of the
b. Boundary following items except:
c. Stress a. Data entry methods
d. Invalid b. Control functions
4. An example of interface software functionality is: c. Performance of testing in production database
d. Invalid data
a. The entry of blood components into the blood bank
database 11. Complete the truth table below for a negative antibody
b. The transmission of patient information from the HIS screen using two screening cells (SCI, SCII) at the im-
into the blood bank system mediate spin (IS), 37°C (37), and antihuman globulin
c. The printing of a workload report (AHG) phases.
d. Preventing access to the system by an unauthorized
user
Phase SCI SCII Interpretation
5. Backup copies of the information system:
a. Can be used to restore the information system data and IS
software if the production system is damaged
37°C
b. Are used to maintain hardware components
c. Are performed once a month AHG
d. Are created any time changes are made to the system
CC
6. User passwords should be:
a. Shared with others
b. Kept confidential
c. Posted at each terminal
d. Never changed
6888_Ch28_590-606 29/10/18 5:07 PM Page 605

Chapter 28 Laboratory Information Systems in the Blood Bank 605

12. During validation testing, a computer user entered the 4. The Transfusion History screen display will indicate the
following results for an antibody screen test: patient has been transfused and will display the date of
the last transfusion.
SCI IS 37 AHG CC Interpretation 5. Printed reports will indicate relevant units were assigned
Result 0 0 0 + Negative transfused status.

After the user verified the entries, the monitor displayed Name of Section C. ________________________________
the following message: “Invalid test results.” What Section D
caused the error message to display?
1. Attempt to assign transfused status to the following units:
a. An invalid entry was made in the check cells (CC)
a. Quarantined blood component
column
b. Selected (but not issued) blood component
b. The truth table was set up incorrectly
c. Transfused blood component
c. The interpretation does not correlate with the test
entries 2. Selection of units from issued inventory list
d. The interface to the laboratory computer system is 3. Manual entry of issued blood components
down Name of Section D. ________________________________
13. The following test plan has been created to validate the
Section E
blood bank computer function used to update the status
of blood units that have been transfused. The test plan 1. Operator will input blood component information and
contains each of the sections, lettered A through H, re- select blood components.
quired for a thorough test plan. Evaluate each section 2. Operator will input selected patient information.
and, using the list below, assign a name to each section. 3. Blood bank computer will update the patient and unit
record.
Section Names
• Acceptance • Data entry methods Name of Section E. ________________________________
• Acceptance criteria • Documentation methods Section F
• Control functions • Result review
• Corrective action • Test cases The following screen displays and printed reports will be
verified for accuracy:
Test Plan Function: Assigning Transfused Status
Section A Screen Displays Printed Reports
Patient Information Patient History Report
Description: This function is used to change the status of is- Unit Information Transfusion Listing
sued units to a transfused status. All records pertaining to Unit History
the unit and patient will be updated: Transfusion History
Name of Section A. ________________________________
Name of Section F. _________________________________
Section B
Section G
1. Preventing the assignment of transfused status to a quar-
antined blood unit. The acceptability of the results of each test case will be de-
2. Preventing the assignment of transfused status to a blood termined by the blood bank manager and documented on
unit that has already been transfused. the validation documentation form. If the software does not
3. Preventing the assignment of transfused status to a blood perform as expected, the problem must be recorded on a
component that has not been issued. computer problem report and the supervisor alerted. A re-
medial action plan will be devised with the assistance of the
Name of Section B. ________________________________ blood bank computer system vendor.
Section C Name of Section G. _________________________________
1. The computer will beep and display **Unit Has Not Been Section H
Issued** when a blood unit number that has not been is-
Blood bank director signature: ____ Date: _____________
sued is entered.
Comments: _______________________________________
2. The computer will beep and display **Unit Is in Quar- Name of Section H. _______________________________
antined Status** when a blood component that is in quar-
antine status is entered.
3. The computer will beep and display **Unit Has Been
Transfused** when a blood component that has already
been transfused is entered.
6888_Ch28_590-606 29/10/18 5:07 PM Page 606

606 PART V Quality Management and Compliance

References 9. General principles of software validation; final guidance for in-


dustry and fda staff. Food and Drug Administration. January 11,
1. Delgado MA, ISBT 128 for blood components an introduction 6th 2002. https://www.fda.gov/downloads/MedicalDevices/Device
ed. San Bernardino (CA): ICCBBA; 2018. Available from: http:// RegulationandGuidance/GuidanceDocuments/UCM085371
www.iccbba.org/uploads/4c/e4/4ce45e99017ab8b3767d01d5da113f .pdf
c6/IN-003-ISBT-128-for-Blood-Components-An-Introduction-v6 10. Medical Devices 510(k) Clearances. Food and Drug Adminis-
.pdf tration. https://www.fda.gov/MedicalDevices/ProductsandMedical
2. Verlicchi FR, Pacilli PA, Braglianni AR, Rapuano SI, Dini DA, Procedures/DeviceApprovalsandClearances/510kClearances/
Vincenzi DA. Electronic remote blood issue combined with a default.htm accessed 07-23-18.
computer_controlled, automated refrigerator for major surgery 11. Petrides AK, Bucha I, Goonan EM, Bates DW, Shaykeuich S,
in operating theatres at a distance from the transfusion service. Lipsitz SR, et al. The benefits and challenges of an interfaced
Transfusion. 2018;58(2):372-8. electronic health record and laboratory information system:
3. Standards for Blood Banks and Transfusion Services 31st ed. effects on laboratory processes. Arch Pathol Lab Med. 2017;
AABB, Bethesda MD. 2018. 141(3):410-7.
4. Guidance for industry: blood establishment computer system 12. ISBT Guidelines for validation of automated systems in blood
validation in the user’s facility. Food and Drug Administration. establishments. Vox Sang.2010;98(S1):1-19.
April 2013. https://www.fda.gov/downloads/BiologicsBlood 13. Wong KI. Virtual blood bank. J Pathol Inform. 2011;(2):6.
Vaccines/GuidanceComplianceRegulatoryInformation/Guidances/ 14. Adams ES, Longhurst CA, Pageler NM, Widen E, Franzon D,
Blood/ucm078815.pdf Cornfield DN. Computerized physician order entry with deci-
5. Guidance for industry: “computer crossmatch” (computerized sion support decreases blood transfusions in children. Pedi-
analysis of the compatibility between the donor’s cell type and the atrics. 2011;127, e1112–e1119.
recipient’s serum or plasma type). Food and Drug Administration. 15. Bennett_Guerrero EL, Zhao YU, O’Brien SM, Ferguson TB,
April 2011. https://www.fda.gov/downloads/BiologicsBlood Peterson ED, Gammie JS, et.al.Variation in use of blood trans-
Vaccines/GuidanceComplianceRegulatoryInformation/Guidances/ fusion in coronary artery bypass graft surgery. JAMA 2010;
Blood/UCM252894.pdf 304(14):1568-75.
6. Bordowitz RI, Electronic health records: a primer. Lab Medicine. 16. Collins RA, Triulzi DJ, Waters JH, Reddy VI, Yazer MH. Eval-
2008;39(5):301-6. uation of real_time clinical decision support systems for
7. Savage W. Implementing a blood utilization program to optimize platelet and cryoprecipitate orders. Am. Journ..Clin. Path.2014;
transfusion practices. Hematology Am Soc Hematol Educ Pro- 14(1):78-84.
gram. 2015;2015:444-7. 17. Hibbs SP, Noel S, Miles DO, Staves PE, Murphy MF. The impact
8. Foss ML, Stubbs JR, Jones G. Integrating quality, education, lean, of electronic decision support and electronic remote blood
and performance management into a culture of continuous issue on transfusion practice. Trans. Med. 2014;24(5):274-9.
improvement. Transfusion. 2011; 51(7 Pt 2):1598-603.
6888_Ch29_607-616 29/10/18 5:07 PM Page 607

Chapter 29
Medicolegal and Ethical Aspects of Providing
Blood Collection and Transfusion Services
Kathleen Sazama, MD, JD, MS, MT(ASCP) and Matthew Montgomery, MD

Introduction Invasion of Privacy Under Civil Case Studies


Focus of Current Legal Issues Case Law Case Study 29-1
Sources of Law Basis of Liability: HIPAA Case Study 29-2
Statutes and Regulations Restrictions on Plaintiff Suits and Case Study 29-3
Case Law Recovery Case Study 29-4
Basis of Liability: Torts Statutes of Limitations Case Study 29-5
Intentional Tort of Battery Doctrine of Charitable Immunity Case Study 29-6
Doctrine of Informed Consent Tort Reform Summary Chart
Negligence Risk Management Review Questions
Voluntary and Mandatory Standards Specific Donor Issues References
Blood Banking as a Medical Profession Processing, Labeling, and Distribution Bibliography
Strict Liability Medical Malpractice
Intentional Infliction of Emotional Ethics and Transfusion Medicine
Distress Emerging Issues

OBJECTIVES
1. Describe the legal and ethical parameters for providing blood collection and transfusion services.
2. Describe the legal bases for liability for providing transfusion medicine services.
3. Explain the necessity of establishing and following standard operating procedures similar to those of other comparable facilities
throughout the United States.
4. Identify the evolving legal and ethical concerns that are likely to accompany the increasing complexity of providing blood
during the early 21st century.
5. List the two reasons why patients sue for transfusion injury.
6. Describe the steps that blood bank professionals can take to avoid or minimize litigation.

Introduction reducing the likelihood of being found liable, when possible.


Emerging concerns for the 21st century are also identified. This
Patients who are injured during, or as a result of, transfusion chapter is not intended as a substitute for legal advice, which
may seek reparation through legal channels, a complaint that is necessary in particular situations; rather, it is intended to
is usually filed by their families. This has been of particular provide the reader with some general principles and definitions
concern in blood banking and transfusion medicine because so that ethically and legally sound practices continue within
of the possibility of disease transmission, especially from pre- transfusion medicine.
viously unknown sources, and because mistakes may mean
death. Basing practices on sound ethical principles provides Focus of Current Legal Issues
a good foundation to limit liability when these unforeseen
events occur. This chapter briefly describes the sources of At the center of legal issues for the transfusion medicine pro-
law and theories of liability, including practical hints for fessional in the 21st century are ABO errors, acute lung injury,
607
6888_Ch29_607-616 29/10/18 5:07 PM Page 608

608 PART V Quality Management and Compliance

and patient privacy. There are also concerns about possible applied in particular instances. In the absence of federal law,
transfusion-transmitted diseases, including babesiosis, zika, state law can provide precedence for future decisions. However,
and dengue fever or other exotic diseases that are emerging states may choose to adapt, follow, or ignore decisions made in
globally. Concern has also focused on issues regarding trans- sister state courts, whereas federal law is applicable to all states.
fusion indications, informed consent, and other medically rel- The details of how laws are to be put into action are provided
evant topics. A growing discipline called patient blood in regulations. Regulations, both federal and state, can be ap-
management continues to gain prominence in light of the on- plied only if they have been established according to a formal
going emphasis on evidence-based medical practice. Many process called the Administrative Procedure Act (APA).3 Federal
studies have now been published questioning the safety of regulations that apply specifically to blood banking are found
blood transfusions, both allogeneic and autologous, with con- in the Title 21, Code of Federal Regulations, Parts 600, 601,
cerns regarding the age of the red blood cells at the time of 606, 608, 610, 630, 640, and 660, 210–211, and 820, published
transfusion and recognition that patient anemia may play a annually.4 Blood banks and transfusion services are also feder-
larger role in patient outcomes than previously appreciated. ally regulated by provisions of the Clinical Laboratories Im-
However, even before transfusion-transmitted AIDS provement Act of 1988 (CLIA 88) and the Medicare provisions
(TTAIDS) became the basis for numerous lawsuits against of the Social Security Act. In addition, the 1996 federal law pro-
blood centers, hospitals, and physicians, there was litigation tecting health information (Health Insurance Portability
because of death and serious injury caused by transfusion- and Accessibility Act [HIPAA]), including the 2008 Health In-
transmitted hepatitis B virus (HBV) and a few because of formation Technology for Economic and Clinical Health
donor injury.1 (See Perlmutter v. Beth David Hospital, 308 NY (HITECH) Act that extends these protections to electronic
100, 123 N.E.2d 792 [1954] in which the court decided that records and transmissions and the 2008 federal law, the Genetic
transactions involving blood were not sales but were inci- Information Nondiscrimination Act (GINA), also apply.
dental to the provision of medical services. This decision pre- State legislatures also enact laws and publish explanatory
cludes the application of commercial law, particularly that of regulations about blood banking, clinical laboratories, and
warranties, to blood transfusions.) transfusion practices, principally covering licensure of facil-
The HBV cases stimulated nearly every state legislature to ities and personnel.
enact protection for blood banks through blood shield
statutes.2 (California was the first state to enact such protection Case Law
for blood banks in 1955. Only New Jersey lacks such a law, but
it provides similar protection solely through judicial decisions.) Case law is established by court decisions, sometimes related
Because of these blood shield statutes, most of which extended to interpretations and applications of statutes and regulations.
protection without amendment or modification for TTAIDS Patients generally believe that medical treatment administered
and HBV, many TTAIDS lawsuits have been either dismissed to them (after obtaining their informed consent) will, on bal-
or unsuccessful for the person suing (the plaintiff). ance, be beneficial. When transfusion causes harm (e.g.,
Recently, however, this premise has come under new chal- through identification error, lung injury, and other causes),
lenge in the courts. As most TTAIDS cases have been unsuc- patients have understandably reacted by seeking reparation in
cessful in compensating plaintiffs (usually patients or their the courts. The legal bases for such suits are generally civil
families), new theories of liability are increasingly being (not criminal) actions for tort. Tort is defined as any wrong-
raised. When questions of appropriateness of transfusion, doing for which action for damages may be brought.
availability of blood components, and informed consent U.S. civil law depends on each competent adult in our
arise, the ethical bases of transfusion medicine practice are society behaving reasonably (i.e., not negligently and not ag-
focused more sharply. gressively) toward every other person, respecting other peo-
Although blood shield statutes generally protect against ple’s rights. Civil lawsuits arise because someone disrespects
application of strict liability, tort liability remains the basis another’s rights by:
for most lawsuits. 1. Striking or threatening to strike another person (battery
and assault)
Sources of Law 2. Being careless or reckless (negligence)
3. Failing to complete an agreement (breach of contract)
Laws are created by society through legislation called
4. Intruding on another’s property or privacy
statutes (passed by either the U.S. Congress or by individual
5. Misbehaving in other, similar ways
state legislatures) or by court decisions, referred to as case
law (in federal courts, including the U.S. Supreme Court, or Civil lawsuits can also arise because of violation of
in state courts through their own highest state court). statutes or regulations that require certain types of actions.

Statutes and Regulations Basis of Liability: Torts


Federal law (enacted by Congress or by decision in federal Torts arise from improper interactions between individu-
courts, including the Supreme Court) may frequently supersede als. Some of the common torts are described on the fol-
state law, but both federal and state laws can be and often are lowing page.
6888_Ch29_607-616 29/10/18 5:07 PM Page 609

Chapter 29 Medicolegal and Ethical Aspects of Providing Blood Collection and Transfusion Services 609

Intentional Tort of Battery history is obtained have emphasized more face-to-face oral
questioning, not just self-administration of these question-
Any touching without consent, including deliberate blows naires, until the recent availability of information technology.
and intentional striking, is the legal definition of battery. For One part of the donation process has always been for donors
transfusion medicine, this concept is used when a donor or a to sign a statement of consent or assent to donate.
patient claims that he or she never agreed to have the needle Until the late 1980s, donors were protected from sub-
placed in his or her arm. If significant harm occurs as a result poena in cases of transfusion-transmitted diseases. However,
of the needle, this legal theory may be upheld. Generally, an increasing number of plaintiffs in state courts are insisting
however, the fact that the donor or patient allowed the needle that the donor be subject to questioning regarding his or her
to be placed is sufficient evidence that he or she agreed to the donation. This questioning may completely or partially pro-
procedure. Informed consent is generally not an issue in bat- tect a donor’s identity or may require that donor to appear
tery but is fundamental in the tort of negligence. in open court. Because no one donates blood expecting to
have to defend such an altruistic act at some future date, the
Doctrine of Informed Consent impact of these decisions on the future availability of blood
for transfusion is uncertain.
Particularly in the special circumstances of the practice of
medicine, the issue of whether a patient (or, in the case of
blood collection, a donor) agreed to undergo the procedure Negligence
actually performed, with full knowledge of the possible ben-
Probably the most common tort is that of negligence. This
efits, alternatives, and harm that may accompany it, has
is the basis for lawsuits when injury occurs.
come to be known as the doctrine of informed consent. This
doctrine protects the patient (or donor) by requiring that in- Elements
formation be provided in a manner understandable to the
Liability for negligence is found when all of the following
patient under circumstances that permit the patient to ask
elements are present:
questions or express concerns and to receive answers to
them (Canterbury v. Spence, 464 F.2d 772 [DC Cir 1972]). 1. A duty was owed to the injured party.
For TTAIDS transfusion recipients, this issue first came 2. The duty was not met by the injuring party.
into sharp focus in the case of Kozup v. Georgetown Univer- 3. Because the duty was not met, the injured party was
sity, 663 F. Supp. 1048 (DC 1987), 851 F.2d 437 (DC Cir harmed.
1988), 906 F.2d 783 (DC Cir 1990). In this case, an infant 4. Failure to meet the duty owed was directly responsible
brought by his parents to Georgetown University Hospital for or could have been predicted to cause the harm suf-
for medical care contracted TTAIDS and died. His parents fered by the injured party.
argued that they were insufficiently informed about the harm 5. Some measurable (compensable) harm (called damages)
of the transfusions and did not specifically agree to transfu- occurred. To be successful in a negligence action, the
sions as part of the care given their child. The District of plaintiff has the responsibility to prove all these factors
Columbia court ruled that their actions in bringing the child against the person being sued (the defendant).
to the hospital and not objecting to transfusions that they
observed at the time of infusion amounted to implied con- Standard of Care
sent. The issue of whether specific consent is required for There are two standards of care applicable to blood collection
transfusion and who should obtain such consent remains and transfusion services: ordinary and professional.
controversial. Generally, physicians have been held respon-
sible (Ritter v. Delaney, 790 S.W.2d 29 [Tex. App. San Antonio, Ordinary Standard of Care. When the alleged negligence involves
1990] and Howell v. Spokane, 785 P.2d 815 [Wash 1990]; ordinary things that anyone may encounter in daily life (e.g.,
Hoemke v. New York Blood Center, 90–7182 [2d Cir. 1990] injuries caused by traffic accidents, fistfights, etc.), a jury or
and Gibson v. Methodist Hospital, 01-89-00645-CV [Tex. Ct. judge can consider the facts and decide whether the behavior
App. 1991]). However, in 1996, the Joint Commission of the defendant was reasonable—that is, did the defendant
specifically required hospitals to obtain informed consent for meet the standard of care required in the situation? For or-
transfusion in some situations.5 dinary negligence, this standard of care depends only on
Another arena in which the informed consent is the key what the average person (e.g., a juror) believes is acceptable
to resolving disputes is that of donor rights. With the onset in our society, or the ordinary standard of care (i.e., the jury
of AIDS, the language of donor histories has been subjected decides whether a reasonable person, in the same circum-
to continuous revising and updating to include information stances as the defendant, would have acted the same way as
about the disease, how it is acquired, and under what cir- the defendant did). If the defendant acted reasonably in the
cumstances a person may donate blood. In the United States, circumstances, the plaintiff will be unsuccessful in the law-
unlike other countries, there is no right to donate blood or suit and vice versa.
other tissues. Instead there has been increasing emphasis on In situations in which larger organizations are involved,
educating donors about the restrictions on blood donation. the question of who is liable for the actions of employees has
Federal and voluntary requirements for the way in which the been resolved under the doctrine of respondeat superior.
6888_Ch29_607-616 29/10/18 5:07 PM Page 610

610 PART V Quality Management and Compliance

Under this doctrine, the actions of employees are attributable processing, and distributing blood may be considered dif-
to the employer or person who directs their actions. The per- ferently from the crossmatching, issuance, and transfusion
son responsible for transfusion services has been defined by of blood to individual patients. Redefining blood banking
federal regulation and general practice to be a physician, a as not medical practice removes the extra protection pro-
definition that has been reinforced by judicial decision in vided by the requirement for expert medical testimony to
many states for blood centers as well as for hospitals. The establish the standard of care, leaving defendants to be
advantage of having a physician as the responsible employer judged by the ordinary negligence standard. There is little
is that the principles and regulations related to medical mal- dispute that loss of medical professional stature would sig-
practice usually apply, including the requirement for estab- nificantly alter the practice of blood banking. None of the
lishing a professional standard of care. protections of medical malpractice reform would be avail-
able, and blood centers may even find themselves subject
Professional Standard of Care. When the negligence lawsuit to strict liability.
involves professionals such as physicians and scientists, in-
cluding laboratory professionals, nurses, or other allied Strict Liability
health practitioners, the definition of what is reasonable, the
“standard of care,” depends on expert testimony from other Manufacturers and distributors of goods used in everyday
professionals in the same field (e.g., physicians, nurses, or life have been defined by law to have certain responsibilities
scientists) about what should have been done by other rea- in their activities to protect consumers. Among other re-
sonable practitioners (usually of the same specialty). The law quirements, there are certain warranties that the product
makes an extra requirement: in discharging his or her duty purchased—for example, a television set—will actually work
to the plaintiff, the defendant must apply the special knowl- and will continue to do so for some fixed period, with small
edge and ability he or she possesses by virtue of the profes- risk of harm from such things as electric shock or blowing
sion. This increased professional standard of care is not just up. These warranties, actual or implied, exist for virtually
what a reasonable person such as the judge or members or anything a consumer buys and uses. If a product fails to per-
the jury would have done but also what other professionals form as expected or creates harm when none was expected,
(the expert witnesses) testify should have been done. The the consumer has the right to have a replacement or, if the
judge or jury is not permitted to decide what they would manufacturer denies responsibility, to sue for negligent man-
have done but must depend upon testimony by expert wit- ufacturing or distribution.
nesses of the same profession as the defendant who define In addition, for some items (such as dynamite), the dan-
what that reasonable professional standard of care is. For the ger from proper use is so great that manufacturers are legally
complex scientific, technical, and medical issues involved in liable for all harm that occurs, which is called strict liability.
TTAIDS litigation, this distinction has been a key element in This means that anyone who is harmed when properly using
protecting blood bankers. dynamite does not have to prove that the manufacturer or
distributor was negligent; he or she has only to show that he
Voluntary and Mandatory Standards or she was injured while properly using it. Imagine how rare
and expensive blood transfusions would become if these
The testimony of experts should generally be supportable by practices were applied to it. Instead, nearly every state has
authorities such as statutes, regulations, or other bodies of enacted specific protection—the blood shield statutes
published knowledge, including published scientific articles described previously—to exclude harm from blood transfu-
and texts. The existence of voluntary standards—particularly sions from suit under these legal theories. It is important that
those provided by the AABB (formerly known as the American blood bankers avoid implying or stating that blood transfu-
Association of Blood Banks)6 but also those from the College sion is completely safe because such statements may be con-
of American Pathologists (CAP), the American Association strued as creating a warranty, invoking these theories of
of Tissue Banks (AATB), the American Society for Histo- liability.
compatibility and Immunogenetics (ASHI), the Joint Com-
mission (TJC), and other organizations—are helpful in Intentional Infliction of Emotional Distress
establishing the professional standard of practice for trans-
fusion medicine. In fact, some state laws and federal regula- As the phrase intentional infliction of emotional distress sug-
tions cross-reference the AABB standards specifically. Blood gests, a plaintiff must show that what the defendant did to
bankers and transfusion services that can show that they cause actual and severe emotional distress was intentional,
acted in conformance with these standards are more likely usually some extreme or outrageous conduct that was cal-
to be found nonnegligent than those that do not follow such culated to deliberately cause harm to the plaintiff. This can
guidelines. take the form of the plaintiff’s relative claiming wrongful
death, particularly in some TTAIDS cases.
Blood Banking as a Medical Profession
Invasion of Privacy Under Civil Case Law
The question of whether blood banking is a medical profes-
sion is being relitigated in courts today with conflicting re- Health-care providers, including blood bankers, are required
sults.7 One possible chilling result is that collecting, to respect personal privacy and to maintain patient and
6888_Ch29_607-616 29/10/18 5:07 PM Page 611

Chapter 29 Medicolegal and Ethical Aspects of Providing Blood Collection and Transfusion Services 611

donor confidentiality. Plaintiffs may claim remuneration for patient records and test results from wherever they are), the
loss of privacy under four theories: rise in the adoption rate of these technologies increases the
potential security risks. On February 17, 2009, the Health
1. Intruding upon the plaintiff’s seclusion or solitude or into
Information Technology for Economic and Clinical Health
his or her private affairs
(HITECH) Act, enacted as part of the American Recovery
2. Publicly disclosing embarrassing facts
and Reinvestment Act of 2009, was signed into law. This law
3. Publicly placing plaintiff in a “false light”
was designed to strengthen the privacy and security protec-
4. Appropriating plaintiff’s name or likeness for defendant’s
tions for health information established under the Health In-
benefit
surance Portability and Accountability Act of 1996 (HIPAA).
These categories protect a patient or donor from illegal or The privacy of individually identifiable health information
inadvertent disclosure of his or her personal information, of known as “protected health information” (PHI) is protected
particular concern with HIV because of the risk of loss of by HIPAA. The “21st Century Cures Act of 2016 (Cures Act)
employment, housing, insurance, and other benefits of so- Mandate” issued by the OCR requires the Secretary of the
ciety. When information is exciting or noteworthy, the media Department of HHS to issue “Guidance Related to Stream-
may become aware of and publish private information. lining Authorization” under HIPAA for uses and disclosures
of protected health information (PHI) for research. In accor-
Basis of Liability: HIPAA dance with the Cures Act, authorizations for the use or dis-
closure of PHI for future research (or other purposes) must
In 1996, the U.S. Congress responded to the information age include a description of each purpose of the requested use
by enacting a law specifically directed toward protecting per- or disclosure.
sonal health information (PHI): the Health Information Although cases have been filed, blood banks and transfu-
Portability and Accessibility Act (HIPAA). The regulations sion services will be involved in suits under the preexisting
implementing the provisions of this act occurred in segments civil cases if they are responsible (through negligence or by
beginning in 1996. The U.S. Department of Health and intention) for releasing confidential data of a donor or pa-
Human Services (HHS) published the HIPAA Privacy Rule tient. Great care should be exercised by blood-banking pro-
in 2000 and the HIPAA Security Rule in 2003. The Privacy fessionals to ensure that private information (whether about
Rule, or Standards for Privacy of Individually Identifiable donors, patients, or relatives) be kept confidential and not
Health Information, establishes national standards for the released without written authorization. Procedures to safe-
protection of certain health information. The Security Stan- guard such release should include proper use of copy and
dards for the Protection of Electronic Protected Health In- facsimile machines and direct electronic transfer via infor-
formation (the Security Rule) establish a national set of mation systems.
security standards for protecting certain health information
that is held or transferred in electronic form. For example, Restrictions on Plaintiff Suits
laboratories must ensure confidentiality when transmitting and Recovery
health information electronically. This rule requires health-
care organizations (laboratory test sites are part of this), in- Even when harm or injury occurs, the plaintiff may be unable
surers, and payers using any electronic means of storing to pursue legal redress for a number of reasons.
patient data and performing claims submission to comply
with the Final Rule for National Standards for Electronic Statutes of Limitations
Transactions published in 2009.
With this new federal requirement, information about Some protection for defendants arises because of a statutorily
donors and patients must be kept in such a manner that in- defined limit of time during which a lawsuit can be filed, re-
advertent “use or disclosure” does not occur. The Security ferred to as the statute of limitations. States often have dif-
Rule operationalizes the protections contained in the Privacy ferent limits, depending on the legal requirements for
Rule by addressing the technical and nontechnical safe- initiating suits. Statutes of limitations for medical malprac-
guards that organizations (“covered entities”) must put in tice are generally shorter (approximately 2 years from the
place to secure individuals’ “electronic protected health in- date that the injury should have been discovered in adults)
formation” (e-PHI). Within HHS, the Office for Civil Rights than limitations for other kinds of negligence (2 to 6 years
(OCR) has responsibility for enforcing the Privacy and Se- is common).
curity Rules with voluntary compliance activities and civil
money penalties. Doctrine of Charitable Immunity
Today, providers are using clinical applications such as
computerized physician order entry (CPOE) systems, elec- Historically, courts provided immunity for nonprofit orga-
tronic health records (EHR), and radiology, pharmacy, and nizations such as hospitals from excess liability because they
laboratory systems. Health plans are providing access to perform charitable acts. This is referred to as the doctrine of
claims and care management, as well as member self-service charitable immunity. Many state legislatures have enacted
applications. While this means that the medical workforce and continue to support statutes to provide protection for
can be more mobile and efficient (i.e., physicians can check boards of directors and volunteers using this common law
6888_Ch29_607-616 29/10/18 5:07 PM Page 612

612 PART V Quality Management and Compliance

rationale. A 1996 decision in New Jersey redefined this pro- attention to the process of donor screening is appropriate.
tection in that state (Snyder v. AABB, 144 N.J. 269 [1996]). Balancing the real threat to patient care that would arise if
Fortunately for blood bankers, other states have been loath blood components were not available with the serious na-
to accept the Snyder decision. Also, many health-care insti- ture of litigation will continue to challenge blood-banking
tutions rely less on this doctrine and more on insurance for professionals.
protection.
Donations Requested by Patients
Tort Reform Several monetary settlements in the range of hundreds of
thousands to millions of dollars have resulted from either
State legislatures, recognizing the need to protect some spe- failing to offer directed donor services or from improperly
cialties such as obstetrics, have been active in seeking limits characterizing them. With the advent of the HIPAA, patient-
on damages against physicians, protecting them from abu- requested donations from friends and family require close
sive litigation. For transfusion practices, it is vital that these attention to ensure that inadvertent or overt disclosure of
protections be afforded. protected health information does not occur.

Risk Management Untimely Notification


When recipients received notification in years, rather than in
Avoiding liability for TTAIDS and other possible harm
weeks or months, after blood centers knew (or should have
from practicing transfusion medicine, whether in hospitals
known) that the recipient had received an HIV-reactive unit,
or blood centers, depends on having well-established
many of them or their families were angry enough to file suit
policies and procedures that are consistent with quality
on the basis that they should have been informed sooner. Pro-
principles; that comply with recognized authorities, regu-
cedures for look-back and recipient notification were not well
lations, and statutes; and that have some measurement of
established for several years following application of specific
how persons engaged in all activities actually follow those
testing in most blood collection organizations. It was not until
procedures. Complying with accreditation requirements
1996 that the federal government, through the Center
(e.g., AABB, TJC, CAP, and state laws) for quality systems
for Health Care Financing Administration (now called Cen-
will assist in limiting risk. To avoid being negligent, one
ters for Medicaid and Medicare Services, or CMS) and the
must behave reasonably. Reasonable behavior for transfu-
FDA, issued specific regulations for HIV look-back notifica-
sion medicine practice includes continually obtaining and
tion by hospitals. Parallel requirements for HCV, HBV, and
applying new knowledge from all possible sources that will
other transfusion-transmissible infections have also been
safeguard the donor during collection, the component dur-
developed.
ing handling and delivery, and the patient before and during
transfusion. Component Collection
Specific Donor Issues Occasionally, lawsuits have occurred from injury that donors
received during the collection process. Generally, the injuries
Although it is possible to sue another party for many reasons, are more severe than a simple bruise at the needle site and
the most common are discussed here. involve such things as nerve damage, slip-and-fall incidents,
and other similar severe reactions. Also, although donor
Screening deaths continue to be reported at a rate of approximately two
What the AIDS epidemic has taught us is that every person per year, these infrequently result in litigation.
who volunteers to donate does not have an unqualified
right to do so. In fact, for several years before March 1985 Processing, Labeling, and Distribution
(when a test for HIV antibodies in blood first became avail-
able), the best safeguard against TTAIDS was improved Lack of Standard Protocol for Implementing Testing
donor education and more pertinent questioning regarding Several successful TTAIDS lawsuits awarded millions of dol-
behaviors that might put that donor at risk for acquiring lars against blood-collecting organizations (Belle Bonfils
HIV. Although numerous lawsuits have been filed against Memorial Blood Bank v. Denver District Court, 723 P.2d 1003
blood collection agencies for improper donor screening, [Colo. 1988]) because they had no standard protocol for im-
few have been successful when collection facilities could plementing new testing to ensure that all available compo-
show that they had implemented written procedures and nents (including those distributed or in active inventory)
had properly trained employees who followed those proce- were test-negative before transfusion once the test kits and
dures and that proper documentation of each screen equipment were received by the collection facility. The lack
occurred. Problems occurred when breaches in procedure, of a written plan to implement such testing, including train-
typically failures to follow standard operating procedures ing of personnel, validating instrumentation and reagents,
or to properly document actual practice, were discovered. and establishing the necessary information service support,
However, with several state courts demanding release of was seen as negligent by the jury (even with expert testimony
donor identity or access to donors for questioning, further to the contrary).
6888_Ch29_607-616 29/10/18 5:07 PM Page 613

Chapter 29 Medicolegal and Ethical Aspects of Providing Blood Collection and Transfusion Services 613

Failure to Perform Surrogate Testing


BOX 29–1
Despite concerted efforts, rarely has a plaintiff prevailed
when alleging that blood collection facilities should have Ethical Principles and Definitions
performed more or different surrogate tests between 1983 Autonomy: The right of each person to make decisions based on
and 1985, when a specific HIV antibody test was first avail- that person’s values and beliefs, having adequate information and
able (Baker v. JK and Susie Wadley Research Institutes and an understanding of the choices available to him or her and
Blood Bank, aka The Blood Center at Wadley, 86-2728-C [Tex. lacking any compulsion by external forces.
Jud. Dist. Ct. 1988] and Clark v. United Blood Services, CV Beneficence: In the health-care setting, professionals seek the
well-being of each patient.
88–6981 [Nev. 2d Jud. Dist. Ct. 1990]).
Justice: Patients should be treated fairly, with equal, need-based
access to beneficial treatment.
Failure to Properly Perform Testing
Testing personnel should be constantly alert to proper per-
formance and documentation of all required testing of blood
components. There are multiple opportunities for errors to
occur during the processes of collecting and transfusing type of therapy and the source of the blood they receive. Al-
blood components. In addition to mistakes in doing the re- though autonomy is not unrestricted, it must form part
quired testing, failure to document is as damning as failure of the health-care relationship. When transfusions are of-
to perform at all. The FDA has expended considerable effort fered, patients expect that the treatment will benefit them.
to inform facilities and to enforce requirements for proper When the benefit is marginal or even questionable and, in
testing for virally transmissible diseases in blood compo- particular, if something harmful occurs, the decision of the
nents. Likewise, private accrediting organizations emphasize health-care professional may be challenged both legally and
proper performance and documentation of these activities. ethically.
The ethical principle of beneficence will be balanced
Informed Consent against autonomy in every instance. Fortunately, the ethical
In TTAIDS cases arising in the early 1980s, it was frequently principle of justice is rarely invoked because there are only
alleged that patients were insufficiently warned of the haz- rare instances in which transfusions are inequitably distrib-
ards of transfusion because transfusion experts failed to warn uted, generally because of shortage of collections. In times
hospitals and ordering physicians about them. Few cases of blood shortages, rationing begins with canceling elective
were successful because the state of scientific knowledge, es- surgical procedures for which transfusion may be indicated
tablished by expert testimony relying on published data, was and rarely reaches a point at which emergency needs are
limited. Also, the early HBV cases had established a record compromised. An example of balancing ethical principles
that supported the defense position that transfusions were that has caused legal action is when patients have suffered
already known by hospitals and other physicians to be un- serious or fatal consequences and were not given the choice
avoidably unsafe, particularly for transfusion-transmitted of donating for themselves or selecting their own blood
viral diseases. Some states (e.g., California) enacted specific donors from among family members, church or social
legislation about informed consent for transfusion. groups, or coworkers.

Medical Malpractice Emerging Issues


Several multimillion-dollar lawsuits, decided for the plaintiff, In addition to the concerns listed previously, new issues
resulted from successful allegations that either the patient focused on outcomes related to the concepts of patient
did not give adequate informed consent, did not need a blood management challenge the appropriateness of exist-
transfusion at all, could have waited until test-negative blood ing transfusion practices that are not fundamentally
was available before receiving a transfusion, or required evidence-based. These include ongoing debate surround-
transfusion solely because of something the physician did. ing the age of red blood cells when transfused, issues sur-
In all these cases, the basis for fault was negligence by the rounding provision of autologous services, use of newer
treating physician. therapies such as recombinant erythropoietin, standards
for provision of perioperative blood collection and reinfu-
Ethics and Transfusion Medicine sion, and control over the use of gene therapy to treat cer-
tain transfusion-dependent illnesses. In the TTAIDS area,
Biomedical ethical principles that must be balanced when suits have been filed for false AIDS test results and for fear
considering appropriateness of and informed consent for of AIDS exposure through transfusion or via a health-care
transfusion include autonomy, beneficence, and justice (see worker. There are also ongoing concerns over handling of
definitions in Box 29–1). look-back. New concerns about bacterial contamination
In transfusion medicine, autonomy is increasingly impor- and testing of platelets and issues surrounding emerging
tant as patients become more aware of the harm, and the transfusion-transmissible agents (e.g., babesiosis, Zika
benefit, of transfusions and insist on the right to choose the virus, and dengue fever) will surface.
6888_Ch29_607-616 29/10/18 5:07 PM Page 614

614 PART V Quality Management and Compliance

CASE STUDIES donate the platelets. The donor is shocked and embar-
rassed by the news and storms out of the center. Two
Case 29-1 weeks later, the donor sues the blood center for inten-
tional infliction of emotional distress.
A 26-year-old woman is critically injured in a two-car col-
lision. She is rushed via helicopter to the nearest trauma 1. Is the donor likely to be successful?
center, where 10 units of uncrossmatched O-negative red 2. Are there other bases on which suit can be brought?
blood cells (RBCs), 12 units of crossmatch-compatible 3. Are there established standards for preventing such an
A-negative RBCs, four A-negative apheresis platelet packs, occurrence?
and 8 units of group A fresh-frozen plasma are adminis-
Case 29-4
tered within the first 24 hours of her care. Soon after her
admission, she is taken to the operating room, where a A 59-year-old man was notified in 1999 that he had re-
splenectomy is performed, liver and left kidney lacera- ceived a unit of RBCs in 1989 that was negative by first-
tions are repaired, numerous fractures of both lower ex- generation HCV antibody testing, but the donor was
tremities are reset, and a chest tube is placed for a found to be positive when subsequently tested by second-
collapsed left lung. She requires only 4 more units of A- generation methods. He was tested by the blood center in
negative RBCs during the next several days in the inten- late 1999 and found to be HCV-positive. He filed a lawsuit
sive care unit and recovers sufficiently to be discharged against the treating physician, the hospital, and the blood
home 15 days after the accident to recuperate. Two years center in 2002.
later, during her first prenatal visit for a second pregnancy,
she tests positive for HIV. Her obstetrician tells her that 1. What legal theory can this man use?
the blood center reported that one of the units she re- 2. What is the likely outcome of this lawsuit?
ceived came from a donor who was test-positive for HIV
Case 29-5
a month ago.
1. Was the blood center negligent? A 67-year-old woman, crippled by degenerative arthritis,
2. Was the physician who transfused the patient negligent? requests that her own blood and blood from family mem-
3. Would using an “ordinary” versus a “professional” bers be used during her hip replacement surgery. The
standard of care yield a different answer? blood center describes its donation procedures, which do
not permit directed donations. Although she is disap-
Case 29-2 pointed, she agrees to donate for herself. When her first
unit is tested, it is reactive for hepatitis B surface antigen
A 43-year-old man received a 1-unit transfusion of RBCs and for antibody to hepatitis B core antigen. The patient-
during an emergency coronary artery bypass operation. donor is advised that she may no longer donate for herself
He experienced sudden unexplained hemolysis, uncon- and that the unit she has already donated will not be avail-
trollable coagulopathy, and renal shutdown, from which able for her surgery. Surgery proceeds. She receives 4 units
he could not be resuscitated despite massive efforts by the of volunteer blood and develops TTAIDS 3 years later. She
cardiac surgeon and anesthesiologist. During the course sues the blood-collecting organization.
of the resuscitation, when a new blood sample was sent
for additional crossmatch, it was discovered that the pa- 1. On what legal grounds can this suit be brought?
tient’s blood group was O-positive; the original unit of 2. What is the current standard of care regarding patient-
RBCs was A-positive. His spouse sued his cardiologist for directed donations?
lack of informed consent and negligence and sued the 3. Can/should health-care organizations deny autologous
hospital and transfusion service physician for negligence. donor-patients access to their own test-reactive blood?

1. Which issue is likely to be successful for this plaintiff? Case 29-6


2. Would the result be different if ordinary rather than a
professional standard of care were applied? On July 5, a Monday afternoon, two trauma victims, both
group O-negative, arrive in the same hospital emergency
Case 29-3 room. Both patients have massive injuries requiring large-
volume RBC transfusions. Because of the time of year, the
A 32-year-old male repeat whole blood donor is found to hospital transfusion service is low on O-negative RBCs.
be positive for HIV through nucleic acid testing, but he As the requests for RBCs rise, units crossmatched for to-
has been test-negative for HIV 1/2 antibodies and for HIV morrow’s elective surgery are taken to be used for the
p24 antigen on several prior donations. Before all testing trauma patients. The transfusion medicine physician
is completed, he returns to the blood collection center to (TMP) contacts the department of surgery to cancel elec-
donate HLA-matched platelets. At registration, the staff tive surgery cases for Tuesday, July 6. The transfusion
person notices that his prior record indicates his deferral service manager (TSM) puts in an emergency call to the
status. She informs the donor that he is not eligible to regional blood center. The blood center informs the TSM
6888_Ch29_607-616 29/10/18 5:07 PM Page 615

Chapter 29 Medicolegal and Ethical Aspects of Providing Blood Collection and Transfusion Services 615

that it, too, is short of O-negative RBCs but that it will financial support for his spouse, two college-age chil-
canvas the neighboring hospitals and put in an emergency dren, and widowed mother; the other is a 28-year-old
request to the blood exchange network. gas service attendant who is unmarried. Both patients
As several hours pass, during which the trauma sur- are critically injured, but with adequate transfusion sup-
geons and OR team work diligently to correct the injuries port, both are deemed likely to recover by their treating
and restore vital organs, it is clear that there will be suffi- physician.
cient RBCs available to adequately support only one pa-
1. Which ethical principle is being emphasized?
tient. The chair of the hospital ethics committee is
2. How will a decision favoring one patient over the
consulted for assistance in determining how to proceed.
other be viewed ethically? Legally?
Additional information available includes this: One
patient is a 55-year-old bank president who is the sole

SUMMARY CHART
 Tort liability is the basis for most lawsuits.  Under the doctrine of respondeat superior, the actions
 Federal regulations that apply specifically to blood bank- of employees are attributable to the employer or per-
ing are found in Title 21, Code of Federal Regulations. son who directs their actions; for example, in transfu-
 Claims of the intentional tort of battery is used in sion medicine, this would be a physician.
transfusion medicine when a donor or a patient claims  The concept of product liability implies that a warranty
that he or she never agreed to have the needle placed exists for virtually any product a consumer buys, and
into his or her arm. if the product fails to perform or creates harm when
 The doctrine of informed consent protects the patient none was expected, the consumer has a right to a
(or donor) by requiring that information be provided replacement or, if denied a replacement, to sue for
in a manner understandable to the patient under cir- negligent manufacturing or distribution.
cumstances that permit the patient to ask questions  Blood centers may be liable for invasion of privacy law-
and to receive answers to any questions or concerns. suits if confidentiality is breached via public disclosure
 The elements of negligence include: of embarrassing facts (e.g., HIV-positive results) or for
violation of federal law (the HIPAA).
• Duty was owed to the injured party.
• The duty was not met by the injuring party.  Traditionally, hospitals and other nonprofit organiza-
• Because the duty was not met, the injured party was tions are protected under the doctrine of charitable im-
harmed. munity from excess liability because they perform
• Failure to meet the duty owed was directly respon- charitable acts.
sible for or could have been predicted to cause the  Biomedical ethical principles that must be balanced
harm suffered by the injured party. when considering appropriateness of and informed
• Some measurable (compensable) harm occurred consent for transfusion include autonomy, benefi-
(damages). cence, and justice.

3. The reasons patients have sued for transfusion injury


Review Questions
include:
1. Transfusion-transmitted diseases can result in lawsuits a. Failure to perform surrogate testing
claiming: b. Failure to properly test blood components
c. Failure to properly screen donors
a. Battery
d. All of the above
b. Invasion of privacy
c. Negligence 4. Blood banking professionals may increase the threat of
d. a, b, and c litigation by:
2. Laws applicable to blood banking and transfusion medi- a. Following published regulations and guidelines
cine can arise: b. Knowing the legal bases for liability
c. Disclosing all information about patients and donors
a. In state and federal courts
d. Practicing good medicine
b. In the U.S. Congress, state legislatures, and state and
federal courts
c. In state legislatures and courts
d. In state legislatures and the U.S. Congress
6888_Ch29_607-616 29/10/18 5:07 PM Page 616

616 PART V Quality Management and Compliance

5. Issues about transfusion-transmitted diseases: 6. AABB. Standards for blood banks and transfusion services. 31st
edition. Bethesda, (MD): AABB; 2018.
a. Are evolving and will continue to result in litigation in
7. Kelly C, Barber JP. Legal issues in transfusion medicine: is blood
the foreseeable future banking a medical profession? Clin Lab Med. 1992;12:819.
b. Frequently result in plaintiff verdicts
c. Have all been litigated Bibliography
d. Are known and avoidable
Cooper JS, Rodrigue JE. Legal issues in transfusion medicine. Lab
Med. 1992;23:794.
References Crigger, B-J, editor. Cases in bioethics:. Selections from the Hastings
1. Randall, CH Jr. Medicolegal problems in blood transfusion. Wash- Center Report 3rd ed. New York: Bedford/St. Martin’s Press;
ington (DC): Joint Blood Council; 1962. Reprinted Chicago: 2012.
American Medical Association, Committee on Blood; 1963. Hewitt PE. Medicolegal aspects of transfusion practice. In: Murphy
2. Rabkin B, Rabkin MS. Individual and institutional liability for MF, Pamphilon DH, Heddle NM, editors. Aspects of transfusion
transfusion-acquired disease: an update. JAMA. 1986;256:2242. practice, 4th ed. John Wiley & Sons Ltd. 2013.
3. Administrative Procedure Act, 60 Statutes 237–244, 5 USC Sec- Rabkin B, Rabkin MS. Individual and institutional liability for
tions 551–559, 1988. transfusion-acquired disease: an update. JAMA. 1986;256:2242.
4. Code of Federal Regulations. Food and Drugs, Title 21, Parts Schwartz VE, Kelly K, Partlett DF. Prosser, Wade and Schwartz’s
210,211 and 600, 601, 606, 607, 610, 630, 640, 660, 820, Title Torts: cases and materials. 13th ed. Mineola, (NY): Foundation
42, Part 493. Washington (DC): U.S. Government Printing Press; 2015.
Office; 2017. Stowell C, Sazama K, editors. Informed consent. Bethesda (MD):
5. Sazama K. Practical issues in informed consent for transfusion. AABB; 2008.
Am J Clin Pathol. 1997;107:572.
6888_Answerkey_617-628 29/10/18 11:26 AM Page 617

Answer Key

Chapter 1 child’s Rh negative phenotype. The mother is known


to be Rh positive, but the blood type of the father is
Review Questions
unknown. Another possible explanation is that the
1. c 5. d 9. b 13. d mother inherited one RHD gene and the father is Rh neg-
2. c 6. c 10. d 14. a ative. Neither parent would be required to be Rh negative
3. b 7. d 11. d 15. b (as in answers a and b) for child 2 to be Rh negative.
4. c 8. a 12. a Review Questions
1. d 5. b 9. c 13. d
2. b 6. b 10. a 14. c
Chapter 2
3. a 7. c 11. b 15. d
Case Study 2-1
4. d 8. d 12. b 16. c
1. d. The offspring inherit an ABO gene from each parent.
Assuming the inheritance of normal ABO genes, with a
group A mother and offspring that are group A (1st child) Chapter 3
and group O (2nd child), the mother could only be geno-
type AO, not AA. If she were genotype AA, she would Review Questions
have contributed an A gene to the 2nd child. Since 1. a 6. c 11. a 16. d
the 2nd child is group O (genotype OO), the father could 2. c 7. b 12. d 17. a
be genotype AO, BO, or OO and this would explain the
3. b 8. d 13. c 18. b
possibility of both group A and group O offspring. These
are the possible genotypes for each family member: 4. d 9. c 14. a 19. d
5. d 10. a 15. b
Mother Father Child 1 Child 2

Phenotype A Not tested A O Chapter 4


Possible Case 4-1
A, B, or O 1. The anti-C was an alloantibody. The anti-C was identified
Possible AO AO AA or AO OO in the autoadsorbed plasma, meaning the antibody speci-
Genotype(s) ficity did not match an antigenic determinant on the
patient’s RBCs. The antibody was misidentified as an
autoantibody because the patient serologically typed as
AO BO AO OO C positive. The r′S haplotype has an SNP that codes for
valine as the 245 amino acid and produces a partial C
antigen. Patient’s that are e and VS positive may have
a weakened e if the valine 245 is in cis with the alanine
AO OO AO OO 226. These patients may become alloimmune to the e
antigen.
2. This patient should receive both E and C negative units.
Case 4-2
1. The HSCT recipient is D–, E–, whereas the donor is D+,
2. c. If both parents express a D antigen on their red cells, E+. The recipient immune system produced the anti-E
it is still possible for the offspring to have D negative red and the anti-D during his relapse.
cells if each parent inherited only one copy of the RHD 2. The IC may have been due to a dual cell population of
gene. With the absence of the RHD gene on the other both the donor and recipient. The recipient was negative
haplotype, if each of them contributed the haplotype for the E and positive for the N SNPs, whereas the donor
without RHD to the 2nd child, that would explain the was positive for the E, and negative for the N SNPs.

617
6888_Answerkey_617-628 29/10/18 11:26 AM Page 618

618 Answer Key

Case 4-3 Although anti-A1 is considered to be nonreactive at 37°C,


1. The first platform used allele-specific probes to predict it was decided to transfuse type O–positive cells to this
the patient’s Kidd status. Because the antithetical Jka and patient since there was ample supply.
Jkb antigens are determined by only one SNP, the probe Review Question
only analyzed the 838G>A SNP. The SNP was adenine,
1. d 4. b 7. a 10. b
and the software called the patient Jkb positive. The se-
quencing identified the c.1038 deletion that caused a stop 2. a. 5. c 8. a
codon in the coding sequence of the final transmembrane 3. d. 6. b 9. c
region, most likely causing a truncated protein to be im-
properly expressed on the membrane surface. This is why
the patient’s immune system identified the Jkb antigen as Chapter 7
foreign.
Case 7-1
Review Questions 1. The patient’s ABO/Rh is O Rh-negative.
1. a 5. d 9. c 13. c 2. The antibody screen is positive at the indirect antiglobu-
2. c 6. a 10. b lin test (IAT) phase with the Rh-positive screening cells.
3. Anti-D is present in the patient’s plasma. All D-positive
3. c 7. d 11. b
cells are reactive 2+ in both the screen and the antibody
4. b 8. d 12. d ID panel cells, and all D-negative cells are nonreactive. In
addition, all other common alloantibodies are ruled out
Chapter 5 using at least one double-dose reagent cell.
4. Select RBCs for transfusion that are O-negative, cross-
Review Questions match compatible.
1. d 5. b 9. b 13. a
Case 7-2
2. c 6. b 10. a 14. d
1. The patient’s ABO/RH is O-Positive
3. a 7. d 11. c 15. b 2. Anti-c and anti-E are present. Panel cell numbers 1, 2, 3,
4. a 8. a 12. c 16. d and 11 are c+E- and react 2+ at IAT. Cell numbers 8, 9,
and 10 are c-E+ and react 2+ at IAT. Cells 4, 5, 6 and 7 are
c-E- and are nonreactive at IAT. In addition, the patient
Chapter 6 lacks both the c and E antigens, and the patient serum
tested with the patient’s own cells is negative at IAT.
Case 6-1
3. In this case study, antibodies are excluded ONLY if the
1. The results indicate an ABO discrepancy in the reverse patient’s plasma does not react with panel cells that pos-
grouping since there is only a 2+ reaction with the reagent sess a double-dose expression of the antigen (homozy-
A1 red blood cells. Normally, ABO forward and reverse gous expression).
testing show strong 3+ to 4+ reactions. 4. Test the patient’s plasma against selected cells that are
2. Since the antibody screen was negative, the blood bank c-E-K+k-.
technologist proceeded to type the patient’s RBCs with 5. The most probable genotype is DCe/DCE in Fisher-Race
anti-A1 lectin, Dolichos biflorus. This was decided be- and R1R1 in Weiner nomenclature.
cause, based on the negative antibody screen and patient 6. The patient’s antigen type confirms the antibody identi-
history, an alloantibody was most likely not causing the fication in terms of the anti-c and anti-E as the patient
ABO discrepancy. lacks both c and E antigens. The patient is K-, so he is
3. The reaction with anti-A1 lectin indicates the patient’s capable of producing anti-K and the antibody should be
cells are not type A1. The additional serum studies ruled out.
show that the antibody is not directed toward a possible 7. In the event of transfusion, select O-positive crossmatch
alloantibody reacting at room temperature and that the compatible RBCs that are c-, E-.
antibody is not anti-A, in that only anti-A would react
with A2 cells. If a cold autoantibody or alloantibody was Case 7-3
causing the discrepancy, the patient’s serum would likely 1. Patient’s ABO/Rh is O-negative. Weak D testing is not
react with the reagent O cells. Results indicate the patient required for patients.
is a likely group A2 and has formed an anti-A1. 2. It appears that there may be anti-D and anti-C. However,
4. During surgery, the OR ordered 2 units of packed RBCs. we cannot rule out the possibility that an anti-G exists.
Two type O-positive packed RBCs were crossmatched Cell numbers 2, 3, and 4 are D+C- and react 2+ at IAT.
with the patient serum and found to be compatible. Only Cell numbers 7 and 8 are D-C+ and react 2+ at IAT. Cell
1 unit was transfused without episode. The patient was numbers 1 and 5 are D+C+ and react 2+ at IAT. Cell num-
discharged 4 days later. In this case, there was an ABO bers 6, 9, 10, and 11 are D-C- and are all nonreactive at
discrepancy caused by anti-A1 in the reverse grouping. IAT. In addition the patient cells, which are D-C-, also are
6888_Answerkey_617-628 29/10/18 11:26 AM Page 619

Answer Key 619

nonreactive at IAT. The D antigen resides on cells that disease. Fetal anemia in anti-K HDFN is associated
have D and/or C antigens. Therefore an anti-D can react with suppression of erythropoiesis due to destruction
with cells that have G, D, or C antigens. It is possible that of erythroid precursor cells, which can be additional to
there is anti-G alone; an anti-G with an anti-D; an anti-G destruction of circulating antigen-positive RBCs. Kell
with anti-C; an anti-G with both anti-D and anti-C; or glycoprotein is expressed on fetal RBCs at a much earlier
two separate antibodies anti-D and anti-C without the stage of erythropoiesis than Rh antigens.
anti-G present.
Case 8-2
3. All other common blood group alloantibodies are ruled
out using double-dose cells. 1. The patient’s ABO, Rh is O Rh-positive, with the antibody
4. Absorption and elution studies are indicated to distin- test (screen) showing 3+ reactivity in both cells.
guish and identify distinct antibody specificities using 2. Due to the negative auto control, the antibody appears to
cells that are D+C- and D-C+ in separate tests to identify be an alloantibody.
the anti-G. 3. Based on the phenotype, the ethnicity of the patient
5. This is an obstetric patient, so it is important to know if appears to be African American due to the increased
she has an anti-D or anti-G. Since she is Rh-negative, she frequency of ccDee; Fy(a-b-); Le(a-b-); and S-s- in the
should be given Rh-immune globulin to prevent anti-D African American population.
HDFN if she has not already produced an anti-D. How- 4. Based on the patient’s phenotype, anti-U should be con-
ever, if there is a G present, it can mislead the technologist sidered.
to conclude that the patient has made a separate anti-D 5. Genotype testing can be used to detect the presence of
and anti-C that could result in the patient not being of- the GATA gene. The GATA gene disrupts the binding
fered Rh Immune Globulin. In addition, if the patient has site for mRNA transcription in the RBC. Consequently,
an anti-G and the fetus has either the D, C, or G antigens, Fy(a–b–) blacks do not express Fyb on their RBCs
there is a real risk of HDFN caused by the anti-G. Anti-G but rather express Fyb in other tissues. The presence of
is IgG class, so it can readily cross the placenta. Fyb in tissues presumably precludes the recognition
of Fyb as foreign; thus, no anti-Fyb is made by these
Review Questions individuals.
1. a 4. a 7. d 10. c 6. Adsorption of the anti-U using cell phenotypically similar
2. c 5. c 8. c to the patient except positive for S and/or s antigen will
allow for the rule-outs of the remaining alloantibodies.
3. b 6. c 9. a
7. The first 26 amino acids on GPB are identical to the first
11. 26 amino acids on the N form of GPA. The N activity of
GPB is denoted as ‘N’ (N-quotes) to distinguish it from
Weiner Fisher-Race Rosenfield Antigens Present
the N activity of GPA. Because the M+N-S-s- phenotype
a. R1r DCe/ce RH: 1,2,-3,4,5 D, C, c, e lacks both the N from GPA and the ‘N’ from GPB, a potent
anti-N can be made by the rare individual whose RBC
b. R2R0 DcE/Dce RH: 1,-2,3,4,5 D, E, c, e
type is M+N–S–s–.
c. RzR1 DCE/DCe RH: 1,2,3,-4,5 D, C, E, e
Review Questions
d. r⬘r Ce/ce RH: -1, 2, -3, 4, 5 C, c, e 1. d 7. a 13. d 19. c
2. b 8. d 14. c 20. d
12. d 3. a 9. b 15. c 21. b
13. a 4. c 10. c 16. b 22. a
14. c 5. a 11. a 17. c 23. c
15. b 6. d 12. c 18. b

Chapter 8 Chapter 9
Case 8-1 Case 9-1
1. The patient’s ABO, Rh is A Rh-positive, with the antibody 1. The patient’s ABO, Rh is B Rh-positive with the antibody
test (screen) showing a 2+ reactivity on screening cell test (screen) showing 1+ reactivity on screening cell num-
number one. ber one.
2. The antibody identified with panel testing is an anti-K. 2. All the common alloantibodies have been ruled out.
3. The K antigen can be detected on fetal RBCs as early as 3. The removal of reactivity by chloroquine treatment of the
10 weeks and is well developed at birth. cell is consistent with an antibody to the HLA antigen
4. Anti-K is also associated with severe HDFN. The antibody known as anti-Bg. Anti-Bg antibodies are common in
titer does not always accurately predict the severity of multiparous women.
6888_Answerkey_617-628 29/10/18 11:26 AM Page 620

620 Answer Key

4. Due to the increased expression of HLA class I antigens Lua would have enhanced antigen expression. K, Kpa, and
on platelets, Bg antibodies can be adsorbed from the Jsa would be unaffected by routine enzymes.
serum/plasma using platelet concentrate. 3. The specificity of the antibodies was unclear after the ini-
5. Bg antibodies are usually considered insignificant, but tial panel. After repeating the panel using ficin-treated
although rare, Bg antibodies have been reported to cause cells, it appears that the antibodies present are anti-K and
immediate and delayed HTRs. HLA antibodies have not anti-Fyb. The enzyme treatment destroyed the Fyb anti-
been implicated in HDFN. gens, thereby eliminating the anti-Fyb reactivity, which
6. Since Bg antibody was detected in this patient’s specimen allows the anti-K to present clearly. Anti-C and anti-Jka
at the antiglobulin phase, units that are crossmatch- were also not eliminated by the initial panel; however,
compatible at the antiglobulin phase should be selected one would expect these antibodies to demonstrate
for transfusion, if needed. enhanced reactivity with the ficin-treated cells. Anti-Jka
7. HLA antibodies play a significant role in transfusion- may be excluded using cell 8 of the ficin panel. Whereas
associated acute lung injury (TRALI). anti-C cannot be excluded using a homozygous cell,
however, the pattern of reactivity and lack of response to
Case 9-2
enzyme treatment suggest it is not present in this sample.
1. The patient’s ABO, Rh is O Rh-negative with a negative 4. Testing a C+, c–, K–, Fyb– cell would be necessary to
antibody screen. exclude anti-C.
2. A DAT and AHG phase crossmatch should be performed
on the transfused unit suspected in the transfusion Case 10-2
reaction. 1. The positive reactions on this panel are all the same
3. Since the panel and antibody screen was negative, the pres- strength. Cell 10, the only cell that failed to react with
ence of a high-prevalence antibody can be ruled out. The the patient’s serum, has been identified as being Yta
antibody specificity is likely to a low-prevalence antigen. negative. Since Yta is a high-prevalence antigen, one may
4. The likely antibody is anti-Wra. Anti-Wra is a relatively be suspicious that this is the antibody present in this
common antibody in donors and patients; some are di- specimen.
rectly agglutinating, but most require the antiglobulin 2. For this case, the ideal solution is to test two additional
phase of testing to be detected. Anti-Wra has caused se- Yta-negative cells, which would provide 95% confidence
vere HTRs. Knb is a low-prevalence antigen, and Knops for correct antibody identification and allow for addi-
system antibodies are usually clinically insignificant for tional exclusions. If antigen-negative cells are not readily
both transfusion and HDFN. Both Wrb and Gya antigens available, it may be necessary to consult a reference
are high-prevalence antigens that should have been laboratory that maintains a stock of rare cells. Any other
detected on the panel testing. antibodies not excluded following this testing may
5. An antiglobulin crossmatch needs to be performed on any require patient phenotyping or absorption studies to con-
additional units. firm their presence or absence. The services of a reference
laboratory may be required to antigen-type the patient for
Review Questions
the Yta antigen.
1. d 6. b 11. c 16. a 3. When working up any antibody identification, it is good
2. c 7. d 12. c 17. a practice to get the patient’s history of transfusions, trans-
3. c 8. c 13. d 18. d plantations, and if female, pregnancies. A list of medica-
tions and the patient’s diagnosis may prove helpful.
4. d 9. a 14. a 19. c
Knowing the ethnicity of the patient may also give clues
5. b 10. d 15. c 20. a as to the identity of the antibody, as antigen frequencies
vary among races. One example of a high-prevalence
antigen associated with a specific ethnicity is the U anti-
Chapter 10 gen, which is present in virtually all whites and all but
approximately 1% of blacks. Anti-U is produced mainly
Case 10-1
in multiparous black females or those patients who have
1. There is inconsistent reactivity (1+ to 3+), which suggests been repeatedly transfused.
more than one antibody may be present. The pattern of
reactivity appears to fit anti-Fyb; however, one homozy- Case 10-3
gous cell (cell 2) reacts 3+ while other homozygous 1. The low-prevalence antigen that is found on cell 4 is Jsa.
cells (cells 8 and 9) react only 2+. This inconsistency is 2. Testing two additional Jsa antigen-positive cells will be
seen in the cells with heterozygous antigen expression required to confirm the specificity of the antibody (the
as well. Cell 7 reacts 2+, while cells 1, 6, and 11 react 3 and 3 rule); all other antibody specificities have been
only 1+. excluded, with the exception of anti-Lua, which is
2. Anti-C, -Cw, -K, -Kpa, -Jsa, -Fyb, -Jka, and -Lua have not also an antibody to a low-prevalence antigen. Referring
been excluded. Fyb antigens would be destroyed or to Figure 10-14, none of the panel cells express the
diminished after treatment with enzymes. C, Cw, Jka, and Lua antigen, hence anti-Lua, if present, would not be
6888_Answerkey_617-628 29/10/18 11:26 AM Page 621

Answer Key 621

detected with this panel. If the patient has not been Kell, LW, Dombrock, Knops, and Cromer antigens. Once
transfused within the last 3 months and antisera is avail- the autoantibody has been removed from the cells, the
able, phenotyping the patient for the Jsa antigen should treated cells can be incubated with the patient’s serum at
also be performed. A reference laboratory may need to 37°C to absorb autoantibody from the serum. Multiple
be consulted if additional antigen-positive Jsa panel cells steps are necessary to treat the cells and adsorb the
or antisera are not readily available. autoantibody, which makes this procedure very time-
consuming. Once the autoantibody is removed from the
Case 10-4
serum, it can be tested for underlying alloantibodies.
1. All cells (except the I negative cord blood cells) are When patient cells are limited or when the patient has
positive at the immediate spin phase of testing, and the been recently transfused, allogeneic cells are used for
autocontrol is positive. The reactivity does not persist adsorption. The cells used for allogeneic adsorption
into the AHG phase of testing. (phenotypically matched or differential) are usually treated
2. The use of cord blood cells, which lack the I antigen, con- with enzymes or with ZZAP to enhance expression of
firms the presence of anti-I in this sample. certain antigens and facilitate autoantibody removal.
3. Many laboratories avoid detection of cold autoantibodies 4. The pattern of reactivity in the “Absorbed serum” column
by omitting the immediate spin phase of the antibody of Figure 10–12 matches that of anti-K. Positive reactions
screen (and panel) and by using monospecific anti-IgG with the autoabsorbed serum were observed with cells 2,
Coombs’ reagent. 6, and 10, which are positive for the K antigen. All other
One of the least complex methods used to prevent cold antibody specificities are excluded except for Cw, Kpa, Jsa,
autoantibodies from interfering in antibody detection or and Lua. These are low-prevalence antigens and generally
identification tests at the AHG phase of testing is the pre- do not need to be excluded. Additional phenotyping
warm technique. strategies should be employed to determine if the patient
A more complex method, discussed previously, is ad- is negative for the K antigen.
sorption. The patient’s autologous cells may be incubated 5. RBC units negative for the K antigen, which are least in-
with the patient’s serum at 4°C to remove the autoantibod- compatible with unabsorbed serum at the antiglobulin
ies before antibody detection steps are performed. This crossmatch, should be provided for transfusion.
method provides autoantibody-free serum for antibody de-
tection and identification procedures. If the autoantibody Review Questions
is particularly strong or if the patient has been recently 1. d 3. c 5. c 7. d
transfused (within the last 3 months), RESt or allogeneic 2. a 4. b 6. c
adsorbing cells may be used to perform the adsorption in-
stead of the patient’s RBCs. RESt-adsorbed serum is not
suitable for crossmatch as anti-B may be removed.
Chapter 11
Case 10-5
Case Study 11-1
1. Warm autoantibody with an underlying alloantibody. The
1. Select an O-negative red blood cell unit.
reactivity with all cells, including the autocontrol, at the
2. Perform an immediate spin crossmatch to detect ABO
AHG phase of testing is typical for a warm autoantibody.
incompatibilities.
The increased reactivity on cells 2, 6, and 10 suggests that
an additional antibody may be present. With the patient’s Case Study 11-2
transfusion history, one must consider the possible pres- 1. Select an O-positive, K-negative red blood cell unit.
ence of an alloantibody. 2. Select an O-negative, K-negative unit or an O-negative
2. Typically, with an antibody to a high-prevalence antigen, unit compatible at the antiglobulin phase of testing with
the autocontrol will be negative. However, if the patient either the infant’s or mother’s plasma.
has been recently transfused with RBCs that possess the
high-prevalence antigen, the autocontrol may be positive Review Questions
as the antibody will bind (in vivo) antigen-positive donor 1. d 5. b 9. d 13. c
RBCs remaining in circulation. 2. a 6. b 10. b
3. Autoadsorption is the method of choice when the patient
3. b 7. c 11. e
has not been recently transfused (transfusion in the past
3 months), and when a sufficient quantity of autologous 4. c 8. a 12. c
RBCs are available. The patient’s RBCs should first be
stripped of autoantibody before they are used to adsorb
autoantibody from the serum. Partial elution using chem- Chapter 12
ical methods is used to produce an antibody-free cell.
Review Questions
ZZAP is one chemical that works particularly well for
this. It is composed of DTT and papain, resulting in the 1. a 3. d 5. a 7. c
destruction of enzyme-sensitive antigens and denaturing 2. d 4. b 6. b 8. c
6888_Answerkey_617-628 29/10/18 11:26 AM Page 622

622 Answer Key

Chapter 13 whether this is the first surgical procedure on this hip


(redo surgery uses more blood), and whether the patient
Case Study 13-1 has any pretransfusion compatibility problems.
1. Yes, if the implicated unit was transfused in December 2. Considerations should be the patient’s general health, he-
2009, that would fall within the 6-week incubation moglobin and hematocrit levels, amount of time between
period. request for donation and the time of surgery, and whether
2. Symptoms include fever, chills, fatigue, joint and muscle the patient has infectious diseases that might interfere
aches, and headache. (e.g., bacterial infections).
3. No, they are more strict than the current AABB require- 3. Tests should include PT, PTT, and platelet count. If these
ments, which is indefinite deferral. tests are normal and von Willebrand’s disease is sus-
4. The donor must be tested for Babesia antibodies to deter- pected, vWF workup should be considered.
mine if the donor is infected, and then a decision must 4. If the patient has moderate to severe von Willebrand’s
be made regarding eligibility to donate. disease, the patient may need factor VIII concentrate
known to contain vWF.
Case Study 13-2
1. The donor would be tested for T. cruzi infection using a Case 16-2
licensed test. Donors whose blood tests positive or inde- 1. Both answers can be justified. If the answer is yes, her
terminate on a licensed supplemental test would be per- hemoglobin is close to 6 g/dL, which is the criterion for
manently deferred; if the test is negative, the donor would RBC transfusion. She will be less tired and weak if she
be considered for reentry into the donor pool. receives a transfusion. If the answer is no, she is not in
2. Chagas disease any acute distress. She is iron deficient (from chronic
blood loss) and should be treated with iron replacement
Review Questions rather than with transfusion, which has higher risk of
1. c 5. b 9. c 13. b complications. The safest, least-expensive, and the most
2. b 6. e 10. a 14. a effective long-term strategy is to prescribe iron and not
3. a 7. b 11. c 15. a transfuse.
2. For adults, each unit of blood should increase the hemo-
4. a 8. a 12. a globin 1 g/dL and the hematocrit 3 percentage points.
For accuracy, this is based on a hypothetical 70-kg man.
For smaller men and women, the increase is greater; for
Chapter 14 larger ones, less.
Review Questions Case 16-3
1. a 8. b 15. a 22. c 1. The platelet count is 5,000/µL; therefore, a platelet trans-
2. b 9. d 16. c 23. a fusion is indicated. Without the platelet transfusion, she
3. d 10. c 17. a 24. d would be at increased risk of bleeding from the site of
4. a 11. c 18. d 25. b the bone marrow biopsy. Although the hemoglobin and
hematocrit levels are lower than normal for a woman of
5. c 12. d 19. c this age, an RBC transfusion is not indicated (hemoglo-
6. b 13. a 20. d bin greater than 6 g/dL in an otherwise healthy young
7. c 14. d 21. b adult).
2. For an adult, each unit (bag) of platelet concentrate
should increase the patient’s platelet count by 5,000
Chapter 15 to 10,000/µL. The platelet count should be at about
20,000/µL because a bone marrow biopsy is an invasive
Review Questions procedure. Thus, one unit of platelets (plateletpheresis or
1. a 5. c 9. a 13. c pool) would be indicated.
2. b 6. e 10. c 3. If it is anticipated that the patient will experience bone
marrow recovery soon (after the chemotherapy), no RBC
3. c 7. b 11. a
transfusion is indicated. If, however, the bone marrow
4. e 8. b 12. b will be suppressed for several weeks, 1 or 2 units are
indicated, with a recheck in a couple weeks for another
possible transfusion. Each unit of RBCs is expected to
Chapter 16 increase the hemoglobin level about 1 g/dL.
Case 16-1 Case 16-4
1. It is necessary to know the patient’s hemoglobin and 1. The blood group for RBC transfusions is O, which is the
hematocrit levels, the surgeon’s usual blood usage, universal blood group.
6888_Answerkey_617-628 29/10/18 11:26 AM Page 623

Answer Key 623

2. Group AB should be selected for plasma and platelet Review Questions


transfusion. If AB platelets are not available, any other 1. c 5. c 9. d 13. b
blood group can be selected.
3. RBCs, plasma, and platelets should be in the amount 2. c 6. b 10. d 14. d
specified by your massive transfusion protocol. 3. b 7. c 11. d 15. d
4. RBCS, plasma, and platelets should be B. The Rh can be 4. b 8. b 12. a
positive because the patient is male. If group B RBCs are
in short supply, group O can continue to be used.
5. Workup of the positive antibody screen should start Chapter 18
as soon as possible, but without delaying provision
of products for the massive transfusion. Transfusing Case 18-1
RBCs in massive hemorrhage is more important than ob- 1. Top five possible diagnoses for this case are:
taining “compatible” units. With massive transfusion, the • Hemolytic uremic syndrome.
patient’s antibody will be removed and diluted by the • Thrombotic thrombocytopenic purpura (TTP).
hemorrhage and the replacement of blood components. • Disseminated intravascular coagulation (DIC).
The “incompatible” RBCs will be removed more slowly • Malignant hypertension.
because the mechanism is extravascular hemolysis. When • Sepsis.
the serologic problem is solved, compatible RBC units can 2. Thrombotic thrombocytopenic purpura (TTP) is the
be obtained and transfused. most likely diagnosis. The patient has normal coagulation
studies (DIC is excluded). There is evidence of hemolysis
Review Questions
(elevated LDH, decreased haptoglobin), and the platelet
1. d 4. c 7. a 10. a count is decreased, with the presence of schistocytes on
2. c 5. a 8. b the peripheral smear. The ADAMTS13 level is signifi-
3. c 6. a 9. d cantly decreased.
3. In classic cases, patients present with a pentad of clinical
and laboratory findings:
Chapter 17 • Microangiopathic hemolytic anemia (MAHA)
• Thrombocytopenia
Case 17-1
• Neurological symptoms and signs
1. A reaction in which signs and symptoms occur during or • Renal function abnormalities
within 24 hours of transfusion. • Fever
2. Hemolysis. 4. Main treatment is plasma exchange with fresh frozen
3. Volume of incompatible blood transfused. plasma (FFP).
4. Look for matching or discrepant information between the
information on the labels on the pre- and post-transfusion Review Questions
samples, transfused units, and patient transfusion service 1. d 4. a 7. b 10. d
records. 2. a 5. c 8. a
Case 17-2 3. b 6. d 9. c
1. Communication with other health professionals served
as the key to resolving this incident.
2. Chemical or mechanical damage such as improper ship- Chapter 19
ping, storage temperatures, incomplete deglycerolization
Case Study 19-1
of frozen red blood cells, inappropriate needle bore size
for transfusion rapid infusion, improper use of blood 1. This is a bidirectional ABO mismatch. The blood group
warmers, unapproved fluid infusion. for RBC transfusions is O, which is the only blood group
compatible with both the donor and the recipient. If
Case 17-3 group A is chosen, the cells will be hemolyzed or have
1. Laboratory workup for the diagnosis of TAS must rule a shortened life span because of the preexisting anti-A in
out hemolysis and perform a Gram stain and culture of the recipient. If group B is chosen, then the engraftment
the implicated component and patient. The culture will be difficult to determine.
must be obtained from the container and not the seg- 2. Group AB FFP and platelets should be selected for trans-
ments attached to the product. Blood cultures from the fusion. If group A is selected, then the anti-B in the com-
patient should be drawn from a site other than the site ponent plasma is incompatible with the recipient’s B
of transfusion. antigens on the RBC and tissues. The anti-A in group B
2. Symptoms implicated with TAS include a fever with products is also incompatible with the donor RBCs and,
greater than 2°C increase in body temperature, rigors, and if selected, may contribute to delayed engraftment of RBC
hypotension. precursors.
6888_Answerkey_617-628 29/10/18 11:26 AM Page 624

624 Answer Key

3. On day 147, the ABO should be interpreted as Group B. 2. The titer at 8 weeks is 128, which is a critical value. The
While there is a weakened mixed field result in the for- antibody titer should be repeated every 4 weeks.
ward typing, there is still demonstrable B antigen as well 3. The fetus is only 8 weeks’ gestational age, so it is too early
as anti-A in the reverse type. Monitoring the RBC ABO in the pregnancy to perform any intervention such as
antigens and plasma ABO antibodies will help inform MCA-PSV, cordocentesis, or intrauterine transfusion. The
transfusion decisions. earliest an intervention can take place with the fetus is
4. The blood group interpretation on day 156 is inconclu- 16 weeks’ gestational age.
sive. The sample has a forward type of group O and a 4. The MCA-PSV should be performed at 16 to 18 weeks’
reverse type of group B. This is an expected result gestational age to determine if the test is positive.
as the recipient RBCs slowly diminish and the group A 5. The mother is NOT a candidate for RHIG. The purpose
HPC engrafts. The recipient’s B antigens are no longer of prenatal administration of RHIG is to prevent a mother
detectable, but there is still anti-A showing in the re- from producing anti-D. This mother has already pro-
verse typing. It is unclear as to the source of the anti-A duced the anti-D; therefore, there is no clinical value to
because it could be from the recipient or anti-A administer RHIG.
in ABO incompatible platelet transfusions. On day 200,
Case 20-3
the ABO is not normal, but should be interpreted
as group A. In the forward grouping, the weak result 1. The most probable cause of the second child being
with anti-A shows that donor A antigens are detectable affected is ABO HDFN. Group O mothers produce anti-
because the HPC has engrafted and is producing group IgG Anti-A,B which can cross the placenta. The mother
A RBCs in the patient. The reverse type is negative, is RhD positive, which rules anti-D as a likely cause.
because the recipient is no longer producing anti-A 2. No. Anti-Lea does not cause HDFN because the Lewis
and the HPC engraftment has not resulted in produc- antigens are poorly developed at birth.
tion of ABO antibodies from the recipient (in this case,
Case 20-4
anti-B). The change in ABO type represents the patient
moving into transplant phase III in which the donated 1. The blood types of the cord blood and the heel stick spec-
HCPs are becoming fully engrafted and functional. imens are different A negative cord blood versus O nega-
tive heel stick. The severely anemic infant appears to have
Review Questions HDFN. The DAT is only +/- on the cord blood sample,
1. a 4. c 7. b 10. a but the hemoglobin result is very low (4.3g/dL). The
2. d 5. c 8. b ABO group performed at 4oC shows what is likely the
baby’s true ABO type: group B. Incubating at 4oC can en-
3. d 6. c 9. a
hance the reactions for ABO antigens. Although the DAT
is weak (+/-) an eluate should be performed on the in-
fant’s RBCs. Recall that DAT strength is not an indicator
Chapter 20 of severity of HDFN. So further testing must include an
Case 20-1 elution. The infant is anemic because of maternal anti-
1. The most probable cause is that the mother received pro- body, most likely anti-D. The titer of the anti-D in the
phylactic Rh Immune Globulin (RHIG) at 28 weeks’ ges- mother is 256, which is clinically significant and suggests
tation. The antibody screen was negative at 28 weeks’ that this child could be RhD positive. Maternal IgG class
gestation prior to the administration of RHIG. The anti- anti-D crosses the placenta to bind to RhD positive RBCs,
body titer of anti-D due to RHIG is typically less than 1:4. which are removed extravascularly, resulting in anemia.
You can confirm this suspicion by asking the patient, or That is why the intrauterine transfusion was necessary.
her health-care provider if the RHIG was administered at 2. The Kleihauer Betke test measures the presence of fetal
28 weeks. cells and is expressed as a ratio of fetal cells/adult cells. If
2. Yes, unless the newborn infant is RHD negative a cord blood sample is collected correctly, then there
should be a very high percentage of fetal cells and we
Case 20-2 would not expect to see a result of 0/1,000. This result
1. The maternal history and the fact that the father is indicates that the cord blood was not collected properly.
the same for all pregnancies suggest that the fetus is Cord blood sample should be collected by needle and
RhD positive. HDFN due to anti-D is RBC stimulated syringe from the umbilical cord vein. Collecting the
so the first pregnancy would have been unaffected. In specimen by allowing blood from the placenta or cord to
the second pregnancy, although there is no mention of drip into the specimen tube can contaminate the sample
a positive antibody screen, the mother may have pro- with maternal blood. In this case the specimen tube
duced a very low titer anti-D which was enough to labeled as “cord blood” could either be a mislabeled sam-
cause a weak positive DAT but not high enough to ple collected from the mother, or it could be the result of
cause the baby to be affected. The third pregnancy an improperly collected cord blood sample. The
was stillborn. Anti-D HDFN can cause fetal death if left Kleibauer-Betke result for the heel stick sample does
untreated. show fetal cells, although the percentage is lower than
6888_Answerkey_617-628 29/10/18 11:26 AM Page 625

Answer Key 625

what we would expect from a normal heel stick, which anti-B is present, but there is no anti-A. This resolves the
should be all baby cells. However, this infant received in- ABO serologic discrepancy. The antibody screening per-
trauterine transfusions 3 weeks prior. The RBCs used for formed with the cold autoadsorbed serum also confirms
intrauterine transfusion would be collected from an adult that group O RBCs are nonreactive and that there is no
blood donor, which explains the low count of fetal cells. alloantibody present.
In addition, the RBCs used for intrauterine transfusion 6. The cold autoadsorbed serum is suitable for cross-
would be group O Rh-negative, which would help to ex- matching group A RBCs for transfusion (if still deemed
plain the problems with performing the ABO on this in- necessary).
fant and the weak D typing. As discussed in this chapter, 7. A cold agglutinin titer and thermal amplitude would be
transfusion may cause suppression of fetal erythropoiesis very useful to investigate whether the patient has cold
resulting in the production of few fetal RBCs. The fetal B hemagglutinin disease (CHD), as suggested by the pre-
antigen is only detectable after an incubation at 4oC. The liminary testing and by his symptoms. If a diagnosis of
eluate on the baby’s cells shows an anti-D, which is the CHD were made, such testing might avoid having to
likely cause of this HDFN. endure the bone marrow biopsy.
Review Questions Case 21-3
1. c 4. d 7. b 10. b 1. The patient’s RBCs are group B Rh-positive. There is noth-
2. a 5. c 8. c ing unusual about the blood grouping.
2. The antibody screening is negative, indicating no alloanti-
3. b 6. d 9. a
bodies present.
3. Because the antibody screening is negative, crossmatches
of the patient’s serum with three units of group B Rh-
Chapter 21 positive units of red cells would be expected to be com-
Case 21-1 patible. The positive auto control and the color of the
1. The patient is group O Rh-positive. patient’s serum are unexpected. Although there is no history
2. Her antibody screening is positive with all three screening of recent transfusion, a DAT could be helpful if the patient
cells, as well as with the autologous control, at the indi- is anemic due to an autoimmune process. If the DAT is
rect antiglobulin phase. None of the 4 units crossmatched positive, preparing and testing an eluate of the patient’s
is compatible. There is some variability in the strength of RBCs could be informative.
the IAT reactivity. Because the patient has no history 4. The DAT shows that the patient’s RBCs are coated with
of recent transfusion (within the last 3 months) the pos- IgG and complement. These results could be seen in a
itive auto control by IAT suggests a warm autoantibody normal individual. (Approximately 1 in 1,000 healthy
is present. The variability of reactivity suggests alloanti- blood donors have positive DATs.) But because this
body may also be present. patient has severe anemia that developed in 1 week and
3. A direct antiglobulin test should be done. Warm auto- frank hemolysis noted in the sample, there is likely to be
adsorption of the patient’s serum should also be done, more clinical significance to the DAT results.
and an antibody identification panel tested with the 5. The eluate is nonreactive with the three antibody detec-
adsorbed serum. tion cells, but reacts with the patient’s own RBCs even
after they have been EGA-treated to remove in vivo IgG.
Case 21-2 The results are consistent with a drug-induced antibody
1. There is an ABO serologic discrepancy and a positive that reacts with the patient’s drug-coated RBCs. Typical
albumin control, so ABO/Rh results are invalid. results seen in cases like this, called “penicillin-type”
2. The antibody screen shows a strongly reactive immediate drug-induced immune hemolytic anemia, are a negative
spin (room temperature) reading and weak reactivity at antibody screen and negative eluate in a patient with a
LISS 37C phase. These preliminary results suggest a cold positive DAT and clinical signs of acute hemolysis. In
agglutinin. Because the patient’s RBCs are also reactive these cases, adsorption of the drug by the patient’s RBCs
with his own serum, this appears to be a cold autoanti- causes them to react selectively with the drug antibody
body, which would also affect the ABO serum typing in the patient’s serum and/or eluate.
(tested at room temperature). 6. Determining what drug the patient has been receiving is
3. The patient’s RBCs are complement coated. key to knowing what drug antibody to suspect. In this
4. The most likely cause of these atypical results is a cold case, the patient was unaware of taking any drugs or an-
autoantibody. To obtain valid ABO/Rh typing results, the tibiotics during or following her hospitalization for labor
patient’s RBCs should be warmed to 37°C and washed and delivery by C-section. A careful review of the surgical
with warm saline. His serum/plasma should be cold au- record revealed that she was given a single dose of cefote-
toadsorbed and used for repeating the ABO serum typing tan during surgery, presumably to prevent postoperative
and antibody screen. infection.
5. The patient’s warm-washed RBCs type group is A Rh- Cefotetan-coated RBCs were tested with the patient’s
positive. The cold autoadsorbed serum confirms that serum and eluate in parallel with the same donor RBCs
6888_Answerkey_617-628 29/10/18 11:26 AM Page 626

626 Answer Key

not treated with cefotetan. Patients with DIIHA have Chapter 25


high-titered antibodies (e.g., >10,000) so dilutions of the
serum and eluate are tested. Also, many normal individ- Case Study 25-1
uals have naturally occurring anti-cefotetan antibodies 1. Step 7: Patient care technician identifies patient; Step 12:
(cephalosporins are added to cattle food) that agglutinate Patient care technician returns transfusion requisition to
cefotetan-coated RBCs, but naturally occurring antibodies nurse.
are of low titer, so testing for DIIHA employs the use of 2. What this high number indicates is that if the patient care
diluted serum and eluate: technician (PCT) mislabeled the specimen, unless the
Review Questions nurse was at the bedside with the PCT when the speci-
men was being collected and observed the PCT correctly
1. d 6. d 11. d 16. b identify the patient, the nurse wouldn’t know that the
2. b 7. b 12. b 17. c specimen had been mislabeled. At this theoretical hospi-
3. a 8. b 13. d 18. d tal, PCTs collect all blood specimens. Looking at the
occurrence rank, if this situation occurred once a year,
4. a 9. c 14. a
the rank would be 5. The severity rank would be 10 be-
5. a 10. a 15. c cause it affects patient safety; detectability would rank as
10 since the failure would be undetectable unless the
patient had an historical blood type. Multiply the three
Chapter 22 numbers: 5 × 10 × 10 = 500.
Review Questions 3. RCA is looking at an error that has already occurred, and
FMEA is predicting how to prevent a future error.
1. d 4. b 7. d 10. d
2. b 5. b 8. b Case Study 25-2
3. c 6. c 9. d Answers will vary.
Review Questions
1. a 4. b 7. c 10. d
Chapter 23
2. d 5. c 8. a
Review Questions
3. d 6. d 9. b
1. c 4. a 7. d 10. b
2. c 5. d 8. a
3. b 6. b 9. d Chapter 26
Case Study 26-1
1. A blood utilization management program is intended
Chapter 24 to ensure blood components are administered appro-
Case 24-1 priately, in accordance with hospital guidelines and
1. b evidence-based criteria. Its primary focus is to improve
2. a patient safety by ensuring patients are not unnecessarily
exposed to the risks of transfusion. If there is a large
Case 24-2 deviation from standard practice in a facility, reduction
1. c in inappropriate ordering may be reflected in reduced
2. d expenditure for blood components. However, if this is
3. c. For DNA polymorphisms, the mutation rate can be as not the case, a utilization management program may still
high as 2 per 1,000; thus, a single mismatch, when all reduce cost in terms of expenditures for treating trans-
other systems are consistent with paternity, may represent fusion-associated adverse events, impact on length of
a mutation. A minimum of two mismatches is necessary stay and clinical course, and improved blood component
before an interpretation of exclusion is rendered. availability.
2. First steps include recruiting a multidisciplinary cross-
Review Questions
functional team. This team should define the program’s
1. b 2. a 3. b 4. a overall goal, then assess the current state and perform a gap
6888_Answerkey_617-628 29/10/18 11:26 AM Page 627

Answer Key 627

analysis. This process will involve using evidence-based cri- 2. Further auditing revealed multiple types of waste with
teria and guidelines. Once improvement opportunities are plasma usage among patients awaiting cardiac surgery
identified, the team may prioritize interventions to promote and procedures. In nearly every case, there was an ex-
evidence-based practice. cess of plasma ordered in advance of the procedure that
3. The blood utilization management team should include was thawed by the blood bank but was either never dis-
the blood bank medical director and the blood bank pensed or never transfused. Regarding the dispensed
supervisor. Additional members will depend upon the plasma, most of these patients had an average of 2 units
individual facility, but representation from nursing, clin- of unused thawed plasma. In a few cases, the product
ical staff (especially from areas with greater blood use), was improperly returned and had to be discarded. This
and quality should be considered. Participation by an in- overproduction type of waste increased the number of
formation technology associate may be particularly useful expired plasma components, resulting in increased in-
in developing tools for queries and to track the metrics ventory waste. Further waste was identified through a
the team agrees on. Administrative support, if not active detailed medical chart audit, which demonstrated that
participation, is critical. nearly half of the plasma transfused was likely inappro-
4. Appropriate utilization criteria should be gleaned from priately administered when compared to the institution’s
evidence-based literature and the recommendations of transfusion guidelines.
consensus panels from various subspecialties. Active dis- 3. Solutions and improvements arise from the collaborative
cussion of proposed guidelines with clinical staff is nec- input of multiple sources such as the medical director of
essary to gain approval and promote compliance. Where the blood bank, the blood bank supervisor, members of
necessary, exceptions for specific clinical populations or the transfusion committee, and appropriate members from
diagnoses should be included. The literature upon which administration. If a specific unit or specialty is identifiable,
the guidelines are based should be widely disseminated as in this case, input should also be requested from clinical
and readily available to clinical staff. staff in that area. In this case, after investigation by the
5. Any of the types of review (retrospective, concurrent, medical director, the new coronary unit was employing
targeted, or discontinuous prospective) may be utilized antiquated order sets copied from another institution.
depending on the facility. However, for a medium- or These order sets included template standing plasma orders
small-sized facility, retrospective review is most likely to that were not appropriate for a large institution with read-
be the primary method, due to resource limitations. Large ily available thawed plasma. Unfortunately, in spite of
facilities such as academic institutions are more likely to discussion with the involved practitioners, many were un-
have the available personnel to conduct concurrent and willing to change their practice patterns. Following review
prospective review actions. Targeted or discontinuous by the transfusion committee, the antiquated order sets
prospective review may be appropriate for smaller facili- were removed and a discontinuous prospective review was
ties, since the benefits of real-time intervention are bal- adopted, focusing on plasma orders for coronary care pa-
anced by the limited scope. tients. In addition, educational measures were employed
in the form of several short lectures on current plasma
Case Study 26-2
guidelines presented to the coronary unit’s physicians and
1. The first step is to determine the gap between the current nursing staff.
and desired state. This gap defines the scope of the prob- 4. Over the following three quarters, the transfusion safety
lem and should be qualified and quantified as succinctly officer continued the discontinuous prospective review
as possible. Review of the value stream will help identify of the coronary care patients in addition to the monthly
the source of the problem. In this case, review of the nurs- retrospective reporting. At the end of the third quarter,
ing units and physician services show that the plasma blood utilization appeared to be at the desired state and
trend appears to be related to a recent expansion in the the labor-intensive discontinuous prospective review
hospital’s coronary care unit. This is further supported by strategy was retired. It is important to always reassess and
the observation that the increased usage and waste trends reevaluate the blood utilization management process to
vanish when the data from the coronary unit are excluded determine the effectiveness of corrective strategies and to
from the utilization report. Review of the timing of the identify new areas of waste.
blood orders showed that the majority of the increased
utilization appeared just prior to the performance of car-
diac procedures and surgeries.
6888_Answerkey_617-628 29/10/18 11:26 AM Page 628

628 Answer Key

Chapter 26 Chapter 29
Review Questions Case Studies 29-1 through 29-6
1. d 4. c 7. b 10. e Editor’s Note: Because of the nature of these cases, answers
2. d 5. c 8. d are not provided. Answers to these questions would be
decided by a judge or jury.
3. b 6. d 9. a
Review Questions
1. d
Chapter 27 2. b
Review Questions 3. d
4. c
1. b 4. d 7. d 10. b
5. a
2. c 5. c 8. a
3. d 6. a 9. b

Chapter 28
Review Questions
1. c 4. b 7. d 10. c
2. a 5. a 8. a
3. d 6. b 9. b

11.
Phase SCI SCII Interpretation

IS 0 0 Negative

37ºC 0 0

AHG 0 0

CC + +

12. b
13. A. Control functions
B. Acceptance criteria
C. Test cases
D. Data entry methods
E. Documentation methods
F. Result review
G. Corrective action
H. Acceptance
6888_color plate 1 25/10/18 5:53 PM Page 1
6888_color plate 1 25/10/18 5:53 PM Page 2

Red Cell Antigen-


Serologic
Macroscopic
0 W+ to 1+ 2+
Tiny Small
Medium-sized
No agglutination agglutinates agglutinates
turbid turbid agglutinates-clear
or hemolysis
background background background

Weaker

Stronger

This chart is modified from


the Red Cell Antigen–Anti-
body Reactions Interpreta-
tion Scheme with
permission from Organon Microscopic
Teknika, BCA Blood Bank
Products, P.O. Box 15969,
Durham, North Carolina
27704-0969 and Organon
Teknika, n.v. Veedijk Negative Positive
58-2300 Turnhout–Belgium.

Plate 1. Red cell antigen-antibody reactions: serologic grading and macroscopic evaluation.
6888_color plate 1 25/10/18 5:53 PM Page 3

Antibody Reactions
Grading
Evaluation
3+ 4+
Several large
One solid
agglutinates—clear
agglutinate
background
Negative: No aggregates

Negative: No aggregates (Microscopic)

Pseudoagglutination or strong
rouleaux (2+)

Note: Partial
or complete
hemolysis is a
positive reaction
Rouleaux: Microscopic (original
magnification x10; enlarged 240%)
Note: The “stack of coins” appearance
of the agglutinates
6888_color plate 1 25/10/18 5:53 PM Page 4
6888_CP2_ch6 29/10/18 10:18 AM Page 2

Step 1: Label test tubes. Step 2: Make a 2–5% patient red cell suspension.

Step 3: Add reagent antisera* Step 3A: Add reagent Anti-A antisera* Step 3B: Add Anti-B reagent antisera*
(approximately 1 drop). (approximately 1 drop). (approximately 1 drop).

Step 4: Add one drop of 2–5% Step 5: Mix and centrifuge


suspension of patient red cells to (approximately 20 seconds).
each tube.
* Consult manufacturer's package insert for specifics.

Figure 6–1.
6888_CP2_ch6 29/10/18 10:18 AM Page 3

Step 6: Read and record agglutination reactions.

Group B Group B
4+ Agglutination with Anti-B 4+ Agglutination with Anti-B
0 Agglutination with Anti-A 0 Agglutination with Anti-A

Group A Group A
4+ Agglutination with Anti-A 4+ Agglutination with Anti-A
0 Agglutination with Anti-B 0 Agglutination with Anti-B

Group AB Group AB
4+ Agglutination with Anti-A and Anti-B 4+ Agglutination with Anti-A and Anti-B

Group O Group O
No Agglutination with Anti-A or Anti-B No Agglutination with Anti-A and Anti-B

Figure 6–1.—cont’d
6888_CP2_ch6 29/10/18 10:18 AM Page 4

Step 1: Label test tubes. Step 2: Add two drops of


patient serum to each tube.

Step 3: Add one drop of reagent cells* Step 3A: Add one drop of Step 3B: Add one drop of
to each test tube. reagent A1 cells. reagent B cells.

Step 4: Mix and centrifuge


(approximately 20 seconds).
* Consult manufacturer's package insert for specifics.

Figure 6–2.
6888_CP2_ch6 29/10/18 10:18 AM Page 5

Step 5: Resuspend cells button; interpret and record results.

Group A Group B
4+ Agglutination with B Cells 4+ Agglutination with A1 Cells
0 Agglutination with A1 Cells 0 Agglutination with B Cells

Group O Group AB
4+ Agglutination with A1 Cells 0 Agglutination with A1 Cells
3+ Agglutination with B Cells 0 Agglutination with A1 and B Cells

Figure 6–2.—cont’d
6888_Glossary_629-652 29/10/18 11:32 AM Page 629

Glossary

Abruptio placentae Premature detachment of normally Agranulocytosis An acute disease in which the white
situated placenta. blood cell count drops to extremely low levels, and
Absorbed anti-A1 If serum from a group B individual that neutropenia becomes pronounced.
contains anti-A plus anti-A1 is incubated with A2 cells, Albumin Protein found in the highest concentration in
the anti-A will adsorb onto the cells. Removal of the human plasma; used as a diluent for blood typing
cells yields a serum containing only anti-A1; thus, it is antisera and a potentiator solution in serologic testing
absorbed anti-A1. to enhance antigen-antibody reactions.
Absorption Removal of an unwanted antibody. Aldomet See Methyldopa.
Acid-citrate-dextrose (ACD) An anticoagulant and preser- Alkaline phosphatase (ALP) A red blood cell enzyme used
vative solution that was once used routinely for blood as an identification marker in paternity testing and
donor collection but now is used only occasionally. criminal investigation.
Acid phosphatase (ACP) A red blood cell enzyme used as Allele One of two or more different genes that may occupy
an identification marker in paternity testing and crimi- a specific locus on a chromosome.
nal investigation. Allo- Prefix indicating differences within a species (e.g., an
Adenosine deaminase (ADA) A red blood cell enzyme alloantibody is produced in one individual against the
used as an identification marker in paternity testing and red blood cell antigens of another individual).
criminal investigation. Allogeneic Transplant donor who is related or unrelated to
Adenosine triphosphate (ATP) A compound composed of the recipient.
adenosine (nucleotide containing adenine and ribose) Allograft A tissue transplant between individuals of the
and three phosphoric acid groups, which, when split same species.
by enzyme action, produces energy that can be used to
Allosteric change A change in conformation that exposes a
support other reactions.
new reactive site on a molecule.
Adenylate kinase (AK) A red blood cell enzyme used as an
Alpha-adrenergic receptor A site in autonomic nerve
identification marker in paternity testing and criminal
pathways wherein excitatory responses occur when
investigation.
adrenergic agents such as norepinephrine and epineph-
Adjuvant One of a variety of substances that, when com- rine are released.
bined with an antigen, enhance the antibody response
Alum precipitation A method for obtaining an enhanced
to that antigen.
response when producing an antibody; see also
Adsorption Providing an antibody with its corresponding Adjuvant.
antigen under optimal conditions so that the antibody
Aminoacyl-tRNA synthetase An enzyme involved in pro-
will attach to the antigen, thereby removing the anti-
tein synthesis by attaching a specific amino acid to
body from the serum; often used interchangeably with
transfer RNA (tRNA), based on a three-nucleotide se-
absorption.
quence present in the loop area of the tRNA called anti-
Agammaglobulinemia A rare disorder in which gamma codon. Once the amino acid is attached to the tRNA, it
globulin is virtually absent. may be joined in the ribosome with the growing peptide
Agarose Seaweed extract used to make gels used in elec- chain.
trophoresis. Molten agarose (0.5% to 3.5%, depending Amniocentesis Transabdominal puncture of the amniotic
on application) is poured into a mold where a plastic sac, using a needle and syringe, in order to remove am-
comb is suspended. As it cools, the agarose hardens to niotic fluid. The material may then be studied to detect
form a porous gel slab. genetic disorders or fetomaternal blood incompatibility.
Agglutination The clumping together of red blood cells or Amniotic fluid Liquid or albuminous fluid contained in
any particulate matter resulting from interaction of anti- the amnion.
body and its corresponding antigen.
Amorph A gene that does not appear to produce a de-
Agglutinin An antibody that agglutinates cells. tectable antigen; a silent gene, such as Jk, Lu, O.
Agglutinogen A substance that stimulates the production Amplicon The product of polymerase chain reaction.
of an agglutinin, thereby acting as an antigen.

629
6888_Glossary_629-652 29/10/18 11:32 AM Page 630

630 Glossary

Anamnestic response An accentuated antibody response Antigen A substance recognized by the body as being for-
following a secondary exposure to an antigen. Antibody eign, which can cause an immune response. In blood
levels from the initial exposure are not detectable in the banking, antigens are usually, but not exclusively, found
patient’s serum until the secondary exposures, when a on the red blood cell membrane.
rapid rise in antibody titer is observed. Antihemophilic factor See Hemophilia A.
Anaphylaxis An allergic hypersensitivity reaction of the Antihemophilic globulin See Hemophilia A.
body to a foreign protein or drug.
Antihistamine Drug that opposes the action of histamine.
Anastomosis A connection between two blood vessels,
Anti-H lectin A reagent anti-H produced from the seeds of
either direct or through connecting channels.
the plant Ulex europaeus.
Anemia A condition in which there is reduced oxygen
Antihuman globulin or antiglobulin serum See Antihuman
delivery to the tissues; may result from increased
serum.
destruction of red blood cells, excessive blood loss,
or decreased production of red blood cells. Antihuman globulin or antiglobulin test (AGT) Test to
ascertain the presence or absence of red blood cell
Angina pectoris Severe pain and constriction about the
coating by immunoglobulin G (IgG) or complement or
heart caused by an insufficient supply of blood to the
both; uses a xenoantibody (rabbit antihuman serum)
heart.
or monoclonal antibody (to IgG or complement) to act
Anion An ion carrying a negative charge. as a bridge between sensitized cells, thus yielding agglu-
Annealing Process in which single strands of DNA anneal tination as a positive result. Direct antihuman globulin
to each other by formation of hydrogen bonds, under test (DAT): Used to detect in vivo cell sensitization.
certain temperature conditions, due to complementarity. Indirect antihuman globulin test (IAT): Used to detect
Annealing, renaturation, and hybridization are chemi- antigen-antibody reactions that occur in vitro.
cally the same processes. Two strands of DNA, separated Antihuman serum An antibody prepared in rabbits or
by heat denaturation, will start annealing (renaturation) other suitable animals that is directed against human
upon cooling to approximately 5°C (or more) below immunoglobulin or complement or both; used to
DNA melting point (Tm). This phenomenon is used perform the antihuman globulin or Coombs’ test.
in polymerase chain reaction when oligonucleotide The serum may be either polyspecific (anti-IgG plus
primers must anneal to denatured DNA prior to starting anticomplement) or monospecific (anti-IgG or
DNA synthesis by the enzyme polymerase. anticomplement).
Antecubital In front of the elbow, at the bend of the elbow; Anti-M lectin A reagent anti-M serum produced from the
usual site for blood collection. plant Iberis amara.
Antenatal Occurring before birth. Anti-nl An antibody implicated in warm autoimmune
Anti-A1 lectin A reagent anti-A1 serum produced from the hemolytic anemia, which reacts with all normal Rh cells
seeds of the plant Dolichos biflorus; reacts with A1 cells except Rhnull cells and deleted Rh cells.
but not with A subgroup cells, such as A2, A3, and so Anti-N lectin A reagent anti-N serum produced from the
on; reacts weakly with Aint cells. plant Vicia graminea.
Anti-B lectin A reagent anti-B serum produced from the Anti-pdl An antibody implicated in warm autoimmune
seeds of the plant Bandeiraea simplicifolia. hemolytic anemia, which reacts with all normal Rh cells
Antibody A protein substance secreted by plasma cells that and deleted Rh cells but not with Rhnull cells.
is developed in response to, and interacting specifically Antipyretic An agent that reduces fever.
with, an antigen. In blood banking, it is found in serum,
Antiserum A reagent source of antibody, as in a commer-
from either a commercial manufacturer or a patient.
cial antiserum.
Antibody screen Testing the patient’s serum with group O
Antithetical Referring to antigens that are the product of
reagent red blood cells in an effort to detect atypical
allelic genes (e.g., Kell [K] and Cellano [k]).
antibodies.
Apheresis A method of blood collection in which whole
Anticoagulant An agent that prevents or delays blood
blood is withdrawn, a desired component separated
coagulation.
and retained, and the remainder of the blood is
Anticodon A sequence of three nucleotides in the loop returned to the donor. See also Plateletpheresis and
region of the transfer RNA (tRNA), which aligns Plasmapheresis.
in the ribosome with the corresponding mRNA codon
Aplasia Failure of an organ or tissue to develop
and attaches the amino acid to the growing peptide
normally.
chain.
Aplastic anemia Anemia caused by aplasia of bone
Anti-dl An antibody implicated in warm autoimmune
marrow or bone marrow’s destruction by chemical
hemolytic anemia, which reacts with all Rh cells,
agents or physical factors.
including Rhnull and Rh-deleted cells.
6888_Glossary_629-652 29/10/18 11:32 AM Page 631

Glossary 631

Apoptotic Adjective of apoptosis, the programmed (non- Bar code reader An optical input device that reads and in-
traumatic) cell death. A natural process occurring in terprets data from a bar code for entry into a computer
senescent cells. system.
Arachis hypogaea A peanut lectin used to differentiate T Beta globin chain A structural peptide subunit of hemo-
polyagglutination from Tn polyagglutination. globin molecule that consists of two beta globin and
Asphyxia Condition caused by insufficient intake of two alpha globin chains. Alpha and beta chains are
oxygen. coded for by different genes located on different
chromosomes.
Asthma Paroxysmal dyspnea accompanied by wheezing
caused by a spasm of the bronchial tubes or by swelling Bilirubin The orange-yellow pigment in bile carried to the
of their mucous membrane. liver by the blood; produced from hemoglobin of red
blood cells by reticuloendothelial cells in bone marrow,
Atypical antibodies Any antibody other than anti-A,
spleen, and elsewhere. Direct bilirubin: The conjugated
anti-B, or anti-A,B.
water-soluble form of bilirubin. Indirect bilirubin: The
Australia antigen Old terminology referring to the unconjugated water-insoluble form of bilirubin.
hepatitis B–associated antigen.
Bilirubinemia Pathological condition in which excessive
Auto- Prefix indicating self (e.g., an autoantibody is reac- destruction of red blood cells occurs, increasing the
tive against one’s own red blood cell antigens); usually amount of bilirubin found in the blood.
associated with a disease state.
Binding constant The “goodness of fit” in an antigen-
Autoabsorption A procedure to remove a patient’s anti- antibody complex.
body, using the patient’s own cells.
Biphasic Reactivity occurring in two phases.
Autoimmune hemolytic anemia Shortened RBC survival
Blood bank information system Computer system that has
mediated through the immune response of humoral an-
been developed specifically to assist blood bank profes-
tibody directed at “self” antigenic determinants. Ac-
sionals in the management of the patient, donor, and
quired disorder characterized by premature erythrocyte
blood component information.
destruction owing to abnormalities in the individual’s
immune system. Blood gases Determination of pH, PCO2, PO2, and HCO3;
performed on a blood gas analyzer.
Autologous Donor and recipient are the same person.
Blood group genotyping DNA typing aimed at genes
Autologous control Testing the patient’s serum with his or
coding for blood group antigens.
her own cells in an effort to detect autoantibody activity.
Blood group–specific substances (BGSSs) Soluble anti-
Autologous transfusion Blood taken from a patient to be
gens present in fluids that can be used to neutralize
used for the same patient.
their corresponding antibodies; systems that demon-
Autoradiography An image recorded on a photographic strate BGSSs include ABO, Lewis, and P blood group
film or plate produced by the radiation emitted from a systems.
specimen, such as DNA labeled with radioactive phos-
Blood order sets A means of standardization to clarify
phorus isotope.
indications and enforce guidelines during physician
Autosomal-recessive pattern An inheritance pattern in order entry.
which a condition or disease determined by a gene lo-
Blood utilization management A process incorporating
cated on an autosomal (nonsex) chromosome occurs
blood utilization review with particular intervention
when both genes on the homologous chromosomes are
strategies to reduce inappropriate transfusion and im-
affected. Individuals with only one gene affected are
prove patient safety.
considered carriers and do not have symptoms due to
proper function of the unaffected gene on the other Blood utilization review A focused audit of a population
chromosome. or subpopulation of transfused patients to determine
the appropriateness of transfusion.
Autosome Any chromosome other than the sex (X and Y)
chromosomes. Blunt ends The ends of a DNA sequence resulting from re-
striction enzymes that cut both strands in the middle of
Bacterial artificial chromosomes (BACs) In recombinant
the target sequence, leaving no “overhangs.” Blunt ends
DNA technology, vectors capable of carrying DNA frag-
may be rejoined using enzyme ligase.
ments (inserts) of up to 350 kb.
Bombay Phenotype occurring in individuals who possess
Bactericidal Destructive to or destroying bacteria.
normal A or B genes but are unable to express them
Bacteriophage A virus that infects and reproduces in bacte- because they lack the gene necessary for production
rial cells. Often carries genes coding for antibiotic resis- of H antigen, the required precursor for A and B. These
tance. Basic tools used in molecular biology (vectors) persons often have a potent anti-H in their serum,
frequently used to introduce and clone (multiply) for- which reacts with all cells except other Bombays.
eign genes or their fragments inside of a bacterium. Also known as Oh.
Bandeiraea simplicifolia See Anti-B lectin.
6888_Glossary_629-652 29/10/18 11:32 AM Page 632

632 Glossary

Bovine Pertaining to cattle. Central dogma A theory developed originally by Francis


Bradykinin A plasma kinin. Crick, claiming that basic information of life flows from
DNA through RNA to proteins.
Bromelin A proteolytic enzyme obtained from the
pineapple. Central processing unit (CPU) The part of a computer
that contains the semiconductor chips that process the
Bromophenol blue A tracking dye mixed with DNA sam-
instructions of the computer programs.
ple and used in electrophoresis to monitor its progress
in order to prevent the escape of bands out of the gel. In Central venous pressure The pressure within the superior
a 1% gel, it runs at around 500 bp. vena cava reflecting the pressure by which the blood is
returned to the right atrium.
Buffy coat Light stratum of a blood clot seen when the
blood is centrifuged or allowed to stand in a test tube. Chain termination method (Sanger sequencing) The most
The red blood cells settle to the bottom, and between common method of DNA sequencing, developed by
the plasma and the red blood cells is a light-colored Fred Sanger, which uses DNA synthesis terminating
layer that contains mostly white blood cells. ddNTPs.
Burst-forming unit committed to erythropoiesis (BFU-E) Chargaff’s rules Established in 1950 by chemist Erwin
A primitive progenitor cell committed to erythropoiesis Chargaff, these rules describe the quantitative ratios of
and believed to be a precursor to the CFU-E. nitrogen-containing bases, adenine, thymine, cytosine,
and guanine, in DNA. The first rule stated that the
C3a A biologically active fragment of the complement C3
amount of adenine in a given molecule was always
molecule that demonstrates anaphylactic capabilities
equal to the amount of thymine and that the amount of
upon liberation.
cytosine was always equal to the amount of guanine.
C3b A biologically active fragment of the complement C3 The second rule stated that the amount of all pyrimi-
molecule that is an opsonin and promotes immune dine bases (T and C) in a given DNA molecule was
adherence. equal to the amount of all purine bases (A and G) in
C3d A biologically inactive fragment of the C3b comple- that molecule.
ment component formed by inactivation by the C3b Chemically modified anti-D IgG anti-D reagent antisera in
inactivator substance present in serum. which the immunoglobulin has been chemically modi-
C4 A complement component present in serum that fied to react in the saline phase of testing by breaking
participates in the classic pathway of complement disulfide bonds at the hinge region of the molecule,
activation. converting the Y-shaped antibody structure to a
C5a A biologically active fragment of the C5 molecule, T-shaped form through the use of sulfhydryl-reducing
which demonstrates anaphylactic capabilities and reagents.
chemotactic properties upon liberation. This fragment is Chemotaxis Movement toward a stimulus, particularly
also a potent aggregator of platelets. that movement displayed by phagocytic cells toward
Cadaveric Source of organs and tissues from a person who bacteria and sites of cell injury.
has been declared dead. Chimera An individual who possesses a mixed cell
Capillary electrophoresis The electrophoresis platform population.
consisting of a small capillary filled with some form of Chloroquine diphosphate Substance that dissociates IgG
separation medium that fractionates a mixture of mole- antibody from red blood cells with little or no damage
cules based upon size or charge. to the red blood cell membrane.
Capillary gel electrophoresis The process of electrophore- Chorionic villus sampling A prenatal procedure of
sis that occurs in a thin glass capillary filled with poly- retrieval of tissue from the chorionic villi (the villi in
acrylamide or other synthetic polymer rather than in a the external membrane surrounding the fetus).
slab. Chromogen Any chemical that may be changed into
Cardiac output The amount of blood discharged from the coloring matter.
left or right ventricle per minute. Chromosome The structures within a nucleus that
Catecholamines Biologically active amines, epinephrine contain a linear thread of DNA, which transmits genetic
and norepinephrine, derived from the amino acid tyro- information. Genes are arranged along the strand of
sine. They have a marked effect on nervous and cardio- DNA and constitute portions of the DNA.
vascular systems, metabolic rate, temperature, and Cis position The location of two or more genes on the
smooth muscle. same chromosome of a homologous pair.
Cathode ray tube (CRT) A display device in an informa- Citrate Compound of citric acid and a base; used in
tion system. anticoagulant solutions.
Cation An ion carrying a positive charge. Citrate-phosphate-dextrose (CPD) The anticoagulant
CD34 Cell membrane marker of stem cells. preservative solution that replaced ACD in routine
6888_Glossary_629-652 29/10/18 11:32 AM Page 633

Glossary 633

donor collection. It has been replaced by CPDA-1 in Complementarity A phenomenon occurring in nucleic
routine use. acids, resulting from formation of noncovalent hydro-
Citrate-phosphate-dextrose-adenine (CPDA-1) The anti- gen bonds between the nitrogen-containing bases in
coagulant preservative solution in current use. It has the nucleotides on the opposite strands of the double
extended the shelf life of blood from 21 days (ACD and helix. Due to this process, adenine (A) always pairs
CPD) to 35 days. by two hydrogen bonds with thymine (T) or uracil (U),
and guanine (G) always pairs by three hydrogen bonds
Clone A group of genetically identical cells.
with cytosine (C).
Codominant A pair of genes in which neither is dominant
Complementary DNA (cDNA) An in vitro enzymatically
over the other—that is, they are both expressed.
synthesized DNA from an RNA template. A product of
Codon A sequence of three nucleotides (bases) in the reverse transcription.
strand of DNA that provides the genetic code for spe-
Complement fixation (CF) An immunologic test.
cific amino acid. The complementary triplets are found
in the messenger RNA (mRNA), which is synthesized Component therapy Transfusion of specific components
based on the DNA template in the nucleus, and then (e.g., red blood cells, platelets, plasma) rather than
proceeds to the ribosomes for protein synthesis, where whole blood to treat a patient. Components are separa-
the transfer RNA (tRNA), upon alignment with the ble by physical means such as centrifugation.
mRNA, attaches the corresponding amino acid to the Compound antibody An antibody whose corresponding
growing peptide chain. antigen is an interaction product of two or more
Colloid A gluelike substance, such as protein or starch, antigens.
whose particles, when dispersed in a solvent to the Compound antigen Two or more antigens that interact
greatest possible degree, remain uniformly distributed and are recognized as a single antigen by an antibody.
and fail to form a true solution. Computer crossmatch Comparison of recent serologic
Colony-forming unit committed to erythropoiesis results and interpretations on a computer file for both
(CFU-E) A progenitor cell that is committed to forming the donor and the recipient being matched, establishing
cells of the red blood cell series. compatibility based on the comparison.
Colony-forming unit–culture (CFU-C) Generation of stem Concurrent review A blood utilization review that occurs
cells using tissue culture methods. Current synonym is shortly after transfusion events.
CFU-GM, which is a colony-forming unit committed to Configuration The physical layout and design of the
the production of myeloid cells (granulocytes and central processing unit and the peripheral devices of
monocytes). an information system.
Colostrum Thin yellowish breast fluid secreted 2 to 3 days Conglutinin A substance present in bovine serum that
after birth but before the onset of true lactation; it will agglutinate sensitized cells in the presence of
contains a great quantity of proteins and calories as complement.
well as antibodies and lymphocytes.
Constant region The portion of the immunoglobulin chain
Compatibility test A series of tests to determine if a partic- that shows a relatively constant amino acid sequence
ular blood component is appropriate for transfusion within each class of immunoglobulin. Both light and
to a particular patient. This series of tests usually in- heavy chains have these constant portions, which origi-
cludes the ABO/Rh of the component and the intended nate at the carboxyl region of the molecule.
recipient, antibody detection of the recipient, and a
Convulsion Involuntary muscle contraction and relaxation.
crossmatch (serologic or electronic) of the recipient’s
serum/plasma with the RBCs of the donor unit. Coombs’ serum See Antihuman serum.
Competent cells Cells (usually various strains of E. coli) Coombs’ test See Antihuman globulin test (AGT).
that possess easily altered cell membrane and readily Cord cells Fetal cells obtained from the umbilical cord at
incorporate foreign DNA (are prone to transformation). birth; may be contaminated with Wharton’s jelly.
Competence may be achieved by exposure to calcium Cordocentesis Umbilical cord blood sampling performed
chloride and heat shock. by ultrasound-guided needle insertion through the
Complement A series of proteins in the circulation that, uterine wall.
when sequentially activated, causes disruption of Cosmids Vectors that can accept DNA 28 to 45 kb long.
bacterial and other cell membranes. Activation occurs They have a small region of bacteriophage lambda
via one of two pathways; once activated, the compo- (␭) necessary for packaging viral DNA into ␭ particles.
nents are involved in a great number of immune They are useful for producing large-insert genomic
defense mechanisms, including anaphylaxis, chemo- libraries.
taxis, and phagocytosis. Red blood cell antibodies
Coumarin (Coumadin) A commonly employed anticoagu-
that activate complement may be capable of causing
lant that acts as a vitamin K antagonist to prolong
hemolysis.
prothrombin time.
6888_Glossary_629-652 29/10/18 11:32 AM Page 634

634 Glossary

Counterelectrophoresis (CEP) An immunologic Deglycerolization Removal of glycerol from a unit of red


procedure. blood cells after thawing has been performed; required
Crossmatch The testing of the patient’s serum with the to return the cells to a normal osmolality.
donor red blood cells, including an antiglobulin phase Deletion The loss of a portion of chromosome.
or simply an immediate spin phase to confirm ABO Denaturation In molecular biology, a term that describes
compatibility. separation of two DNA strands upon thermal or
Cross-reacting antibody Antibody that reacts with chemical treatment, which destroys the noncovalent
antigens functionally similar to its specific antigen. hydrogen bonds between the complementary nitrogen-
Cryoprecipitate A concentrated source of coagulation containing bases. Thermal denaturation is reversible
factor VIII prepared from a single unit of donor blood; (see Renaturation). The temperature at which a given
it also contains fibrinogen, factor XIII, and von double-stranded DNA denatures is proportional to
Willebrand’s factor. the number of hydrogen bonds in the molecule and
increases with the increase of G-C pair content. In
Cryopreservation Preservation by freezing at very low
reference to DNA, denaturation is a chemical or thermal
temperatures.
process of breaking hydrogen bonds between the
Cryoprotectant A substance that protects blood cells from opposite nucleotides in the double helix, resulting in
damage caused by freezing and thawing. Glycerol and reversible separation of the two strands. Denaturation
dimethyl sulfoxide are examples. is the first step of polymerase chain reaction (PCR).
Cryptantigens Hidden receptors that may be exposed Deoxyribonucleic acid (DNA) The chemical basis of
when normal erythrocyte membranes are altered by heredity and the carrier of genetic information for all
bacterial or viral enzymes. organisms, except RNA viruses. Structured as a double
Crystalloid A substance capable of crystallization; opposite helix of polymers of nucleotides, each consisting of one
of colloid. of the nitrogen-containing bases (A, T, C, and G), sugar
Cyanosis Slightly bluish or grayish skin discoloration deoxyribose, and a phosphate.
resulting from accumulations of reduced hemoglobin Deoxyribonucleoside triphosphates (dNTPs) Chemical
or deoxyhemoglobin in the blood caused by oxygen compounds (nucleotides) used during DNA synthesis.
deficiency or carbon dioxide buildup. They consist of one of the nitrogen-containing bases
Cycle sequencing A variation of Sanger DNA sequencing (A, T, C, and G), sugar deoxyribose, and three phos-
conducted in a thermocycler and during the elongation phates. They become monophosphates once they are
(extension) step of a modified polymerase chain built into the DNA molecule.
reaction. Dexamethasone A topical steroid with anti-inflammatory,
Cytapheresis A procedure performed using a machine by antipruritic, and vasoconstrictive actions.
which one can selectively remove a particular cell type Dextran A plasma expander that may be used as a substi-
normally found in peripheral blood of a patient or donor. tute for plasma; can be used to treat shock by increasing
Cytokines A family of signaling molecules, such as inter- blood volume. Rouleaux may be observed in the recipi-
ferons or interleukins, secreted by certain cell types and ent’s serum or plasma.
triggering various physiological processes by interacting Diagnosis-related group (DRG) Classification system that
with receptors located either on cell membranes or organizes short-term, general hospital inpatients into
inside of cells. statistically stable groups based on age and illness.
Cytomegalovirus (CMV) One of a group of species- Diaphoresis Profuse sweating.
specific herpesviruses. Diastolic pressure The point of least pressure in the
Cytotoxicity Ability to destroy cells. arterial vascular system; the lower or bottom value
Cytotoxicity testing Procedure commonly used in HLA of a blood pressure reading.
typing and crossmatching. Dideoxyribonucleoside triphosphates (ddNTPs)
Dane particle Hepatitis B virion. Deoxyribonucleoside triphosphates deprived of an
oxygen atom located on the third sugar in the deoxyri-
Database An organized group of files in which information
bose molecule. Lack of this oxygen prevents polymer-
is stored in an information system.
ization of nucleotides during DNA synthesis. Used as
Degenerate In molecular biology, the quality of the genetic terminators of DNA synthesis in the process of DNA
code that refers to each amino acid being encoded by sequencing.
more than one nucleotide triplet (codon) in messenger
Dielectric constant A measure of the electrical conductivity
RNA (mRNA) corresponding to a triplet in DNA.
of a suspending medium.
Degeneracy is also called redundancy of the genetic
code. For example, the amino acid leucine is coded Differential count Counting 100 leukocytes to ascertain
by UUA, UUG, CUU, CUC, CUA, or CUG codons. the relative percentages of each.
6888_Glossary_629-652 29/10/18 11:32 AM Page 635

Glossary 635

2,3-Diphosphoglycerate (2,3-DPG) An organic phosphate DNA typing Also called genetic profiling, this is a process
in red blood cells that alters the affinity of hemoglobin in which it is established which polymorphic version or
for oxygen. Blood cells stored in a blood bank lose variant of a gene (allele, locus) is present in an individ-
2,3-DPG, but once infused, the substance is resynthe- ual. Typing of multiple loci is called DNA fingerprinting.
sized or reactivated. Dolichos biflorus See Anti-A1 lectin.
Diploid Having two sets of 23 chromosomes, for a total Domain Portions along the immunoglobulin chain that
of 46. show specific biological function.
Direct antiglobulin test (DAT) Test that detects in vivo Dominant A trait or characteristic that will be expressed in
coating of an individual’s red blood cells with IgG or the offspring even though it is carried on only one of
complement (C3). the homologous chromosomes.
Direct transfusion Transfer of blood directly from one Donath-Landsteiner test A test usually performed in
person to another. the blood bank to detect the presence of the Donath-
Discontinuous prospective review A temporary, focused Landsteiner antibody, which is a biphasic immunoglob-
prospective blood utilization review strategy that is per- ulin G antibody with anti-P specificity found in patients
formed on a combination of particular product types, suffering from paroxysmal cold hemoglobinuria.
DRGs, clinical services, or individual clinicians that can Donor An individual who donates a pint of blood.
be employed in areas of increased inappropriate blood
Dopamine A catecholamine synthesized by the adrenal
utilization.
gland, used especially in the treatment of shock.
Disk drive A hardware device in an information system
Dosage A phenomenon whereby an antibody reacts more
that contains a disk on which data are stored; provides
strongly with a red blood cell carrying a double dose
for quick access to storing or retrieving data.
(homozygous inheritance of the appropriate gene)
Disseminated intravascular coagulation (DIC) Clinical than with a red blood cell carrying a single dose
condition of altered blood coagulation secondary to a (heterozygous inheritance) of an antigen.
variety of diseases.
Dot blotting In molecular biology, a technique in which
Dithiothreitol (DTT) A sulfhydryl compound used to the nucleic acid extracted from a specimen is immobi-
disrupt the disulfide bonds of immunoglobulin M, lized on a membrane (blot) in a form of a dot. This
yielding monomeric units rather than the typical procedure does not require electrophoresis as do other
pentameric molecule. blotting techniques (Southern, Northern, or Western).
Diuresis Secretion and passage of large amounts of urine. The membrane is subsequently incubated in a solution
Diuretic An agent that increases the secretion of urine, with labeled probes, complementary to sequences, ex-
either by increasing glomerular filtration or by pected to be present in the tested specimen. Microarrays
decreasing reabsorption from the tubules. are an example of a modified (reversed) dot blotting
technique.
Dizygotic twins Twins who are the product of two
fertilized ova (also called fraternal twins). Drug-induced immune hemolytic anemia Shortened RBC
survival caused by an antibody to a therapeutic agent
DMSO Dimethyl sulfoxide, a cryoprotectant used for
(e.g., antibiotic) that has become bound to the RBC
hematopoietic progenitor cells.
membrane or that has altered the RBC membrane.
DNA See Deoxyribonucleic acid.
Dyscrasia An old term now used as a synonym for disease.
DNA fingerprinting DNA typing of multiple loci simulta-
Ecchymosis A form of macula appearing in large irregu-
neously in order to establish a genetic profile unique
larly formed hemorrhagic areas of the skin; first blue-
for every individual. A method initially developed by
black, then changing to greenish brown or yellow.
Alex Jeffreys and performed using restriction enzymes
and DNA probes complementary to short repetitive Edema A local or generalized condition in which the body
sequences. Later, improved by the polymerase chain tissues contain an excessive amount of tissue fluid.
reaction (PCR). Electrolyte A substance that in solution conducts an
DNA libraries Collections of clones (cells produced in electric current; common electrolytes are acids, bases,
molecular cloning) that contain all genomic or mRNA and salts.
sequences of a particular cell type. Electrophoresis The movement of charged particles
DNA polymerase An enzyme that catalyzes the template— through a medium (paper, agar gel) in the presence of
dependent on synthesis of DNA. Also known as the an electrical field; useful in the separation and analysis
HBeAg of the hepatitis B virion. of proteins.
DNA polymerase III Bacterial enzyme synthesizing a new Eluate See Elution.
DNA strand using deoxyribonucleoside triphosphates Elution A process whereby cells that are coated with
(dNTPs) as substrates, double-stranded DNA as antibody are treated in such a manner as to disrupt
template, and magnesium as an enzyme cofactor. the bonds between the antigen and antibody. The freed
6888_Glossary_629-652 29/10/18 11:32 AM Page 636

636 Glossary

antibody is collected in an inert diluent such as saline UV visualization. Mutagenic and carcinogenic is being
or 6% albumin. This antibody serum then can be tested replaced by safer SYBR green dye.
to identify its specificity using routine methods. The Ethylenediaminetetraacetic acid (EDTA) An anticoagulant
mechanism to free the antibody may be physical useful in hematologic testing and preferable when direct
(heating, shaking) or chemical (ether, acid), and the antihuman globulin testing is indicated.
harvested antibody-containing fluid is called an eluate.
Euglobulin lysis Coagulation procedure testing for
Embolism Obstruction of a blood vessel by foreign sub- fibrinolysin.
stances or a blood clot.
Exchange transfusion Transfusion and withdrawal of
Embolus A mass of undissolved matter present in a blood small amounts of blood, repeated until blood volume is
or lymphatic vessel, brought there by the blood or almost entirely exchanged; used in infants born with
lymph circulation. hemolytic disease.
Endemic A disease that occurs continuously in a particular Exclusion An exclusion occurs when a child has inherited
population but has a low mortality rate; used in contrast a genetic marker not detected in an alleged parent.
to epidemic. Exclusions can be direct or indirect, which are distin-
Endogenous Produced or arising from within a cell or guished in the following way: A direct exclusion refers
organism. to the presence of a genetic marker in the child that
Endothelium A form of squamous epithelium consisting is not detectable in the alleged parent. An indirect
of flat cells that line the blood and lymphatic vessels, exclusion occurs when the alleged parent has a marker
the heart, and various other body cavities; derived from that should be transmitted to all his or her children,
mesoderm. but the child in question lacks that genetic marker.
Indirect exclusions can result when mutations occur
Endotoxemia The presence of endotoxin in the blood;
during meiosis or when the gene transmitted to the
endotoxin is present in the cells of certain bacteria (e.g.,
child is silent or “null.”
gram-negative organisms).
Exogenous Originating outside an organ or part.
Engraftment The successful establishment, proliferation,
and differentiation of transplanted hematopoietic stem Exon The coding part of a gene in eukaryotes. Only this
cells. part of the gene is translated into protein product.
In genomic DNA, exons are separated by introns.
Enzyme A substance capable of catalyzing a reaction; pro-
teins that induce chemical changes in other substances Expression library A DNA library generated in a vector
without being changed themselves. that not only replicates itself, but also drives protein
synthesis in Escherichia coli (an expression vector).
Enzyme-linked immunosorbent assay (ELISA) An
immunologic test. Extracorporeal Outside of the body.
Enzyme treatment A procedure in which red blood cells Extravascular Outside of the blood vessel.
are incubated with an enzyme solution that cleaves Factor assay Coagulation procedure to assay the concen-
some of the membrane’s glycoproteins, then washed tration of specific plasma coagulation factors.
free of the enzyme and used in serologic testing. Factor VIII concentrate A commercially prepared source of
Enzyme treatment cleaves some antigens and exposes coagulation factor VIII.
others.
Febrile reaction A transfusion reaction caused by leukoag-
Epistaxis Hemorrhage from the nose; nosebleed. glutinin that is characterized by fever; usually observed
Epitope The portion of the antigen molecule that is in multiply transfused or multiparous patients.
directly involved in the interaction with the antibody; Fetal and neonatal alloimmune thrombocytopenia
the antigenic determinant. (FNAIT) A rare condition that occurs when a mother
Equivalence zone The zone in which antigen and antibody becomes sensitized to a foreign antigen of paternal
concentrations are optimal and lattice formation is most origin that is present on fetal thrombocytes (platelets).
stable. These platelet antigens provoke production of maternal
Erythroblast A precursor form of nucleated red blood cell antibodies that cross the placenta and destroy fetal
that is not normally seen in the circulating blood. platelets.
Erythroblastosis fetalis See Hemolytic disease of the fetus Fibrin A whitish filamentous protein or clot formed by
and newborn (HDFN). the action of thrombin on fibrinogen, converting it to
fibrin.
Erythrocyte The blood cell that transports oxygen and
carbon dioxide; a mature red blood cell. Fibrinogen A protein produced in the liver that circulates
in plasma. In the presence of thrombin, an enzyme
Ethidium bromide (EB) A fluorescent dye intercalating
produced by the activation of the clotting mechanism,
between the grooves of the double-stranded DNA.
fibrinogen is cleaved into fibrin, which is an insoluble
Used for staining of DNA in electrophoretic gels for
protein that is responsible for clot formation.
6888_Glossary_629-652 29/10/18 11:32 AM Page 637

Glossary 637

Fibrinolysin The substance that has the ability to dissolve GM-CSF Granulocyte macrophage–colony stimulating
fibrin; also called plasmin. factor, sargramostim.
Fibrinolysis Dissolution of fibrin by fibrinolysin, caused G6PD (glucose-6-phosphate dehydrogenase) A red cell
by the action of a proteolytic enzyme system that is enzyme involved in the glycolytic pathway.
continually active in the body but that is increased Gamete A mature male or female reproductive cell.
greatly by various stress stimuli.
Gamma globulin A protein found in plasma and known to
Fibroblast Cells found throughout the body that synthe- be involved in immunity.
size connective tissue.
Gamma marker Allotypic marker on the gamma heavy
Ficin A proteolytic enzyme derived from the fig. chain of the IgG immunoglobulin.
Ficoll A macromolecular additive that enhances the Gel test A blood group serology test method that uses a
agglutination of red blood cells. microtube containing gel (with or without antisera or
Ficoll-Hypaque A density-gradient medium used to antiglobulin sera) that acts as a reaction vessel for
separate and harvest specific white blood cells, most agglutination.
commonly lymphocytes. Gene A unit of inheritance within a chromosome.
Fluorescent antibody Antibody reaction made visible Gene chips Microarrays.
by incorporating a fluorescent dye into the antigen-
Gene expression A term generally describing the processes
antibody reaction and examining the specimen with a
of transcription and translation. A gene that is not tran-
fluorescent microscope.
scribed (not expressed) is considered “silent.”
Fluorescent in situ hybridization (FISH) A technique in
Gene therapy An introduction of new genetic material into
molecular biology in which fluorescent DNA probes are
the cells of an organism for therapeutic purposes.
used to detect homologous DNA or RNA sequences in
preserved chromosomes or intact cells. Genetic code A key according to which a sequence of sets
of three DNA nucleotides within a gene is transcribed
Fluorescent resonance energy transfer (FRET) probes A
into a sequence of sets of three nucleotides within
type of DNA probe used in real-time PCR. Instead of a
mRNA (codon) and then translated into amino acids,
quencher, attached to the TaqMan probes or molecular
the building blocks of a peptide or protein.
beacons, in the FRET system the fluorescence “donor”
and “acceptor” are attached to two separate hybridiza- Genetic transformation Process of foreign DNA uptake by
tion probes designed to bind to adjacent DNA sequences a cell, usually enforced by use of chemicals, electricity,
within the amplified region. Once the PCR product or liposomes that enable penetration of cell membrane.
starts to accumulate in the tube, the probes bind to the Genotype An individual’s actual genetic makeup.
sequences within the product in close proximity and Gestation In mammals, the length of time from conception
the energy from the donor is transferred to the reporter, to birth.
which results in emission of light that may now be
Globin A protein constituent of hemoglobin. There are
detected.
four globin chains in the hemoglobin molecule.
Formaldehyde A disinfectant solution.
Glomerulonephritis A form of nephritis in which the
Forward grouping Testing unknown red blood cells lesions involve primarily the glomeruli.
with known reagent antisera to determine which ABO
Glutamic pyruvate transaminase A liver enzyme used to
antigens are present.
monitor liver function; also called serum glutamic pyru-
Frameshift mutation A change in which a message is read vate transaminase (SGPT) or alanine transferase (ALT).
incorrectly either because a base is missing or an extra
Gluten enteropathy A condition associated with
base is added; this results in an entirely new polypep-
malabsorption of food from the intestinal tract.
tide because the triplet sequence has been shifted
one base. Glycerol A cryoprotective agent.
Fresh-frozen plasma (FFP) A frozen plasma product Glycerolization Adding glycerol to a unit of red blood cells
(from a single donor) that contains all clotting factors, for the purpose of freezing.
especially the labile factors V and VIII; useful for Glycine soja Soybean extract or lectin used to differentiate
clotting factor deficiencies other than hemophilia A, different forms of polyagglutination.
von Willebrand’s disease, and hypofibrinogenemia. Glycophorin A A major glycoprotein of the red blood cell
Freund’s adjuvant Mixture of killed microorganisms, membrane. MN antigen activity is found on it.
usually mycobacteria, in an oil-and-water emulsion. Glycophorin B An important red blood cell glycoprotein:
The material is administered to induce antibody forma- SsU antigen activity is found here.
tion and yields a much greater antibody response.
Glycoprotein A protein with linked carbohydrates.
Furosemide (Lasix) An oral diuretic.
Glycosyl transferase A protein enzyme that promotes
G-CSF Granulocyte-colony stimulating factor, filgrastim. the attachment of a specific sugar molecule to a
6888_Glossary_629-652 29/10/18 11:32 AM Page 638

638 Glossary

predetermined acceptor molecule. Many blood group Hematopoietic progenitor cell Stem cells that are commit-
genes code for transferases, which reproduce their ted to produce blood cells.
respective antigens by attaching sugars to designated Hematuria Blood in the urine.
precursor substances.
Heme The iron-containing protoporphyrin portion of the
Goodpasture’s syndrome A disease entity that represents a hemoglobin wherein the iron is in the ferrous (Fe2+)
rapidly progressive glomerulonephritis associated with state.
pulmonary lesions. Usually the patients possess an anti-
Hemodialysis Removal of chemical substances from the
body to the basement membrane of the renal glomeruli.
blood by passing it through tubes made of semiperme-
Graft-versus-host (GVH) disease A disorder in which the able membranes that are continually bathed by solu-
grafted tissue attacks the host tissue. tions that selectively remove unwanted material; used
Granulocytopenia Abnormal reduction of granulocytes in to cleanse the blood of patients in whom one or both
the blood. kidneys are defective or absent and to remove excess
Griffith’s transformation A famous genetic experiment accumulation of drugs or toxic chemicals in the blood.
performed in 1928 by Frederick Griffith using two Hemodilution An increase in the volume of blood plasma,
strains of Streptococcus pneumoniae. Griffith demon- resulting in reduced relative concentration of red blood
strated that a heat-resistant “transforming principle” cells.
from one strain could change the virulence of another Hemoglobin The iron-conjugated protein in the red blood
strain. This was the first event that eventually led to cells whose function is to carry oxygen from the lungs
discovery of DNA as carrying the genetic information to the tissues. This protein contains heme plus globin.
on molecules.
Hemoglobinemia Presence of hemoglobin in the blood
Hageman’s factor Synonym for coagulation factor XII. plasma.
Half-life The time that is required for the concentration Hemoglobin-oxygen dissociation curve The relationship
of a substance to be reduced by one half. between the percent saturation of the hemoglobin
Haploid Possessing half the normal number of chromo- molecule with oxygen and the environmental oxygen
somes found in somatic or body cells; seen in germ tension.
cells (sperm and ova). Hemoglobinuria The presence of hemoglobin in the urine
Haplotype A term used in HLA testing to denote the five freed from lysed red blood cells, which occurs when
genes (HLA-A, HLA-B, HLA-C, HLA-D, HLA-DR) on the hemoglobin from disintegrating red blood cells or from
same chromosome. rapid hemolysis of red blood cells exceeds the ability of
Haptene The portion of an antigen containing the group- the blood proteins to combine with the hemoglobin.
ing on which the specificity depends. Hemolysin An antibody that activates complement,
Haptoglobin A mucoprotein to which hemoglobin leading to cell lysis.
released into plasma is bound; it is increased in certain Hemolysis Disruption of the red blood cell membrane and
inflammatory conditions and decreased in hemolytic the subsequent release of hemoglobin into the suspend-
disorders. ing medium or plasma.
Hardware Components of an information computer sys- Hemolytic anemia Anemia caused by hemolysis of red
tem that are the tangible, physical pieces of equipment, blood cells, resulting in reduction of normal red blood
such as the central processing unit, cathode ray tube, cell life span.
and keyboard. Hemolytic disease of the fetus and newborn (HDFN) A
HBcAg Hepatitis core antigen, referring to the nucleocapsid disease, characterized by anemia, jaundice, enlargement
of the virion. of the liver and spleen, and generalized edema (hydrops
HBeAg Hepatitis DNA polymerase of the nucleus of the fetalis), that is caused by maternal IgG antibodies cross-
virion. ing the placenta and attacking fetal red blood cells when
there is a fetomaternal blood group incompatibility
HBsAg Hepatitis B surface antigen.
(usually ABO or Rh antibodies). Synonym is erythro-
Hemangioma A benign tumor of dilated blood vessels. blastosis fetalis.
Hemarthrosis Bloody effusion into the cavity of a joint. Hemolytic transfusion reaction (HTR) A reaction from red
Hematinic Pertaining to blood; an agent that increases the blood cell destruction caused by patient’s antibody(ies)
amount of hemoglobin in the blood. directed to donor red blood cell antigen(s).
Hematocrit The proportion of red blood cells in whole Hemophilia A A hereditary disorder characterized by
blood, expressed as a percentage. greatly prolonged coagulation time ((PTT). The blood
Hematoma A swelling or mass of blood confined to an fails to clot and bleeding occurs; caused by inheritance
organ, tissue, or space and caused by a break in a blood of a factor VIII deficiency, it occurs almost exclusively
vessel. in males.
6888_Glossary_629-652 29/10/18 11:32 AM Page 639

Glossary 639

Hemophilia B “Christmas disease,” a hemophilia-like Hormone A substance originating in an organ or gland


disease caused by a lack of factor IX. that is conveyed through the blood to another part
Hemopoiesis Formation of blood cells. Synonym is of the body, chemically stimulating it to increase func-
hematopoiesis. tional activity and increase secretion.
Hemorrhage Abnormal internal or external bleeding; may Hoshin kanri In LEAN, a total quality management
be venous, arterial, or capillary; from blood vessels into principle to prioritize the elimination of waste via
the tissues or out of the body. policy deployment and control.
Hemorrhagic diathesis Uncontrolled spontaneous bleeding. Hyaluronidase An enzyme found in the testes; present in
semen.
Hemosiderin An iron-containing pigment derived from
hemoglobin from disintegration of red blood cells; a Hybrid gene A gene that results from combining two
method of storing iron until it is needed for making different genes.
hemoglobin. Hybridization Formation of hydrogen bonds between
Hemostasis Arrest of bleeding; maintaining blood flow complementary strands of nucleic acids to form either
within vessels by repairing rapidly any vascular break DNA:DNA or DNA:RNA hybrids. Chemically, a process
without compromising the fluidity of the blood. identical with renaturation or annealing. Nucleic acid
hybridization is a fundamental tool in molecular
Hemotherapy Blood transfusion as a therapeutic
genetics that takes advantage of the ability of individual
measure.
single-stranded nucleic acid molecules to form double-
Heparin An anticoagulant used for collecting whole blood stranded molecules—that is, to hybridize to each other.
that is to be filtered for the removal of leukocytes. The interacting single-stranded molecules must have a
Hepatitis Inflammation of the liver. sufficiently high degree of base complementarity.
Hepatitis-associated antigen (HAA) Older terminology Standard nucleic acid hybridization assays involve
currently replaced by HBsAg. using a labeled nucleic acid probe to identify related
DNA or RNA molecules—ones with a significantly
Hepatitis B immunoglobulin (HBIg) An immune serum
high degree of sequence similarity—within a complex
given to individuals exposed to the hepatitis B virus.
mixture of unlabeled nucleic acid molecules, the target
Hereditary spherocytosis An inherited anemia character- nucleic acid.
ized by fragile, spherical RBCs prone to hemolysis.
Hybridization protection (HPA) A molecular detection
The condition occurs due to mutations within genes
technology in which ssDNA probes labeled with
coding for cytoskeleton proteins like spectrin, ankyrin,
chemiluminescent molecules are added to form hybrids
or others.
with the amplified RNA molecules produced during
Heterogenous RNA (hnRNA) See pre-mRNA. transcription-mediated amplification (TMA). Light is
Heterozygote An individual with different alleles on a gene emitted upon hybridization of the probes to the RNA
for a given characteristic. and captured by a luminometer.
Heterozygous Possessing different alleles at a given gene Hybridoma A hybrid (cross) between a plasmacytoma
locus. cell and a spleen (or Ab-producing) cell that produces
High-prevalence antigen Also known as high-incidence a monoclonal antibody, resulting in a cell line that can
antigen; antigen whose frequency in the population is grow indefinitely in culture and can produce high
98% to 99%. quantities of Ab. This antibody is monoclonal because
only one Ab-producing cell combined with the plasma-
High protein anti-D Reagent anti-D consisting of human
cytoma cell is present.
source IgG anti-D with potentiators (bovine albumin
and macromolecular additives) to enhance antibody Hydatid cyst fluid Source of P1 substance.
reactivity on direct testing. Hydrocortisone A corticosteroid with anti-inflammatory
Histocompatibility The ability of cells to survive without properties.
immunologic interference; especially important in blood Hydrogen bonds Noncovalent, heat-labile chemical bonds
transfusion and transplantation. that occur between a hydrogen atom and either a nitrogen
HLA Human leukocyte antigen. or oxygen atom of the adjacent molecule. In DNA, hydro-
gen bonding holds together the two strands of double
Homeostasis State of equilibrium of the internal environ-
helix by formation of two bonds between adenine and
ment of the body that is maintained by dynamic
thymine and three bonds between cytosine and guanine.
processes of feedback and regulation.
Hydrops fetalis See Hemolytic disease of the fetus and
Homozygote An individual developing from gametes with
newborn (HDFN).
similar alleles and thus possessing like pairs of genes
for a given hereditary characteristic. Hydroxyethyl starch (HES) A red blood cell sedimenting
agent used to facilitate leukocyte withdrawal during
Homozygous Possessing a pair of identical alleles.
leukapheresis.
6888_Glossary_629-652 29/10/18 11:32 AM Page 640

640 Glossary

Hypertension Increase in blood pressure. Immunoglobulin (Ig) One of a family of closely related
Hyperventilation Rapid breathing that results in carbon though not identical proteins that are capable of acting
dioxide depletion and that accompanies hypotension, as antibodies: IgA, IgD, IgE, IgG, and IgM. IgA is the
vasoconstriction, and fainting. principal immunoglobulin in exocrine secretions such
as saliva and tears. IgD may play a role in antigen
Hypogammaglobulinemia Decreased levels of gamma
recognition and the initiation of antibody synthesis.
globulins seen in some disease states.
IgE, produced by the cells lining the intestinal and
Hypotension Decrease in blood pressure. respiratory tracts, is important in forming reagin. IgG
Hypothermia Having a body temperature below normal. is the main immunoglobulin in human serum. IgM is
Hypovolemia Diminished blood volume. formed in almost every immune response during the
early period of the reaction.
Hypoxia Deficiency of oxygen.
Immunologic memory The development of T and B mem-
Iberis amara See Anti-M lectin.
ory cells that have been sensitized by exposure to an
Icterus A condition characterized by yellowish skin, antigen and that respond rapidly under subsequent
whites of the eyes, mucous membranes, and body fluids encounters with the antigen.
caused by increased circulating bilirubin resulting from
Immunologic unresponsiveness Development of a toler-
excessive hemolysis or from liver damage due to hepati-
ance to certain antigens that would otherwise evoke an
tis. Synonym is jaundice.
immune response.
Idiopathic Pertaining to conditions without clear patho-
Immunoprecipitin An antigen-antibody reaction that
genesis, or disease without recognizable cause, as of
results in precipitation.
spontaneous origin.
Inclusion The opposite of an exclusion, in which all of
Idiopathic thrombocythemia An increase in blood
the genetic information inherited by the child from the
platelets of unknown etiology.
parent whose identity is in question is present in the
Idiopathic thrombocytopenic purpura (ITP) Bleeding alleged parent.
owing to a decreased number of platelets; the etiology
Incubation In vitro combination of antigen and antibody
is unknown, with most evidence pointing to platelet
under certain conditions of time and temperature to
autoantibodies.
allow antigen-antibody complexes to occur.
Idiotype The portion of the immunoglobulin variable re-
Indirect transfusion Transfusion of blood from a donor
gion that is the antigen-combining site, which interacts
to a suitable storage container and then to a patient.
with the antigenic epitope.
Initiation The deposition of N-formylmethionine on the
Immune response The reactions of the body to substances
ribosome, which begins the synthesis of all proteins.
that are foreign or are interpreted as being foreign.
Cell-mediated or cellular immunity pertains to tissue In Lu A rare dominant gene that inhibits the production
destruction mediated by T cells, such as graft rejection of all Lutheran antigens as well as i, P1, and Aua
and hypersensitivity reactions. Humoral immunity (Auberger). The quantity of antigen on the red blood
pertains to cell destruction response during the early cell is markedly reduced in the presence of In Lu; it
period of the reaction. may be virtually undetectable.
Immune serum globulin Gamma globulin protein fraction Interface Software that allows a computer system to send
of serum-containing antibodies. data to or receive data from another computer system.
Immunoblast A mitotically active T or B cell. Intergenic recombination Recombination of genetic
material in noncoding regions of DNA.
Immunoblotting A technique for detecting proteins
extracted from cells, separated by polyacrylamide gel Intracellular Occurs within a cell.
electrophoresis, transferred from the gel onto a filter Intraoperative salvage A procedure to reclaim a patient’s
membrane and then incubated with a specific labeled blood loss from an operation by reinfusion.
antibody. Intrauterine transfusion Transfusion of blood into a fetus
Immunodeficiency A decrease from the normal concentra- in utero.
tion of immunoglobulins in serum. Intravascular Within the blood vessel.
Immunodominant sugar In reference to glycoprotein or Intron A noncoding part of the gene. This fragment of the
glycolipid antigens, the sugar molecule that gives the gene is transcribed into RNA but is excised out during
antigen its specificity (e.g., galactose, which confers the process of splicing so it is not translated into a
B antigen specificity). protein product.
Immunogen Any substance capable of stimulating an In utero Within the uterus.
immune response.
Inversion The breaking of a chromosome during division,
Immunogenicity The ability of an antigen to stimulate an with subsequent reattachment occurring in an inverted
antibody response. or upside-down position.
6888_Glossary_629-652 29/10/18 11:32 AM Page 641

Glossary 641

In vitro Outside the living body, as in a laboratory develops into a genetically modified organism missing
setting. the desired structural or functional protein.
In vivo Inside the living body. Labile Capable of deteriorating rapidly upon storage.
Ion exchange resin Synthetic organic substances of Lambda (λ) vectors Vectors that contain the part of the
high molecular weight. They replace certain positive bacterial virus Lambda genome necessary for lytic
or negative ions, which they encounter in solutions. replication in Escherichia coli and one or more restric-
Ionic strength Refers to the number of charged particles tion sites for insertion of the DNA fragment of interest.
present in a solution. They can carry foreign DNA 5 to 14 kb long. The re-
combinant DNA is “packaged” into viral particles and
Ir genes Immune response genes found within the region
used to infect E. coli in order to produce multiple copies
of the major histocompatibility complex. Ir genes in
of the fragment.
humans are likely to exist; preliminary evidence shows
genes at the D-related locus may be analogous to the LEAN A systematic approach for identifying actions
Ir genes of mice. within a process that create value, then optimizing the
alignment of these actions to the customer.
Iron-deficiency anemia Anemia resulting from a greater
demand on stored iron than can be met. Lectin Proteins present in plants (usually seeds) that
bind specifically to carbohydrate determinants and
Irradiation Gamma or electron treatment of a cellular
agglutinate erythrocytes through their cell surface of
blood product for protection against graft-versus-host
oligosaccharide determinants.
disease.
Leukemia Malignant proliferation of leukocytes, which
Ischemia Local and temporary deficiency of blood supply
spill into the blood, yielding an elevated leukocyte
caused by obstruction of the circulation to a cell, tissue,
count.
or organ.
Leukoagglutinin Antibodies to white blood cells.
Isoagglutinins The ABO antibodies anti-A, anti-B, and
anti-A,B. Ligases A class of enzymes that catalyze the linkage of
two molecules, generally utilizing ATP as the energy
Isoimmune An antibody produced against a foreign
donor. In recombinant DNA technology, these are
antigen in the same species.
enzymes linking the adjacent nucleotides between two
Isotype The subclasses of an immunoglobulin molecule. DNA fragments being joined after restriction enzyme
Jaundice See Icterus. digestion.
Kaizen In LEAN, a continuous improvement process Ligature Process of binding or tying; a band or bandage; a
required for long-term success. thread or wire for tying a blood vessel or other structure
Karyotype A photomicrograph of a single cell in the in order to constrict it.
metaphase stage of mitosis that is arranged to show LightCycler A special type of thermocycler with built-
the chromosomes in descending order of size. in fluorescence detector, designed for real-time PCR
kb (or kbp) A measure of DNA length. One thousand applications.
(“kilo”) nucleotides (or “bases” for simplification) in Linkage The association between distinct genes that occupy
single-stranded DNA. In double-stranded DNA, this is closely situated loci on the same chromosome, resulting
referred to as 1,000 base pairs. in an association in the inheritance of these genes.
Kernicterus A form of icterus neonatorum occurring in Linkage disequilibrium Genes associated in a haplotype
infants, developing at 2 to 8 days of life; prognosis poor more often than would be expected on the basis of
if untreated. This condition is due to an increase in chance alone.
unconjugated bilirubin. Locus The site of a gene on a chromosome.
Kinin A group of polypeptides that have considerable Low ionic-polycation test A compatibility test that
biological activity (e.g., vasoactivity). incorporates both glycine (low ionic) and protamine
Kleihauer-Betke technique Quantitative procedure used (polycation) in an effort to obtain maximal sensitivity
to determine the amount of fetal cells present in the and to minimize the need for antibody screening.
maternal circulation. Low ionic strength solution (LISS) A type of potentiating
Km Light chain marker on the kappa light chains of IgG medium in use for serologic testing. Reducing the ionic
(formerly known as InV). strength of the red blood cell–suspending medium in-
Knockout transgenic animal A model to study the conse- creases the affinity of the antigen for its corresponding
quences of lack of function of a gene that is blocked, antibody such that sensitivity can be increased and
or “knocked out” (e.g., by RNA interference) and incubation time decreased. LISS contains glycine or
introduced into the egg using transgenic technology glucose in addition to saline.
(a microinjection of external/modified genetic material). Low melting point agarose Agarose specially formulated to
The egg, upon implantation into a surrogate mother, melt in temperatures much lower than normal, which
6888_Glossary_629-652 29/10/18 11:32 AM Page 642

642 Glossary

allows for easy dissolving of the gel in order to isolate contain half the number of chromosomes present in
and purify DNA that was run in it. somatic cells.
Low-prevalence antigen Also known as low-incidence anti- Melting curve analysis A postamplification analysis of
gen; antigen whose frequency in a random population is the amplicon produced in the real-time PCR by
very low—less than 10%. slowly increasing the temperature and monitoring the
Luminometer An instrument that detects and quantifies decrease of fluorescence as a result of DNA strand
the amount of light emitted as a result of chemical separation from the probes used in the reaction. The
reaction (chemiluminescence). fluorescent probes detach from the PCR product at
lower temperatures if the product has a mutation.
Lymphocyte A type of white blood cell involved in the
This method may be used to distinguish between
immune response. Lymphocytes normally total 20% to
homo- and heterozygosity underlying certain
45% of total white blood cells. T lymphocytes mature
conditions (the presence of the mutation in one or
during passage through the thymus or after interaction
both homologous chromosomes).
with thymic hormones; these cells function both in
cellular and humoral immunity. Subsets include helper Melting point (Tm) The temperature at which half of all
T cells (Th), which enhance B-cell antibody production, hydrogen bonds in the double-stranded DNA molecule
and suppressor T cells (Ts), which inhibit B-cell anti- are broken, while the other half are still intact. It deter-
body production. B-lymphocyte cells are not processed mines how much energy is needed to keep the strands
by the thymus. Through morphologic and functional of the helix apart. As a general rule, temperatures above
differentiation, they mature into plasma cells that se- the Tm promote the separation of the strands. Knowing
crete immunoglobulin. the Tm is necessary for calculating temperatures for
hybridization-based assays and for annealing primers
Lymphoma A solid tumor of lymphocyte cells.
in the polymerase chain reaction. The primers generally
Lysosomes Part of an intracellular digestive system that anneal to DNA at about 5°C (or more) below the Tm.
exists as separate particles in the cell. Even though If the temperature is decreased more, they will start to
their importance in health and disease is certain, all anneal to sequences with only partial complementarity,
the precise ways lysosomes effect changes are not which affects the specificity of the reaction. For short
understood. DNA sequences, the simplified formula to calculate the
Macroglobulinemia Abnormal presence of high-molecular- Tm is 2AT + 4GC, where AC is the total number of
weight immunoglobulins (IgM) in the blood. adenines and thymines and GC is the total number of
Macrophages End-stage development for the blood mono- guanines and cytosines in the molecule. Based on this
cyte; these cells can ingest (phagocytose) a variety of formula, it is evident that GC-rich molecules will have
substances for subsequent digestion or storage and are higher Tm than AT-rich sequences because there are
located in a number of sites in the body (e.g., spleen, always three hydrogen bonds between guanines and
liver, lung), existing as free mobile cells or as fixed cells. cytosines, while there are only two bonds between
Functions include elimination of senescent blood cells adenines and thymines.
and participation in the immune response. Mendel’s laws The classical principles of inheritance de-
Major ABO incompatibility ABO antibody in the recipient fined by the Moravian monk Gregor Mendel in 1866.
that is incompatible with the donor. The first law of genetics (the law of segregation) stated
that the hereditary characteristics are determined by
Major histocompatibility complex (MHC) Present in all
particulate units (presently called genes) that occur in
mammalian and ovarian species; analogous to HLA
an individual as pairs (diploid), but in the formation of
complex. HLA antigens are within the MHC at a locus
germ cells/gametes they segregate so the gamete con-
on chromosome 6.
tains only one member of the pair (haploid). The sec-
Malaria An acute and sometimes chronic infectious disease ond law (the law of independent assortment) stated that
caused by the presence of a parasite within red blood the particulate units (genes) that determine different
cells. The parasite is Plasmodium (Plasmodium vivax, characteristics are inherited independently of other
Plasmodium falciparum, Plasmodium malariae, Plasmod- units (now we know that if the genes are close to one
ium ovale), which is introduced through bites of in- another on the chromosome, they will be inherited
fected female Anopheles mosquitoes or through blood together).
transfusion.
Menorrhagia Excessive menstrual bleeding in number of
Maternal antibody Antibody produced in the mother and days or amount of blood or both.
transferred to the fetus in utero.
2-Mercaptoethanol (2-ME) A sulfhydryl compound used
Megaloblastic anemia Anemia in which megaloblasts are to disrupt the disulfide bonds of immunoglobulin M,
found in the blood. yielding monomeric units rather than the typical
Meiosis Type of cell division of germ cells in which two pentameric units.
successive divisions of the nucleus produce cells that Messenger RNA (mRNA) See Ribonucleic acid.
6888_Glossary_629-652 29/10/18 11:32 AM Page 643

Glossary 643

Metastasis Movement of bacteria or body cells, especially amplicon, occurs upon unfolding of the loop structure,
cancer cells, from one part of the body to another; which physically separates the quencher.
change in location of a disease or of its manifestations Molecular cloning Reproduction of recombinant DNA
or transfer from one organ or part of another not molecules in host cells that, due to universality of the
directly connected. Spread is by the lymph or blood genetic code, replicate the foreign DNA along with its
circulation. own. The daughter cells of a single cell carrying the
Methemoglobin An abnormal form of hemoglobin wherein recombinant molecule propagate to produce a clone.
the ferrous (Fe2+) iron has been oxidized to ferric (Fe3+) Monoclonal Antibody derived from a single ancestral
iron. antibody-producing parent cell.
Methyldopa (Aldomet) Common drug used to treat hyper- Monocytes See Macrophage.
tension; frequently the cause of a positive direct
Monospecific antiglobulin reagent Antiserum specific
Coombs’ test result.
for one type of immunoglobulin (e.g., anti-IgG).
Microaggregates Aggregates of platelets and leukocytes
Monozygotic twins Two offspring that develop from a
that accumulate in stored blood.
single fertilized ovum.
Microarrays (gene chips) Devices consisting of glass slides
Mosaic An antigen composed of several subunits, such as
or membrane filters with fragments of DNA printed by a
the Rh0(D) antigen. A mixture of characteristics that
robot in small spots at high density. These DNA frag-
may result from a genetic crossover or mutation.
ments serve as multiple probes that will bind (hybridize
with) the complementary regions of DNA (or RNA) Multiparous Having borne more than one child.
present in the analyzed specimen applied to the chip. Multiple myeloma A neoplastic proliferation of plasma cells,
The chips are scanned with a laser to identify the re- which is characterized by very high immunoglobulin
gions detected by the probes. levels of monoclonal origin.
Microglobulin (β2) A protein synthesized by all nucleated Multiplex assay In molecular biology, refers primarily to
cell types; an integral part of the class I MHC antigens. multiplex polymerase chain reaction (PCR). Multiplex
Microsatellites DNA regions composed of back-back- PCR is a concurrent amplification of several DNA frag-
repeated units of 1 to 8 nucleotides (also called short ments from the same DNA target using several sets of
tandem repeats; STRs). primers that must be carefully designed so that they
don’t interfere with each other.
Microspherocytes Red blood cells, small and spherical, in
certain kinds of anemia (i.e., hereditary spherocytosis). Mutation A change in a gene potentially capable of being
transmitted to offspring.
Miniprep A rapid method for making a small preparation
of purified plasmid DNA from 1 to 5 mL of bacterial Myelofibrosis Replacement of bone marrow by fibrous
culture. tissue.
Minisatellites DNA regions composed of back-to-back re- Myeloproliferative An autonomous, purposeless increase
peated units ranging in size from 9 to 80 bp (also called in the production of the myeloid cell elements of
VNTR; variable number of tandem repeats). the bone marrow that includes granulocytic, erythro-
cytic, and megakaryocytic cell lines as well as the
Minor ABO incompatibility ABO antibody in the donor
stromal connective tissue.
that is incompatible with the recipient.
N-acetylneuraminic acid (NANA) See Sialic acid.
Mitosis Type of cell division in which each daughter cell
contains the same number of chromosomes as the par- Naturally occurring antibody Antibody present in
ent cell. All cells except sex cells undergo mitosis. a patient without known prior exposure to the
corresponding red blood cell antigen.
Mixed-field agglutination A type of agglutination pattern
in which numerous small clumps of cells exist amid a Neonatal alloimmune neutropenia (NAN) A rare condi-
sea of free cells. This usually occurs when there is more tion that occurs when a mother becomes sensitized
than one population of RBCs present in the sample to a foreign antigen of paternal origin that is present
(e.g., in a recently transfused individual). on fetal granulocytes. These fetal granulocyte antigens
provoke antibody production. The maternal IgG anti-
MLC Mixed lymphocyte culture.
body crosses the placenta and destroys fetal granulo-
MLR Mixed lymphocyte reaction. cytes. This neutropenia is typically self-limiting and
Modem Hardware device that provides the ability to attach lasts for several weeks but can persist for as long as
to a computer system via telephone communication 6 months. During this period, neonates are at high risk
lines. of developing infections.
Molecular beacons Fluorescent DNA probes structured Neonate A newborn infant up to 4 months of age.
like a hairpin loop with reporter and quencher; used Network Configuration of personal computers linked to-
in real-time PCR. The emission of fluorescent light gether with cables; allows all of the personal computers
from the reporter, indicating the presence of an
6888_Glossary_629-652 29/10/18 11:32 AM Page 644

644 Glossary

to access common data and software located on a file Orthostatic Concerning an erect position.
server. Osmolality The osmotic concentration of a solution
Neuraminidase An enzyme that cleaves sialic acid from the determined by the ionic concentration of dissolved
red blood cell membrane. substances per unit of solvent.
Neutralization Inactivating an antibody by reacting it with Ouchterlony diffusion An immunologic procedure in
an antigen, against which it is directed. which antibody and antigen are placed in wells of a
Neutrophil A leukocyte that ingests bacteria and small gel medium plate and allowed to diffuse in order to
particles and plays a role in combating infection. visualize the reaction by a precipitin line.
Nondisjunction Failure of a pair of chromosomes to Oxyhemoglobin The combined form of hemoglobin and
separate during meiosis. oxygen.
Nonresponder An individual whose immune system does P50 The partial pressure of oxygen or oxygen tension at
not respond well in antibody formation to antigenic which the hemoglobin molecule is 50% saturated with
stimulation. oxygen.
Normal serum albumin See Albumin. PAGE Polyacrylamide gel electrophoresis.
Northern blotting A variation of Southern blotting where, Palindromic sequences Four to 8 nucleotide DNA se-
instead of DNA, RNA is run on the electrophoretic gel quences that read the same in the 5⬘ to 3⬘ direction on
and transferred onto a blot for detection of a specific both strands of the double helix—for example,
RNA sequence. 5⬘AAATTT3⬘ or 5⬘GAATTC3⬘.
Nucleic acid testing (NAT) General term used to describe Pallor Paleness; lack of color.
diagnostic tests (predominately amplification tests: Panagglutinin An antibody capable of agglutinating all red
NAATs) based on detection of nucleic acid sequences blood cells tested, including the patient’s own cells.
unique for the organism (pathogen) in question. Pancytopenia A reduction in all cellular elements of the
Nucleosides Compounds consisting of one of the nitrogen- blood, including red blood cells, white blood cells, and
containing bases (A, T or U, C, and G) and sugar de- platelets.
oxyribose or ribose. Depending on the base (adenine, Panel A large number of group O reagent red blood cells
thymine, uracil, cytosine, and guanine), the nucleosides that are of known antigenic characterization and are
are named: adenosine, thymidine, uridine, cytidine, used for antibody identification.
and guanosine. Upon phosphorylation, they become
Papain A proteolytic enzyme derived from papaya.
nucleotides and are building blocks of nucleic acids:
DNA or RNA. Paragloboside The immediate precursor for the H and
P antigens of the red blood cell.
Nucleotides Compounds consisting of one of the
nitrogen-containing bases (A, T or U, C, and G), Parentage testing The analysis of one or more genetic
sugar deoxyribose or ribose, and one, two, or three markers in a group of individuals thought to be related
phosphates. Chemically, they are referred to as nucleo- as parent and child. The group normally consists of a
side phosphates. Nucleotides are the building blocks of known parent (often a mother), a child, and an alleged
nucleic acids: DNA or RNA. parent (often an alleged father). The goal of the testing
is to confirm or refute the alleged relatedness of the
Oh See Bombay.
tested individuals as parents and child.
Oligo-dT primers Universal primers starting cDNA
Paroxysm A sudden, periodic attack or recurrence of
synthesis from messenger RNA (mRNA) in the process
symptoms of a disease.
of reverse transcription. These primers consist of
several thymine nucleotides that are complementary to Paroxysmal cold hemoglobinuria (PCH) A type of cold
the poly-A tail of mRNA. These primers will not start autoimmune hemolytic anemia usually found in chil-
cDNA synthesis from rRNA or tRNA because these dren suffering from viral infections in which a biphasic
molecules are missing the poly-A tail. immunoglobulin G antibody can be demonstrated with
anti-P specificity. See also Donath-Landsteiner test.
Oligonucleotide A short synthetic segment of DNA, ap-
proximately 20 nucleotides in length, used as a probe. Paroxysmal nocturnal hemoglobinuria (PNH) An intrinsic
defect in the red blood cell membrane, rendering it
Oliguria Diminished amount of urine formation.
more susceptible to hemolysins in an acid environment;
Opsonin A substance in serum that promotes immune characterized by hemoglobin in the urine following
adherence and facilitates phagocytosis by the periods of sleep.
reticuloendothelial system.
Partial D Red blood cells that are missing normal RhD
Origin of replication (Ori) In DNA, the sequence that epitopes, resulting in a qualitative difference in the
signals the beginning of replication. In plasmids, they RhD protein. Individuals with partial D can make
drive the replication of the foreign DNA fragments anti-D.
along with their own.
6888_Glossary_629-652 29/10/18 11:32 AM Page 645

Glossary 645

Passenger lymphocytes Donor lymphocytes in the Plasmapheresis A procedure using a machine to remove
transplanted organ or HPC (hereditary progenitor cell) only plasma from a donor or patient.
product. Plasma protein fraction (PPF) Also known as Plasmanate;
Paternity index Term that refers to a statement of “weight” sterile pooled plasma stored as a fluid or freeze-dried
concerning the probability a tested individual, who and used for volume replacement.
cannot be excluded as the parent of the child, is the true Plasmid Bacterial circular genetic element, 2 to 4 kb long,
parent. The paternity index represents a likelihood ratio that replicates independently from the chromosome.
that compares two mutually exclusive hypotheses. The Used as vectors in recombinant DNA technology to
numerator of the ratio reflects the probability the tested carry up to 15 kb foreign DNA into host cells. A vast
alleged parent is the true parent of the child and the selection of plasmids are commercially available that are
denominator reflects the probability someone random useful for different purposes, such as DNA sequencing,
and unrelated to the alleged parent is the true parent. protein expression in bacteria, and protein expression in
Patient blood management A multidisciplinary systems- mammalian cells.
based process encompassing all processes related to Plasminogen A protein in many tissues and body fluids
blood transfusion to limit and prevent inappropriate or important in preventing fibrin clot formation.
unnecessary transfusions while promoting strategies to
Plasmodium See Malaria.
reduce the overall transfusion requirements of patients.
Also: a multidisciplinary process to apply strategies to Plasmodium knowlesi A parasite that causes malaria in
avoid or at least minimize a patient’s transfusion needs monkeys.
from the preoperative period through discharge. Platelet A round or oval disk, 2 to 4 ␮m in diameter, that
Perfusion Supplying an organ or tissue with nutrients is derived from the cytoplasm of the megakaryocyte, a
and oxygen by passing blood or another suitable fluid large cell in the bone marrow. Platelets play an impor-
through it. tant role in blood coagulation, hemostasis, and blood
thrombus formation. When a small vessel is injured,
Perioral paresthesia Tingling around the mouth occasion-
platelets adhere to each other and to the edges of the
ally experienced by apheresis donors, resulting from
injury, forming a “plug” that covers the area and
the rapid return of citrated plasma, which contains
initially stops the blood loss.
citrate-bound calcium and free citrate.
Platelet concentrate Platelets prepared from a single unit
Peroxidase An enzyme that hastens the transfer of oxygen
of whole blood or plasma and suspended in a specific
from peroxide to a tissue that requires oxygen; this
volume of the original plasma; also known as random-
process is essential to intracellular respiration.
donor platelets.
Phagocytosis Ingestion of microorganisms, other cells,
Plateletpheresis A procedure using a machine to remove
and foreign particles by a phagocyte.
only platelets from a donor or patient.
Phenotype The outward expression of genes (e.g., a blood
Platelet refractoriness Failure to yield an increase in
type). On blood cells, serologically demonstrable anti-
recipient’s platelet count on transfusion of suitably
gens constitute the phenotype, except those sugar sites
preserved platelets. HLA alloimmunization is a common
that are determined by transferases.
cause.
Phenylthiocarbamide (PTC) A chemical used in studying
Point mutation A change in a base in DNA that can lead
medical genetics to detect the presence of a marker
to a change in the amino acid incorporated into the
gene. About 70% of the population inherits the ability
polypeptide; identifiable by analysis of the amino
to taste PTC, which tastes bitter; the remaining 30%
acid sequences of the original protein and its mutant
finds PTC tasteless. The inheritance of this trait is due
offspring.
to a single dominant gene of a pair.
Polarity (3⬘ and 5⬘ ends) Also called DNA “directionality,”
Phlebotomy The procedure used to draw blood from a
determines the direction of DNA replication and tran-
person.
scription. Results from the antiparallel way that the two
Phosphoglyceromutase A red blood cell enzyme. strands of nucleotides ran in opposite directions. The
Phototherapy Exposure to sunlight or artificial light for labels 3⬘ and 5⬘ refer to the number assigned by conven-
therapeutic purposes. tion to the deoxyribose carbon atom linked to
Plasma The liquid portion of whole blood, containing either hydroxyl or phosphate group in DNA molecule.
water, electrolytes, glucose, fats, proteins, and gases. Polyacrylamide gel A polymer of acrylamide, used as a
Plasma contains all the clotting factors necessary for matrix for gel electrophoresis that provides better
coagulation but in an inactive form. Once coagulation resolution than agarose electrophoresis.
occurs, the fluid is converted to serum. Polyagglutination A state in which an individual’s red
Plasma cell A B lymphocyte–derived cell that secretes blood cells are agglutinated by all sera, regardless of
immunoglobulins or antibodies. blood type.
6888_Glossary_629-652 29/10/18 11:32 AM Page 646

646 Glossary

Polyagglutinins Naturally occurring immunoglobulin protein. When added to a solution of the antigen, it
antibodies that are found in most normal human brings about precipitation. The injected protein is the
adult sera. antigen; the antibody produced is the precipitin.
Poly-A tail A long chain of adenine nucleotides that is Precursor substance A substance that is converted to
added in the reaction of polyadenylation to a messenger another substance by the addition of a specific
RNA (mRNA) molecule during RNA processing to constituent (e.g., a sugar residue).
increase the stability of the molecule immediately Pre-mRNA Immature messenger RNA right after synthesis
after a gene in a eukaryotic cell is transcribed. in the nucleus. Also called heterogeneous RNA
Polybrene A positively charged polymer that causes (hnRNA) because it consists of sequences transcribed
normal red blood cells to aggregate spontaneously by from both exons and introns. The introns (the noncod-
neutralizing the negative surface charge contributed by ing fragments) are removed during mRNA processing
sialic acid. (maturation) by excision (splicing).
Polyclonal Antibodies derived from more than one Preseroconversion window A period of time during which
antibody-producing parent cell. a person may be infected with a viral pathogen (HIV,
Polycythemia vera A chronic life-shortening myeloprolifer- HCV, HBV, etc.) but does not yet produce levels of
ative disorder involving all bone marrow elements, antibodies detectable by serologic methods.
characterized by an increase in red blood cell mass and Pretransfusion compatibility testing Series of testing
hemoglobin concentration. procedures and processes with the ultimate objective of
Polylinker In plasmid, a region with a series of recognition ensuring the best possible results of a blood transfusion,
sequences for different restriction endonucleases that including recipient and donor identification, ABO
may be used to introduce a foreign DNA fragment testing, clerical checks, and so on.
cut out of its original source with the corresponding Primer A short segment of single-stranded DNA, usually
enzymes. 17 to 25 nucleotides long, used to initiate DNA
Polymer Combination of two or more molecules of the replication in PCR.
same substance. Primers (forward and reverse) Short, synthetic segments
Polymerase chain reaction (PCR) An in vitro method of single-stranded DNA, usually 15 to 25 nucleotides
of amplifying a specific fragment of DNA using ther- long, used to initiate DNA replication in PCR. Primers
mostable DNA polymerase enzyme and short synthetic are designed to anneal to complementary DNA se-
primers that attach to separated strands of DNA and quences at the 3⬘ end of each strand of the DNA frag-
are extended by the addition of deoxyribonucleotides ment desired to be amplified and extended toward the
in numerous cyclic changes of temperature. Each 5⬘ end. The forward primer anneals to the antisense
PCR cycle consists of denaturation, annealing, and strand, and the reverse primer anneals to the sense
extension (elongation). strand.
Polymorphism A genetic system that possesses numerous Private antigen An antigenic characteristic of the red
allelic forms, such as a blood group system. blood cell membrane that is unique to an individual
or a related family of individuals and therefore is not
Polyspecific Coombs’ sera A reagent that contains
commonly found on all cells (usually less than 1% of
antihuman globulin sera against immunoglobulin G
the population).
and C3d.
Probability of exclusion (PE) Refers to the strength of
Polyvinylpyrrolidone (PVP) A neutral polymeric sub-
the test battery to exclude a falsely accused alleged
stance used to increase blood volume in patients with
parent.
extensive blood loss; also used to enhance antigen-
antibody reactions in vitro. Probability of parentage The probability of parentage is
produced from the likelihood ratio (paternity index
Portal hypertension Increased venous pressure in the
for example) using Bayesian statistical logic and reflects
portal vein as a result of obstruction of the flow of
the level of conviction that the included alleged parent
blood through the liver.
is the true parent of the child.
Postpartum Occurring after childbirth.
Probe A fragment of DNA that is labeled and hybridized
Potentiator A substance that, when added to a serum and to diagnostic material to locate a complementary
cell mixture, will enhance antigen-antibody interactions. strand of DNA.
Precipitation The formation of a visible complex (pre- Prodrome A symptom indicative of an approaching
cipitate) in a medium containing soluble antigen disease.
(precipitinogen) and the corresponding antibody
Propositus The initial individual whose condition led to
(precipitin).
investigation of a hereditary disorder or to a serologic
Precipitin An antibody formed in the blood serum of an evaluation of family members. Feminine form is
animal by the presence of a soluble antigen, usually a proposita. Synonyms are proband and index case.
6888_Glossary_629-652 29/10/18 11:32 AM Page 647

Glossary 647

Prospective blood utilization review A blood utilization upon exposure to cold or emotional stress. A history of
review that occurs prior to transfusion events and is symptoms for at least 2 years is necessary for diagnosis.
often used to determine the appropriateness of a trans- Real-time PCR A variation of polymerase chain reaction
fusion order request. in which the product formed during each cycle of am-
Prospective validation Validation testing of software; done plification is detected by fluorescence at the same time
before implementation of the computer system. that it is produced, instead of being detected after
Prosthesis An artificial substitute for a missing part, such the reaction is finished. In addition to primers, the
as an artificial extremity. real-time reaction mixture contains DNA probes com-
plementary to the region between the primers (called
Protamine A polycation with applications similar to those
TaqMan or molecular beacons or FRET probes) labeled
of polybrene.
with fluorophores that emit fluorescent light when
Protamine sulfate A substance used to neutralize the binding to the newly synthesized amplicon.
effects of heparin.
Recessive A type of gene that, in the presence of its domi-
Prothrombin complex A concentrate of coagulation factors nant allele, does not express itself; expression occurs
II, VII, IX, and X in lyophilized form. when it is inherited in the homozygous state.
Provirus A DNA form of a retrovirus upon integration Recipient A patient who is receiving a transfusion of blood
into the genome of the infected (host) cell. or a blood product.
Prozone Incomplete lattice formation caused by an excess Recombinant DNA technology A process of recombining
of antibody molecules relative to the number of antigen two DNA fragments from different species and inserting
sites, resulting in false-negative reactions. such recombinant molecule into a host organism in
PRP Platelet-rich plasma. order to produce new genetic combinations that are of
Public antigen An antigen characteristic of the red blood value in medicine, science, and industry.
cell membrane found commonly among individuals, Recombinant proteins Proteins produced by translation of
usually more than 98% of the population. recombinant genes (created by the recombinant DNA
Pulmonary artery wedge pressure Pressure measured in technology).
the pulmonary artery at its capillary end. Refractory Obstinate; stubborn; resistant to ordinary
Pulse pressure The difference between the systolic and the treatment; resistant to stimulation (said of a muscle or
diastolic pressures. nerve).
Quality assurance (QA) A set of planned actions to Relationship testing See Parentage testing.
provide confidence that systems and elements that Renaturation See Annealing.
influence the quality of the product or service are Replication The process of DNA synthesis based on the
working as expected, individually and collectively. existing DNA molecule sequence (template) by enzyme
Radioimmunoassay (RIA) A very sensitive method for DNA polymerase, which recognizes the 3⬘ end of the
determining substances present in low concentrations template and starts adding the nucleotides, synthesizing
in serum or plasma by using specific antibodies and the new strand in the 5⬘ to 3⬘ direction (See polarity).
radioactively labeled or tagged substances. The place where the two original strands are separating
Random man not excluded (RMNE) Refers to the opposite is called a replication fork—one strand is synthesized in
of probability of exclusion, which is the probability that a continuous manner and is called a leading strand,
someone who is falsely accused of being the parent while the other strand’s synthesis occurs in short frag-
would not be excluded by a particular genetic test. The ments called Okazaki fragments, named after a Japanese
relationship between PE and RMNE is PE + RMNE = 1. researcher. This strand is called a lagging strand because
it is formed slightly slower due to the polymerase
Random primers (random hexamers) Six-nucleotide-long
constantly jumping toward the fork. DNA replication
sequences used as cDNA (See complementary DNA)
is said to be semiconservative.
synthesis primers in the process of reverse transcription.
Random primers will attach to any RNA molecule pres- Respiratory distress syndrome (RDS) A condition, formerly
ent in the reaction tube. The resulting cDNA represents known as hyaline membrane disease, accounting for more
the total RNA isolated from the cell, not just mRNA, than 25,000 infant deaths per year in the United States.
which is reverse transcribed using oligo-dT primers. Clinical signs, including delayed onset of respiration and
low Apgar score, are usually present at birth.
Rapid passive hemagglutination assay (RPHA) A third-
generation procedure used in hepatitis testing. Restriction endonucleases Bacterial enzymes that cleave
DNA at specific sequences and allow scientists to cut
Rapid passive latex assay (RPLA) A second-generation
DNA in a controlled and predictable way.
procedure used in hepatitis testing.
Restriction enzyme mapping Finding the sequences in
Raynaud’s disease A peripheral vascular disorder charac-
a given DNA recognized by restriction enzymes and
terized by abnormal vasoconstriction of the extremities
establishing the distance between such sites.
6888_Glossary_629-652 29/10/18 11:32 AM Page 648

648 Glossary

Restriction fragment length polymorphism (RFLP) Refers Rhnull Rare Rh phenotype in which no Rh antigens are ex-
to a specific molecular tool that can be used to demon- pressed on the red blood cell; results from a mutation in
strate nucleotide sequence polymorphisms in chromo- the RHAG gene (regulator-type Rhnull) or by a deletion
somal DNA. A single nucleotide polymorphism can of the RHD gene (the amorphic type) and a mutation in
alter the spatial arrangement of restriction enzyme RHCE genes.
recognition sequences in chromosomal DNA that will Rh-positive Red blood cells possessing one particular Rh
be revealed following restriction enzyme digestion as antigen, the D antigen.
an altered pattern of restriction fragments.
Ribonucleic acid (RNA) A nucleic acid that controls pro-
Reticulocyte Also known as neocyte, the last stage of devel- tein synthesis in all living cells. There are three different
opment before becoming a mature erythrocyte. The types, and all are derived from the information encoded
reticulocyte has lost its nucleus but retains some resid- in the DNA of the cell. Messenger RNA (mRNA) carries
ual RNA in its cytoplasm, which is stainable by special the code for specific amino acid sequences from the
techniques. It may be slightly larger than the mature red DNA to the cytoplasm for protein synthesis. Transfer
blood cell. RNA (tRNA) carries the amino acid groups to the ribo-
Reticuloendothelial system (RES) The fixed phagocytic some for protein synthesis. Ribosomal RNA (rRNA),
cells of the body, such as the macrophage, having the which exists within the ribosomes, is thought to assist
ability to ingest particulate matter. in protein synthesis.
Retrospective blood utilization review A blood utilization Ribosome A cellular organelle that contains ribosomal
review that occurs after the transfusion events. RNA and protein and functions to synthesize protein.
Retrospective validation Validation testing of software, Ribosomes may be single units or clusters called
which is done after the computer system has been polyribosomes or polysomes.
implemented. Rickettsia Any of the microorganisms belonging to the
Retrovirus A virus in which the genetic material consists genus Rickettsia.
of two identical molecules of RNA that, upon entering Ringer’s lactated injection An aqueous solution suitable
the infected cell, are converted into DNA by the enzyme for intravenous use.
reverse transcriptase to form a “provirus” that may be RNA interference Gene-regulating enzymatic activity of
integrated into the host’s own genomic DNA. RNA molecules first discovered in nematode Caenorhab-
Reverse genetics Technology in which genes can be ditis elegans. Also referred to as gene silencing. Interfering
inactivated (“knocked out”) in the germ line of a mouse RNAs prevent mRNA from being translated into protein.
or other animal model in order to study systemic conse- RNase RNA endonuclease, a ubiquitous enzyme that
quences of lack of function of the protein coded by destroys RNA.
that gene. The resulting model animal is a “knockout
Rouleaux Coinlike stacking of red blood cells in the
transgenic animal.”
presence of plasma expanders or abnormal plasma
Reverse grouping Testing a patient’s serum with commer- proteins.
cial or reagent A and B red blood cells to determine
Saline anti-D A low-protein (6% to 8% albumin) im-
which ABO antibodies are present.
munoglobulin M anti-D reagent.
Reverse transcription An enzymatic process, naturally
Salvia horminum Plant lectin used in the differentiation of
occurring in retroviruses, in which RNA sequence is
various forms of polyagglutination.
transcribed into the DNA sequence. In vitro reverse
transcription may be performed using isolated or Salvia sclarea Plant lectin with anti-Tn activity used in the
synthetic enzymes (reverse transcriptases) to obtain a differentiation of various forms of polyagglutination.
complementary DNA (cDNA). Screening cells Group O reagent red blood cells that are
Rh immunoglobulin (RhIg) A concentrated, purified used in antibody detection or screening tests.
anti-Rh0(D) prepared from human serum (of immu- SD Serologically defined antigens.
nized donors) that is given to Rh0(D)-negative mothers SDS-PAGE An electrophoretic technique using a polyacryl-
after they have given birth to an Rh0(D)-positive baby amide gel (PAGE), in which proteins are denatured by
or after abortion or miscarriage. It acts to prevent the the negatively charged detergent sodium dodecyl sulfate
mother from becoming immunized to any Rh0(D)- (SDS), which masks their intrinsic electrical charge so
positive fetal cells that may have entered her circulation that they are separated according to size, not electrical
and thereby prevents formation of anti-Rh0(D) by the charge.
mother.
Secretor An individual who is capable of secreting soluble,
Rhmod Rare Rh phenotype resulting from mutations in glycoprotein ABH-soluble substances into saliva and
the RHAG gene, it can result in missing or significantly other body fluids.
altered (reduced) RhD and RhCE antigen expression.
Semiconservative Term referring to the DNA replication
Rh-negative Red blood cells lacking the D antigen. model (proved by Meselson and Stahl), in which each
6888_Glossary_629-652 29/10/18 11:32 AM Page 649

Glossary 649

double-stranded DNA molecule upon replication con- all organs and tissues. Shock may be caused by a variety
tains one parental and one newly synthesized comple- of conditions, including hemorrhage, infection, drug
mentary strand. reaction, trauma, poisoning, myocardial infarction, or
Sensitization A condition of being made sensitive to a spe- dehydration. Symptoms include paleness of skin (pal-
cific substance (e.g., an antigen) after the initial expo- lor), a bluish gray discoloration (cyanosis), a weak and
sure to that substance. This results in the development rapid pulse, rapid and shallow breathing, or blood
of immunologic memory that evokes an accentuated pressure that is decreased and perhaps immeasurable.
immune response with subsequent exposure to the Short tandem repeats (STR) A type of genetic polymor-
substance. phism demonstrated as back-to-back repeats of sets of
Sepsis Pathological state, usually febrile, resulting from 1 to 8 nucleotides. The number of times each set is
the presence of microorganisms or their toxins in the repeated is characteristic for each individual and is
bloodstream. useful in kinship and crime investigations and in
monitoring of graft success or rejection. See also
Septicemia Presence of pathogenic bacteria in the blood.
microsatellites.
Sequence-specific oligonucleotide probe (SSOP) hy-
Sialic acid A group of sugars found on the red blood cell
bridization A molecular method used in HLA testing,
membrane attached to a protein backbone; the major
in which the individual’s DNA is immobilized on a
source of the membrane’s net negative charge.
membrane as a dot-blot and incubated with a labeled
complementary DNA probe designed to detect specific Sickle cell anemia Hereditary, chronic hemolytic anemia
HLA sequence. characterized by large numbers of sickle-shaped red
blood cells occurring almost exclusively in black
Sequence-specific PCR (SSP) A polymerase chain reaction
people.
method variant in which one of the primers is supposed
to align exactly where the anticipated mutation is lo- Sickle trait Blood that is heterozygous for the gene coding
cated (sequence-specific primer). for the abnormal hemoglobin of sickle cell anemia.
Sequence-specific primers (1) In reverse transcription: Siderosis Increase of iron in the blood that can lead to
sequences used as cDNA (complementary DNA) syn- organ damage.
thesis primers when only a specific region of cDNA is Single-donor platelets Platelets collected from a single
desired (not cDNA representing total RNA or messenger donor by apheresis.
(mRNA). (2) In PCR: primers designed to anneal to Single nucleotide polymorphisms (SNPs) Single-
the specific mutated region of the DNA that is supposed nucleotide differences between DNA sequences. The
to be amplified. In absence of mutation, the PCR vast majority of these polymorphisms have no biologi-
product will either not be produced at all or will have cal effect and are useful in genetic variation studies
a different length than the product resulting from the relevant to crime or kinship investigations.
mutated DNA.
Sodium dodecyl sulfate (SDS) An anionic detergent
Sequencing A manual or automated process of deciphering that renders a net negative charge to substances it
the order of nucleotides in DNA to find various poly- solubilizes.
morphisms (mutations) relevant in biotechnology or
Software Written instructions for a computer, which result
clinical applications.
in information being stored, manipulated, and retrieved.
Serologic test for syphilis (STS) First developed in 1906
Solid phase test A blood group serology test method that
by Wassermann, present tests are of three main types
uses red blood cell adherence on an endpoint instead of
based on complement fixation, flocculation, and
agglutination.
detection of specific antitreponemal antibodies.
Somatic gene therapy Gene therapy in which the cells into
Serotonin A chemical present in platelets that is a potent
which a new genetic material is introduced are the so-
vasoconstrictor.
matic cells (not the gametes) so that the material is not
Serum The fluid that remains after whole blood has transmitted into the next generation.
clotted.
Southern blotting Technique, developed by E. M. South-
Sex chromosome Chromosomes associated with determi- ern, in which DNA is cut with one or more restriction
nation of sex. endonucleases, the fragments are separated by agarose
Sex linkage A genetic characteristic located on the X or Y gel electrophoresis, transferred onto a membrane (blot),
chromosome. and incubated with a labeled detection probe (a short
Shelf life The amount of time blood or blood products fragment of DNA complementary to the desired region).
may be stored upon collection. Specificity The affinity of an antibody and the antigen
Shock A clinical syndrome in which the peripheral blood against which it is directed.
flow is inadequate to return sufficient blood to the heart Spectrin A dimeric structural protein of the red blood cell
for normal function, particularly transport of oxygen to (RBC) cytoskeleton that maintains the shape of the cell
6888_Glossary_629-652 29/10/18 11:32 AM Page 650

650 Glossary

by forming a network with other proteins such as actin Systemic lupus erythematosus (SLE) A disseminated au-
and ankyrin. Spectrin defects lead to hereditary ellipto- toimmune disease characterized by anemia, thrombocy-
cytosis or spherocytosis. topenia, increased immunoglobulin G levels, and the
Splenomegaly Enlargement of the spleen. presence of four immunoglobulin G antibodies: antinu-
clear antibody, antinucleoprotein antibody, anti-DNA
Splicing The process of excision of introns (noncoding
antibody, and antihistone antibody; believed to be
sequences) from the immature, newly synthesized RNA
caused by suppressor T-cell dysfunction.
(pre-mRNA) in eukaryotes.
System manager A specially trained person who is respon-
Spontaneous agglutination Direct agglutination of
sible for the maintenance of an information system.
antibody-coated RBCs without the presence of anti-
serum; this usually occurs when RBCs are heavily Systolic pressure Maximum blood pressure that occurs at
coated with IgM or IgG autoantibody. Spontaneous ventricular contraction; upper value of a blood pressure
agglutination differs from cold agglutination in that reading.
it is not dispersed when the sample is warmed to 37°C. Tachycardia Abnormally rapid heart action, usually de-
Steatorrhea Increased secretion of the sebaceous glands. fined as a heart rate greater than 100 beats per minute.
Stem cell An unspecialized cell, capable of self-renewal, Tachypnea Abnormally rapid respirations.
that gives rise to a group of differential cells, such as the TaqMan probes Also called cleavage or hydrolysis DNA
hematopoietic cells. probes; used to detect the amplicon produced in real-
Steroid hormones Hormones of the adrenal cortex and the time PCR. These probes, labeled with reporter and
sex hormones. quencher molecules, bind to their complementary area
in a single strand of DNA region between the primers
Stertorous Pertaining to laborious breathing.
and get in the way of the Taq polymerase synthesizing
Sticky (cohesive) ends The ends of DNA sequence result- the new strand. The polymerase, in order to continue
ing from restriction enzymes that cut both strands, the synthesis, degrades the probe and the reporter and
leaving “overhangs” that are complementary to any end quencher molecules become separated, which results in
cut by the same enzyme. These ends will spontaneously emission of light.
join with complementary ends and do not require
Targeted review A type of prospective blood utilization
DNA ligase.
review focusing on one or a few specifically selected
Storage lesion A loss of viability and function associated products or indications.
with certain biochemical changes that are initiated
Template bleeding time The elapsed time a uniform inci-
when blood is stored in vitro.
sion made by a template and blade stops bleeding,
STR Small tandem repeat, or short tandem repeat. The which is a test of platelet function, assuming a normal
name describes the molecular nature of the DNA that platelet count.
harbors this type of genetic marker, consisting of
Tetany A nervous affliction characterized by intermittent
tetra- or pentanucleotide sequences that are tandemly
spasms of the muscles of the extremities.
repeated a variable number of times in unrelated
individuals. Thalassemia major The homozygous form of deficient
beta-chain synthesis, which is very severe and presents
Stroma The red blood cell membrane that is left after
itself during childhood. Prognosis varies; however, the
hemolysis has occurred.
younger the child at disease onset, the less favorable the
Subgroup Antigens within the ABO group that react less outcome.
strongly with their corresponding antisera than do A
Thermal amplitude The range of temperature over which
and B antigens.
an antibody demonstrates serologic and/or in vitro
Survival studies A measure of the in vivo survival of trans- activity.
fused blood cells; usually performed with radioactive
Thermocycler (or thermal cycler) A programmable heat-
isotopes. Normal red blood cells survive approximately
ing and cooling machine used to conduct polymerase
100 to 120 days in circulation.
chain reaction.
SYBR Green Fluorescent stain used in UV visualization of
Thrombin An enzyme that converts fibrinogen to fibrin so
nucleic acids in electrophoretic gels or in real-time PCR
that a soluble clot can be formed.
to detect the amplified product of the reaction.
Thrombocytopenia A reduction in platelet count below the
Syncytiotrophoblast The outermost fetal component of the
normal level, which is associated with spontaneous
placenta that allows for exchange of nutrients between
hemorrhage.
the fetus and the mother.
Thrombotic thrombocytopenic purpura (TTP) A coagula-
Syngeneic Possessing identical genotypes, as monozygotic
tion disorder characterized by (1) increased bleeding
twins.
owing to a decreased number of platelets, (2) hemolytic
Synteny Genes that are closely situated on a chromosome anemia, (3) renal failure, and (4) changing neurological
but cannot be shown to be linked.
6888_Glossary_629-652 29/10/18 11:32 AM Page 651

Glossary 651

signs. The characteristic morphological lesion is throm- ribosomes, during which a codon within messenger
botic occlusion of small arteries or capillaries in various RNA (mRNA) is aligned with an anticodon from the
organs. transfer RNA (tRNA), which attaches a corresponding
Thymidine An essential ingredient used in DNA synthesis amino acid into the growing peptide chain.
and incorporated by T lymphocytes undergoing blast Translocation Transfer of a portion of one chromosome to
transformation in response to foreign HLA-D antigens its allele.
in the mixed lymphocyte culture test. Transmembrane protein A protein that crosses the red
Titer A measure of the strength of an antibody by testing blood cell membrane and is present on both sides of
its reactivity at increasing dilutions against the appro- the membrane.
priate antigen. The reciprocal of the highest dilution Transposition The location of two genes on opposite chro-
that shows agglutination is the titer. mosomes of a homologous pair.
Titer score A method used to evaluate more precisely than Trypsin A proteolytic enzyme formed in the intestine.
simple dilution by comparing the titers of an antibody.
Type and screen Testing a patient’s blood for ABO, Rh,
Agglutination at each higher dilution is graded on a
and unexpected antibodies (antibody screen). If no
continuous scale; the total is the titer score.
abnormalities exist in the ABO and Rh and no unex-
Trait A characteristic that is inherited. pected antibodies are detected in the antibody screen,
Trans The location of two or more genes on opposite chro- then the recipient blood sample is retained in the event
mosomes of a homologous pair. that subsequent serologic crossmatching is necessary.
Transcription The synthesis of RNA based on the sequence Ulex europaeus See Anti-H lectin.
of DNA by the enzyme RNA polymerase. Process based Ultracentrifugation Rapid and prolonged centrifugation,
on the principle of complementarity. The first stage of used to separate by density gradient those substances of
deciphering of the genetic code. various specific gravities.
Transcription factor A protein that binds to DNA at a Urticaria A vascular reaction of the skin similar to hives.
specific nucleotide sequence (a promoter or
Vaccine A suspension of infectious organisms or compo-
enhancer sequence) to influence the process of
nents of them that is given as a form of passive immu-
DNA transcription.
nization to establish resistance to the infectious disease
Transcription mediated amplification (TMA) Developed caused by that organism.
initially at Gen-Probe, TMA is a molecular method of
Validation A systematic process of testing the hardware,
detecting an organism by isothermal amplification of its
software, and user components of an information sys-
RNA using enzyme reverse transcriptase to produce
tem to ensure that they are functioning correctly for
complementary DNA (cDNA), which is subsequently
their intended purpose.
transcribed into multiple RNA molecules, which then
undergo another reverse transcription. This cycle re- Value stream In LEAN, identification of all the steps neces-
peats several times, resulting in abundance of RNA to sary to bring a finished product to the customer.
the levels that may be detected by chemiluminescent Valvular Relating to or having a valve.
DNA probes. Variable number of tandem repeats (VNTR) DNA
Transferase An enzyme that catalyzes the transfer of atoms regions composed of back-to-back repeated units
or groups of atoms from one chemical compound to ranging in size from 9 to 80 bp. Like other polymor-
another. phisms, the VNTRs are useful in genetic fingerprinting
Transfer RNA (t-RNA) See Ribonucleic acid. performed in kinship and crime investigations. See also
minisatellites.
Transfuse To perform a transfusion.
Variable region That portion of the immunoglobulin light
Transfusion The injection of blood, a blood component,
and heavy chains where amino acid sequences vary
saline, or other fluids into the bloodstream.
tremendously, thereby permitting the different im-
Transfusion guidelines Systematically developed evidence- munoglobulin molecules to recognize different anti-
based statements relevant to transfusion therapy, such genic determinants. In other words, the variable region
as thresholds and indications, intended to assist determines the antigen against which the antibody will
clinicians in their decisions related to transfusion. react, thus providing each antibody molecule with its
Transfusion reaction An adverse response to a transfusion. unique specificity. The variable region is located at the
Transfusion safety officer A specialized member of the pa- amino terminal region of the molecule.
tient blood management team involved with coordina- Vasculitis Inflammation of a blood or lymph vessel.
tion, audits of transfusion records, quality assurance, Vasoconstriction Constriction of blood vessels.
and educational functions.
Vasodilation Dilation of blood vessels, especially small
Translation The production of protein, according to the arteries and arterioles.
genetic code. An enzymatic process occurring in the
6888_Glossary_629-652 29/10/18 11:32 AM Page 652

652 Glossary

Vasovagal syncope Syncope resulting from hypotension classification used to describe mutations in the RHD
caused by emotional stress, pain, acute blood loss, fear, gene that cause changes in amino acids present in the
or rapid rising from a recumbent position. transmembrane or intracellular region of the RhD
Vector In recombinant DNA technology, a molecule of protein, thus causing quantitative differences in the
known nucleotide sequence that is used to carry a RhD protein.
foreign DNA fragment into a host organism in order to Western blotting See Immunoblotting.
produce multiple copies using the host’s replication. Wharton’s jelly A gelatinous intercellular substance con-
Most common vectors are extrachromosomal, circular sisting of primitive connective tissue of the umbilical
DNA plasmids, or bacterial viruses (bacteriophages). cord.
Venesection See Phlebotomy. X chromosome The chromosome that determines
Venipuncture Puncture of a vein for any purpose. female sex characteristics. Biologically, a female has
Veno-occlusive disease Disease involving the veins of the two X chromosomes, and a male has an X and a
liver associated with GVHD. Y chromosome.
Venule A tiny vein continuous with a capillary. Xeno- Prefix indicating differing species. For example, a
xenoantibody is an antibody produced in one species
Viability Ability of a cell to live or to survive for a reason-
against an antigen present in another species. Synonym
ably normal life span.
is hetero-.
Vicia graminea See Anti-N lectin.
Xenogeneic Transplantation between species.
Virion A complete virus particle; a unit of genetic material
Yaws An infectious nonvenereal disease caused by the
surrounded by a protective coat that serves as a vehicle
spirochete Treponema pertenue and found mainly in
for its transmission from one cell to another.
humid equatorial regions.
VNTR Variable number of tandem repeats.
Zeta potential The difference in charge density between
von Willebrand’s disease A congenital bleeding disorder. the inner and outer layers of the ionic cloud that
von Willebrand’s factor Coagulation factor VIII. surrounds red blood cells in an electrolyte solution.
WAIHA Warm autoimmune hemolytic anemia. A he- Zygosity testing The process through which DNA se-
molytic anemia caused by the patient’s autoantibody quences are compared to assess whether individuals
that reacts at 37°C. born from a multiple gestation (twins, triplets, etc.) are
Waste Also called Muda, as used within LEAN, an action monozygotic (identical) or dizygotic (fraternal); often
that does not create value as defined by the customer. used to identify a suitable donor for organ transplanta-
tion or to estimate disease susceptibility risk if one
Weak D A general term used to describe individuals with
sibling is affected.
weakened expression of RhD. It represents a specific
6888_Index_653-670 29/10/18 11:32 AM Page 653

Index
Page numbers followed by f indicate figures; t, tables, b, boxes.

A caused by low-prevalence antibodies Administrative actions, 582, 583t


α-3-N-acetylgalactosaminyltranferase, 124 in the reagent antisera, 140t Adsol, 9, 9t
AABB (American Association of Blood caused by rouleaux formation, Adsorption
Banks), 3, 257, 282–283, 418, 479 140–141, 140t allogenic, 245
standards, 375, 485–486t between forward and reverse grouping, in antibody identification, 243, 245–246
Standards for Blood Banks and 143, 143t–144t autoadsorption, 245, 246f
Transfusion Services, 402, 533–534 group I, 136–138, 138t commercial reagents for, 245
tissue banking standards, 488, 489t group II, 139–140, 139t, 140t differential, 245–246
A antigen group III, 140–141, 140t, 141f immunoadsorption, 410–411
formation of, 123, 125f group IV, 141–142, 141f, 142t phenotypically matched, 245
soluble, formation of, 126–127 groups IV, 142–143 RBC, 245–246
AATB (American Association of Tissue resolution of, 136 Adverse effects
Banks), 477, 485–486t, 488, simplified summary of, 145f of apheresis, 411–412, 412b
490, 490t technical errors, 136, 136b metabolic effects, 387–388
ABC. See American Blood Commission with weak or missing antibodies, 138t of tissue transplantation, 491–494
(ABC) ABO gene, 125 Adverse reactions. See Transfusion reactions
ABH antigens, 186 ABO grouping Advisory actions, 582, 583t
in disease, 135–136 of cord blood, 434 African ethnicity, 156, 156–157t
formation of, 123, 124f, 125f in pretransfusion testing, 259 Agarose, 84, 85f
on red blood cells, comparison of, with selection order for transfusion, 260t Agglutination, 65
A, B, and C soluble substances, ABO incompatibility in ABO testing, 121–122
126–127, 127t HDFN caused by, 428–429, 428t, mixed-field, 455
soluble, formation of, 126 430, 431 Agglutination reactions, 121
ABH-soluble substances, 126–127, 127t in hematopoietic progenitor cell antigen-antibody ratio and, 66–67
ABO antibodies, 121–122 transplantations, 421–423, antihuman globulin reagents and, 70
ABO blood group system, 57, 78, 422t, 423t centrifugation and, 66
119–148 ABO isoagglutinin, 141–142, 142t enhancement media and, 67–68
Bombay phenotypes, 124b, 134–135 ABO truth table, 597f, 597t factors influencing, 66–70
B subgroups in, weak, 132–134, 133t ABO typing immunoglobulin type and, 67f
case study on, 145 cold-reactive autoantibodies and, low ionic strength solutions and, 68, 70
discovery of, 2 444–445 pH effect and, 67
ethnic frequency of blood groups in, warm autoantibodies and, 454 polybrene and, 70
121, 122t Absorption, selective, 410–411 polyethylene glycol and, 70
forward grouping, 120, 121t, 122t ABTI antigen, 225 protein media and, 68
genes for, interaction of Hh genes with, Achromatin, 29 proteolytic enzymes and, 70
124–125, 126f Acid-citrate-dextrose (ACD) preservative temperature and, 67
genotypes and phenotypes of, 123b solution, 3, 8t Agglutinogen theory, 151f
grouping/typing in Acquired B phenomenon, 135 AGT. See Antiglobulin test (AGT)
forward, ABO discrepancies between Acquired immunity, 47t, 48, 50 AHG. See Antihuman globulin (AHG)
reverse grouping and, 143, Acrodose PL System, 17 AHG reagents, 235
143–144t Activated factor VII (factor VIIa), 348 Alanine aminotransferase (ALT), 308
reverse, ABO discrepancies between Acute hemolytic transfusion reaction Albumin, 112–113, 363, 385
forward grouping and, 143, (AHTR), 378–379, 378t Aliquots, 345–346, 346b
143–144t Acute lung injury, transfusion-related, Alleles, 26, 125, 175, 500
historical perspective on, 120–121 16b, 368, 380–381, 380b, 381b, Allele-specific probes, 93
inheritance of, 122–127 382f, 504, 508 Allergic transfusion reactions (ATRs),
introduction to, 119–120 Acute normovolemic hemodilution 384–385, 384t, 385b, 386f
molecular genetics of, 125 (ANH), 293–295 Alloantibodies, 46, 63, 142, 142t, 175, 501
phenotype frequencies, 122t Adaptive immunity, 47t clinically significant, 233
reverse grouping, 121t, 122t ADCC. See Antibody-dependent cell- detection and identification
routine testing, 120–121, 121t mediated cytotoxicity (ADCC) of, 457–458, 460t
subgroups, 127–134 Additive solutions (AS) immune, 233
A subgroups of, 127–132, 127t, 128f, in platelet preservation, 15, 16b naturally occurring, 233
129t, 131b in red blood cell preservation, 9, 9b, optimal phase of reactivity for
weak, 131–132, 131t, 133f 9t, 10t common, 240t
ABO discrepancies, 136–145 improved, 11 providing compatible blood products
algorithm for resolving, 137b Adenine, 32, 33f, 80 in presence of, 248
categories of, 136–143 Adenosine deaminase, 88 Alloantigens, 54
caused by acquired B antigen, 139t Adenosine triphosphate (ATP), 4, 6 Alloanti-P, 184

653
6888_Index_653-670 29/10/18 11:32 AM Page 654

654 Index

Allogeneic HPC transplantation, 418–419 case studies, 248–252 Antigen-antibody reactions, 50


Allogenic adsorption, 245 direct antiglobulin test and elution characteristics of, 63–65
Allograft failure, 492 techniques, 246–247 detection of RBC, 65
Alloimmunization, to RBC antigens, 377 introduction to, 233 factors affecting, 46b
Alloimmunized patients, extensive blood serologic systems used in, 68t types of, 64f
group typing of donors for, 97 tube method of, 233–235 Antigenic determinants, 50
Allosteric changes, 7 warm autoantibodies and, 455 Antigen-presenting cells (APCs), 52
Allotypic variation, 58 in disease, 135–136 Antigens, 46, 50
Alphanumeric terminology, 151–152 to high-prevalence antigen, 249, 249f ABH, 123, 124f, 125f, 126–127,
ALT. See Alanine aminotransferase (ALT) against HLA, 501–502 135–136
Alternative complement pathway, immune, 62 antithetical, 50
59–60, 60f incomplete, 104 blood group, 62b
Alternative procedures, 584 to low-prevalence antigen, 250, 250f characteristics of, 61, 61b
American Association of Blood Banks monoclonal, 61–62 compound, 165
(AABB), 3, 257, 282–283, 418, 479 multiple, 248–249 high-prevalence, 175
standards, 375, 402, 533–545 naturally occurring, 62 pseudoantigens, 135
tissue banking standards, 485–486t, passively acquired, 233 specificity of, 50, 51
488, 489t polyclonal, 61–62 Antiglobulin crossmatch, 261–262
American Association of Tissue Banks primary, 71 Antiglobulin test (AGT), 103–118, 141
(AATB), 477, 488, 490, 490t primary and secondary case studies, 115–116
tissue banking standards, 485–486t responses, 52, 53f direct, 104, 109–112, 110f, 110t, 161,
American Blood Commission (ABC), 351 as probes for proteins, 93 163–164
American Rare Donor Program properties, 63–64 factors affecting, 112–114
(ARDP), 163 required in AHG, 107–108 history of, 104
American Red Cross, blood banks, 3 summary of characteristics, 205t indirect, 104, 112, 112t
American Society for Blood and Marrow unexpected, 62–63, 233 introduction to, 104
Transplantation (ASBMT), 418 Antibody-dependent cell-mediated modified and automated techniques,
Amino acids, 33 cytotoxicity (ADCC), 60 114–115
sequences of, 80 Antibody identification principles of, 109
Aminoacyl tRNA synthetases, 80–81 See also Antibody screen reaction mediums, 112–113
Amino terminal, 56 adsorption in, 243, 245–246, 246f sources of error in, 114, 114b
Amniocentesis, 97, 433 alterations in pH and, 246 Anti-H, 448–449
Amorphic alleles, 175 enzymes in, 241–242, 243t Anti-H antisera, 130f
Amorphs, 30, 122 evaluation of panel results in, Anti-HBc, 308
Amplicon, 88 238–241 Antihemophilic factor, cryoprecipitated,
Amplification techniques, 92–93 exclusion in, 238 342, 343b, 358t
AMR. See Antibody-mediated rejection neutralization in, 242–243, 243t, 244f Anti-H lectin, 130f
(AMR) patient history in, 237–238 Antihuman globulin (AHG), 104
Anaerobic glycolytic pathways, 6 reagents in, 238 addition of, in antiglobulin test, 113
Anaphase, 30, 31t selected cell panels in, 241, 242f antibodies required in, 107–108
Anaphylaxis, 72 techniques for resolving, 241–246 comparison of methodologies, 115, 115t
Anastomosis, 138 Antibody identification panel, 238, monoclonal, 106–107
Anemia 238f, 239f monospecific, 107, 108–109
autoimmune hemolytic, 97, 111t, Antibody-mediated rejection (AMR), polyclonal, 106f
441–475 501, 505 polyspecific, 105–106, 108–109
correction of, 565 Antibody screen preparation of, 105–107, 106f, 107f
HDFN and, 430, 430t See also Antibody identification Antihuman globulin (AHG) reagents, 70,
iron deficiency, 565 interpretation of results, 236 104–105
sickle cell, 80, 97 limitations of, 236–237 FDA licensed, 105t
Annealing, 80 other methods for, 236 monospecific, 105, 105t
Antenatal, 432 in pretransfusion testing, 259–260 polyspecific, 104–105, 105t
Antibiotics, used in bacterial cloning, 85b results with possible causes, 237f Antihuman globulin (AHG) test, 110f
Antibodies, 46, 50, 51, 52 tube method of, 233–235, 234f Antihuman serum, 104
See also Immunoglobulins Antibody titration, 247–248, 432 Anti-I, 185–186, 448
ABO, 121–122 titer and score of anti-D, 247t Anti-i, 186, 448
alloantibodies, 63 Anticoagulant preservative solutions Anti-IgG, 105, 107–108
anticomplement, 108 chemicals in, 9t Anti-IH, 448–449
antigen-antigen reactions, factors in red blood cell preservation, 8, Anti-Jk3, 201
affecting, 46b 8t, 9t Anti-Jka and Jkb, 200
anti-IgG, 107–108 Anticoagulants, 2 Anti-K, 194
autoantibodies, 63 Anticoagulation, 398 Anti-Ku, 195
blood group, 61–63 Anticodons, 81 Anti-Lu3, 204
to blood group antigens, 260t Anticomplement antibodies, 108 Anti-Lua, 202–203
to compound antigens, 184, 187 Anticomplement reagents, 105 Anti-Lub, 203
detection and identification, 3, Anti-Cw, 164–165 Anti-M, 190
232–255. see also Antibody Anti-D, 165 Anti-N, 190
identification; Antibody screen Anti-Fya, 198 Anti-P1, 183
benign cold autoagglutinins and, Anti-Fyb, 198 Antiparallel, 32
446–447 Antigen-antibody ratio, 66–67 Antipenicillin antibody, 463–464
6888_Index_653-670 29/10/18 11:32 AM Page 655

Index 655

Anti-PP1Pk, 184 Autoimmune hemolytic anemia(s) Baxter/Fenwal Amicus, 400f


Anti-S, 190–191 (AIHAs), 441–475, 453f Beneficence, 613
Anti-s, 190–191 antibody characteristics in, 468t Beta globin chain, 80
Antithetical antigens, 50 autoantibodies in, 442–462 Bidirectional ABO incompatibility,
Antithrombin III concentrates, 350–351, blood group typing of patients with, 97 138–139
363 case studies of, 468–471 Bilirubin, 430–431
AnWj antigen, 224 comparison of warm and cold, 463t Biochemical barriers, 48
Apheresis, 3, 15, 396–416 DAT-negative, 462 Biological product manufacturing
adverse effects of, 411–412, 412b drug-induced, 462–468 additional requirements of, 585–586
case study of, 412–413 idiopathic cold, 449–450, 450b, 450f contract, 584–585
centrifugation methods in, 399–401, introduction to, 442 current good manufacturing practice
399f, 400f mixed-type autoantibodies, 462 regulations, 583–584
component collection in, 402–405 paroxysmal cold hemoglobinuria, enforcement actions and, 582–583, 583t
donation, 296–297 451–452, 453t FDA inspection process for, 581–582
donor, 397 positive DAT and, 111t regulation of, 575–581
erythrocytapheresis, 397, 409 secondary cold, 451 short supply arrangements, 585
granulocytes, 357t warm, 453–462 Biologics (biological products)
history and development of, 397 Autologous blood/red blood cell recovery definition of, 577, 577t
HPC, 419 techniques, 565–566 deviation reporting, 585, 585t
instruments used for, 403t Autologous donation, 292–295 drugs compared with, 577t
introduction to, 396–397 acute normovolemic hemodilution manufacturing. See Biological product
leukapheresis, 397, 409 and, 293–295 manufacturing
membrane filtration in, 401–402 intraoperative collection, 295 recalls of, 585–586
methodology, 399–402 permission form, 294f registration and licensure of, 579–580
photophesis, 411 postoperative blood salvage, 295 regulatory process for, 576–578
physiology of, 398 preoperative collection, 293, 565–566 Biologics Cadre, 581
plasma, 341 Autologous HPC transplantation, 418–419 Biologics Control Act, 575
plasmapheresis, 397, 408–409, 408b, Autologous tissue, 482–485 Biology, systems, 94–95
409–410, 410–411 implantation of, 484–485 BioMerieux BacT/ALERT, 17, 18t
plateletpheresis, 397, 409 label requirements for, 484b Bio-Rad Erytype S Blood Group
procedure types and application, 397t preparation of, 483–484 System, 275f
special procedures in, 410–411 sterilization and testing of, 484 Bio-Rad Solidscreen II, 276f
technology, 397 storage of, 484 Biotechnology, 78
therapeutic, 397, 405–410, 406–407t Autologous transfusion, 10, 365, 366f Bite cells, 4, 5f
Apparent opposite homozygosity, 522 Automated fluorescent cycle sequencing Blood
Application software, 593, 594–599 method, 90, 90f administration of, 368–369, 368f
Appraisal costs, 546 Autonomy, 613 demand for, 3
ARDP. See American Rare Donor Program Autosomal inheritance, 28–29, 28f, 29t donation. See Blood donation
(ARDP) Autosomal-recessive pattern, 80 establishments handling, registration
Arteriocyte, 11 Autosomal recessive traits, 123 of, 579, 579t
Artificial oxygen carriers, 12–13, 12b, 13t Avidity, 63–64 transfusion.Ssee Transfusion(s)
ASBMT. See American Society for Blood umbilical cord, 141, 419, 434
and Marrow Transplantation B whole. See Whole blood
(ASBMT) β-2-mercaptoethanol (2-ME), 57, 70 Blood banking
Aseptic technique, 298 Babesia microti, 322–323 blood group systems and, 204
Asian ethnicity, 156 Babesiosis, 290–291, 322–323 clinical effects of prolonged, 388t
Assessments, 542–543 tests for, 327t lectins used in, 129b
A subgroups BacT/ALERT, 17, 18t as medical profession, 610
in ABO blood group system, 127–132, Bacterial artificial chromosomes RBC storage legion, 388t
127t, 128f, 129t, 131b (BACs), 86 serologic testing for, diseases important
weak, 131–132, 131t, 133f Bacterial cloning, antibiotics used in, 85b in, 71–73, 72b
ATRs. See Allergic transfusion reactions Bacterial contamination Blood banks
(ATRs) clinical manifestations and pathology, current status of, 3
Audits, 542–543, 544f 320–321 first, 3
Augustine blood group system, 221 epidemiology and transmission, 321 laboratory information systems in,
Autoadsorption, 245, 246f infection from, 320–322 590–606
Autoagglutinins, 442 laboratory diagnosis, 321 quality management in, 531–551
See also Autoantibodies overview of, 320 virtual, 602, 603f
Autoantibodies, 46, 63, 201, 233, 462 platelet storage and, 16–17 Blood bank testing technologies, 268–280,
characterization of, 442–443 prophylaxis and treatment, 321 277–278t
cold-reactive, 250, 251f, 443–452, Bacterial infections, transfusion- Bio-Rad solid-phase protein A
447t, 448t transmitted, 389–390 technology, 274–276, 275f,
definition of, 442 Bacteriophages, 80 277–278t
evaluation of, 455–457 B antigen column agglutination technology,
formation of, 466–467 formation of, 123 268–271, 277f
warm, 251–252, 453–462. See also soluble, formation of, 126–127 comparison of, 276–279, 277–278t
Warm autoantibodies Barcoded information, 592, 592f equipment in, 276
Auto control, 137–138 Basophils, 53 introduction to, 268–269
Autoimmune diseases, 73 Battery, 609 quality control and, 278
6888_Index_653-670 29/10/18 11:32 AM Page 656

656 Index

sensitivity and specificity of, 278–279 Diego, 213–214 factor VIIa concentrates, 362b
solid-phase technology, 272–274 discovery of, 3 factor VIII concentrates, 362–363,
test reactions and procedures, 276 Dombrock, 215 362b
Blood collection Duffy, 197–201 frozen, deglycerolized RBCs, 359
arm preparation methods, 298b Erytype S10, 275 granulocyte pheresis, 361
aseptic technique for, 298 FORS, 220 immune globulin, 363–364
donor identification in, 297–298 genetic basis of, 95 leukocyte-reduced RBCs, 359, 359t
donor reactions to, 299 Gerbich, 217, 217t manufacturing and quality control,
procedure for, 298, 298b Gill, 219 335–348
Blood components, 357–358t Globoside, 182–185 aliquots, 345–346, 346b
See also Blood products; specific I, 185–187, 185t, 186f, 187t cryo-poor plasma, 342
components Indian, 218 cyroprecipitated antihemophilic
cellular, 325 introduction to, 175 factor, 342, 343b
characteristics of, 343–344t ISBT, 176–178, 177–178t, 179t, frozen, deglycerolized RBCs, 347–348
CMV-negative, 364 221–223, 221t granulocyte concentrates, 342
collection, by apheresis, 402–405 ISBT 700 series, 223–224, 223t irradiation, 342, 344
irradiated, 342, 344, 364–365 ISBT 901 series, 224–225, 224t leukoreduction, 339–341
issuance of, 598 John Milton Hagen, 219 plasma, 341–342
labeling of, 351–352, 351f JR, 220 platelets, 337–339
leukocyte-reduced cellular, 364 Kell, 176t, 193–197, 193–194t, 195t, pooling, 346
management of, 595–596, 596f 196f, 197t red blood cells, 336–337
plasma derivatives, 348–351 Kidd, 29, 176t, 199–201, 200f, 200t washing, 344–345
pooling, 346 Knops, 218 whole blood, 335–336, 337b
preparation of, 333–354 LAN, 220 plasma, 361–362
centrifuges in, 334, 334b Landsteiner Wiener, 166–167 platelets, 359–361, 360b
documentation of manufacturing Landsteiner-Wiener, 216, 216t providing compatible, when
steps, 334 Lewis, 176t, 178–182, 179t alloantibody is present, 248
introduction to, 334 Lutheran, 201–204, 202t, 203f, red blood cells, 356, 359
laboratory equipment for, 334 203t, 204t rejuvenated RBCs, 359
plasma expressors in, 334, 335f MNS, 176t, 187–193 thawed plasma, cryoprecipitate
plasma freezers in, 335 Ok, 218 reduced, 361–362
scales in, 334–335 P, 182–185, 182t, 183f in transfusion therapy, 356–364
sterile connection devices in, 335 P1Pk, 182–185 whole blood, 356
storage devices in, 335 parentage testing and, 518 Blood shield statutes, 608
tubing sealers in, 335 polymorphisms Blood utilization management (BUM),
regulations pertaining to, 577, 578b determining population frequency 552–573
reservation, 598 of, 97 See also Patient blood management
washing, 344–345 molecular basis of, 95 (PBM)
Blood donation Raph, 218–219 Blood utilization review, 560–563, 599
current status of, 3–4 Rh, 149–172 concurrent review, 562
donor screening tests, 4t Rh-associated glycoprotein, 219–220 feedback, 563
process, 3–4, 3b Rodgers, 36 intervention strategies, 563–564
Blood donors. See Donor(s) Scianna, 215 prospective review, 562–563
Blood drives, 3 screening for rare phenotypes, 97 retrospective review, 561–562
Blood establishment computer software terminology, 175–178, 176t review criteria, 560–561
(BECS), 575 uncommon, 212–231 Blood volume, extracorporeal, 408
Blood facilities, registration and licensure Vel, 220–221 Blunt end cloning, 85
of, 579–581, 579t with antibodies that exhibit dosage, B lymphocytes (B cells), 50–51, 53–54
Blood group antibodies, characteristics 235b Bombay phenotypes, 134–135, 134b
of, 61–63 Xg, 214–215 Bone marrow, 52t, 53, 419
Blood Group Antigen Gene Mutation Yt, 214 Boundary testing, 602
Data Base (BGMUT), 95 Blood group typing Bromelin, 70
Blood group antigens, 62b of donors for alloimmunized Bromophenol blue dye, 85
Blood group-specific soluble substances patients, 97 B subgroups
(BGSS), 135–136, 140 of patients with autoimmune in ABO blood group system, weak,
Blood group systems, 78, 96t, 173–211 hemolytic anemia and other 132–134, 133t
ABO, 2, 78, 119–148 diseases, 97 Bw4, 500t
antibodies, summary of characteristics, Blood loss, minimizing iatrogenic, 567 Bw6, 500t
205t Blood management. See Patient blood
antithrombin III concentrates, 363 management (PBM) C
applications to routine blood banking, Blood order sets, 559–560, 559f Calculated PRA (cPRA), 506
204, 225 Blood pharming, 11–12 Calmodulin, 5
Augustine, 221 Blood preservation research, research, 2 Cancer
case studies, 206–207, 226–227 Blood products treatments, 35
CD59, 221 See also Blood components; specific viral causes of, 81
Chido, 36 blood products C antigen, 163
Chido/Rodgers, 216–217 albumin, 363 Capillary electrophoresis, 90, 520
Colton, 215 cryoprecipitate, 362 Carboxyl terminal, 56
Cromer, 217–218 factor IX concentrates, 362b, 363 Carriers, 80
6888_Index_653-670 29/10/18 11:32 AM Page 657

Index 657

Cascade, 59 Chorionic villus sampling, 97 Colloids, 68


Case law, 608 Chromatin, 29, 32 Colloid solutions, 350b
CAT. See Column agglutination Chromosomes Colony-stimulating factor (CSF), 54
technology (CAT) bacterial artificial, 86 Colton blood group system, 215
Caucasian MPG, 156t–157t cell division and, 30 Column agglutination technology (CAT),
CCI. See Corrected count increment (CCI) definition of, 29 268–272, 269f, 270f, 271f
CD3 complex, 54 number of, 29 advantages and disadvantages, 271
CD34-positive cells, 52 structure of, 32 applications of, 269
CD59 blood group system, 221 Chronic lymphocytic leukemia (CLL), 135 automation of, 271–272
CDC. See Complement-dependent Circulatory overload, 3 history of, 269
cytotoxicity (CDC) transfusion-associated, 381–383, introduction to, 269
CDRs. See Complementary determining 382t, 383f principle of, 269–271
regions (CDRs) Cis-AB inheritance, 142–143, 143f procedural steps of, 277f
Cefotetan-induced hemolysis, 464–465 Citrate-phosphate-dextrose-adenine Commonly used abbreviations, 575b
Cell control, 137–138 preservative solution, 3, 8t Compatibility tests, 259–260
Cell division, 30, 31–32, 32t Citrate-phosphate-double-dextrose See also Pretransfusion testing
Cells preservative solution, 8t cold-reactive autoantibodies and, 447
bite, 4, 5f Citrate toxicity, 387, 387b, 411–412 Competent bacteria, 86
host, 86 Classical complement pathway, 59, 60f Complementarity, 80
immune system, 50–52 Classic genetics, 25 Complementary determining regions
lineages and markers, 52–54 Classic systems, 516 (CDRs), 58
ratio of serum to, 112 Clinical Laboratory Improvement Complementary DNA (cDNA), 81
Cellular components, 325 Amendments of 1988 (CLIA’88), Complement components, 46
Cellular genetics, 29–32 256, 608 Complement-dependent cytotoxicity
cell division cycle, 31–32 Clinically significant alloantibodies, 233 (CDC), 504–505
meiosis, 30–31 Clinically significant antibodies, 57 Complements
mitosis, 30, 32 Cloned genes, expression of, 88 activation of, 108
terminology, 29–30 Cloning antibodies capable of binding, 108t
Cellular immunity, 50 bacterial, 85b Complement system, 48, 59–61, 60f
Cellular loss, in apheresis, 398 DNA, 83–87 alternative complement
Cellular mechanisms, 46 molecular, 81, 83 pathway, 59–60
Cellular therapy, 417–426 Clotting, 2 binding of complement by RBC
See also Hematopoietic progenitor cell Clotting factor concentrates, 290 antibodies, 61
(HPC) transplantation Clusters of differentiation (CD), 50 classical complement pathway, 59
viral testing requirements for, 420, 420b CMS. See Centers for Medicare and lectin complement pathway, 60
Center for Biologics Evaluation and Medicaid Services (CMS) membrane attack complex, 60–61
Research (CBER), 578 CMV-negative cellular blood Complex branched chains, 129
Centers for Disease Control and components, 364 Compliance inspections, 532
Prevention (CDC), 575 Coagulation factor deficiencies, 367–368 Compliance programs, 532
Centers for Medicare and Medicaid COBE Spectra, 400f Component manufacturing
Services (CMS), 256–257, 575, 580 Code of Federal Regulations (CFR), 282, laboratory, 334
Central dogma, of molecular biology, 37, 402, 485, 487–488, 575 Components. See Blood components
78, 81 Codominant alleles, 175 Component therapy, 3
Central processing unit (CPU), 591 Codominant inheritance, 25 Compound antigens, 165
Central system unit, 591 Codons, 33, 81 antibodies to, 184, 187
Centrifugation, 66, 114, 336b start, 33 Computer crossmatch, 262–263, 263f
continuous flow, 400–401, 400f stop, 33 Computer downtime, 600
intermittent flow, 399, 399f Cold agglutinin disease, 141f Computerized physician order entry
methods of, 399–401 Cold autoantibodies, 142t, 250, 251f, (CPOE), 560, 611
Centrifuges, 334, 334b 443–452, 447t, 448t Computer systems. See Laboratory
Cephalosporins, 464, 466 ABO typing and, 444–445 information systems
CFC. See Continuous flow centrifugation antibody detection and identification Concurrent blood utilization review, 562
CGMP. See Current good manufacturing and, 446–447 Conformance costs, 546–547
practice (CGMP) benign, 444–449, 444t, 445t Constant region, 56
Chagas disease, 290–291, 323 characteristics of, 444t Continual improvement, 543
screening donor blood for, 303 compatibility testing and, 447 Continuous flow centrifugation (CFC),
tests for, 326t laboratory tests affected by, 444 400–401, 400f
Chain termination method, 90 other, 449 Continuous improvement, 568–569
Chargaff’s rules, 79 pathological, 444t, 445t, 449–452 Continuous noninvasive hemoglobin
Charitable immunity, doctrine related to infection, 451 monitoring, 565–566
of, 611–612 RhD typing, 445–446 Contract manufacturing, 584–585
Chase, Martha, 79 specificity of, 447–448 Convulsions, in severe donor
CHD. See Cold hemagglutinin disease typical reactions of, 187t reactions, 299
Chemokines, 54 Cold hemagglutinin disease (CHD), Coombs’ serum, 235
Chido/Rodgers blood group system, 36, 449–452, 450b, 450f, 453t Coombs’ test, 103–118, 141
216–217 Collection 210 (unnamed), 222 Cord blood samples, 141
Chikungunya virus (CHIKV), 319–320 College of American Pathologists (CAP), Cord blood testing, 434
Chimera, 64 16, 283, 479 Cordocentesis, 97, 433
Chimerism, 138, 138t standards, 375 Corrected count increment (CCI), 15
6888_Index_653-670 29/10/18 11:32 AM Page 658

658 Index

Corrective action, 543–544 Denaturation, 80 Disseminated intravascular coagulation


Corticosteroids, for WAIHA, 460 Dendritic cells, 51 (DIC), 368
Cosmids, 86 Dengue virus (DENV), 320 Disulfide bonding, 56
Cost collection 205, 222 Deoxyribonucleic acid (DNA), 25, 29, Dithiothreitol (DTT), 57, 70
Cost of conformance, 546–547 32–37, 78 DMAIC (defined, measure, analyze,
Cost of good quality (COGQ), 546–547 See also DNA entries improve, control), 544–545, 545f
Cost of poor quality (COPQ), 546 amplification techniques, 92–93 DNA. See Deoxyribonucleic acid (DNA)
Crawford antigen, 166 cloning tools, 83–87 DNA polymerase III, 80
Creutzfeldt-Jacob disease (CJD), 289–290, complementary, 81 DNA polymorphisms, 516, 519, 523
290t, 324 as genetic material, 78–81 DNA sequence-based typing (SBT),
Crick, Francis, 32 hybridization, 32 503–504
Cromer blood group system, 217–218 isolation, 36–37 DNA typing, 93–94
Crossmatch tests, 261–262 junk, 36 Doctrine of charitable immunity, 611–612
computer, 262–263, 263f melting point, 80 Doctrine of informed consent, 609
serologic, 261–262 microarrays, 91–92 Documentation, of blood component
virtual, 507 mitochondrial, 522 manufacturing, 334
Crossover, 501 mutations, 35–36, 36t, 37f Document control system, 540, 540f
Cross-reactive groups (CREGs), 502, 502f profiling, 94 Documents, 539–540
Cross reactivity, 502 recombinant, 81–90 Domains, of immunoglobulins, 56
Cruz-Gregurek model, 554, 555f repair, 34–35, 35b Dombrock blood group system, 215
Cryoprecipitate, 362 replication, 34, 80 Donath-Landsteiner test, 451, 452t
Crystalloids, 68, 350b sequencing, 90, 90f, 93–94 Donation facility software, 594–596
Current good manufacturing practice structure of, 32–33, 33f, 79–80 Donor apheresis, 397
(CGMP), 375, 488, 537, 575, viral, 81 Donor management, 594–595
583–584 Deoxyribonucleoside triphosphates Donor release form, 292f
Cut and patch repair, 35 (dNTPs), 80 Donor rights, 609
Cw antigen, 164–165 Department of Health and Human Donor(s)
Cycle sequencing method, 90 Services (DHHS), 575 for alloimmunized patients, extensive
Cryo-poor plasma, 342 D epitopes on RhCE protein, 160–161, blood group typing of, 97
Cryoprecipitate 161f arm preparation methods, 298b
compatibility testing and, 261 DG Gel 8, 269, 271 autologous, 292–295
pooled, 346, 358t DHTR. See Delayed hemolytic and deferral of, 286b, 287f
Cryoprecipitated antihemophilic factor, serologic transfusion reaction HPC, 419–420
342, 343b, 358t (DHTR) identification of, 297–298
Cryoprecipitation, 3 Dideoxyribonucleoside triphosphates informed consent by, 402
Cryoprotective agents, 10 (ddNTPs), 90 minimum age for, 283t
Cytokines, 48, 49t–50t, 54 Diego blood group system, 213–214 postdonation instructions
Cytomegalovirus, 318 Dielectric constant, 68 for, 299, 302f
Cytomegalovirus (CMV), 364, 421 Di(ethylhexyl)-phthalate (DEHP), 8 processing of, 299, 301–303
Cytosine, 32, 33f, 80 Differential adsorption, 245–246 reactions of, to blood collection, 299
Differentiated cells, 54 records of, 299, 303f
D DIIHA. See Drug-induced immune recruitment of, 595
Damages, 609 hemolytic anemia (DIIHA) registration of, 594–595
Dane particle, 310f Dimers, 35 Rh testing requirements for, 162
D antigen, 158, 158t Dimethyl sulfoxide (DMSO), 19 screening, 612
expression of variations of, 159–161, 2,3-Diphosphoglycerate, 6–7 screening, for rare blood group
159f, 160f Diploids, 26 phenotypes, 97
partial or D mosaic, 159f, 160, 160f Direct antiglobulin test (DAT), 63, 104, suitability of, 595
Darwin, Charles, 25 109–112, 110f, 110t, 111t, testing for disease, 307–308
DAT. See Direct antiglobulin test (DAT) 163–164, 268, 442 Donor selection and screening, 281–306
Data archiving, 600 in antibody detection and agencies governing, 282–283
Databases, 593, 593f identification, 246–247 apheresis donation and, 296–297
DAT-negative autoimmune hemolytic benign cold autoagglutinins and, 446 autologous donation and, 292–295
anemia, 462 of cord blood, 434 case studies, 303–304
DCE terminology, 150–151, 151f DAT panel, 109, 110t, 111 directed donation and, 295–296
Decision support, 560 evaluation of positive, 111–112 informed consent in, 292, 292f
Defendant, 609 organic solvent methods, 247 introduction to, 282
Defense Advanced Research Projects pH method, 247 medical history questionnaire
Agency (DARPA), 11 in pregnancy, 161 in, 283–284, 284–285f
Define, measure, analyze, improve, principle and application, 109 medical history questions in, 285–291
control (DMAIC), 544–545, 545f temperature-dependent methods, 247 physical examination in, 291–292
Degeneration, 81 warm autoantibodies and, 455 registration in, 283
Deglycerolization, 10–11, 348b Direct bilirubin, 430 for tissue banking, 487–488
Del, 159 Direct Coombs’ test, 63 whole blood collection and, 297–299
Delayed hemolytic transfusion reaction Directed donation, 295–296, 296f, 612 Donor-specific anti-HLA antibodies
(DHTR), 379–380 Direct exclusion, 522 (DSA), 504
Delayed serologic transfusion reaction Discontinuous prospective review, 563 Donor units
(DSTR), 379–380 Disease, ABH antigens and antibodies in, ABO/Rh testing, 299, 301
Deletions, 166 135–136 antibody screening of, 301
6888_Index_653-670 29/10/18 11:32 AM Page 659

Index 659

HBV screening of, 301 Er collection 208, 222 Fingerprinting, DNA, 94


issuance of, 264 Erythroblastosis, 430 Fisher-Race nomenclature, 150–151,
processing of, 299, 301–303 Erythrocytes. See Red blood cells (RBCs) 151f, 151t, 154t, 155t
screening of, 301–303 Erythrocytopheresis, 397, 409 Flowcharts, 538, 539f
selection of, 260–261 Erythropoiesis, 430 Flow cytometric crossmatch, 505
testing, 260 Erytype S10 blood group system, 275 Flow cytometry, 54, 71, 505–506, 505f
Dosage effect, 66 Ethics, 613, 613b Fluid replacement, 409–410
Dot blotting, 93 Ethidium bromide, 85 Fluid shifts, in apheresis, 398
Double RBC apheresis, 297 Ethylenediaminetetraacetic acid Fluorescence-assisted cell sorting
Drug-adsorption (hapten) mechanism, (EDTA), 65 (FACS), 71
463–465, 464f, 464t Euchromatin, 29 Fluorescent in situ hybridization, 92
Drug-dependent mechanism, 465 Eukaryotic organisms, 29 Fluosol, 13
Drug-induced immune hemolytic anemia E variants, 166 FMEA. See Failure modes and effects
(DIIHA), 442, 462–468 Exchange transfusion, 434–435 analysis (FMEA)
autoantibody formation in, 466–467 Excision repair, 35 FNHTR. See Febrile nonhemolytic
hapten mechanism in, 463–465, Exons, 82f, 83 transfusion reaction (FNHTR)
464f, 464t Expression libraries, 87 Food and Drug Administration (FDA),
immune complex mechanism External assessments, 543 256, 308, 575
in, 465, 465f External inspections, 543 Code of Federal Regulations, 402
mechanisms leading to development Extracorporeal blood volume (EBV), 408 donor selection and, 282
of, 467t Eye Bank Association of America (EBAA), enforcement actions and, 583t
membrane modification in, 466, 485t–486t, 491 enforcement actions of, 582–583
466f, 466t information system regulations, 599
serologic reactions with, 466t F inspection process of, 581–582
treatment of, 467–468 f (ce) antigen, 165 labeling regulations, 351–352
Drugs, biologics compared with, 577t FACS. See Fluorescence-assisted cell qualification of tissue vendors and,
DSTYR. See Delayed serologic transfusion sorting (FACS) 481–482
reaction (DSTR) FACT. See Foundation for Accreditation recalls, 493–494, 494b, 585–586
Duffy antigen group, 54 for Cellular Therapy (FACT) role of, 576
Duffy blood group system, 197–201 Factor IX concentrates, 349, 362b, summary of fatalities issued by, 374
biochemistry of, 198 363, 367 transfusion safety and, 576
common phenotypes, 198t Factor VIIa concentrates, 362b Food and Drug Administration
genetics of, 198–199 Factor VIII concentrates, 348–349, Modernization Act (FDAMA), 576
Dynamic files, 593 362–363, 362b, 367 Food and Drug Administration
Dyspnea, transfusion-associated, 383 Factor XIII concentrates, 349 Safety and Innovation Act
Failure modes and effects analysis (FDASIA), 576
E (FMEA), 547–549 Forms, 540
eBDS, 17, 18t Fainting, 412 FORS blood group system, 220
Ebola virus, 303, 320 Familial hypercholesterolemia, 411 Forward and reverse primers, 88–89, 89f
Education, in utilization management, Fatal hemolytic transfusion reactions, Forward grouping, 120, 121t
567–568 120b, 120t ABO discrepancies between reverse
Effector cells, 51 Fatalities grouping and, 143, 143–144t
Ehrlichiosis, 322 from apheresis, 412 Foundation for Accreditation for Cellular
Electrophoresis reporting, 586 Therapy (FACT), 418
definition of, 84 transfusion-related, 586t Frameshift mutations, 36
gel, 84–85 FDA. See Food and Drug Administration Franklin, Rosalind, 79
Electrostatic forces, 63 (FDA) Freezing
Elution techniques, 246–247, 434 FDA Amendments Act (FDAAA), 576 platelets, 19
Emergency transfusions, 365–366 Febrile nonhemolytic transfusion of red blood cells, 10–11, 11t
Emergent transfusions, 263 reaction (FNHTR), 383–384 Fresenius AS-104, 401f
Emm antigen, 224 Federal Food, Drug, and Cosmetic Fresh-frozen plasma (FFP), 341, 358t,
Emotional distress, intentional infliction (FD&C) Act, 575–576, 578, 579 361–362, 410
of, 610 Federal laws, on tissue banking, 485, FRET probes, 92
En(a-) phenotype, 192 487–488 Frozen, deglycerolized RBCs, 347–348,
Enhancement media, agglutination Federal Register, 578 359
reactions and, 67–68 Federal regulatory requirements, 574–589 Fucosyltransferase, 124
Enhancement reagents, 235 current good manufacturing practice Functional affinity, 63
Enzyme immunoassay (EIA), 65 regulations, 583–584 Fy3 antigen and antibody, 199
Enzyme-linked immunosorbent assay enforcement actions and, 582–583 Fy5 antigen and antibody, 199
(ELISA), 65 history of, 575–576 Fya and Fyb antigens, 198
Enzymes, 78 introduction to, 575 Fyx, 199
in antibody identification, 241–242, registration and licensure, 579–581
243t regulatory process and, 576–578 G
parentage testing and, 518 Fetal DNA typing, 95, 97 Galactose, 127
proteolytic, 70, 241–242, 243t Fetal ultrasound, 433, 433f Gametes, 29, 30–31
recombinant, 88 Fetomaternal hemorrhage, 429 Gamma globulin fraction, 135
Eosinophils, 53 FFP. See Fresh-frozen plasma (FFP) G antigen, 165
Epitopes, 50, 125 Fibrinogen, 3, 362 Gatekeeping, 563–564
Epstein-Barr virus (EBV), 318 Ficin, 70 Gel electrophoresis, 84–85, 85f
6888_Index_653-670 29/10/18 11:32 AM Page 660

660 Index

Gel test, 115, 115t Growth factors, 52 Hematopoietic progenitor cells


CAT, 268–271, 270f Guanine, 32, 33f, 80 (HPCs), 410
Gene array technology, 25 Guanine triphosphate (GTP), 39 processing and storage of, 420–421
Gene chips, 91–92 Guidance documents, 578 sources of, 419
Gene expression, 78 Guideline for the Uniform Labeling of Hematopoietic stem cells (HSCs), 11, 13,
Gene polymorphism, techniques for Blood and Blood Components, 351 478, 508–509
studying, 93–94 GVHD. See Graft-versus-host disease Hematopoietic stem cell transplantation,
General system applications, 598–599 (GVHD) 508–509
Genes, 29, 78 Hemizygous, 30
cloning, 83–87, 86f, 87f H Hemoglobin-based oxygen carriers
coding sequence of, 83 H-active antigenic structures, 130f (HBOCs), 12, 12b, 13t
expression of cloned, 88 Haemonetics Centrifuge Bowl, 399f Hemoglobinemia, 378t, 379
hybrid, 160 Haemonetics eBDS, 17, 18t Hemoglobin-oxygen dissociation curve,
silent, 30 Haemonetics MCS Plus, 399f 6–7, 7f
Gene therapy, 88 H antigen Hemoglobinuria, 379–380, 391, 443, 449
Genetic code, 33, 33f, 78, 80–81 formation of, 123, 124f paroxysmal cold, 184, 451–452, 453t
Genetic information, expression soluble, formation of, 126–127 HemoLink, 13t
of, 80–81 Haploid number, 30 Hemolysis, 65–66, 378t, 430
Genetics Haplotype, 501 Hemolytic anemia, autoimmune. See
basic, 24–44 Hapten mechanism, in drug-induced Autoimmune hemolytic anemia(s)
cellular, 29–32 immune hemolytic anemia, (AIHAs)
classic, 25 463–465, 464f, 464t Hemolytic disease of the fetus and
future role of, 41 Haptens, 61 newborn (HDFN), 57, 64, 73, 164,
Hardy-Weinberg formulas, 24–25, 27b Hardware, 591–592, 591f 233, 427–440
Hardy-Weinberg principle, 27–28 Hardy-Weinberg formulas, 24–25, 27b bilirubin and, 430–431
immune system, 54–55 Hardy-Weinberg principle, 27–28 case studies of, 436–437
inheritance patterns, 28–29, 28f HBOCs. See Hemoglobin-based oxygen caused by ABO, 428–429, 428t,
introduction to, 24–25 carriers (HBOCs) 430, 431
Mendelian, 24–25, 25–27, 26f, 27f, 122 HBV testing, 301 caused by RBC alloimmunization,
molecular, 32–41. See also Molecular HDFN. See Hemolytic disease of the fetus 429–430, 431–433
genetics and newborn (HDFN) diagnosis of, 431–433, 431f
population, 25–29 Health Care Financing Administration ABO, RhD, and antibody screen,
reverse, 94 (HCFA), 580 431–432
Rh blood group system, 154–157, 155f Health Information Technology for antibody identification, 432
techniques in modern, 40–41 Economic and Clinical Health antibody titers, 432
Genome, 29 (HITECH) Act, 608, 611 fetal DNA testing, 433
Genotypes, 29–30, 175 Health Insurance Portability and fetal ultrasound, 433, 433f
Genotyping Accessibility Act (HIPAA), paternal phenotype and genotype,
red blood cell, 94–97 608, 611 432–433
clinical applications of, 95, 97–98 Heart transplantation, 510 postnatal, 431
Gerbich blood group system, 217, 217t Heat-shock proteins, 40 etiology of, 428
Gill blood group system, 219 Helix structure, of DNA, 79–80 hemolysis, anemia, and erythropoiesis
Globoside blood group system, 182–185 Hemagglutination, 65 and, 430, 430t
Gluboside collection, 222 HemAssist, 12, 13t introduction to, 427–428
Glucose, as preservative solution, 2–3 Hematomas, 299 management of
Glycans, 123, 125 Hematopoiesis, 135 amniocentesis in, 433
Glycerol, 10 Hematopoietic growth factors, 410 cord blood testing in, 434
high-concentration vs. low- Hematopoietic progenitor cell (HPC) cordocentesis in, 433
concentration technique, 10t transplantation, 417–426 exchange transfusion in, 434–435
removal of, 10–11 ABO-incompatible, 421–423, 422t, 423t fetal ultrasound in, 433
Glycophorin A (GPA), 189–190, accrediting bodies for, 418 in fetus, 433–434
190f, 191 allogeneic, 418–419 in infant, 434–435
Glycophorin B (GPB), 189–190, autologous, 418–419 intrauterine transfusion in, 433–434
190f, 191 case study on, 424 intravenous immune globulin in, 435
Glycoprotein-soluble substances, 127 clinical uses of, 418–421 phototherapy in, 435
Glycosylation, 40 complications of, 418–419 simple transfusion in, 435
Glycosyltransferases, 123, 124t, 175 donor considerations, 419–420 pathogenesis of, 428–431, 429f
Government agencies, regulating donor introduction to, 418 positive DAT and, 111t
selection, 282–283 overview of, 418 prevention of, 435–436
GPA. See Glycophorin A (GPA) passenger lymphocyte syndrome, 423 RhD, 428t
GPB. See Glycophorin B (GPB) platelet refractoriness, 423 Hemolytic transfusion reaction (HTR), 368
Graft-versus-host disease (GVHD), processing and storage in, 420–421 acute, 378–379, 378t
364–365, 385–387, 386f, 405, Rh incompatible, 423 delayed, 379–380
418–419, 421 special transfusion requirements in, Hemophilia V, 367
Graft-versus-host reactions, 72 421–423 Hemopure, 12, 13t
Granulocyte pheresis, 361 syngeneic, 418 Hemostasis, optimization of, 566
Granulocytes, 52, 53 transfusion-associated graft-versus- HemoTech, 13t
collection, by apheresis, 404–405 host disease and, 421 Hepatitis, 290
concentrates, 342 transfusion-transmitted infectious diagnostic tests for, 311t
Griffith’s transformation, 78–79, 78f diseases and, 421 transfusion-associated, 308–313
6888_Index_653-670 29/10/18 11:32 AM Page 661

Index 661

Hepatitis A, transfusion-associated, genetics, 500–501, 500f Hypogammaglobulinemia, 135


308–309, 309t, 311t introduction to, 497–498 Hypotensive transfusion reactions, 383
Hepatitis B nomenclature, 498–500, 500f Hypovolemia, 412
tests for, 326t parentage testing and, 518–519
transfusion-associated, 308t, 309–310, Hodgkin’s disease, 135 I
309t, 311t, 312f Homozygous inheritance, 234, 235f, 235t I antigen, 185–187, 185t, 186f
Hepatitis C, 301 Hospital tissue banks, 477–481 i antigen, 185–187, 185t, 186f
tests for, 326t See also Tissue banking IAT. See Indirect antiglobulin test (IAT)
transfusion-associated, 308t, 310, acquisition of allografts and inventory Iatrogenic blood loss, 567
311t, 312 by, 479–480 I blood group system, 185–187, 185t,
Hepatitis D, transfusion-associated, common tissues in, 478 186f, 187t
311t, 312 function of, 476, 477 biochemistry and genetics of, 186
Hepatitis E, transfusion-associated, introduction to, 476–477 disease associations, 187
311t, 312 oversight of, 479 Idiopathic cold AIHA, 449–450
Hepatitis G, transfusion-associated, 313 record-keeping and traceability in, Idiotypic regions, 56
Hereditary spherocytosis (HS), 78 480–481 Idiotypic variation, 58
Hershey, Alfred, 79 standard operating procedure (SOP) ID-MTS gel, 269, 271
Hershey-Chase phase experiment, 79, 79f checklist, 480b IgA deficiency, allergic transfusion
Heterochromatin, 29 storage in, 480 reactions and, 385b
Heterogeneous RNA (hRNA), 82f, 83 Hospital transfusion committee, 369–370 IgG. See Immunoglobulin G (IgG)
Heterozygous inheritance, 234, 235f, 235t Host cells, 86 IgM. See Immunoglobulin M (IgM)
Hh genes, interaction of ABO Host factors, 64, 64b Immediate hypersensitivity, 72
and, 124–125, 126f HPA. See Hybridization protection assay Immediate spin crossmatch, 261
High-concentration glycerol technique, (HPA) solid-phase red cell adherence (SPRCA),
10, 10t HPC-apheresis (HPC-A), 419, 419b 273–274, 274f
High-prevalence antigens, 175 HPC-cord blood (HPC-C), 419, 419b Immune alloantibodies, 233
High-protein anti-D reagents, 161–162 HPC-marrow (HPC-M), 419, 419b Immune antibodies, 62
High responders, 54–55 HPCs. See Hematopoietic progenitor cells Immune complex mechanism, 465, 465f
HIPAA. See Health Insurance Portability (HPCs) Immune globulin, 363–364
and Accessibility Act (HIPAA) HSCs. See Hematopoietic stem cells Immune hemolytic anemia, 442
HIPAA Privacy Rule, 611 (HSCs) See also Autoimmune hemolytic
HIPAA Security rule, 611 HTR. See Hemolytic transfusion anemia(s) (AIHAs); Drug-induced
Histocompatibility testing technique, reaction (HTR) immune hemolytic anemia
503–507 Human genome, 93 (DIIHA)
HLA antibody detection techniques, 504 Human Genome Project, 87 Immune response
HLA genotyping, 503–505, 503f Human herpesvirus 6 (HHV-6), 319 antigens, 61
microlymphocytotoxicity, 504–507, Human herpesvirus 8 (HHV-8), 319 host factors, 64, 64b
504f Human immunodeficiency virus (HIV), innate, 46–48
Histones, 29, 32 81, 290 tolerance and, 64–65
HIV. See Human immunodeficiency blood safety and, 12 traditional laboratory testing methods,
virus (HIV) screening donor blood for, 302 65–66
HIV-1 Group 0 risk, 291, 291b tests for, 326t Immune serum globulin, 349
HLA. See Human leukocyte antigens transfusion-associated, 308t Immune suppression, 73
(HLA) types 1 and 2, 313–314, 313f, 313t Immune system
HLA alleles, associated through Human leukocyte antigens (HLA), 51, antigen-antibody reactions, 63–65
disequilibrium, 502t 55, 78, 418 blood group antibodies, 61–63
HLA antibodies, detection antibodies against, 501–502 blood product transfusions and, 73
techniques, 504 function of, 498, 498f cells and organs of, 50–54
HLA antigens, on RBCs, 225 nomenclature, 498–500, 500f cellular and humoral components
HLA genetic region, 500–501 testing, 497–498 of, 47t
HLA genetics, 500–501, 500f Human T-cell lymphotropic virus (HTLV) complement system, 59–61
HLA genotyping, 503–505, 503f screening donor blood for, 302 cytokines, 54
HLA haplotype, 501, 501f transfusion-associated, 308t genetics, 54–55
HLA serologic specificities, 499t, 500t Human T-cell lymphotropic viruses major mechanisms of, 47t
HLA system, 497–514 types I/II (HTLV-I/II), 314–315, maturation, 52
clinical significance of, 507–510 316f, 326t overview of, 46–48
disease associations, 507–508, 507t Humoral immunity, 50 Immunetics BacTX, 18t
heart and lung transplantation, 510 Humoral mechanisms, 46 Immunity
hematopoietic stem cell Hybrid formation, 36 acquired or adaptive, 47t, 48, 50
transplantation, 508–509 Hybrid genes, 160 cellular, 50
kidney transplantation, 509–510 Hybridization, 32 definition of, 46
liver transplantation, 510 Hybridization-based assays, 80 humoral, 50
pancreas and islet cell Hybridization protection assay (HPA), 92 innate or natural, 47t, 48
transplantation, 510 Hydrogen bonding, 56, 63, 79, 79–80 Immunoadsorption, 410–411
paternity testing, 507 Hydrophilic bonds, 63 Immunoblotting, 93
platelet transfusion, 508 Hydrophobic bonds, 63 Immunodeficiency, 71–72
transfusion-related acute lung Hyperkalemia, 387, 387b Immunodeficiency diseases, 135
injury, 508 Hypersensitivity, 72 Immunodominant sugars, 124t
United Network for Organ Hypervariable regions, 58 Immunofluorescence (IF), 65
Sharing, 510 Hypocalcemia, 387, 387b Immunogen, 61, 104
6888_Index_653-670 29/10/18 11:32 AM Page 662

662 Index

Immunoglobulin Fc receptors, 59 Interface software, 593–594 Kell blood group system, 176t, 193–197
Immunoglobulin G (IgG), 52, 104 Interferon (IFN), 54 antibodies, 194–195
agglutination reactions and, 67–68 Interleukin (IL), 54 antigens, 193–195, 193–194t,
chemical reduction of, 70 Intermittent flow centrifugation, 399, 399f 197, 197t
domain structure of, 56, 57f Intermolecular binding forces, 63 autoantibodies, 197
subclasses, 57–58, 58t Internal and external failure costs, 546 biochemistry of, 195
Immunoglobulin M (IgM), 52, 57, 57f, Internal assessments, 542–543 common phenotypes, 194t
104 Internal audits, 542–543, 544f genetics of, 195
agglutination reactions and, 67–68 International Council for Commonality McLeod phenotype and syndrome and,
chemical reduction of, 70 in Blood Banking Automation 195–196
Immunoglobulins, 46, 50 (ICCBBA), 351 Kidd blood group system, 29, 176t,
See also Antibodies International Society for Cellular 199–201, 200f, 200t
characteristics of, 55–59, 55t Therapy (ISCT), 418 Kidney transplantation, 509–510
significant for blood banking, 56–58 International Society of Blood Kleihauer-Betke test, 436
structure of, 55–56, 56f, 57f Transfusion (ISBT), 95, 175 Knockout transgenic animals, 94
type of, agglutination reactions and, 67 700 series, 223–224, 223t Knops blood group system, 218
variations, 58 901 series, 224–225, 224t Ko phenotype, 195
Immunohematology, 58, 78, 82–83t, 92 blood group collections of, 221–223, Kpa antigen, 194
Immunology 221t Kpb antigen, 194
fundamentals of, 45–76 Committee on Terminology for Red Kpc antigen, 194
introduction to, 46 Cell Surface Antigens, 152, 154 Kx antigen, 195
self vs. nonself concept in, 46 labeling regulations, 351 Kx system, 193–197
Immunomodulatory effects, of Interphase, 30, 31t
transfusion, 388–389 Intraoperative collection, 295 L
Immunoregulatory molecules, 54 Intrauterine transfusions, 264, 433–434 Laboratory information systems
Immunosuppressive drugs, for Intravenous immune globulin (IVIG), acronyms for, 591t
WAIHA, 460 385, 435 backups of, 600
Incomplete antibodies, 104 Introns, 82f, 83 in blood banks, 590–606
Incubation time, AGT and, 113 Invalid test cases, 602 blood bank software applications,
Indian blood group system, 218 Invasion of privacy, 610–611 594–599
Indirect antiglobulin test (IAT), 104, Ionic cloud concept, 69f computer downtime and, 600
110f, 112, 112t Iron deficiency anemia, 565 error management in, 601
polyspecific vs. monospecific AHG in, Irradiated cellular blood components, future trends for, 602
108–109 364–365 hardware maintenance and, 600
Indirect bilirubin, 430 Irradiation, 342, 344 introduction to, 590–591
Indirect exclusion, 522 ISBT. See International Society of Blood personnel training for, 601
Individual quality control plan Transfusion (ISBT) regulatory and accreditation
(IQCP), 535f ISBT blood group system, 176–178, requirements for, 599–600
Infection, tissue transplantation and, 492 177–178t, 179t security maintenance and, 600–601
Infectious transfusion reactions, 389–390 Islet cell transplantation, 510 software validation, 601–602
Inflammation, innate immunity and, 48 Isoagglutinins, 62, 135 system components, 591–594
Information management, 540 Isotype, 56 hardware, 591–592, 591f
Information processing, 590 Isotype variation, 58 people, 594
Information storage hardware, 592 Isotypic class, 55 software, 592–594
Information systems. See Laboratory IT antibody, 187 system management, 600–602
information systems IT antigen, 187 types of, 591
Informed consent, 292, 292f, 402, IV immunoglobulins, for WAIHA, 460 virtual blood banks and, 602, 603f
609, 613 Laboratory testing
Ingram, Vernon, 80 J flow cytometry, 54, 71
Inheritance nontraditional, 71
JACIE, 418
of ABO blood groups, 122–127 traditional methods, 65–66
Jka antigen, 199–200
autosomal-recessive, 80 Lambda vectors, 86
Jk(a–b–) phenotype, 201
cis-AB, 142–143, 143f LAN blood group system, 220
Jk allele, 201
codominant, 25 Landsteiner-Wiener blood group system,
Jkb antigen, 199–200
genes and, 29–30 166–167, 216, 216t
John Milton Hagen blood group system,
Mendel’s laws of, 24–25, 25–27, 26f, Large granular lymphocytes, 54
219
27f, 78 Latency period, of immune response, 52
Joint Commission, 476
patterns, 28–29, 28f, 29t Lattice-type structure, 65, 66f
HCT/P-related standards of, 485–486t
Initiation factor IF2, 39 Laws, 577
tissue banking standards, 490–491
In(Jk) allele, 201 See also Medicolegal issues in
JR blood group system, 220
Injunctions, 582, 583t transfusion medicine
Jsa and Jsb antigens, 194
Innate immunity, 46–48, 47t, 48 case, 608
Judicial actions, 582–583, 583t
Innocent bystander mechanism, 465 sources of, 608
Junk DNA, 36
Innocent VII, 2 LEAN approach, 545–546, 550
Justice, in transfusion medicine, 613
Input/output devices, 592 Lectin complement pathway, 60
Inspection process, FDA, 581–582 Lectins, used in blood banking, 129b
Intentional infliction of emotional K Legal issues, in transfusion medicine,
distress, 610 K and k antigens, 193–194 607–616
Intentional tort of battery, 609 Karyotype, 30 Leishmaniasis, 290
INTERCEPT Blood System, 18, 339 Kell antibodies, 194–195 Leukapheresis, 297, 397, 409
6888_Index_653-670 29/10/18 11:32 AM Page 663

Index 663

Leukemia, 135 Major ABO incompatibility, 138 Minisatellites, 94


serologic reactions typical of, 139t Major histocompatibility complex Minor ABO incompatibility, 138
Leukocyte-reduced cellular blood (MHC), 51–52, 55, 500 Mismatch repair, 34–35
components, 364 Malaria, 289, 289f, 323–324 Missense point mutations, 36
Leukocyte-reduced RBCs, 359, 359t, 364 Malignancy, tissue transplantation Mitochondrial DNA (mDNA), 522
Leukocytes, 48 and, 492 Mitomycin C, 35
Leukoreduction, 339–341 MAM antigen, 225 Mitosis, 30, 30f, 31t, 32
Level 2 guidance documents, 578 Management assistance software, 599 Mixed-field, 132
Level I guidance documents, 578 Mandatory standards, 610 Mixed-field agglutination, 135, 455
Lewis antibodies, 180 M antigen, 189 Mixed-type autoantibodies, 462
Lewis antigens, 179–180, 180t, 181f Manufacturers, 576–577 Mk phenotype, 192
development of, 181–182 See also Biological product Mn CHO collection 213, 222–223
Lewis blood group system, 176t, 178–182 manufacturing MNS antigens
antibodies, 180 registration and licensure of, 579–581 M and N, 189
antigens, 179–180, 180t, 181–182, 181f regulatory oversight of, 576–577, 581t S and s, 189–190
genetics and biosynthesis, 180–181, Massive transfusions, 263–264, 366, 366t summary of, 188t–189t
180f Maternal factors, in HDFN, 429 MNS blood group system, 176t, 187–193
phenotypes of, 179t McLeod phenotype and syndrome, antigens, summary of, 188t–189t
Lewis’s substances, 244f 195–196 anti-M, 190
L-fucose, 124 Medical devices, 586 anti-N, 190
Liability Medical history questionnaire, for anti-S and anti-s, 190–191
basis of, 608–611 donors, 283–284, 284–285f autoantibodies in, 193
HIPAA and, 611 Medical history questions, for donors, biochemistry of, 191
strict, 610 285–291 disease associations, 193
License suspension/revocation, 582, 583t Medical laboratories, quality genetics of, 191, 191f, 192f
Licensure, 579–580 management system for, 546, 546f GPA- and GPB-phenotypes, 191–192
Li collection 207, 222 Medical malpractice, 613 other antibodies in, 192–193
Ligands, 7 Medication deferral list, 286, 287f prevalence of common MN and Ss
Ligases, 83 Medicolegal issues in transfusion phenotypes, 188t
LightCycler, 92 medicine, 607–616 Molecular beacons, 92
Linkage disequilibrium, 501 case studies, 614–615 Molecular biology
Linnaeus, Carolus, 25 current, 607–608 advances in, 41, 94–95
LISS. See Low ionic strength solutions emerging issues in, 613 central dogma of, 78, 81
(LISS) ethics and, 613, 613b concepts in, 77–101
LISS-tube testing, 115t HIPAA and, 611 introduction to, 78
Liver transplantation, 510 introduction to, 607 systems biology, 94–95
Lock and key mechanism, 50 liability as, torts as basis of, 608–611 Molecular cloning, 81, 83
Locus, 29, 175 restrictions on plaintiff suits and Molecular diagnostics, 78
Long-term memory, 592 recovery as, 611–612 Molecular genetics, 32–41
Look-back, 325–326 risk management as, 612–613 See also Deoxyribonucleic acid (DNA);
Low-concentration glycerol technique, sources of law on, 608 Genetics
10, 10t Meiosis, 30–31, 31f, 31t of ABO, 125
Low ionic strength solutions (LISS), 68, Melting curve analysis, 92 deoxyribonucleic acid, 32–37
70, 113 Melting point (Tm), 80 ribonucleic acid, 37–40
Low-melting-point agarose, 85 Membrane attack complex (MAC), 60–61 Molecular immunohematology, 82–83t
Low responders, 54–55 Membrane filtration, 401–402 Molecular techniques, features relevant
Lua antigen, 202 Membrane modification, 466 to, 79–80
Lu(a–b–)phenotypes, 203–204, 204t Memory cells, 52 Monoclonal AHG production, 106–107,
Lub antigen, 202 Mendel, Gregor, 25 107f
Luebering-Rapaport shunt, 6 Mendelian genetics, 122 Monoclonal antibodies, 61–62
Luke (LKE) antigen, 184–185 Mendel’s laws of inheritance, 24–25, Monoclonal anti-D (MAb-D), 160
Luminometer, 92 25–27, 26f, 27f, 78 Monoclonal gammopathies, 72–73
Lung transplantation, 510 Messenger RNA (mRNA), 33, 38, 80, 81 Monoclonal reagents, 70–71
Lutheran blood group system, 201–204, Metabolic effects, adverse, to transfusion, Monocytes, 53
202t, 203f, 204t 387–388 Monokines, 54
Lyme disease, 322 Metabolic pathways, of RBCs, 5–7 Mononuclear phagocytic system (MPS),
Lymph nodes, 52t Metaphase, 30, 31t 52, 53
Lymphocytes, 50–51, 53–54 Methemoglobin reductase pathway, 6 Monospecific antiglobulin reagents, 443
Lymphocytic cells, 53 MHC. See Major histocompatibility Monospecific antihuman globulin, 70
Lymphocytotoxicity, 504–507 complex (MHC) in IAT, 108–109
Lymphoid lineage, 52–53 MHC class I, 51–52 Monospecific antihuman globulin
Lymphoid organs, 52, 52t MHC class II, 51–52 reagents, 105, 105t, 107
Lymphokines, 50, 54 Microarrays, DNA polymerase III, 91–92 Most probable genoytpes, 156–157, 156t,
Lysis, 119 Microlymphocytotoxicity, 504–507, 504f, 156–157t
518–519 MP4OX Hemospan, 13t
M Micro-RNA, 38 M phase, 32
MAC. See Membrane attack complex Microsatellites, 94 MPS. See Mononuclear phagocytic
(MAC) Middle cerebral artery peak systolic system (MPS)
Macrophages, factors affecting activity velocity (MCA-PSV), 433, 433f Mucosa-associated tissues, 52t
of, 454b Miniprep, 86 Multiplex assay, 92
6888_Index_653-670 29/10/18 11:32 AM Page 664

664 Index

Multiplex single-antigen bean Opsonization, 48 education, 567–568


immunoassay (SAB), 506–507, Optisol, 9, 9t goals of, 552t
506f Ordinary standard of care, 609–610 intervention strategies, 563–564, 564f
Mutagens, 35 Origin of replication, 85 introduction to, 552–555
Mutations Ortho Gel Matrix, 270f key players in, 556–559, 556t
DNA, 35–36, 36t, 37f O type RBCs, formation of, 11 transfusion committee, 557
duplications, 36 Oxyglobin, 13t transfusion guidelines, 557–559,
frameshift, 36 558t
missense point, 36 P transfusion safety officer, 556–557
nonsense, 36 P1 antibody, 184 patient-focused care and, 564–567
transversions, 36 P1 antigen, 183, 184 tools, 552t
Myeloid lineage, 52–53 P1Pk blood group system, 182–185 utilization auditing and review,
Packed RBCs, 3 560–563
N PAGE. See Polyacrylamide gel Patient-focused care, 564–567
electrophoresis (PAGE) Patient history, in antibody identification,
N-acetyl-D-galactosamine (GalNAc)
Palindromic sequences, 83 237–238
sugar, 124, 128
Pancreas transplantation, 510 Patient management, 596–597
NANEX technology, 11
Panel-reactive antibody (PRA), 505 Patient testing, 597–598
N antigen, 189
Pan Genera Detection (PGD) test, 17 Pauling, Linus, 80
National Blood Collection and Utilization
Papain, 56, 70 P blood group, 182–185, 182t, 183f
Survey (NBCUS), 3
Para-Bombay phenotypes, 135 biochemistry of, 183
National Center for Biotechnology
Paragloboside, 123 disease associations, 185
Information (NCBI), 93
Parentage index (PI), 523 genetics of, 183–184
National Institutes of Health
Parentage testing, 515–530 PBM. See Patient blood management
(NIH), 41, 575
accreditation and quality issues in, 527 (PBM)
Natural immunity, 47t
advantages and disadvantages of PCH. See Paroxysmal cold
Natural killer (NK) cells, 48, 51
genetic systems in, 526–527 hemoglobinuria (PCH)
Naturally occurring alloantibodies, 233
case studies, 528 Pedigree analysis, 28
Naturally occurring antibodies, 62
classical systems in, 518–519, 522 Peer review, 479
Natural selection, 25
criteria for selection of genetic systems PEG-tube testing, 115t
Negligence, 609–610
in, 517 PEL antigen, 225
Neonatal transfusions, 264, 366–367,
DNA polymorphisms and, 519, 523 Penicillin-induced hemolysis, 463–464
367b
genetic systems and technology used Pentose phosphate pathway, 6
Neutralization, in antibody identification,
in, 517–522, 517b, 518f, 518t Peptide bond synthesis, 40, 40f
242–243, 243t, 244f
goals of, 517 Peptides, 39
Neutrophils, 53
history of, 516–517 Perfluorocarbons, 12–13, 14t
NIH. See National Institutes
HLA system and, 518–519 Peripheral devices, 591
of Health (NIH)
inclusionary calculations, 523–526 Peripheral proteins, 4
Nonagglutinating antibodies, 104
introduction to, 515–516 Perlmutter v. Beth David Hospital, 608
Nonconformance costs, 546
nonclassic situations, 526 Personalized medicine, 41, 264
Nonconformance management, 541–542,
RFLP analysis, 519–520, 519f, 520t pH, agglutination reactions and, 67
541t, 542f
single nucleotide polymorphisms, 522 Phagocytic cells, 48, 53
Nonself, 46
social and legal issues in, 527 Phagocytosis, 48, 53
Nonsense mutations, 36
STR analysis, 520–522, 521f Pharmacogenomics, 41
Normal serum albumin (NSA), 350
terminology and interpretation of Phenotypes, 29–30, 175
Normal testing, 602
results, 522–523 writing, 175–176
Northern blotting technique, 91
Paroxysmal cold hemoglobinuria (PCH), Phenotypically matched adsorption, 245
NSA. See Normal serum albumin (NSA)
184, 451–452, 453t Photopheresis, 411
Nucleic acids, 32, 78
Partial-D antigens, 159f, 160, 160f Photoreactivation, 35
detection of, 91–93
Parvovirus B19, 319 Phototherapy, 435
hybridization, 91–92
PAS-C (Intersol), 15 Physical barriers, 48
Nucleic acid testing (NAT), 92–93
PAS-F (Isoplate), 15 Physical examination, of donors,
Nucleosomes, 32
Passenger lymphocyte syndrome, 423 291–292
Nucleotides, 79–80
Passively acquired antibodies, 233 Plaintiffs, 608
Nutricel, 9, 9t
PASSPORT study, 339 restrictions on plaintiff suits and
Paternity, 507 recovery, 611–612
O See also Parentage testing Plasma, 3, 357t
Obstetric patients, Rh testing Paternity index, 524–525 apheresis, 341
requirements for, 162 Pathogen inactivation (PI), 18, 325 collection, by apheresis, 403–404
Office of Blood Research and Review Pathogens, procedures to reduce and compatibility testing and, 261
(OBRR), 578 inactivate, 11 cryo-poor, 342
Okazaki fragments, 34 Pathological cold autoagglutinins, fresh-frozen, 341, 358t, 361–362, 410
Ok blood group system, 218 449–452 manufacturing and quality control,
Oligo-dT nucleotides, 83 Patient blood management (PBM), 264, 341–342
Oligonucleotides, 87 552–573 PF24, 358t, 361–362
Oligosaccharide chain, 124 blood order sets, 559–560, 559f PF24RT24, 358t
Oncology, transfusion in, 367 case studies, 569 pooled, 346
On the Origin of Species (Darwin), 25 continuous improvement in, 568–569 preparation of, from whole blood, 338b
Operating system software, 593 decision support and computerized thawed, 358t, 361–362
Opsonins, 48 order entry, 560 in transfusion therapy, 361–362
6888_Index_653-670 29/10/18 11:32 AM Page 665

Index 665

Plasma cells, 51 Polyclonal reagents, 70–71 compatibility tests, 259–260


Plasma derivatives, 325, 348–351, Polyethylene glycol (PEG), 70, 113 crossmatch tests, 261–262
357t–358t PolyHeme, 12, 13t donor unit selection in, 260–261
activated factor VII, 348 Polylinkers, 85 introduction to, 256
adverse reactions to, 385 Polymerase chain reaction (PCR), 25, issuance of blood components and, 264
antithrombin III concentrates, 80, 517 platelets, thawed plasma, and
350–351 allele-specific probes and, 93 cryoprecipitate, 261
factor IX concentrates, 349 in DNA replication, 88–90, 89f recipient testing in, 259–260
factor VIII concentrates, 348–349 real-time, 92 regulatory and accreditation
factor XIII concentrates, 349 reverse transcriptase, 92 requirements for, 256–257
immune serum globulin, 349 sequence-specific, 93 review of records for, 259
normal serum albumin, 350 Polymorphonuclear cells (PMNs), 52 Rh testing requirements for, 162
plasma protein factor, 350 Polyspecific antiglobulin reagents, 443 Rh typing in, 259
Rho(D) immune globulin, 350 Polyspecific antihuman globulin, 70, 106f special circumstances in, 263–264
synthetic volume expanders, 350 in IAT, 108–109 specimen collection in, 257–259
Plasma expressors, 334, 335f preparation of, 105–106 steps in, 257t
Plasma freezers, 335 Polyspecific antihuman globulin Preventive action, 543–544
Plasmapheresis, 296–297, 397, 408–409, reagents, 104–105, 105t Primary antibodies, 71
408b, 409–410, 410–411 Pooling, blood components, 346 Primers, 34
Plasma protein factor, 350 Population genetics, 25–29 Printers, 592
Plasma protein interactions, 412 Porcine factor VIII, 349 Prion disease, 324
Plasmids Postdonation instructions, 299, 302f Privacy
in DNA cloning, 85–86 Posterior probability of parentage or HIPAA Privacy Rule, 611
isolation of, 86–87 relatedness, 525–526 invasion of, 610–611
Plasmodium species, 323–324 Postoperative blood salvage, 295 Probability of exclusion (PE), 523, 526
Plastic storage bags, 15b Post-transfusion purpura (PTP), 387 Probability of paternity (PP), 523, 525–526
Platelet functional disorders, 368 Postzone effect, 66 Process, 537
Plateletpheresis, 296, 359–361, 397, 409 Potassium toxicity, 387, 387b Process control, 537, 538–539, 538f, 599t
Platelet refractoriness, 423 Potentiators, 69t Process improvement, 543–546
Platelets, 3, 357t PRA. See Panel-reactive antibody (PRA) LEAN, 545–546, 550
clinical use of, 15 Precipitation reaction, 65 Six Sigma, 544–545
collection, by apheresis, 404 Precision medicine, 41 Processing hardware, 591–592
compatibility testing and, 261 Predictive modeling, 567 Product recalls, 585–586
frozen, 19 Pregnancy Proenzymes, 48
manufacturing and quality control, Rh antibodies in, 161 Professional standard of care, 610
337–339 Rh testing requirements for, 162 Proficiency testing, 538, 543
pathogen reduction in, 18–19 Pre-mRNA, 82f, 83 Profiling, DNA, 94
performance criteria for concentrate Preoperative collection, 293 Progenitor cells, 478
collections, 16t Prescription Drug and Device User Fee Prokaryotic organisms, 29
pooled, 346 programs, 576 Prophase, 30, 31t
preparation of, from whole blood, Preseroconversion, 52, 92–93 Prosecution, 582–583, 583t
338b Preservation Prospective blood utilization review,
preservation of, 14–19 early research on, 2–3 562–563
additive solutions, 16b of platelets, 14–19 Protein media, agglutination reactions
refractoriness to transfusion and at 1°C to 6°C, new approaches to, 19 and, 68
alloimmunization, 387 additive solutions, 15 Proteins, 80
screening for bacterial contamination, bacterial contamination and, 16–17, antibodies as probes for, 93
17, 17b, 18t 17b, 18t detection of, 91–93
storage lesion, 14–15, 15t current trends and research, 19 heat-shock, 40
substitutes, 19 freezing, 19 parentage testing and, 518
testing and quality control monitoring, pathogen reduction and, 18–19 peripheral, 4
15–16 viability and functional properties of recombinant, 88, 88b
in transfusion therapy, 359–361, stored platelets, 16 transmembrane, 157, 157f
360b, 508 of red blood cells, 7–12 Protein synthesis, 40, 40f
uses of, 14 additive solutions, 9, 10t Proteolytic enzymes, 70, 241–242, 243t
viability and functional properties of anticoagulant preservative solutions, Provirus, 81
stored, 16 8, 8t, 9t Prozone effect, 66
in vitro assays, 15b current trends and research, 11–14 Pseudoantigens, 135
Platelet swirl, 14, 16 freezing, 10–11, 11t Pseudo-B antigen, 139
PMNs. See Polymorphonuclear cells rejuvenation, 11 Pseudogenes, 36
(PMNs) Preservative solutions, development Public Health Service Act, 575, 578, 579
Polarity, 79 of, 2–3 Purines, 32, 33f
Polyacrylamide gel electrophoresis Prestorage pooled WBD platelets, 17 PX2 antigen, 185
(PAGE), 520 Presurgical autologous blood donation Pyrimidines, 32, 33f
Poly-A tail, 83 (PAD), 565–566
Polybrene, 70 Pretransfusion testing, 256–267 Q
Polyclonal AHG production, 105–106, ABO grouping in, 259 Quality
106f antibody screening in, 259–260 cost of, 546–547
Polyclonal antibodies, 61–62 case studies, 264–265 foundation of, 532–534
Polyclonal gammopathies, 72–73 causes of positive, 262–263t in relationship testing, 527
6888_Index_653-670 29/10/18 11:32 AM Page 666

666 Index

Quality assessment, 533 Reconstituted whole blood, 346 social and legal issues in, 527
Quality assurance, 533, 598–599 Records, 539–540 terminology and interpretation of
Quality control (QC), 532–533, 532f, Records management, 540 results, 522–523
583–584 Red blood cell agglutination, 65 Relevant transfusion-transmitted
blood bank testing and, 278 Red blood cell alloimmunization, HDFN infections (RTTIs), 575
blood product manufacturing and, caused by, 429–430, 431–433 Replication, 80
335–348 Red blood cell antibodies, binding of DNA, 34
for deglycerolization, 348b complement by, 61 semiconservative, 34, 80
platelets, 15–16 Red blood cell antibody specificity, 429, Respiratory movement, 7
in tissue banking, 481 429t Respondeat superior, 609–610
Quality control plan (QCP), 533 Red blood cell exchange, 409 Responders, 54–55
Quality management Red blood cells (RBCs), 357t Restriction endonucleases, 83–84, 84t
in blood bank, 531–551 adsorption, 245–246 Restriction enzyme mapping, 84
vs. compliance, 532 antigen-antibody reactions in, Restriction fragment length
introduction to, 532 detection of, 65 polymorphism (RFLP), 84,
structure of, 532–534, 532f antigens, formation of, 123 93, 93f
Quality management system, 533–546, biology of, 4–7 analysis, 519–520, 519f, 520t
536f collection, by apheresis, 403 testing, 516–517, 516f
case studies, 547–549 deformability of, 4 Restriction mapping, 25
documents and records, 540f freezing, 10–11, 10t, 11t, 347t Retrospective blood utilization review,
essentials for, 534–543, 536b frozen, deglycerolized, 347–348, 359 561–562
assessments, 542–543 genotyping, 94–97 Retroviruses, 81
continual improvement, 543 clinical applications of, 95, 97–98 Reverse genetics, 94
customer focus, 535 hemolysis of, 453–454 Reverse grouping, 121t
documents and records, 539–540 laboratory creation of, 11–12 ABO discrepancies between forward
equipment, 537 leukocyte-reduced, 359, 359t, 364 grouping and, 143, 143t–144t
facilities and safety, 536 manufacturing and quality control, Reverse transcriptase PCR (RT-PCR), 92
information management, 540 336–337 Reverse transcription, 81, 82f
organization, 534–535 membrane of, 4–5, 5f RFLP. See Restriction fragment length
personnel, 536–537, 537b metabolic pathways, 5–7, 6f polymorphism (RFLP)
process management, 537–539 O type, formation of, 11 Rh17 (Hr0) antigen, 165
purchasing and inventory, 537 packed, 3 Rh23 antigen, 165
importance of, 547 permeability of, 4–5 Rh30 antigen, 165
for medical laboratories, 546, 546f preservation of, 7–12 Rh32 antigen, 166
nonconformance management, 541–542 rejuvenated, 11, 359 Rh33 (Har) antigen, 165
process improvement and, 543–546 substitutes, 12–13, 12b, 12t, 13t, 14t Rh40 antigen, 165
Quality reviews, 584 tissue engineering of, 13 Rh43 (Crawford) antigen, 166
Quality system essentials (QES), in transfusion therapy, 356, 359 Rh52 antigen, 165
533–534, 534b washed, 113, 359 RHAG gene, 154–155, 164
Quarantine, 325–326 zeta potential of, 66, 67–68 Rh antibodies
Red blood cell storage lesion, 7, 7t case study on, 167–168
R Registration characteristics of, 161
Radio frequency identification (RFID), 602 of blood establishments, 579, 579t detecting and identifying, 162–163
Random access memory (RAM), 591 donor, 283 in hemolytic disease of the fetus and
Random hexamers, 83 Regulations newborn, 164
Raph blood group system, 218–219 federal, 574–589, 578b in pregnancy, 161
RBC reagents, 234–235 on information systems, 599–600 testing for, 161–163
RBCs. See Red blood cells (RBCs) as source of laws, 608 in transfusion reactions, 163–164
Reaction mediums, for AGT, 112–113 Regulatory overview, of tissue banking, Rh antigen D (RhD), 158, 158t, 427
Read-only memory (ROM), 591 485–491, 485–486t Rh antigens, 158t
Reagents Regulatory process, 576–578 biochemistry of, 157–158
See also specific types Rejuvenated RBCs, 11, 359 characteristics of, 158
for adsorption, 245 Relationship index (RI), 523, 524–525, common (C, E, c, e), 158, 158t, 159t
in antibody identification, 238 525t D antigen, 158, 158t, 427
enhancement, 235 Relationship testing, 515–530 frequency of, 158t
monoclonal vs. polyclonal, 70–71 accreditation and quality issues in, 527 rare, 164–166
RBC, 234–235 advantages and disadvantages of testing for, 161–163
Rh typing, 161–162 genetic systems in, 526–527 Rh-associated glycoprotein (RHAG),
saline reactive, 161 case studies, 528 154–155
Real-time PCR, 92 classical systems in, 518–519, 522 Rh-associated glycoprotein blood group
Recalls, 493–494, 494b, 585–586 criteria for selection of genetic systems system, 219–220
Recessive traits, 28 in, 517 Rh blood group system, 149–172
Recipient tracing, 325–326 DNA polymorphisms, 523 antigens of, by four nomenclatures,
Recombinant DNA, 81–90 genetic systems and technology used 153–154t
in clinical use, 88 in, 517–522, 517b, 518f, 518t biochemistry of, 157–158
libraries, 87 goals of, 517 case studies, 167–169
Recombinant enzymes, 88 history of, 516–517 clinical considerations for, 163–164
Recombinant factor VIII, 349 inclusionary calculations, 523–526 D antigen expression variations in,
Recombinant proteins, 88, 88b introduction to, 515–516 159–161, 159–160f, 161f
Recombination repair, 35 nonclassic situations, 526 genetics of, 154–157, 155f
6888_Index_653-670 29/10/18 11:32 AM Page 667

Index 667

history of, 150 Saline, for washing, 113 Solidscreen II assay, 275–276
introduction to, 150 Saline reactive reagents, 161 SOLX, 9, 9t
Rh deficiency syndrome and, 164 Saline-tube testing, 115t Somatic gene therapy, 88I
terminology, 150–154, 152t, 153–154t S antigen, 189–190 SOS repair, 35
alphanumeric, 151–152 s antigen, 189–190 Southern blotting technique, 91, 91f
DCE, 150–151, 151f Satellite bags, 9 Species, 25
Rh-Hr, 151 Scales, 334–335 Specificity, 64
updated numeric, 152 Scianna blood group system, 215 Specimen collection
unusual phenotypes and rare alleles, Sda antigen, 224–225 patient identification and specimen
164–166 SDS-PAGE electrophoresis, 93 labeling in, 257–258, 258f
RHCE gene, 154, 164 Secondary cold AIHA, 451 in pretransfusion testing, 257–259
RhCE protein, D epitopes on, 160–161, Secretor ABH glycoprotein substances, 126f specimen requirements in, 258–259
161f Secretory studies, 65 Spectra Optia, 401f
Rh deficiency syndrome, 164 Security Spectrin, 78
RHD gene, 154, 155–156 HIPAA Security Rule, 611 Spherocytes, 4, 5f
RhD hemolytic disease of the information system, 598, 600–601 Spleen, 52t
fetus/newborn, 428t Sedimented blood sample, 397f Splenectomy, for WAIHA, 460
RhD typing Seizure, 582, 583t SSO. See Sequence-specific
cold-reactive autoantibodies and, Selected cell panels, in antibody oligonucleotides (SSO)
445–446 identification, 241, 242f SSP. See Sequence-specific PCR (SSP)
of cord blood, 434 Selective absorption, 410–411 Standard of care, 609–610
warm autoantibodies and, 454–455 Self, 46 Standard operating procedures, 538
Rh genes, 154, 155f Semiconservative replication, 34, 80 Standards for Blood Banks and Transfusion
Rh-Hr terminology, for Rh blood group Sensitization, 104 Services, 402, 533–534
system, 151, 151f, 152t, 153–154t Sensitizing events, 501 Start codons, 33
rhi (Ce) antigen, 165 Sequence-specific oligonucleotide probe State laws, on tissue banking, 488
Rh immune globulin (RhiG), 363–364, (SSOP), 93, 93f Static files, 593
427, 435–436, 436f Sequence-specific oligonucleotides Statutes, 577, 608
Rhmod syndrome, 164 (SSO), 503 blood shield, 608
Rh-negative phenotypes, 155–156 Sequence-specific PCR (SSP), 93, 94f of limitations, 611
Rhnull syndrome, 164 Sequence-specific primers (SSP), 83, 503 Stem cells, 419, 478
Rho(D) immune globulin, 350 Sequencing, 25 hematopoietic, 11, 13
Rh-positive phenotypes, 155 Serologic activity, 498, 499–500 Sterile connection devices, 335
Rh transplant incompatibility, 423 Serologic crossmatch, 261–262 Stop codons, 33
Rh typing, in pretransfusion testing, 259 Serologic systems, 68t Storage devices, 335
Rh typing reagents, 161–162 Serologic testing, diseases important Storage lesion, 388t
false reactions with, 163t in, 71–73, 72b platelet, 14–15, 15t
Ribonucleic acid (RNA), 25, 37–40, Serum, ratio of, to cells, 112 RBC, 7, 7t
78, 80 Short supply arrangements, 585 Storage time, 388t
heterogeneous, 82f, 83 Short tandem repeats (STRs), 94, 517 STR analysis, 520–522, 521f
isolation, 37 Sickle cell anemia, 80, 97 Strict liability, 610
messenger, 33 Sickle cell disease (SCD), 377 Substitutes
ribosomal, 38 Sigmoid-curve relationship, 7 platelet, 19
structure of, 37 Silent genes, 30 RBC, 12–13, 12b, 12t, 13t, 14t
transcription, 29, 38–39, 39f Single-antigen bean immunoassay (SAB), Superantigens, 54
translation, 39–40, 39f, 40f 506–507, 506f Surgical blood order schedule, type, and
types of, 37–38 Single nucleotide polymorphism (SNP), screen, 365, 365b
Ribosomal RNA (rRNA), 38 25, 92, 522 Surrogate markers, 308
Ribosomes, 38, 39, 80 Six Sigma, 544–545 SYBR Green, 85
Risk assessment (RA), 533, 601 Small tandem repeats (STRs), 517 Syngeneic HPC transplantation, 418
Risk management, 612–613 SNP. See Single nucleotide polymorphism Synthetic volume expanders, 350
medical malpractice and, 613 (SNP) Syphilis, 321–322
processing, labeling, and distribution, Society for the Advancement of Blood screening donor blood for, 302–303,
612–613 Management (SABM), 555 308t
specific donor issues, 612 Sodium citrate, 2 tests for, 326t
RNA. See Ribonucleic acid (RNA) Sodium dodecyl sulfate (SDS), 86 System index (SI), 524–525, 525t
RNA virus, 303 Sodium hydroxide, 86 System management, 600–602
RNC antigens, alloimmunization Sodium phosphate, 2 System managers, 594
to, 377 Software, 592–599 Systems biology, 94–95
Rocky Mountain spotted fever backup of, 600
(RMSF), 322 validation, 601–602 T
Rodgers blood group, 36 Solid-phase technology, 114–115, 115t, TA. See Therapeutic apheresis (TA)
Rouleaux formation, 140t 272–274 TACO. See Transfusion-associated
RTTIs. See Relevant transfusion- Bio-Rad solid-phase protein A circulatory overload (TACO)
transmitted infections (RTTIs) technology, 274–276, 275f, TaqMan, 92
277–278t Targeted prospective review, 562–563
S Immucor solid-phase red cell T cell-independent antigens, 54
SAB. See Single-antigen bean adherence, 273–274, 274f, 277f, T-cell receptor (TCR), 54
immunoassay (SAB) 277–278t TCP/IP protocol, 592
Safety. See Transfusion safety introduction to, 272 T cytotoxic cells, 51–52
6888_Index_653-670 29/10/18 11:32 AM Page 668

668 Index

Team Biologics, 581 Tolerance, 64–65 Transfusion-related acute lung injury


Telophase, 30, 31t Torts, 608–611 (TRALI), 16b, 368, 380–381, 380b,
Temin, Howard, 81 battery, 609 381b, 382f, 504, 508
Temperature negligence, 609–610 Transfusion-related fatalities, 586, 586t
agglutination reactions and, 67 reform, 612 Transfusion-related immunomodulation
AGT and, 113 Total process control, 537, 538f (TRIM), 73
Terminal galactose, 124 Traceability, in tissue banking, 480–481 Transfusion(s)
Test cases, 601–602 Transcription, 29, 78 ABO-incompatible, 120b
Test plan, 601 reverse, 81 adverse effects of, 373–395. see also
Thalassemia, 135 RNA, 38–39, 39f Transfusion reactions
Thawed plasma, cryoprecipitate reduced, Transcription factors, 78 autologous, 10, 292–295, 365, 366f
361–362 Transcription-mediated amplification blood administration, 368–369, 368f
T helper cells, 51–52 (TMA), 92–93 coagulation factor deficiencies and,
Therapeutic apheresis (TA), 397, 405–410 Transfer RNA (tRNA), 38, 38f, 80 367–368
erythrocytapheresis, 409 Transformation, 86 compatible blood for, selecting, 163
fluid replacement, 409–410 Transfusing facility software, 596–598 current status of, 3–4
general considerations for, 405–406, Transfusion-associated circulatory donor selection for, 281–306
408 overload (TACO), 368, 381–383, agencies governing, 282–283
indication categories for, 406–407t 382t, 383f case studies, 303–304
leukapheresis, 409 Transfusion-associated dyspnea informed consent in, 292, 292f
physiological considerations for, 408 (TAD), 383 introduction to, 282
plasmapheresis, 408–409, 408b, Transfusion-associated fatalities, 374, 375t medical history questionnaire in,
409–410, 410–411 Transfusion-associated graft-versus-host 283–284, 284–285f
plateletpheresis, 409 disease (TA-GVHD), 364–365, medical history questions inn,
vascular access in, 406, 408 385–387, 386f, 421 285–291
Therapeutic plasma exchange (TPE), Transfusion-associated hepatitis, 308–313 physical examination in, 291–292
408–409, 408b, 410–411 Transfusion-associated parasites, 322–324 registration in, 283
Thermocycler, 89 Transfusion audit, 369–370, 369–370t whole blood collection
3′ and 5′ ends, 80 Transfusion committee, 557 and, 297–299
Thrombocytopenia, 15 Transfusion guidelines, 557–559, 558t emergency, 365–366
Thymine, 32, 33f, 35, 80 Transfusion medicine emergent, 263
Thymus, 52t, 53 ethics and, 613 exchange, 434–435
Tick-borne bacterial agents, 322 future of, 264 fatalities, 369t
Tissue, 491 medicolegal issues in, 607–616 general practices for, 368–370, 368f
Tissue banking, 476–496 types of waste in, 554, 555f historical overview, 2–3
See also Tissue transplantation Transfusion reactions, 373–395 immune system and, 73
acquisition of allografts and inventory, acute hemolytic, 378–379, 378t incompatible, 2
479–480 adverse metabolic effects, 387–388 intrauterine, 264, 433–434
autologous tissue, 482–485 allergic, 384–385, 384t, 385b, 386f massive, 263–264, 366, 366t
case studies on, 494 alloimmunization to RBC antigens, 377 neonatal, 264, 366–367, 366b
common tissues in, 478 case studies, 390–392 in oncology, 367
hospital tissue bank for, 477–481 delayed hemolytic and serologic, rapid, 369
introduction to, 476–477 379–380 risk mitigation strategies, 553–554, 554f
oversight of, 479 fatal hemolytic, 120b, 120t risks of, 374
qualification of vendors fatalities, 374, 375t surgical blood order schedule, type,
in, 481–482, 482t febrile nonhemolytic, 383–384 and screen, 365, 365b
quality assurance in, 481 hypotensive, 383 in WAIHA, 458, 460
record-keeping and traceability, immunomodulatory effects, 388–389 Transfusion safety, 574–589
480–481 incidence of, 374, 374b See also Federal regulatory
regulatory overview of, 485–491, infectious, 389–390 requirements
485–486t introduction to, 374 enforcement actions and, 582–583
state laws governing, 482t noninfectious, 377–389 FDA inspection process and, 581
storage in, 480 to plasma-derived products, 385 introduction to, 575
tissue dispension, 481 post-transfusion purpura, 387 Transfusion safety officer, 556–557
Tissue dispensing service, 476 recognition and evaluation Transfusion therapy, 355–372
Tissue engineering, of RBCs, 13 of, 374–375, 375f, 376f blood products in, 356–364
Tissue transplantation refractoriness to platelet transfusion case studies, 370–371
adverse events involving, 491–494 and alloimmunization, 387 introduction to, 356
allograft failure, 492 regulatory and accreditation in special conditions, 365–368
infection, 492 requirements related to, 375, special considerations for, 364–365
malignancy, 492 376t, 377b Transfusion-transmitted AIDS (TTAIDS),
recalls, 493–494, 494b Rh blood group system and, 163–164 608, 613
reporting and investigation signs and symptoms of, 375f Transfusion-transmitted bacterial
of, 492–493 transfusion-associated circulatory infections (TTBIs), 389–390,
case studies on, 494 overload, 381–383, 382t, 383f 389t, 390t
history and overview of, 477 transfusion-associated dyspnea, 383 Transfusion-transmitted diseases (TTDs),
Titers, 121–122 transfusion-associated graft-versus- 307–332
T lymphocytes (T cells), 50–51, 53–54 host disease, 385–387, 386f babesiosis, 322–323
TMA. See Transcription-mediated transfusion-related acute lung injury, bacterial, 320–322, 389–390,
amplification (TMA) 380–381, 380b, 381b, 382f 389t, 390t
6888_Index_653-670 29/10/18 11:32 AM Page 669

Index 669

Chagas disease, 323 U Warm autoimmune hemolytic anemia


chikungunya virus, 319–320 Ultraviolet (UV) radiation, 35 (WAIHA), 453–462
Creutzfeldt-Jacob disease, 324 Umbilical cord blood, 141, 419, 434 clinical findings in, 453–454
cytomegalovirus, 318 Unbranched straight chains, 129 compared with cold, 463t
dengue virus, 320 Unexpected antibodies, 233 diseases associated with, 453b
donor testing and, 307–308 United Network for Organ Sharing RBC hemolysis in, 453–454
Ebola virus, 320 (UNOS), 510 serologic characteristics and laboratory
Epstein-Barr virus, 318 Unregistered transfusion services, tests affected by, 454–458, 456t
hepatitis as, 308–313, 309f, 309t, 580–581 transfusion in, selection of blood for,
310f, 311t U- phenotype, 192 458, 460
herpesvirus 6 and 8, 319 Urticarial reactions, 72 treatment of, 460
HIV, 313–314, 313f, 313t U.S. Food and Drug Administration. See Warning Letters, 582, 583t
HPC transplantation and, 421 Food and Drug Administration Washing, 344–345
human T-cell lymphotropic viruses (FDA) Watson, James, 32
types I/II (HTLV-I/II), 314–315, Utilization management, 552–573 Watson-Crick base pairing, 32
316f See also Patient blood management Weak A subgroups, 131–132, 131t, 133f
introduction to, 307 (PBM) Weak B subgroups, 132–134
laboratory tests for, 326–327t Weak D antigen, 159f
malaria, 323–324 D antigen, expression of variations of,
V
parasites, 322–324 159–161
parvovirus B19, 319 Valency, 55, 64 Weiner haplotype terminology, 152t
pathogen inactivation and, 325 Validation, 538 Western blotting (WB), 65, 93
prion disease, 324 Van der Waals forces, 56 West Nile virus (WNV), 315–317
quarantine and, 325–326 V antigens, 166 screening donor blood for, 302, 308t
recipient tracing and, 325–326 Variable number of tandem repeats tests for, 327t
required tests for, 308b (VNTRs), 94, 516 Whole blood, 357t
syphilis, 321–322 Variable regions, 56 components of, 3
tick-borne, 322 Variances, 584 fractionation procedure, using
West Nile virus, 315–317 Vascular access, in therapeutic apheresis, centrifugation, 336b
Zika virus, 317–318 406, 408, 412 manufacturing and quality control,
Transfusion-transmitted infectious Vasovagal reactions, 412 335–336, 337b
diseases, 421 Vasovagal syncope, 412 reconstituted, 346
Translation, 39–40, 39f, 40f, 78, vCJD, 289–290, 290t in transfusion therapy, 356
80–81 Vectors, 85–86 Whole blood collection, 297–299
Transmembrane proteins, 157, 157f viral, 88 arm preparation methods, 298b
Transmissible spongiform Vel blood group system, 220–221 aseptic technique for, 298
encephalopathies (TSE), 289 Vendors, tissue, 481–482, 482t donor reactions to, 299
Transplantation Verax PGD test, 18t, 339 procedure for, 298, 298b
hematopoietic progenitor cell. see Viral vectors, 88 Whole blood-derived platelets (WBD), 15
Hematopoietic progenitor cell Virtual blood banks, 602, 603f Wiener terminology, 151, 151f
(HPC) transplantation Virtual crossmatch, 507 Wilkins, Maurice, 79
tissue. See Tissue transplantation Virtual private networks (VPNs), 592 Window period, of immune response, 52
Transversions, 36 Viruses Work process, 538
TRICC trial, 264 provirus, 81 World Health Organization Nomenclature
Trima Accel Automated Collection retroviruses, 81 Committee for Factors of the HLA
System, 402f tumor, 81 System, 498–499
True chimerism, 138 Viscoelastic blood testing, 566 World War II, 2–3
Trypanosoma cruzi, 323, 323f Vitamin K, 367–368
screening donor blood for, 303, 308t VNTRs. See Variable number tandem X
Trypsin, 70 repeats (VNTR)
Voluntary standards, 610 Xg blood group system, 214–215
TTDs. See Transfusion-transmitted X-linked inheritance, 28–29, 28f, 29t
diseases (TTDs) Von Willebrand’s disease, 367
Tube antibody detection VS antigens, 166
screen, 233–235, 234f
Y
Tubing sealers, 335 W Yt blood group system, 214
Tumor necrosis factor (TNF), 54 WAIHA. See Warm autoimmune
Tumor viruses, 81 hemolytic anemia (WAIHA) Z
21st Century Cures Act, 576 Warm autoantibodies, 251–252, Z-DNA, 32
Type 2 precursor chain, 124, 124f 453–462 Zeta potential, 66, 67–68
Type H oligosaccharide (glycan) IgM, warm autoimmune hemolytic Zika virus, 317–318
chains, 125 anemia with, 462 screening donor blood for, 303, 308t
Type III reactions, 72 with underlying alloanti-E, 459t tests for, 327t
Type II reactions, 72 with underlying alloanti-K and -Jka, Zone of equivalence, 66
Type I reactions, 72 461t ZZAP reagent, 70
6888_Index_653-670 29/10/18 11:32 AM Page 670
6888_IBC 25/10/18 2:31 PM Page 2

Antigen-Antibody Characteristic Chart*


ANTIGENS
Antigen RBC Antigen Antigen Antigen
Antigen Antigen ISBT Freq. % Expression Distrib. Demonstrates Modification
System Name Name W B at Birth Plasma/RBC Dosage Enzyme/Other
Jka JK1 77 91 strong RBC only yes Enz. ↑

Kidd
Jkb JK2 73 43 strong RBC only yes Enz. ↑

JK3 100 100 strong RBC no Enz. ↑


Lea LE1 22 23 nil Plasma/RBC no Enz. ↑
Lewis

Leb LE2 72 55 nil Plasma/RBC no Enz. ↑


P1 P1PK1 79 94 moderate RBC, platelets, individual Enz. ↑ AET → ZZAP ↑
WBC variation
P GLOB1 100 100 moderate RBC, platelets, no Enz. ↑ AET → ZZAP ↑
WBC
†P

‡Pk P1PK3 100 100 moderate RBC, platelets, no no


fibroblast
M MNS1 78 74 strong RBC only yes Enz. ↓ AET → ZZAP ↓

N MNS2 72 75 strong RBC only yes Enz. ↓ AET → ZZAP →

MNS

S MNS3 55 31 strong RBC only yes Enz.V AET → ZZAP ↓


s MNS4 89 93 strong RBC only yes Enz.V AET → ZZAP ↓
U MNS5 99.9 99.9 strong RBC only no Enz. → AET → ZZAP →
Lua LU1 8 5 poor RBC only yes Enz. → AET → ZZAP ↓

Lutheran

Lub LU2 99.8 99.9 poor RBC only yes Enz. → AET → ZZAP ↓
LU3 >99.8 99.9 poor RBC only no Enz. → AET → ZZAPV

*This chart is to be used for general information only. Please refer to the appropriate chapter for more detailed information.
AET = 2-aminoethylisothiouronium bromide; ↑ = enhanced reactivity; → = no effect; ↓ = depressed reactivity; occ = occasionally; HDN = hemolytic disease of the newborn;
HTR = hemolytic transfusion reaction; NRBC = non-red blood cell; RBC = red blood cell; WBC = white blood cell; ZZAP = dithiothreitol plus papain. PCH = paroxysmal cold
hemoglobinuria; ↑ = enhanced reactivity; V = variable
†In the P system, phenotype P1 contains both P1 P, and Pk antigens; phenotype P2 contains only P antigens; phenotype p lacks both P1 P, and Pk antigens.
‡The Pk antigen is typically converted to P; therefore there is no Pk antigen detectable on adult cells. There are rare individuals (P1k, P2k) where pk antigen is not converted to P.
6888_IBC 25/10/18 2:31 PM Page 3

ANTIBODIES
Immunoglobin Clinical
Serology Comp. Class Optimum Significance
Stimulation Saline AHG Binding IgM IgG Temperature HTR HDN Comments
RBC no yes yes no yes warm yes yes Kidd antibodies seem to disappear
rapidly both in vivo and in vitro.
They are often associated with
delayed HTR.
RBC no yes yes no yes warm yes yes Jk(a–b–) RBCs are resistant to lysis by
2M urea.
RBC rare yes yes no yes warm yes yes
NRBC yes some yes yes occ cold rare no Sometimes a pattern of unusual
agglutination sheeting occurs.
NRBC yes some yes yes occ cold no no Antigen expression for the Lewis
system may be lost during pregnancy.
NRBC yes no some yes rare cold yes no Commonly occurring antibody in
P2 individuals.
NRBC yes occ yes most few cold yes rare Anti-P may occur “naturally” in Pk
individuals; it may occur as an
auto-antibody in PCH, in which case
it is a cold reactive IgG antibody and
causes in vivo hemolysis.
NRBC yes yes yes cold rare Anti-PP1Pk may occur in p individuals
and can cause HTR and HDN.
NRBC yes some rare yes occ cold rare rare M–N–cells may also be En(a–), in
which case the cells are resistant to
invasion by P. falciparum merozoites.
NRBC yes no rare yes occ cold rare rare Anti–N–like antibodies may be pro-
duced in renal dialysis patients where
the dialysis machine has been steril-
ized with formaldehyde (i.e., Anti-NF).
RBC some yes some occ yes warm yes yes
RBC rare yes occ occ yes warm yes yes
RBC no yes no no yes warm yes yes
NRBC yes some some yes occ cold no v.mild Lu(a–b–) cells may result from in-
heritance of the recessive Lu gene or
from the dominant In(Lu)gene. These
cells are labile and hemolyze easily
on storage. The antibody Anti-Lua
demonstrates a characteristic loose
mixed-field agglutination pattern.
RBC occ yes some yes yes warm yes mild

You might also like