You are on page 1of 67

Congenital and Acquired Bone Marrow

Failure 1st Edition Edition Mahmoud


Aljurf
Visit to download the full and correct content document:
https://ebookmass.com/product/congenital-and-acquired-bone-marrow-failure-1st-edit
ion-edition-mahmoud-aljurf/
CONGENITAL
AND ACQUIRED
BONE MARROW
FAILURE
Edited by

M.D. Aljurf
Adult Hematology and Bone Marrow Transplantation
Oncology Center, King Faisal Specialist Hospital
and Research Center, Riyadh, Saudi Arabia

E. Gluckman
Eurocord, Saint Louis Hospital, Paris, France

C. Dufour
Hematology Unit
G. Gaslini Children’s Hospital, Genova, Italy

 
Academic Press is an imprint of Elsevier
125 London Wall, London EC2Y 5AS, United Kingdom
525 B Street, Suite 1800, San Diego, CA 92101-4495, United States
50 Hampshire Street, 5th Floor, Cambridge, MA 02139, United States
The Boulevard, Langford Lane, Kidlington, Oxford OX5 1GB, United Kingdom

Copyright © 2017 Elsevier Inc. All rights reserved.

No part of this publication may be reproduced or transmitted in any form or by any means, electronic or mechanical,
including photocopying, recording, or any information storage and retrieval system, without permission in writing from the
publisher. Details on how to seek permission, further information about the Publisher’s permissions policies and our arrange-
ments with organizations such as the Copyright Clearance Center and the Copyright Licensing Agency, can be found at our
website: www.elsevier.com/permissions.

This book and the individual contributions contained in it are protected under copyright by the Publisher (other than as may
be noted herein).

Notices
Knowledge and best practice in this field are constantly changing. As new research and experience broaden our understand-
ing, changes in research methods, professional practices, or medical treatment may become necessary.

Practitioners and researchers must always rely on their own experience and knowledge in evaluating and using any informa-
tion, methods, compounds, or experiments described herein. In using such information or methods they should be mindful
of their own safety and the safety of others, including parties for whom they have a professional responsibility.

To the fullest extent of the law, neither the Publisher nor the authors, contributors, or editors, assume any liability for any
injury and/or damage to persons or property as a matter of products liability, negligence or otherwise, or from any use or
operation of any methods, products, instructions, or ideas contained in the material herein.

Library of Congress Cataloging-in-Publication Data


A catalog record for this book is available from the Library of Congress

British Library Cataloguing-in-Publication Data


A catalogue record for this book is available from the British Library

ISBN: 978-0-12-804152-9

For information on all Academic Press publications


visit our website at https://www.elsevier.com/

Publisher: Mica Haley


Acquisition Editor: Tari Broderick
Editorial Project Manager: Kathy Padilla
Production Project Manager: Karen East and Kirsty Halterman
Designer: Victoria Pearson

Typeset by Thomson Digital


List of Contributors

S.O. Ahmed Adult Hematology and Bone Marrow S. Elmahdi Department of Pediatrics, Graduate
Transplantation, Oncology Center, King Faisal School of Medicine, Nagoya University, Nagoya,
Specialist Hospital and Research Center, Riyadh, Japan
Saudi Arabia
P. Farruggia Pediatric Hematology and Oncology
G. Aldawsari Adult Hematology and Bone Marrow Unit, A.R.N.A.S. Ospedale Civico, Palermo, Italy
Transplantation, Oncology Center, King Faisal
F. Fioredda Hematology Unit, G. Gaslini Children’s
Specialist Hospital and Research Center, Riyadh,
Hospital; Unità di Ematologia Istituto Giannina
Saudi Arabia
Gaslini, Genova, Italy
M.D. Aljurf Adult Hematology and Bone Marrow
S. Gandhi Department of Haematological Medicine,
Transplantation, Oncology Center, King Faisal
King’s College Hospital/King’s College London,
Specialist Hospital and Research Center, Riyadh,
London, United Kingdom
Saudi Arabia
S. Giraudier Hematology Laboratory, Henri–
H. Alzahrani Adult Hematology and Bone Marrow
Mondor Hospital, Paris–Est–Creteil University,
Transplantation, Oncology Center, King Faisal
Paris, France
Specialist Hospital and Research Center, Riyadh,
Saudi Arabia E. Gluckman Eurocord, Hospital Saint-Louis, Paris,
France
M. Ayas Pediatric Hematology–Oncology and Stem
Cell Transplantation, King Faisal Specialist Hospital F. Grimaldi Hematology, Department of Clinical
and Research Center, Riyadh, Saudi Arabia Medicine and Surgery, Federico II University,
Naples, Italy
A. Bacigalupo Istituto di Ematologia, Policlinico
Universitario A. Gemelli, Universita’ Cattolica del B. Höchsmann Institute of Transfusion Medicine,
Sacro Cuore, Roma, Italy University of Ulm; Institute of Clinical Transfusion
Medicine and Immunogenetics, German Red Cross
F. Ciceri Hematology and BMT Unit, IRCCS San
Blood Transfusion Service Baden–Württemberg–
Raffaele Scientific Institute; University Vita-
Hessia, and University Hospital Ulm, Ulm,
Salute San Raffaele, IRCCS San Raffaele Scientific
Germany
Institute, Milano, Italy
K. Hosokawa National Institutes of Health, National
J.N. Cooper National Institutes of Health, National
Heart, Lung, and Blood Institute, Bethesda, MD,
Heart, Lung, and Blood Institute, Bethesda, MD,
United States
United States
S. Kojima Department of Pediatrics, Graduate
J.H. Dalle Hematology–Immunology Pediatric
School of Medicine, Nagoya University, Nagoya,
Department, Robert–Debre Hospital, Paris,
Japan
France
T. Leblanc Hematology–Immunology Pediatric
C. Dufour Hematology Unit, G. Gaslini Children’s
Department, Robert–Debre Hospital, Paris, France
Hospital; Unità di Ematologia Istituto Giannina
Gaslini, Genova, Italy

ix
x List of Contributors

J.C.W. Marsh Department of Haematological P. Scheinberg Oncology Center, Hospital São José
Medicine, King’s College Hospital/King’s College National Institutes of Health, National Heart, Lung,
London, London, United Kingdom and Blood Institute, Bethesda, MD, United States
D. Meyran Hematology–Immunology Pediatric H. Schrezenmeier Institute of Transfusion
Department, Robert–Debre Hospital, Paris, France Medicine, University of Ulm; Institute of Clinical
Transfusion Medicine and Immunogenetics,
M. Miano Hematology Unit, G. Gaslini Children’s
German Red Cross Blood Transfusion Service
Hospital; Unità di Ematologia Istituto Giannina
Baden–Württemberg–Hessia, and University
Gaslini, Genova, Italy
Hospital Ulm, Ulm, Germany
G.J. Mufti Department of Haematological
S. Sica Istituto di Ematologia, Policlinico
Medicine, King’s College Hospital/King’s College
Universitario A. Gemelli, Universita’ Cattolica del
London, London, United Kingdom
Sacro Cuore, Roma, Italy
J.R. Passweg Division of Hematology, University
M.T.L. Stanghellini Hematology and BMT Unit,
Hospital Basel, Basel, Switzerland
IRCCS San Raffaele Scientific Institute, Milano,
R. Peffault de Latour BMT Unit, French Reference Italy
Center for Aplastic Anemia and PNH, Saint-Louis
A. Tichelli Hematology, University Hospital of
Hospital, Paris, France
Basel, Basel, Switzerland
A.M. Risitano Hematology, Department of Clinical
M.T. Van Lint Divisione di Ematologia e Trapianto
Medicine and Surgery, Federico II University,
di Midollo Osseo, IRCCS AOU San Martino IST,
Naples, Italy
Genova, Italy
A. Rovó Hematology, University Hospital of Bern,
A.J. Warren Cambridge Institute for Medical
Bern, Switzerland
Research; The Department of Haematology,
A. Ruggeri Eurocord, Hospital Saint-Louis; University of Cambridge; Wellcome Trust–Medical
Hematology Department, Hospital Saint Antoine, Research Council Stem Cell Institute, University of
Paris, France Cambridge, Cambridge, United Kingdom

S. Samarasinghe Department of Haematology, N.S. Young National Institutes of Health, National


Great Hormond Street Hospital, London, United Heart, Lung, and Blood Institute, Bethesda, MD,
Kingdom United States

 
Introduction

From the first descriptions by Ehrlich over a haploidentical and cord blood transplants, and
century ago to where we are today, the under- in supportive care of hematopoietic stem cell
standing of the pathogenesis and biology and transplantation have lead not only to the expan-
of the diseases that are encompassed under sion of the potential donor pool, but also to re-
the umbrella of the “bone marrow failure syn- markable improvements in the outcome of he-
dromes” have witnessed tremendous progress. matopoietic stem cell transplantation for bone
This has been coupled with significant improve- marrow failure from related and unrelated stem
ments in the clinical care and outcomes of both cell sources with excellent rates of cure, par-
congenital and acquired forms of bone marrow ticularly in the younger patients, when a fully
failure. matched unrelated donor is used.
When considering acquired bone marrow Telomere shortening and telomeropathies
failure, in the last 4 decades, aplastic anemia have been increasingly recognized as contrib-
has progressed from a disease that was almost uting factors for bone marrow failure and stem
universally fatal to one in which the majority cell exhaustion in acquired states and causative
of patients can now expect to be cured and lead factors for the increasing number of entities of
normal lives. We have gained noteworthy in- congenital bone marrow failure.
sights in understanding the immune mechanism Several new treatment modalities have
of hematopoietic stem cell destruction, stem cell emerged during the last decade, most notable
niche, and T-cell dysregulation. This knowledge of which is the introduction of thrombopoietin
has placed immunosuppressive therapy at the mimetic agents. Possible interventions to rescue
center stage in the treatment of aplastic anemia, telomere length are being actively investigated.
and has facilitated the initiation of clinical trials Given the exciting progress in the field, this
for the use of new potential therapies to treat ac- book is a timely educational initiative produced
quired aplastic anemia. jointly by the Working Party of Severe Aplas-
Substantial progress in the knowledge about tic Anemia of the European Society for Blood
the role of somatic mutations and their relation and Marrow Transplantation and the European
to clonal hematopoiesis in acquired bone mar- School of Haematology.
row failure has been facilitated by the use of the The contents are a collection of up-to-date
next generation sequencing technologies, and contributions from world-renowned experts in
has allowed us to understand their relationship the field of congenital and acquired bone mar-
with myelodysplastic syndromes, paroxysmal row failure. The chapters in this book provide
nocturnal hemoglobinuria, and the risk of dis- extensive coverage for all aspects of congenital
ease progression. and acquired bone marrow failure, including bi-
Advances in HLA-typing technology, prog- ology, pathology, and treatments involving he-
ress in the field of alternate donor availability, matopoietic stem cell transplantation.

xi
xii Introduction

We hope this book will be an important re- With the endorsement of the European Soci-
source for scientists, clinicians, nurses, and all ety for Blood and Marrow Transplantation and
other health-related professionals involved in the European School of Haematology
research and patient care with the bone marrow
failure syndromes. Mahmoud Aljurf, Eliane Gluckman, Carlo Dufour

 
C H A P T E R

1
Epidemiology of Acquired Bone
Marrow Failure
F. Grimaldi*, S. Gandhi**, A.M. Risitano*
*Hematology, Department of Clinical Medicine and Surgery, Federico II University,
Naples, Italy; **Department of Haematological Medicine, King’s College Hospital/
King’s College London, London, United Kingdom

INTRODUCTION studies to gather data on the epidemiology of


AA. Based on the two epidemiological studies
Paul Ehrlich in 1888 gave the first seminal carried out in Europe and Asia that used the
description of aplastic anemia (AA) in a preg- same methodology, the incidence of the disease
nant woman, where the normal hemopoietic is two- to threefold higher in Asia than in the
tissue was replaced by a fatty marrow and West [2–12]. This variability in incidence rates
empty spaces, the “hypocellular” marrow that may reflect differences in exposure to environ-
resulted in pancytopenia. Idiopathic AA is mental factors including viruses, drugs and
a rare form of acquired bone marrow failure, chemicals, genetic background, diagnostic cri-
where improved supportive care, early insti- teria, and study designs.
tution of immunosuppressive treatment, and In the following sections, a short review of
hematopoietic stem cell transplantation have studies performed and available in literature, in-
led to better treatment outcomes [1]. Several cluding case series and reports to determine the
retrospective studies from Europe, United epidemiology and demographics of AA across
States, South America, and Asia suggest that different centers of the world is presented. Dis-
the incidence is 0.6–6.1 cases per million popu- cussion is restricted to acquired cases of AA, and
lation. In addition, the incidence of AA shows hence the inherited bone marrow failure syn-
geographical variability, with lower rates re- dromes, the inevitable cases of marrow aplasia
ported in Europe, North America, and Brazil that follow intentional chemoirradiation treat-
and higher rates in Asia. However, the rarity of ments and cytopenias of nutritional deficiencies
disease means there have been few prospective or other causes have been excluded.

Congenital and Acquired Bone Marrow Failure


http://dx.doi.org/10.1016/B978-0-12-804152-9.00001-4
1 Copyright © 2017 Elsevier Inc. All rights reserved.
2 1. Epidemiology of Acquired Bone Marrow Failure

INCIDENCE OF AA IN DIFFERENT South Asia, in this study were found to have an


GEOGRAPHICAL REGIONS incidence even higher than their counterparts
AND RACE from East Asia, at 7.3 per million [13]. This study
suggested that Asian children have an increased
The annual incidence of AA varies from 0.6 incidence of AA, possibly as a result of a genetic
to 6 per million populations per annum across predisposition. Indeed, in a hospital based case
centers in different continents. Most findings control study from Lucknow (India), the annual
are from retrospective studies, and even retro- incidence of childhood AA was determined to be
spective reviews of death registries. However, 6.8 per million [14].
this incidence masks the variability that is seen Benzene has been found to be toxic to the
across continents and different ethnic groups; hemopoietic progenitor cells. In large collab-
for example, reports from the Barcelona group orative studies between the National Cancer
(2008), which was a detailed prospective study Institute and American and Chinese institutions
by Montané et al. [2] had an incidence of 2.34 per [15], hematologic susceptibility to benzene has
million population. This incidence rate is simi- been correlated to nucleotide polymorphism
lar to the 2.0 per million reported by the Inter- in key drug metabolic patways [16]. Thus, he-
national Agranulocytosis and Aplastic Anemia matotoxicity from exposure to benzene may be
Study (IAAAS) [3], which was conducted in Eu- particularly evident among genetically suscep-
rope and Israel from 1980 to 1984, and to rates re- tible populations, and it is plausible that the
ported in smaller national studies in Europe that increased incidence of AA seen in Asia may be
included United Kingdom [4], France [5], Scan- related not only to increased exposure but also
dinavia [6], and in South Americas and Brazil [7]. to key polymorphism in genes regulating the
The incidence was accurately determined to be 4 metabolism and cytokine expression.
cases per million population in Bangkok [8], but Similarly, although AA incidence reported
based on prospective studies, it may actually be in a multicenter Latin American study remains
closer to 5.6 cases per million population in the very low at 1.6 per million; this study corrobo-
rural areas of Thailand (Khonkaen region) [9]. rated the association between risk of exposure to
In the prospective Chinese Epidemiologic Study benzene, chloramphenicol, and also azithromy-
Group of Leukemia and Aplastic Anemia survey, cin and predisposition to AA in this area [17,18].
7.4 per million was reported as a national inci-
dence, which clearly is much on the higher side,
but may have been overestimated, as stringent AGE AND GENDER RELATED
criteria for the diagnosis of AA, as bone marrow DEMOGRAPHICS OF AA
study, were not strictly applied [10]. Increased
incidence in Eastern countries may be related to In nearly all modern studies of AA, the sex
environmental factors, such as increased expo- ratio has been close to 1:1, which is unusual for
sure to toxic chemicals and pesticides on agricul- immune-mediated diseases [2,19]. An exception
tural farms, practiced in the Far East and South to this has been a study from the Sabah prov-
Asia. However, the incidence of AA in children ince in Malaysia [12], where an unusually high
of immigrants from the East Asia in a pediat- male to female ratio was noted at 3:4. Similarly
ric population from the ages 0 to 14 years, in a a male preponderance was also noted in stud-
study from British Columbia (Canada) was sig- ies from Thailand [9], India [20], and Pakistan
nificantly higher at 6.9 per million, as compared [21]. This may reflect the underreporting of cases
to children of white/mixed ethnic descent at 1.7 of AA among females and access to adequate
per million [11]. Children of immigrants from healthcare services in Asia. However, it remains

 
AA and association with toxins/drugs 3
unclear why a female preponderance is not seen tries and Europe, where recently an incidence of
in a quintessential autoimmune disorder, such as 5.4%, stable across the years, has been reported
AA among studies in Europe and United States. in a large registry study from EBMT [25].
In all the largest studies available, including a Cases of AA associated with HAV, HBV, HGV,
series of 300 patients reported by Clinical Center parvovirus B19 [26–29], Epstein–Barr virus
at NIH by Young et al. [22], the Barcelona report (EBV) [30], transfusion-transmitted virus (TTV)
[2] and epidemiologic study from Thailand [9], [31] or echovirus [32] infections have been re-
2 patient age peaks of incidence are constantly ported. In a German–Austrian study of 213 pe-
observed, one among young adults and the sec- diatric cases aged 17 years or less of AA, 80% of
ond in the elderly. This characteristic biphasic cases were idiopathic, 9% followed posthepatitis
distribution shows two peaks, one from 10 to AA, 7% were following viral infections, and 4%
25 years and the second above 60 years. Within were associated with drugs/toxins [33].
the younger age group, a small peak in the in- Parvovirus B19 is the causative agent for fifth
cidence is observed in childhood, probably due disease, usually in the immunocompetent host.
to overlap with inherited marrow-failure syn- However, transient aplastic crises have been re-
dromes featured by a less penetrating pheno- ported in chronic hemolytic anemias, such as
type, where classical physicalanomalies of the sickle cell disease owing to reticulocytopenia,
inherited marrow-failure syndromes are not ob- and cases of severe AA have also been reported
vious. On the other hand, the second incidence in normal individuals during an acute episode
peak of AA seen above 60 years may reflect the of infection [34]. The actual incidence of acute
smaller pool of hematopoietic stem cell reserve parvovirus B19 infection at presentation in AA
left, with age-related telomeric attrition and its is not known, but in single case series of 27 pa-
capacity to maintain normal hemopoiesis, in the tients with AA from India, parvovirus B19 IgM
face of an immune insult against the hematopoi- and viral DNA was detected in nearly 40% of the
etic precursor cells [23]. cases [35]. This maybe an important etiological
factor, especially in immunocompromised host
or patients with chronic hemolytic anemias.
POSTHEPATITIS AA AND AA Very recently retrospective observations
OCCURRING AFTER VIRAL about eight AA cases occurring during HIV in-
INFECTIONS fection have been reported [36]. Even if AA ap-
pears to be a late rare complication in HIV pa-
Posthepatitis AA is a stereotypical syndrome, tients, report from Pagliuca et al. [36] highlights
where pancytopenia often presents 2–3 months that immunosuppressive therapy is a feasible
after an acute attack of seronegative severe but strategy in AA patients and that better outcome
self-limited liver inflammation. This distinct is observed in patients eligible to transplant,
variant has been commonly seen in 5–10% of while death for infection remain the principle
“classical” AA cases, typically occurring in ado- cause of mortality in undertreated patients.
lescent boys and young men [24]. Severe imbal-
ance of the T-cell immune system as seen with
“classical” AA, human leukocyte antigen (HLA) AA AND ASSOCIATION WITH
association, and effective response to immuno- TOXINS/DRUGS
suppressive therapy strongly suggest an im-
mune-mediated mechanism. As for other form There is no discernible difference in the de-
of AA, higher incidence is noted in East Asia mographics or clinical behavior, including re-
(4–10%), when compared to the Western coun- sponse to immunosuppressive therapy, between

 
4 1. Epidemiology of Acquired Bone Marrow Failure

patients classified as having “drug or toxin in- clined to the point that it has not been reported
duced” versus “idiopathic” AA [37]. as significant risk factor in any recent systematic
Benzene is the most widely studied and im- epidemiologic study of AA in Western countries.
plicated amongst toxins causing AA. The rela- Even in Thailand where the need for such effec-
tionship was initially brought to light by a case tive and inexpensive antibiotic is substantial, and
where a series of workers exposed through their usage is reported 100 times greater than in the
specific occupations [15] and has since been West, association with AA is infrequent, prob-
detected in some, but not all population-based ably due to lower-doses prescription [9].
case-control studies. An association has been In the IAAAS study [3] approximately 25%
seen in some case-control studies but even when of the identified AA cases were related to
present, the proportion of cases that can be at- drug use. Major drug associations were with
tributed to this chemical has been small. Studies gold salt (relative risk, RR of 29), antithyroid
on American workers earlier in this century sug- drugs (RR of 11), and nonsteroidal antiinflam-
gested that the risk of AA was about 3% in men matory agents (RR of 8.2 for Indomethacin).
exposed to concentration higher than 300 ppm, Similarly in 235 patients with AA prospec-
and in the more recent IAAAS study [3], benzene tively followed by the Barcelona group [2],
was accounted for about 1–3% of AA recorded 67 cases (28.5%) had a history of exposure to
cases. Similarly in Thailand population, benzene drugs or toxic agents that have been associated
carried a relative risk of 3.5 but accounted for an with AA, sometime in the preceding 6 months.
etiologic fraction of only 1% [9]. Forty nine (20.8%) cases had been exposed to
Pesticides have been associated with AA in a the following drugs (Table 1.1). In addition, 21
large number of medical records. In the Indian (8.9%) cases had been exposed to toxic agents:
cohort of pediatric AA, although significantly insecticides (n = 8), benzene (n = 6), and other
higher blood levels of organochlorine com- solvents (n = 10).
pounds were detected suggesting an associa-
tion, they were not entirely supported by statis- TABLE 1.1 Exposures to Drugs Reported to be
tical methods [14]. Anecdotal case reports of AA Associated With Aplastic Anemia in the
following use and pesticides, such as dichloro- 2–6 Months Prior to Hospital Admission
Among 235 Cases (Montané et al. [2])
diethyly-trichlorthane (DDT), chlordane or lin-
dane, or following exposure to organic solvents, Drug N (%)
such as toluene and other molecules resembling
Allopurinol 9 (3.8)
benzene, or containing benzene ring, again point
merely to an association. Unfortunately, system- Indomethacin 9 (3.8)
atic population case-control studies correlating Gold salts 9 (3.8)
level and duration of exposure of these identi- Sulfonamides 9 (3.8)
fied toxins and AA onset are lacking. Carbamazepine 5 (2.1)
Initially suggested by accumulation of case re- Ticlopidine 4 (1.7)
ports, specific drug associations have been estab-
Chloramphenicol 3 (1.2)
lished in different population based study and
have changed across time, mainly due to chang- Oxyphenbutazone 3 (1.2)
es in drugs diffusion and utilization. Chloram- Phenylbutazone 3 (1.2)
phenicol, for example, that gained notoriety for Penicillamine 3 (1.2)
its prominent association with AA in the 1950s Clopidogrel 2 (0.8)
and for decades was considered the common-
Methimazole 2 (0.8)
est cause of the disease, has progressively de-

 
AA and association with HLA genes 5
Finally, incidence of drug-associated AA ap- Nimer et al. [39]. Subsequently, positive asso-
pears to be lower in East Asia [8,9]. ciation between HLA-DR2 (precisely DRB1*15
allele) and AA was confirmed in Chinese [40],
Japanese [41], Turkish [42], Pakistani [43], and
AA AND ASSOCIATION Malaysian [44] series. These data depict a clear
WITH HLA GENES role for HLA-DR2 gene as risk factor for AA,
and different distribution of this gene across
The (HLA) system is a crucial group of genes human population may probably account for
responsible for starting and regulating immune different incidence seen for AA inspecific geo-
response. Since antigen presentation and T-cell graphical areas and ethnic groups. Interestingly,
activation through HLA may represent the early when Maciejewski et al. [45] analyzed distribu-
step that precede global hematopoietic stem cell tion of HLA-DR2 (mainly DRB1*15 allele) across
destruction in AA, HLA polymorphism may disease subgroups (i.e., AA, hemolytic PNH
contribute to pathogenesis of the disease by: (1) and AA/PNH), they found an increased fre-
increasing or decreasing susceptibility to AA, quency in the ones where bone marrow failure
particularly for specific ethnic or age groups, was associated with a PNH clone. Other DR2 al-
(2) facilitating activity of drugs or viral antigen leles have been correlated with AA; in a single
to break immune tolerance and starting the dis- case report, Nakao et al. [46] showed a specific
ease, (3) influencing response to immunosup- T-cell cytotoxic response associated with the
pressive therapy. DRB1*0405 allele. The DRB1*07 allele has been
As for other autoimmune disease, a large reported in a single cohort of Iranian subjects
number of studies demonstrated an association [47], and DRB1*0901 in Chinese children [48].
between polymorphism of HLA class II genes Finally, evidences available correlate AA even
and AA susceptibility (Table 1.2, a). with other HLA-II class genes, such as Dpw3
An increased frequency of HLA-DR2 was [49] and DR4 [22].
first described in several studies for European Less data are available for HLA class I genes.
and American Caucasian patients [38], and Early reports suggested a correlation with HLA-
finally confirmed in a multiethnic cohort by A2 gene [50,51] and with HLA-B7 and HLA-B14

TABLE 1.2 Summary of AA and Incidence of Specific HLA Genes or Alleles


a. HLA and b. HLA and
increased risk increased risk c. HLA and d. HLA and e. HLA and drug/ f. HLA and
of AA (Class II) of AA (Class I) reduced risk of AA onset age of AA viral related AA AA outcome

HLA-DRB1*1501 HLA-A2 HLA-DRB1*13 HLA-DRB*09 Haplotype: -B38, HLA-DR2


-DR4, -DQ3
HLA-DRB1*0405 HLA-A*0206 HLA-DRB1*03:01 HLA-A26 Haplotype: HLA-DRB1*1501
-DRB1*0402,
-DQB1*0302
HLA-DRB1*07 HLA-B7 HLA-DRB1*11:01 HLA-B14 Haplotype: HLA-DR4-Ala74
-DRB1*1601,
-DQB1*0502
HLA-DRB1*0901 HLA-B14 HLA-DRB1*51:01 HLA-B48 HLA-DRB1*08
HLA-Dpw3 HLA-B*4002 HLA-DRB1*03
HLA-DR4 HLA-DRB1*13:02

 
6 1. Epidemiology of Acquired Bone Marrow Failure

genes at least in European patients [52,53]. More- tentially different HLA landscape in children
over, association between HLA-B14 alleles, HLA- with AA, even if further investigation involving
Cw7, and AA has been confirmed by Maciejewski a higher number of patient are needed to specifi-
et al. [45] in a multiethnic cohort of 212 American cally address this question (Table 1.2, c).
individuals. Finally Shichishima et al. [54] report- Association between agranulocytosis and
ed higher incidence of HLA-B*4002 and HLA- HLA genes has been reported for drugs expo-
A*0206 in 78 Japanese AA patients, suggesting sure in specific ethnic groups, suggesting that
a potential role of HLA class I genes even in Far certain alleles may facilitate the initiation of AA
East countries. through direct presentation of drug-derived an-
HLA genes revealing a protective role in devel- tigens to T-cells [37]. HLA-class II haplotypes
oping AA have been identified too (Table 1.2, b). have been reported to be associated with clozap-
Even if data refer mainly to small series of pa- ine-induced agranulocytosis in Askenazi Jewish
tients, and may reflect natural polymorphism [60,61] individuals, and association between
in HLA system, again a key role for HLA class HLA-DRB1*08 alleles and thionamide-induced
II genes and its alleles is suggested. HLA- AA has been described for Japanese patients
DRB1*13 appeared to be protective in a cohort [62]. A similar mechanism of disease can be sug-
of patients of Turkish origin [55], as well as gested for posthepatitis AA too, where associa-
HLA-DRB1*03:01, HLA-DRB1*11:01, and HLA- tion between HLA class I, specifically HLA-B8,
B1*51:01 alleles appear to be protective in co- and hematological disease have been document-
hort of Chinese children [48]. In a small cohort ed [24] (Table 1.2, d).
of Pakistani patients [43], HLA-DRB1*03 had an Finally, some groups have shown that re-
higher frequency in healthy control, suggesting sponse to immunosuppressive therapy can be
a putative protective role, and a similar observa- related to specific HLA genes. HLA-DRB1*1501
tion is available for allele DRB1*1302 in Korean is the most clearly allele associated with a better
population [56]. response to Cyclosporine [41], and the presence
Even if AA has a typical bimodal age of inci- of HLA-DR2 and a PNH clone independently
dence, majority of studies did not look specifi- predict response to therapy in patients under
cally into HLA frequency and age differences, immunosuppression [45]. On the other hand
mostly due to their retrospective nature and HLA-DRB1*1502 allele [63], that represents a dif-
rarity of disease. Therefore, a good assump- ferent allele variant of HLA-DR2, doesn’t seem
tion could be that data on HLA allele frequency to have same influence on treatment, even if its
could be inferred to the pediatric setting. How- incidence is increased among older Japanese pa-
ever Fuhrer et al. [57] in a recent study involv- tients with AA. High-resolution genotyping of
ing 181 Caucasian children observed a positive HLA-DRB1 showed that the HLA-DRB1*04 al-
association between HLA-A26 and HLA-B14 lele coding for alanine position 74 (HLA-DR4-
alleles, but not a higher incidence of HLA-DR2. Ala74) [64] predispose to AA independently
Similarly Chen et al. [48] in a retrospective sur- from HLA-DRB1*15 and that HLA-DRB1*04 al-
vey conducted on 80 Chinese children showed leles are associated with worse response to cy-
an higher than expected incidence of class I closporine and poorest prognosis (Table 1.2, e).
HLA-B48 alleles and class II HLA-DRB*09 al- Association between AA and HLA remains
lele. Finally, a frequency of HLA-DR2 B15 allele an intriguing field of interest. However studies
not different from normal population has been with larger patients populations from different
reported by Kook et al. [58] in North Korean AA ethnic backgrounds are needed to better address
children and by Yoshida et al. [59] in Japanese complex relations between HLA, environmental
AA children. These limited data suggest a po- factors, ethnicity, and AA incidence.

 
AA during pregnancy 7

AA AND AUTOIMMUNE among AA patients. Stalder et al. in a single cen-


DISORDERS ter series [71] of 253 individuals showed that
5.3% of patients with AA had a previous diagno-
Although AA is idiopathic in most cases, as- sis of AIDS, and 4.5% developed an AIDS after
sociations with other autoimmune disease (AID) AA was diagnosed. Similarly 4% of patients re-
have been shown in numerous single-case re- corded in EBMT database [72] for AA have been
ports. In rheumatic diseases, such as systemic found diagnosed with a previous AIDS in a ret-
lupus erythematosus (SLE) and rheumatoid ar- rospective analysis. Most frequently diagnosed
thritis (RA), there is a recognized associations AIDS were autoimmune gastritis and thyroid-
with AA [65], with a true incidence not known, itis in Stalder series, and RA in EBMT analysis,
but presumed to be very low. respectively. Interestingly, in both studies pa-
Peripheral autoimmune cytopenias are com- tient’s age at diagnosis of AA seems to be higher
monly seen in SLE, and they are listed among (>50 years) than normal. Impact of immunosup-
diagnostic criteria for the disease in the revised pressive treatment for AA on concomitant AIDS,
American College of Rheumatology classifica- and effects on its natural history, remains at the
tion. Similarly, anemia of chronic disease, and moment a controversial topic.
cytopenias in relation to the use of cytotoxic
drugs are commonly seen in SLE patients [66].
Less frequently, case reports and small series AA DURING PREGNANCY
documented bone marrow abnormalities consis-
tent with AA, suggesting that in rare cases bone There are no prospective studies about the
marrow may also be a target organ of the disease, incidence of AA during pregnancy. Curiously
showing a full picture of acquired bone marrow AA was first described in a pregnant women
failure [67]. Very recently Chalayer et al. [68] re- by Ehrlich in 1888. Since then, there have been a
ported a MEDLINE methanalysis of 25 patients few published reports of AA during pregnancy.
with SLE and AA. In 12 of these 25 patients, di- Tichelli et al. [73] described a case series of 36
agnosis of AA was associated with long story of pregnancies in women with AA. Their study
SLE, extensive disease, and drug exposure, sug- showed that successful pregnancy with normal
gesting that drug-induced AA may represent a outcome is possible in women with AA who
consistent type of AA in these patients. have been previously treated with immuno-
Similarly the association of RA with AA is suppression. Nineteen percent of women were
better recognized because of the use of thera- found to have relapse of AA and a further 14%
peutic agents in RA, such as Methotrexate, gold needed transfusion support at the time of de-
salts, penicillamine, and Eternacept [69] causing livery. Complications appear to be more likely
AA, likely drug induced. in patients with low platelet counts and par-
Correlation with immune disorders is also oxysmal nocturnal hemoglobinuria-associated
highlighted by the strong link observed with AA. Similarly, Choudhry et al. [74] described
eosinophilic fasciitis (Shulman disease), a rare 10 cases of AA and concomitant pregnancies,
sclerodermiform syndrome that, in most cases, reporting successful delivery in 10 of 11 cases,
resolves spontaneously or after corticosteroid with adverse outcome related to fatal bleeding
therapy, and that is frequently reported in asso- in only 2 patients.
ciation with hematological diseases, especially Although pregnancy remains an immuno-
AA [70]. modulatory state, with an higher incidence of
Two large different studies have specifically autoimmune disease reported, causative rela-
looked into incidence of autoimmune conditions tionship with AA remains controversial.

 
8 1. Epidemiology of Acquired Bone Marrow Failure

AA POSTVACCINATION Ready availability of nucleotide variants


from the human genome project have defined
There have been three case reports of AA, new polymorphisms that are definitely patho-
following hepatitis B vaccination [75]. System- geneic and disease causing as opposed to vari-
atic case control studies for the incidence of AA ants that are disease predisposing and require
following vaccination are not known. Patients the interplay of other external factors for disease
with AA who have been treated with immu- causation. Furthermore, genetic tests have also
nosuppressive treatment, should better avoid defined how environmental influences, such as
vaccinations, if possible, including influenza, benzene play a role in the pathogenesis of AA
as there remains a theoretical risk of disease re- because of nucleotide polymorphisms that affect
lapse [1]. key drug metabolism pathways or cytokine sig-
naling molecules.
Following this theme, future epidemiological
PROBLEMS WITH studies for AA, are likely to better explain the
EPIDEMIOLOGICAL STUDIES IN association to causality in AA, precise diagno-
AA AND FUTURE STRATEGIES sis and staging prognostic information and also
unravel key molecular pathways for therapeutic
AA remains a rare disease with an annual inci- exploitation.
dence between 0.6 and 6 per million per annum. It
is innately difficult to conduct a population based References
study in a rare disease, such as AA. Reporting of
[1] Killick S, Brown N, Cavenagh J, et al. Guidelines for the
cases with AA is likely to be only from areas and
diagnosis and management of adult aplastic anemia. Br
centers that have a high coverage of health servic- J Haematol 2016;172:187–207.
es. It’s likely that there had been underreporting [2] Montané E, Ibanez L, Vidal X, et al. Epidemiology of
of cases with AA in regions with poor access to aplastic anaemia: a prospective multicenter study. Hae-
tertiary health care services or facilities for diag- matologica 2008;93:518–23.
[3] Kaufman DW, Kelly JP, Levy M, et al. The drug etiol-
nostic tests, including special diagnostic tests for
ogy of agranulocytosis and aplastic anemia. New York:
genomic and molecular analysis. Oxford University Press; 1991.
Drug recording and reporting to the medi- [4] Cartwright RA, McKinney PA, Williams L, et al. Aplas-
cines regulatory body for association studies tic anaemia incidence in parts of the United Kingdom
is not uniformly practiced, leading to under in 1985. Leuk Res 1988;12:459–63.
[5] Mary JY, Baumelou M, Guiguet M. The French Coop-
or over estimation of association and causality
erative Group for Epidemiological Study of Aplastic
(Chloramphenicol remains a case in point for an Anemia. Epidemiology of aplastic anemia in France: a
example of the latter). prospective multi-centric study. Blood 1990;75:1646–53.
The distinction between acquired and inher- [6] Clausen N, Kreuger A, Salmi T, et al. Severe aplastic
ited disease may present a clinical challenge, es- anaemia in the Nordic countries: a population based
study of incidence, presentation, course, and outcome.
pecially among pediatric cases.
Arch Dis Child 1996;74:319–22.
Multicenter clinical trials for newer thera- [7] Maluf EM, Pasquini R, Eluf JN, et al. Aplastic anemia
peutic agents or treatment strategies in AA may in Brazil: incidence and risk factors. Am J Hematol
provide on different countries a uniform panel 2002;71:268–74.
of recording data, including family history for [8] Issaragrisil S, Sriratanasatavorn C, Piankijagum A, et al.
Incidence of aplastic anemia in Bangkok. The Aplastic
inherited bone marrow failure syndromes, and
Anemia Study Group. Blood 1991;77(10):2166–8.
special tests, including T-cell subset repertoire [9] Issaragrisil S, Kaufman DW, Anderson T, et al. The
analysis and next-generation technique molecu- epidemiology of aplastic anemia in Thailand. Blood
lar analysis. 2006;107:1299–307.

 
REFERENCES 9
[10] Yang C, Zhang X. Incidence survey of aplastic anemia [26] Gonzalez-Casas R, Garcia-Buey L, Jones EA, et al. Sys-
in China. Chin Med Sci J 1991;6:203–7. tematic review: hepatitis-associated aplastic anaemia—
[11] Szklo M, Sesenbrenner L, Markowitz J, et al. Incidence a syndrome associated with abnormal immunological
of aplastic anemia in Metropolitan Baltimore. A popu- function. Aliment Pharmacol Ther 2009;30:436–43.
lation based study. Blood 1985;66:115–9. [27] Adachi Y, Yasui H, Yuasa H, Ishi Y, Imai K, Kato Y. Hep-
[12] Yong AS, Goh M, Rahman J, et al. Epidemiology of atitis B virus-associated aplastic anemia followed by
aplastic anemia in the state of Sabah, Malaysia. Cell Im- myelodysplastic syndrome. Am J Med 2002;112:330–2.
munol 1996;1284:S75. [28] Byrnes JJ, Banks AT, Piatack M Jr, Kim JP. Hepatitis G-
[13] McCahon E, Tang K, Rogers PC, McBride ML, et al. The associated aplastic anaemia. Lancet 1996;348:472.
impact of Asian descent on the incidence of acquired [29] Pardi DS, Romero Y, Mertz LE, Douglas DD. Hepatitis-
severe aplastic anaemia in children. Br J Haematol associated aplastic anemia and acute parvovirus B19
2003;121(1):170–2. infection: a report of two cases and a review of the lit-
[14] Ahamed M, Anand M, Kumar A, Siddiqui MK. Child- erature. Am J Gastroenterol 1998;93:468–70.
hood aplastic anaemia in Lucknow, India: incidence, [30] Lau YL, Srivastava G, Lee CW, et al. Epstein–Barr virus
organochlorines in the blood and review of case re- associated aplastic anaemia and hepatitis. J Paediatr
ports following exposure to pesticides. Clin Biochem Child Health 1994;30:74–6.
2006;39(7):762–6. [31] Poovorawan Y, Tangkijvanich P, Theamboonlers A,
[15] Qing L, Luoping Z, Guilan L, et al. Hematotoxicity et al. Transfusion transmissible virus (TTV) and its pu-
in workers exposed to low levels of benzene. Science tative role in the etiology of liver disease. Hepatogas-
2004;306(5702):1774–6. troenterology 2001;48:256–60.
[16] Qing L, Luoping Z, Min S, et al. Polymorphisms in cy- [32] Imai T, Itoh S, Okada H, et al. Aplastic anemia follow-
tokine and cellular adhesion molecule genes and sus- ing hepatitis associated with echovirus 3. Pediatr Int
ceptibility to hematotoxicity among workers exposed 2002;44:522–4.
to benzene. Cancer Res 2005;65:9574–81. [33] Führer M, Rampf U, Baumann I, et al. Immunosup-
[17] Maluf E, Hamerschlak N, Cavalcanti AB, et al. In- pressive therapy for aplastic anemia in children: a
cidence and risk factors of aplastic anemia in Latin more severe disease predicts better survival. Blood
American countries: the LATIN case-control study. 2005;106:2102–4.
Haematologica 2009;94(9):1220–6. [34] Young NS, Brown KE. Mechanisms of disease: parvovi-
[18] Young N, Kaufman D. The epidemiology Of acquired rus B19. N Engl J Med 2004;350:586–97.
aplastic anemia. Haematologica 2008;93:489–92. [35] Mishra B, Malhotra P, Ratho RK, et al. Human parvovi-
[19] Heimpel H. Epidemiology and aetiology of aplastic rus B19 in patients with aplastic anemia. Am J Hematol
anaemia. In: Schrezenmeier H, Bacigalupo A, editors. 2005;79:166–7.
Aplastic anaemia: pathophysiology and treatment. [36] Pagliuca S, Gérard L, Kulasekararaj A, et al. Character-
Cambridge, UK: Cambridge University Press; 2000. istics and outcomes of aplastic anemia in HIV patients:
p. 97–116. a brief report from the severe aplastic anemia work-
[20] Mahapatra M, Singh PK, Agarwal M, et al. Epidemiol- ing party of the European Society of Blood and Bone
ogy, clinico-haematological profile and management of Marrow Transplantation. Bone Marrow Transplant
aplastic anaemia: AIIMS experience. J Assoc Physicians 2016;51:313–5.
India 2015;63(3 Suppl):30–5. [37] Young NS, Alter BP, Young NS. . In: Young NS, Alter BP,
[21] Adil SN, Kakepoto GN, Khurshi M. Epidemiological editors. Aplastic anemia, acquired and inherited, drugs
features of aplastic anaemia in Pakistan. J Pak Med As- and chemicals. Philadelphia: W.B. Saunders; 1994.
soc 2001;51:443. p. 100–32.
[22] Young NS. Bone marrow failure syndromes. Philadel- [38] Chapuis B, Von Fliedner VE, Jeannet M, et al. Increased
phia: W.B. Saunders; 2000. frequency of DR2 in patients with aplastic anaemia and
[23] Hao LY, Armanios M, Strong MA, et al. Short telomeres, increased DR sharing in their parents. Br J Haematol.
even in the presence of telomerase, limit tissue renewal 1986;63:51–7.
capacity. Cell 2005;123:1121–31. [39] Nimer SD, Ireland P, Meshkinpour A, et al. An in-
[24] Brown KE, Tisdale J, Barrett AJ, et al. Hepatitis-associ- creased HLA DR2 frequency is seen in aplastic anemia
ated aplastic anemia. N Engl J Med 1997;336:1059–64. patients. Blood 1994;84:923–7.
[25] Locasciulli A, Bacigalupo A, Bruno B. Hepatitis-as- [40] Shao W, Tian D, Congyan L, et al. Aplastic anemia is
sociated aplastic anaemia: epidemiology and treat- associated with HLA-DRB1*1501 in northern Han Chi-
ment results obtained in Europe. A report of The nese. Int J Hematol 2000;71:350–2.
EBMT aplastic anaemia working party. Br J Haematol [41] Nakao S, Takamatsu H, Chuhjo T, et al. Identification
2010;6:890–5. of a specific HLA class II haplotype strongly associated

 
10 1. Epidemiology of Acquired Bone Marrow Failure

with susceptibility to cyclosporine-dependent aplastic [57] Fuhrer M, Durner J, Brunnler G, et al. HLA association
anemia. Blood 1995;84:4257–61. is different in children and adults with severe acquired
[42] Ilhan O, Beksac M, Arslan O, Koc H, et al. HLA-DR aplastic anemia. Pediatr Blood Cancer 2007;48:186–91.
frequency in Turkish aplastic anemia patients and the [58] Kook H, Hwang TJ, Seo JJ, et al. The frequency of
impact of HLA-DR2 positivity in response rate in pa- HLA alleles in Korean children with aplastic anemia
tients receiving immunosuppressive therapy. Blood and the correlation with the response to immunosup-
1995;86:2055. pressive treatment. Korean J Pediatr Haematol Oncol
[43] Rehman S, Saba N, Khalilullah, et al. The frequency 2003;10:177–88.
of HLA class I and II alleles in Pakistani patients with [59] Yoshida N, Yagasaki H, Takahashi Y, et al. Clinical im-
aplastic anemia. Immunol Investig 2009;38:251–4. pact of HLA-DR15, a minor population of paroxysmal
[44] Dhaliwal JS, Wong L, Kamaluddin MA. Susceptibility nocturnal haemoglobinuria-type cells, and an aplastic
to aplastic anemia is associated with HLA-DRB1* 1501 anaemia-associated autoantibody in children with ac-
in an aboriginalpopulation in Sabah, Malaysia. Hum quired aplastic anemia. Br J Haematol 2008;142:427–35.
Immunol 2011;72:889–92. [60] Yunis JJ, Corzo D, Salazar M, et al. HLA associa-
[45] Maciejewski JP, Follmann D, Nakamura R. Increased tions with clozapine-induced agranulocytosis. Blood
frequency of HLA-DR2 in patients with paroxysmal 1995;86:1777.
nocturnal hemoglobinuria and the PNH/aplastic ane- [61] Corzo D, Yunis JJ, Salazar M, et al. The major histo-
mia syndrome. Blood 2001;98:3513–9. compatibility complex region marked by HSP70-1 and
[46] Nakao S, Takami A, Takamatsu H. Isolation of a T-cell HSP70-2 varisnts is associated with clozapine induced
clone showing HLA-DRB1*0405-restricted cytotoxicity agranulocytosis in two different ethnic groups. Blood
for hematopoietic cells in a patient with aplastic ane- 1995;86:3835–40.
mia. Blood 1997;89:3691. [62] Tamai H, Sudo T, Kimura A, et al. Association be-
[47] Yari F, Sobhani M, Vaziri MZ, et al. Association of aplas- tween the DRB1*08032 histocompatibility antigen
tic anemia and Fanconi’s disease with HLA-DRB1 al- and methimazole-induced agranulocytosis in Japa-
leles. Int J Immunogenet 2008;35:453–6. nese patients with Graves disease. Ann Intern Med
[48] Chen C, Lu S, Luo M, et al. Correlations between HLA- 1996;124:490–4.
A, HLA-B and HLA-DRB1 allele polymorphism to ac- [63] Sugimori C, Yamazaki H, Feng X, et al. Roles of
quired aplastic anemia. Acta Haematol 2012;128:23–7. DRB1*1501 and DRB1*1502 in the pathogenesis of
[49] Odum N, Platz P, Morling N, et al. Increased frequency aplastic anemia. Exp Hematol 2007;35:13–20.
of HLA-DPw3 in severe aplastic anemia. Tissue Anti- [64] Kapustin SI, Popova TI, Lyshchov AA, et al. HLA-
gens 1987;29:184–5. DR4-Ala74 beta is associated with high risk and poor
[50] Albert E, et al. HLA antigens and haplotypes in outcome of severe aplastic anemia. Ann Haematol
200 patients with aplastic anemia. Transplantation 2001;80:66–71.
1976;22:528–31. [65] Bacigalupo A. Aetiology of severe aplastic anaemia and
[51] Dausset J, Gluckman E, et al. Excess of HLA-A2 and outcome after allogeneic bone marrow transplantation
HLA-A2 homozygotes in patients with aplastic anemia or immunosuppression. Eur J Haematol 1996;57(Sup-
Fanconi’s anemias. Nouv Rev Fr Hematol Blood Cells pl):16–9.
1977;18:315–24. [66] Bhatt AS, Berliner N. Hematologic Manifestations of
[52] Gluckman E. HLA markers in patients suffering from SLE. In: Schur P, Massarotti E, editors. Lupus erythe-
aplastic anaemia. Haematologia 1981;14:165–72. matosus: clinical evaluation and treatment. New York:
[53] D’Amaro J, et al. HLA associations in Italian and non- Springer; 2012. p. 127–40.
Italian Caucasoid aplastic anemia patients. Tissue Anti- [67] Alishiri GH, Saburi A, Bayat N, et al. The initial pre-
gens 1983;21:184–91. sentation of systemic lupus erythematosis with aplastic
[54] Shichishima T, Noji H, Ikeda K, et al. The frequency of anemia successfully treated with rituximab. Clin Rheu-
HLA class I alleles in Japanese patients with bone mar- matol 2012;31(2):381.
row failure. Haematologica 2006;91:857. [68] Chalayer É, Ffrench M, Cathébras P. Aplastic anemia as
[55] Oguz FS, Yalman N, Diler S, et al. HLA-DR15 and a feature of systemic lupus erythematosus: a case report
pediatric aplastic anemia patients. Haematologica and literature review. Rheumatol Int 2015;35(6):1073–
2002;87:772–4. 82.
[56] Song EY, Park S, Lee DS, et al. Association of human [69] Kozak N, Friedman J, Schattner A. Etanercept-asso-
leukocyte antigen-DRB1 alleles with disease suscep- ciated transient bone marrow aplasia: a review of
tibility and severity of aplastic anemia in Korean pa- the literature and pathogenetic mechanisms. Drugs
tients. Hum Immunol 2008;69:354–9. 2014;14(2):155–8.

 
REFERENCES 11
[70] De Masson. Severe aplastic anemia associated with eo- [73] Tichelli A, Socié G, Marsh J, et al. Outcome of preg-
sinophilic fasciitis: report of 4 cases and review of the nancy and disease course among women with aplastic
literature. Medicine (Baltimore) 2013;2:69–81. anemia treated with immunosuppression. Ann Intern
[71] Stalder MP, Rovó A, Halter J, et al. Aplastic anemia Med 2002;137(3):164.
and concomitant autoimmune disease. Ann Hematol [74] Choudhry VP, Gupta S, Gupta M, et al. Pregnancy asso-
2009;88:659–65. ciated aplastic anemia—a series of 10 cases with review
[72] Cesaro S, Marsh JC, Tridelli G, et al. Retrospective of the literature. Haematology 2002;7:233–8.
survey on the prevalence and outcome of prior au- [75] Shah C, Lemke S, Singh V, et al. Case reports of aplas-
toimmune diseases in patients with aplastic anemia tic anemia after vaccine administration. Am J Hematol
reported to the registry of the European group for 2004;77(2):204.
blood and marrow transplantation. Acta Haematol
2010;124:19–22.

 
C H A P T E R

2
Pathophysiology of Acquired Bone
Marrow Failure
K. Hosokawa*, P. Scheinberg**, N.S. Young*
*National Institutes of Health, National Heart, Lung, and Blood Institute, Bethesda,
MD, United States; **Oncology Center, Hospital São José National Institutes of
Health, National Heart, Lung, and Blood Institute, Bethesda, MD, United States

INTRODUCTION: EVIDENCE AND therapy (IST) alone is strong evidence of an im-


INFERENCES FROM THE CLINIC mune mechanism in most patients with AA: 60–
70% respond to one course of horse hATG/cyclo-
Acquired aplastic anemia (AA) is the proto- sporine (CsA) and an additional 30% of primary
typical bone marrow (BM) failure syndrome. nonresponders will respond to a second course
AA is characterized by pancytopenia of pe- of IST. Similarly, patients’ blood counts often are
ripheral blood and BM hypoplasia. Low blood dependent on continued administration of CsA,
counts and “empty” marrow pathology implies as would be expected of a T-cell driven disease.
the absence of stem and progenitor cells, which Eltrombopag, a thrombopoietin (TPO) mimetic,
is consistent with, the success of BM transplan- has activity in refractory SAA as single agent
tation (BMT), in which replacement of hemato- and in increasing the response rate and com-
poietic stem cells (HSCs) (and immune cells) is pleteness of the response when combined with
adequate to cure disease. There is little evidence IST as first therapy. These results suggest that
that AA is mediated by BM stromal cells. If stem there are residual HSCs that can repopulate BM,
cell absence were the only defect, twin donor or even if they are not detectable. Recent genomic
syngeneic transplants should be successful with data applying whole exome sequencing in AA
infusion of BM, but a large proportion suffer show that hematopoiesis can be sustained from
graft failure; that conditioning eliminates graft a single or very few HSC clones. Clonal hemato-
failure suggests an immune pathophysiology poiesis has also been inferred from the frequent
[1]. Immunosuppressive regimens were devel- and unique association of paroxysmal nocturnal
oped in the context of graft failure and now are hemoglobinuria (PNH), the origin of which is a
widely employed when stem- cell transplant is somatic mutation in an X-linked gene, with ac-
not feasible. The efficacy of immunosuppressive quired AA.

Congenital and Acquired Bone Marrow Failure


http://dx.doi.org/10.1016/B978-0-12-804152-9.00002-6
13 Copyright © 2017 Elsevier Inc. All rights reserved.
14 2. Pathophysiology of Acquired Bone Marrow Failure

In vitro experiments with patients’ cells are fected patients at presentation. There are few or
consistent with clinical observations in support- no CD34+ cells on flow cytometry [8].
ing an immune mechanism of AA. The presence Recent HSC fate-mapping analyses in mice
of PNH or acquired copy number-neutral loss of have suggested that progenitors and not HSCs
heterozygosity of the 6p arms (6pLOH) clones are fundamental for hematopoiesis under ho-
support clonal escape from immune-mediated meostatic conditions [11,12]. Physiological
BM destruction. Immune-mediated destruction studies to experimentally test this hypothesis
of BM can be modeled in animals and animal ex- are not possible in normal human subjects.
periments employing myelotoxic drugs or with However, certain disease states may indicate
myeloablative transplants show that very limit- the consequences of HSC loss on progenitors
ed numbers of HSCs can support hematopoiesis and the role of HSCs in human blood produc-
for very prolonged time periods. tion under nontransplant conditions. Using a
Clonal evolution, the development of myelo- flow cytometric gating scheme to define MPPs
dysplastic syndrome (MDS) and acute myeloid (CD34+CD38–Thy1–CD45RA–CD49f–), CMPs
leukemia (AML) in a patient with typical AA, of- (CD34+CD38+CD10–FLT3+CD45RA–), and MEPs
ten after successful IST, demonstrates genomic (CD34+CD38+CD10–FLT3- CD45RA–), the pro-
instability in the setting of immune or inflamma- genitor hierarchy was examined in a few cases
tory disease environment (and occurs in other of AA [13]. Consistent with previous reports,
immune diseases, such as inflammatory bowel the proportion of CD34+ cells within the over-
disease and chronic hepatitis). Short telomeres all mononuclear cell pool was much lower
appear to predispose to genomic instability, in AA compared with normal BM [6,9]. The
with tissue culture and animal model experi- CD34+CD38– stem-cell compartment in AA was
ments providing a mechanism of chromosome more significantly depleted compared with the
derangement. In some cases of clonal evolution, CD34+CD38+ progenitor compartment. HSCs
there is evidence of origin from a tiny clone of and MPPs were virtually undetectable in the
cells harboring a recurrent mutation in an MDS/ residual CD34+CD38– compartment, confirming
AML candidate gene. that HSCs are lost in AA, as determined by phe-
notype. Despite the loss of phenotypic HSCs,
the CD34+CD38+ compartment was detectable
in all cases. The percentage of myeloid progeni-
PATHOPHYSIOLOGY tors was equivalent compared with normal BM.
In contrast, erythroid progenitors were low, like
Hematopoiesis in AA HSCs, in all patients. These results suggest that
AA is a BM failure syndrome characterized ongoing erythropoiesis is more reliant on HSC
by pancytopenia of the peripheral blood and input compared with myelopoiesis. Consistent
BM hypoplasia [2–4]. Profound reduction in he- with these findings, clinical data suggested that
matopoietic stem and progenitor cells (HSPCs) the baseline absolute reticulocyte count (ARC)
is a consistent finding [5–9]. Stem cells/early and absolute lymphocyte count (ALC) together
progenitor cells can be assayed by long-term serve as a simple predictor of response to IST
culture-initiating cell assays (LTC-ICs) [7,9], or and greater rate of 5-year survival [14].
cobblestone area-forming cells [10], these cells In AA, BM is not truly empty but replaced by
are also markedly deficient in AA. The scant fat cells [15]. BM adipocytes were reported to
numbers of LTC-ICs per mononuclear cell sug- be possible negative regulators in the hemato-
gest that only a small percentage of residual poietic microenvironment [16]. To examine the
early hematopoietic cells remain in severely af- role of adipocytes in BM failure, recent study

 
Pathophysiology 15
investigated peroxisomal proliferator-activated of the AA patients, in a limited number of genes
receptor gamma (PPARγ), a key transcription and at low initial variant allele frequency [26].
factor in adipogenesis, utilizing an antagonist of Clonal hematopoiesis was detected in 47% of the
this factor [17]. While PPARγ antagonists inhib- AA, most frequently as acquired mutations. The
ited adipogenesis as expected, it also suppressed prevalence of the mutations increased with age.
T-cell infiltration of BM, reduced plasma in- DNMT3A-mutated and ASXL1-mutated clones
flammatory cytokines, decreased expression of tended to increase in size over time; the size of
multiple inflammasome genes, and ameliorated BCOR- and BCORL1-mutated and PIGA-mutat-
marrow failure. These results suggested that ed clones decreased or remained stable. How-
PPARγ antagonists acted as negative regulators ever, clonal dynamics were highly variable and
of T cells in addition to their inhibition of BM did not determine the response to therapy and
adipogenesis. long-term survival among individual patients.
AA is strongly associated with PNH [18]. These results were consistent with those of re-
PNH is a rare acquired disorder of HSCs, char- cent studies in which candidate-gene targeted
acterized by hemolytic anemia, BM failure, and sequencing also showed recurrent mutations in
venous thrombosis. The etiology of PNH is a a similar spectrum of genes [28,29]. Two hun-
somatic mutation in the X-linked phosphati- dred and nineteen genes were screened in 39 pa-
dylinositol glycan class A gene (PIG-A), result- tients, and found somatic mutations in 9 (23%),
ing in global deficiency of glycosyl phosphati- comprising ASXL1, DNMT3A, and BCOR. The
dylinositol–anchored proteins (GPI–APs) [19]. median allele burden was <10% in 7 patients
Clinically, AA may coexist or appear to evolve [28]. In 2 of 38 patients (SLIT1, and SETBP1 with
to other hematologic diseases that are character- ASXL1) using a smaller panel of 42 genes, 3 mu-
ized by proliferation of distinctive cell clones, as tations were found. However, the patient with
in PNH or MDS [2]. Nearly half of AA patients SETBP1 and ASXL1 was tested at time of pro-
have clonal populations of cells lacking GPI–APs gression to MDS.
because of somatic mutations in the PIG-A gene; These results show parallels between BM
these are called PNH clones [20,21]. Most clones failure and normal aging of the hematopoietic
are small and do not lead to clinical manifesta- compartment. The characteristic mutation sig-
tions of hemolysis or thrombosis [22], but classic nature and correlation of mutations with patient
PNH can be dominated by marrow failure (the age suggested age-related, spontaneous conver-
“AA/PNH syndrome”). PIG-A mutant cells can sion of methylated cytosine to thymidine at CpG
support hematopoiesis for long term, despite sites as major source of nucleotide alternations
accumulation of somatic mutations associated in AA [30]. Similar C-to-T conversion muta-
with clonal evolution to MDS/AML [23–25]. tions accumulate in hematopoietic progenitors
Recent studies have demonstrated clonal he- in healthy persons [31–33]. Mutations generally
matopoiesis in the majority of AA [26,27]. To appeared at a low variant allele frequency and
clarify the origin, importance, and dynamics involved common mutational targets in my-
over time of clonal hematopoiesis in AA, and its eloid cancers, which suggests that the origin and
relationship to the development of MDS, AML, clonal selections of these mutations are similar
or both, targeted deep-sequencing, SNP array to those in AA.
karyotyping, and whole-exome sequencing, However, the exact mechanism of selection
were performed to identify genetic alterations in of mutated cells in AA is unclear. DNMT3A is
AA and described their dynamics over long clin- essential for hematopoietic stem-cell differen-
ical courses [26]. Somatic mutations in myeloid tiation [34]. DNMT3A loss predisposes murine
cancer candidate genes were present in one-third HSCs to malignant transformation [35]. Deletion

 
16 2. Pathophysiology of Acquired Bone Marrow Failure

of ASXL1 results in myelodysplasia in vivo BM [44–50]; CD8+ CTLs are expanded


[36,37]. Cells containing DNMT3A or ASXL1 in AA, leading to the production of
mutations may preferentially self-renew rather proinflammatory cytokines (e.g., IFN-γ) that
than differentiate in response to extrinsic sig- induces apoptosis of CD34+ cells [47,50].
nals. In contrast, striking overrepresentation of 2. Oligoclonal skewing of the T-cell repertoire
BCOR and BCORL1 and PIGA mutations as well indicating expansion of pathogenic CD8+
as frequent 6pUPD involving the specific HLA T cells [51–53]. In general, patients at
classes suggest a mechanism of protection of presentation demonstrate oligoclonal
mutated cells from immune-mediated destruc- expansions of a few Vβ subfamilies, which
tion by pathogenic T cells [38,39]. diminish or disappear with successful
IST; original clones reemerge with relapse,
sometimes accompanied by new clones,
Immune Mechanisms in AA consistent with spreading of the immune
Clinical Data response. Very occasionally, a large clone
An immune mechanism was inferred de- persists in remission, perhaps evidence of
cades ago from the recovery of hematopoiesis T-cell tolerance.
in patients who failed to engraft after stem-cell 3. A reduction of regulatory T cells (Treg) and
transplantation, when renewal of autologous an increase in Th17 related T cells resulting
blood-cell production was credited to the con- in a high Th17/Treg ratio at diagnosis which
ditioning regimen. Also, the majority of synge- tends to normalize in responding patients
neic transplantations in which BM was infused to IST [54,55]; reduction in Treg numbers
without conditioning failed due to rejection im- correlates with disease severity, and the
plying a disease immune mechanism [40]. The defect is most prominent in severe and very
responsiveness of AA to IST in most patients severe AA [56].
is the best evidence of an underlying immune 4. Increased transcription of Th1-related genes
pathophysiology: the majority of patients show in activated T cells of AA patients [57,58];
hematologic improvement after only transient recent study in BM failure mouse models
T-cell depletion by ATGs; relapse also usually identified NOTCH signaling as a primary
responds to ATG; and dependence of adequate driver of Th1-mediated pathogenesis in
blood counts on administration of very low dos- AA and may represent a novel target for
es of CsA is not infrequent [2]. therapeutic intervention.
5. Confirmation in murine models on the role
T Cells and Cytokines of Th1 and Th17 cells and related cytokines
In early laboratory experiments, removal of to producing destruction of marrow
lymphocytes from aplastic BMs improved colony progenitor cells and the positive effects of
numbers in tissue culture, and their addition to Th17 blocking antibodies and infusion of
normal marrow inhibited hematopoiesis in vitro regulatory T cells in reversing BM failure in
[41]. Immunity to HSCs by activated T cells has these models [55,59–63].
been considered to be responsible for the patho- The impact of T-cell attack on BM can be mod-
genesis of AA [42,43]. Laboratory in vitro data eled in vitro and in vivo. IFN-γ (and tumor necro-
has further reinforced the immune pathogenesis sis factor-α) in increasing doses reduce numbers
in AA with the principal findings including: of human hematopoietic progenitors assayed in
1. An increased cytokine (IFN-γ) of activated vitro; the cytokines efficiently induce apoptosis
T cells identified both in the blood and in CD34+ target cells, at least partially through

 
Pathophysiology 17
the Fas-dependent pathway of cell death [43,64]. Immune Escape Clones (PNH, 6pLOH)
In long-term culture of human BM, in which Certain clones may escape the immune attack
stromal cells were engineered to constitutively within the BM environment and proliferate and
express IFN-γ, the output of long-term-culture- attain a survival advantage over normal HSCs.
initiating cells (LTCI-ICs) was markedly dimin- The global absence of a large number of cell-sur-
ished, despite low concentrations of the cytokine face proteins in PNH has been hypothesized to
in the media, consistent with local amplification allow escape and survival of a preexisting mutant
of toxicity in the marrow milieu [49]. clone. Association of present PNH clone with a
Measurements of soluble circulating mediat- predictor of responsiveness to IST suggests that
ing factors in BM failure were limited largely to the escape is from immune attack [20,80–83].
one or two cytokines in AA [65–68]. High TPO Small PNH clones present at diagnosis usually
levels have been observed in patients with AA, remain stable over time, but may expand suffi-
and these abnormal levels correlate with disease ciently to produce symptomatic hemolysis [84].
severity [66,68]. Comprehensive analysis of 31 Comparison by microarray shows that residual
cytokines by an immuno-bead-based multiplex cells of normal phenotype in the PNH BM upreg-
assay (Luminex) identified that high levels of ulate the same apoptosis and cell-death genes as
TPO and granulocyte colony-stimulating fac- do CD34+ cells in aplastic marrow, while the PIG-
tor, with low levels of CD40 ligand, CXCL5, A mutant clone appears transcriptionally similar
CCL5, CXCL11, epidermal growth factor, vascu- to CD34+ cells from healthy donors [85]. Alter-
lar endothelial growth factor, and CCL11 were natively, NK cell mediated cytotoxicity may play
a signature profile for AA [69]. An increase in a role; immunoglobulin-like receptors (KIR) may
IL-17-producing Th17 cells in the peripheral be differentially expressed in PNH compared to
blood and BM of patients with AA has also been normal, resulting in cytotoxicity of normal HSCs
reported [55,70,71]. Recent study showed that [86]. Recent work has suggested expansion of au-
TPO and IL-17 levels are useful for differentiat- toreactive, CD1d-restricted, GPI-specific T cells
ing hypocellular refractory cytopenia of child- in PNH [38]. These data suggested important
hood (RCC) from pediatric AA [72]. roles for cell-extrinsic factors in clonal expansion
of PNH cells.
HLA and Cytokine Gene Polymorphisms
It was recently reported that AA patients po­
There have been a number of studies on the ssessing clonal/oligoclonal hematopoiesis who
human leukocyte antigens (HLA) and their as- had specifically lost either HLA haplotype con-
sociation with AA. HLA-DR2 is overrepresent- taining the HLA-B*40:02, HLA-A*31:01, HLA-
ed among patients [73], suggesting a role for A*02:01, and HLA-A*02:06 [87]. Approximately
antigen recognition, and its presence is predic- 10% of AA have acquired copy number neutral
tive of a better response to CsA [74]. Further re- loss of heterozygosity (CN-LOH) in chromo-
search showed HLA-DRB1*1501 was associated some arm 6p (6pLOH), postulated to emerge by
with a good response to IST in Japanese cohorts immune selection against specific HLA alleles
[75,76]. [87–89]. This clonal hematopoiesis may repre-
Polymorphisms in cytokine genes, associated sent a signature of an escape from cytotoxic T-
with an increased immune response, also may cells autoimmunity targeting autoantigens and
be more prevalent: a nucleotide polymorphism strengthen the hypothesis of the immune-me-
in the TNF-α (TNF2) promoter at −308 [77], ho- diated pathogenesis of AA, although the exact
mozygosity for a variable number of dinucleo- mechanism is still unclear. Recent report showed
tide repeats in the gene encoding IFN-γ [78], and the PNH patient that had acquired CN-6pLOH
polymorphisms in the CTLA4 [79]. in GPI–AP+ granulocytes, but not in GPI–AP−

 
18 2. Pathophysiology of Acquired Bone Marrow Failure

granulocytes, supporting the hypothesis that a matory bowel disease. Recent work described
hostile immune environment drives selection previously unknown potential regulatory roles
of resistant hematopoietic cell clones [90]. Re- of the miR-145-5p and miR-126-3p in T-cell ac-
cent study reported successful isolation of HLA- tivation in AA, in which MYC and PIK3R2 are
B*40:02-restricted CTLs specific for HSCs that the respective targets of these miRNA [99]. Dys-
were present in AA patient peripheral blood or regulated miR-145-5p and miR-126-3p promote
BM [91]. T-cell proliferation and increase GZMB and IFN-
γ production. Targeting or employing miRNA
STAT3 Mutant Clones mimics might be novel molecular therapeutic
Large granular lymphocyte leukemia (LGL) approaches in AA.
is often associated with immune cytopenias and
can occur in BM failure, such as AA and MDS Autoantibodies
[92,93]. STAT3 mutations in LGL clonal expan- Autoantibodies are frequently detected in
sions are detected [94,95]. STAT3 clones can be patients with AA [100–103]. Antimoesin [100],
not only in known LGL concomitant cases, but diazepam-binding inhibitor-related protein 1
in a small population of other BM failure cases [101], kinectin [102], postmeiotic segregation in-
(7% AA and 2.5% MDS) [72]. In STAT3-mutat- creased 1 [102], and HNRNPK antibodies [103]
ed AA patients, trend toward better responses were reported to be expressed in AA. Recent
of IST and an association with the presence of study using SEREX identified autoantibodies
HLA-DR 15 were found. STAT3-mutant clones that are expressed in AA accompanied by im-
may facilitate a persistently dysregulated auto- mune abnormality [104]. Eight candidates were
immune activation, responsible for the primary identified: CLIC1, SLIRP, HSPB11, NHP2L1,
induction of BM failure in a subset of AA and SLC50A1, RPL41, RPS27, and SNRPF.
MDS.
Immune-Mediated BM Failure
Innate Immunity Mouse Models
Transcriptional analysis of T cells from AA Mouse models of AA, produced by the de-
has implicated some components of innate im- struction of BM cells using radiation, cytotoxic
munity in AA, including toll-like receptors and drugs, and immune cells, have been useful in
natural killer cells [96]. defining the hematopoietic stem cell and illus-
There are some experimental results that sup- trating the potency of small numbers of lympho-
port the natural killer cells involvement in AA cytes in specifically inducing apoptosis of BM
[97,98]. KIR and KIR ligand (KIR-L) genotype targets and their cytokines (e.g., IFN-γ-) as nega-
study showed that AA and PNH showed de- tive effector molecules [105–108]. Murine mod-
creased frequency of KIR-2DS1 and KIR-2DS5 els mimicking AA have used exposure to agents
genes [98]. The reduced frequency of these KIRs that result in marrow destruction through a
in AA and PNH may indicate an immunogenetic direct toxic effect, but models that explore anti-
relationship between these diseases. genic disparities between strains have resulted
in immune-mediated destruction of the marrow,
microRNAs more closely modeling human AA [109]. Infu-
There is emerging evidence that microRNA sion of parental lymph node cells into F1 hybrid
(miRNA) play crucial roles in controlling and donors caused pancytopenia, profound mar-
modulating immunity. Dysregulation of miRNA row aplasia, and death [63]. Not only a murine
can lead to autoimmune diseases, such as rheu- version of ATG and CsA but also monoclonal
matoid arthritis, multiple sclerosis, and inflam- antibodies to IFN-γ and tumor necrosis factor

 
Pathophysiology 19
abrogated hematologic disease, rescuing ani- and continued until days 91 and 112 posttreat-
mals. A powerful “innocent bystander” effect, ment [113].
in which activated cytotoxic T cells kill geneti-
cally identical targets, was present in second-
ary transplantation experiments [62]. In a minor Genetic Risk Factors in AA
histocompatibility antigen-discordant model, Telomeres are DNA sequences with a struc-
marrow destruction resulted from activity of an ture that protects chromosomes from erosion and
expanded H60 antigen-specific T-cell clone [61]. that a specific enzyme, telomerase, is involved
Treatment with the CsA abolished H60-specific in their repair after mitosis [114,115]. Telomeres
T-cell expansion and rescued animals from fatal are repeated nucleotide sequences that cap the
pancytopenia. The development of BM failure ends of chromosomes and protect them from
was associated with a significant increase in ac- damage. Acquired and congenital AA have been
tivated CD4+CD25+ T cells that did not express linked molecularly and pathophysiologically
intracellular FoxP3, whereas inclusion of normal by abnormal telomere maintenance [116,117].
CD4+CD25+ regulatory T cells in combination Telomeres are eroded with cell division, but in
with C57BL/6 LN cells aborted H60-specific T- HSCs, maintenance of their length is mediated
cell expansion and prevented BM destruction. by telomerase. Accelerated telomere shorten-
Trafficking of T cells to the marrow has also been ing is virtually universal in dyskeratosis con-
shown to be important in AA pathogenesis in genita, caused by mutations in genes encoding
murine models [110]. components of telomerase or telomere-binding
protein (TERT, TERC, DKC1, NOP10, or TINF2)
Mouse Models of Chemical and Drug [118–122]. Short telomeres were found in leuko-
Hematopoietic Toxicity cytes from approximately one-third of patients
In a few instances, mouse models have been with AA, especially those who do not have a re-
utilized to examine chemical and drug toxicity sponse to IST [123,124]. Systematic screening of
for hematopoiesis. Industrial exposure to ben- patients with apparently AA showed a few pa-
zene has numerous deleterious hematologic tients with TERT or TERC mutations [125,126].
effects in human workers. When benzene was Mutation of SBDS underlie Shwachman–
subcutaneously injected into CD1 mouse, they Diamond syndrome (SDS), inherited syndrome
showed lethargy, irritability, and weight loss, featuring BM failure [127]. Heterozygosis for
with decreased hemoglobin, erythrocytes, leu- 258 + 2 T > C SBDS gene was associated with
kocytes, and BM cells indicative of BM failure AA (4 of 91 AA) and telomere shortening of
[111]. leukocytes [128]. These mutations cause low
Chronic, delayed hematotoxicity of the che- telomerase activity, accelerated telomere short-
motherapy drug busulfan was recapitulated in ening, and diminished proliferative capacity of
a mouse model: following a course of therapy, hematopoietic progenitors. Sex hormones in-
animals maintained normal blood and BM cell crease telomerase activity by upregulating the
counts for 1 year before developing pancytope- TERT gene [129]. Blood count improvement can
nia and frank marrow aplasia, with significant be obtained with androgen therapy in patients
decline in splenic colony-forming units (CFU) with mutation in telomere repair complex genes
[112]. BALB/c mice were treated 8 times with [130]. For more information about telomeres,
busulfan over 23 days and found reductions please refer Chapter 15.
in nucleated marrow cells, granulocyte-macro- Germ-line GATA2 gene mutations, lead-
phage (CFU-GM), CFU-erythroid, erythrocytes, ing to haploinsufficiency, have been identi-
leukocytes, platelets, and reticulocytes on day 1 fied in patients with familial MDS/AML [131],

 
20 2. Pathophysiology of Acquired Bone Marrow Failure

monocytopenia and mycobacterial infections dyskeratosis congenita (DC), and Diamond–


[132,133], Emberger syndrome [134], and den- Blackfan anemia (DBA) are sequenced by this
dritic cell, monocyte, B-, and NK-cell deficien- method [144].
cy [135,136]. GATA2 mutations have also been
identified in a subset of patients presenting with
chronic neutropenia [137], and young adults Clonal Evolution in AA
with AA [138,139], highlighting the clinical With improved survival, the late develop-
heterogeneity and variable hematologic phe- ment of MDS, AML, or both has been noted in
notypes associated with a single genetic defect. about 15% of AA patients and termed “clonal
The BM from patients with GATA2 deficiency evolution” [145]. AA patients with clonal cy-
is typically hypocellular, with varying degrees togenetic patterns are heterogenous; unlike in
of dysplasia. The marrow had severely reduced primary MDS, aberrancies of chromosome 5
monocytes, B cells, and NK cells; absent hema- and 20 were infrequent [146]. The clinical course
togones; and inverted CD4:CD8 ratios. Atypi- depended on the specific abnormal cytogenetic
cal megakaryocytes and abnormal cytogenetics pattern. Most deaths related to leukemic trans-
were more common in GATA2 marrows. Routine formation occur in patients with abnormalities
BM flow cytometry, morphology, and cytogenet- of chromosome 7 or complex cytogenetic altera-
ics in patients who present with cytopenias can tions or both [146]. In contrast, +8 and del13q,
identify patients for whom GATA2 sequencing may appear in AA that is responsive to IST and
is indicated [139]. If GATA2 mutations are iden- associated with good prognosis [146–149]. In AA
tified, it is important to screen family members patients with PNH clone, cytogenetic abnormal-
who may be potential donors, as BM transplan- ities usually occur in hematopoietic cells that are
tation is the only definitive therapy for GATA2 normal phenotype (GPI–AP positive), suggest-
deficiency [140,141]. ing these cells have different origin [25,148].
Familial AA is an extremely rare inherited Telomere dynamics play a role in the devel-
subtype affecting multiple individuals in a fam- opment of myeloid cancers in patients with AA
ily. Patients typically only have features of AA; not associated with a telomeropathy. In adult
the absence of any somatic features making it patients with severe AA undergoing IST (with-
distinct from other inherited AA. By exome se- out a known genetic telomeropathy), pretreat-
quencing, the causative homozygous MPL mu- ment telomere length in the bottom quartile for
tation in a family with familial AA is reported age was a significant risk factor for evolution to
[142]. Biallelic constitutional mutations in MPL MDS [150]. Patients with the shortest telomeres
have been described in congenital amegakaryo- had more uncapped telomere-free chromosome
cytic thrombocytopenia (CAMT) [143]. MPL ends as compared to the patients with the lon-
mutations can be found in children with famil- gest telomeres [151]. Analysis of patients, with-
ial AA in whom CAMT was not diagnosed or out telomeropathies and with normal telomere
suspected. Additional studies will be needed to length at the time of diagnosis, who developed
further clarify the relationship between CAMT, to monosomy 7 found these patients had dra-
AA, and MPL. matically accelerated telomere attrition before
For screening, genomic panels of large num- developing MDS [152]. Rapid telomere loss
bers of genes are now available and routinely led to an accumulation of individual chromo-
used in the clinic to molecularly characterize somes bearing extremely short telomeres prior
patients with suspected inherited bone marrow to the development of monosomy 7 as detected
failure syndromes (IBMFS); multiple genes from by STELA. Dependence on a limited stem-cell
pathways involved in, Fanconi anemia (FA), pool to support hematopoiesis would require

 
Treatments for AA 21
an increased rate of cell division and accelerate were not associated with the development of
telomere attrition. MDS-defining cytogenetic abnormalities [156].
In recent years, a number of studies have In contrast, clones carrying mutations in DN-
reported the presence of acquired somatic mu- MT3A, ASXL1, and a few other genes were more
tations in AA, often associated with low level likely to increase in size over time, and these mu-
clones [28,29,153,154]. These mutations were tations (dominated by DNMT3A and ASXL1) as
recurrent somatic mutations found in MDS/ a group were associated with a poorer response
AML. In a small cohort of predominantly pedi- to IST, inferior overall survival, and progression
atric patients, somatic mutations were detected to MDS, AML, or both. Computational strategy
in 72%, most frequently involved in immune identified patients with better overall survival
escape (PIGA, 6pLOH) and signal transduction (those with PIGA, BCOR, and BCORL1) and pa-
(STAT5B, CAMK2G), and MDS-associated SM tients with worse overall survival (those with
were found in only 9% of patients [27]. A large ASXL1, DNMT3A, TP53, RUNX1, and CSMD1)
cohort of 150 AA patients with no morphologi- than overall survival in the unmutated group,
cal evidence of MDS identified a subgroup (19%) and patients with better progression-free surviv-
with relevant somatic mutations in a small num- al (those with PIGA, BCOR, and BCORL1) and
ber of genes (ASXL1 in 12 patients, DNMT3A in patients with worse progression-free survival
8, BCOR in 6, and (those with ASXL1, DNMT3A, RUNX1, JAK2,
1 each for SRSF2, U2AF1, TET2, MPL, IKZF1, and JAK3) than progression-free survival in the
and ERBB2) [155]. Somatic mutations, when ex- unmutated group.
amined together, predicted for risk of later evo- A more detailed analysis of clonal evolution
lution to MDS; the risk was 38% compared to 6% was obtained by the sequencing of serial samples
in the absence of a somatic mutation, and if the from 35 AA patients that spanned years [26]. In
disease duration of the AA was >6 months, the most patients, clonal hematopoiesis originated
risk of MDS was even more significant at 40% from a minor clone that was already present at
compared to 4% without a somatic mutation. the time of diagnosis. However, the subsequent
ASXL1 and DNMT3A mutations were associated temporal course of these clones was highly vari-
with evolution to monosomy 7 in 4 AA patients. able. Close monitoring of clonal hematopoiesis
They also showed that presence of a somatic by means of both deep sequencing and SNP ar-
mutation was associated with shorter telomere ray karyotyping will need to be combined with
length compared to patients who lacked a somat- clinical evaluation to estimate prognosis and to
ic mutation. In a combined USA and Japanese guide treatment of patients with AA.
study targeted sequencing of 106 genes in 439
AA patients [26], various sets of mutations show
distinct behavior and clinical effect; BCOR-mu- TREATMENTS FOR AA
tant and BCORL1-mutant clones and PIGA-mu-
tant clones tended to disappear or remain small,
BMT With Matched Sibling Donors
were associated with a better response to IST,
and predicted favorable outcomes. These data HSCT is the treatment of choice in newly di-
are compatible with previous studies focusing agnosed patients up to 40 years of age eligible
on the significance of PIG-A mutant clones. The for HSCT with a histocompatible donor [157].
vast majority of patients tended to equilibrate at For all other patients IST with horse ATG/CsA
a clone size, even after IST [23,84]. Recent study is the preferred treatment modality. The long-
reported that 6p CN-LOH clones were present term survival for either therapy is about 80% for
in 11.3% of AA, remained stable over time, and patients of all ages, with younger patients (<20)

 
22 2. Pathophysiology of Acquired Bone Marrow Failure

faring better in general [158]. Standard treat- significantly improved [165,166]. Recent study
ment for patients who have a matched sibling reported that the outcome of UD transplants for
donor is HSCT which provides a cure in about AA is currently not statistically inferior when
80–90% of patients [159]. Cyclophosphamide compared to sibling transplants, although pa-
(CY) with ATG as a conditioning regimen and tients are at greater risk of acute and chronic
the CsA plus methotrexate (MTX) as GVHD pro- GVHD [167]. The risk of death of UD grafts was
phylaxis represent an effective treatment, with a higher, but not significantly higher, compared to
rate of engraftment of 90–95% and overall sur- a sibling donor. The strongest negative predic-
vival near 80–90% [159]. These rates tend to be tor of survival was the use of peripheral blood
worst in older patients (>40 years of age) ap- as a stem cell source, followed by an interval of
proximating 50–60% [160]. diagnosis to transplant of 180 days or more, pa-
BM source, conditioning with CY + ATG, and tient age 20 years or over, no ATG n in the con-
GVHD prophylaxis by CsA + MTX represent the ditioning, and donor/recipient cytomegalovirus
gold standard in transplantation for AA in pa- serostatus, other than negative/negative.
tients receiving transplantations from an HLA-
identical sibling donors. Given these excellent
results, survival is thus no more the sole concern Experimental HSCT
then prevention and early detection of late com- Other alternative source of stem cells in-
plications after BMT is the main objective [161]. cludes umbilical cord blood and a haploiden-
After transplantation from an HLA-identical sib- tical family donor [168]. The difficult situation
ling donor or from an unrelated donor, the use encountered is when a patient with refractory
of peripheral blood stem cells must be strongly AA has no suitably matched UD, and this is not
discouraged because they have been systemati- uncommon. The options for these individuals
cally associated with an increased incidence of include a second course of IST, an alternative
chronic GVHD compared with the use of BM as immunosuppressive drug or novel agent, or an
a stem cell source, leading to an unacceptably experimental form of transplantation using an
higher risk of treatment-related mortality in this alternative donor source, namely cord blood or
setting. a haploidentical family donor. HSCT offers pos-
sibilities of cure, but risks of alternative donor
HSCT remain graft rejection and GVHD, espe-
BMT From Alternative Donors cially chronic GVHD, which affects mortality
For patients who lack an HLA identical sib- and quality of life.
ling donor, IST remains the first treatment of The key features of haploidentical graft are
choice. However, 30–40% of the patients will availability for most patients, the time to pro-
be refractory or relapse to IST and will thus be cure the graft is short, and the cost is low. This
considered for transplantation using an alterna- is not a new approach to HSCT for SAA patients
tive donor. HSCT is indicated if refractory AA lacking a matched sibling donor or a suitably
patients are fit and have a suitably matched matched UD. Historically, haploidentical HSCT
donor. Transplantation from an unrelated do- was invariably unsuccessful, with high rates of
nor (UD) is usually considered after failure of graft rejection and GVHD. A recent review on
at least one course of IST [162]. This strategy is 73 patients receiving HSCT between 1976 and
based on a relatively high risk of complications 2011 and mostly using nonmyeloablative regi-
for UD transplant recipients, such as graft rejec- mens showed a 3-year OS of only 37% [169]. A
tion, GvHD, and infections [163,164]. However, novel approach is nonmyeloablative condition-
the outcome of unrelated donor transplants has ing with high-dose CY given on days +3 and +4

 
Treatments for AA 23
after transplantation (PTCy) to prevent GVHD Amounting evidence that AA had an immune
by depleting dividing donor-alloreactive T cells mediated pathogenesis led to the exploration
but sparing quiescent, nonalloreactive T cells. of more immunosuppressive agents in clinical
Such an approach has been reported anecdotally protocols. This development was conducted in
in PNH and is under study in protocols in AA two manners: (1) adding a third immunosup-
[170]. Recent studies from Johns Hopkins and pressive agent to horse ATG/CsA and (2) using
others showed that nonmyeloablative allo-BMT more potent lymphocyte depleting agents than
with PTCy from haploidentical donor appears horse ATG. Neither strategy improved the out-
promising in patients with refractory acquired comes. Adding mycophenolate mofetil or siro-
and inherited AA with acceptable rates of en- limus to horse ATG/CsA did not improve out-
graftment, eradication of preexisting clonal dis- comes in SAA and the use of more lymphocyte
eases, low risk of GVHD, and expansion of the depleting agents (cyclophosphamide, alemtu-
donor pool [171,172]. zumab, rabbit ATG) was equally disappointing
The largest retrospective study of unrelated [180,181]. Cyclophosphamide, despite being an
cord blood transplantation comprised 71 AA active agent in SAA, is associated with prohibi-
patients (9 with PNH) [173]. The main problem tive toxicity impeding its use [182,183]. Toxic-
was engraftment failure, with a cumulative in- ity was considerable, mainly due to prolonged
cidence of neutrophil recovery of only 51% at absolute neutropenia, that occurred regardless
2 months and a 3-year OS of 38%. All those pa- of pretherapy blood counts, and persisted an
tients receiving total body irradiation 12 Gy as average of 2 months [183]. Alemtuzumab as-
part of the conditioning regimen died, indicat- sociated with a low response rate (about 20%)
ing that a RIC rather than myeloablative regi- when given as first therapy [184]. Noteworthy
men is preferable. Significantly improved OS are the disappointing results with rabbit ATG/
was seen in recipients of >3.9 × 107 TNCs/kg CsA when compared directly to horse ATG/
prefreezing. Recent study showed that haplo- CsA in a randomized study [185]. The anticipa-
cord HSCT is an effective treatment option for tion was for a higher response rate with rabbit
severe AA patients who lack an HLA-matched ATG/CsA given its more lymphocyte depleting
donor [174]. properties, Treg inducing property, activity in
horse ATG/CsA refractory cases and superior-
ity to horse ATG in solid organ transplant [186–
IST 189]. However, results showed a lower response
The possibility that the pathophysiology of rate of about 35–40% compared to the expected
AA could be immune mediated was initially 60–70% seen with horse ATG/CsA [185]. This
proposed by Barnes and Mole and later rein- difference resulted in superior survival out-
forced by autologous hematologic recovery af- comes with horse ATG/CsA compared to rab-
ter ATG exposure [109,175]. The combination of bit ATG/CsA [185]. Other observations from
ATG + CsA was shown superior to ATG alone prospective studies confirmed this observation
and became the standard IST option in patients [190]. Therefore, horse ATG/CsA remains the
not eligible for HSCT [176]. Extensive experi- optimal immunosuppressive regimen as first
ence with this regimen showed that 60–70% of line therapy in SAA. The options for patients
patients responded to the combination of horse who fail initial horse ATG/CsA include HSCT
ATG/CsA becoming the standard IST regimen. from a related (in older patients) or unrelated
Across these studies hematologic response (in younger patients) histocompatible donor or
correlated with excellent long-term survival a repeat course of IST [191,192]. Rabbit ATG/
[157,176–179]. CsA or alemtuzumab in this refractory setting

 
24 2. Pathophysiology of Acquired Bone Marrow Failure

can salvage about 30–40% of patients who can titrated up to 150 mg in a IST refractory SAA
achieve a hematologic response [184,188]. The cohort [202]. The overall response rate was 44%
ability to salvage a proportion of patients with with some bi- and trilineage hematologic im-
transplant and nontransplant modalities along provements observed. The degree and quality of
with advances in supportive care have resulted improvement in blood counts with eltrombopag
in improved survival outcomes in IST refractory of patients with refractory AA was unanticipat-
patients over the years [193]. Even among very ed. Responses were striking in several respects:
refractory patients to IST who do not undergo they were not restricted to platelets and were
HSCT long-term survival can be achieved with robust, resulting in transfusion independence.
supportive care measure, such as transfusions, In an extended experience (n = 43) the response
antimicrobials, growth factor, androgens and rate was confirmed at 40% and multilineage in-
iron chelation. Neutrophil count in this setting crement in blood counts were continued to be
are a principal determinant of survival [194]. observed [203]. Discontinuation of eltrombopag
When neutrophils are adequate in numbers was possible in a few patients who had achieved
that prevent recurrent life-threatening infec- a robust hematologic recovery without worsen-
tions long-term survival with supportive care ing of the counts. Eight of 43 patients developed
measures can be achieved [194]. A longer, taper- new cytogenetic abnormalities. The most com-
ing course of CsA following horse ATG up to mon chromosomal changes were chromosome
24 months as initial therapy delayed but did not 7 abnormalities, which developed in five of the
ultimately prevent relapses [195]. Therefore, ef- eight evolvers. This clonal transformation is as-
forts to improve beyond the outcomes achieved sociated with a poor outcome, but all patients
with horse ATG plus CsA were ineffective until who developed chromosome 7 changes were
recently. successfully transplanted. Increase in bone mar-
row cellularity was observed in some respond-
ing patients suggesting that eltrombopag was
Eltrombopag stimulating a more primitive progenitor popu-
TPO mimetics, such as romiplostim and el- lation leading to improvement in marrow func-
trombopag, were developed to treat patients with tion and increase in blood counts. The protection
refractory immune thrombocytopenia but have and/or stimulation of residual HSCs by eltrom-
been investigated for the treatment of BM failure bopag is a strategy with many applications.
syndromes [196]. TPO is the main regulator for Multiple clinical trials of thrombopoietin mi-
platelet production and its receptor (c-Mpl) is metics, eltrombopag, and romiplostim are in
present on megakaryocytes and HSCs [197,198]. progress. Eltrombopag is being combined with
Historically the use of G-CSF and erythropoietin standard horse ATG and CsA in treatment-naive
have not been effective in systematic AA studies severe AA [204]. In these previously untreated
and did not change the natural history of the dis- patients, the combination of eltrombopag with
ease [199]. The use of a TPO agonist was of par- horse ATG and CsA yielded overall response
ticular interest given the expression of the TPO rate at 3 and 6 months of 80 and 85%, respec-
agonist receptor in marrow progenitor cells and tively, which are 20–30% higher than histori-
the reduced numbers of HSCs in murine TPO cal rates with the same regimen without Tpo
receptor knockout models [200]. However, the agonist in AA. Clonal cytogenetic evolution oc-
very high endogenous TPO levels in SAA could curred in 7 of 88 patients, similar to prior rates
render this approach ineffective [201]. observed with standard IST. Longer and more
In a pilot study (n = 26) eltrombopag was mature data regarding this novel combination
investigated at an initial dose of 50 mg/day is awaited to better define the durability of

 
REFERENCES 25
response and the impact on long-term outcomes mechanistic insights into normal hematopoiesis,
of relapse and clonal evolution. relevant to leukemia and normal aging.

Supportive Care References


Management of AA has been dramatically [1] Gerull S, Stern M, Apperley J, Beelen D, Brinch L,
Bunjes D, et al. Syngeneic transplantation in aplastic
improved since late 1970s with the introduction
anemia: pre-transplant conditioning and peripheral
of allogeneic stem-cell transplantation (allo- blood are associated with improved engraftment: an
SCT) and IST as well as optimized supportive observational study on behalf of the Severe Aplastic
care [194]. Supportive care has impacted posi- Anemia and Pediatric Diseases Working Parties of the
tively these different treatment modalities. The European Group for Blood and Marrow Transplanta-
tion. Haematologica 2013;98(11):1804–9.
advent of oral antifungals with activity against
[2] Young NS, Calado RT, Scheinberg P. Current concepts
Aspergillus sp. (voriconazole, posaconazole) in the pathophysiology and treatment of aplastic ane-
has allowed for continued outpatient therapy mia. Blood 2006;108(8):2509–19.
against these pathogens that represent the dead- [3] Young NS, Bacigalupo A, Marsh JC. Aplastic anemia:
liest infectious culprit in SAA [194,205]. The use pathophysiology and treatment. Biology of blood
and marrow transplantation: journal of the American
of granulocyte transfusion in selected cases has
Society for Blood and Marrow Transplantation 2010;
allowed for better control of fungal infections 16(1 Suppl.):S119–125.
when neutropenia is severe and persistent and [4] Killick SB, Bown N, Cavenagh J, Dokal I, Foukaneli
antifungals are not resolutive alone [206,207]. T, Hill A, et al. Guidelines for the diagnosis and man-
Growth factors can be effective in some patients agement of adult aplastic anaemia. Br J Haematol
2016;172(2):187–207.
with increments in neutrophil and/or hemoglo-
[5] Scopes J, Bagnara M, Gordon-Smith EC, Ball SE, Gib-
bin levels lessening transfusion requirements son FM. Haemopoietic progenitor cells are reduced in
and the risk associated with these cytopenias. aplastic anaemia. Br J Haematol 1994;86(2):427–30.
Oral iron chelators have allowed for continued [6] Marsh JC, Chang J, Testa NG, Hows JM, Dexter TM.
long-term transfusion programs in very refrac- The hematopoietic defect in aplastic anemia assessed by
long-term marrow culture. Blood 1990;76(9):1748–57.
tory patients minimizing the risks associated
[7] Maciejewski JP, Selleri C, Sato T, Anderson S, Young
with iron accumulation in the long term [208]. In NS. A severe and consistent deficit in marrow and
the aggregate these advances have allowed for circulating primitive hematopoietic cells (long-term
patients who failed IST and are not HSCT eligi- culture-initiating cells) in acquired aplastic anemia.
ble to be supported for long periods of time with Blood 1996;88(6):1983–91.
[8] Matsui WH, Brodsky RA, Smith BD, Borowitz MJ,
few complications associated with the persistent
Jones RJ. Quantitative analysis of bone marrow CD34
cytopenias. In general, the neutrophil count is cells in aplastic anemia and hypoplastic myelodys-
an important determinant in how these patients plastic syndromes. Leukemia 2006;20(3):458–62.
who are kept on supportive care fare long term. [9] Rizzo S, Scopes J, Elebute MO, Papadaki HA, Gor-
don-Smith EC, Gibson FM. Stem cell defect in aplas-
tic anemia: reduced long term culture-initiating cells
(LTC-IC) in CD34+ cells isolated from aplastic anemia
CONCLUSIONS patient bone marrow. Hematol J 2002;3(5):230–6.
[10] Schrezenmeier H, Jenal M, Herrmann F, Heimpel H,
Nature of evidence, hematopoiesis, immune Raghavachar A. Quantitative analysis of cobblestone
pathophysiology, genetic risk factors, and risk area-forming cells in bone marrow of patients with
aplastic anemia by limiting dilution assay. Blood
factors for clonal evolution for AA were sum-
1996;88(12):4474–80.
marized. Better understanding of hemato- [11] Sun J, Ramos A, Chapman B, Johnnidis JB, Le L, Ho
poiesis and immune mechanisms in AA may YJ, et al. Clonal dynamics of native haematopoiesis.
provide new therapeutic strategies, and novel Nature 2014;514(7522):322–7.

 
26 2. Pathophysiology of Acquired Bone Marrow Failure

[12] Busch K, Klapproth K, Barile M, Flossdorf M, Hol- [25] Sloand EM, Fuhrer M, Keyvanfar K, Mainwaring L,
land-Letz T, Schlenner SM, et al. Fundamental proper- Maciejewski J, Wang Y, et al. Cytogenetic abnormali-
ties of unperturbed haematopoiesis from stem cells in ties in paroxysmal nocturnal haemoglobinuria usually
vivo. Nature 2015;518(7540):542–6. occur in haematopoietic cells that are glycosylphos-
[13] Notta F, Zandi S, Takayama N, Dobson S, Gan OI, phatidylinositol-anchored protein (GPI-AP) positive.
Wilson G, et al. Distinct routes of lineage development Br J Haematol 2003;123(1):173–6.
reshape the human blood hierarchy across ontogeny. [26] Yoshizato T, Dumitriu B, Hosokawa K, Makishima H,
Science 2016;351(6269):aab2116. Yoshida K, Townsley D, et al. Somatic mutations and
[14] Scheinberg P, Wu CO, Nunez O, Young NS. Predict- clonal hematopoiesis in aplastic anemia. N Engl J Med
ing response to immunosuppressive therapy and 2015;373(1):35–47.
survival in severe aplastic anaemia. Br J Haematol [27] Babushok DV, Perdigones N, Perin JC, Olson TS, Ye
2009;144(2):206–16. W, Roth JJ, et al. Emergence of clonal hematopoiesis in
[15] Takaku T, Malide D, Chen J, Calado RT, Kajigaya S, the majority of patients with acquired aplastic anemia.
Young NS. Hematopoiesis in 3 dimensions: human Cancer Genet 2015;208(4):115–28.
and murine bone marrow architecture visualized by [28] Lane AA, Odejide O, Kopp N, Kim S, Yoda A, Erlich
confocal microscopy. Blood 2010;116(15):e41–55. R, et al. Low frequency clonal mutations recoverable
[16] Naveiras O, Nardi V, Wenzel PL, Hauschka PV, Fahey by deep sequencing in patients with aplastic anemia.
F, Daley GQ. Bone-marrow adipocytes as negative Leukemia 2013;27(4):968–71.
regulators of the haematopoietic microenvironment. [29] Heuser M, Schlarmann C, Dobbernack V, Panagiota V,
Nature 2009;460(7252):259–63. Wiehlmann L, Walter C, et al. Genetic characterization
[17] Sato K, Feng X, Chen J, Li J, Muranski P, Desierto MJ, of acquired aplastic anemia by targeted sequencing.
et al. PPARγ antagonist attenuates mouse immune- Haematologica 2014;99(9):e165–167.
mediated bone marrow failure by inhibition of T cell [30] Alexandrov LB, Nik-Zainal S, Wedge DC, Apari-
function. Haematologica 2016;101(1):57–67. cio SA, Behjati S, Biankin AV, et al. Signatures of
[18] Young NS, Maciejewski JP, Sloand E, Chen G, Zeng mutational processes in human cancer. Nature
W, Risitano A, et al. The relationship of aplastic ane- 2013;500(7463):415–21.
mia and PNH. Int J Hematol 2002;76(Suppl. 2):168–72. [31] Welch JS, Ley TJ, Link DC, Miller CA, Larson DE, Ko-
[19] Takeda J, Miyata T, Kawagoe K, Iida Y, Endo Y, Fu- boldt DC, et al. The origin and evolution of mutations
jita T, et al. Deficiency of the GPI anchor caused by in acute myeloid leukemia. Cell 2012;150(2):264–78.
a somatic mutation of the PIG-A gene in paroxysmal [32] Jaiswal S, Fontanillas P, Flannick J, Manning A, Grau-
nocturnal hemoglobinuria. Cell 1993;73(4):703–11. man PV, Mar BG, et al. Age-related clonal hematopoi-
[20] Dunn DE, Tanawattanacharoen P, Boccuni P, Nagaku- esis associated with adverse outcomes. N Engl J Med
ra S, Green SW, Kirby MR, et al. Paroxysmal nocturnal 2014;371(26):2488–98.
hemoglobinuria cells in patients with bone marrow [33] Genovese G, Jaiswal S, Ebert BL, McCarroll SA. Clonal
failure syndromes. Ann Intern Med 1999;131(6):401–8. hematopoiesis and blood-cancer risk. N Engl J Med
[21] Scheinberg P, Marte M, Nunez O, Young NS. Parox- 2015;372(11):1071–2.
ysmal nocturnal hemoglobinuria clones in severe [34] Challen GA, Sun D, Jeong M, Luo M, Jelinek J, Berg JS,
aplastic anemia patients treated with horse antithy- et al. Dnmt3a is essential for hematopoietic stem cell
mocyte globulin plus cyclosporine. Haematologica differentiation. Nat Genet 2012;44(1):23–31.
2010;95(7):1075–80. [35] Mayle A, Yang L, Rodriguez B, Zhou T, Chang E, Cur-
[22] Wang H, Chuhjo T, Yasue S, Omine M, Nakao S. Clini- ry CV, et al. Dnmt3a loss predisposes murine hemato-
cal significance of a minor population of paroxysmal poietic stem cells to malignant transformation. Blood
nocturnal hemoglobinuria-type cells in bone marrow 2015;125(4):629–38.
failure syndrome. Blood 2002;100(12):3897–902. [36] Abdel-Wahab O, Gao J, Adli M, Dey A, Trimarchi T,
[23] Shen W, Clemente MJ, Hosono N, Yoshida K, Przy- Chung YR, et al. Deletion of Asxl1 results in myelo-
chodzen B, Yoshizato T, et al. Deep sequencing reveals dysplasia and severe developmental defects in vivo. J
stepwise mutation acquisition in paroxysmal nocturnal Exp Med 2013;210(12):2641–59.
hemoglobinuria. J Clin Invest 2014;124(10):4529–38. [37] Wang J, Li Z, He Y, Pan F, Chen S, Rhodes S, et al. Loss
[24] Katagiri T, Kawamoto H, Nakakuki T, Ishiyama K, of Asxl1 leads to myelodysplastic syndrome-like dis-
Okada-Hatakeyama M, Ohtake S, et al. Individual ease in mice. Blood 2014;123(4):541–53.
hematopoietic stem cells in human bone marrow of [38] Gargiulo L, Papaioannou M, Sica M, Talini G, Chaidos
patients with aplastic anemia or myelodysplastic syn- A, Richichi B, et al. Glycosylphosphatidylinositol-spe-
drome stably give rise to limited cell lineages. Stem cific, CD1d-restricted T cells in paroxysmal nocturnal
Cells 2013;31(3):536–46. hemoglobinuria. Blood 2013;121(14):2753–61.

 
REFERENCES 27
[39] Murakami Y, Kosaka H, Maeda Y, Nishimura J, Inoue memory stem cells in acquired aplastic anemia. J Im-
N, Ohishi K, et al. Inefficient response of T lympho- munol 2016;196(4):1568–78.
cytes to glycosylphosphatidylinositol anchor-negative [51] Risitano AM, Maciejewski JP, Green S, Plasilova M,
cells: implications for paroxysmal nocturnal hemoglo- Zeng W, Young NS. In-vivo dominant immune re-
binuria. Blood 2002;100(12):4116–22. sponses in aplastic anaemia: molecular tracking of pu-
[40] Hinterberger W, Rowlings PA, Hinterberger-Fischer tatively pathogenetic T-cell clones by TCR beta-CDR3
M, Gibson J, Jacobsen N, Klein JP, et al. Results of sequencing. Lancet 2004;364(9431):355–64.
transplanting bone marrow from genetically identical [52] Kook H, Risitano AM, Zeng W, Wlodarski M, Lot-
twins into patients with aplastic anemia. Ann Intern temann C, Nakamura R, et al. Changes in T-cell
Med 1997;126(2):116–22. receptor VB repertoire in aplastic anemia: effects
[41] Young NS. Hematopoietic cell destruction by immune of different immunosuppressive regimens. Blood
mechanisms in acquired aplastic anemia. Semin He- 2002;99(10):3668–75.
matol 2000;37(1):3–14. [53] Zeng W, Maciejewski JP, Chen G, Young NS. Limited
[42] Nakao S, Takami A, Takamatsu H, Zeng W, Sugimori heterogeneity of T cell receptor BV usage in aplastic
N, Yamazaki H, et al. Isolation of a T-cell clone show- anemia. J Clin Invest 2001;108(5):765–73.
ing HLA-DRB1*0405-restricted cytotoxicity for hema- [54] Solomou EE, Rezvani K, Mielke S, Malide D, Keyvan-
topoietic cells in a patient with aplastic anemia. Blood far K, Visconte V, et al. Deficient CD4+ CD25+
1997;89(10):3691–9. FOXP3+ T regulatory cells in acquired aplastic ane-
[43] Maciejewski J, Selleri C, Anderson S, Young NS. mia. Blood 2007;110(5):1603–6.
Fas antigen expression on CD34+ human marrow [55] de Latour RP, Visconte V, Takaku T, Wu C, Erie AJ,
cells is induced by interferon gamma and tumor Sarcon AK, et al. Th17 immune responses contribute
necrosis factor alpha and potentiates cytokine-me- to the pathophysiology of aplastic anemia. Blood
diated hematopoietic suppression in vitro. Blood 2010;116(20):4175–84.
1995;85(11):3183–90. [56] Kordasti S, Marsh J, Al-Khan S, Jiang J, Smith A,
[44] Zoumbos NC, Gascon P, Djeu JY, Young NS. Inter- Mohamedali A, et al. Functional characterization of
feron is a mediator of hematopoietic suppression in CD4+ T cells in aplastic anemia. Blood 2012;119(9):
aplastic anemia in vitro and possibly in vivo. Proc 2033–43.
Natl Acad Sci USA 1985;82(1):188–92. [57] Solomou EE, Keyvanfar K, Young NS. T-bet, a Th1
[45] Zoumbos NC, Gascon P, Djeu JY, Trost SR, Young NS. transcription factor, is up-regulated in T cells from
Circulating activated suppressor T lymphocytes in patients with aplastic anemia. Blood 2006;107(10):
aplastic anemia. N Engl J Med 1985;312(5):257–65. 3983–91.
[46] Gascon P, Zoumbos NC, Scala G, Djeu JY, Moore JG, [58] Roderick JE, Gonzalez-Perez G, Kuksin CA, Dongre
Young NS. Lymphokine abnormalities in aplastic ane- A, Roberts ER, Srinivasan J, et al. Therapeutic target-
mia: implications for the mechanism of action of anti- ing of NOTCH signaling ameliorates immune-mediat-
thymocyte globulin. Blood 1985;65(2):407–13. ed bone marrow failure of aplastic anemia. J Exp Med
[47] Sloand E, Kim S, Maciejewski JP, Tisdale J, Follmann 2013;210(7):1311–29.
D, Young NS. Intracellular interferon-gamma in cir- [59] Tang Y, Desierto MJ, Chen J, Young NS. The role of
culating and marrow T cells detected by flow cytom- the Th1 transcription factor T-bet in a mouse model
etry and the response to immunosuppressive therapy of immune-mediated bone-marrow failure. Blood
in patients with aplastic anemia. Blood 2002;100(4): 2010;115(3):541–8.
1185–91. [60] Omokaro SO, Desierto MJ, Eckhaus MA, Ellison
[48] Sato T, Selleri C, Young NS, Maciejewski JP. Inhibition FM, Chen J, Young NS. Lymphocytes with aberrant
of interferon regulatory factor-1 expression results in expression of Fas or Fas ligand attenuate immune
predominance of cell growth stimulatory effects of in- bone marrow failure in a mouse model. J Immunol
terferon-gamma due to phosphorylation of Stat1 and 2009;182(6):3414–22.
Stat3. Blood 1997;90(12):4749–58. [61] Chen J, Ellison FM, Eckhaus MA, Smith AL, Keyvan-
[49] Selleri C, Maciejewski JP, Sato T, Young NS. Inter- far K, Calado RT, et al. Minor antigen h60-mediated
feron-gamma constitutively expressed in the stro- aplastic anemia is ameliorated by immunosuppres-
mal microenvironment of human marrow cultures sion and the infusion of regulatory T cells. J Immunol
mediates potent hematopoietic inhibition. Blood 2007;178(7):4159–68.
1996;87(10):4149–57. [62] Chen J, Lipovsky K, Ellison FM, Calado RT, Young
[50] Hosokawa K, Muranski P, Feng X, Townsley DM, Liu NS. Bystander destruction of hematopoietic progeni-
B, Knickelbein J, et al. Memory stem T cells in auto- tor and stem cells in a mouse model of infusion-in-
immune disease: high frequency of circulating CD8+ duced bone marrow failure. Blood 2004;104(6):1671–8.

 
28 2. Pathophysiology of Acquired Bone Marrow Failure

[63] Bloom ML, Wolk AG, Simon-Stoos KL, Bard JS, [75] Sugimori C, Yamazaki H, Feng X, Mochizuki K, Kon-
Chen J, Young NS. A mouse model of lymphocyte do Y, Takami A, et al. Roles of DRB1 *1501 and DRB1
infusion-induced bone marrow failure. Exp Hematol *1502 in the pathogenesis of aplastic anemia. Exp He-
2004;32(12):1163–72. matol 2007;35(1):13–20.
[64] Maciejewski JP, Selleri C, Sato T, Anderson S, Young [76] Nakao S, Takamatsu H, Chuhjo T, Ueda M, Shiobara
NS. Increased expression of Fas antigen on bone mar- S, Matsuda T, et al. Identification of a specific HLA
row CD34+ cells of patients with aplastic anaemia. Br class II haplotype strongly associated with susceptibil-
J Haematol 1995;91(1):245–52. ity to cyclosporine-dependent aplastic anemia. Blood
[65] Hirayama Y, Sakamaki S, Matsunaga T, Kuga T, Ku- 1994;84(12):4257–61.
roda H, Kusakabe T, et al. Concentrations of throm- [77] Demeter J, Messer G, Schrezenmeier H. Clinical rele-
bopoietin in bone marrow in normal subjects and in vance of the TNF-alpha promoter/enhancer polymor-
patients with idiopathic thrombocytopenic purpura, phism in patients with aplastic anemia. Ann Hematol
aplastic anemia, and essential thrombocythemia cor- 2002;81(10):566–9.
relate with its mRNA expression of bone marrow stro- [78] Dufour C, Capasso M, Svahn J, Marrone A, Haupt
mal cells. Blood 1998;92(1):46–52. R, Bacigalupo A, et al. Homozygosis for (12) CA re-
[66] Marsh JC, Gibson FM, Prue RL, Bowen A, Dunn peats in the first intron of the human IFN-gamma
VT, Hornkohl AC, et al. Serum thrombopoietin lev- gene is significantly associated with the risk of aplas-
els in patients with aplastic anaemia. Br J Haematol tic anaemia in Caucasian population. Br J Haematol
1996;95(4):605–10. 2004;126(5):682–5.
[67] Kojima S, Matsuyama T, Kodera Y, Nishihira H, Ueda [79] Svahn J, Capasso M, Lanciotti M, Marrone A, Haupt
K, Shimbo T, et al. Measurement of endogenous plas- R, Bacigalupo A, et al. The polymorphisms -318C > T
ma granulocyte colony-stimulating factor in patients in the promoter and 49A > G in exon 1 of CTLA4 and
with acquired aplastic anemia by a sensitive chemilu- the risk of aplastic anemia in a Caucasian population.
minescent immunoassay. Blood 1996;87(4):1303–8. Bone Marrow Transplant 2005;35(Suppl. 1):S89–92.
[68] Kojima S, Matsuyama T, Kodera Y, Tahara T, Kato T. [80] Nakao S, Sugimori C, Yamazaki H. Clinical signifi-
Measurement of endogenous plasma thrombopoietin cance of a small population of paroxysmal nocturnal
in patients with acquired aplastic anaemia by a sensi- hemoglobinuria-type cells in the management of bone
tive enzyme-linked immunosorbent assay. Br J Hae- marrow failure. Int J Hematol 2006;84(2):118–22.
matol 1997;97(3):538–43. [81] Sugimori C, Chuhjo T, Feng X, Yamazaki H, Takami A,
[69] Feng X, Scheinberg P, Wu CO, Samsel L, Nunez O, Teramura M, et al. Minor population of CD55-CD59-
Prince C, et al. Cytokine signature profiles in acquired blood cells predicts response to immunosuppressive
aplastic anemia and myelodysplastic syndromes. therapy and prognosis in patients with aplastic ane-
Haematologica 2011;96(4):602–6. mia. Blood 2006;107(4):1308–14.
[70] Du HZ, Wang Q, Ji J, Shen BM, Wei SC, Liu LJ, et al. [82] Kulagin A, Lisukov I, Ivanova M, Golubovskaya I,
Expression of IL-27, Th1 and Th17 in patients with Kruchkova I, Bondarenko S, et al. Prognostic value of
aplastic anemia. J Clin Immunol 2013;33(2):436–45. paroxysmal nocturnal haemoglobinuria clone pres-
[71] Gu Y, Hu X, Liu C, Qv X, Xu C. Interleukin (IL)-17 pro- ence in aplastic anaemia patients treated with com-
motes macrophages to produce IL-8, IL-6 and tumour bined immunosuppression: results of two-centre pro-
necrosis factor-alpha in aplastic anaemia. Br J Haema- spective study. Br J Haematol 2014;164(4):546–54.
tol 2008;142(1):109–14. [83] Hosokawa K, Sugimori N, Katagiri T, Sasaki Y, Saito
[72] Elmahdi S, Hama A, Manabe A, Hasegawa D, Mura- C, Seiki Y, et al. Increased glycosylphosphatidylino-
matsu H, Narita A, et al. A cytokine-based diagnostic sitol-anchored protein-deficient granulocytes define
program in pediatric aplastic anemia and hypocellu- a benign subset of bone marrow failures in patients
lar refractory cytopenia of childhood. Pediatr Blood with trisomy 8. Eur J Haematol 2015;95(3):230–8.
Cancer 2016;63(4):652–8. [84] Sugimori C, Mochizuki K, Qi Z, Sugimori N, Ishiya-
[73] Chapuis B, Von Fliedner VE, Jeannet M, Merica H, ma K, Kondo Y, et al. Origin and fate of blood cells
Vuagnat P, Gratwohl A, et al. Increased frequency deficient in glycosylphosphatidylinositol-anchored
of DR2 in patients with aplastic anaemia and in- protein among patients with bone marrow failure. Br
creased DR sharing in their parents. Br J Haematol J Haematol 2009;147(1):102–12.
1986;63(1):51–7. [85] Chen G, Zeng W, Maciejewski JP, Kcyvanfar K, Bill-
[74] Maciejewski JP, Follmann D, Nakamura R, Saunth- ings EM, Young NS. Differential gene expression in
ararajah Y, Rivera CE, Simonis T, et al. Increased fre- hematopoietic progenitors from paroxysmal noctur-
quency of HLA-DR2 in patients with paroxysmal noc- nal hemoglobinuria patients reveals an apoptosis/im-
turnal hemoglobinuria and the PNH/aplastic anemia mune response in ’normal’ phenotype cells. Leukemia
syndrome. Blood 2001;98(13):3513–9. 2005;19(5):862–8.

 
REFERENCES 29
[86] van Bijnen ST, Withaar M, Preijers F, van der Meer A, isoforms of inhibiting superfamily receptors. Blood
de Witte T, Muus P, et al. T cells expressing the activat- 2005;106(7):2399–408.
ing NK-cell receptors KIR2DS4, NKG2C and NKG2D [98] Howe EC, Wlodarski M, Ball EJ, Rybicki L, Maciejew-
are elevated in paroxysmal nocturnal hemoglobinuria ski JP. Killer immunoglobulin-like receptor geno-
and cytotoxic toward hematopoietic progenitor cell type in immune-mediated bone marrow failure syn-
lines. Exp Hematol 2011;39(7). 751-62 e1–751-62 e3. dromes. Exp Hematol 2005;33(11):1357–62.
[87] Katagiri T, Sato-Otsubo A, Kashiwase K, Morishima S, [99] Hosokawa K, Muranski P, Feng X, Keyvanfar K,
Sato Y, Mori Y, et al. Frequent loss of HLA alleles asso- Townsley DM, Dumitriu B, et al. Identification of nov-
ciated with copy number-neutral 6pLOH in acquired el microRNA signatures linked to acquired aplastic
aplastic anemia. Blood 2011;118(25):6601–9. anemia. Haematologica 2015;100(12):1534–45.
[88] Babushok DV, Xie HM, Roth JJ, Perdigones N, Olson [100] Takamatsu H, Feng X, Chuhjo T, Lu X, Sugimori
TS, Cockroft JD, et al. Single nucleotide polymor- C, Okawa K, et al. Specific antibodies to moesin, a
phism array analysis of bone marrow failure patients membrane-cytoskeleton linker protein, are frequently
reveals characteristic patterns of genetic changes. Br J detected in patients with acquired aplastic anemia.
Haematol 2014;164(1):73–82. Blood 2007;109(6):2514–20.
[89] Afable MG II, Wlodarski M, Makishima H, Shaik M, [101] Feng X, Chuhjo T, Sugimori C, Kotani T, Lu X, Takami
Sekeres MA, Tiu RV, et al. SNP array-based karyo- A, et al. Diazepam-binding inhibitor-related protein
typing: differences and similarities between aplastic 1: a candidate autoantigen in acquired aplastic ane-
anemia and hypocellular myelodysplastic syndromes. mia patients harboring a minor population of par-
Blood 2011;117(25):6876–84. oxysmal nocturnal hemoglobinuria-type cells. Blood
[90] Ueda Y, Nishimura J, Murakami Y, Kajigaya S, 2004;104(8):2425–31.
Kinoshita T, Kanakura Y, et al. Paroxysmal nocturnal [102] Hirano N, Butler MO, Guinan EC, Nadler LM, Kojima
hemoglobinuria with copy number-neutral 6pLOH in S. Presence of antikinectin and anti-PMS1 antibodies
GPI (+) but not in GPI (−) granulocytes. Eur J Haema- in Japanese aplastic anaemia patients. Br J Haematol
tol 2014;92(5):450–3. 2005;128(2):221–3.
[91] Inaguma Y, Akatsuka Y, Hosokawa K, Maruyama [103] Qi Z, Takamatsu H, Espinoza JL, Lu X, Sugimori
H, Okamoto A, Katagiri T, et al. Induction of HLA- N, Yamazaki H, et al. Autoantibodies specific to
B*40:02-restricted T cells possessing cytotoxic and hnRNP K: a new diagnostic marker for immune
suppressive functions against haematopoietic progen- pathophysiology in aplastic anemia. Ann Hematol
itor cells from a patient with severe aplastic anaemia. 2010;89(12):1255–63.
Br J Haematol 2016;172(1):131–4. [104] Goto M, Kuribayashi K, Takahashi Y, Kondoh T,
[92] Loughran TP Jr. Clonal diseases of large granular lym- Tanaka M, Kobayashi D, et al. Identification of auto-
phocytes. Blood 1993;82(1):1–14. antibodies expressed in acquired aplastic anaemia. Br
[93] Saunthararajah Y, Molldrem JL, Rivera M, Williams J Haematol 2013;160(3):359–62.
A, Stetler-Stevenson M, Sorbara L, et al. Coincident [105] de Bruin AM, Voermans C, Nolte MA. Im-
myelodysplastic syndrome and T-cell large granular pact of interferon-γ on hematopoiesis. Blood
lymphocytic disease: clinical and pathophysiological 2014;124(16):2479–86.
features. Br J Haematol 2001;112(1):195–200. [106] de Bruin AM, Demirel O, Hooibrink B, Brandts CH,
[94] Koskela HL, Eldfors S, Ellonen P, van Adrichem AJ, Nolte MA. Interferon-γ impairs proliferation of he-
Kuusanmaki H, Andersson EI, et al. Somatic STAT3 matopoietic stem cells in mice. Blood 2013;121(18):
mutations in large granular lymphocytic leukemia. N 3578–85.
Engl J Med 2012;366(20):1905–13. [107] Chen J, Feng X, Desierto MJ, Keyvanfar K, Young NS.
[95] Jerez A, Clemente MJ, Makishima H, Koskela H, Leb- IFN-γ-mediated hematopoietic cell destruction in mu-
lanc F, Peng Ng K, et al. STAT3 mutations unify the rine models of immune-mediated bone marrow fail-
pathogenesis of chronic lymphoproliferative disor- ure. Blood 2015;126(24):2621–31.
ders of NK cells and T-cell large granular lymphocyte [108] Lin FC, Karwan M, Saleh B, Hodge DL, Chan T,
leukemia. Blood 2012;120(15):3048–57. Boelte KC, et al. IFN-γ causes aplastic anemia by al-
[96] Zeng W, Kajigaya S, Chen G, Risitano AM, Nunez O, tering hematopoietic stem/progenitor cell composi-
Young NS. Transcript profile of CD4+ and CD8+ T tion and disrupting lineage differentiation. Blood
cells from the bone marrow of acquired aplastic ane- 2014;124(25):3699–708.
mia patients. Exp Hematol 2004;32(9):806–14. [109] Scheinberg P, Chen J. Aplastic anemia: what have we
[97] Poggi A, Negrini S, Zocchi MR, Massaro AM, Gar- learned from animal models and from the clinic. Se-
barino L, Lastraioli S, et al. Patients with paroxysmal min Hematol 2013;50(2):156–64.
nocturnal hemoglobinuria have a high frequency [110] Arieta Kuksin C, Gonzalez-Perez G, Minter LM.
of peripheral-blood T cells expressing activating CXCR4 expression on pathogenic T cells facilitates

 
30 2. Pathophysiology of Acquired Bone Marrow Failure

their bone marrow infiltration in a mouse model of [125] Yamaguchi H, Calado RT, Ly H, Kajigaya S, Baerlo-
aplastic anemia. Blood 2015;125(13):2087–94. cher GM, Chanock SJ, et al. Mutations in TERT, the
[111] Velasco Lezama R, Barrera Escorcia E, Munoz Torres gene for telomerase reverse transcriptase, in aplastic
A, Tapia Aguilar R, Gonzalez Ramirez C, Garcia Lo- anemia. N Engl J Med 2005;352(14):1413–24.
renzana M, et al. A model for the induction of aplastic [126] Yamaguchi H, Baerlocher GM, Lansdorp PM, Chanock
anemia by subcutaneous administration of benzene in SJ, Nunez O, Sloand E, et al. Mutations of the human
mice. Toxicology 2001;162(3):179–91. telomerase RNA gene (TERC) in aplastic anemia and
[112] Morley A, Blake J. An animal model of chronic aplas- myelodysplastic syndrome. Blood 2003;102(3):916–8.
tic marrow failure. I. Late marrow failure after busul- [127] Dror Y. Shwachman-Diamond syndrome. Pediatr
fan. Blood 1974;44(1):49–56. Blood Cancer 2005;45(7):892–901.
[113] Gibson FM, Andrews CM, Diamanti P, Rizzo S, [128] Calado RT, Graf SA, Wilkerson KL, Kajigaya S, An-
Macharia G, Gordon-Smith EC, et al. A new model of cliff PJ, Dror Y, et al. Mutations in the SBDS gene in
busulphan-induced chronic bone marrow aplasia in acquired aplastic anemia. Blood 2007;110(4):1141–6.
the female BALB/c mouse. Int J Exp Pathol 2003;84(1): [129] Calado RT, Yewdell WT, Wilkerson KL, Regal JA,
31–48. Kajigaya S, Stratakis CA, et al. Sex hormones, acting
[114] Szostak JW, Blackburn EH. Cloning yeast telomeres on the TERT gene, increase telomerase activity in hu-
on linear plasmid vectors. Cell 1982;29(1):245–55. man primary hematopoietic cells. Blood 2009;114(11):
[115] Greider CW, Blackburn EH. Identification of a specific 2236–43.
telomere terminal transferase activity in Tetrahymena [130] Ziegler P, Schrezenmeier H, Akkad J, Brassat U, Vank-
extracts. Cell 1985;43(2 Pt 1):405–13. ann L, Panse J, et al. Telomere elongation and clini-
[116] Calado RT, Young NS. Telomere maintenance and hu- cal response to androgen treatment in a patient with
man bone marrow failure. Blood 2008;111(9):4446–55. aplastic anemia and a heterozygous hTERT gene mu-
[117] Calado RT, Young NS. Telomere diseases. N Engl J tation. Ann Hematol 2012;91(7):1115–20.
Med 2009;361(24):2353–65. [131] Hahn CN, Chong CE, Carmichael CL, Wilkins EJ,
[118] Heiss NS, Knight SW, Vulliamy TJ, Klauck SM, Wie- Brautigan PJ, Li XC, et al. Heritable GATA2 mu-
mann S, Mason PJ, et al. X-linked dyskeratosis con- tations associated with familial myelodysplastic
genita is caused by mutations in a highly conserved syndrome and acute myeloid leukemia. Nat Genet
gene with putative nucleolar functions. Nat Genet 2011;43(10):1012–7.
1998;19(1):32–8. [132] Vinh DC, Patel SY, Uzel G, Anderson VL, Freeman
[119] Armanios M, Chen JL, Chang YP, Brodsky RA, AF, Olivier KN, et al. Autosomal dominant and spo-
Hawkins A, Griffin CA, et al. Haploinsufficiency of radic monocytopenia with susceptibility to mycobac-
telomerase reverse transcriptase leads to anticipation teria, fungi, papillomaviruses, and myelodysplasia.
in autosomal dominant dyskeratosis congenita. Proc Blood 2010;115(8):1519–29.
Natl Acad Sci USA 2005;102(44):15960–4. [133] Hsu AP, Sampaio EP, Khan J, Calvo KR, Lemieux JE,
[120] Vulliamy T, Marrone A, Goldman F, Dearlove A, Patel SY, et al. Mutations in GATA2 are associated with
Bessler M, Mason PJ, et al. The RNA component of the autosomal dominant and sporadic monocytopenia
telomerase is mutated in autosomal dominant dys- and mycobacterial infection (MonoMAC) syndrome.
keratosis congenita. Nature 2001;413(6854):432–5. Blood 2011;118(10):2653–5.
[121] Walne AJ, Vulliamy T, Marrone A, Beswick R, Kirwan [134] Ostergaard P, Simpson MA, Connell FC, Steward
M, Masunari Y, et al. Genetic heterogeneity in auto- CG, Brice G, Woollard WJ, et al. Mutations in GATA2
somal recessive dyskeratosis congenita with one sub- cause primary lymphedema associated with a predis-
type due to mutations in the telomerase-associated position to acute myeloid leukemia (Emberger syn-
protein NOP10. Hum Mol Genet 2007;16(13):1619–29. drome). Nat Genet 2011;43(10):929–31.
[122] Savage SA, Giri N, Baerlocher GM, Orr N, Lansdorp [135] Dickinson RE, Griffin H, Bigley V, Reynard LN, Hus-
PM, Alter BP. TINF2, a component of the shelterin sain R, Haniffa M, et al. Exome sequencing identi-
telomere protection complex, is mutated in dyskera- fies GATA-2 mutation as the cause of dendritic cell,
tosis congenita. Am J Hum Genet 2008;82(2):501–9. monocyte, B and NK lymphoid deficiency. Blood
[123] Ball SE, Gibson FM, Rizzo S, Tooze JA, Marsh JC, 2011;118(10):2656–8.
Gordon-Smith EC. Progressive telomere shortening in [136] Bigley V, Haniffa M, Doulatov S, Wang XN, Dickinson
aplastic anemia. Blood 1998;91(10):3582–92. R, McGovern N, et al. The human syndrome of den-
[124] Brummendorf TH, Maciejewski JP, Mak J, Young NS, dritic cell, monocyte, B and NK lymphoid deficiency. J
Lansdorp PM. Telomere length in leukocyte sub- Exp Med 2011;208(2):227–34.
populations of patients with aplastic anemia. Blood [137] Pasquet M, Bellanne-Chantelot C, Tavitian S, Prade
2001;97(4):895–900. N, Beaupain B, Larochelle O, et al. High frequency

 
REFERENCES 31
of GATA2 mutations in patients with mild chronic [150] Scheinberg P, Cooper JN, Sloand EM, Wu CO, Calado
neutropenia evolving to MonoMac syndrome, my- RT, Young NS. Association of telomere length of pe-
elodysplasia, and acute myeloid leukemia. Blood ripheral blood leukocytes with hematopoietic relapse,
2013;121(5):822–9. malignant transformation, and survival in severe
[138] Townsley DM, Hsu A, Dumitriu B, Holland SM, aplastic anemia. JAMA 2010;304(12):1358–64.
Young NS. Regulatory mutations in GATA2 associ- [151] Calado RT, Cooper JN, Padilla-Nash HM, Sloand EM,
ated with aplastic anemia. Blood 2012;120(21). Wu CO, Scheinberg P, et al. Short telomeres result in
[139] Ganapathi KA, Townsley DM, Hsu AP, Arthur DC, chromosomal instability in hematopoietic cells and
Zerbe CS, Cuellar-Rodriguez J, et al. GATA2 deficien- precede malignant evolution in human aplastic ane-
cy-associated bone marrow disorder differs from idio- mia. Leukemia 2012;26(4):700–7.
pathic aplastic anemia. Blood 2015;125(1):56–70. [152] Dumitriu B, Feng X, Townsley DM, Ueda Y, Yoshizato
[140] Grossman J, Cuellar-Rodriguez J, Gea-Banacloche J, T, Calado RT, et al. Telomere attrition and candidate
Zerbe C, Calvo K, Hughes T, et al. Nonmyeloablative gene mutations preceding monosomy 7 in aplastic
allogeneic hematopoietic stem cell transplantation for anemia. Blood 2015;125(4):706–9.
GATA2 deficiency. Biol Blood Marrow Transplant [153] Marsh JC, Mufti GJ. Clinical significance of acquired
2014;20(12):1940–8. somatic mutations in aplastic anaemia. Int J Hematol
[141] Cuellar-Rodriguez J, Gea-Banacloche J, Freeman AF, 2016;.
Hsu AP, Zerbe CS, Calvo KR, et al. Successful allo- [154] Ogawa S. Clonal hematopoiesis in acquired aplastic
geneic hematopoietic stem cell transplantation for anemia. Blood 2016;.
GATA2 deficiency. Blood 2011;118(13):3715–20. [155] Kulasekararaj AG, Jiang J, Smith AE, Mohamedali
[142] Walne AJ, Dokal A, Plagnol V, Beswick R, Kirwan M, AM, Mian S, Gandhi S, et al. Somatic mutations iden-
de la Fuente J, et al. Exome sequencing identifies MPL tify a subgroup of aplastic anemia patients who prog-
as a causative gene in familial aplastic anemia. Hae- ress to myelodysplastic syndrome. Blood 2014;124(17):
matologica 2012;97(4):524–8. 2698–704.
[143] Ballmaier M, Germeshausen M, Schulze H, Cherkaoui [156] Betensky M, Babushok D, Roth JJ, Mason PJ, Biegel JA,
K, Lang S, Gaudig A, et al. c-mpl mutations are the Busse TM, et al. Clonal evolution and clinical signifi-
cause of congenital amegakaryocytic thrombocytope- cance of copy number neutral loss of heterozygosity
nia. Blood 2001;97(1):139–46. of chromosome arm 6p in acquired aplastic anemia.
[144] Khincha PP, Savage SA. Genomic characterization of Cancer Genet 2016;209(1–2):1–10.
the inherited bone marrow failure syndromes. Semin [157] Scheinberg P, Young NS. How I treat acquired aplastic
Hematol 2013;50(4):333–47. anemia. Blood 2012;.
[145] Socie G, Rosenfeld S, Frickhofen N, Gluckman E, [158] Scheinberg P, Wu CO, Nunez O, Young NS. Long-
Tichelli A. Late clonal diseases of treated aplastic ane- term outcome of pediatric patients with severe aplas-
mia. Semin Hematol 2000;37(1):91–101. tic anemia treated with antithymocyte globulin and
[146] Maciejewski JP, Risitano A, Sloand EM, Nunez O, cyclosporine. J Pediatr 2008;153(6):814–9.
Young NS. Distinct clinical outcomes for cytogenetic [159] Scheinberg P, Young NS. How I treat acquired aplastic
abnormalities evolving from aplastic anemia. Blood anemia. Blood 2012;120(6):1185–96.
2002;99(9):3129–35. [160] Gupta V, Eapen M, Brazauskas R, Carreras J, Aljurf M,
[147] Ishiyama K, Karasawa M, Miyawaki S, Ueda Y, Gale RP, et al. Impact of age on outcomes after bone
Noda M, Wakita A, et al. Aplastic anaemia with marrow transplantation for acquired aplastic anemia
13q-: a benign subset of bone marrow failure respon- using HLA-matched sibling donors. Haematologica
sive to immunosuppressive therapy. Br J Haematol 2010;95(12):2119–25.
2002;117(3):747–50. [161] Socie G. Allogeneic BM transplantation for the treat-
[148] Hosokawa K, Katagiri T, Sugimori N, Ishiyama K, ment of aplastic anemia: current results and expand-
Sasaki Y, Seiki Y, et al. Favorable outcome of patients ing donor possibilities. Hematol Am Soc Hematol
who have 13q deletion: a suggestion for revision Educ Prog 2013;2013:82–6.
of the WHO ’MDS-U’ designation. Haematologica [162] Bacigalupo A, Marsh JC. Unrelated donor search and
2012;97(12):1845–9. unrelated donor transplantation in the adult aplastic
[149] Holbro A, Jotterand M, Passweg JR, Buser A, Tichelli anaemia patient aged 18–40 years without an HLA-
A, Rovo A. Comment to “Favorable outcome of pa- identical sibling and failing immunosuppression.
tients who have 13q deletion: a suggestion for revi- Bone Marrow Transplant 2013;48(2):198–200.
sion of the WHO ’MDS-U’ designation”. Haemato- [163] Deeg HJ, Amylon ID, Harris RE, Collins R, Beatty
logica 2012;97(12):1845–9. Haematologica. 2013;98(4): PG, Feig S, et al. Marrow transplants from unrelated
e46–e47. donors for patients with aplastic anemia: minimum

 
32 2. Pathophysiology of Acquired Bone Marrow Failure

effective dose of total body irradiation. Biol Blood combined with a single umbilical cord blood unit.
Marrow Transplant 2001;7(4):208–15. Blood 2015;126(23).
[164] Kojima S, Matsuyama T, Kato S, Kigasawa H, Ko- [175] Barnes DW, Mole RH. Aplastic anaemia in sublethally
bayashi R, Kikuta A, et al. Outcome of 154 patients irradiated mice given allogeneic lymph node cells. Br
with severe aplastic anemia who received transplants J Haematol 1967;13(4):482–91.
from unrelated donors: the Japan Marrow Donor Pro- [176] Frickhofen N, Kaltwasser JP, Schrezenmeier H,
gram. Blood 2002;100(3):799–803. Raghavachar A, Vogt HG, Herrmann F, et al. Treat-
[165] Maury S, Balere-Appert ML, Chir Z, Boiron JM, ment of aplastic anemia with antilymphocyte globulin
Galambrun C, Yakouben K, et al. Unrelated stem cell and methylprednisolone with or without cyclospo-
transplantation for severe acquired aplastic anemia: rine. The German Aplastic Anemia Study Group. N
improved outcome in the era of high-resolution HLA Engl J Med 1991;324(19):1297–304.
matching between donor and recipient. Haematologi- [177] Bacigalupo A, Bruno B, Saracco P, Di Bona E, Locas-
ca 2007;92(5):589–96. ciulli A, Locatelli F, et al. Antilymphocyte globulin,
[166] Viollier R, Socie G, Tichelli A, Bacigalupo A, Korthof cyclosporine, prednisolone, and granulocyte colony-
ET, Marsh J, et al. Recent improvement in outcome of stimulating factor for severe aplastic anemia: an up-
unrelated donor transplantation for aplastic anemia. date of the GITMO/EBMT study on 100 patients. Eu-
Bone Marrow Transplant 2008;41(1):45–50. ropean Group for Blood and Marrow Transplantation
[167] Bacigalupo A, Socie G, Hamladji RM, Aljurf M, Mas- (EBMT) Working Party on Severe Aplastic Anemia
chan A, Kyrcz-Krzemien S, et al. Current outcome of and the Gruppo Italiano Trapianti di Midolio Osseo
HLA identical sibling versus unrelated donor trans- (GITMO). Blood 2000;95(6):1931–4.
plants in severe aplastic anemia: an EBMT analysis. [178] Kojima S, Hibi S, Kosaka Y, Yamamoto M, Tsuchida
Haematologica 2015;100(5):696–702. M, Mugishima H, et al. Immunosuppressive therapy
[168] Marsh JC, Kulasekararaj AG. Management of the re- using antithymocyte globulin, cyclosporine, and da-
fractory aplastic anemia patient: what are the options? nazol with or without human granulocyte colony-
Blood 2013;122(22):3561–7. stimulating factor in children with acquired aplastic
[169] Ciceri F, Lupo-Stanghellini MT, Korthof ET. Haploidenti- anemia. Blood 2000;96(6):2049–54.
cal transplantation in patients with acquired aplastic ane- [179] Rosenfeld SJ, Kimball J, Vining D, Young NS. Inten-
mia. Bone Marrow Transplant 2013;48(2):183–5. sive immunosuppression with antithymocyte globu-
[170] Dezern AE, Luznik L, Fuchs EJ, Jones RJ, Brodsky RA. lin and cyclosporine as treatment for severe acquired
Post-transplantation cyclophosphamide for GVHD aplastic anemia. Blood 1995;85(11):3058–65.
prophylaxis in severe aplastic anemia. Bone Marrow [180] Scheinberg P, Wu CO, Nunez O, Scheinberg P, Boss
Transplant 2011;46(7):1012–3. C, Sloand EM, et al. Treatment of severe aplastic
[171] DeZern AE, Dorr D, Luznik L, Bolanos-Meade J, Gam- anemia with a combination of horse antithymocyte
per C, Symons HJ, et al. Using haploidentical (haplo) globulin and cyclosporine, with or without sirolim-
donors and high-dose post-transplant cyclophospha- us: a prospective randomized study. Haematologica
mide (PTCy) for refractory severe aplastic anemia 2009;94(3):348–54.
(SAA). Blood 2015;126(23). [181] Scheinberg P, Nunez O, Wu C, Young NS. Treat-
[172] Esteves I, Bonfim C, Pasquini R, Funke V, Pereira NF, ment of severe aplastic anaemia with combined im-
Rocha V, et al. Haploidentical BMT and post-transplant munosuppression: anti-thymocyte globulin, ciclo-
Cy for severe aplastic anemia: a multicenter retrospec- sporin and mycophenolate mofetil. Br J Haematol
tive study. Bone Marrow Transplant 2015;50(5):685–9. 2006;133(6):606–11.
[173] Peffault de Latour R, Purtill D, Ruggeri A, Sanz G, Mi- [182] Tisdale JF, Dunn DE, Maciejewski J. Cyclophospha-
chel G, Gandemer V, et al. Influence of nucleated cell mide and other new agents for the treatment of severe
dose on overall survival of unrelated cord blood trans- aplastic anemia. Semin Hematol 2000;37(1):102–9.
plantation for patients with severe acquired aplastic [183] Scheinberg P, Townsley D, Dumitriu B, Scheinberg P,
anemia: a study by eurocord and the aplastic anemia Weinstein B, Daphtary M, et al. Moderate-dose cyclo-
working party of the European group for blood and phosphamide for severe aplastic anemia has signifi-
marrow transplantation. Biol Blood Marrow Trans- cant toxicity and does not prevent relapse and clonal
plant 2011;17(1):78–85. evolution. Blood 2014;124(18):2820–3.
[174] Purev E, Aue G, Kotecha R, Wilder J, Khuu HM, Stron- [184] Scheinberg P, Nunez O, Weinstein B, Scheinberg P,
cek DF, et al. Excellent engraftment and long-term Wu CO, Young NS. Activity of alemtuzumab mono-
survival in patients with severe aplastic anemia (SAA) therapy in treatment-naive, relapsed, and refractory
Undergoing allogeneic hematopoietic stem cell trans- severe acquired aplastic anemia. Blood 2012;119(2):
plantation (HSCT) with Haplo-identical CD34 + cells 345–54.

 
REFERENCES 33
[185] Scheinberg P, Nunez O, Weinstein B, Scheinberg P, Bi- [196] Townsley DM, Desmond R, Dunbar CE, Young NS.
ancotto A, Wu CO, et al. Horse versus rabbit antithy- Pathophysiology and management of thrombocy-
mocyte globulin in acquired aplastic anemia. N Engl J topenia in bone marrow failure: possible clinical ap-
Med 2011;365(5):430–8. plications of TPO receptor agonists in aplastic ane-
[186] Gaber AO, First MR, Tesi RJ, Gaston RS, Mendez R, mia and myelodysplastic syndromes. Int J Hematol
Mulloy LL, et al. Results of the double-blind, random- 2013;98(1):48–55.
ized, multicenter, phase III clinical trial of Thymo- [197] Kaushansky K. The molecular mechanisms that
globulin versus Atgam in the treatment of acute graft control thrombopoiesis. J Clin Invest 2005;115(12):
rejection episodes after renal transplantation. Trans- 3339–47.
plantation 1998;66(1):29–37. [198] Zeigler FC, de Sauvage F, Widmer HR, Keller GA, Do-
[187] Feng X, Kajigaya S, Solomou EE, Keyvanfar K, Xu nahue C, Schreiber RD, et al. In vitro megakaryocy-
X, Raghavachari N, et al. Rabbit ATG but not horse topoietic and thrombopoietic activity of c-mpl ligand
ATG promotes expansion of functional CD4 + CD- (TPO) on purified murine hematopoietic stem cells.
25highFOXP3+ regulatory T cells in vitro. Blood Blood 1994;84(12):4045–52.
2008;111(7):3675–83. [199] Marsh JC, Ganser A, Stadler M. Hematopoietic growth
[188] Scheinberg P, Nunez O, Young NS. Retreatment with factors in the treatment of acquired bone marrow fail-
rabbit anti-thymocyte globulin and ciclosporin for pa- ure states. Semin Hematol 2007;44(3):138–47.
tients with relapsed or refractory severe aplastic anae- [200] Kuter DJ. Biology and chemistry of thrombopoietic
mia. Br J Haematol 2006;133(6):622–7. agents. Semin Hematol 2010;47(3):243–8.
[189] Scheinberg P, Fischer SH, Li L, Nunez O, Wu CO, [201] Feng X, Scheinberg P, Samsel L, Rios O, Chen J, McCoy JP
Sloand EM, et al. Distinct EBV and CMV reactivation Jr, et al. Decreased plasma cytokines associate with low
patterns following antibody-based immunosuppres- platelet counts in aplastic anemia and immune thrombo-
sive regimens in patients with severe aplastic anemia. cytopenic purpura. J Thromb Haemost 2012;.
Blood 2007;109(8):3219–24. [202] Olnes MJ, Scheinberg P, Calvo KR, Desmond R, Tang
[190] Marsh JC, Bacigalupo A, Schrezenmeier H, Tichelli Y, Dumitriu B, et al. Eltrombopag and improved he-
A, Risitano AM, Passweg JR, et al. Prospective study matopoiesis in refractory aplastic anemia. N Engl J
of rabbit antithymocyte globulin and ciclosporin for Med 2012;367(1):11–9.
aplastic anemia from the EBMT Severe Aplastic Ane- [203] Desmond R, Townsley DM, Dumitriu B, Olnes MJ,
mia Working Party. Blood 2012;. Scheinberg P, Bevans M, et al. Eltrombopag restores
[191] Bacigalupo A, Socie G, Hamladji RM, Aljurf M, Mas- trilineage hematopoiesis in refractory severe aplastic
chan A, Kyrcz-Krzemien S, et al. Current outcome of anemia that can be sustained on discontinuation of
HLA identical sibling versus unrelated donor trans- drug. Blood 2014;123(12):1818–25.
plants in severe aplastic anemia: an EBMT analysis. [204] Townsley DM, Dumitriu B, Scheinberg P, Desmond
Haematologica 2015;100(5):696–702. R, Feng XM, Rios O, et al. Eltrombopag added to
[192] Marsh JC, Pearce RM, Koh MB, Lim Z, Pagliuca A, standard immunosuppression for aplastic anemia ac-
Mufti GJ, et al. Retrospective study of alemtuzumab celerates count recovery and increases response rates.
vs ATG-based conditioning without irradiation for Blood 2015;126(23).
unrelated and matched sibling donor transplants in [205] Valdez JM, Scheinberg P, Young NS, Walsh TJ. Infec-
acquired severe aplastic anemia: a study from the tions in patients with aplastic anemia. Semin Hematol
British Society for Blood and Marrow Transplantation. 2009;46(3):269–76.
Bone Marrow Transplant 2014;49(1):42–8. [206] O’Donghaile D, Childs RW, Leitman SF. Blood con-
[193] Valdez JM, Scheinberg P, Nunez O, Wu CO, Young NS, sult: granulocyte transfusions to treat invasive as-
Walsh TJ. Decreased infection-related mortality and pergillosis in a patient with severe aplastic anemia
improved survival in severe aplastic anemia in the past awaiting mismatched hematopoietic progenitor cell
two decades. Clin Infect Dis 2010;52(6):726–35. transplantation. Blood 2012;119(6):1353–5.
[194] Valdez JM, Scheinberg P, Nunez O, Wu CO, Young NS, [207] Quillen K, Wong E, Scheinberg P, Young NS, Walsh
Walsh TJ. Decreased infection-related mortality and TJ, Wu CO, et al. Granulocyte transfusions in severe
improved survival in severe aplastic anemia in the past aplastic anemia: an eleven-year experience. Haemato-
two decades. Clin Infect Dis 2011;52(6):726–35. logica 2009;94(12):1661–8.
[195] Scheinberg P, Rios O, Scheinberg P, Weinstein B, Wu [208] Kohgo Y, Urabe A, Kilinc Y, Agaoglu L, Warzocha K,
CO, Young NS. Prolonged cyclosporine administra- Miyamura K, et al. Deferasirox decreases liver iron
tion after antithymocyte globulin delays but does not concentration in iron-overloaded patients with myelo-
prevent relapse in severe aplastic anemia. Am J He- dysplastic syndromes, aplastic anemia and other rare
matol 2014;89(6):571–4. anemias. Acta Haematol 2015;134(4):233–42.

 
C H A P T E R

3
Diagnosis of Acquired
Aplastic Anemiaa
A. Rovó*, C. Dufour**, A. Tichelli†
*Hematology, University Hospital of Bern, Bern, Switzerland; **Hematology Unit,
G. Gaslini Children’s Hospital; Unità di Ematologia Istituto Giannina Gaslini,
Genova, Italy; †Hematology, University Hospital of Basel, Basel, Switzerland

INTRODUCTION edge stresses thus the need of expertise to estab-


lish the diagnosis accurately without missing
Aplastic anemia (AA) is a rare, severe, non- relevant aspects which may eventually impact
malignant disease caused by autoimmune patient management.
destruction of early hematopoietic cells. AA AA is defined by the presence of pancyto-
presents with geographic rate variability, with penia with an empty bone marrow (Figs. 3.1
a global incidence rate range 0.7–7.4 cases per and 3.2), by exclusion of marrow aplasia due to
million inhabitants per year, with two- to three- direct effect of chemotherapy or radiotherapy
fold higher rates in Asia than Europe and the [3]. Diagnosis difficulty is due to the number
United States [1]. The low incidence of its oc- of marrow failure syndromes having a similar
currence as well as the overlapping with other presentation. Furthermore, acquired AA has
bone marrow failure syndromes makes its di- no specific disease markers; the bone marrow
agnosis a real challenge. The lack of experience histology showing reduced cellularity with fat
approaching such a patient can lead to unnec- replacement provides the basis of the diagnosis,
essary delay for diagnosis and treatment initia- but the final diagnosis is mainly reached by ex-
tion. During the last years, the continuous in- clusion of other entities. The better comprehen-
crease of new information has contributed to a sion of AA will lead to significant revision in
better understanding of the current landscape the near future: accelerated telomere attrition,
of bone marrow failures; we now know that acquired mutations of myeloid-related genes,
constitutional and acquired diseases are not so somatic mutations, and cytokine profiles are
clearly delineated [2]. The extent of the knowl- some of the new players highlighting profound

a
All the authors have worked on the behalf of the SAA-WP EBMT.

Congenital and Acquired Bone Marrow Failure


http://dx.doi.org/10.1016/B978-0-12-804152-9.00003-8
35 Copyright © 2017 Elsevier Inc. All rights reserved.
36 3. Diagnosis of Acquired Aplastic Anemiaa

FIGURE 3.1 Histology and immunohistology of bone marrow from a patient with SAA. (A) PAS staining of a patients
with SAA; (B) HE staining, hotspot of a patient with SAA; (C) immunostaining with CD34 of the hotspot, showing absence of
CD34 positive cells. Source: Picture courtesy of Professor Stephan Dirnhofer and Professor Alexandar Tzankov, Institute of Pathology,
University Hospital of Basel, Basel, Switzerland.

changes in the way to approach diagnosis of lymphocyte (LGL) leukemia, and autoimmune
bone marrow failures. In this chapter, we will diseases should be systematically considered.
focus on the current approach to make diagno- A congenital marrow failure syndrome, such as
sis of AA in adults, future diagnostic challenges Fanconi anemia (FA) and dyskeratosis congenita
will be also discussed. (DKC) as cause for the aplasia is more likely in
younger adults (generally below 50 years) pre-
senting with suggestive clinical findings or hav-
APPROACH TO DIAGNOSIS ing a positive family history. Patient age, family
OF APLASTIC ANEMIA history, exposure to toxic substances or medica-
ments, infections, as well as occupation, clini-
In adult patients during the diagnostic work- cal presentation, and comorbidities are relevant
up process, a number of diseases such as par- information which helps to set priorities in the
oxysmal nocturnal hemoglobinuria (PNH), hy- diagnostic process. During this process, first the
poplastic myelodysplastic syndromes (MDS), diagnosis has to be confirmed, second the mar-
hypoplastic acute leukemia, large granular row failure syndrome needs to be characterized

 
Diagnosis confirmation 37

FIGURE 3.2 Cytomorphology of an aspiration of bone marrow from a patients with aplastic anemia. (A) Hotspot with
a nest of erythroid cells; (B) reactive lymphoid infiltrate; (C) reactive plasma cells; (D) mast cells.

and finally the severity of the disease must be Complete Blood Count
settled (Fig. 3.3).
Pancytopenia is the main manifestation in
the peripheral blood, but at least two cell lines
DIAGNOSIS CONFIRMATION should be decreased for the diagnosis. In early
stages isolated cytopenia, particularly thrombo-
cytopenia can be seen. Anemia is the most fre-
Clinical Examination
quent cytopenia, this anemia is due to decreased
Clinical examination is a part of the diag- red cell production presenting with reticulocy-
nostic procedure. Except for findings related topenia. Macrocytosis is a common feature; red
to bleeding or infections, the examination pres- blood cells (RBCs) classically do not show rel-
ents mainly negative characteristics: absence of evant anisocytosis and/or poikilocytosis. The
lymphadenopathy, no enlarged spleen or liver, association with iron deficiency might generate,
and no infiltration of any other organ. Such find- however, other changes in the peripheral blood.
ings, if present, would render the diagnosis of However, iron deficiency is rare in AA, and
AA most unlikely. should suggest the presence of an active PNH

 
38 3. Diagnosis of Acquired Aplastic Anemiaa

FIGURE 3.3 Stepwise diagnostic phases of acquired aplastic anemia.

 
Diagnosis confirmation 39
clone. Leukopenia is variable, but the total leu- stromal cells, such as plasma cells, lymphocytes
kocyte count can be normal. For the evaluation forming follicles, and mast cells are frequent but
of leukocyte differentiation absolute numbers confounding findings. The increment of stromal
and not relative percentage should be analyzed. cells can mimic the picture of a marrow with
Neutropenia is frequent, it can occur at different normal cellularity, and thus, stromal cells have
degree of severity. Lymphocyte count is usually to be excluded in the global estimation for cel-
preserved. Monocytopenia can be present and lularity. The presence of nests of erythropoiesis
imposes the differential diagnosis of hairy cell [4], conforming the so-called “hot spots” are
leukemia. A careful examination of blood film frequent in AA marrow; they may show a cer-
is needed to assess morphology abnormalities tain degree of dyserythropoiesis, mainly with
of RBCs, dysplastic changes of the neutrophils, macro/megaloblastic changes (Fig. 3.2). The
the presence of erythro- and/or myelopoesis overinterpretation of this finding particularly in
precursors and of blasts, abnormal platelets, or aspirates may guide falsely to the diagnosis of
other abnormal cells, such as hairy cells. Any MDS. Megakaryocytes are usually strongly de-
of these findings would be a strong argument creased or absent. Immunostaining allows the
against the diagnosis of AA. Fetal hemoglobin identification and assessment of the topographi-
(Hb) can be increased in AA [4]; in children a cal distribution of blasts, megakaryocytes, ab-
pretransfusional increase of fetal Hb imposes normal cells, and infiltrates (Fig. 3.1). It might
the differential diagnosis of a myeloprolifera- eventually identify the unusual association with
tive/MDS like juvenile myelomonocytic leuke- lymphoma [8,9]. Fibrosis is not present in AA.
mia or other subtype of MDS [5,6]. Repeat BM biopsy, may be necessary and is rec-
ommended in any unclear case. Flow cytometry
of BM aspiration may contribute identifying
Bone Marrow Examination abnormal populations. Immunostained BM tis-
Bone marrow (BM) aspiration and biopsy are sues can be successfully mapped by multicolor
of paramount relevance to perform the diagno- immunofluorescence using confocal reflection
sis of AA. Both evaluations are complementary; microscopy. BM architecture through three-di-
aspiration allows a better discrimination of cel- mensional images can be assessed qualitatively
lular morphology, particularly by the assess- and quantitatively to appreciate the distribution
ment of dysplasia. Trephine biopsy is crucial to of cell types and their interrelationship, with
assess overall cellularity, topography of hemato- minimal perturbations. Confocal microscopy is
poietic cells, and abnormal infiltrates. A BM bi- currently only used for basic laboratory investi-
opsy containing at least five to six intertrabecu- gation and their potential contribution for clini-
lar spaces, providing a core of 20–30 mm length cal diagnosis in BM failure is promising [10].
is considered as representative [7]. Dry tap is
unusual and suggests diagnoses other than AA.
Cytogenetic
Overall cellularity is more often reduced rather
than completely absent. Aplastic and hypocel- Cytogenetic abnormalities can be present in
lular BM is defined by <10% (empty marrow) up to 12–15% of otherwise typical AA patients;
and 30% of hematopoietic cells, respectively. therefore, cytogenetic investigations should be
This cut off has been established mainly for chil- systematically performed in all AA patients [11].
dren and young adults. Diagnostic in elderly Due to hypocellular bone marrow, it frequently
patients is discussed later in this chapter. Beside occurs that there are insufficient metaphases for
the variable amounts of residual hematopoietic an adequate analysis. In such cases, FISH tar-
cell, there are prominent fat spaces. Increase in geting specific anomalies should be considered.

 
40 3. Diagnosis of Acquired Aplastic Anemiaa

Most frequent anomalies in AA include trisomy to IST, and had longer and a higher rate of over-
8, uniparental disomy of the 6p (6pUPD) [12], all and progression-free survival; in contrast,
5q-, anomalies of chromosome 7 and 13. Abnor- mutations in a subgroup of genes that includ-
mal cytogenetic clones often are small at diag- ed DNMT3A and ASXL1 were associated with
nosis, and may arise during the course of the worse outcomes. The pattern of somatic clones
disease or may be transient and disappear after in individual patients was however variable
immunosuppression (IST) [13,14]. While abnor- and frequently unpredictable [19]. The emerg-
malities of chromosome 7 and 5 even in absence ing data showing clonal hematopoietic expan-
of dysplasia turn the diagnosis more likely in sion in pediatric and young adult AA patients is
MDS, other cytogenetic findings are less cat- strikingly different from healthy hematopoiesis.
egorical. An abnormal cytogenetic clone does It also seems that between pediatric patients
however not necessarily imply the diagnosis and older adults, there are potential differenc-
of MDS or AML. Del(13q) has been reported in es in the mutational spectrum since adults are
patients with MDS and other hematologic ma- more likely to carry age-related somatic muta-
lignancies cases [15] as well as in patients with tions associated with malignancy [20]. In clinical
bone marrow failure syndrome. Patients pre- practice, the finding of somatic mutations tends
sented with del(13q) were reported mainly asso- to be interpreted as signature for malignancy;
ciated with good response to IST [16,17]. Trans- this belief might be true for certain mutations,
formation to MDS and AML may occurs usually but not necessary for all of them. Further data
many months to years after the diagnosis of AA, are needed to clarify the mutational profiles and
therefore cytogenetic monitoring during follow- disease outcomes in AA.
up is recommended.
Telomere Length Measurement
Molecular Analysis Telomere attrition offers an interesting prog-
Molecular analysis including next-genera- nostic tool in acquired AA. Telomere shortening
tion sequencing (NGS) is increasingly used to in AA patients was associated with both, numer-
understand disease pathophysiology. Kulasek- ical and structural chromosome abnormalities.
araraj et al. postulated that somatic mutations Patients with shorter telomeres were at higher
are present in a subset of AA and might pre- risk of malignant transformation. Shorter aver-
dict malignant transformation. Clonal hema- age telomere lengths inversely correlated with
topoiesis was identified in a fifth of evaluated monosomy 7 at diagnosis [21]. Thirteen SAA
AA patients showing specific gene mutations patients were analyzed for acquired mutations
associated with transformation to MDS [18]. A in myeloid cells at the time of evolution to −7
larger report evaluating 668 blood samples ob- and all had a dominant hematopoietic stem and
tained from 439 patients with AA showed that progenitor cell clones bearing specific acquired
somatic mutations in myeloid cancer candidate mutations. Mutations in genes associated with
genes were present in one third of the evaluated MDS/AML were however present in only four
AA patients. Furthermore clonal hematopoiesis cases. Patients who evolved to MDS and AML
was detected in 47% of the patients, most fre- showed marked progressive telomere attrition
quently as acquired mutations. The prevalence before the emergence of −7 [22]. This result
of the mutations increased with age, and muta- might have clinical implications because affected
tions had an age-related signature. This study individual may benefit from therapies that even-
also showed that mutations in PIGA and BCOR tually eliminate the shortest (and dysfunction-
and BCORL1 correlated with a better response al) telomeres. Telomere length measure, unless

 
Characterization of aplastic anemia 41
there is a suspicion of congenital bone marrow failure. To exclude FA, tests to demonstrate in-
failure does not belong yet to standard screen- creased sensitivity to chromosomal breakage
ing in acquired AA. Due to constraints on health with mitomycin C or diepoxybutane should be
care budgets telomere length measurement will performed. All patients with bone marrow fail-
be not systematically covered by insurance. In- ures who are candidates to hematopoietic stem
clusion of patients in clinical trials addressing cell transplantation should undergo this test,
this aspect is strongly recommended. since conditioning regimen in FA patients has to
be adapted because of the defective DNA repair
mechanisms, and therefore the higher suscepti-
HLA-Typing bility to chemotherapy. However, even for IST-
Human leukocyte antigen (HLA) typing of the treated patients it is reasonable to perform the
patient and his family belongs to the diagnostics tests, first in order to be aware due to the higher
of patients with marrow failure syndrome. To- risk of secondary cancers in these patients and
day, HLA typing should no longer be restricted second because many of them do not respond to
to children and younger adults. Indeed, early standard IST with antithymocyte globulin (ATG)
knowledge of the HLA-type of the patient and and cyclosporine (CSA), but sometimes show
identification of a possible sibling donor allows response to androgens. Screening should also
to include early transplantation in the treatment include sibling donors of FA patients. Telomere
decision process. Even patients without sibling shortening is a consistent and typical finding of
donor and older fit patients should be nowadays telomeropathies [29]. Telomere length measure-
HLA-tested. Matched unrelated donor trans- ment of leukocytes from peripheral blood can
plantation looks consistent front line option in be performed as screening test by suspicion of
children [23]. Wider applicability of alternative- a telomeropathy. TERC and TERT gene muta-
donor transplantation for AA is under investiga- tions cause telomeropathies in both children and
tion this will be discussed in several chapters of adults [30,31]. In adult patients, symptoms and
the book [24,25]. clinical signs are often milder than in children,
mucocutaneous findings and other physical
anomalies are infrequent. The pattern of organ
abnormalities is extremely variable among af-
CHARACTERIZATION fected individuals. Indeed, a family history with
OF APLASTIC ANEMIA blood count abnormalities or hematologic dis-
ease is often lacking. Given the higher frequency
Exclusion of Congenital Bone of congenital bone marrow failure syndromes in
Marrow Failures children, an age tailored diagnostic work up for
A positive family history including other AA in pediatric age is included in Chapter 11.
members affected with cytopenia or malignan-
cy, suggests an inherited bone marrow failure Differential Diagnosis of AA From the
syndrome. The presence of unusual clinical fea-
Hypocellular Variant of MDS
tures (liver, lung, bone disease) should alert the
possibility of a congenital form of bone marrow The distinction between AA and hypoplastic
failure. However, a normal clinical examination MDS is certainly the most difficult diagnostic
does not definitively rule out “cryptic” telome- task. Both diseases present with markedly hy-
ropathies [26,27] or a nonclassical FA [28]. Bone pocellular bone marrow, and increased fat cells
marrow morphology in such a disease cannot [32,33]. Dysplasia of erythropoiesis may be pres-
be distinguished from acquired bone marrow ent in both entities and therefore may not help

 
Another random document with
no related content on Scribd:
occur from asphyxia from the second to the fourth day near the
beginning of an outbreak or the illness may last twenty days, after
the more susceptible birds have been killed off.
In investigating a series of outbreaks of roup in chickens in
America, Dr. V. A. Moore found a non-motile bacillus allied to the
colon bacillus which proved much more deadly to rabbits and guinea
pigs than to chickens, and which was not found in the blood nor
internal organs but only in the local lesions where inoculated. The
disease tended to assume a chronic type in place of the acute form as
seen in Europe. Three inoculated chickens escaped the disease
altogether. It would appear therefore that we have here a disease
distinct from that described by Lœffler, or that there was an absence
of some unknown predisposing or contributing conditions that were
present in the European outbreaks. In both diseases however
infection is an undoubted factor and similar measures of prevention
and even of treatment may be followed.
Prevention. The first consideration is the seclusion of flocks from
outside animals in affected localities. Newly purchased birds or those
returning from a poultry show should be placed in strict quarantine
for a few weeks until the absence of infection shall have been
demonstrated. Different flocks should not be allowed to mingle, nor
the members of a healthy flock to wander where the manure of
another flock has been laid. Birds having diarrhœa, or any discharge
from eyes, nose or beak, or any false membrane on such parts should
be excluded from the flock, and the house and yard disinfected. It
should not be forgotten that rabbits, guinea pigs and mice may be
bearers of the infection, and that it may be introduced on the feet of
dogs or their masters. Pigeons, buzzards, and carrion crows are
especially dangerous as possible bearers of the infection. Cleanliness
as regards food and water, buildings and yards is of vital importance.
Treatment. The sick birds should be strictly secluded and handled
by a special attendant. When the lesions appear on visible mucosæ
they should be painted several times a day with a 5 per cent. solution
of phenol, or a saturated solution of boric acid, or salicylic acid,
potassium permanganate, iodine, or some other germicide may be
used. For the bowel affection one or two drops phenol in water may
be given daily, and the drinking water should be slightly charged
with the same. Cleanliness, pure air, warmth, dryness and sloppy
food are all important.
ACUTE CATARRHAL ENTERITIS IN
SOLIPEDS.
Definition. Causes: Irritants swallowed, debility, improper, insufficient food,
congestions, parasitisms, impaired innervation or circulation, iced water, chills,
perspirations, fatigue, hot, damp weather, overfeeding, cryptogams, bacteria,
newly harvested fodder, septic, or fermented food, leafy fodder, toxins, stagnant,
septic water, lack of pepsin, muriatic acid and bile, diseased teeth or jaws or
salivary glands. Lesions: Gastritis, congestion of small intestine and colon, in striæ,
thickening, ecchymosis, ulceration, necrosis, excess of mucus with pus, villi,
follicles and glands swollen. Symptoms: Fever, high colored urine, costiveness,
coated tongue, red eyes, inappetence, sluggishness, emaciation, weakness,
unthriftiness, colics, rumbling, diarrhœa; or more fever, suffering, anorexia,
icterus, hurried breathing, pleuritic ridge, arched back, tender abdomen, rumbling,
flatus, diarrhœa, critical or bloody, anxiety, debility, prostration, collapse.
Prognosis. Treatment: In mild cases, careful diet, and laxatives with antiferments,
in severe cases, laxatives, anodynes, antiseptics, demulcents, stimulants of
peristalsis, enemata, counter-irritants, fomentations, compresses, mustard, in
profuse diarrhœa antiseptics, anodynes, demulcents, calomel and chalk, bismuth,
astringents, boiled flour or starch, gums. Dieting during convalescence.
Definition. Inflammation of the intestinal mucosa.
Causes. Irritants of all kinds taken in with food, or as medicine or
otherwise and acting on the mucosa. Debilitating conditions (chronic
disease, starvation, overwork, close indoor life) which lower the tone
of the system at large, and local debilitating conditions like coarse,
dry, fibrous, innutritious food, congestions, parasitisms, impaired
innervation and troubles of the circulation are strongly predisposing.
Drinking iced water may operate by lowering the tone of the
intestines but seems to habitually act rather by inducing reaction and
congestion. Chills of the surface, especially when perspiring and
fatigued, act in the same way. The relaxation and atony attending on
long continued hot weather, predisposes to enteritis, but is doubtless
even more injurious by the abundance of ferments which it
propagates in food and water.
Overfeeding and stimulating aliments thrown on an alimentary
canal in such an atonic condition become especially hurtful. The
injury, however, comes most commonly from food that contains an
excess of cryptogams or bacterial ferments or from water similarly
charged. Newly harvested fodders in which the microbes are still in a
state of vigorous life, when added to the poisonous principles in
certain immature seeds (leguminosæ, gramineæ, etc.); fodders that
have undergone fermentative changes (rotten potatoes, turnips,
musty hay or oats); fodders that are leafy and harbor an excess of
microbes (alfalfa, sainfoin, cowpea, clover) are especially dangerous
at times. If musty or otherwise altered they often contain besides,
dangerous toxins.
In taking into account the fungi and microbes in spoiled foods, we
need not give exclusive attention to the particular species of microbe
present. An extended observation shows that the same ferments may
be present in the dry, well cured, wholesome fodder, and in the
musty or spoiled specimen, the main difference being in the excess
found in the latter case as compared with the former. With the excess
too, there is always present a large amount of toxins, ptomaines and
other more or less poisonous products, which, acting on the
intestinal mucosa or even on the system at large, tend to reduce its
vitality and to lay it open to the attacks of bacteria which had
otherwise remained perfectly harmless. Porcher and Desoubry,
Achard and Phulpin and Wurz have shown experimentally that
intestinal microbes can enter the chyle and blood from even a
healthy bowel. The streptococcus of pneumoenteritis equi of Galtier
and Violet appears to be a common fodder and intestinal microbe,
which has become pathogenic, because of its excess or on account of
a lack of resistance on the part of the animal. In the same way the
various intestinal cocci and the common colon bacillus may become
pathogenic when the normal antagonism of the bowels and their
contents is lessened.
In the same way stagnant and septic water may be harmless to one
animal of great vigor and good tone and pathogenic to another which
lacks these qualities; or the excess of the ferment and its toxins may
overcome the natural resistance of the animal. The lack of the
natural antiferments of the intestine, pepsin and hydrochloric acid,
on the one hand and bile on the other, will also conduce to
multiplication of the microbes and their products, so that they can
successfully attack the mucous membrane.
Other accessory causes operate more or less, thus any impairment
of the process of mastication, through diseased teeth or jaws tends to
the escape of undigested food through the stomach, as a specially
favorable culture media for the microbes, and irritants of the
mucosa.
Lesions. As the disease very often implicates the stomach (gastro-
enteritis), the usual lesions of gastritis will be seen. Most commonly
the lesions are best marked in the small intestine, and again in other
cases in the colon, but usually there is more or less change in all
parts of the intestinal canal. The small intestine is almost devoid of
aliments and the mucosa deeply congested in patches or striæ, with
at points thickening, softening so that it crushes under the finger,
hemorrhagic discoloration, and even ulceration, and necrotic
changes. It is covered with a layer of mucus, thin and mucilaginous
or thick and glutinous, containing many granular and pus cells. The
villi are swollen, the follicles of Lieberkuhn puffed up, and the
agminated and solitary glands widely dilated, filled with exudate, and
surrounded by an area of congestion. Proliferation of small, round
cells has produced embryonic tissue in the mucosa and especially
between the glands.
Symptoms. In the mildest form there is hyperthermia, thirst,
insensible loins, scanty, high colored urine, costive bowels, a few
small pellets only being passed at a time, hard, dry and covered with
a mucous film, hot, clammy mouth, coated tongue, with redness
along the edges and tip, yellowish red eyes, impaired appetite, dull,
sluggish habit, a tendency to hang back on the halter, and a steady
loss of flesh and increased dryness and unthriftiness of the coat.
Slight, intermittent colics occurring especially after meals and
attended by loud rumbling of the bowels are marked features. This
may be followed by slight relaxation of the bowels and recovery in
about a week, unless it should become complicated by intestinal
indigestion or impaction, or should merge into the acute form.
In the more intense forms all symptoms are aggravated. There is
anorexia and even refusal of water, dullness and prostration are well
marked, the head carried low, and the gait is unsteady. The mouth is
hot with tenacious mucus, and a fœtid odor; the tongue is furred and
red at the tip and margins, the eye is sunken, the conjunctiva icteric,
the face pinched, and the pulse accelerated. Hyperthermia may reach
104° F. Breathing may be almost normal, or with fever, may become
rapid and accompanied by a pleuritic ridge on the flank. The back is
slightly arched and rigid, the belly drawn up and tender, after meals
it may be tympanitic, and colics set in or are aggravated, pawing,
uneasy movements of the hind limbs, and lying down to rise again
shortly, with frequent looking at the flanks being noticeable.
Defecations are at first abundant and coated with mucus, later the
balls are small and scanty and expelled with much effort. From the
first the everted rectum is of a very deep red. Toward the end of the
first day or later the intestinal rumbling increases, flatus passes
freely, and diarrhœa may set in and prove critical. This usually
indicates disease in the colon and tends to recovery; it may be
entirely absent if the inflammation is confined to the duodenum, the
effused liquid being re-absorbed from the cæcum and colon. If the
diarrhœa should prove critical there is a return of appetite and spirit,
the fæcal discharges become firmer and recovery takes place in a
week. If, however, the diarrhœa becomes more profuse and bloody,
the colics more intense, the eyes more sunken and hopeless, the face
more pinched and anxious, and the temperature reduced to or below
the normal, with great weakness and debility, the near approach of
death may be feared. This state of collapse may be further marked by
extreme coldness, or dropsy of the limbs, increased icterus, hurried
breathing and rapid loss of flesh.
A prominent icterus indicates implication of the liver from the
ascent of the infecting germs through the bile ducts, or the passage of
microbes or their products or both through the portal vein. In either
case it is a serious complication.
Prognosis. In its uncomplicated form the disease is not very fatal
to vigorous, mature horses, though more trying to the young. If
infective germs or their products implicate the liver producing
marked jaundice, or if the general system is poisoned by the
microbes or their toxins, producing marked depression and
prostration the danger is enormously enhanced.
Treatment. In the mildest cases a limitation of the food to
moderate bran mashes, and a dose of ½ lb. of sodic sulphate, with
salicylate of soda (3–4 drs.) or bismuth will usually suffice.
In severe cases, at the outset, while constipation exists give 3 or 4
drs. of cape aloes, or ½ lb. Glauber salts or ½ pint olive oil,
combined with 2 drs. of extract of hyoscyamus or belladonna and 3
drs. salicylate of soda. This serves to deplete from the inflamed
vessels and the whole portal system, to soothe suffering, to expel
much of the offensive and infective matters from the bowels, and to
check fermentation in that which remains. They should be given
with, or followed by mucilaginous liquids like solutions of slippery
elm or gum arabic, flaxseed tea, or well boiled farinaceous gruels.
Pilocarpin, 3 grs., or eserine, 2 grs., or both have been
recommended and may be resorted to when action of the bowels is
urgently demanded. They need not supersede the other laxatives. In
manifest impaction of the large intestines, salts, aloes, pilocarpin and
eserine may form an effective combination.
Copious enemata with mucilaginous liquids or warm soap suds
should be given at frequent intervals.
Counter-irritants and derivatives to the abdomen are most
important. Hot fomentations may be persisted in for an hour at a
time, or a damp compress around the abdomen covered closely by
dry blankets and held in place by elastic circingles. Mustard pulp
made with cold water rubbed in against the hair and at once covered
by paper and a thick blanket is often of great value as drawing blood
and nervous action to the skin and relieving the suffering intestine.
In all cases the diet and drink must be carefully supervised. A little
thoroughly scalded wheat bran, or farina, and decoctions of flaxseed,
farinas, slippery elm or mallow, or a solution of gum arabic will
refresh the animal without overloading the digestive organs or
favoring further fermentation.
In case of the onset of diarrhœa which threatens to prove excessive
and persistent, the giving by mouth and anus of antiseptics and
anodynes with mucilaginous agents may be resorted to. Calomel may
be given in 10 grain doses twice daily mixed with five times the
amount of chalk. Or 2 drs. each of nitrate of bismuth and salicylate of
soda and ½ oz. of laudanum may be given three times a day. Or
quinine, 2 drs. and nux vomica 10 grs. may be added to the above. A
choice may be made of other anodynes, (hyoscyamus, belladonna),
antiseptics, (salol, chloral, naphthol, naphthalin, creolin), and
bitters, (gentian, calumba, cascarilla).
Antiseptic and even astringent injections must be given, and well
boiled farinas and mucilaginous agents may be given by the mouth.
Wheat flour boiled for several hours; starch prepared with boiling
water as for the laundry, (1 pint); gum arabic, or slippery elm may
suffice as examples.
The patient should have a dry comfortable box and warm clothing
according to the season of the year. He must be kept for a week on
linseed gruel or other equally simple demulcent agent and brought
back to his customary food by slow degrees.
CHRONIC CATARRHAL ENTERITIS IN
SOLIPEDS.

Causes: As in acute: troubles of circulation, heart, lungs, verminous embolism,


parasitism, skin disease. Lesions: thickening of mucosa, pigmentation, rigidity,
hypertrophy of villi, follicles and glands, ulceration, polypi. Symptoms: impaired
appetite, buccal fœtor, retracted flank, unthrifty skin, pallid mucosæ, colics,
tympanies, rumblings, irregular bowels, emaciation, perspiration, fatigue.
Treatment: dietetic, tonic, bitters, salines, aromatics, enemata, bismuth,
laudanum, calomel and chalk, iron, astringents, counter-irritants, electricity,
sunshine.

Causes. This may occur from a continuance of the same causes as


in the acute, or from an imperfect recovery from the acute form. It
may result from troubles in the circulation, as valvular disease of the
heart, or emphysema of the lungs, which forces the blood back on the
venous system, including the liver and portal vein. Or the lesions that
come from verminous embolism may leave such alteration in the
intestinal walls as entail chronic congestion of the mucosa, or
intestinal parasites may be the cause. Severe and inveterate skin
diseases appear to affect the intestinal mucosa by sympathy, just as
diseases of that mucosa usually entail skin diseases.
Lesions. Attenuation of the coats of the small intestine and
thickening of the mucosa of the large have been noticed. The mucosa
is darkly pigmented and covered with excess of mucus. The
thickening of the mucosa may extend into the submucous tissue,
giving a firm leathery feeling to the part, and entailing a loss of
elasticity. The villi are hypertrophied and the follicles of Lieberkuhn
and Peyers’ patches may be congested, ulcerated or otherwise
altered. Polypoid growths are not uncommon on the mucosa, and the
mesenteric glands are enlarged and pigmented.
Symptoms are by no means very definite. Disturbance of the
digestive functions, capricious or impaired appetite, dry fœtid
mouth, tucked up abdomen, dry hair and skin, pallor of the visible
mucous membranes, slight intermittent colics and tympanies, loud
rumblings in the bowels, and relaxed bowels, or alternate costiveness
and diarrhœa, with some tenderness on manipulation of the
abdomen are the usual symptoms. The animal loses flesh, has dry,
unthrifty coat, and sweats and is easily exhausted at work.
Treatment. Dietary care is the first essential. Boiled oats, barley,
rye or bran, in small amount and flaxseed tea may indicate the kind.
These should be given in small amount often, and at regular
intervals.
A failing appetite may be stimulated by nux vomica (10 grains)
twice daily, or by gentian or other bitter, along with common salt and
aromatics.
Constipation may be combated by fresh green food in small
quantities, or by an ounce each of Glauber salt and common salt
given every morning before feeding, in a drink of water (half to a
bucket, if possible), and 10 to 20 grains of nux vomica may be
advantageously added. Soapy injections with salt or glycerine may
also be given.
Diarrhœa may be moderated or checked by nitrate of bismuth (2
drachms), with laudanum (1 ounce), repeated as may be demanded.
A combination of calomel and chalk (1:12) will often serve a good
purpose in drachm doses several times a day. For persistent
diarrhœa Cadeac recommends the following: Iron carbonate 4
drachms, lime water 10 ounces, alum 1 drachm, powdered oak bark 1
ounce, given in water and farina.
Sepsis and fermentation must be combated by the same means as
in the acute type, and the same counter-irritants may be resorted to.
A life in the open air or sunshine, but without undue exertion is of
great importance.
ACUTE CATARRHAL ENTERITIS IN CATTLE.
Causes: atony, debility, starvation, overfeeding, innutritious food, close, foul
buildings, ill health, over-exertion, hot weather, sudden changes, chills, privation
of water, irritants, spoiled and newly harvested grain, foul water, parasitism, chest
diseases, thrombosis. Lesions; in small intestine mainly, tympany, congestion,
thickened mucosa, epithelial degeneration, desquamation, enlarged villi, follicles
and glands, erosions, ulcers, perforations. Symptoms: solid masses in rumen,
impaired rumination and appetite, rumbling, tenderness, costiveness, fever,
arched back, tender, tucked up abdomen, colics, in severe cases, agalactia, tremors,
rigors, drooping head, ears, eyelids, tender abdomen, straining, expulsion of
mucus, foul eructations, later diarrhœa, critical or exhausting. Death from
tympany, bleeding, infection, inanition. Diagnosis: by concurrence of symptoms,
hyperthermia, tender abdomen, no blood nor coccidia in stools, no frothy bloody
mucus with tenesmus. Treatment: dietetic, friction, synapism, atropia, chloral
hydrate by rectum, salines, demulcents, nux, tartar emetic, eserine, pilocarpin,
sulphites, salol, sodium salicylate, naphthol, etc., bitter tonics, carminatives,
stimulants, sodium chloride, ipecacuan, hygiene during convalescence.
Causes. As in solipeds the various conditions which lower the
general tone, and those which especially debilitate the bowels
predispose to catarrhal inflammation. Underfeeding and
overfeeding, fibrous, innutritious, indigestible food, an indoor life in
close, foul stables, chronic and debilitating diseases, overwork,
overdriving, long continued hot weather, sudden changes of weather,
chills, long railway journeys without water, exposure in hot
stockyards in midsummer, all lessen the resisting power of the
system and of the bowels.
As more direct irritants, may be named irritant plants, weedings
and culls from gardens, musty and spoiled fodders of all kinds, newly
harvested grain, and putrid drinking water. Also intestinal
parasitism, diseases of the heart and lungs, and thrombosis of the
mesenteric arteries.
Lesions. These predominate in the small intestine in catarrhal
enteritis as they do in the large intestine in dysentery. The small
intestine and cæcum may be distended by gas, and reddened more or
less deeply on their outer surface. The mucosa is the seat of
congestion, punctiform and ramified redness, thickening, infiltration
and softening so that the epithelium breaks down into a pulp under
the pressure of the finger. Desquamation may be extensive leaving a
raw angry surface. The villi are infiltrated, erect, and ulcerated
showing dark bloody points, and ecchymoses, and circumscribed
sloughs and eschars are present. The solitary glands are congested,
hypertrophied and projecting. The submucosa is infiltrated with a
gelatinoid material and the same may be found around the swollen
and congested mesenteric glands. Perforations have been met with in
some cases, and coexistent inflammatory lesions in the stomachs are
common.
Symptoms. In the mildest forms there is inactivity of the rumen,
aggregation of the contents into hard masses, easily felt through the
surrounding gases, appetite and rumination are greatly impaired,
and there is much rumbling and considerable tenderness of the right
side of the abdomen, and more or less costiveness, with hard, glazed
mucus-covered fæces. There is some rise of temperature, ardent
thirst, injected mucous membranes, dry, hot muzzle, weeping eyes, a
small, hard, weak pulse, arched back, tender to pinching, and tucked
up abdomen. There may be slight colicy pains, uneasy movements of
the hind feet and tail, and sometimes lying down and rising at short
intervals.
In more severe cases the impaction and tympany of the rumen are
more marked, the hyperthermia runs high, appetite and rumination
cease, the milk dries up, rigors and tremors appear, the head and
ears droop, the eyes are sunken, the mouth is clammy and fœtid, the
colicy pains are severe or extreme, the right side of the abdomen is
very tender, defecation may be altogether suspended and rumbling
in the right side of the abdomen ceases or becomes rare. Straining
may continue but seldom is anything but mucus passed. Eructations
from the rumen are distinctly fœtid.
After the third day the violence of the pains may abate, and
sometimes diarrhœa sets in and may be regarded as critical, and
portending recovery. If rumbling in the right side is resumed, if the
fever subsides, the spirits revive, and some appetite and rumination
return they will herald improvement.
If on the other hand the pulse becomes smaller, the temperature
higher, the eyes sunken and fixed, the urine scanty, red and acid, the
animal constantly recumbent on its left side, if when raised it omits
the healthy stretching of its hind limbs, and walks sluggishly and
painfully with frequent moaning, if when down it rests its head on
the ground, the prospects are very unfavorable.
Death may occur early from tympany and asphyxia; it may follow
profuse intestinal hæmorrhage; or it may be the result of general
infection and inanition.
Diagnosis must depend on the combination and succession of the
above-named symptoms. From acute intestinal congestion it is
distinguished by the more moderate type of the colic, and the more
gradual advance of the disease. From acute indigestion and tympany
of the rumen by the early and marked tenderness of the right side of
the abdomen, and the decided hyperthermia. From hemorrhagic
enteritis by the absence of the black sanguineous discharges from the
bowels at an early stage of the malady, and of coccidia from the
droppings. From dysentery it is distinguished by the absence of the
mucous and bloody discharges, which are passed with much
straining in that affection from the beginning.
Treatment. The first consideration is dietetic and hygienic. If the
animal will still eat, he ought to have boiled flax seed or other well
boiled gruel, rendered palatable by salt. Even if he refuses food, this
may be diluted largely and will be taken on account of the thirst. If he
refuses all, a bottle may be given at intervals to refresh him. Or better
—milk may be given from a bottle in the same way. Active friction to
the abdomen with straw, or the application of oil of turpentine or
mustard may abstract blood to the skin and favor the restoration of
the intestinal functions. To calm the pains and control spasms,
sulphate of atropia (½ gr.) may be given subcutem and repeated if
there is no action on the pupil in fifteen minutes. Or extract of
belladonna (2 drs.), or chloral hydrate (½ oz.) may be given by rectal
injection.
To overcome the intestinal torpor 1 lb. each Glauber and common
salt may be given in four to six quarts of warm water and followed by
frequent mucilaginous drinks, as much as the animal will take, but
only two or three quarts at a time. The addition of ½ dr. nux vomica
will serve to rouse peristaltic action. Harm advises 1½ dr. tartar
emetic by rectal injection for the same end. Next to Glauber salts,
Castor oil (1 qt.) is to be recommended. Along with these or
independently of them sulphate of eserin (1½ gr.) or pilocarpin (2
gr.) may be employed subcutem.
Frequent rectal injections of soap or mucilaginous liquids, with or
without laxatives will be useful.
As antiferments beside the salt may be used bisulphite of soda in
½ oz. doses, salol 3½ drs., salicylate of soda 3 drs., betol 3½ drs., or
naphthol 3½ drs., by the mouth and rectum.
When free movement of the bowels has been secured, attempts
should be made to restore appetite and rumination by tonics and
stimulants: gentian ½ oz., nux vomica ¼ dr., ipecacuan 2 drs.,
common salt 1 oz., may be given three times a day.
The diet should at first be restricted to flax seed gruel or that of
other farinas, with a mere handful of fresh grass or bran mash and
the restoration of the previous diet should be slow and gradual, care
being taken meanwhile that no costiveness of the bowels supervenes.
CHRONIC CATARRHAL ENTERITIS IN
CATTLE.
Causes: As in acute chest diseases, abdominal tuberculosis. Lesions: Thinning,
discoloration, degeneration of mucosa, fœtid, mucous contents, black baked
masses, lymph glands pigmented. Symptoms: Impaired appetite, irregular bowels,
tympanies, lies with nose on right flank, unthrifty coat, prostration, emaciation,
weakness, tender flank. Treatment: Dietetic, laxative, stimulant of peristalsis,
bitters, antiseptics, aromatics, muriatic acid, treat concurrent disease.
Causes. This may result from a continuance of the causes that are
operative in the acute, or from the latter merging into the chronic
form. Chronic diseases of the heart and lungs, local disturbances of
the circulation, and tumors or tubercles of the intestines or
mesentery are additional causes.
Lesions. These embrace attenuation of the intestinal walls at
Peyers’ patches, a dark, slaty discoloration of the mucosa, more or
less congestion, an accumulation of fœtid mucus in the small
intestines, of mucus and black baked fæcal matters in the large, and
discoloration of the mesenteric glands. Inter-dependent diseases of
the heart, lungs and liver are not uncommon.
Symptoms. Following the acute form there remain impaired or
capricious appetite and rumination, costiveness alternates with
relaxation of the bowels, intermittent slight tympanies occur, the
subject inclines to lie much with his nose in his flank, has dull coat,
erect on back and neck, sunken eyes, drooping ears, and rapidly loses
flesh and strength. Tenderness of the right side of the abdomen when
the fist or knee is pressed into it is a marked feature.
Treatment. The diet must be cared for as in the acute form, yet
fresh green grass, a little at a time, is calculated to stimulate appetite
and rumination and to prove laxative to the bowels. The same
purgatives may be given in one-fourth the doses and repeated daily
or reduced as may be found best to secure a moderate secretion and
discharge from the bowels; eserine or pilocarpin may be used for the
same purpose; the bitters and antiseptics may be given in the same
way. As calmative aromatics, oil of peppermint 30 drops, powdered
anise ½ ounce, or ginger ½ ounce, may be given twice or thrice
daily.
Cadeac strongly recommends a drink slightly acidulated with
hydrochloric acid to assist the digestion and stimulate the stomach to
action.
Attention must of course be given to any curable concurrent or
inter-dependent disease.
DYSENTERY OF CATTLE.
Definition. Attacks ox mainly. Causes: accessory causes, chills, rain storms, night
dews, hoar frost, foul or iced water, alimentary irritants, spoiled fodder, over
exertion, hot damp weather, odors of carrion, crowding, swamps, foul stables,
germs or pathogenic ferment, in man catarrhal, diphtheritic and amœbic, amœba
dysenterica, other microbes, effect of better hygiene. Symptoms: attack sudden,
languor, trembling, weakness, weeping eyes, fever, buccal epithelial softening,
erosions, tenesmus, fœtid, liquid stools, involuntary defecation, hemorrhoidal
congestion, open anus, colics, tender right flank, splashing on handling, anorexia,
salivation, unthrifty skin, hide-bound, cracked muzzle, later prostration, low
temperature, sunken glazed eyes, drooping head, ears, eyelids, weakness,
emaciation, alkaline, fœtid, frothy, bloody, mucous stools, with sloughs, saliva
acid, gastric liquids alkaline, bile suppressed. Duration: three days to three weeks
or chronic. Mortality 50 to 80 per cent. Complications: mostly septic, abscess,
gangrene of other organs, lungs, joints, glands, etc. Lesions: rapid sepsis, blood
deep red, coagulum loose, venous congestion, large intestines congested, tumefied,
softened, desquamated, eroded, sloughing, necrotic, folds perforated, cicatrizing,
contents mucopurulent, bloody, putrid, microbes, glandular lesions, implication of
small intestines, stomach mouth, liver, spleen, hepatic abscess. Diagnosis: from
rinderpest by tardiness and comparative weakness of contagion, absence of general
mucous congestion and epithelial concretions, from toxic enteritis by same.
Prevention: avoidance of causes, separation of sick, disinfection, careful feeding.
Treatment: Demulcents, antiseptics, astringent tonics, opiates, ipecacuan, calomel,
sodium sulphate with antiseptics, antiseptic enemata of glycerine, phenol, creolin,
iron sulphate, silver nitrate, salicylic and boric acids, rest, gravitation, careful
dieting.
Definition. An infective, ulcerative inflammation of the large
intestine but especially of the colic and rectal mucosæ.
It is well recognized as a disease of the ox, and the older writers
allege its existence also in horses, swine and dogs, though this is not
admitted in all modern veterinary works.
Causes. It was formerly ascribed to improper hygiene, chills, cold
rain storms, night dews, hoar frost, malarious emanations, putrid,
stagnant or iced water, irritants in food, green, fermented or musty
food, a too liberal diet after starvation, overtaxation in very hot
weather, and bad odors from decomposing carcasses. These can only
be accepted as predisposing causes begetting a general debility, or
debility of the alimentary canal and laying that open to the attacks of
specific microbes. The close aggregation of cattle on ship-board, in
besieged cities, and in the parks of armies in the field, has apparently
contributed to the propagation of the dysentery. The removal of a
victim to a herd with free healthy range seldom starts a new center of
the disease. In all infected herds huddled in small compass there is
every facility for the propagation of a germ already present, and
especially in the commissariat of a belligerent army there are enough
privations, over-exertions and other trying conditions to favor
predisposition. Faulty food like stale bread, musty hay, have been
supposed to cause it. For man and beast alike dysentery is
preëminently a disease of the tropics, and of hot seasons, and will
often subside on the advent of cold weather.
Its propagation on given (swampy) soils, and in particular (foul)
stables strongly suggests a special germ, though for the cow this has
not been perfectly identified. Gerlach vainly attempted to inoculate
it, and it does not often propagate itself beyond the foul and infected
localities or stables, yet its persistence in them for years bespeaks
unequivocally the operation of a special pathogenic ferment.
It may also be fairly assumed that it is not necessary that the same
factor should be present in all cases alike, but that one operates
predominantly in one case and another in another. In other words
dysentery must be recognized as not one disease, but several, of
which the true pathogenic microbes have not yet been fully
demonstrated, but which are classed together because of the
similarity of the attendant lesions.
In man three distinct forms are recognized. 1. Catarrhal
dysentery, with frequent small stools of rosy mucus, and blood; and
later pus, scybala, passed with tenesmus, but no sloughs and little
odor; 2. Diphtheritic dysentery, with thin watery bloody
discharges having a pronounced cadaveric odor; also tenesmus,
sloughs, and increasingly offensive smell; 3. Amœbic dysentery
with frequent bloody mucus stools, tenesmus sloughs and fœtor, but
with distinct remissions or intermissions. With the latter, amœbæ
are found abundantly and more so in the more acute cases with
alkaline stools. They are found in the fresh warm stools, 5 to 8 times
the size of a red blood globule and oval, pyriform or irregular in
form, with nucleus and nucleolus. Kartulis and Hlava succeeded in
inducing dysentery in cats and dogs by injecting pure culture of the
amœba, and the former testifies that dogs in Egypt take the disease
spontaneously, and their stools contain the same amœba coli as is
found in man.
Cunningham who investigated the subject in India found amœba
in the bowels of healthy men, and also abundantly in the fæces of
horses and cows, which have naturally the requisite alkaline reaction.
The mere presence of the amœba therefore may not be sufficient to
cause the disease, but with the requisite predisposition and an
alkaline condition of the intestinal contents, it is manifestly an
important factor in the disease.
The causative microbes in other forms of dysentery have not been
identified, but under the requisite irritation and local debility one
can easily conceive of the ordinary bacterial ferments of the intestine,
concurring with others introduced from without, in determining the
morbid condition. With better hygiene the disease is steadily
diminishing in man and beast, though violent epizootics (in cattle)
still appear in connection with wars (siege of Belfort, 1870, Zundel),
and carriage by sea in hot climates (Mediterranean trade, Bouley).
Symptoms. The disease sets in suddenly, yet prodromata may
occasionally be observed, such as dullness, langor, trembling over
the flanks and elbows, weakness, prominent, weeping, congested
eyes, and low moans when moved. Then follow hyperthermia, at first
slight, heat of the mouth without injection, epithelial concretions or
erosions and diarrhœa (sometimes there is straining without passage
at first). Then follows a period of profuse and fœtid discharge, with
relaxed or open anus, the liquids escaping involuntarily and
smearing the tail, perineum, thighs and hocks, and the protruded
mucosa showing dark red congestions and even commencing
erosions. Colicy pains, slight at first, have now become intense, and
the right side of the abdomen is very tender to the touch and
fluctuates noisily when manipulated. Appetite and rumination are
lost. Salivation may be present, the saliva falling in films to the
ground. The buccal epithelium is softened, loosened and easily
detached by the finger, leaving raw sores. The temperature which has
risen slowly (not as in rinderpest abruptly) may reach 106°. The hair
becomes dry, the skin harsh, rigid, and firmly adherent to the deeper
parts, and often cold, while the muzzle is hot, dry and even cracked.
At a still more advanced stage, the pulse is small, the temperature
lowered, the animal very weak and unsteady and inclined to lie, great
emaciation, sunken glazed eye, drooping head, ears and eyelids, and
a general fœtor from the skin as well as the dejections, which attracts
crowds of flies. By this time there may be passed only bloody mucus
mixed with eschars, and having a most repulsive odor.
It is a noticeable fact that the fæces are alkaline, and in man the
saliva is acid and destitute of its glycogenic properties, the stomach
secretions are alkaline and no longer peptogenic, and the secretion of
bile is arrested until improvement sets in.
Course. Duration. Some mild cases recover in two or three days,
and in violent cases death may occur at this early date. More
commonly the disease continues for two or three weeks before
ending in death or recovery. Some merge into the chronic form and
may last for months and die in a condition of marasmus. These last
cases become mere walking skeletons, with pallid mucosæ, sunken
eyes, scurfy hide-bound skins covered with vermin, and the
frequently everted rectum is congested and covered with ulcers and
eschars. Mortality is from 50 to 80 per cent.
Complications. Most complications are in the direct line of septic
infection. Among the most common are hepatic abscess, gangrenous
pneumonia, and extensive gangrene of the intestinal walls. In man
arthritis, paralysis, parotitis and other secondary affections are seen.
In the animal as well we may expect necrotic centres in any organ,
from the supervention of a general septicæmia.
Lesions. The carcasses putrefy with extraordinary rapidity being as
a rule pervaded by septic microbes and their toxins. The blood is of a
deep red, loosely and imperfectly coagulated, and accumulated in the
veins and subcutaneous connective tissue.
The large intestines are the special seat of the disease, the walls
being found hyperæmic, with concentration especially on the mucous
surface which is red, congested, infiltrated, tumefied so that it is
easily detached under pressure by the finger, or broken down into a
putrid pulp. At other points the epithelium or the whole mucosa has
been detached leaving ulcers, varying from mere erosions to the deep
and even perforating sores, through which the putrid contents escape
into the abdominal cavity. At other points the surface is covered by
necrotic masses surrounded by a swollen margin of living mucosa.

You might also like