Anda di halaman 1dari 14

The n e w e ng l a n d j o u r na l of m e dic i n e

Review Article

Dan L. Longo, M.D., Editor

Aplastic Anemia
Neal S. Young, M.D.​​

A 
plastic anemia is a disease with a long history. The first case From the Hematology Branch, National
description was published by Paul Ehrlich in 1888, the term “anemia aplas- Heart, Lung, and Blood Institute, Bethesda,
MD. Address reprint requests to Dr.
tique” originated with Louis Henri Vaquez in 1904, and clinical features Young at the Mark Hatfield Clinical Re-
were described by Richard C. Cabot and other pathologists in the early 20th cen- search Center, 10 Center Dr., Bethesda, MD
tury. What was once a terrifying, mysterious disease, most often sudden in onset 20892-1202, or at ­youngns@​­mail​.­nih​.­gov.

and occurring in young persons, can now be successfully treated in almost all N Engl J Med 2018;379:1643-56.
patients. An understanding of the pathophysiology of aplastic anemia, gained in DOI: 10.1056/NEJMra1413485
Copyright © 2018 Massachusetts Medical Society.
the research laboratory, has guided the development of effective therapies. Marrow
failure syndromes have been linked to viral infection and environmental toxins,
inherited and acquired genetic mutations, early events in leukemogenesis, and the
hematopoiesis of normal aging.

Defini t ions
The long history of aplastic anemia has resulted in confusing terminology. The
term “anemia” derives from the early ability to measure red cells in a hematocrit.
Most patients with aplastic anemia have pancytopenia, with decreased platelets,
white cells, and erythrocytes. “Aplastic” refers to the failure of marrow to form
blood, and hematopoietic failure is the end-organ effect of diverse pathophysiolog-
ical mechanisms. Yet a seemingly “empty” bone marrow may be entirely capable
of supporting normal hematopoiesis. Conversely, bone marrow failure can occur
without low marrow cellularity, as in the myelodysplastic syndromes and paroxys-
mal nocturnal hemoglobinuria (PNH).

Pathoph ysiol o gic a l Fe at ur e s


There are three main pathophysiological mechanisms that can lead to an “empty”
bone marrow (Fig. 1).

Direct Damage to Marrow


Damage to the bone marrow occurs most often iatrogenically, from chemotherapy
or radiation therapy. The effects are dose dependent and transient at conventional
doses. Other organ systems are affected, and spontaneous recovery is expected.
Benzene also impairs hematopoiesis, and industrial exposure to benzene figured
prominently in the early literature on aplastic anemia. Benzene exposure is now a
negligible risk factor, accounting for only a small fraction of cases of marrow
failure in most countries.1,2 However, in China, a country characterized by rapid
industrialization and slower regulation, benzene remains a workplace toxin.3,4
Dosage is critical; workers with less intense or less prolonged exposure to benzene
appear to have milder cytopenias, and they recover after termination of the expo-
sure. Marrow failure is a proximate effect, not a late consequence, of benzene
exposure.

n engl j med 379;17 nejm.org  October 25, 2018 1643


The New England Journal of Medicine
Downloaded from nejm.org at The University of Texas at El Paso on October 24, 2018. For personal use only. No other uses without permission.
Copyright © 2018 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

Idiopathic
Cytotoxic drugs Telomere disease
Seronegative hepatitis
CAUSES Radiation Fanconi anemia
Eosinophilic fasciitis
Benzene Other germline gene mutations
Thymoma

PATHOPHYSIOLOGY Chemical and physical damage Immune destruction Constitutional genetic defects

APLASTIC ANEMIA

PATHOLOGY NORMAL BONE MARROW

Supportive care Immunosuppressive therapy Sex hormones


Hematopoietic growth factors Hematopoietic stem-cell Hematopoietic stem-cell
TREATMENTS transplantation transplantation
Hematopoietic stem-cell
transplantation Eltrombopag

Figure 1. Diverse Pathophysiological Features that Lead to a Common Pathologic Process in Bone Marrow Failure.
Replacement of hematopoietic cells by fat is the distinctive pathologic feature of aplastic anemia. A hypocellular marrow can result from
chemical or physical damage; the most frequent causes are iatrogenic, such as after cytotoxic chemotherapy or irradiation and exposure
to benzene, usually in industry. In certain constitutional genetic defects that affect hematopoietic stem-cell function and the immune
system, marrow failure and a hypocellular marrow are prominent. The disease immune aplastic anemia results from destruction, mainly
by T cells, of hematopoietic stem cells and progenitor cells. Treatments are different for marrow damage, genetic defects that produce
multiorgan effects, and the immune disease.

Constitutional Syndromes mune regulation, such as in those due to cyto-


Marrow failure results from specific loss-of- toxic T-lymphocyte–associated antigen 4 (CTLA-4)
function germline mutations, usually inherited mutations8 and deficiency of adenosine deami-
(Table 1 and Fig. 2A). There is a spectrum of nase 2 (DADA2).9 Constitutional syndromes are
genetic lesions that diminish the capacity for classically manifested in childhood, often with
hematopoietic stem cells to repair DNA, as in characteristic physical anomalies; typically, mul-
Fanconi anemia (replication-dependent removal tiple organs are involved. The family history may
of interstrand DNA cross-links)5 and dyskeratosis disclose affected relatives. However, genetic and
congenita (telomere maintenance and repair),6 or genomic testing has revealed that genetic defi-
interfere with differentiation and self-renewal ciencies can appear in adults, who may present
pathways, as in GATA2 deficiency.7 Marrow fail- without these typical features. Recognition of a
ure also may occur in syndromes affecting im- germline cause is critical for guiding therapy

1644 n engl j med 379;17 nejm.org October 25, 2018

The New England Journal of Medicine


Downloaded from nejm.org at The University of Texas at El Paso on October 24, 2018. For personal use only. No other uses without permission.
Copyright © 2018 Massachusetts Medical Society. All rights reserved.
Aplastic Anemia

Table 1. Constitutional Marrow Failure Syndromes in Adults.*

Syndrome Hematologic Presentation Clinical Features Genetic Features Pathophysiological Features


Telomere diseases SAA in childhood; MAA, Early hair graying, pulmonary DKC1, TERT, TERC, Deficient telomere repair
macrocytic anemia, fibrosis, hepatic cirrhosis RTEL1, other rare ­(telomerase enzyme complex),
thrombocytopenia mutations ­inadequate telomere protection
in adulthood (shelterin proteins)
Fanconi anemia SAA in childhood; rare Short stature, café-au-lait 17 FANC genes Deficient repair of interstrand DNA
­presentation as bone spots, skeletal and uro­ cross-links
marrow failure, MDS, genital anomalies
AML in adulthood
GATA2 deficiency SAA, MDS, AML Persistent and unusual infec- GATA2 Unknown
tion (e.g., warts)
CTLA4 deficiency AA with low IgG Intestinal disease, adenopathy, CTLA4 Immune de-repression
infection, autoimmunity

* AA denotes aplastic anemia, AML acute myeloid leukemia, MAA moderate aplastic anemia, MDS myelodysplastic syndrome, and SAA severe
aplastic anemia.

and has consequences for family members. Fatal not have a clear cause and have been labeled
graft rejection has followed inadvertent use of a idiopathic.
graft from an affected sibling10 and persistent Immune aplastic anemia lies on a spectrum
marrow failure after transplantation in patients of bone marrow and blood-cell diseases (Fig. 2B).
with mutations in the gene encoding the growth Cytotoxic T cells have been the focus in studies
factor thrombopoietin.11 Results of published of samples from affected patients and in vitro
surveys from specialty clinics are influenced by studies. These cells appear to be functionally and
referral patterns and case definitions. In a study phenotypically activated,18,19 skewed to produce
involving 98 children and adults with aplastic type 1 cytokines (e.g., interferon-γ),20,21 induce
anemia, 5% had genetic mutations on screening.12 apoptosis through Fas and the Fas ligand,22 and
Of 173 patients with bone marrow failure of circulate as oligoclones.23 Acquired, somatic mu-
suspected inherited origin, most of whom were tations in the signal transducer and activator of
under 18 years of age, about 50% had muta- transcription 3 (STAT3) signaling pathway may be
tions.13 At the National Institutes of Health, pathogenic in some cases of aplastic anemia, as
among children and adults referred for protocol they are in large granular lymphocytosis, producing
treatments, unexpected pathogenic mutations constitutively activated T cells.24 Regulatory T (Treg)
were very unusual in patients with severe aplastic cells are decreased in patients with aplastic ane-
anemia but were much more prevalent in those mia and increase with a hematologic response.25,26
with moderate bone marrow failure (Rodrigues F: Aplastic anemia is associated with specific
personal communication). histocompatibility antigens.27 The presence of
“escape clones” (granulocytes with loss of the
Immune Aplastic Anemia region of chromosome 6 that encompasses HLA
Almost all sporadic cases of aplastic anemia, alleles) in 10 to 15% of patients is striking28,29;
especially when severe and acute, appear to be cells selected because of the absence of HLA,
immune-mediated. The strongest, most relevant acquired by 6p loss of heterozygosity (LOH) or
evidence for an immune mechanism is improve- somatic mutations, sustain hematopoiesis by
ment in blood counts after a variety of immuno- means of clonal expansion.30 Immune escape has
suppressive therapies and dependence of adequate also been hypothesized to explain clonal expan-
counts after recovery on a maintenance calci- sion of “PNH cells” in aplastic anemia; red cells
neurin inhibitor (usually cyclosporine).14 Aplastic and white cells deficient in glycosylphosphoino-
anemia is associated with immunologic diseases sitol (GPI)–anchored proteins originate from a
(particularly seronegative hepatitis,15 eosinophilic stem cell with an acquired mutation in PIGA
fasciitis,16 and thymoma17), but most cases do (phosphatidylinositol glycan class A).31 It has been

n engl j med 379;17 nejm.org  October 25, 2018 1645


The New England Journal of Medicine
Downloaded from nejm.org at The University of Texas at El Paso on October 24, 2018. For personal use only. No other uses without permission.
Copyright © 2018 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

A
Paroxysmal
nocturnal
hemoglobinuria
Fanconi anemia
GATA2
RUNX1
CTLA4
Large granular
MPL APLASTIC lymphocytosis
ANEMIA

Telomere
disease
Myelodysplastic Acute myeloid leukemia
syndromes
Hypocellular
myelodysplastic
syndrome

B
B-CELL IMMUNITY

Autoimmune Large granular


hemolytic
anemia lymphocytosis

Autoimmune Immune Myelodysplastic


neutropenia APLASTIC
thrombocytopenia ANEMIA syndromes
purpura
Pure red-cell
aplasia
Pure white-cell aplasia
T-CELL IMMUNITY

Paroxysmal
nocturnal Hemophagocytic
hemoglobinuria lymphohistiocytosis
Amegakaryocytic thrombocytopenia

SINGLE LINEAGE PANCYTOPENIA

Figure 2. Aplastic Anemia in Relationship to Other Diseases.


The Venn diagram in Panel A highlights the overlap of aplastic anemia, both diagnostically and pathophysiologically, with paroxysmal
nocturnal hemoglobinuria, myelodysplastic syndromes, and constitutional marrow failure syndromes. Many clinical and pathophysiolog-
ical features of aplastic anemia parallel those of other immune-mediated diseases in which a single organ is targeted. Panel B shows the
spectrum of immune cytopenias. The dominant immune effectors are delineated on the y axis; for example, autoimmune destruction of
peripheral-blood cells is mainly antibody-mediated, whereas T cells have been implicated in marrow destruction. However, the immune
response is almost certainly complex in all these diseases. (For schematic purposes, the circles in each Venn diagram have been scaled
to the relative numbers of PubMed citations.)

1646 n engl j med 379;17 nejm.org October 25, 2018

The New England Journal of Medicine


Downloaded from nejm.org at The University of Texas at El Paso on October 24, 2018. For personal use only. No other uses without permission.
Copyright © 2018 Massachusetts Medical Society. All rights reserved.
Aplastic Anemia

suggested that the GPI anchor itself is a target functional changes. Clonal populations may be
of the immune response.32 Autoantibodies of un- easier to detect in patients with marrow failure
certain clinical relevance have been identified by than in healthy persons with many more active
high-throughput screening of serum samples,33,34 stem cells. Mutated clones need not be malig-
but the inciting antigen (or antigens) for the nant. In aplastic anemia, benign clonal popula-
dominant T-cell response remains unknown. tions of granulocytes deficient in GPI-anchored
Immune aplastic anemia can be modeled in proteins or lacking HLA expression are com-
mice: infusion of mismatched donor lympho- mon, presumably selected for their survival un-
cytes leads to rapid hematopoietic failure and der immune attack. Indeed, healthy persons have
death.35,36 Limited numbers of T cells specifi- tiny numbers of leukocytes with PIGA mutations,
cally attack marrow cells, causing apoptosis and chromosomal clonal mosaicism is present in
through Fas engagement; type 1 cytokines have many normal tissues. Clonal evolution in aplas-
an active role in targeted cell death, directly for tic anemia is development of the myelodysplastic
interferon-γ and indirectly for tumor-necrosis syndrome (MDS) or acute myeloid leukemia
factor α (TNF-α); and Treg cells are inhibitory. (AML), usually characterized by aneuploidy, es-
pecially loss of all or a portion of chromosome 7.
Hem at op oie sis Abnormalities of chromosome 7 are a feature of
both acquired39 and constitutional40 aplastic ane-
Stem-Cell Number mia, suggesting that the environment of marrow
Aplastic anemia has long been regarded as the failure itself confers a predisposition to their
result of a profound deficit in hematopoietic selection.
stem and progenitor cells.37 The marrow is de- With next-generation sequencing, clonality is
void of morphologic precursors to erythrocytes, detected in leukocytes with a mutation in a spe-
granulocytes, and platelets. Cells bearing the cific gene. In most targeted genomic studies,
antigen CD34, which contain stem and progeni- mutations are queried in about 50 “candidate”
tor cells, are almost completely absent in fixed genes (i.e., genes known to be recurrently mu-
biopsy specimens or on flow-cytometric analysis. tated in MDS and AML). (These mutations are
Colony-forming cells for differentiated lineages acquired, not inherited, and are present in a
and more immature multipotent cells are ex- hematopoietic stem cell and its progeny.41) Such
tremely low in number. None of the available clonal populations are present in about one third
measurements of hematopoiesis accurately quan- of patients with aplastic anemia, but in contrast
tify stem cells, and they also do not correlate to MDS and AML, a very limited set of genes
closely with blood counts. A return to normal (DNMT3A, ASXL1, and BCOR) is involved, and the
blood counts and bone marrow function after clone size (variant allele fraction) is small.42,43
immunosuppression, and even more dramatically Mutated clones are associated with prognosis
with growth factor stimulation, indicates the (favorable with BCOR and PIGA and unfavorable
presence of residual stem cells in an “empty” with DNMT3A and ASXL1).42 Clones containing
bone marrow. mutations in these genes can be stable over a
period of many years,42 and mutated clones in-
Stem-Cell Clonality frequently appear to drive evolution to a myeloid
Hematopoiesis in aplastic anemia is clonal, but cancer.44,45
this is not a well-defined term.38 Cancer is
clonal: a tumor is derived from a single malig- Telomeres
nant cell. Normal hematopoiesis is also clonal: Extremely short telomeres are a typical finding
a single stem cell gives rise to many differenti- in patients with a genetic telomere disease. In im-
ated progeny. In a tumor or leukemia, mutations mune aplastic anemia, telomere length may be
in specific genes alter cell function and in com- decreased as a result of increased mitotic demand
bination produce the malignant phenotype. Muta- on a limited pool of stem cells.46 Telomere length
tions also accumulate in normal cells as “pas- at diagnosis has correlated with outcomes,45,47
sengers,” in genes that do not lead to gross response to immunosuppression,48 and evolution

n engl j med 379;17 nejm.org  October 25, 2018 1647


The New England Journal of Medicine
Downloaded from nejm.org at The University of Texas at El Paso on October 24, 2018. For personal use only. No other uses without permission.
Copyright © 2018 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

to MDS and AML.47 Accelerated telomere attrition Hypoplastic Myelodysplastic Syndrome


precedes progression to monosomy 7.44 versus Aplastic Anemia
Acquired mutations are detected on genomic
screening of recurrently mutated genes in MDS
Di agnosis
and AML. Such testing is valuable when MDS is
The fatty bone marrow remains a basic diag- suspected. Hypocellular MDS may be suggested
nostic feature of aplastic anemia, but sophisti- from the appearance of the bone marrow, espe-
cated testing can now be directed at distin- cially dyspoietic megakaryocytes,51 and a normal
guishing among diverse pathophysiological or increased number of CD34 cells is not consis-
disorders and discriminating among similar, tent with aplastic anemia. Flow cytometry enu-
sometimes overlapping diseases in the differ- merates CD34 cells and identifies anomalous
ential diagnosis (Fig. 2B). Accurate diagnosis is phenotypes indicating aberrant differentiation.52
required for appropriate therapy and effective Genomic testing may be useful, since spliceo-
management. some gene mutations and multiple mutated genes
are characteristic of MDS but not of aplastic
Constitutional versus Acquired Bone anemia.53 However, the genomic pattern of hypo-
Marrow Failure plastic MDS — involvement of specific genes,
Genomic screening complements functional test- the likelihood of a single gene mutation, and
ing for Fanconi anemia (indicated by chromo- clone size — is both similar to the pattern in
somal damage after clastogenic stress) and aplastic anemia and distinct from that in typical
telomeropathies (indicated by shortened telo- persons and patients with normocellular and
meres). However, comprehensive germline screen- hypercellular types of MDS.54 The finding of a
ing adds to the cost of the evaluation, and the DNMT3A- or ASXL1-mutated clone does not alter
results may not be available to the clinician for the diagnosis of aplastic anemia or the likeli-
several weeks. Screening for the approximately hood of a response to therapy.
50 genes that cause constitutional marrow fail-
ure is particularly valuable in moderate and chron- PNH and Aplastic Anemia Syndrome
ic pancytopenia, thrombocytopenia, and macro- Screening for PNH is performed by means of
cytic anemia; in children and adolescents; and in flow cytometry, which precisely measures the
patients in whom immunosuppressive therapy proportion of GPI-anchored, protein-deficient
has failed. In patients with severe pancytopenia erythrocytes and leukocytes. In classic hemolytic
who do not have a family history, clinical fea- PNH, the PNH clone is large, above 50% and
tures (Table 1), or evidence of organ involvement sometimes approaching representation of all
beyond the marrow, screening is not likely to be circulating cells from the mutated clone. A large
positive. Commercial testing reports “patho- clone correlates with an increased risk of cata-
genic” mutations, a determination based on strophic venous clot and is an indication for
amino acid changes and their location in con- anticomplement therapy with eculizumab, which
served or functionally critical regions of a gene, corrects intravascular hemolysis and is effective
and requires continual reannotation of the litera- prophylaxis against thrombosis. Clones are gen-
ture for genotype–phenotype correlations. Some erally small in aplastic anemia, requiring moni-
base substitutions are infrequent polymorphisms toring but not treatment. Clinical PNH usually
in certain ethnic populations, and their clinical does not develop from tiny clones or in the ab-
meaning may be uncertain. Conversely, exome sence of a clone at diagnosis.55
sequencing of candidate genes may not detect
critical mutations in regulatory regions.49,50 Cor-
T r e atmen t s
relation of genomic screening with functional
testing is desirable, but some patients with Approaches to the treatment of aplastic anemia
telomeropathy have normal telomere length, in children and adults are shown in Figure 3.
short telomeres not below the first percentile
can be difficult to interpret, and mosaicism Bone Marrow Transplantation
due to reversion of a Fanconi anemia gene can Replacement of failed bone marrow is curative
lead to a normal chromosomal study in periph- of the underlying disease. Historically, transplan-
eral blood. tation has been limited by its complications

1648 n engl j med 379;17 nejm.org  October 25, 2018

The New England Journal of Medicine


Downloaded from nejm.org at The University of Texas at El Paso on October 24, 2018. For personal use only. No other uses without permission.
Copyright © 2018 Massachusetts Medical Society. All rights reserved.
Aplastic Anemia

A Children (<18 yrs) B Adults


Diagnosis and support (days): <40 yrs and
Control infection HLA-matched
Transfusions sibling
HLA-matched
sibling
Immunosuppressive therapy Allogeneic bone marrow
+ eltrombopag transplantation

Immunosuppressive therapy Allogeneic Response Treatment


Definitive therapy (weeks): +/– eltrombopag bone marrow (6 mo) failure
Restore blood counts transplantation
Response Treatment
(3–6 mo) failure

Treatment
Second failure Matched unrelated donor
Alternative donor hematopoietic immunosuppressive or haploidentical bone
stem-cell transplantation therapy marrow transplantation
(matched unrelated donor,
umbilical cord blood) Response
Long-term monitoring
(months to years): Relapse
After immunosuppressive
Observation Observation Observation Observation
therapy: Recurrent relapse Evolution to
Monitor for relapse or evolution to −7, MDS, or AML
and evolution −7, MDS, or AML
After bone marrow
transplantation:
Manage graft-versus-host
disease, infection, and
late complications

Figure 3. Treatment Algorithms for Patients with Immune Aplastic Anemia.


Treatment algorithms are shown for children (Panel A) and adults (Panel B). The algorithms are intended for guidance. Age thresholds
for transplantation are approximate, since clinical decisions in individual patients often depend on multiple factors, such as history, co-
existing conditions, and HLA matching. For simplicity, coexisting conditions are not incorporated in the figure, despite their importance.
Since hematologists supervise treatment, some secondary and tertiary treatment branches have also been omitted for simplicity. In ad-
dition, treatment strategies are not fixed because of developing results. Eltrombopag is likely to be incorporated into first-line treatment
and, as a result, will be used less frequently for disease that is refractory to immunosuppressive therapy. Some alternative donor ap-
proaches (transplantation from a matched unrelated donor and transplantation of umbilical cord blood) are supported by substantial
evidence; other alternative approaches — in particular, transplantation from haploidentical donors — are promising but are based on
much less experience, and long-term results have not yet been reported. Shading indicates the stages of treatment: days to diagnose
severe pancytopenia and stabilize the patient’s condition; weeks to months to select, implement, and complete definitive therapy; and
years of monitoring for responses and complications. The ability to manage severe neutropenia56 is fundamental for good long-term
outcomes. Undesirable delays from diagnosis to transplantation should be avoidable when outcomes of immunosuppressive therapy
are clear in 3 to 6 months. Patients who do not have a response to immunosuppressive therapy57-59 or who have complications60 can do
well with second-line transplantation. MDS denotes myelodysplastic syndromes.

— graft rejection and graft-versus-host disease in older adults, the results have not improved
(GVHD) — and the need for suitable donors. for several decades, with a survival rate of about
Expanded donor options are major recent ad- 50% for recipients older than 40 years of age70
vances. and a relative risk of death that is almost three
For immune aplastic anemia in a young pa- times as high for older adults as it is for chil-
tient, transplantation is always the preferred dren in registry data.71 Black patients also have
treatment. When transplantation is undertaken poorer outcomes than white patients.72 Marrow
expeditiously after diagnosis, with the use of a is the preferred source, since the risk of GVHD
graft from a histocompatible sibling donor, the is higher with the use of peripheral blood.73,74
results are excellent, with a long-term survival Rabbit antithymocyte globulin (ATG) is often
rate of more than 90% among young children61,62 added to the conditioning regimen,64,75 and radia-
and more than 80% among adolescents63 and a tion therapy, especially in children, is avoided.
low rate of short- and long-term complications Histocompatible sibling donors are unavail-
(Table 2). Although transplantation of grafts able for most patients, but large donor registries
from sibling donors has become more common provide the option of an HLA match with an

n engl j med 379;17 nejm.org October 25, 2018 1649


The New England Journal of Medicine
Downloaded from nejm.org at The University of Texas at El Paso on October 24, 2018. For personal use only. No other uses without permission.
Copyright © 2018 Massachusetts Medical Society. All rights reserved.
1650
Table 2. Hematopoietic Stem-Cell Transplantation for Severe Aplastic Anemia.*

No. of Overall Acute and Graft


Study Transplant Source and Recipient Status Patients Age Conditioning and Prophylaxis Survival Chronic GVHD† Failure

yr % % %
IBMTR prospective RCT, MFD — 50% of recipients had no previ- 70 Median, 23 Conditioning: Cy, ATG 80 at 5 yr Acute: 11 16
1994–200164‡ ous treatment Prophylaxis: CsA, MTX Chronic: 32
King’s College retrospective MFD — most recipients had no previous 100 Median, 18 Conditioning: Alemtuzumab+Cy 90 at 5 yr Acute: 29 9
study, 1999–200965§ treatment; MUD — most recipients Prophylaxis: CsA for MSD, FLU+Cy Chronic: 3
had refractory disease for MUD, FLU+Cy+TBI for mis-
matched MUD
EGBMT registry, children, MFD — no previous treatment 396 Range, 0–12 Conditioning: mainly Cy, some 87 at 3 yr Acute: 8 2
The

2000–200961 Cy+FLU Chronic: 6


Prophylaxis: CsA±ATG±MTX
EGBMT registry, adolescents, MFD — no previous treatment 394 Median, 15 Conditioning: mainly Cy, some 86 at 3 yr Acute: 12 8
2000–200963 Cy+FLU±ATG Chronic: 8
Prophylaxis: mostly MTX+CsA, some
CsA+MMF
EGBMT registry, 2005–200966 MFD — previous recipient treatments not 940 50% >20 — 83 at 5 yr Acute: 13 9
described Chronic: 6
EGBMT registry, 2005–200966 MUD — previous recipient treatments 508 53% >20 — 76 at 5 yr Acute: 26 9
not described Chronic: 11
French national prospective UCB — refractory SAA 26 Median, 16 Conditioning: FLU+Cy+ATG+TBI 85 at 2 yr Acute: 46 12
study, 2011–201567 Prophylaxis: CsA Chronic: 36
n e w e ng l a n d j o u r na l

The New England Journal of Medicine


of

JSHCT registry, 2001–201268 UCB — refractory adult SAA 69 Median, 49 Conditioning: mainly 69 at 3 yr Acute: 32 29
FLU+melphalan+low-dose TBI Chronic: 21
Prophylaxis: MTX or MMF±
glucocorticoids±CIN

n engl j med 379;17 nejm.org  October 25, 2018


Eurocord and EBMT retrospec- Sibling UCB with or without bone marrow 20 Median, 5.6 Conditioning: variable but mainly 81 at 7 yr Acute: 6 10

Copyright © 2018 Massachusetts Medical Society. All rights reserved.


tive registry, 1988–201469 — mainly for refractory disease Cy+FLU Chronic: 8
m e dic i n e

Prophylaxis: variable but mainly


CsA+glucocorticoids

* ATG denotes antithymocyte globulin, CIN calcineurin inhibitor, CsA cyclosporine, Cy cyclophosphamide, EBMT European Society for Bone Marrow Transplantation, EGBMT European
Group for Bone Marrow Transplant, FLU fludarabine, GVHD graft-versus-host disease, IBMTR International Bone Marrow Transplant Registry, JSHCT Japan Society for Hematopoietic
Cell Transplantation, MFD matched family donor, MSD matched sibling donor, MTX methotrexate, MUD matched unrelated donor, RCT randomized, controlled trial, TBI total-body ir-
radiation, and UCB umbilical cord blood.
† Data for acute GVHD are for grades II, III, and IV disease. Data for chronic GVHD are usually for grades II, III, and IV (for extensive disease) and occasionally for all chronic GVHD.
‡ This RCT compared Cy alone with Cy plus ATG. There were no significant differences between the conditioning regimens with respect to outcomes. For simplicity, only data for the
group that received CTX plus ATG are shown, since this is the more common conditioning regimen.
§ Only data from alemtuzumab-treated patients are shown; MSD and MUD are combined, as they were in the original report.

Downloaded from nejm.org at The University of Texas at El Paso on October 24, 2018. For personal use only. No other uses without permission.
Aplastic Anemia

unrelated donor at molecular resolution for most ment for children.92 In Europe, with an average
white patients.76-78 In a comprehensive report on 1-year survival rate of about 74%, haploidentical
more than 500 transplantations, survival after transplantation is recommended as second-line
receipt of a transplant from an HLA-matched therapy.93 Current results are promising, but be-
unrelated donor was not statistically inferior to cause of the relatively limited numbers of cases
survival after receipt of a transplant from a reported and the unknown long-term effects of
matched sibling donor, but the frequency of seri- complicated regimens and a mismatched im-
ous GVHD was twice as high.77 Young age is a mune system, haploidentical transplantation is
favorable factor for transplantation of grafts regarded as experimental in the United States
from unrelated donors, as it is for transplanta- and Europe.
tion of grafts from sibling donors. For children For the constitutional marrow failure syn-
in whom other therapies have failed, grafts from dromes, transplantation is the only curative
unrelated donors are associated with excellent option for marrow failure or acute leukemia.
survival; the survival rate was 95% in a multi- Although recipients have the advantage of young
center British study.57 Outcomes are better when age, specific considerations relate to the under-
marrow is transplanted rather than blood cells, lying biology of the syndrome: patients with
ATG is included in the conditioning regimen, Fanconi anemia have an inherent sensitivity to
donors are young, and the interval between diag- alkylating agents in conditioning regimens and
nosis and transplantation is short.77,79 Outcomes an increased risk of late cancers, and patients
have been so good in children that some experts with telomere disease may have hepatic and pul-
have advocated for first-line use of transplants monary deterioration after transplantation. Both
from unrelated donors,80,81 despite sometimes the decision to perform transplantation and the
protracted delays in identifying and collecting timing of the procedure can be difficult because
donor cells. Late complications are more fre- of the slow progression of moderate hematopoi-
quent in recipients of transplants from unrelated etic failure, the uncertainty of variable disease
donors than in recipients of transplants from outcomes without intervention, and the unpre-
siblings.82 dictable risk of transformation to leukemia.
Umbilical cord blood transplantation has been Nevertheless, in some clinical circumstances,
successful in aplastic anemia, mainly in children improvement has been seen in survival and mor-
because of the favorable relationship between bidity over historically poor outcomes.94-96 For
the cell numbers in the inoculum and the re- almost 100 patients with constitutional marrow
cipient’s weight, with survival approximating failure who underwent cord-blood stem-cell
90%.69,83 Rates of GVHD are low. The major transplantation, the 7-year survival rate was 86%
disadvantages of cord blood are delayed engraft- and most of the patients did not have GVHD.69
ment and prolonged neutropenia. Some proto- Children with Fanconi anemia who received
cols combine cord blood and mismatched bone transplants from unrelated donors had good
marrow.58 outcomes, similar to those with transplants
A potential donor who is half-matched to the from matched family donors.97 Even among
patient should be present in virtually every fam- adults with Fanconi anemia who received matched
ily. Since even single antigen disparities mark- transplants from a sibling after a reduced-inten-
edly affect the outcome of transplantation, over- sity conditioning regimen, the survival rate was
coming major histocompatibility differences had 85% at 3 years.98
seemed an insuperable barrier. T-cell–depleting
strategies — administration of cytotoxic drugs Immunosuppression
and biologic agents before transplantation and In the early years of transplantation, occasional
cyclophosphamide afterward84 — have been autologous recovery of bone marrow despite
used to prevent GVHD. The results have been graft failure suggested that the antilymphocyte
encouraging on the basis of extensive experience globulin used in conditioning regimens might
in Chinese centers, with much smaller series of have had a salutary effect. Combined with cyclo-
transplant recipients in the United States and sporine, ATG leads to hematologic responses in
Europe (Table 3). Haploidentical transplantation about two thirds of patients.14,93 Complex mix-
has been advocated in China as first-line treat- tures of antibodies to human proteins that have

n engl j med 379;17 nejm.org  October 25, 2018 1651


The New England Journal of Medicine
Downloaded from nejm.org at The University of Texas at El Paso on October 24, 2018. For personal use only. No other uses without permission.
Copyright © 2018 Massachusetts Medical Society. All rights reserved.
1652
Table 3. Haploidentical Hematopoietic Stem-Cell Transplantation for Severe Aplastic Anemia.*

No. of Median Acute and Chronic


Study Patients Age Conditioning and Prophylaxis Overall Survival GVHD† Graft Failure

yr % % no. of cases
85
The

Prospective study in Korea, 2009–2010 4 18 Conditioning: Cy, FLU, ATG 100 at 19 mo 0 0


Prophylaxis: CsA, MMF; graft depleted of CD3 or
CD3–CD19 cells
King’s College study86 6 30 Conditioning: Cy, FLU, low-dose TBI 67% at 1 yr Acute: 17 (skin) 2 primary
Prophylaxis: Cy (after transplantation), tacrolimus, MMF; Chronic: 0
GCSF-mobilized peripheral blood
Retrospective study in Brazil, 16 17 Conditioning: Cy, FLU, low-dose TBI 67% at 1 yr Acute: 13 1 primary,
2010–201487 Prophylaxis: Cy (after transplantation), CNI, MMF Chronic: limited, 20; 1 secondary
severe, 7
Multicenter prospective study in China, 101 19 Conditioning: BU, Cy, ATG 89 at 3 yr Acute: 34 2 secondary
2012–201588 Prophylaxis: CsA, MMF, MTX Chronic: 20 (extensive, 9)
Prospective pediatric study in Beijing, 52 9 Conditioning: BU, Cy, ATG 85 at 3 yr Acute: 14 (grade III–IV) 3 secondary
n e w e ng l a n d j o u r na l

The New England Journal of Medicine


2007–201589 Prophylaxis: CsA, MMF, MTX Chronic: 13
of

Multicenter retrospective study in China, 89 Conditioning: BU, Cy, ATG 86 at 3 yr Acute: 30 1 primary
2012–201590 Prophylaxis: CsA, MMF, MTX Chronic: 3
Johns Hopkins study, 2011–201659 13 30 Conditioning: Cy, FLU, low-dose TBI, ATG 100 at 21 mo 0 0

n engl j med 379;17 nejm.org  October 25, 2018


Prophylaxis: Cy (after transplantation), MMF, tacrolimus

Copyright © 2018 Massachusetts Medical Society. All rights reserved.


m e dic i n e

Retrospective study in Langfang, China, 41 13 Conditioning: Cy, FLU, BU, ATG, and GCSF-mobilized 80 at 3 yr Acute: 44 0
2012–201691 bone marrow and peripheral blood Chronic: 12
Prophylaxis: CIN, MMF, MTX

* BU denotes busulfan, G-CSF granulocyte colony-stimulating factor, and MMF mycophenolate mofetil.
† Data for acute GVHD are for grade II, III, or IV disease. Patients with chronic GVHD may or may not have had grade II, III, or IV acute GVHD.

Downloaded from nejm.org at The University of Texas at El Paso on October 24, 2018. For personal use only. No other uses without permission.
Aplastic Anemia

not been fully defined, ATGs have a relatively without Eltrombopag as Front-line Therapy for
mild lymphocyte-depleting effect, but subtle dif- Severe Aplastic Anemia Patients; ClinicalTrials.gov
ferences in the mechanism of action appear to number, NCT02099747), a multicenter European
be important for efficacy. For example, rabbit trial, is testing eltrombopag in combination with
ATG was much less efficacious than was horse ATG in a randomized protocol.
ATG in a randomized, controlled trial, in which Increases in bone marrow cellularity and the
a major difference in biologic effect was more numbers of CD34 and progenitor cells suggest a
severe depletion of CD4 cells and Treg cells after direct effect of eltrombopag on marrow stem
the administration of rabbit ATG.26 Even in pa- cells.45 Thrombopoietin concentrations in blood
tients with a response, blood counts do not usu- from patients with aplastic anemia are very
ally return to normal. Relapse occurs in 30%99 to high,107 but eltrombopag may evade a block to
60%100 of patients and usually responds to fur- receptor engagement in the presence of inter­
ther immunosuppression, but years of continued feron-γ (Alvarado L, Larochelle A: personal com-
cyclosporine therapy are required.101 Responses munication).
and outcomes are better in children102 than in
older adults.103 Patients who do not have a re- Androgens
sponse to horse ATG may have improvement Androgens have long been used as therapy for
with second-line rabbit ATG or alemtuzumab, a marrow failure syndromes. Although generally
pan–T-cell monoclonal antibody.101,104 regarded as much less efficacious than immuno-
suppressive strategies for the treatment of severe
Stem-Cell Stimulation aplastic anemia, androgens are standard care for
Many attempts to improve on ATG by adding many constitutional syndromes. Sex hormones
androgens, granulocyte colony-stimulating factor, increase expression of the gene for telomerase in
mycophenolate mofetil, or sirolimus have not cell culture108 and in mice.109 In a recent prospec-
altered response rates or long-term outcomes.14 tive trial, high doses of danazol, a synthetic an-
Hematopoietic growth factors are ineffective in drogen, improved blood counts in patients with
aplastic anemia. It was therefore unexpected when telomere disease and also appeared to reverse
eltrombopag, a synthetic mimetic of thrombo- accelerated telomere attrition.110
poietin, showed activity in patients with refrac-
tory aplastic anemia. Approximately half the pa- C onclusions
tients had robust trilineage improvements in
blood counts, which in most cases were durable The development of understanding and treat-
after discontinuation of the drug.105,106 When ment of aplastic anemia is a success story of the
eltrombopag was added to initial standard im- laboratory and clinic, with implications beyond
munosuppressive therapy, it markedly increased bone marrow failure. The causes of aplastic ane-
the overall response rate to about 80% and the mia are related to common environmental tox-
complete response rate to about 50%, with pa- ins, specific viral infections, and genes affecting
tients often having rapid hematologic recovery.45 basic cellular mechanisms. The role of the im-
To date, the rates of relapse and evolution to mune system has been recognized as both po-
myeloid cancers appear to be similar to or lower tent and subtle. Most gratifying, treatments for
than the rates among historical controls treated patients with immune aplastic anemia have im-
with immunosuppression alone. Eltrombopag has proved remarkably over the past several decades
been approved for use in patients with refractory because of the development of better transplan-
severe aplastic anemia, and an application has tation and immunosuppressive regimens. Trans-
been submitted to the Food and Drug Adminis- plantation can be beneficial in all types of mar-
tration for the use of eltrombopag in combina- row failure, but in the future, gene editing and
tion with immunosuppressive agents as initial restoration of function offer hope for constitu-
therapy for aplastic anemia. In both cases, data tional diseases.
came from a single center and limited numbers of Disclosure forms provided by the author are available with the
patients. RACE (A Prospective Randomized Multi- full text of this article at NEJM.org.
I thank my colleagues James Cooper, Valentine Giudice, Clare
center Study Comparing Horse Antithymocyte Sun, Philip Scheinberg, and Danielle Townsley for their careful
Globuline [hATG] + Cyclosporine A [CsA] with or reading of an earlier version of the manuscript.

n engl j med 379;17 nejm.org  October 25, 2018 1653


The New England Journal of Medicine
Downloaded from nejm.org at The University of Texas at El Paso on October 24, 2018. For personal use only. No other uses without permission.
Copyright © 2018 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

References
1. Kaufman DW, Kelly JP, Levy M, Sha­ Latour R, et al. Severe aplastic anemia as- 30. Imi T, Katagiri T, Hosomichi K, et al.
piro S. The drug etiology of agranulocyto- sociated with eosinophilic fasciitis: report Sustained clonal hematopoiesis by HLA-
sis and aplastic anemia. New York:​Ox- of 4 cases and review of the literature. lacking hematopoietic stem cells without
ford University Press, 1991. Medicine (Baltimore) 2013;​92:​69-81. driver mutations in aplastic anemia. Blood
2. Issaragrisil S, Kaufman DW, Ander- 17. Bernard C, Frih H, Pasquet F, et al. Adv 2018;​2:​1000-12.
son T, et al. The epidemiology of aplastic Thymoma associated with autoimmune 31. Luzzatto L, Notaro R. The “escape”
anemia in Thailand. Blood 2006;​107:​1299- diseases: 85 cases and literature review. model: a versatile mechanism for clonal
307. Autoimmun Rev 2016;​15:​82-92. expansion. Br J Haematol 2018 January 24
3. Yang C, Zhang X. Incidence survey of 18. Zoumbos NC, Gascón P, Djeu JY, (Epub ahead of print).
leukemia in China. Chin Med Sci J 1991;​6:​ Trost SR, Young NS. Circulating activated 32. Gargiulo L, Zaimoku Y, Scappini B,
65-70. suppressor T lymphocytes in aplastic ane- et al. Glycosylphosphatidylinositol-specific
4. Gross SA, Irons RD, Schnatter AR, mia. N Engl J Med 1985;​312:​257-65. T cells, IFN-γ-producing T cells, and patho-
et al. A hospital-based case control study 19. Hosokawa K, Muranski P, Feng X, et al. genesis of idiopathic aplastic anemia.
of aplastic anemia in Shanghai, China. Memory stem T cells in autoimmune dis- Blood 2017;​129:​388-92.
Chem Biol Interact 2010;​184:​165-73. ease: high frequency of circulating CD8+ 33. Feng X, Chuhjo T, Sugimori C, et al.
5. Garaycoechea JI, Patel KJ. Why does memory stem T cells in acquired aplastic Diazepam-binding inhibitor-related pro-
the bone marrow fail in Fanconi anemia? anemia. J Immunol 2016;​196:​1568-78. tein 1: a candidate autoantigen in acquired
Blood 2014;​123:​26-34. 20. Solomou EE, Keyvanfar K, Young NS. aplastic anemia patients harboring a mi-
6. Savage SA, Dufour C. Classical inher- T-bet, a Th1 transcription factor, is up- nor population of paroxysmal nocturnal
ited bone marrow failure syndromes with regulated in T cells from patients with hemoglobinuria-type cells. Blood 2004;​
high risk for myelodysplastic syndrome aplastic anemia. Blood 2006;​107:​3983-91. 104:​2425-31.
and acute myelogenous leukemia. Semin 21. Smith JN, Kanwar VS, MacNamara KC. 34. Hirano N, Butler MO, Von Bergwelt-
Hematol 2017;​54:​105-14. Hematopoietic stem cell regulation by Baildon MS, et al. Autoantibodies fre-
7. Wlodarski MW, Collin M, Horwitz MS. type I and II interferons in the pathogen- quently detected in patients with aplastic
GATA2 deficiency and related myeloid esis of acquired aplastic anemia. Front anemia. Blood 2003;​102:​4567-75.
neoplasms. Semin Hematol 2017;​54:​81-6. Immunol 2016;​7:​330. 35. Chen J. Animal models for acquired
8. Kuehn HS, Ouyang W, Lo B, et al. Im- 22. Liu CY, Fu R, Wang HQ, et al. Fas/FasL bone marrow failure syndromes. Clin Med
mune dysregulation in human subjects in the immune pathogenesis of severe Res 2005;​3:​102-8.
with heterozygous germline mutations in aplastic anemia. Genet Mol Res 2014;​13:​ 36. Chen J, Feng X, Desierto MJ, Keyvan-
CTLA4. Science 2014;​345:​1623-7. 4083-8. far K, Young NS. IFN-γ-mediated hemato-
9. Michniacki TF, Hannibal M, Ross CW, 23. Giudice V, Feng X, Lin Z, et al. Deep poietic cell destruction in murine models
et al. Hematologic manifestations of defi- sequencing and flow cytometric charac- of immune-mediated bone marrow failure.
ciency of adenosine deaminase 2 (DADA2) terization of expanded effector memory Blood 2015;​126:​2621-31.
and response to tumor necrosis factor in- CD8+CD57+ T cells frequently reveals T-cell 37. Maciejewski JP, Selleri C, Sato T, Ander-
hibition in DADA2-associated bone mar- receptor Vβ oligoclonality and CDR3 ho- son S, Young NS. A severe and consistent
row failure. J Clin Immunol 2018;​38:​166- mology in acquired aplastic anemia. Hae- deficit in marrow and circulating primi-
73. matologica 2018;​103:​759-69. tive hematopoietic cells (long-term culture-
10. Fogarty PF, Yamaguchi H, Wiestner A, 24. Jerez A, Clemente MJ, Makishima H, initiating cells) in acquired aplastic ane-
et al. Late presentation of dyskeratosis et al. STAT3 mutations indicate the pres- mia. Blood 1996;​88:​1983-91.
congenita as apparently acquired aplastic ence of subclinical T-cell clones in a sub- 38. Cooper JN, Young NS. Clonality in con-
anaemia due to mutations in telomerase set of aplastic anemia and myelodysplas- text: hematopoietic clones in their mar-
RNA. Lancet 2003;​362:​1628-30. tic syndrome patients. Blood 2013;​122:​ row environment. Blood 2017;​130:​2363-72.
11. Seo A, Ben-Harosh M, Sirin M, et al. 2453-9. 39. Maciejewski JP, Selleri C. Evolution of
Bone marrow failure unresponsive to bone 25. Kordasti S, Costantini B, Seidl T, et al. clonal cytogenetic abnormalities in aplas-
marrow transplant is caused by mutations Deep phenotyping of Tregs identifies an tic anemia. Leuk Lymphoma 2004;​45:​433-
in thrombopoietin. Blood 2017;​130:​875- immune signature for idiopathic aplastic 40.
80. anemia and predicts response to treat- 40. Pezeshki A, Podder S, Kamel R, Corey
12. Keel SB, Scott A, Sanchez-Bonilla M, ment. Blood 2016;​128:​1193-205. SJ. Monosomy 7/del (7q) in inherited bone
et al. Genetic features of myelodysplastic 26. Scheinberg P, Nunez O, Weinstein B, marrow failure syndromes: a systematic
syndrome and aplastic anemia in pediat- et al. Horse versus rabbit antithymocyte review. Pediatr Blood Cancer 2017;​64:​64.
ric and young adult patients. Haemato- globulin in acquired aplastic anemia. 41. Ogawa S. Clonal hematopoiesis in ac-
logica 2016;​101:​1343-50. N Engl J Med 2011;​365:​430-8. quired aplastic anemia. Blood 2016;​128:​
13. Bluteau O, Sebert M, Leblanc T, et al. 27. Deng XZ, Du M, Peng J, et al. Asso- 337-47.
A landscape of germ line mutations in a ciations between the HLA-A/B/DRB1 poly- 42. Yoshizato T, Dumitriu B, Hosokawa K,
cohort of inherited bone marrow failure morphisms and aplastic anemia: evidence et al. Somatic mutations and clonal hema-
patients. Blood 2018;​131:​717-32. from 17 case-control studies. Hematology topoiesis in aplastic anemia. N Engl J Med
14. Scheinberg P, Young NS. How I treat 2018;​23:​154-62. 2015;​373:​35-47.
acquired aplastic anemia. Blood 2012;​120:​ 28. Katagiri T, Sato-Otsubo A, Kashiwase 43. Kulasekararaj AG, Jiang J, Smith AE,
1185-96. K, et al. Frequent loss of HLA alleles asso- et al. Somatic mutations identify a sub-
15. Gonzalez-Casas R, Garcia-Buey L, ciated with copy number-neutral 6pLOH group of aplastic anemia patients who
Jones EA, Gisbert JP, Moreno-Otero R. in acquired aplastic anemia. Blood 2011;​ progress to myelodysplastic syndrome.
Systematic review: hepatitis-associated 118:​6601-9. Blood 2014;​124:​2698-704.
aplastic anaemia — a syndrome associ- 29. Babushok DV, Duke JL, Xie HM, et al. 44. Dumitriu B, Feng X, Townsley DM, et al.
ated with abnormal immunological func- Somatic HLA mutations expose the role of Telomere attrition and candidate gene mu-
tion. Aliment Pharmacol Ther 2009;​30:​ class I–mediated autoimmunity in aplas- tations preceding monosomy 7 in aplastic
436-43. tic anemia and its clonal complications. anemia. Blood 2015;​125:​706-9.
16. de Masson A, Bouaziz JD, Peffault de Blood Adv 2017;​1:​1900-10. 45. Townsley DM, Scheinberg P, Winkler T,

1654 n engl j med 379;17 nejm.org  October 25, 2018

The New England Journal of Medicine


Downloaded from nejm.org at The University of Texas at El Paso on October 24, 2018. For personal use only. No other uses without permission.
Copyright © 2018 Massachusetts Medical Society. All rights reserved.
Aplastic Anemia

et al. Eltrombopag added to standard 58. Purev E, Tian X, Aue G, et al. Alloge- et al. Long-term outcomes of cord blood
­immunosuppression for aplastic anemia. neic transplantation using CD34+ selected transplantation from an HLA-identical
N Engl J Med 2017;​376:​1540-50. peripheral blood progenitor cells com- sibling for patients with bone marrow
46. Townsley DM, Dumitriu B, Young NS. bined with non-mobilized donor T cells failure syndromes: a report from Euro-
Bone marrow failure and the telomeropa- for refractory severe aplastic anaemia. Br J cord, Cord Blood Committee and Severe
thies. Blood 2014;​124:​2775-83. Haematol 2017;​176:​950-60. Aplastic Anemia Working Party of the
47. Scheinberg P, Cooper JN, Sloand EM, 59. DeZern AE, Zahurak M, Symons H, European Society for Blood and Marrow
Wu CO, Calado RT, Young NS. Associa- Cooke K, Jones RJ, Brodsky RA. Alterna- Transplantation. Biol Blood Marrow Trans-
tion of telomere length of peripheral tive donor transplantation with high-dose plant 2017;​23:​1939-48.
blood leukocytes with hematopoietic re- post-transplantation cyclophosphamide for 70. Giammarco S, Peffault de Latour R,
lapse, malignant transformation, and sur- refractory severe aplastic anemia. Biol Sica S, et al. Transplant outcome for pa-
vival in severe aplastic anemia. JAMA Blood Marrow Transplant 2017;​23:​498-504. tients with acquired aplastic anemia over
2010;​304:​1358-64. 60. Kim SY, Le Rademacher J, Antin JH, the age of 40: has the outcome improved?
48. Narita A, Muramatsu H, Sekiya Y, et al. et al. Myelodysplastic syndrome evolving Blood 2018;​131:​1989-92.
Paroxysmal nocturnal hemoglobinuria and from aplastic anemia treated with immu- 71. Gupta V, Eapen M, Brazauskas R, et al.
telomere length predicts response to im- nosuppressive therapy: efficacy of hema- Impact of age on outcomes after bone
munosuppressive therapy in pediatric aplas- topoietic stem cell transplantation. Hae- marrow transplantation for acquired aplas-
tic anemia. Haematologica 2015;​100:​1546- matologica 2014;​99:​1868-75. tic anemia using HLA-matched sibling
52. 61. Dufour C, Pillon M, Sociè G, et al. donors. Haematologica 2010;​95:​2119-25.
49. Aalbers AM, Kajigaya S, van den Outcome of aplastic anaemia in children: 72. Eckrich MJ, Ahn KW, Champlin RE, et
Heuvel-Eibrink MM, van der Velden VHJ, a study by the Severe Aplastic Anaemia al. Effect of race on outcomes after allo-
Calado RT, Young NS. Human telomere and Paediatric Disease Working Parties of geneic hematopoietic cell transplantation
disease due to disruption of the CCAAT the European Group for Blood and Bone for severe aplastic anemia. Am J Hematol
box of the TERC promoter. Blood 2012;​ Marrow Transplantation. Br J Haematol 2014;​89:​125-9.
119:​3060-3. 2015;​169:​565-73. 73. Schrezenmeier H, Passweg JR, Marsh
50. Hsu AP, Johnson KD, Falcone EL, et al. 62. Yoshida N, Kobayashi R, Yabe H, et al. JCW, et al. Worse outcome and more
GATA2 haploinsufficiency caused by mu- First-line treatment for severe aplastic chronic GVHD with peripheral blood pro-
tations in a conserved intronic element anemia in children: bone marrow trans- genitor cells than bone marrow in HLA-
leads to MonoMAC syndrome. Blood 2013;​ plantation from a matched family donor matched sibling donor transplants for
121:​3830-7. versus immunosuppressive therapy. Hae- young patients with severe acquired aplas-
51. Bennett JM, Orazi A. Diagnostic crite- matologica 2014;​99:​1784-91. tic anemia. Blood 2007;​110:​1397-400.
ria to distinguish hypocellular acute my- 63. Dufour C, Pillon M, Passweg J, et al. 74. Eapen M, Le Rademacher J, Antin JH,
eloid leukemia from hypocellular myelo- Outcome of aplastic anemia in adoles- et al. Effect of stem cell source on out-
dysplastic syndromes and aplastic anemia: cence: a survey of the Severe Aplastic Ane- comes after unrelated donor transplanta-
recommendations for a standardized ap- mia Working Party of the European Group tion in severe aplastic anemia. Blood 2011;​
proach. Haematologica 2009;​94:​264-8. for Blood and Marrow Transplantation. 118:​2618-21.
52. Cremers EMP, Westers TM, Alhan C, Haematologica 2014;​99:​1574-81. 75. Kekre N, Zhang Y, Zhang M-J, et al.
et al. Multiparameter flow cytometry is 64. Champlin RE, Perez WS, Passweg JR, Effect of antithymocyte globulin source
instrumental to distinguish myelodys- et al. Bone marrow transplantation for on outcomes of bone marrow transplan-
plastic syndromes from non-neoplastic severe aplastic anemia: a randomized con- tation for severe aplastic anemia. Haema-
cytopenias. Eur J Cancer 2016;​54:​49-56. trolled study of conditioning regimens. tologica 2017;​102:​1291-8.
53. Malcovati L, Gallì A, Travaglino E, Blood 2007;​109:​4582-5. 76. Peinemann F, Grouven U, Kröger N,
et al. Clinical significance of somatic mu- 65. Marsh JC, Pearce RM, Koh MBC, et al. Pittler M, Zschorlich B, Lange S. Unrelated
tation in unexplained blood cytopenia. Retrospective study of alemtuzumab vs donor stem cell transplantation in ac-
Blood 2017;​129:​3371-8. ATG-based conditioning without irradia- quired severe aplastic anemia: a systematic
54. Nazha A, Seastone D, Radivoyevitch T, tion for unrelated and matched sibling review. Haematologica 2009;​94:​1732-42.
et al. Genomic patterns associated with donor transplants in acquired severe 77. Bacigalupo A, Sica S. Alternative do-
hypoplastic compared to hyperplastic my- aplastic anemia: a study from the British nor transplants for severe aplastic ane-
elodysplastic syndromes. Haematologica Society for Blood and Marrow Transplan- mia: current experience. Semin Hematol
2015;​100(11):​e434-e437. tation. Bone Marrow Transplant 2014;​49:​ 2016;​53:​115-9.
55. Bat T, Abdelhamid ON, Balasubrama- 42-8. 78. Eapen M, Horowitz MM. Alternative
nian SK, et al. The evolution of paroxys- 66. Bacigalupo A, Socié G, Hamladji RM, donor transplantation for aplastic ane-
mal nocturnal haemoglobinuria depends et al. Current outcome of HLA identical mia. Hematology Am Soc Hematol Educ
on intensity of immunosuppressive ther- sibling versus unrelated donor transplants Program 2010;​2010:​43-6.
apy. Br J Haematol 2018;​182:​730-3. in severe aplastic anemia: an EBMT analy- 79. Arai Y, Kondo T, Yamazaki H, et al.
56. Valdez JM, Scheinberg P, Nunez O, sis. Haematologica 2015;​100:​696-702. Allogeneic unrelated bone marrow trans-
Wu CO, Young NS, Walsh TJ. Decreased 67. Peffault de Latour R, Chevret S, Jubert plantation from older donors results in
infection-related mortality and improved C, et al. Unrelated cord blood transplan- worse prognosis in recipients with aplastic
survival in severe aplastic anemia in the tation in patients with acquired refractory anemia. Haematologica 2016;​101:​644-52.
past two decades. Clin Infect Dis 2011;​52:​ aplastic anemia: a nationwide phase II 80. Dufour C, Veys P, Carraro E, et al.
726-35. study. Blood 2018;​132:​750-4. Similar outcome of upfront-unrelated and
57. Samarasinghe S, Steward C, Hiwarkar 68. Kuwatsuka Y, Kanda J, Yamazaki H, matched sibling stem cell transplantation
P, et al. Excellent outcome of matched un- et al. A comparison of outcomes for cord in idiopathic paediatric aplastic anaemia:
related donor transplantation in paediat- blood transplantation and unrelated bone a study on behalf of the UK Paediatric
ric aplastic anaemia following failure with marrow transplantation in adult aplastic BMT Working Party, Paediatric Diseases
immunosuppressive therapy: a United King- anemia. Biol Blood Marrow Transplant Working Party and Severe Aplastic Anae-
dom multicentre retrospective experience. 2016;​22:​1836-43. mia Working Party of EBMT. Br J Haema-
Br J Haematol 2012;​157:​339-46. 69. Pagliuca S, Peffault de Latour R, Volt F, tol 2015;​171:​585-94.

n engl j med 379;17 nejm.org  October 25, 2018 1655


The New England Journal of Medicine
Downloaded from nejm.org at The University of Texas at El Paso on October 24, 2018. For personal use only. No other uses without permission.
Copyright © 2018 Massachusetts Medical Society. All rights reserved.
Aplastic Anemia

81. Mackarel J, Iatan M, Kumar L, Storey 90. Xu L-P, Jin S, Wang S-Q, et al. Upfront long-term observation follow-up. Br J Hae-
L, O’Marcaigh A, Smith O. In support of haploidentical transplant for acquired se- matol 2008;​140:​197-205.
upfront stem cell transplantation as first- vere aplastic anemia: registry-based com- 101. Scheinberg P, Nunez O, Weinstein B,
line therapy for paediatric patients with parison with matched related transplant. Scheinberg P, Wu CO, Young NS. Activity
idiopathic severe aplastic anaemia who J Hematol Oncol 2017;​10:​25. of alemtuzumab monotherapy in treat-
lack a sibling donor. Br J Haematol 2017;​ 91. Lu Y, Sun RJ, Zhao YL, et al. Unmanip- ment-naive, relapsed, and refractory se-
177:​806-8. ulated haploidentical hematopoietic stem vere acquired aplastic anemia. Blood 2012;​
82. Buchbinder D, Nugent DJ, Brazauskas cell transplantation achieved outcomes 119:​345-54.
R, et al. Late effects in hematopoietic cell comparable with matched unrelated do- 102. Scheinberg P, Wu CO, Nunez O,
transplant recipients with acquired severe nor transplantation in young acquired Young NS. Long-term outcome of pediat-
aplastic anemia: a report from the Late severe aplastic anemia. Biol Blood Mar- ric patients with severe aplastic anemia
Effects Working Committee of the Center row Transplant 2018 May 14 (Epub ahead treated with antithymocyte globulin.
for International Blood and Marrow Trans- of print). J Pediatr 2008;​153:​814-9.
plant Research. Biol Blood Marrow Trans- 92. Cheng Y, Xu Z, Zhang Y, et al. First- 103. Tichelli A, Socié G, Henry-Amar M,
plant 2012;​18:​1776-84. line choice for severe aplastic anemia in et al. Effectiveness of immunosuppressive
83. Ades L, Mary JY, Robin M, et al. Long- children: transplantation from a haplo­ therapy in older patients with aplastic
term outcome after bone marrow trans- identical donor vs immunosuppressive anemia. Ann Intern Med 1999;​130:​193-
plantation for severe aplastic anemia. therapy. Clin Transplant 2018;​32:​32. 201.
Blood 2004;​103:​2490-7. 93. Bacigalupo A. How I treat acquired 104. Thota S, Patel BJ, Sadaps M, et al.
84. Robinson TM, O’Donnell PV, Fuchs aplastic anemia. Blood 2017;​129:​1428-36. Therapeutic outcomes using subcutane-
EJ, Luznik L. Haploidentical bone marrow 94. Alter BP. Inherited bone marrow failure ous low dose alemtuzumab for acquired
and stem cell transplantation: experience syndromes: considerations pre- and post- bone marrow failure conditions. Br J Hae-
with post-transplantation cyclophospha- transplant. Blood 2017;​130:​2257-64. matol 2017 September 14 (Epub ahead of
mide. Semin Hematol 2016;​53:​90-7. 95. Fioredda F, Iacobelli S, Korthof ET, print).
85. Koh KN, Im HJ, Kim BE, et al. Haplo­ et al. Outcome of haematopoietic stem cell 105. Olnes MJ, Scheinberg P, Calvo KR,
identical haematopoietic stem cell trans- transplantation in dyskeratosis congenita. et al. Eltrombopag and improved hemato-
plantation using CD3 or CD3/CD19 Br J Haematol 2018 July 9 (Epub ahead of poiesis in refractory aplastic anemia.
­depletion and conditioning with f luda­ print). N Engl J Med 2012;​367:​11-9.
rabine, cyclophosphamide and antithymo- 96. Peffault de Latour R, Soulier J. How I 106. Desmond R, Townsley DM, Dumitriu
cyte globulin for acquired severe aplastic treat MDS and AML in Fanconi anemia. B, et al. Eltrombopag restores trilineage
anaemia. Br J Haematol 2012;​ 157:​139- Blood 2016;​127:​2971-9. hematopoiesis in refractory severe aplas-
42. 97. Ebens CL, DeFor TE, Tryon R, Wagner tic anemia that can be sustained on dis-
86. Clay J, Kulasekararaj AG, Potter V, et al. JE, MacMillan ML. Comparable outcomes continuation of drug. Blood 2014;​ 123:​
Nonmyeloablative peripheral blood haplo­ after HLA-matched sibling and alternative 1818-25.
identical stem cell transplantation for re- donor hematopoietic cell transplantation 107. Zhao X, Feng X, Wu Z, et al. Persis-
fractory severe aplastic anemia. Biol Blood for children with Fanconi anemia and se- tent elevation of plasma thrombopoietin
Marrow Transplant 2014;​20:​1711-6. vere aplastic anemia. Biol Blood Marrow levels after treatment in severe aplastic
87. Esteves I, Bonfim C, Pasquini R, et al. Transplant 2018;​24:​765-71. anemia. Exp Hematol 2018;​58:​39-43.
Haploidentical BMT and post-transplant 98. Bierings M, Bonfim C, Peffault De La- 108. Calado RT, Yewdell WT, Wilkerson
Cy for severe aplastic anemia: a multi- tour R, et al. Transplant results in adults KL, et al. Sex hormones, acting on the
center retrospective study. Bone Marrow with Fanconi anaemia. Br J Haematol 2018;​ TERT gene, increase telomerase activity in
Transplant 2015;​50:​685-9. 180:​100-9. human primary hematopoietic cells. Blood
88. Xu L-P, Wang S-Q, Wu D-P, et al. Haplo- 99. Scheinberg P, Rios O, Scheinberg P, 2009;​114:​2236-43.
identical transplantation for acquired se- Weinstein B, Wu CO, Young NS. Pro- 109. Bär C, Huber N, Beier F, Blasco MA.
vere aplastic anaemia in a multicentre pro- longed cyclosporine administration after Therapeutic effect of androgen therapy in
spective study. Br J Haematol 2016;​172:​ antithymocyte globulin delays but does a mouse model of aplastic anemia pro-
265-74. not prevent relapse in severe aplastic ane- duced by short telomeres. Haematologica
89. Xu LP, Zhang XH, Wang FR, et al. mia. Am J Hematol 2014;​89:​571-4. 2015;​100:​1267-74.
Haploidentical transplantation for pediat- 100. Saracco P, Quarello P, Iori AP, et al. 110. Townsley DM, Dumitriu B, Liu D,
ric patients with acquired severe aplastic Cyclosporin A response and dependence et al. Danazol treatment for telomere dis-
anemia. Bone Marrow Transplant 2017;​ in children with acquired aplastic anae- eases. N Engl J Med 2016;​374:​1922-31.
52:​381-7. mia: a multicentre retrospective study with Copyright © 2018 Massachusetts Medical Society.

images in clinical medicine


The Journal welcomes consideration of new submissions for Images in Clinical
Medicine. Instructions for authors and procedures for submissions can be found
on the Journal’s website at NEJM.org. At the discretion of the editor, images that
are accepted for publication may appear in the print version of the Journal,
the electronic version, or both.

1656 n engl j med 379;17 nejm.org  October 25, 2018

The New England Journal of Medicine


Downloaded from nejm.org at The University of Texas at El Paso on October 24, 2018. For personal use only. No other uses without permission.
Copyright © 2018 Massachusetts Medical Society. All rights reserved.

Anda mungkin juga menyukai