Penugasan - Diagnosis and Treatment of Aplastic Anemia

Download as pdf or txt
Download as pdf or txt
You are on page 1of 22

HHS Public Access

Author manuscript
Curr Treat Options Oncol. Author manuscript; available in PMC 2018 November 16.
Author Manuscript

Published in final edited form as:


Curr Treat Options Oncol. ; 18(12): 70. doi:10.1007/s11864-017-0511-z.

Diagnosis and Treatment of Aplastic Anemia


Scott A. Peslak, MD, PhD1, Timothy Olson, MD, PhD2,3, and Daria V. Babushok, MD, PhD1,2
1Division
of Hematology and Oncology, Department of Medicine, Hospital of the University of
Pennsylvania, Perelman Center for Advanced Medicine, 3400 Civic Center Blvd, Philadelphia, PA
19406, USA
2Comprehensive Bone Marrow Failure Center, Children’s Hospital of Philadelphia, 3615 Civic
Author Manuscript

Center Boulevard, Philadelphia, PA 19104


3Division
of Oncology, Department of Pediatrics, Children’s Hospital of Philadelphia, 3615 Civic
Center Boulevard, Philadelphia, PA 19104

Opinion Statement
Acquired aplastic anemia (AA) is a rare, life-threatening bone marrow failure (BMF) disorder that
affects patients of all ages and is caused by lymphocyte destruction of early hematopoietic cells.
Diagnosis of AA requires a comprehensive approach with prompt evaluation for inherited and
secondary causes of bone marrow aplasia, while providing aggressive supportive care. The choice
of frontline therapy is determined by a number of factors including AA severity, age of the patient,
donor availability, and access to optimal therapies. For newly diagnosed severe aplastic anemia,
bone marrow transplant should be pursued in all pediatric patients and in younger adult patients
Author Manuscript

when a matched sibling donor is available. Frontline therapy in older adult patients and in all
patients lacking a matched sibling donor involves immunosuppressive therapy (IST) with horse
antithymocyte globulin and cyclosporine A. Recent improvements in upfront therapy include
encouraging results with upfront closely matched unrelated donor transplants in younger patients
and the emerging benefits of eltrombopag combined with initial IST, with randomized studies
underway. In the refractory setting, several therapeutic options exist, with improving outcomes of
matched unrelated donor and haploidentical bone marrow transplantation as well as the addition of
eltrombopag to the non-transplant AA armamentarium. With the recent appreciation of frequent
clonal hematopoiesis in AA patients and with the growing use of next-generation sequencing in
the clinic, utmost caution should be exercised in interpreting the significance of somatic mutations
in AA. Future longitudinal studies of large numbers of patients are needed to determine the
prognostic significance of somatic mutations and to guide optimal surveillance and treatment
Author Manuscript

approaches to prevent long term clonal complications.

Corresponding Author: Daria Babushok, M.D. Ph.D., Division of Hematology-Oncology, Department of Medicine, Hospital of the
University of Pennsylvania, Philadelphia, Pennsylvania, PCAM 12 South, 3400 Civic Center Blvd, Philadelphia PA 19104,
[email protected], Phone: 215-614-1847, Fax: 215-615-5888.
Compliance with Ethics Guidelines
Conflict of Interest
The authors have no conflicts of interests to disclose.
Human and Animal Rights and Informed Consent
This article does not contain any studies with human or animal subjects performed by any of the authors.
Peslak et al. Page 2

Keywords
Author Manuscript

Aplastic anemia; bone marrow failure; eltrombopag; cyclosporine A; antithymocyte globulin;


immunosuppressive therapy; bone marrow transplant; clonal hematopoiesis

Introduction
Aplastic anemia (AA) is a rare, immune-mediated hematopoietic disorder associated with
significant morbidity and mortality. In patients with suspected AA, rapid and accurate
diagnosis and concomitant supportive care are critical. Historically, immunosuppressive
therapy (IST) and bone marrow transplantation (BMT) in eligible patients have been the
mainstay of AA treatment [1]. However, new frontline and salvage therapies are
fundamentally changing how we approach therapy of AA, particularly in adult patients [2–
Author Manuscript

4]. In pediatric patients, new transplant strategies and improvements in supportive care have
led to greatly improved outcomes and increasing use of BMT in both upfront and refractory
settings [5]. Furthermore, recent recognition of frequent clonal hematopoiesis in AA has
changed our understanding of this immune-mediated blood disorder, reframing how we view
somatic changes and a diagnosis of myelodysplastic syndrome (MDS) in patients with AA
[6, 7]. Here, we present a comprehensive review of the diagnosis and treatment of AA,
focusing on recent studies.

Clinical presentation and epidemiology


AA should be suspected in patients presenting with pancytopenia and a hypocellular bone
marrow. Typical symptoms include fatigue and easy bruising or bleeding; infections may be
present, but generally there is no long-standing illness. There is a well-recognized bimodal
Author Manuscript

age distribution with one peak in mid to late childhood and another in the elderly [8]. The
estimated annual incidence of AA is ~2 cases per million in Europe and North America,
with a 2–3 fold higher incidence in East Asia [8]. In ~10% of patients, a history of non-viral
hepatitis can precede the onset of AA [9]; an uncommon association with eosinophilic
fasciitis has also been reported [10]. With rare exceptions, such as chloramphenicol,
antiepileptics, and the emerging link to immunotherapies [8, 11], a causal relationship to
medications or toxins can be difficult to establish.

Diagnosis and severity stratification


When AA is suspected, a comprehensive evaluation should be performed rapidly to exclude
other mimicking conditions (Figure 1, Table 1). A baseline evaluation requires a full history
Author Manuscript

and physical exam, a complete blood count with differential, a blood smear, a reticulocyte
count, and a bone marrow aspirate with a core biopsy, with ancillary studies including
cytogenetics and fluorescence in situ hybridization (FISH).

The search for alternative etiologies (Figure 1, Table 1) should focus on ascertainment of
drug and toxin exposures, signs and symptoms suggestive of autoimmune or rheumatologic
diseases, family and/or personal history suggestive of an inherited BMF disorder, infections,
and nutritional deficiencies. Exclusion of inherited BMF is particularly relevant in children

Curr Treat Options Oncol. Author manuscript; available in PMC 2018 November 16.
Peslak et al. Page 3

and younger adults, where, at a minimum, an evaluation should include a detailed family
Author Manuscript

history looking for lifelong cytopenias, congenital anomalies, cancers, and lung and liver
pathology; in addition, patients should be screened for Fanconi anemia by testing the
patient’s lymphocytes for sensitivity to crosslinking agents and for Dyskeratosis congenita
by measuring lymphocyte telomere lengths [12]. Lymphocyte telomere lengths may also be
low in AA, particularly hepatitis-associated AA, requiring careful interpretation [13].
Additional causes of acquired BMF include autoimmune marrow aplasia due to a clonal T-
or NK- large granular lymphocyte (LGL) expansion [14], which can be evaluated by T cell
receptor rearrangement studies paired with lymphocyte flow cytometry. Morphologic and
cytogenetic analyses are used to evaluate for hypoplastic MDS [15], although limited
cellularity frequently precludes informative morphology assessment. Because of their
association with acquired AA, detection of a paroxysmal nocturnal hemoglobinuria (PNH)
clone (seen in up to 50% of AA patients) or copy number-neutral loss of heterozygosity of
Author Manuscript

chromosome arm 6p (6p CN-LOH, seen in about 12% of AA patients) can be helpful in
supporting the diagnosis of AA.

Once the diagnostic evaluation is complete, treatment is guided by the AA severity,


established by the Camitta criteria (Figure 1) [16, 17]. For younger patients with severe
aplastic anemia (SAA) or very severe aplastic anemia (VSAA), a transplant evaluation
should be rapidly initiated. A referral to a tertiary center that specializes in the care of AA
patients should be strongly considered.

Supportive Care
Throughout the diagnostic and treatment process, patients must be provided aggressive
supportive care. Generally, restrictive transfusion targets (hemoglobin > 7 g/dL, platelets >
Author Manuscript

10,000 cells/μL) are preferred, especially in potential transplant candidates, given the risk of
alloimmunization and transfusional iron overload [18]. Irradiated blood products should be
used to prevent transfusion-associated graft-versus-host disease (GVHD). Because of the
high mortality due to invasive mold infections, particularly Aspergillus species, antifungal
prophylaxis with voriconazole or posaconazole should be used in patients with severe
neutropenia (absolute neutrophil count < 500 cells/μL) [18]. Pneumocystis jirovecii
pneumonia (PJP) prophylaxis should be used during the period of lymphopenia following
ATG therapy, ideally selecting an alternative to trimethoprim-sulfamethoxazole because of
its myelosuppressive effects. Antimicrobial prophylaxis with quinolone antibiotics in
patients with VSAA can reduce the risk of gram-negative sepsis, but routine use of
prophylactic antibiotics in patients with higher neutrophil counts is not advised in order to
limit antibiotic resistance. Because granulocyte-colony-stimulating factor (G-CSF) does not
Author Manuscript

improve overall survival when added to IST [19, 20], routine G-CSF use outside of episodes
of febrile neutropenia remains controversial [21]. The benefits and risks of vaccines in AA
also remain controversial due to the risk of immune activation, with some AA guidelines
recommending against vaccinations outside of the post-transplant setting [21].

Curr Treat Options Oncol. Author manuscript; available in PMC 2018 November 16.
Peslak et al. Page 4

Transplant-based therapies for SAA/VSAA


Author Manuscript

Patient selection
In patients with SAA and VSAA eligible for transplant-based therapy, age remains the major
factor predicting survival after matched sibling donor (MSD) allogeneic transplantation. A
retrospective analysis from the Center for International Blood and Marrow Transplant
Research (CIBMTR) of over 1,300 patients receiving MSD-BMT showed the adjusted 5-
year overall survival (OS) of 53% in patients over the age of 40 years, as compared to 82%
for patients under 20 years and 72% for patients aged 20–40 years [22]. The differences
were primarily due to increased GVHD, infections, and delayed platelet recovery in the
older cohort. In addition, these patients were more likely to have received prior IST, and/or
to have additional comorbidities with poorer performance status and a longer interval
between diagnosis and BMT [22]. Although outcomes in older patients transplanted with
Author Manuscript

fludarabine-containing regimens have been more encouraging, these data are limited to
retrospective analyses [23]. Thus, the current standard of care for patients older than 40
years is frontline IST, while BMT is the treatment of choice for children and young adults
with SAA who have a MSD (Figure 1).

Donor choice
Historically, frontline transplantation for SAA in patients under 40 years of age has been
largely limited to MSD transplants [24, 25]. However, a recent retrospective analysis of
approximately 1,450 patients with AA transplanted between 2005 and 2009 showed no
significant difference in OS between MSD and matched unrelated donor (MUD) transplant,
although rates of acute and chronic GVHD were higher with MUD-BMT [26]. An analysis
of 29 pediatric patients treated with Fludarabine/Cyclophosphamide/ATG (FCC)
Author Manuscript

conditioning followed by unrelated donor transplantation showed similar overall and


progression-free survival as compared to historical MSD-BMT controls, and superior
outcomes compared to IST [5], suggesting that front-line therapy with MUD-BMT may be
considered upfront in selected patients under age 20. Randomized trials are underway to
compare outcomes of upfront MUD-BMT versus IST in pediatric patients without a
matched sibling donor (Pediatric Blood and Marrow Transplant Consortium and the North
American Pediatric Aplastic Anemia Consortium); studies exploring frontline MUD-BMT
in adults under 40 are also ongoing (Blood and Marrow Transplant Clinical Trials Network).
Pending these prospective studies, IST remains the standard upfront AA therapy in patients
without MSD [26, 5].

The outcomes of mismatched or haploidentical donor transplantation in AA have also


improved. In a prospective multicenter study of 101 AA patients receiving haploidentical
Author Manuscript

transplants in China, 94% of patients achieved successful engraftment with 3-year overall
and failure free survival of 89% and 86%, respectively [27]. A registry-based comparison of
upfront haploidentical and MSD transplantation in 158 consecutive SAA patients in China
have shown similar high rates of engraftment and OS, but significantly higher rates of grade
III–IV acute GVHD (10% versus 1.5%) and chronic GVHD (31% versus 4.4%) for
haploidentical transplants [28]. A more recent study of 16 patients receiving haploidentical
or unrelated donor transplants with post-transplant cyclophosphamide showed encouraging

Curr Treat Options Oncol. Author manuscript; available in PMC 2018 November 16.
Peslak et al. Page 5

results with 100% engraftment and no instances of grade 3 or higher GVHD [29]. Novel
Author Manuscript

approaches including co-infusion of mesenchymal stem cells [30] and selective T cell
Receptor αβ depletion [31] are being explored.

Graft source
Bone marrow grafts have been shown to produce superior OS compared to peripheral blood
stem cell (PBSC) grafts in both pediatric [32] and adult [33] AA patients, due to lower rates
of GVHD. More recent efforts to improve outcomes with PBSC showed encouraging results
with partial T cell depletion [34]; larger randomized prospective studies are needed to
confirm the efficacy and safety of this approach.

Conditioning Regimens
The standard conditioning regimen for MSD-BMT in younger patients is 200 mg/kg
Author Manuscript

cyclophosphamide with antithymocyte globulin (ATG), with three-year survival rates of 92%
[35]. However, subsequent studies in older transplant recipients (age > 30) did not show a
survival benefit when compared to IST [36]. To reduce toxicity in older patients, newer
regimens have incorporated fludarabine with lower-dose cyclophosphamide and with ATG
(FCA) or alemtuzumab (FCC), with improved OS [37–39]. A CIBMTR analysis of 833 AA
bone marrow transplants evaluated the role of ATG source on transplant outcomes, and
demonstrated that rabbit ATG (Thymoglobulin, Sanofi, France) results in lower rates of
acute and chronic GVHD for MSD transplants, improves survival, and lowers rates of acute
GVHD for MUD transplants [40]. Conditioning for MUD and haploidentical transplants
also includes 200 cGy total body irradiation [41].

Non-transplant therapy of AA
Author Manuscript

Immunosuppression
For patients older than 40 years with newly-diagnosed SAA/VSAA or younger patients
without an MSD, immunosuppression with ATG and cyclosporine A (CsA) continues to be
the recommended frontline therapy (Figure 1) [42], offering outcomes comparable to
allogeneic BMT with reduced morbidity in older patients [43, 44]. Horse ATG is the
recommended ATG source, based on a randomized-controlled trial of 120 patients showing a
superior overall response (68% compared to 37%) and OS (96% compared to 76%) for horse
ATG-based IST compared to rabbit ATG-based IST [45].

Role of Eltrombopag in Frontline Therapy


One of the most promising recent treatments for AA is the oral thrombopoietin (TPO)
Author Manuscript

receptor agonist eltrombopag, previously approved for treatment of chronic idiopathic


thrombocytopenic purpura [46]. Studies in mouse models showed that signaling via the TPO
receptor c-mpl is necessary for hematopoietic stem cell maintenance and downstream
expansion and differentiation [47, 48]. Interestingly, patients with congenital
amegakaryocytic thrombocytopenia who have mutations in c-mpl can progress to aplastic
anemia, suggesting that a deficiency in TPO signaling may play a role in AA pathogenesis
[49]. Based on these studies, eltrombopag was initially tested as monotherapy in a Phase 2
study of 43 patients with relapsed/refractory SAA with persistent thrombocytopenia

Curr Treat Options Oncol. Author manuscript; available in PMC 2018 November 16.
Peslak et al. Page 6

following IST [2, 3]. The overall response rate after 3–4 months of therapy was 40% (17 of
Author Manuscript

43). Remarkably, one patient achieved a trilineage response, and 8 patients had neutrophil
responses including 4 with severe neutropenia. With long-term treatment, 7 patients
achieved trilineage responses. An early signal of increased clonal evolution in this high-risk
population was also noted with 19% (8 of 43) patients developing cytogenetic abnormalities
during follow-up [2, 3].

Based on these encouraging data, a phase 1/2 study evaluated whether the addition of
eltrombopag to frontline IST can further improve patient outcomes [4]. 92 patients were
assigned to receive daily eltrombopag in addition to horse ATG and CsA in three separate
cohorts starting eltrombopag at either day 0 or day 14 and continuing for 3 or 6 months.
Because of the concern for karyotypic evolution [2, 3], patients with cytogenetic
abnormalities were excluded. The overall response rate at 6 months in the three cohorts
ranged between 80–94%, as compared to 66% in the composite historical cohort of 102
Author Manuscript

patients [50, 45]. There was also an encouraging improvement in the rate of complete
response, at 36% with eltrombopag as compared to a historical estimate of 10% with
standard IST. During an initial phase of the study CsA was discontinued at 6 months, with a
relapse rate of 32%–54%, leading to a protocol amendment to extend CsA duration to 2
years [4]. Encouragingly, the rates of chromosomal aberrations were similar to those in
historical controls, ~8% at 2 years of follow-up [4]. Several prospective randomized trials
combining eltrombopag therapy with IST are underway to confirm these findings and to
better assess long-term efficacy and safety, particularly clonal evolution (Table 2). However,
given these encouraging early results and low observed toxicity, we believe that in selected
newly diagnosed SAA/VSAA patients without pre-existing karyotypic abnormalities,
addition of six months of eltrombopag to upfront standard IST should be considered.
Author Manuscript

Cyclosporine Maintenance and Taper


Although there are no definitive data on the optimal duration of CsA, it is clear that early
discontinuation leads to a high rate of early relapse. An 11-year follow-up of a randomized
trial of horse ATG with or without CsA showed that CsA maintenance delays relapses. 26%
of patients required CsA for greater than 6 months due to recurrent cytopenias on
discontinuation [43]. Follow-up of two National Institutes of Health cohorts totaling 102
patients treated with standard IST for whom CsA taper was started at 6 months showed a
cumulative relapse rate of 33% at 5 years of follow-up, with a median time to relapse of 2
years [51]. Compared to historical cohorts that discontinued CsA at 6 months, a tapering
regimen delayed relapse by approximately 1 year [51]. The high relapse rates of 32%–54%
in the recent study of eltrombopag added to upfront IST were also attributed to early CsA
discontinuation, and were improved with extending CsA to a 2 year maintenance [4]. There
Author Manuscript

are limited data on the rate of CsA taper, although an analysis of 33 pediatric AA patients
suggested that a slower taper of <0.3mg/kg/month may lead to fewer relapses [52]. Putting
these data together, our practice (Figure 1) is to continue full dose CsA, targeting therapeutic
CsA trough of 200–300 mcg/L, for approximately 12 months after horse ATG therapy and
until achievement of stable and maximally-improved blood counts, at which time we initiate
a slow taper with no more than 10% dose reduction at a time over the course of
approximately one year.

Curr Treat Options Oncol. Author manuscript; available in PMC 2018 November 16.
Peslak et al. Page 7

Treatment of nonsevere aplastic anemia (NSAA)


Author Manuscript

Unlike the fairly defined guidelines for frontline treatment of SAA/VSAA (Figure 1), the
approach to nonsevere aplastic anemia (NSAA) is more nuanced. The natural history of
patients with NSAA has been evaluated in several retrospective analyses, showing
progression of cytopenias in ~20–67% of NSAA patients [53–56]. Interestingly, NSAA
patients managed in more recent years (1997–2002) appear to have worse outcomes with a
56% 10-year survival as compared to 70% in NSAA patients treated between 1991–1996;
this was associated with a significantly longer interval from diagnosis to treatment in the
more recent cohort (52 versus 102 days) [57]. Expert AA guidelines recommend treating
NSAA patients if they have transfusion dependence or neutropenia [21]. A prospective
randomized study compared horse ATG and CsA to CsA monotherapy in 114 NSAA
patients, showing significantly higher overall response (74% versus 46%) in the ATG and
CsA arm [58]. Transfusion independence was achieved in 90% of ATG/CsA-treated patients
Author Manuscript

compared to only 67% of patients receiving CsA alone; 5-year OS was equivalent in both
groups [58]. The outcomes of a recent cohort of 95 Japanese NSAA patients treated with
horse ATG and CsA were less encouraging, showing a lower 6-month response rate of 55%
and a 10-year failure free survival of 44%, with the majority of patients needing second- and
third-line therapies. The median time to initial treatment was 47 days [59].

More recently, eltrombopag has been proposed as a potential option for NSAA patients, with
a number of ongoing studies studying the safety and efficacy of eltrombopag in combination
with CsA in NSAA (Table 2) [60]. Given the excellent tolerability and efficacy of
eltrombopag in the relapsed/refractory and first-line SAA/VSAA settings (Table 2) [2–4],
we anticipate that eltrombopag-containing regimens would be similarly beneficial in NSAA
and may allow for improved outcomes with lower toxicities in this population.
Author Manuscript

Several other treatments have been investigated in NSAA; however, to date, none have been
demonstrated to be superior to standard IST. In 45 patients with NSAA treated with a
recombinant humanized anti-IL2 receptor antibody daclizumab, 42% achieved a
hematologic response at 3 months [61], although only 25% achieved transfusion
independence at ~5 years of follow-up [62]. An antihelminthic agent levamisole, associated
with immunomodulatory activity, was tested in combination with CsA in 118 Chinese
patients with NSAA; the study found a nearly 100% overall response rate in 42 patients with
newly-diagnosed NSAA and 87% overall response rate in chronic NSAA [63], suggesting
that CsA combined with levamisole may be a promising therapy to be evaluated in future
randomized studies.
Author Manuscript

Clonal evolution
The improvement in long-term survival of AA patients led to an increased appreciation of
the long-term clonal sequelae of AA. ~15% of AA patients treated with IST go on to
develop the late complications of MDS and acute myeloid leukemia (AML) [64, 65, 43].
Approximately 10% of AA patients (range 3–26%) develop cytogenetic changes during the
course of their disease (reviewed in [7]), most commonly monosomy 7/del (7q) and trisomy
8, as well as del (13q), and trisomies of chromosomes 6, 15 and 21. In the context of AA,

Curr Treat Options Oncol. Author manuscript; available in PMC 2018 November 16.
Peslak et al. Page 8

monosomy 7 has been found to correlate with a poor prognosis, including a worse response
Author Manuscript

to IST and increased progression to MDS, while del (13q) and trisomy 8 are associated with
an improved response to IST and a better prognosis (reviewed in [6, 7]).

Newer techniques combining single nucleotide polymorphism arrays (SNP-A) and next-
generation sequencing (NGS) have allowed to more precisely evaluate clonal hematopoiesis
in AA. The majority of AA patients, including over 60% of children with AA, develop
clonal genetic changes [66, 67]. Several recent reviews comprehensively addressed this topic
[6, 7, 68]. The most common clonal abnormality in AA is the development of PNH clones,
which can be detected by flow cytometry as cells lacking glycophophatidyl-inositol-linked
proteins due to a somatic mutation in the PIGA gene [69, 70], found in up to 50% of AA
patients. The second most common is somatic loss of human leukocyte antigen (HLA) loci,
that are detected as either regions of acquired 6p CN-LOH or as inactivating mutations in
HLA class I genes in approximately 17% of AA patients [71–74]. Both PNH clones and
Author Manuscript

HLA loss are hypothesized to occur due to immune escape, and, if present, can be helpful in
corroborating a diagnosis of AA. The presence of even a subclinical PNH clone has been
found to correlate with an improved response to IST [75–79, 70]. In contrast to PNH, the
prognostic impact of somatic HLA loss is less clear [71, 72], with emerging data suggesting
that HLA loss may be best viewed as a marker of a higher immune pathogenicity of a
patient’s inherited HLA alleles [74, 73].

Using targeted NGS of genes recurrently mutated in hematologic malignancies, several


groups identified somatic mutations in MDS-associated genes in AA, detected in up to a
third of adult AA patients [80–83, 66]. The most commonly mutated malignancy-associated
genes in AA are ASXL1, BCOR/BCORL1, and DNMT3A [66, 81, 82]. The prognostic
implications of somatic mutations in MDS-associated genes are not clear, due to the lack of
Author Manuscript

prospective, long-term studies of large numbers of patients carrying these mutations.


Nevertheless, available data suggest that mutations in BCOR and BCORL1 may be
predictive of an improved response to IST [66], and together with mutations in PIGA,
comprise a group with “favorable” prognosis [66]. In the comprehensive study of clonal
hematopoiesis in AA by Yoshizato and colleagues, no other genes were identified to have
prognostic significance individually, perhaps due to the limited statistical power, although
several genes (DNMT3A, ASXL1, TP53, RUNX1, and CSMD1) were associated with
worse OS when analyzed in aggregate [66]. Putting together the available data, it is clear
that the majority of pediatric and adult AA patients develop clonal hematopoiesis, with a
large fraction of adult AA patients having detectable somatic mutations in genes that are
frequently altered in aging and MDS. However, only ~15% of AA patients progress to the
late complications of MDS and leukemia after 10 years of follow-up [64]. Thus, in the
Author Manuscript

absence of prospective longitudinal studies evaluating the prognosis of AA patients carrying


specific MDS-associated somatic mutations, we advise caution when factoring the presence
of somatic mutations into therapeutic decisions in this patient population.

Salvage therapy for relapsed and refractory aplastic anemia


Despite overall improvement in AA outcomes, ~33–35% of patients who initially respond to
IST will relapse during or after CsA taper, while another 35% will be refractory to frontline

Curr Treat Options Oncol. Author manuscript; available in PMC 2018 November 16.
Peslak et al. Page 9

IST [45, 1]. Most patients (~60–68%) who relapse following an initial response to IST can
Author Manuscript

be salvaged with full-dose CsA monotherapy and/or a second course of IST with rabbit ATG
and CsA [84, 52, 85, 42, 86] or transplant. Alternatives to ATG and CsA in relapsed disease
include alemtuzumab, which, in a single-arm prospective study of 25 patients with relapsed
AA, was shown to produce a hematologic response in 56% of patients, with 86% 3-year
survival [87]. Because most patients will respond to a second round of IST [84], in adults,
transplant therapies are usually reserved for relapsed patients who failed an attempt of
salvage with a second course of immunosuppression, while excellent outcomes in children
with salvage BMT after IST failure make MUD-BMT a reasonable second-line option [88].

Compared to relapsed AA patients who previously responded to IST, patients with primary
refractory AA have worse outcomes, and only ~30% of refractory AA patients can be
salvaged with rabbit ATG and CsA [84]. The failure-free survival in refractory pediatric
patients treated with second-line IST can be as poor as 9.5%, as compared to >80% for
Author Manuscript

salvage SCT [89]. Thus, refractory AA patients should be evaluated for salvage allogeneic
transplant options, which may include HLA identical sibling, matched unrelated, or
haploidentical bone marrow transplantation, depending on donor availability. Among non-
transplant therapies, eltrombopag has hematologic response rates of ~40% in refractory AA,
including some trilineage responses, and represents an important option, particularly for
older adults or patients who are poor transplant candidates [2, 3].

There are several additional second- and third-line treatment options for refractory AA, of
which danazol and alemtuzumab are more commonly used. In 48 patients with refractory
AA randomized to receive alemtuzumab versus rabbit ATG with CsA, alemtuzumab was
comparable to rabbit ATG arm, with a hematologic response of 37% and a 3-year survival of
83% [87]. Although androgen therapy has not been found to improve survival in
Author Manuscript

combination with first-line IST [90, 91], a study of 16 refractory AA patients suggests that
androgens may be helpful, particularly in female patients [92]. Cyclophosphamide in
moderate to high doses also has efficacy in refractory AA [93–95], but significant toxicity
with prolonged neutropenia and high rates of infectious have largely limited its use [96, 97].

Summary
AA is a rare, life-threatening, BMF syndrome that requires a systematic and timely approach
to diagnosis and treatment. For a younger patient with a MSD, allogeneic BMT remains the
standard frontline therapy, while other patients should receive frontline immunosuppression
with horse ATG and CsA. Emerging data suggest that addition of eltrombopag to frontline
IST can further improve outcomes and that outcomes following upfront MUD BMT may
now be equivalent to MSD-BMT, at least in pediatric patients. Long-term prospective studies
Author Manuscript

are underway to confirm the safety and efficacy of these approaches. As the outcomes of
MUD and haploidentical transplantation improve and with emergence of eltrombopag as an
effective agent in refractory AA, we expect that outcomes of patients with refractory AA
will improve. Finally, with the recent findings of frequent clonal hematopoiesis in the
majority of AA patients, utmost caution should be exercised in the interpretation of
molecular changes which are common in this patient population and, in the absence of long-
term prospective studies, do not have well-defined prognostic implications.

Curr Treat Options Oncol. Author manuscript; available in PMC 2018 November 16.
Peslak et al. Page 10

Acknowledgments
Author Manuscript

This work was supported by NHLBI T32 HL007439-38 grant to S.A.P, NHLBI K08 HL122306 to T.O., and
NHLBI K08 HL132101-01 and AA & MDS International Foundation Research Grant to D.V.B, and and NIH/
NIDDK R24DK103001 for Penn/CHOP AA studies.

References and Recommended Reading


Papers of particular interest, published recently, have been highlighted as:

• Of importance

•• Of major importance

1. Scheinberg P, Young NS. How I treat acquired aplastic anemia. Blood. 2012; 120(6):1185–96. DOI:
10.1182/blood-2011-12-274019 [PubMed: 22517900]
2. Olnes MJ, Scheinberg P, Calvo KR, Desmond R, Tang Y, Dumitriu B, et al. Eltrombopag and
Author Manuscript

improved hematopoiesis in refractory aplastic anemia. N Engl J Med. 2012; 367(1):11–9. DOI:
10.1056/NEJMoa1200931 [PubMed: 22762314]
••3. Desmond R, Townsley DM, Dumitriu B, Olnes MJ, Scheinberg P, Bevans M, et al. Eltrombopag
restores trilineage hematopoiesis in refractory severe aplastic anemia that can be sustained on
discontinuation of drug. Blood. 2014; 123(12):1818–25. doi:10.1182/blood-2013-10-534743. A
follow-up of a phase 2 study of eltrombopag monotherapy in patients with refractory SAA
showing an overall response of 40%, with a subset of patients achieving trilineage hematopoietic
responses. [PubMed: 24345753]
••4. Townsley DM, Scheinberg P, Winkler T, Desmond R, Dumitriu B, Rios O, et al. Eltrombopag
Added to Standard Immunosuppression for Aplastic Anemia. N Engl J Med. 2017; 376(16):
1540–50. doi:10.1056/NEJMoa1613878. A prospective phase 1–2 study of immunosuppressive
therapy plus eltrombopag in newly-diagnosed SAA showing that addition of eltrombopag
improves hematologic response as compared to historical IST controls. [PubMed: 28423296]
•5. Dufour C, Veys P, Carraro E, Bhatnagar N, Pillon M, Wynn R, et al. Similar outcome of upfront-
unrelated and matched sibling stem cell transplantation in idiopathic paediatric aplastic anaemia.
Author Manuscript

A study on behalf of the UK Paediatric BMT Working Party, Paediatric Diseases Working Party
and Severe Aplastic Anaemia Working Party of EBMT. Br J Haematol. 2015; 171(4):585–94.
doi:10.1111/bjh.13614. A retrospective analysis of 29 consecutive children who underwent
unrelated donor transplantation with Fludarabine/Cyclophosphamide/Alemtuzumab (FCC)
conditioning in an upfront setting, which showed outcomes similar to historical controls treated
with matched sibling donor transplantation and superior to IST and to unrelated donor transplant
after IST failure, suggesting that front-line therapy with unrelated donor BMT may be considered
in selected pediatric patients without a matched sibling donor. [PubMed: 26223288]
6. Ogawa S. Clonal hematopoiesis in acquired aplastic anemia. Blood. 2016; 128(3):337–47. DOI:
10.1182/blood-2016-01-636381 [PubMed: 27121470]
7. Stanley N, Olson TS, Babushok DV. Recent advances in understanding clonal haematopoiesis in
aplastic anaemia. Br J Haematol. 2017; 177(4):509–25. DOI: 10.1111/bjh.14510 [PubMed:
28107566]
8. Young NS, Kaufman DW. The epidemiology of acquired aplastic anemia. Haematologica. 2008;
93(4):489–92. DOI: 10.3324/haematol.12855 [PubMed: 18379007]
Author Manuscript

9. Brown KE, Tisdale J, Barrett AJ, Dunbar CE, Young NS. Hepatitis-associated aplastic anemia. N
Engl J Med. 1997; 336(15):1059–64. DOI: 10.1056/NEJM199704103361504 [PubMed: 9091802]
10. Kim SW, Rice L, Champlin R, Udden MM. Aplastic anemia in eosinophilic fasciitis: responses to
immunosuppression and marrow transplantation. Haematologia (Budap). 1997; 28(3):131–7.
[PubMed: 9283913]
11. Helgadottir H, Kis L, Ljungman P, Larkin J, Kefford R, Ascierto PA, et al. Lethal aplastic anemia
caused by dual immune checkpoint blockade in metastatic melanoma. Ann Oncol. 2017; doi:
10.1093/annonc/mdx177

Curr Treat Options Oncol. Author manuscript; available in PMC 2018 November 16.
Peslak et al. Page 11

12. Wilson DB, Link DC, Mason PJ, Bessler M. Inherited bone marrow failure syndromes in
adolescents and young adults. Ann Med. 2014; 46(6):353–63. DOI:
Author Manuscript

10.3109/07853890.2014.915579 [PubMed: 24888387]


13. Babushok DV, Grignon AL, Li Y, Atienza J, Xie HM, Lam HS, et al. Disrupted lymphocyte
homeostasis in hepatitis-associated acquired aplastic anemia is associated with short telomeres.
Am J Hematol. 2016; 91(2):243–7. DOI: 10.1002/ajh.24256 [PubMed: 26615915]
14. Jerez A, Clemente MJ, Makishima H, Rajala H, Gomez-Segui I, Olson T, et al. STAT3 mutations
indicate the presence of subclinical T-cell clones in a subset of aplastic anemia and
myelodysplastic syndrome patients. Blood. 2013; 122(14):2453–9. doi:blood-2013-04-494930
[pii] 10.1182/blood-2013-04-494930. [PubMed: 23926297]
15. Nakao S, Gale RP. Are mild/moderate acquired idiopathic aplastic anaemia and low-risk
myelodysplastic syndrome one or two diseases or both and how should it/they be treated?
Leukemia. 2016; 30(11):2127–30. DOI: 10.1038/leu.2016.206 [PubMed: 27585953]
16. Camitta BM. Pathogenesis and treatment of aplastic anemia. Rinsho Ketsueki. 1984; 25(4):459–69.
[PubMed: 6381789]
17. Rovo A, Tichelli A, Dufour C. Diagnosis of acquired aplastic anemia. Bone Marrow Transplant.
Author Manuscript

2013; 48(2):162–7. DOI: 10.1038/bmt.2012.230 [PubMed: 23165495]


18. Hochsmann B, Moicean A, Risitano A, Ljungman P, Schrezenmeier H. Supportive care in severe
and very severe aplastic anemia. Bone Marrow Transplant. 2013; 48(2):168–73. DOI: 10.1038/
bmt.2012.220 [PubMed: 23208312]
19. Teramura M, Kimura A, Iwase S, Yonemura Y, Nakao S, Urabe A, et al. Treatment of severe
aplastic anemia with antithymocyte globulin and cyclosporin A with or without G-CSF in adults: a
multicenter randomized study in Japan. Blood. 2007; 110(6):1756–61. DOI: 10.1182/
blood-2006-11-050526 [PubMed: 17526862]
20. Tichelli A, Schrezenmeier H, Socie G, Marsh J, Bacigalupo A, Duhrsen U, et al. A randomized
controlled study in patients with newly diagnosed severe aplastic anemia receiving antithymocyte
globulin (ATG), cyclosporine, with or without G-CSF: a study of the SAA Working Party of the
European Group for Blood and Marrow Transplantation. Blood. 2011; 117(17):4434–41. DOI:
10.1182/blood-2010-08-304071 [PubMed: 21233311]
•21. Killick SB, Bown N, Cavenagh J, Dokal I, Foukaneli T, Hill A, et al. Guidelines for the diagnosis
and management of adult aplastic anaemia. Br J Haematol. 2016; 172(2):187–207. doi:10.1111/
Author Manuscript

bjh.13853. The 2015 update of expert AA guidelines from the British Society for Haematology
(BSH). [PubMed: 26568159]
22. Gupta V, Eapen M, Brazauskas R, Carreras J, Aljurf M, Gale RP, et al. Impact of age on outcomes
after bone marrow transplantation for acquired aplastic anemia using HLA-matched sibling
donors. Haematologica. 2010; 95(12):2119–25. DOI: 10.3324/haematol.2010.026682 [PubMed:
20851870]
23. Shin SH, Jeon YW, Yoon JH, Yahng SA, Lee SE, Cho BS, et al. Comparable outcomes between
younger (40 years) and older (>40 years) adult patients with severe aplastic anemia after HLA-
matched sibling stem cell transplantation using fludarabine-based conditioning. Bone Marrow
Transplant. 2016; 51(11):1456–63. DOI: 10.1038/bmt.2016.171 [PubMed: 27348538]
24. Bacigalupo A, Brand R, Oneto R, Bruno B, Socie G, Passweg J, et al. Treatment of acquired severe
aplastic anemia: bone marrow transplantation compared with immunosuppressive therapy--The
European Group for Blood and Marrow Transplantation experience. Semin Hematol. 2000; 37(1):
69–80. [PubMed: 10676912]
Author Manuscript

25. Yoshida N, Kobayashi R, Yabe H, Kosaka Y, Yagasaki H, Watanabe K, et al. First-line treatment
for severe aplastic anemia in children: bone marrow transplantation from a matched family donor
versus immunosuppressive therapy. Haematologica. 2014; 99(12):1784–91. DOI: 10.3324/
haematol.2014.109355 [PubMed: 25193958]
•26. Bacigalupo A, Socie G, Hamladji RM, Aljurf M, Maschan A, Kyrcz-Krzemien S, et al. Current
outcome of HLA identical sibling versus unrelated donor transplants in severe aplastic anemia:
an EBMT analysis. Haematologica. 2015; 100(5):696–702. doi:10.3324/haematol.2014.115345.
A retrospective analysis of AA patients who underwent transplant between 2005 and 2009 based
on the EBMT registry showing that unrelated donor transplants are not statistically inferior to

Curr Treat Options Oncol. Author manuscript; available in PMC 2018 November 16.
Peslak et al. Page 12

sibling transplants, although they have a higher risk of GVHD. The strongest predictor of
survival continues to be the use of peripheral blood stem cell graft. [PubMed: 25616576]
Author Manuscript

27. Xu LP, Wang SQ, Wu DP, Wang JM, Gao SJ, Jiang M, et al. Haplo-identical transplantation for
acquired severe aplastic anaemia in a multicentre prospective study. Br J Haematol. 2016; 175(2):
265–74. DOI: 10.1111/bjh.14225 [PubMed: 27352174]
28. Xu LP, Jin S, Wang SQ, Xia LH, Bai H, Gao SJ, et al. Upfront haploidentical transplant for
acquired severe aplastic anemia: registry-based comparison with matched related transplant. J
Hematol Oncol. 2017; 10(1):25.doi: 10.1186/s13045-017-0398-y [PubMed: 28107815]
29. DeZern AE, Zahurak M, Symons H, Cooke K, Jones RJ, Brodsky RA. Alternative Donor
Transplantation with High-Dose Post-Transplantation Cyclophosphamide for Refractory Severe
Aplastic Anemia. Biol Blood Marrow Transplant. 2017; 23(3):498–504. DOI: 10.1016/j.bbmt.
2016.12.628 [PubMed: 28013015]
30. Liu Z, Zhang Y, Xiao H, Yao Z, Zhang H, Liu Q, et al. Cotransplantation of bone marrow-derived
mesenchymal stem cells in haploidentical hematopoietic stem cell transplantation in patients with
severe aplastic anemia: an interim summary for a multicenter phase II trial results. Bone Marrow
Transplant. 2017; 52(5):704–10. DOI: 10.1038/bmt.2016.347 [PubMed: 28067873]
Author Manuscript

31. Bertaina A, Merli P, Rutella S, Pagliara D, Bernardo ME, Masetti R, et al. HLA-haploidentical
stem cell transplantation after removal of alphabeta+ T and B cells in children with nonmalignant
disorders. Blood. 2014; 124(5):822–6. DOI: 10.1182/blood-2014-03-563817 [PubMed: 24869942]
32. Schrezenmeier H, Passweg JR, Marsh JC, Bacigalupo A, Bredeson CN, Bullorsky E, et al. Worse
outcome and more chronic GVHD with peripheral blood progenitor cells than bone marrow in
HLA-matched sibling donor transplants for young patients with severe acquired aplastic anemia.
Blood. 2007; 110(4):1397–400. DOI: 10.1182/blood-2007-03-081596 [PubMed: 17475907]
33. Bacigalupo A, Socie G, Schrezenmeier H, Tichelli A, Locasciulli A, Fuehrer M, et al. Bone
marrow versus peripheral blood as the stem cell source for sibling transplants in acquired aplastic
anemia: survival advantage for bone marrow in all age groups. Haematologica. 2012; 97(8):1142–
8. DOI: 10.3324/haematol.2011.054841 [PubMed: 22315497]
34. Purev E, Tian X, Aue G, Pantin J, Vo P, Shalabi R, et al. Allogeneic transplantation using CD34+
selected peripheral blood progenitor cells combined with non-mobilized donor T cells for
refractory severe aplastic anaemia. Br J Haematol. 2017; 176(6):950–60. DOI: 10.1111/bjh.14448
[PubMed: 28169418]
Author Manuscript

35. Storb R, Etzioni R, Anasetti C, Appelbaum FR, Buckner CD, Bensinger W, et al.
Cyclophosphamide combined with antithymocyte globulin in preparation for allogeneic marrow
transplants in patients with aplastic anemia. Blood. 1994; 84(3):941–9. [PubMed: 8043876]
36. Doney K, Leisenring W, Storb R, Appelbaum FR. Primary treatment of acquired aplastic anemia:
outcomes with bone marrow transplantation and immunosuppressive therapy. Seattle Bone
Marrow Transplant Team. Ann Intern Med. 1997; 126(2):107–15. [PubMed: 9005744]
37. Grimaldi F, Potter V, Perez-Abellan P, Veluchamy JP, Atif M, Grain R, et al. Mixed T Cell
Chimerism After Allogeneic Hematopoietic Stem Cell Transplantation for Severe Aplastic Anemia
Using an Alemtuzumab-Containing Regimen Is Shaped by Persistence of Recipient CD8 T Cells.
Biol Blood Marrow Transplant. 2017; 23(2):293–9. DOI: 10.1016/j.bbmt.2016.11.003 [PubMed:
27816648]
38. Maury S, Bacigalupo A, Anderlini P, Aljurf M, Marsh J, Socie G, et al. Improved outcome of
patients older than 30 years receiving HLA-identical sibling hematopoietic stem cell
transplantation for severe acquired aplastic anemia using fludarabine-based conditioning: a
comparison with conventional conditioning regimen. Haematologica. 2009; 94(9):1312–5. DOI:
Author Manuscript

10.3324/haematol.2009.006916 [PubMed: 19734425]


39. Bacigalupo A. How I treat acquired aplastic anemia. Blood. 2017; 129(11):1428–36. DOI:
10.1182/blood-2016-08-693481 [PubMed: 28096088]
•40. Kekre N, Zhang Y, Zhang MJ, Carreras J, Ahmed P, Anderlini P, et al. Effect of antithymocyte
globulin source on outcomes of bone marrow transplantation for severe aplastic anemia.
Haematologica. 2017; 102(7):1291–8. doi:10.3324/haematol.2017.164459. A retrospective
analysis of 833 patients who underwent bone marrow transplant for severe AA, showing that
conditioning regimens with rabbit ATG achieve lower rates of GVHD and improved survival as
compared to horse ATG. [PubMed: 28341733]

Curr Treat Options Oncol. Author manuscript; available in PMC 2018 November 16.
Peslak et al. Page 13

41. Tolar J, Deeg HJ, Arai S, Horwitz M, Antin JH, McCarty JM, et al. Fludarabine-based conditioning
for marrow transplantation from unrelated donors in severe aplastic anemia: early results of a
Author Manuscript

cyclophosphamide dose deescalation study show life-threatening adverse events at predefined


cyclophosphamide dose levels. Biol Blood Marrow Transplant. 2012; 18(7):1007–11. DOI:
10.1016/j.bbmt.2012.04.014 [PubMed: 22546497]
42. Frickhofen N, Kaltwasser JP, Schrezenmeier H, Raghavachar A, Vogt HG, Herrmann F, et al.
Treatment of aplastic anemia with antilymphocyte globulin and methylprednisolone with or
without cyclosporine. The German Aplastic Anemia Study Group. N Engl J Med. 1991; 324(19):
1297–304. DOI: 10.1056/nejm199105093241901 [PubMed: 2017225]
43. Frickhofen N, Heimpel H, Kaltwasser JP, Schrezenmeier H. German Aplastic Anemia Study G.
Antithymocyte globulin with or without cyclosporin A: 11-year follow-up of a randomized trial
comparing treatments of aplastic anemia. Blood. 2003; 101(4):1236–42. DOI: 10.1182/
blood-2002-04-1134 [PubMed: 12393680]
44. Bacigalupo A, Bruno B, Saracco P, Di Bona E, Locasciulli A, Locatelli F, et al. Antilymphocyte
globulin, cyclosporine, prednisolone, and granulocyte colony-stimulating factor for severe aplastic
anemia: an update of the GITMO/EBMT study on 100 patients. European Group for Blood and
Marrow Transplantation (EBMT) Working Party on Severe Aplastic Anemia and the Gruppo
Author Manuscript

Italiano Trapianti di Midolio Osseo (GITMO). Blood. 2000; 95(6):1931–4. [PubMed: 10706857]
45. Scheinberg P, Nunez O, Weinstein B, Biancotto A, Wu CO, Young NS. Horse versus rabbit
antithymocyte globulin in acquired aplastic anemia. N Engl J Med. 2011; 365(5):430–8. DOI:
10.1056/NEJMoa1103975 [PubMed: 21812672]
46. Bussel JB, Cheng G, Saleh MN, Psaila B, Kovaleva L, Meddeb B, et al. Eltrombopag for the
treatment of chronic idiopathic thrombocytopenic purpura. N Engl J Med. 2007; 357(22):2237–47.
DOI: 10.1056/NEJMoa073275 [PubMed: 18046028]
47. Alexander WS, Roberts AW, Nicola NA, Li R, Metcalf D. Deficiencies in progenitor cells of
multiple hematopoietic lineages and defective megakaryocytopoiesis in mice lacking the
thrombopoietic receptor c-Mpl. Blood. 1996; 87(6):2162–70. [PubMed: 8630375]
48. Kimura S, Roberts AW, Metcalf D, Alexander WS. Hematopoietic stem cell deficiencies in mice
lacking c-Mpl, the receptor for thrombopoietin. Proc Natl Acad Sci U S A. 1998; 95(3):1195–200.
[PubMed: 9448308]
49. Ballmaier M, Germeshausen M, Krukemeier S, Welte K. Thrombopoietin is essential for the
Author Manuscript

maintenance of normal hematopoiesis in humans: development of aplastic anemia in patients with


congenital amegakaryocytic thrombocytopenia. Ann N Y Acad Sci. 2003; 996:17–25. [PubMed:
12799278]
50. Scheinberg P, Wu CO, Nunez O, Scheinberg P, Boss C, Sloand EM, et al. Treatment of severe
aplastic anemia with a combination of horse antithymocyte globulin and cyclosporine, with or
without sirolimus: a prospective randomized study. Haematologica. 2009; 94(3):348–54. DOI:
10.3324/haematol.13829 [PubMed: 19181786]
•51. Scheinberg P, Rios O, Scheinberg P, Weinstein B, Wu CO, Young NS. Prolonged cyclosporine
administration after antithymocyte globulin delays but does not prevent relapse in severe aplastic
anemia. Am J Hematol. 2014; 89(6):571–4. A comparison of outcomes of NIH AA cohorts who
were treated with IST followed by an 18-month cyclosporine A taper starting 6 months after IST,
as compared to historical controls who discontinued cyclosporine A 6 months after IST, showed
that cyclosporine A taper delayed relapse by approximately one year, but did not eliminate
relapses. [PubMed: 24971433]
52. Saracco P, Quarello P, Iori AP, Zecca M, Longoni D, Svahn J, et al. Cyclosporin A response and
Author Manuscript

dependence in children with acquired aplastic anaemia: a multicentre retrospective study with
long-term observation follow-up. Br J Haematol. 2008; 140(2):197–205. DOI: 10.1111/j.
1365-2141.2007.06903.x [PubMed: 18173756]
53. Kwon JH, Kim I, Lee YG, Koh Y, Park HC, Song EY, et al. Clinical course of non-severe aplastic
anemia in adults. Int J Hematol. 2010; 91(5):770–5. DOI: 10.1007/s12185-010-0601-1 [PubMed:
20524094]
54. Howard SC, Naidu PE, Hu XJ, Jeng MR, Rodriguez-Galindo C, Rieman MD, et al. Natural history
of moderate aplastic anemia in children. Pediatr Blood Cancer. 2004; 43(5):545–51. DOI: 10.1002/
pbc.20131 [PubMed: 15382271]

Curr Treat Options Oncol. Author manuscript; available in PMC 2018 November 16.
Peslak et al. Page 14

55. Brock K, Goldenberg N, Graham DK, Liang X, Hays T. Moderate aplastic anemia in children:
preliminary outcomes for treatment versus observation from a single-institutional experience. J
Author Manuscript

Pediatr Hematol Oncol. 2013; 35(2):148–52. DOI: 10.1097/MPH.0b013e3182755f36 [PubMed:


23128338]
56. Nishio N, Yagasaki H, Takahashi Y, Muramatsu H, Hama A, Yoshida N, et al. Natural history of
transfusion-independent non-severe aplastic anemia in children. Int J Hematol. 2009; 89(4):409–
13. DOI: 10.1007/s12185-009-0302-9 [PubMed: 19343478]
57. Locasciulli A, Oneto R, Bacigalupo A, Socie G, Korthof E, Bekassy A, et al. Outcome of patients
with acquired aplastic anemia given first line bone marrow transplantation or immunosuppressive
treatment in the last decade: a report from the European Group for Blood and Marrow
Transplantation (EBMT). Haematologica. 2007; 92(1):11–8. [PubMed: 17229630]
58. Marsh J, Schrezenmeier H, Marin P, Ilhan O, Ljungman P, McCann S, et al. Prospective
randomized multicenter study comparing cyclosporin alone versus the combination of
antithymocyte globulin and cyclosporin for treatment of patients with nonsevere aplastic anemia: a
report from the European Blood and Marrow Transplant (EBMT) Severe Aplastic Anaemia
Working Party. Blood. 1999; 93(7):2191–5. [PubMed: 10090926]
Author Manuscript

59. Nishikawa E, Yagasaki H, Hama A, Yabe H, Ohara A, Kosaka Y, et al. Long-term outcomes of 95
children with moderate aplastic anemia treated with horse antithymocyte globulin and
cyclosporine. Pediatr Blood Cancer. 2017; 64(5)doi: 10.1002/pbc.26305
60. [Internet] Cg. Efficacy and Safety of Eltrombopag + CSA in Patients With Moderate Aplastic
Anemia (EMAA) (EMAA). 11 May 2016. Bethesda (MD): National Library of Medicine;
61. Maciejewski JP, Sloand EM, Nunez O, Boss C, Young NS. Recombinant humanized anti-IL-2
receptor antibody (daclizumab) produces responses in patients with moderate aplastic anemia.
Blood. 2003; 102(10):3584–6. DOI: 10.1182/blood-2003-04-1032 [PubMed: 12881307]
62. Sloand EM, Olnes MJ, Weinstein B, Wu C, Maciejewski J, Scheinberg P, et al. Long-term follow-
up of patients with moderate aplastic anemia and pure red cell aplasia treated with daclizumab.
Haematologica. 2010; 95(3):382–7. DOI: 10.3324/haematol.2009.013557 [PubMed: 20207845]
63. Li X, Shao Y, Ge M, Shi J, Huang J, Huang Z, et al. A promising immunosuppressive strategy of
cyclosporine alternately combined with levamisole is highly effective for moderate aplastic
anemia. Ann Hematol. 2013; 92(9):1239–47. DOI: 10.1007/s00277-013-1764-7 [PubMed:
23620110]
Author Manuscript

64. Socie G, Rosenfeld S, Frickhofen N, Gluckman E, Tichelli A. Late clonal diseases of treated
aplastic anemia. Semin Hematol. 2000; 37(1):91–101. [PubMed: 10676914]
65. de Planque MM, Bacigalupo A, Wursch A, Hows JM, Devergie A, Frickhofen N, et al. Long-term
follow-up of severe aplastic anaemia patients treated with antithymocyte globulin. Severe Aplastic
Anaemia Working Party of the European Cooperative Group for Bone Marrow Transplantation
(EBMT). Br J Haematol. 1989; 73(1):121–6. [PubMed: 2803967]
••66. Yoshizato T, Dumitriu B, Hosokawa K, Makishima H, Yoshida K, Townsley D, et al. Somatic
Mutations and Clonal Hematopoiesis in Aplastic Anemia. N Engl J Med. 2015; 373(1):35–47.
doi:10.1056/NEJMoa1414799. A comprehensive analysis of clonal hematopoiesis in 439 patients
with aplastic anemia using NGS showing clonal hematopoiesis in half of the patients, with a third
of the patients carrying mutations in myeloid cancer candidate genes. Mutations in PIGA, BCOR
and BCORL1 correlated with better outcomes, while a subgroup of mutations including ASXL1
were associated with worse outcomes. [PubMed: 26132940]
67. Babushok DV, Perdigones N, Perin JC, Olson TS, Ye W, Roth JJ, et al. Emergence of clonal
hematopoiesis in the majority of patients with acquired aplastic anemia. Cancer Genet. 2015;
Author Manuscript

208(4):115–28. DOI: 10.1016/j.cancergen.2015.01.007 [PubMed: 25800665]


68. Marsh JC, Mufti GJ. Clinical significance of acquired somatic mutations in aplastic anaemia. Int J
Hematol. 2016; 104(2):159–67. DOI: 10.1007/s12185-016-1972-8 [PubMed: 27084249]
69. Tichelli A, Gratwohl A, Wursch A, Nissen C, Speck B. Late haematological complications in
severe aplastic anaemia. Br J Haematol. 1988; 69(3):413–8. [PubMed: 3044440]
70. Sugimori C, Chuhjo T, Feng X, Yamazaki H, Takami A, Teramura M, et al. Minor population of
CD55-CD59- blood cells predicts response to immunosuppressive therapy and prognosis in
patients with aplastic anemia. Blood. 2006; 107(4):1308–14. DOI: 10.1182/blood-2005-06-2485
[PubMed: 16179371]

Curr Treat Options Oncol. Author manuscript; available in PMC 2018 November 16.
Peslak et al. Page 15

71. Katagiri T, Sato-Otsubo A, Kashiwase K, Morishima S, Sato Y, Mori Y, et al. Frequent loss of
HLA alleles associated with copy number-neutral 6pLOH in acquired aplastic anemia. Blood.
Author Manuscript

2011; 118(25):6601–9. DOI: 10.1182/blood-2011-07-365189 [PubMed: 21963603]


72. Betensky M, Babushok D, Roth JJ, Mason PJ, Biegel JA, Busse TM, et al. Clonal evolution and
clinical significance of copy number neutral loss of heterozygosity of chromosome arm 6p in
acquired aplastic anemia. Cancer Genet. 2016; 209(1–2):1–10. DOI: 10.1016/j.cancergen.
2015.10.002 [PubMed: 26702937]
•73. Babushok DV, Duke JL, Xie HM, Stanley N, Atienza J, Perdigones N, et al. Somatic HLA
Mutations Expose the Role of Class I-Mediated Autoimmunity in Aplastic Anemia and its Clonal
Complications. Blood Adv. 2017 Oct 10; 1(22):1900–1910. Targeted NGS of HLA class I alleles
and SNP-A analysis in 66 AA patients showed that 17% of patients have somatic HLA loss, with
loss-of-function mutations targeting four specific HLA class I alleles. AA patients who carried
these HLA alleles had a more severe disease course and more clonal evolution, suggesting that
HLA class I-driven autoimmunity is a driving factor in AA pathogenesis, and likely a major force
in clonal evolution in AA. [PubMed: 28971166]
•74. Zaimoku Y, Takamatsu H, Hosomichi K, Ozawa T, Nakagawa N, Imi T, et al. Identification of an
HLA class I allele closely involved in the autoantigen presentation in acquired aplastic anemia.
Author Manuscript

Blood. 2017; 129(21):2908–16. doi:10.1182/blood-2016-11-752378. A flow-cytometric


screening of 28 AA patients with HLA-B*40:02 for leukocytes that lack the HLA-B*40:02 allele
revealed a high frequency of somatic HLA-B*40:02 allele loss, through 6p LOH and somatic
mutations. The high prevalence of somatic loss of HLA-B*40:02 suggests that antigen
presentation through this HLA-B allele plays a key role in AA pathogenesis. [PubMed:
28232583]
75. Narita A, Muramatsu H, Sekiya Y, Okuno Y, Sakaguchi H, Nishio N, et al. Paroxysmal nocturnal
hemoglobinuria and telomere length predicts response to immunosuppressive therapy in pediatric
aplastic anemia. Haematologica. 2015; 100(12):1546–52. DOI: 10.3324/haematol.2015.132530
[PubMed: 26315930]
76. Kulagin A, Lisukov I, Ivanova M, Golubovskaya I, Kruchkova I, Bondarenko S, et al. Prognostic
value of paroxysmal nocturnal haemoglobinuria clone presence in aplastic anaemia patients treated
with combined immunosuppression: results of two-centre prospective study. Br J Haematol. 2014;
164(4):546–54. DOI: 10.1111/bjh.12661 [PubMed: 24261566]
Author Manuscript

77. Zhao X, Zhang L, Jing L, Zhou K, Li Y, Peng G, et al. The role of paroxysmal nocturnal
hemoglobinuria clones in response to immunosuppressive therapy of patients with severe aplastic
anemia. Ann Hematol. 2015; 94(7):1105–10. DOI: 10.1007/s00277-015-2348-5 [PubMed:
25784221]
78. Tutelman PR, Aubert G, Milner RA, Dalal BI, Schultz KR, Deyell RJ. Paroxysmal nocturnal
haemoglobinuria phenotype cells and leucocyte subset telomere length in childhood acquired
aplastic anaemia. Br J Haematol. 2014; 164(5):717–21. DOI: 10.1111/bjh.12656 [PubMed:
24236830]
79. Maciejewski JP, Rivera C, Kook H, Dunn D, Young NS. Relationship between bone marrow failure
syndromes and the presence of glycophosphatidyl inositol-anchored protein-deficient clones. Br J
Haematol. 2001; 115(4):1015–22. [PubMed: 11843844]
80. Heuser M, Schlarmann C, Dobbernack V, Panagiota V, Wiehlmann L, Walter C, et al. Genetic
characterization of acquired aplastic anemia by targeted sequencing. Haematologica. 2014;
99(9):e165–7. DOI: 10.3324/haematol.2013.101642 [PubMed: 24907358]
•81. Kulasekararaj AG, Jiang J, Smith AE, Mohamedali AM, Mian S, Gandhi S, et al. Somatic
Author Manuscript

mutations identify a subgroup of aplastic anemia patients who progress to myelodysplastic


syndrome. Blood. 2014; 124(17):2698–704. A targeted NGS analysis of 150 AA patients
showing a that a fifth of AA patients carry somatic mutations in genes associated with myeloid
malignancies. Most frequently, mutations involve ASXL1, DNMT3A and BCOR. Presence of
somatic mutations was associated with 40% risk of transformation to MDS. DOI: 10.1182/
blood-2014-05-574889 [PubMed: 25139356]
82. Lane AA, Odejide O, Kopp N, Kim S, Yoda A, Erlich R, et al. Low frequency clonal mutations
recoverable by deep sequencing in patients with aplastic anemia. Leukemia. 2013; 27(4):968–71.
doi:leu201330 [pii] 10.1038/leu.2013.30. [PubMed: 23370706]

Curr Treat Options Oncol. Author manuscript; available in PMC 2018 November 16.
Peslak et al. Page 16

83. Huang J, Ge M, Lu S, Shi J, Li X, Zhang J, et al. Mutations of ASXL1 and TET2 in aplastic
anemia. Haematologica. 2015; 100(5):e172–5. DOI: 10.3324/haematol.2014.120931 [PubMed:
Author Manuscript

25596262]
84. Scheinberg P, Nunez O, Young NS. Retreatment with rabbit anti-thymocyte globulin and
ciclosporin for patients with relapsed or refractory severe aplastic anaemia. Br J Haematol. 2006;
133(6):622–7. DOI: 10.1111/j.1365-2141.2006.06098.x [PubMed: 16704436]
85. Kamio T, Ito E, Ohara A, Kosaka Y, Tsuchida M, Yagasaki H, et al. Relapse of aplastic anemia in
children after immunosuppressive therapy: a report from the Japan Childhood Aplastic Anemia
Study Group. Haematologica. 2011; 96(6):814–9. DOI: 10.3324/haematol.2010.035600 [PubMed:
21422115]
86. Fuhrer M, Burdach S, Ebell W, Gadner H, Haas R, Harbott J, et al. Relapse and clonal disease in
children with aplastic anemia (AA) after immunosuppressive therapy (IST): the SAA 94
experience. German/Austrian Pediatric Aplastic Anemia Working Group. Klin Padiatr. 1998;
210(4):173–9. DOI: 10.1055/s-2008-1043875 [PubMed: 9743949]
87. Scheinberg P, Nunez O, Weinstein B, Wu CO, Young NS. Activity of alemtuzumab monotherapy
in treatment-naive, relapsed, and refractory severe acquired aplastic anemia. Blood. 2012; 119(2):
Author Manuscript

345–54. DOI: 10.1182/blood-2011-05-352328 [PubMed: 22067384]


88. Dufour C, Pillon M, Socie G, Rovo A, Carraro E, Bacigalupo A, et al. Outcome of aplastic
anaemia in children. A study by the severe aplastic anaemia and paediatric disease working parties
of the European group blood and bone marrow transplant. Br J Haematol. 2015; 169(4):565–73.
DOI: 10.1111/bjh.13297 [PubMed: 25683884]
89. Kosaka Y, Yagasaki H, Sano K, Kobayashi R, Ayukawa H, Kaneko T, et al. Prospective multicenter
trial comparing repeated immunosuppressive therapy with stem-cell transplantation from an
alternative donor as second-line treatment for children with severe and very severe aplastic anemia.
Blood. 2008; 111(3):1054–9. DOI: 10.1182/blood-2007-08-099168 [PubMed: 17989314]
90. Champlin RE, Ho WG, Feig SA, Winston DJ, Lenarsky C, Gale RP. Do androgens enhance the
response to antithymocyte globulin in patients with aplastic anemia? A prospective randomized
trial. Blood. 1985; 66(1):184–8. [PubMed: 4005428]
91. Doney K, Pepe M, Storb R, Bryant E, Anasetti C, Appelbaum FR, et al. Immunosuppressive
therapy of aplastic anemia: results of a prospective, randomized trial of antithymocyte globulin
(ATG), methylprednisolone, and oxymetholone to ATG, very high-dose methylprednisolone, and
Author Manuscript

oxymetholone. Blood. 1992; 79(10):2566–71. [PubMed: 1586708]


92. Chuhjo T, Yamazaki H, Omine M, Nakao S. Danazol therapy for aplastic anemia refractory to
immunosuppressive therapy. Am J Hematol. 2008; 83(5):387–9. DOI: 10.1002/ajh.21118
[PubMed: 18161784]
93. Tisdale JF, Dunn DE, Geller N, Plante M, Nunez O, Dunbar CE, et al. High-dose
cyclophosphamide in severe aplastic anaemia: a randomised trial. Lancet. 2000; 356(9241):1554–
9. DOI: 10.1016/S0140-6736(00)03126-3 [PubMed: 11075769]
94. Brodsky RA, Sensenbrenner LL, Jones RJ. Complete remission in severe aplastic anemia after
high-dose cyclophosphamide without bone marrow transplantation. Blood. 1996; 87(2):491–4.
[PubMed: 8555470]
95. Zhang F, Zhang L, Jing L, Zhou K, Wang H, Peng G, et al. High-dose cyclophosphamide
compared with antithymocyte globulin for treatment of acquired severe aplastic anemia. Exp
Hematol. 2013; 41(4):328–34. DOI: 10.1016/j.exphem.2013.01.001 [PubMed: 23313080]
96. Brodsky RA, Chen AR, Dorr D, Fuchs EJ, Huff CA, Luznik L, et al. High-dose cyclophosphamide
Author Manuscript

for severe aplastic anemia: long-term follow-up. Blood. 2010; 115(11):2136–41. DOI: 10.1182/
blood-2009-06-225375 [PubMed: 20018919]
97. Scheinberg P, Townsley D, Dumitriu B, Scheinberg P, Weinstein B, Daphtary M, et al. Moderate-
dose cyclophosphamide for severe aplastic anemia has significant toxicity and does not prevent
relapse and clonal evolution. Blood. 2014; 124(18):2820–3. DOI: 10.1182/blood-2014-05-573642
[PubMed: 25185712]
98. [Internet] Cg. Eltrombopag Combined With Cyclosporine as First Line Therapy in Patients With
Severe Acquired Aplastic Anemia (SOAR). 10 May 2017. Bethesda (MD): National Library of
Medicine;

Curr Treat Options Oncol. Author manuscript; available in PMC 2018 November 16.
Peslak et al. Page 17

99. [Internet] Cg. hATG+CsA vs hATG+CsA+Eltrombopag for SAA (RACE). 10 October 2016.
Bethesda (MD): National Library of Medicine;
Author Manuscript
Author Manuscript
Author Manuscript
Author Manuscript

Curr Treat Options Oncol. Author manuscript; available in PMC 2018 November 16.
Peslak et al. Page 18
Author Manuscript
Author Manuscript
Author Manuscript

Figure 1. Approach to diagnosis and treatment of acquired aplastic anemia


Initial screening evaluation of a patient with aplastic anemia is required to document
pancytopenia with a hypocellular marrow, followed by testing to exclude alternative
diagnoses. Aplastic anemia severity and outcomes of a transplant evaluation factor into
determining an optimal treatment strategy. Patients with Severe or Very Severe Aplastic
Anemia (SAA/VSAA) 40 years of age or younger with an HLA-matched sibling donor
should undergo an evaluation for an allogeneic bone marrow transplant; older patients or
Author Manuscript

patients without an HLA-identical sibling donor should be evaluated for frontline


immunosuppressive therapy (IST) with horse ATG and CsA. **Based on recent data
showing superior hematologic outcomes with the addition of eltrombopag [4], addition of 6
months of eltrombopag to standard IST in patients without pre-existing cytogenetic
abnormalities can be considered. Cyclosporine A should be continued for ~12 months of
therapy, followed by a slow taper to reduce relapse rates. Salvage therapies include
alternative transplant modalities and a variety of nontransplant options. AA, aplastic anemia;

Curr Treat Options Oncol. Author manuscript; available in PMC 2018 November 16.
Peslak et al. Page 19

PNH, paroxysmal nocturnal hemoglobinuria; 6p CN-LOH, copy number-neutral loss of


Author Manuscript

heterozygosity of chromosome arm 6p; alloSCT, allogeneic stem cell transplant; NSAA,
nonsevere aplastic anemia; SAA, severe aplastic anemia; VSAA, very severe aplastic
anemia. *Cellularity criteria are determined on adequate bone marrow biopsy, and
hypoplastic marrow can either be diagnosed on total cellularity or on bone marrow biopsy
with <50 percent normal cellularity in which < 30 percent of the cells are hematopoietic.
HLA, human leukocyte antigen; alloBMT, allogeneic bone marrow transplant; IST,
immunosuppressive therapy; hATG, horse antithymocyte globulin; CsA, cyclosporine A;
CR, complete response; PR, partial response; Cy, cyclophosphamide; MRD, matched related
donor; MUD, matched unrelated donor; haplo BMT, haploidentical bone marrow transplant.
Author Manuscript
Author Manuscript
Author Manuscript

Curr Treat Options Oncol. Author manuscript; available in PMC 2018 November 16.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript

Table 1
Diagnostic evaluation of a patient with suspected acquired aplastic anemia
A variety of testing modalities in addition to a detailed personal/family history and exposure history are required both in the initial screening evaluation as
well as the subsequent exclusion of alternative diagnoses.
Peslak et al.

Initial screening evaluation


Peripheral blood CBC with differential, peripheral blood smear, reticulocyte count
Complete metabolic panel, LDH, haptoglobin, coagulation parameters

Bone marrow aspirate and biopsy with Bone marrow aspirate and biopsy
ancillary studies Metaphase cytogenetics and FISH panel for MDS-associated chromosomal abnormalities of chromosomes 5, 7, 8, 20
* Molecular studies

Exclusion of alternative diagnoses


Infectious HIV, Hepatitis B/C, parvovirus B19 PCR, EBV, CMV, bacterial, fungal (+/− mycobacterial testing)

Inherited bone marrow failure Detailed family history focusing on cytopenias, congenital abnormalities, cancers and lung and liver pathology
Chromosomal breakage testing for Fanconi anemia
Lymphocyte telomere length for Dyskeratosis congenita
Additional syndrome-specific testing if personal or family history are suggestive of specific disorders (IBMF, HLH)

Lymphoproliferative Flow cytometry and T-cell receptor rearrangement testing for clonal LGL expansion

Medication or toxin-related Detailed drug and occupational exposure history, with attention to excessive alcohol intake, antibiotics, prior cytotoxic chemotherapeutic agents, and
immune-activating agents (e.g. interferon and checkpoint blockade inhibitors)

Nutritional Vitamin B12, folate, copper, iron studies, ferritin

Rheumatologic Antinuclear antibody, rheumatoid factor, erythrocyte sedimentation rate, C-reactive protein

Others (rare) Exclude other rare etiologies of pancytopenia with a hypocellular marrow: e.g. graft-versus-host disease, HLH

CBC, complete blood count; LDH, lactate dehydrogenase; MPN myeloproliferative neoplasm; FISH, fluorescence in-situ hybridization; MDS, myelodysplastic syndrome; HIV, human immunodeficiency
virus; EBV, Epstein-Barr virus; CMV, cytomegalovirus; DEB, diepoxybutane; SLE, systemic lupus erythematosus; RA, rheumatoid arthritis; IBMF, inherited bone marrow failure; HLH, hemophagocytic
lymphohistiocytosis; LGL, large granular lymphocyte.
*
Although in current clinical practice molecular sequencing panels of somatic mutations in hematologic malignancy-associated genes are frequently included and can provide useful supporting information,
in isolation, presence of somatic mutations in patients with AA should be interpreted with caution due to their high frequency in this patient population and uncertain prognostic significance (see text).

Curr Treat Options Oncol. Author manuscript; available in PMC 2018 November 16.
Page 20
Author Manuscript Author Manuscript Author Manuscript Author Manuscript

Table 2

Eltrombopag in the treatment of aplastic anemia.

Type of study Inclusion/Exclusion Patients, n Median Treatment arms Duration of Follow-up Outcomes Clonal Evolution
age, yrs
Peslak et al.

(range)
Townsley et Phase 1/2 Inclusion: 92 32 (3–82) hATG/CsA + eltrombopag Up to 24 months Eltrombopag + 7/92 (7.6%)
al. (2017) [4] Age >2 with previously hATG/CsA developed clonal
untreated SAA showed ORR of abnormalities on
Exclusion: 80% and CR of cytogenetics
FA, liver impairment, or 36%, improved
abnormal cytogenetics from historical
cohorts with ORR
66%, CR 10%

SOAR trial Phase 2 Inclusion 50 (target) N/A CsA +/− eltrombopag Planned for up to 60 Pending trial; Planned evaluation
(pending) Age ≥6 with SAA months primary end point for clonal evolution
[98] Exclusion: hematologic to PNH, MDS, or
Prior CsA, alemtuzumab, response at 6 acute leukemia
ATG, or TPO-R agonists; months
diagnosis of FA or
abnormal cytogenetics

EBMT-SAA Phase 3 Inclusion: 200 (target) N/A hATG/CsA +/− eltrombopag Planned for at least 2 Pending trial; Planned evaluation
Working Age >14 with SAA/ years primary end point for clonal evolution
Party RACE VSAA complete response to PNH or clonal
trial Exclusion: at 3 months cytogenetics
(pending) Prior ATG; diagnosis of
[99] FA, DC, or MDS

Höchsmann Phase 2/3 Inclusion: 29 (target) N/A CsA +/− eltrombopag Planned for 6 months Pending trial; Planned evaluation
et al. EMAA Age ≥ 2 with NSAA or after last dose primary end point for clonal evolution
trial unilineage BMF; PLT eltrombopag hematologic to PNH or clonal
(pending) transfusion dependent response at 6 cytogenetics
[60] Exclusion: months
ATG therapy in prior 6
months; diagnosis of FA

Olnes et al. Phase 2 with Inclusion: 43 44 (17–77) Eltrombopag Up to 4 years follow up ORR at 3–4 8/43 patients
(2012) extension Age >12 years with SAA months of 40% (18.6%) developed
Desmond et refractory to ≥1 IST (17/43), including clonal abnormalities
al. (2014) [2, regimen; PLT <30K/uL 1 patient with on cytogenetics

Curr Treat Options Oncol. Author manuscript; available in PMC 2018 November 16.
3] Exclusion: trilineage
Diagnosis of FA, familial response, 9
marrow failure, or DC patients with PLT
transfusion
independence, and
8 patients with
neutrophil
response

SAA, severe aplastic anemia; VSAA, very severe aplastic anemia; NSAA, nonsevere aplastic anemia; PLT, platelet; ORR, overall response rate; CR, complete response; FA, Fanconi anemia; DC,
Dyskeratosis congenita, hATG, horse antithymocyte globulin; CsA, cyclosporine A; IST, immunosuppressive therapy; EMBT-SAA, European Society for Blood and Marrow Transplantation Severe Aplastic
Page 21
Author Manuscript Author Manuscript Author Manuscript Author Manuscript
Anemia; RACE, Randomized multicenter study comparing horse Antithymocyte globuline (hATG) + Cyclosporine A (CsA) with or without Eltrombopag; EMAA, Efficacy and Safety of Eltrombopag +
CsA in Patients with Moderate Aplastic Anemia; SOAR, Eltrombopag Combined With Cyclosporine as First Line Therapy in Patients With Severe Acquired Aplastic Anemia.
Peslak et al.

Curr Treat Options Oncol. Author manuscript; available in PMC 2018 November 16.
Page 22

You might also like