You are on page 1of 17

Expert Review of Hematology

ISSN: 1747-4086 (Print) 1747-4094 (Online) Journal homepage: http://www.tandfonline.com/loi/ierr20

Updates on the pathophysiology and treatment of


aplastic anemia: a comprehensive review

Prajwal Chaitanya Boddu & Tapan Mahendra Kadia

To cite this article: Prajwal Chaitanya Boddu & Tapan Mahendra Kadia (2017): Updates on the
pathophysiology and treatment of aplastic anemia: a comprehensive review, Expert Review of
Hematology, DOI: 10.1080/17474086.2017.1313700

To link to this article: http://dx.doi.org/10.1080/17474086.2017.1313700

Published online: 28 Apr 2017.

Submit your article to this journal

Article views: 4

View related articles

View Crossmark data

Full Terms & Conditions of access and use can be found at


http://www.tandfonline.com/action/journalInformation?journalCode=ierr20

Download by: [The UC San Diego Library] Date: 30 April 2017, At: 10:41
EXPERT REVIEW OF HEMATOLOGY, 2017
https://doi.org/10.1080/17474086.2017.1313700

REVIEW

Updates on the pathophysiology and treatment of aplastic anemia: a


comprehensive review
Prajwal Chaitanya Boddu and Tapan Mahendra Kadia
Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA

ABSTRACT ARTICLE HISTORY


Introduction: The past decade or longer has witnessed an acceleration in our understanding of Received 22 December 2016
previously developed immune system and clonal evolution mechanisms, and the genesis of more Accepted 28 March 2017
novel concepts of telomere attrition. Many of these concepts are steadily finding their way into KEYWORDS
translation in various aspects of clinical practice, and provide prospects to improve AA management Aplastic; telomere;
and inform therapeutic strategy development. In this review, we intend to discuss the pathophysiology immunosuppressive; clonal
and treatments with an emphasis on most recent developments to provide an update on our under- evolution; eltrombopag;
standing of disease mechanisms. alemtuzumab;
Areas covered: A literature search was undertaken addressing various aspects of pathophysiology with antithymocyte globulin;
a focus on the role of immune system repertoire, telomeres and mutational events surrounding AA. We transplant
also reviewed upon the temporal evolution of treatment strategies in AA to the contemporary manage-
ment of today and commented briefly upon the more recently investigated novel therapies and their
expanding niche especially in the transplant and salvage setting.
Expert commentary: Immune-mediated destruction of hematopoietic stem and progenitor cells,
leading to a marrow devoid of hematopoietic elements, and peripheral pancytopenia are the hallmarks
of AA. Recent studies have shed light on another facet of the disease – as a clonal disorder character-
ized by karyotypic abnormalities, genomic instability, telomere attrition, and recurrent somatic muta-
tions reminiscent of myeloid malignancies. Further understanding of this underlying pathophysiology
can help in improving prognostication and treatment of this disease.

1. Introduction transplantation regimens, intensifying immunosuppressive strate-


gies and enhancing supportive care, over several decades of
The first formal description dates back to 1888 when Paul Ehrlich
treatment, has transformed AA into a disease now typified by
described a case of a young pregnant woman who had developed
greater than 80% 10-year survival expectancy.
profound anemia, bleeding, and high fever that eventually proved
Aplastic anemia, a misnomer, is an acquired marrow failure
fatal. The term aplastic anemia (AA) was introduced, 16 years later,
syndrome characterized by findings of peripheral pancytopenia
by Chauffard to describe this condition, characterized morpholo-
and a hypoplastic bone marrow [2]. Aplastic anemia shares clinical
gically by a yellow fatty bone marrow. Since its earliest descriptions
manifestations and some of its marrow morphological findings
until mid-1960s, the inexorably pernicious, uniformly fatal clinical
with other acquired (hypoplastic myelodysplastic syndrome
picture of this disease was accurately depicted in Wintrobe’s
(MDS), hypoplastic leukemias such as hairy cell leukemia, large
description ‘. . .more or less rapidly fatal. . . death ensues in the
granular lymphocytosis, paroxysmal nocturnal hemoglobinuria)
course of a few weeks or life may linger as long as six months.’
and congenital (Fanconi anemia, dyskeratosis congenita) bone
The advent of allogenic bone marrow transplantation in the late
marrow failure syndromes which is why diagnosis requires a care-
1960s and immunosuppressive therapy with antilymphocyte sera
fully constructed history and exhaustive exclusion of potential
in the 1970s proved to be major therapeutic successes leading to a
alternative etiologies – a rather difficult task of morphologic dis-
significant alteration in the natural history of the disease and
tinction based on subtleties in marrow findings, sometimes fre-
markedly improved survival outcomes. Early observations
quenting repeat marrow investigations.
spawned a series of investigations clarifying the autoimmune
Both AA and hypoplastic MDS share histological character-
nature of the disease and were subsequently followed by intensi-
istics of a hypocellular marrow and distinction relies heavily
fication efforts on previously existing immunosuppressive strate-
upon other histologic and flow cytometric characteristics such
gies. In this context, various studies observed that the addition of
as megakaryocytic and CD34+ blast numeration which are low
cyclosporine (Cys) to antithymocyte horse sera furnishes as a
in aplastic anemia and high in hypoplastic MDS, respectively
highly effective immunosuppressive strategy establishing the
[3]. Also, h-MDS is characterized by the presence of dysmega-
combination’s role as the current immunosuppressive treatment
karyocytopoiesis while their absence favors the diagnosis of
of choice [1]. The collective progress made in refining
aplastic anemia. A positive family history, abnormal

CONTACT Tapan Mahendra Kadia tkadia@mdanderson.org Department of Leukemia, UT MD Anderson Cancer center, 1515 Holcombe Blvd., Unit 428,
Houston, TX 77030, USA
© 2017 Informa UK Limited, trading as Taylor & Francis Group
2 P. C. BODDU AND T. M. KADIA

clinicomorphologic features, and laboratory findings showing defects in the progenitor cells. These defects remain sustained
increased chromosomal fragility or telomere shortening sug- even after marrow recovery and reconstitution of blood counts
gest a congenital form of AA [4,5]. Additional diagnostic clues [19]. The low LTC-IC numbers, as determined by these assays, do
may be obtained from blood film examination such as the not correlate with marrow recovery and this may be due to the
presence of monocytopenia and hairy cells (in hairy cell leu- massive compensatory reservoir of hematopoietic progeni-
kemia) [6]. Aplastic anemia may be associated with other tors [19].
disease entities such as paroxysmal nocturnal hemoglobinuria, HSCs have been observed to be true first responders, either
hepatitides, and autoimmune disorders and patients with pre- through direct or indirect signaling, to various inflammatory
sumptive AA should be evaluated for the same [7–9]. and infectious insults. Activated HSCs rapidly switch after with-
Pertinently, prompt diagnosis and treatment are essential to drawal of the inciting stressor to a quiescent state, a requisite
optimal management of AA given that short diagnosis to for preservation of self-renewal potential [20]. It is for this
treatment interval times is known to positively affect overall reason that a tight regulatory control of inflammatory cascade
survival [10,11]. signaling is essential for long-term maintenance of the HSC
Severity grading of AA, originally developed in the 1970s populations [21]. Prolonged activation consequent to dysre-
by study groups to select patients most appropriate for bone gulated signaling induces functional fatigue and impairs HSCs’
marrow transplant, has since undergone minor refinements self-renewal potential [22]. This eventually leads to depletion
and continues to find great prognostic and therapeutic rele- of HSC populations, as is observed in acquired AA. The inflam-
vance in current day management. Aplastic anemia is graded matory milieu in the setting of immune destruction of stem
primarily on peripheral blood count values into (1) severe and progenitor cells leads to depletion of HSCs. These defec-
aplastic anemia (SAA): (a) bone marrow cellularity (BMC) tive HPCs have been found to be insensitive to growth factor
<25% (or) <50% normal cellularity in which <30% are hema- treatments and this therapeutic approach may prove detri-
topoietic and (b) two of three additional criteria i.e. absolute mental as they are expected to relatively increase the apop-
reticulocyte count <20k/µL, absolute neutrophil count (ANC) tosis-susceptible proliferative fraction.
<500/µL, or platelet count <20k/µL; (2) very severe aplastic Apart from the more obvious quantitative deficits, HPCs
anemia (VSAA): SAA criteria + ANC <200/µL; (3) moderate/non may also suffer intrinsic qualitative defects. Stem cells in AA
severe: BMC <30% with absence of severe pancytopenia but patients have been demonstrated to show reduced clono-
decrease of at least two of three blood counts below normal genic potential in short-term primary and secondary colony
[12–14]. assays [23]. Detailed transcriptome analyses have identified
upregulation of several cell-cycle inhibitory and proapoptotic
genes and decreased expression of cell cycle promoting genes
2. Pathophysiology
and antiapoptotic genes in AA samples compared with
Bone marrow hematopoiesis constitutes a well-orchestrated healthy controls [24]. Additionally, sublethal DNA damage
developmental process of proliferation and maturation of mar- can manifest as chromosomal aberrations such as mutations
row cellular elements from pluripotent hematopoietic stem cell and unbalanced translocations and pave the way for clonal
(HSC) precursors. HSCs have the remarkable property of robust evolution toward more malevolent complications of marrow
self-renewal and a potential to differentiate along multiple line- injury including myelodysplasia and leukemia.
age pathways. It has been estimated that about 1 × 104 HSCs are Various etiological drivers mediating HSC destruction have
present in the human bone marrow of which only one-tenth are been identified including radiation, chemicals/toxins, drugs,
actively contributing to hematopoiesis at any given time while viruses, among others. Radiation causes HSC injury through a
the remaining fraction dwell in quiescence [15]. Progenitor and multitude of mechanisms including direct lethal damage of
stem cells, although not identifiable morphologically in a bone DNA through induction of double-stranded breaks and free
marrow, can be distinguished flow cytometrically by the pre- radical species formation, acceleration of HSC senescence
sence of CD34, a glycosylated transmembrane protein involved leading to decrease clonogenesis, induction of HSC differen-
in stem-cell stromal adhesion and maintenance of a phenotypi- tiation at the expense of compromising self-renewal capacity,
cally undifferentiated state [16]. In this regard, there also exists a and via destruction of the highly supportive niche surrounding
minor fraction of CD34 negative HSC population with marrow the HSCs [25,26]. Unlike most other mechanisms, radiation-
repopulating capacity. Flow cytometric and immuno-phenotypic mediated HSC depletion represents a more permanent form of
numeration reveal a profound deficit of the hematopoietic and stem-cell injury [26,27].
progenitor cell compartment in AA [17]. These numbers may The list of drugs, toxins, and chemicals implicated in caus-
drop to lower than 1% of the original HSC population in severe ing bone marrow aplasia is exhaustive and the interested
AA [17]. The most profound deficits occur in the mitotically active reader may refer to other comprehensive texts for a detailed
CD34+ Kit+ and uncommitted progenitor (CD34+CD38−) subset review. Chemical-mediated HSC injury may be due to (a) direct
of HPCs but can also involve the more lineage-committed CD34+ dose-dependent or idiosyncratic toxicity mediated by parent
CD38+ fractions [17]. Surrogate in-vitro assays, such as the long- drug or its metabolites, or (b) immune-mediated HSC destruc-
term culture-initiating cells assay (LTC-IC), have been developed tion by drug–antigen formation [2]. Chloramphenicol, a pro-
to study the properties of primitive human marrow progenitors totype for drug-related AA, exhibits two distinct modes of
and correlate well with the kinetic properties of CD34+ stem cells marrow aplasia – (a) a dose-dependent reversible type and
and their primary colony formations [18]. Data available through (b) a dose-independent idiosyncratic type from toxic metabo-
these assay studies confirm marked quantitative and qualitative lite injury [28]. Benzene, a ubiquitous and highly volatile
EXPERT REVIEW OF HEMATOLOGY 3

petrochemical i.e. used extensively in the hydrocarbon/auto- patients and skewed toward oligo clonal patterns with
mobile industry, has a more consistent marrow aplastic effect highly restricted Vbeta chain sequences [30,31]. In fact, the
compared with other well-studied agents. Models of drug- presence, disappearance, and return of these highly skewed
immune system interaction include immune activation via immune-dominant clonotypes may help predict, follow
drug–hapten–antigen formation and direct stimulation of responses to, and detect relapse after immunosuppressive
immune receptors through non-covalent pharmacological therapy, respectively [31,32]. The putative autoantigens
interactions. Drug-induced AA responds to immunosuppres- against which these T cells react are not clearly known
sive treatments and the distinction does not confer a different but serum autoantibody detection studies suggest multiple
prognosis or therapy response compared with other etiologi- potential candidates. Autoantibodies against antigens
cal types of AA. Infections with EBV, CMV, VZV, HHV-6, parvo- namely diazepam-binding inhibitor-related protein 1 (DRS-
virus B19, HIV, hepatitis A and C have been implicated in 1), kinectin (KTN1), postmeiotic segregation increased 1
aplastic anemia, with only human parvovirus B19 and HHV-6 (PMS1), heterogeneous nuclear ribonucleoprotein K (hnRNP
demonstrated to have a direct cytotoxic effect on the progeni- K) among others have been identified [33]. The potential
tors [29]. pathogenic and predictive role of these antibodies in AA
pathophysiology and immune therapy response, respec-
tively, may have clinical relevance and warrants further
2.1. The T-cell repertoire
investigation.
A large body of evidence supports the proposition of AA The role of T helper (CD4+) cells in AA is less well
being an immune-mediated phenomenon. In this regard, AA characterized compared with their CD8+ T counterparts.
serves as a bone marrow prototype of misdirected immune All subsets of T helper cells i.e. IFN-γ-producing Th1 cells,
response. Response to immunosuppressive therapies in IL-4-producing Th2 cells, T-regulatory (T-reg) cells, and Th17
majority of AA cases provides compelling clinical evidence cells have been implicated to play a role in AA. Similar to
attesting to the immune-mediated nature of the disease. CD8+ T cells, T helper cells show clonal expansion with
Clinical evidence is corroborated by multiple studies con- restriction in their TCR sequences [34]. Activated and resting
firming clonal expansions of activated T cells directed T-reg cells are reduced and are functionally defective with a
against marrow progenitors. Although plenty remains to suboptimal ability to suppress effector T-cell activity [34].
be clarified regarding the exact nature of the pathogenic Kordasti et al. were able to characterize a distinct T-reg
immune response and of putative antigens involved, it subsets of predictive value to immunosuppressive therapy
appears that both adaptive and innate immune system (IST) response in AA. The investigators identified a particular
machineries are involved in contributing to hematopoietic T-reg subset, with phenotypic expression of CD95, CCR4,
progenitor injury (Figure 1). and CD45RO within FOXP3high, CD127low, to predominate
Available evidence suggests a gross homeostatic dysre- among treatment responders [35]. In addition, there exists
gulation of the T-cell repertoire. Among the T-lymphocyte a polarization shift in favor of Th-1 over Th-2 response
subsets, CD8+ T cells have the best characterized role in AA- resulting in increased production of IFN-γ, a potent stimu-
immune pathogenesis. CD8+ T cells are increased in AA lator of CD8+ T cells [36]. Finally, although the contribution

Figure 1. A model of T-cell immune mediated stem cell progenitor cell injury. T-cells are actively involved in the disease process with CD8 T cells orchestrating a
direct cellular mediated cytotoxicity. IFN-γ and TNF-Alpha are major cytokine mediators responsible for activating cellular death/apoptotic machinery including Fas/
FasL pathway (highlighted in yellow). NK-cells are also involved, as demonstrated by upregulation of bound NKG2D ligands on the stem cell progenitor and
decrease in soluble NKG2D ligands. Full color available online.
4 P. C. BODDU AND T. M. KADIA

of Th17 cells in AA pathogenesis remains contentious, regulatory factor-1) through a complex network of signal
increased numbers in blood samples of AA patients with intermediates via the Fas/FasL and TRAIL pathways [37,45]
an inverse relationship to counter regulatory T-reg cells (Figure 1). Cytokine polymorphisms involving TNF-α, IFN-γ,
suggest a potential role early in the course of the dis- and other cytokines like TGF-β are overrepresented in cer-
ease [34]. tain populations of AA and may have an influence on dis-
Natural Killer cells are key mediator cells in innate immu- ease expression independent of HLA [46].
nity responses. Liu et al. demonstrated that NK cell subset
of lymphocytes is severely reduced in active AA and
increases dramatically with responses to immunosuppres- 2.3. The role of telomeres
sive therapy [37]. The NKG2D protein–ligand system repre-
Telomeres are short guanine-rich sequences at the ends of
sents one of the best characterized pathways in NK-cell-
linear chromosomes in all eukaryotic cells, varying in size
mediated immunity. Hanaoka et al. demonstrated increased
from 5 to up to 20 kilobases in length depending upon the
expressions of various stress-inducible NKG2D ligands and
age of the organism, activity of preservative telomerases
were able to preserve hematopoietic colony formations by
and the cell’s proliferation history [47]. Telomeric DNA is
using antibodies against these ligands [38]. These observa-
critical for maintenance of chromosome stability and is
tions may have clinical implications in the evaluation of IST
progressively lost with each replication cycle. Telomeric
responses and developing targeted therapies against
DNA loss leads progressively to cellular senescence.
NKG2D ligands.
Telomere loss can also destabilize the genome by promot-
ing unbalanced chromosome translocations and aneuploidy.
2.2. Effector mediators and their role in apoptosis Telomeric length is preserved by telomerases, RNA–protein
complexes which add lost sequences to DNA ends at each
The apoptotic fraction of CD34+ stem cells is increased in
replication cycle. Telomerases are heavily expressed in HSCs
the aplastic marrow and the proportion of apoptotic cells
and their activity is upregulated during critical periods of
relate to the severity of the disease [39]. Various pathologic
rapid stem-cell proliferation. Aplastic anemia may be viewed
and genetic correlates to apoptosis have been identified
as an acquired form of telomere disease. Telomere attrition
and have been demonstrated to play a crucial role in AA
is observed in peripheral granulocyte and lymphocyte cells
pathogenesis. There is an increased expression of Fas, one
in AA patients and the degree of shortening correlates with
of the receptors in the death receptor signaling pathway, on
the severity of the disease [48,49]. Telomere length has also
CD34+ cells in AA [40]. Bone marrow studies on AA samples
been correlated to increased risk for relapse and inferior
have confirmed CD34+ progenitor cells to be more apopto-
survival [50]. It is not entirely clear as to the mechanism
tic in AA patients when compared with normal donors.
behind telomeric shortening in AA but may be the culmina-
Although this is primarily related to the significantly higher
tion of occult DNA damaging effects in the setting of
proportion of Fas+ CD34+ cells in AA, observation of
reduced telomerase expression and activity due to various
increased apoptosis even in CD34+ Fas− cells in a sizeable
poorly understood genetic determinants. In this context,
fraction of AA patients suggests that the Fas system is not
mutations in TERC and TERT, genes encoding the compo-
an indispensible pathway for apoptosis and there might
nents of the telomerase complex, have been identified in
exist alternate, hitherto unknown mechanisms operating
AA cells [51,52]. These have also been identified rarely in
[41]. Fas antigen surface levels on progenitor cells are clo-
healthy adults suggesting that a complete phenotypic
sely regulated by IFN-γ and TNF-α, and its expression is
expression of their deleterious effects may require an appro-
enhanced as a result of exposure of these cells to the two
priate environmental setting. Telomeric length serves as a
pathogenic cytokines which are constitutively secreted by
marker for DNA damage and reflects underlying ongoing
activated T-effector cells [40,42]. IFN-γ expression is
replicative stress on stem cells [53]. Progressive telomere
increased in the marrow of patients with AA. Intracellular
attrition characteristically occurs before emergence of chro-
levels of IFN-γ in marrow-infiltrating T cells decrease with
mosome 7 loss in acute myeloid leukemia (AML) after SAA
response to IST but increase at onset of relapse [43]. Dufour
[54]. Unlike in other congenital telomeropathies where there
et al. reported on a series of 53 AA patients, who had flow
exist constitutional defects in telomere–telomerase com-
cytometry profiling performed on their bone marrow speci-
plexes, it remains to be clarified if telomere attrition plays
mens, before and after IST administration. While in-vitro
an active role in AA pathogenesis or is merely a passive
blockade of either TNF-α or IFN-γ significantly increased
epiphenomenon of underlying disease progression. This will
blast forming unit populations, only TNF-α blockade signifi-
have future therapeutic implications in the development
cantly increased stem-cell colonies over normal controls, in
and role of highly novel therapies targeting this mechanism
the IST responder patients. Nevertheless, levels of both
in AA.
these cytokines were significantly lower in responders
when compared with nonresponders, providing further evi-
dence to the important role of these cytokines in AA patho-
2.4. Clonal evolution
genesis [44].
These two cytokines transduce their potent apoptotic As mentioned before, direct DNA damaging and telomere
signals to activate the downstream caspase cascade and attritive effects can promote genomic instability by creating
upregulate transcription factors (such as interferon mutations, unbalanced translocations, and aneuploidy. In fact,
EXPERT REVIEW OF HEMATOLOGY 5

accelerated telomerase attrition has been shown to precede in the 1970s with the advent of bone marrow transplantation
clonal evolution and may thus serve as a predictive biomarker and non-transplant immunosuppressive therapies. Prognosis
for future clonal evolution [54]. Chromosomal aberrations can varies markedly depending on the severity of aplastic anemia.
eventually lead to the formation of dysplastic clones which The natural history of non-severe AA, although variable, fol-
may evolve over time. These clones develop unique mechan- lows a fairly indolent course and does not generally warrant
isms of T-cell autoimmune escape, such as by losing their AA- specific treatment apart from supportive care. However, Kwon
overrepresented HLA allele loci [55]. Additionally, these clones et al. in their evaluation of 96 NSAA patients, two-thirds of
may acquire apoptosis-resistance mechanisms and hypersen- whom received some form of immunosuppressive treatment,
sitive growth factor signaling receptors thereby gaining a suggested that NSAA treated with immunosuppressives may
survival advantage that selects for their clinical emergence in decrease its risk of progression to SAA or VSAA [78]. They also
a conducive environmental setting. Clonal evolution rates in identified lower baseline white blood cell (WBC) and ANC
AA occur at the rate of 10–15% over a 10-year duration counts, and treatment non-responsiveness to be predictive
[56,57]. Numerous karyotype defects with abnormalities of of an increased risk of evolution to SAA or VSAA. Unlike in
chromosome 7, trisomy 8, structural and numerical abnormal- SAA patients in whom baseline absolute reticulocyte and
ities of chromosome 13, deletion of Y chromosome, and com- absolute lymphocyte counts predict treatment responsiveness
plex cytogenetics have been identified; the most stereotypical to IST, no predictive identifiers were identified for NSAA in this
defects involve chromosome 6, 7, and 8 [56,57]. The type of study. This study also reported progression to SAA/VSAA in
karyotype defect has a prognostic role in response and clinical 20% of the study population. Taking these findings into con-
outcomes, with chromosome 7 abnormalities portending sideration, it is reasonable to treat NSAA patients with more
worse outcomes while trisomy 8 signals a favorable prognosis. severe cytopenias, especially those with transfusion depen-
Importantly, not all cytogenetic abnormalities lead to clonal dence. It is important to identify and clearly define these
evolution; for example, uniparental disomy of the 6p has not ‘higher risk’ NSAA patients for early institution of immunosup-
been described in association with AA-related MDS/AML [58]. pressive treatment since a delay in treatment from the time of
In addition, somatic mutations involving various bone mar- diagnosis can negatively impact survival.
row failure and myeloid cancer-associated genes such as
ASXL1, DNMT3A, PIGA, BCOR have been identified in up to
3.1. Bone marrow transplant-general considerations
one-third of AA patients and their presence has been corre-
lated with longer disease duration and increased risk of trans- The year 1972 marked the beginning of bone marrow trans-
formation to myelodysplasia/AML [59]. Yoshizato et al. were plant era with Donnall Thomas performing the first successful
able to detect clonal hematopoiesis in up to 47% of their 439 procedure in a patient with aplastic anemia [79]. Evidence of
AA study patients and demonstrated the process to be highly prolonged disease-free survival post engraftment with HLA-
variable with little value in predicting responses and long-term matched sibling marrow grafts soon established the impor-
outcomes. However, certain mutations in PIGA, BCOR, and tance of marrow transplantation in severe aplastic anemia.
BCOR1 did correlate with a better response to IST, while Trends in 5-year survival post bone marrow transplant have
mutations in DNMT3A and ASXL1 associated themselves with considerably improved from 1976 to 1992 (66% vs. 48%,
worse outcomes [59]. The early detection of the deleterious respectively) and primarily reflect an improvement in post-
clones argues for consideration for stem-cell transplantation transplantation anti-graft versus host disease (GVHD) prophy-
(SCT) in favor of IST due to the very high risk of disease laxis with agents like cyclosporine [67]. A European bone
transformation. marrow transplant study analysis on 2002 patients, engrafted
between 1976 and 1998, identified patient’s age, donor type
(HLA-identical sibling vs. alternate donor), interval between
3. Treatment
diagnosis and transplant, and female donor for a male recipi-
Response criteria have been developed to assess and compare ent to be important factors predicting survival outcome after
effectiveness of therapies: complete response (CR) defined by BMT [80]. Infection pre-transplant, a heavy transfusion history
normalization of the blood counts i.e. neutrophil are additional risk factors associated with worsened survival
count > 1.5 × 109/L, platelet count > 150 × 109/L, and [81]. Alternatively, younger age, engraftment from a HLA-
Hb > 12 g/dL; partial response (PR) defined by transfusion matched sibling, transfusion and/or treatment naivety before
independency in patients who needed transfusions before transplantation, higher baseline ANCs associate with over 80%
treatment, with either improvement of the severity degree of long-term survival [80,82].
the AA, or neutrophil counts increasing >0.5 × 109/L and The type of graft also has an effect on the risk of chronic
platelet count >20 × 109/L, in case the values were lower GVHD (cGVHD) and mortality. The EBMT group retrospectively
before treatment. Nonresponse is generally defined by the reviewed outcomes in 692 patients, 134 of whom had periph-
persistence of transfusion dependency or of values lower eral blood progenitor cell grafts, and 558 had bone marrow
than those mentioned above [14]. There are minor variations grafts. Disparity in outcomes by the type of graft was observed
in defining response criteria in various studies (Table 1). in patients younger than 20 years of age in whom cGVHD and
In the pretreatment era, the diagnosis of severe and very overall mortality (5-year OS 85% vs. 73%, respectively) were
severe aplastic anemia called for a dismal prognosis with 2- higher after PBC grafts than after bone marrow transplanta-
year mortality rates approaching 80% on supportive care [77]. tion [83]. A later study on outcomes in 1886 AA patients, by
The grim picture of aplastic anemia dramatically transformed Bacigalupo et al., showed the advantages of bone marrow
6
P. C. BODDU AND T. M. KADIA

Table 1. Frontline immunosuppressive therapies in aplastic anemia.


Median
Overall Time to response follow-up
Group Study type Regimen Study population Age Response criteria response (%) (months) (years) Relapse Clonal evolution Survival
Frickhofen Prospective, single center, hATG + Cys + P NAA/SAA/VAA 32 CR: Hg ≥ 13 g/dL, ANC ≥ 1.5 × 109/L, 11 years ORR: Median at 2 months 11.3 38% 25% (both arms 58% at 11.3 years
et al. [60] randomized pt ≥ 150 × 109/L. 70 combined)
N = 84 PR: TI (or) ↑ in 1/3 (Hg by 3 g/dL, ANC by
0.5 × 109/L, pt by 20 × 109/L)
Rosenfeld Prospective, single center hATG + Cys + P SAA N = 122 35 R = 2/3 (ANC ≥ 0.5 × 109/L; 1 year ORR: 60% by 3 months 7 35% 11% 55% at 7 years
et al. [61] pt ≥ 200 × 109/L; and reticulocyte count 58
≥40 × 109/L)
Bacigalupo Prospective, multicenter hATG + Cys + P + G-CSF SAA N = 100 16 CR: Hg ≥ 11 g/dL, ANC ≥ 1.5 × 109/L, 5 years ORR: Median at 3 months 5 12% 11% 87% at 5 years
et al. [1] pt ≥ 100 × 109/L. PR: TI + Hg ≥ 8 g/dL, 77
ANC ≥ 0.5 × 109/L, p ≥ 30 × 109/L
Tichelli et al. Prospective, multicenter, hATG + Cys ± G-CSF SAA N = 192 46 CR: Hg ≥ 11 g/dL, ANC ≥ 1.5 × 109/L, 6 years ORR: – 6 33% 19% 76% at 6 years
[62] randomized pt ≥ 150 × 109/L. PR: TI + not meeting CR 70
criteria
Kojima et al. Prospective, multicenter, hATG + Cys + Danazol ± GCSF VSAA N = 50 9 CR: Hg ≥ 11 g/dL, ANC ≥ 1.5 × 109/L, 3 years ORR: 47% ORRs in 3 months 3 22% 6% 88% at 3 years
[63] randomized MAA/SAA N = 69 pt ≥ 100 × 109/L. PR: TI + Hg ≥ 8 g/dL, 68 for VSAA
ANC ≥ 0.5 × 109/L, p ≥ 20 × 109/L
Kadia et Prospective, single center rATG + Cys + P + GCSF SAA N = 24 55 CR: ANC ≥ 1 × 109/L, Hgb ≥ 100 g/L, 3 years ORR: Median at 3 months 3 – – 70% at 3 years
al. [64] pt ≥ 100 × 109/L. 64
MDS N = 24 PR: TI + ANC ≥ 0.5 × 109/L, pt ≥ 20 × 109/L,
Hgb ≥ 8 g/dL
Scheinberg Single-center, prospective, Cys + P + (hATG vs. rATG) SAA N = 120 31–37 Hematological response by improvement in 6 months 62% and 33% at 2.3 28% hATG, 11% 21% hATG, 14% hATG: 96% at 3 years
et al. [65] randomized blood counts ORR: 68 3 months for Hatg and rATG rATG rATG: 76% at 3 years
hATG; 37 rATG, respectively
rATG
Marsh et Multicenter, prospective rATG + CyS + P NAA N = 9 36 CR: Hg ≥ 12 g/dL, ANC ≥ 1.5 × 109/L, pt 2 years bRR: 34% at 3 months 1.1 34% at 3 months – 68% at 2 years
al. [66] ≥150 × 109/L 60
SAA N = 20 PR: TI (or) ↑ in 1/3(Hb by 3 g/dL, ANC by
VSAA N = 6 0.5 × 109/L, pt by 20 × 109/L)

Only studies involving ATG (either hATG or rATG) + cyclosporine backbone therapy are included in the table. Cys: cyclosporine; ATG: antithymocyte globulin; P: steroids; NAA/SAA/VSAA: non-severe/severe/very severe aplastic
anemia, respectively; CR: complete remission; PR: partial remission; ORR: overall response rates.
EXPERT REVIEW OF HEMATOLOGY 7

transplant to maintain even in patients above 50 years ago. be significantly higher in patients above 40, as compared with
Similar to previous study, acute GVHD (aGVHD) and cGVHD patients aged 20–40 years [96]. Also, investigators determined
rates were more frequent in peripheral blood transplan- the projected 5-year survival rate in this age group to be
tees [84]. similar to that after IST (in responders) [61,96]. Important
The risk of graft failure and GVHD remains significant pro- reasons identified to negatively affect outcomes included a
blem, which can be partially mitigated by appropriate pre- higher proportion of older patients with prior use of IST, poor
and posttransplant immunosuppressive regimens. Historically, performance score, increased time between diagnosis and
graft failure occurred in 11–32% of AA patients who receive HSCT, and use of PBSC grafts [96]. The optimal treatment
bone marrow transplant. Factors associated with a reduced strategy in older patients awaits a randomized study compar-
risk of graft failure include pretransplant irradiation and use of ing upfront immunosuppressive and transplantation therapy.
cyclosporine prophylaxis against GVHD [85]. Factors favoring There exists further prognostic heterogeneity within this age
rejection include a prior transfusion history, low number of group with age >50 years predictive of inferior survival when
marrow cells, and lack of transfused donor buffy coat cells compared with 40–50 years of age [95]. Fludarabine + CYP, as
[86–88]. Unfortunately, addition of buffy coat cells improves opposed to conventional ATG + CYP used in younger patients,
graft failure rates but may worsen GVHD. Shorter disease- offers superior outcomes and hence advocated for older
transplant time, increasing the number of marrow cells for patients. With improvements in conditioning regimens, appro-
transfusion, and using cyclosporine for post transfusion anti- priate patient selection, and supportive care, it is likely the age
GVHD prophylaxis may offer the best chance of decreasing eligibility for HSCT will continue to expand.
graft failure rates [10,86,87]. Acute severe GVHD and sympto- One of the significant limitations of bone marrow trans-
matic cGVHD occur in 11–40% and 21–32% of bone marrow plant consideration is the availability of HLA-matched sibling
transplant recipients, respectively, depending of various fac- donor. Historically, the risk for transplant-related mortality and
tors [85,88]. aGVHD has decreased considerably with the use GVHD is almost doubled in patients engrafted from a mis-
of potent posttransplant immunosuppressives, while cGVHD matched/unrelated donor as compared with a matched-
continues to be a potentially lethal long-term complication. related donor transplant [82], which is why BMT from unre-
Studies on anti-GVHD prophylaxis regimens favor the use of lated/mismatched donors was traditionally reserved as a sal-
cyclosporine mainly due to a comparatively lower risk of vage option in patients who fail frontline ISTs (Table 2). This
interstitial pneumonia and cGVHD compared to other agents historical notion should be reconsidered in light of emerging
[81,89]. evidence showing an encouraging trend in outcomes over the
Cyclophosphamide (CY) with or without antithymocyte glo- years, after unrelated bone marrow transplant, with much
bulin (ATG) has been used to condition AA patients in pre- lower graft failure, acute and cGvHD rates, and this relates
paration for a bone marrow transplant. The addition of ATG to primarily to improved donor matching. Dufour et al. reported
CY has not been demonstrated to have an additional benefit on equally favorable outcomes in a pediatric AA patient popu-
in improving transplant outcomes [85]. However, fludarabine lation receiving matched unrelated donor (MUD) transplant as
in combination with CY conditioning allows engraftment even opposed to matched sibling donor transplant, while also
in heavily transfused patients and has minimal treatment demonstrating that MUD as initial therapy was superior to
related mortality [1]. Failure of engraftment may not always MUD post-IST failure [97]. Appropriate pretransplant condi-
be detrimental due to facilitation of autologous marrow recov- tioning such as fludarabine-based therapy may partly mitigate
ery by CY conditioning. Furthermore, there is mounting evi- the detrimental effect of post-IST failure on MUD out-
dence to suggest that the further addition of alemtuzumab to comes [91].
the fludarabine and CY provides a highly effective condition- Haploidentical (haplo-SCT) transplant offers a viable option
ing regimen associated with significantly lower graft failure in patients who have failed previous ISTs and lack a suitable
and GVHD rates [90,91]. This becomes especially significant matched donor. Miao et al. reported on outcomes in a series of
and merits further evaluation in the era of MUD and haplo- 39 SAA patients who received haplo-SCT after being condi-
SCTs. tioned with Busulfan + CY + ATG. GVHD prophylaxis constituted
Matched sibling donor transplant offers the best choice for CsA, MMF, and methotrexate. aGVHD rates were about 9%;
long-term reconstitution of hematopoiesis with relatively transplant-related mortality occurred in six patients, with overall
favorable transplant-related mortality and GVHD risks when survival rate of 83% [98]. A head-to-head comparison between
compared with other donor-type transplants. Treatment with haplo-HSCT and MRD type showed no significant difference in
upfront HSCT is currently recommended in younger (18– failure-free or overall survival rates, although aGVHD and
40 years) patients with an available HLA-matched sibling cGVHD rates were significantly higher in the former [99].
[92]. Long-term follow-up in this population, across various Along these lines, GVHD and graft failure remain legitimate
studies, have documented 5-year survival rates in excess of concerns with mismatched transplants. Non-myeloablative con-
80% [69,70,73,93]. Transplant survivorship is complicated by ditioning with posttransplant high-dose CY offers an efficacious
long-term side effects including infertility, cataracts, hypothyr- strategy with acceptable graft failure and severe aGVHD or
oidism, secondary solid malignancies among others [94]. The cGVHD rates [100]. Anti-GVHD prophylaxis may be improved
impressive outcomes in the younger age population, however, upon by the addition of high-dose CY in patients at higher risk
do not apply to older patients, who fare poorly after trans- of GVHD, such as those with mismatched transplants [100].
plantation [80,95]. A retrospective CIBMTR study assessing the Novel strategies involving in-vitro depletion of CD3 T cells
effect of age on transplant outcomes found mortality risks to from grafts prior to transplant have demonstrated to have
8
P. C. BODDU AND T. M. KADIA

Table 2. Bone marrow transplantation in aplastic anemia.


Group/ Median Conditioning
Reference Study population age regimen Post-graft prophylaxis Graft failure/rejection Acute GVHD CGVHD Survival, %
[67] MRD = 471 20 Cy ± TBI/ MTX ± other, Cys ± other, At 5 years, 16% At 100 days, 19% At 5 years, 32% At 5 years, 66%
LFI ± ATG, other MTX + CSA, T-cell depletion,
other
[68] MRD = 71 19 Cy ± others ± TBI Cys ± MTX – At 100 days, 30% At median 4.5 years, 35% At 10 years, 73%
[69] MRD = 64 28 – – 12.5% At 100 days, 31% 18.8% At 6 years, 79%
[70] MRD = 81 24 BU + CY Cys + MTX Primary and At 100 days, 37% At median 8 years, 39% At 8 years, 56%
secondary = 22%
[71] MRD = 79 22 CY + TBI ± ATG Cys + MTX Primary and At 100 days, 7% At median 3.2 years, 35% At 5 years, 74%
secondary = 5.1%
[72] MRD = 113 28 CY + PB + ATG Cys + MTX Primary and secondary at At 100 days, 11% At 1.3 years, 12% At 6 years, 89%
1.3 years, 15%
[73] MRD = 81 26 CY Cys + MTX 11% At 100 days, 24% At median 9.2 years, 26% At 6 years, 88%
[74] MUD = 181, 16 CY ± others, MTX ± Cys other, CsA ± other, Primary and secondary at At 100 days, 48% = MUD, At 5 years, At 5 years,
MMRD = 86, TBI ± irradiation T-depletion ± other 1 year, 15–35% = MMRD, 29% = MUD, 39% = MUD,
MMUD = 51 15% = MUD, 37% = MMUD 19–23% = MMRD23% = MMUD 30–
21–25% = MMRD, 49% = MMRD,
18% = MMUD 36% = MMUD
[75] MUD = 79; 17 CY ± TBI/LFI; CCy/ Cys + MTX, Tac + MTX Primary and At 100 days, 29% At 2 years, 30% At 5 years, 56%
MMUD = 75 ATG ± TBI/LFI secondary = 11%
[76] MUD = 62; 19 TBI + CY ± ATG Cys + MTX Primary and At 100 days, 70% = MUD, At median of 7 years, At 5 years, 61%
MMUD = 25 secondary = 5% 75% = MMUD 52% = MUD, 57% = MMUD
Studies with more than 50 patients included in the table. First seven studies were allogenic sibling transplant, and the bottom 3 studies were mismatched/unrelated donor transplants. MRD: matched-related transplant;
MUD: matched unrelated transplant; MMRD: mismatched-related donor transplant; MMUD: mismatched unrelated donor transplant.
EXPERT REVIEW OF HEMATOLOGY 9

excellent outcomes with lower GVHD rates [101]. Additionally, platelet counts before treatment with ATG were found to be
tacrolimus/methotrexate combination regimen has been shown highly predictive of survival in another study in SAA [107].
to be more effective in preventing GVHD in those who receive However, most of the patients in this other study had long
transplant from unrelated donors [102]. With posttransplant duration of aplasia, and only 40% had response to ATG. Data
immunosuppressive strategies improving, this transplant were not available on the predictive value of cytopenia sever-
should be explored more intensively in future clinical trials. ity in ATG responders [107]. These and multiple other studies
Another alternative for HSCT in the absence of suitable recognized the potentially serious side effects concerning ATG
donor is umbilical cord blood transplantation (UCBT), although treatment. This gradually led to steroids being combined with
experience is limited. Latour et al. reported on outcomes with ATG monotherapy, which not only helped abrogate serious
this procedure in 13 marrow failure patients (SAA = 5). In side effects but also were also found to have an additive
multivariate analysis, only factor associated with engraftment beneficial effect in SAA treatment response.
and survival was total nucleated cell dose (>3.9 × 107/kg, Frickhofen et al. first explored the combination of ATG sera
P = .007). A Japanese retrospective study comparing alternate and cyclosporine, as a part of ongoing intensification efforts,
donor transplants demonstrated similar survival of UCBT to be in a prospective trial of 84 BMT ineligible patients randomized
similar to after MUD, in patients younger than 40 years of age to receive either hATG + methylprednisolone or
[103]. While high engraftment failures limit the utility of UCBT, hATG + methylprednisolone + cyclosporine [108]. The latter
it certainly offers an alternative in patients without available regimen proved superior in terms of 3- (39% vs. 65%, P < .03)
UBMT donors. and 6-month (46% vs. 70%, P < .05) overall remission (CR + PR)
rates, and there was a trend toward superior survival in the
cyclosporine group. Superiority in the 6-month response (65%
3.2. Immunosuppressive therapy
vs. 31%, P < .02) and actuarial survival (at 41 months, 80% vs.
Bone marrow transplant and immunosuppressive treatment 44%, P = .077) rates was most pronounced in the SAA/VSAA
differ vastly with respect to treatment-associated side effects category of patients. The superior efficacy of this combination
with complications vary depending on the choice of therapy; was later confirmed in other studies [109]. Similarly, androgens
graft rejection and GVHD being significant risks after BMT, improved responses when added to ATG and 6-methylpredni-
while relapse, post-therapy malignancies, and secondary clo- solone [110].
nal disorders occur more commonly after immunosuppressive
therapy [10,86,88,104]. While the two treatment choices were 3.2.1. Growth factor support in AA management
historically reported to be comparable in efficacy, improve- Growth factors have been explored extensively in AA to has-
ments in HLA matching, conditioning and posttransplant ten recovery of blood counts, thereby potentially averting AA-
GVHD prophylaxis protocols, and supportive care have, over related complications. A 4-drug regimen involving hATG, ster-
time, translated to comparatively superior efficacy of frontline oids, cyclosporine, and G-CSF was explored by Bacigalupo
transplant over frontline IST, both in terms of event-free survi- et al., in a cohort of 100 SAA patients [1]. Response rates
val and overall survival [68]. An EBMT study comparing out- were 77% (48 CR, 29 PR) and 5-year actuarial survival prob-
comes in AA, adolescent (12–18 years) patients treated with ability was 87%. Severity of baseline neutropenia (less than vs.
frontline MRD transplant versus immunosuppressive therapy greater than 0.2 × 109/L) determined 5-year survival (76% vs.
demonstrated superior 3-year event-free survival in patients 98%; P = .001). This regimen was tested in a phase III rando-
receiving transplant over IST (83% vs. 37%, P < .001) [10]. The mized study comparing standard immunosuppressive therapy
‘immune’ hypothesis of aplastic anemia received credibility with or without growth factor support. The addition of leno-
with Speck et al. demonstrating the value of immunosuppres- grastim to the combination resulted in a higher response rate
sive therapy when administered alone in AA patients consid- (83% vs. 44%; P < .0001) and shorter median time to complete
ered ineligible for transplantation [105]. Study investigators neutrophil response (6 vs. 16 weeks; P < .0001). There was no
concluded that horse ATG (hATG) was comparable to hATG difference in survival, response rates, or secondary leukemia
followed by allo-HSCT in terms of hematological response rates at a follow-up of 5 years [111]. This was later confirmed
(40% vs. 42%, respectively) and 1-year survival (58% vs. 54%, in another randomized prospective study in which the addi-
respectively) rates. The therapeutic value of hATG in AA was tion of G-CSF to hATG, Cys, and prednisolone for the
further established, by Champlin et al, in a prospective study treatment of SAA shortened hospital stay and decreased infec-
randomizing AA patients to either hATG or best supportive tion rates but had no effect on overall and event-free survi-
care [106]. Eleven of 21 patients randomized to receive hATG val [62].
had sustained improvements in blood counts within 3 months
of treatment while none of the 21 control patients receiving 3.2.2. Eltrombopag
supportive care alone improved. Since survival was not the Eltrombopag (E-PAG) is a synthetic thrombopoetin (TPO)-
primary end point of this study, most AA patients in the mimetic that binds to and activates the TPO receptor present
supportive care arm were allowed to crossover and receive on stem progenitor cells and megakaryocytes. Eltrombopag
ATG after the 3-month timepoint. Duration of pancytopenia was initially studied in refractory setting where it was shown
prior to treatment correlated inversely with the robustness of to be effective in producing trilineage hematopoiesis. Olnes
response to IST. It was further observed that actuarial survival et al. first conducted a study on 25 refractory SAA patients
outcomes after responding to hATG were not determined by with an objective to assess platelet count recovery [112]. Dose
the pretreatment severity of AA. Conversely, neutrophil and was started at 50 mg daily and titrated based on platelet
10 P. C. BODDU AND T. M. KADIA

response to a maximum of 150 mg daily. Overall response is not currently justified with other less toxic alternatives
rates were seen in 44% of the patients, of whom 8 maintained available [119].
a response at 10 months. Important clinical responses were Frickhofen et al. reported mature data of their initial study
observed across all lineages with the majority of responders cohort sample in an 11-year follow-up update [60,108]. As was
having increased white blood counts, and achieving platelet expected, response rates were higher (70% vs. 41%, with or
and RBC transfusion independence. The study was continued without cyclosporine; P = .015), median time to response was
by Desmond et al., in an expanded cohort of 43 patients [113]. shorter (median, 60 vs. 82 days; P = .019) and the failure-free
Forty percent of patients responded at a median of survival (39% vs. 24% at 11 years; P = .04) was longer in the
3–4 months. Of the five patients who discontinued therapy cyclosporine group. However, overall survival and relapse
at a median of 28.5 months after achieving robust responses rates were not statistically different between the groups. In
to eltrombopag, all maintained stable counts at a median of spite of this, the ATG + Cys + steroid regimen continues to
13 months off the medication. Eight (including six nonrespon- enjoy favor because of lower time at risk from pancytopenic
ders) patients had evidence of clonal evolution (16%), and five complications effects and decreased need for salvage.
of them had a chromosome 7 abnormality. However, none of Outcomes with ATG + Cys have improved considerably, as
them had clinical evolution to MDS/AML. reflected in the survival trends from the EBMT database, over
Based on its promising activity in the relapse setting, the the past 3 decades (1973–80 vs. 2000–07, 49% vs. 72%, respec-
drug has been explored in the frontline setting in conjunction tively) [120]. This is largely attributed to improved care sup-
with IST (i.e. ATG + Cys). Kadia et al. conducted a phase II portive and better informed second-line salvage treatment.
single-arm study serially enrolling 31 untreated AA patients,
the first 17 receiving IST alone while the next 14 patients were 3.2.4. Rabbit ATG
treated with IST + E-PAG [114]. The latter group had patients Unfortunately, 10–20% fail initial treatment, 30–50% even-
who were comparatively older in age and had had signifi- tually relapse in the long term, and a smaller percentage
cantly lower blood counts. Despite these unfavorable charac- may undergo a disease evolution to other disorders [61,121].
teristics, there was a trend toward improved overall response Relapse does not impact overall survival and patients do
rates in patients receiving IST + E-PAG (79% vs. 59%, P = .28). A respond to a second round albeit at lower rates in the refrac-
higher WBC count and ANC, and the presence of a PNH clone, tory setting [121]. Responses to second IST, incorporating
were associated with response and response was associated either hATG or rabbit ATG (rATG), may vary from 22–64%
with an improved overall survival (12-month OS: 100% vs. 58% after failure to first IST with hATG treatment [107,121].
P = .01). The regimen is well tolerated with manageable grade Among the various ATG preparations, horse ATG has usually
3/4 toxicities. This was followed closely by another study by been the first choice due to its larger supply and well-docu-
Townsley et al. similarly evaluating the IST + E-PAG in a phase mented efficacy as first-line therapy. Scheinberg et al. prospec-
2 single-center trial [115]. A total of 88 patients, divided into tively evaluated outcomes comparing horse ATG and rabbit
three cohorts based on differing dosing schema, were enrolled ATG in conventional regimens [65]. Outcomes were superior in
in this study. Of all evaluable patients, ORRs at 3 and 6 months the hATG regimen group both in terms in hematologic rates
were 80% and 85%, respectively. Interestingly, cytogenetic and overall survival [65]. This was followed by another study,
abnormalities had occurred in seven patients. The investiga- by Marsh et al., which similarly demonstrated an inferior effi-
tors concluded that clonal evolution to MDS occurred at a cacy of rATG, with the best response rates being 60% and
similar frequency compared to their historic experience with 67%, 2-year overall survival of 68% and 86% (P = .009), and
standard IST. The role of eltrombopag alone or in conjunction transplant-free survival of 52% and 76% (P = .002), for rATG
with other ISTs will be further established in future and and hATG, respectively [66]. Hence, rATG is generally reserved
ongoing clinical trials [NCT01328587, NCT01703169, for second-line therapy unless hATG is not tolerated or
NCT01891994, NCT01623167, NCT02099747, NCT02773225, unavailable.
and NCT02404025]. Rabbit ATG has been extensively studied in the second-
line setting after failure or relapse after first-line ISTs with
hATG. Its utility, in the relapsed/refractory setting after failure
3.2.3 Other ATG combinations to hATG, has been well documented with responses ranging
Efforts at improving immunosuppression by combining agents from 22% to 77% in various investigational studies [107,121].
like mycophenolate mofetil or CY to ATG and cyclosporine Means et al. first distinguished that immunoglobulin from a
have been abandoned after the combination regimens proved different source (rATG in their study) was able to effect
to be toxic while not producing higher response rates responses in patients who failed initial ATG treatment [122].
[116,117]. Although early studies showed equivalent response The role of rATG as an effective second-line treatment of AA
rates with either modified high-dose CY plus cyclosporine or was further established by Di Bona et al. [123]. Patients with
ATG and cyclosporine [118], more recent data suggest the SAA who had failed frontline hATG, CSA, methylpredniso-
combination of cyclosporine and CY may prove toxic to justify lone, and G-CSF treatment were treated with rATG, CSA,
this regimen as a viable treatment alternative. In fact, a rando- and G-CSF. Of the 30 evaluable patients, 23 patients (77%)
mized control trial comparing high-dose CY + Cys with became transfusion independent, with 9 (30%) and 14 (47%)
ATG + Cys was terminated prematurely due to higher infec- patients achieving CR and PR, respectively. Importantly, no
tions and early deaths in the former group [117]. Toxicities are immediate or late reactions were observed. Scheinberg et al.
considerable even with moderate dose CY, and hence, its use retrospectively reviewed outcomes in 43 relapsed/refractory
EXPERT REVIEW OF HEMATOLOGY 11

patients who were treated with rATG/CSA; 22 were refractory unrelated transplants, and optimization of pretransplant con-
and 21 had relapsed after treatment. Overall response rates ditioning/posttransplant GVHD prophylaxis regimens. The elig-
were far higher in relapsed patients (66%) than in refractory ibility for transplant shouldn’t be solely based on age, and the
patients (27%) in refractory patients. As might be expected, role of allogeneic transplant in the frontline setting needs to
there was a trend toward improved overall survival in be further clarified in the older patients (above 40 years). With
responders compared with nonresponders (1000 day: 90% evidence showing improved survival when transplanted at the
vs. 65%, P = .18) [124]. Given the roughly equivalent earliest in the disease course, the rationale of choosing IST
responses to either ATG preparation as second-line therapy, over unrelated donor transplant as the initial frontline therapy
immunoglobulin from a different animal source may be pre- and the time frame of determining failure to first-line IST
ferred to decrease the risk of acute, potentially life-threaten- before proceeding to transplant as salvage needs to be clearly
ing hypersensitivity reactions. Outcome data with other defined. Although the potential risk of growth factor and
preparations of ATG such as porcine ATG, although limited immunosuppressive therapies in promoting clonal evolution
to a single institutional experience, demonstrate comparable has not been substantiated, the risk of its development
efficacy to rATG and hATG and may offer suitable cost-effec- remains a pertinent concern especially with a prolonged sur-
tive alternative in the future [125]. vival expected in AA.
Rabbit-ATG, in conjunction with other immunosuppres- The development of novel therapies may prove valuable
sants, was also shown to be effective as frontline therapy. Its especially in patients with severe aplastic anemia who fail ISTs
role in the frontline setting was first evaluated by Stein et al. in and are ineligible for transplant. Decades of observational and
57 MAA/SAA patients, 49 of whom had no prior treatment investigational study have led to a vastly improved under-
therapy with hATG. Treatment regimen in the study consisted standing of AA pathogenesis and helped inform and develop
of r-ATG+prednisone ± cyclosporine ± androgens. Of the 57 novel treatment strategies. As a generation of new therapies
patients, responses occurred in 16 (28%), the majority of them emerges, fascinating insights into underlying disease pro-
(46%) occurring in patients who received rATG and cyclospor- cesses may provide exciting opportunities in which to explore
ine [126]. Similarly, another study recently updated by Kadia new therapeutic avenues.
et al. [64] reported data suggesting rATG to be an effective
alternative to hATG. In the updated study, 14 of 22 evaluable
patients had a response (6 CR, 8 PR) within a median of
5. Expert commentary
3 months. However, comparison studies have consistently
reported hATG/CSA to be superior to rATG/CSA across the Bone marrow failure in aplastic anemia leads to profound
AA severity spectrum, both in terms of response rates and pancytopenia, transfusion dependence, and the clinical seque-
long-term survival [65,127,128]. Based on the available evi- lae resulting from cytopenias. About 15% of patients may
dence, rATG is best reserved for second-line therapy after transform to a clonal myeloid malignancy such as MDS or
failure to hATG but may be considered as a first-line option AML. A more durable treatment of aplastic anemia is achieved
if hATG is not tolerated or available. through allogeneic SCT in selected individuals or through
combination immunosuppressive therapy in those who are
3.2.5. Alemtuzumab not candidates for SCT. Importantly, pronounced deficits in
Alemtuzumab is a humanized monoclonal antibody that binds LTC-IC numbers continue to exist irrespective of treatment
CD52, an antigen expressed on B- and T-lymphocytes. Antibody with BMT or SCT, suggestive of persistent suppressive
binding triggers a potent lympholytic effect via antibody and mechanism with potential implications on disease
complement mediated cytotoxicity. Single-agent alemtuzumab relapse [132].
has a modest activity and appears to be an inferior option in It is through the fortuitous discovery of the response to
the treatment-naïve AA setting [129,130]. However, alemtuzu- immunosuppresion that researchers began to understand
mab + cyclosporine combination has demonstrated activity as a the underlying pathophysiology of idiopathic AA. The
first-line approach with results from different groups reporting clinic–pathologic manifestations of AA appear to be the
response rates ranging from 37% to 58% [129,131]. The efficacy result of an auto-directed, T-lymphocyte-mediated immune
of alemtuzumab monotherapy in the R/R setting is more destruction of HSCs. Responses to immunosuppressive ther-
encouraging with comparable response rates to apy with horse ATG in patients with AA were observed in
rATG + cyclosporine (33% vs. 37%). Hematologic toxicities approximately 30% of cases. Initial attempts to improve
such as severe neutropenia, infusion reactions, and infectious immunosuppression with the combination of horse ATG
risks including reactivation of latent cytomegalovirus infection and cyclosporine led to durable responses in over two-thirds
stress the need for vigilance and preventative antimicrobial of patients and continue to be a mainstay of therapy.
strategies while on therapy [130]. Alemtuzumab is an effective Further attempts at intensifying immunosuppression with
salvage option in R/R patients not eligible for transplant. the use of the more potent rabbit ATG or with the addition
of more immunosuppressive drugs have not improved
response rates. In patients who are refractory to frontline
therapy or have relapsed after initial response, different
4. Conclusions and future directions
immunosuppressive therapies such as alemtuzumab and
The role of transplant in AA continues to expand with the small-molecule thrombopoietin-mimetic, eltrombopag,
improvements in supportive care, improved matching of have demonstrated promising activity. Other studies have
12 P. C. BODDU AND T. M. KADIA

provided more information into a different side of the dis- ● There is evidence to suggest multiple simultaneously and
ease. Recurrent cytogenetic abnormalities and, more sequentially operating pathophysiological mechanisms
recently, recurrent somatic mutations point to an underlying including immune mediated damage, a role for telomere
clonal hematopoietic disorder that shares similarities to attrition, and the less clear role of stromal/adipocyte inhibi-
other myeloid malignancies such as MDS or AML. Studies tion – thus providing opportunities for targeting through
on telomere length reveal ongoing telomere attrition in the novel strategies, many of which are still in the early infancy
hematopoietic cells from patients with AA, which may result of development.
from replication in the face of immune attack and result in ● Current hematological parameter based risk stratification
genomic instability. Several of these abnormalities may carry criteria, such as the Camitta criteria, lack firm clinical
prognostic significance, predict for response to immunosup- grounds and are slowly falling out of favour in the modern
pressive therapy, and help select patients who may be era of AA treatment
better candidates for early allogeneic SCT. ● Integration of cytogenetic/molecular markers, and more
recent concepts on telomere attrition to blood count data
may help develop more rational prognostic risk stratifica-
6. Five-year view
tion criteria informing prognosis and choice of treatment in
As we look ahead to state-of-the-art management of aplastic future protocol studies.
anemia, it is critical to continue collaborative research in this
rare disease and build upon the current research and clinical
Funding
experience. Allogeneic SCT has been a curative approach for
selected patients with AA. However, its use in the frontline is This manuscript was supported by the University of Texas MD Anderson
limited to younger patients with matched-related siblings. Cancer Center Support Grant (P30 CA16672) and award P01 CA049639 (Dr.
Richard Champlin) from the National Cancer Institute (NCI).
With advances in supportive care and reduced intensity con-
ditioning, efforts to broaden the eligible age for SCT should be
explored. Similarly, recent experience using MUD transplants
for newly diagnosed AA may prompt broader use of alterna- Declaration of interest
tive donor SCTs in this setting. With the widespread applica- The authors have no relevant affiliations or financial involvement with any
tion of next-generation sequencing, there is an intense organization or entity with a financial interest in or financial conflict with
interest in defining the clonal heterogeneity and clonal evolu- the subject matter or materials discussed in the manuscript. This includes
employment, consultancies, honoraria, stock ownership or options, expert
tion among patients with aplastic anemia. There are already
testimony, grants or patents received or pending, or royalties.
indications that these recurrent abnormalities have prognostic
significance and predictive value when considering immuno-
suppressive therapy. Going forward, wider validation of these
References
genetic abnormalities will allow them to be incorporated into
a more informative risk stratification system that does not rely Papers of special note have been highlighted as either of interest (•) or of
on clinical factors alone. Deciphering the significance of these considerable interest (••) to readers.
genetic abnormalities may stimulate the development of more 1. Bacigalupo A, Bruno B, Saracco P, et al. Antilymphocyte globulin,
cyclosporine, prednisolone, and granulocyte colony-stimulating
specific therapies among defined subsets. Telomere biology in factor for severe aplastic anemia: an update of the GITMO/EBMT
congenital and acquired bone marrow failure states has study on 100 patients. European Group for Blood and Marrow
opened a whole area of basic and clinical research that will Transplantation (EBMT) Working Party on Severe Aplastic Anemia
have important implications for diagnostics, prognostication, and the Gruppo Italiano Trapianti di Midolio Osseo (GITMO). Blood.
and treatment approaches over the next several years. 2000;95(6):1931–1934.
2. Young NS, Maciejewski J. The pathophysiology of acquired aplastic
Integrating these advances and having the appropriate anemia. N Engl J Med. 1997;336(19):1365–1372.
resources to manage these patients will require expertise, 3. Sakuma T, Hayashi Y, Kanomata N, et al. Histological and cytoge-
education, and development of key centers of excellence to netic characterization of bone marrow in relation to prognosis and
treat this disease. diagnosis of myelodysplastic syndromes. Pathol Int. 2006;56
(4):191–199.
4. Auerbach AD. Fanconi anemia and its diagnosis. Mutat Res.
Key issues 2009;668(1–2):4–10.
5. Walne AJ, Dokal I. Dyskeratosis Congenita: a historical perspective.
● Attempts at intensification beyond ATG, steroids and Mech Ageing Dev. 2008;129(1–2):48–59.
cyclosporine have not been fruitful with the standard ATG, 6. Rovó A, Tichelli A, Dufour C. Diagnosis of acquired aplastic anemia.
Bone Marrow Transplant. 2013;48(2):162–167.
Cyclosporine, steroid combination strategy still favored as
7. Young NS, Maciejewski JP, Sloand E, et al. The relationship of
the current frontline immunosuppressive therapy of choice. aplastic anemia and PNH. Int J Hematol. 2002;76(Suppl 2):168–172.
● Expansion of the role of hematopoietic stem cell transplant 8. Brown KE, Tisdale J, Barrett AJ, et al. Hepatitis-associated aplastic
in domains of elderly AA and alternative donor transplant anemia. N Engl J Med. 1997;336(15):1059–1064.
should be addressed in future investigational studies. 9. Stalder MP, Rovó A, Halter J, et al. Aplastic anemia and concomitant
autoimmune diseases. Ann Hematol. 2009;88(7):659–665.
● Among the more recently investigated agents, alemtuzu-
10. Dufour C, Pillon M, Passweg J, et al. Outcome of aplastic anemia in
mab and eltrombopag have been shown to demonstrate adolescence: a survey of the severe aplastic anemia working party
effectiveness and offer valuable salvage options in HSCT of the European Group for Blood and Marrow Transplantation.
ineligible relapsed/refractory SAA patients. Haematologica. 2014;99(10):1574–1581.
EXPERT REVIEW OF HEMATOLOGY 13

11. Ades L, Mary J-Y, Robin M, et al. Long-term outcome after bone 34. Kordasti S, Marsh J, Al-Khan S, et al. Functional characterization of
marrow transplantation for severe aplastic anemia. Blood. 2004;103 CD4+ T cells in aplastic anemia. Blood. 2012;119(9):2033–2043.
(7):2490–2497. 35. Kordasti S, Costantini B, Seidl T, et al. Deep-phenotyping of Tregs
12. Bacigalupo A, Hows J, Gluckman E, et al. Bone marrow transplanta- identifies an immune signature for idiopathic aplastic anemia and
tion (BMT) versus immunosuppression for the treatment of severe predicts response to treatment. Blood. 2016;128:1193–1205.
aplastic anaemia (SAA): a report of the EBMT SAA working party. Br • T-cell repertoire should be explored further to identify clini-
J Haematol. 1988;70(2):177–182. cally valuable predictive and prognostic markers.
13. Camitta BM, Rappeport JM, Parkman R, et al. Selection of patients 36. Giannakoulas NC, Karakantza M, Theodorou GL, et al. Clinical rele-
for bone marrow transplantation in severe aplastic anemia. Blood. vance of balance between type 1 and type 2 immune responses of
1975;45(3):355–363. lymphocyte subpopulations in aplastic anaemia patients. Br J
14. Rozman C, Marín P, Nomdedeu B, et al. Criteria for severe aplastic Haematol. 2004;124(1):97–105.
anaemia. Lancet. 1987;2(8565):955–957. 37. Liu C, Li Z, Sheng W, et al. Abnormalities of quantities and func-
15. Catlin SN, Busque L, Gale RE, et al. The replication rate of human tions of natural killer cells in severe aplastic anemia. Immunol
hematopoietic stem cells in vivo. Blood. 2011;117(17):4460–4466. Invest. 2014;43(5):491–503.
16. Olweus J, Lund-Johansen F, Terstappen LW. Expression of cell sur- 38. Hanaoka N, Nakakuma H, Horikawa K, et al. NKG2D-mediated
face markers during differentiation of CD34+, CD38-/lo fetal and immunity underlying paroxysmal nocturnal haemoglobinuria and
adult bone marrow cells. Immunomethods. 1994;5(3):179–188. related bone marrow failure syndromes. Br J Haematol. 2009;146
17. Maciejewski JP, Anderson S, Katevas P, et al. Phenotypic and func- (5):538–545.
tional analysis of bone marrow progenitor cell compartment in 39. Philpott NJ, Scopes J, Marsh JC, et al. Increased apoptosis in aplas-
bone marrow failure. Br J Haematol. 1994;87(2):227–234. tic anemia bone marrow progenitor cells: possible pathophysiolo-
18. Sutherland HJ, Eaves CJ, Eaves AC, et al. Characterization and gic significance. Exp Hematol. 1995;23(14):1642–1648.
partial purification of human marrow cells capable of initiating 40. Maciejewski JP, Selleri C, Sato T, et al. Increased expression of Fas
long-term hematopoiesis in vitro. Blood. 1989;74(5):1563–1570. antigen on bone marrow CD34+ cells of patients with aplastic
19. Maciejewski JP, Kim S, Sloand E, et al. Sustained long-term hema- anaemia. Br J Haematol. 1995;91(1):245–252.
tologic recovery despite a marked quantitative defect in the stem 41. Killick SB, Cox CV, Marsh JC, et al. Mechanisms of bone marrow
cell compartment of patients with aplastic anemia after immuno- progenitor cell apoptosis in aplastic anaemia and the effect of anti-
suppressive therapy. Am J Hematol. 2000;65(2):123–131. thymocyte globulin: examination of the role of the Fas-Fas-L inter-
20. Wilson A, Laurenti E, Oser G, et al. Hematopoietic stem cells rever- action. Br J Haematol. 2000;111(4):1164–1169.
sibly switch from dormancy to self-renewal during homeostasis 42. Maciejewski J, Selleri C, Anderson S, et al. Fas antigen expression
and repair. Cell. 2008;135(6):1118–1129. on CD34+ human marrow cells is induced by interferon gamma
21. Sato T, Onai N, Yoshihara H, et al. Interferon regulatory factor-2 and tumor necrosis factor alpha and potentiates cytokine-
protects quiescent hematopoietic stem cells from type I interferon- mediated hematopoietic suppression in vitro. Blood. 1995;85
dependent exhaustion. Nat Med. 2009;15(6):696–700. (11):3183–3190.
22. Passegué E, Wagers AJ, Giuriato S, et al. Global analysis of prolif- 43. Sloand E, Kim S, Maciejewski JP, et al. Intracellular interferon-
eration and cell cycle gene expression in the regulation of hema- gamma in circulating and marrow T cells detected by flow cyto-
topoietic stem and progenitor cell fates. J Exp Med. 2005;202 metry and the response to immunosuppressive therapy in patients
(11):1599–1611. with aplastic anemia. Blood. 2002;100(4):1185–1191.
23. Maciejewski JP, Selleri C, Sato T, et al. A severe and consistent 44. Dufour C, Svahn J, Bacigalupo A. Front-line immunosuppressive
deficit in marrow and circulating primitive hematopoietic cells treatment of acquired aplastic anemia. Bone Marrow Transplant.
(long-term culture-initiating cells) in acquired aplastic anemia. 2013;48(2):174–177.
Blood. 1996;88(6):1983–1991. 45. Kakagianni T, Giannakoulas NC, Thanopoulou E, et al. A probable
24. Zeng W, Chen G, Kajigaya S, et al. Gene expression profiling in role for trail-induced apoptosis in the pathogenesis of marrow
CD34 cells to identify differences between aplastic anemia patients failure. Implications from an in vitro model and from marrow of
and healthy volunteers. Blood. 2004;103(1):325–332. aplastic anemia patients. Leuk Res. 2006;30(6):713–721.
25. Wang J, Sun Q, Morita Y, et al. A differentiation checkpoint limits 46. Zhang J-Y, Chang H, Meng W-T. [The polymorphism of interferon
hematopoietic stem cell self-renewal in response to DNA damage. gamma gene CA short tandem repeat is associated with aplastic
Cell. 2012;148(5):1001–1014. anemia]. Sichuan Da Xue Xue Bao Yi Xue Ban. 2008;39(1):23–25, 58.
26. Mauch P, Rosenblatt M, Hellman S. Permanent loss in stem cell self 47. Blackburn EH. Switching and signaling at the telomere. Cell.
renewal capacity following stress to the marrow. Blood. 1988;72 2001;106(6):661–673.
(4):1193–1196. 48. Brümmendorf TH, Maciejewski JP, Mak J, et al. Telomere length in
27. Shao L, Luo Y, Zhou D. Hematopoietic stem cell injury induced by leukocyte subpopulations of patients with aplastic anemia. Blood.
ionizing radiation. Antioxid Redox Signal. 2014;20(9):1447–1462. 2001;97(4):895–900.
28. Malkin D, Koren G, Saunders EF. Drug-induced aplastic anemia: 49. Lee JJ, Kook H, Chung IJ, et al. Telomere length changes in patients
pathogenesis and clinical aspects. Am J Pediatr Hematol Oncol. with aplastic anaemia. Br J Haematol. 2001;112(4):1025–1030.
1990;12(4):402–410. 50. Scheinberg P, Cooper JN, Sloand EM, et al. Association of telomere
29. Morinet F, Leruez-Ville M, Pillet S, et al. Concise review: anemia length of peripheral blood leukocytes with hematopoietic relapse,
caused by viruses. Stem Cells. 2011;29(11):1656–1660. malignant transformation, and survival in severe aplastic anemia.
30. Xing L, Liu C, Fu R, et al. CD8+HLA-DR+ T cells are increased in patients Jama. 2010;304(12):1358–1364.
with severe aplastic anemia. Mol Med Rep. 2014;10(3):1252–1258. 51. Yamaguchi H, Calado RT, Ly H, et al. Mutations in TERT, the gene
31. Risitano AM, Maciejewski JP, Green S, et al. In-vivo dominant for telomerase reverse transcriptase, in aplastic anemia. N Engl J
immune responses in aplastic anaemia: molecular tracking of puta- Med. 2005;352(14):1413–1424.
tively pathogenetic T-cell clones by TCR beta-CDR3 sequencing. 52. Yamaguchi H, Baerlocher GM, Lansdorp PM, et al. Mutations of the
Lancet. 2004;364(9431):355–364. human telomerase RNA gene (TERC) in aplastic anemia and mye-
32. Schuster FR, Hubner B, Führer M, et al. Highly skewed T-cell recep- lodysplastic syndrome. Blood. 2003;102(3):916–918.
tor V-beta chain repertoire in the bone marrow is associated with 53. Houben JM, Moonen HJJ, Van Schooten FJ, et al. Telomere length
response to immunosuppressive drug therapy in children with very assessment: biomarker of chronic oxidative stress? Free Radic Biol
severe aplastic anemia. Blood Cancer J. 2011;1(3):e8. Med. 2008;44(3):235–246.
33. Goto M, Kuribayashi K, Takahashi Y, et al. Identification of auto- 54. Dumitriu B, Feng X, Townsley DM, et al. Telomere attrition and
antibodies expressed in acquired aplastic anaemia. Br J Haematol. candidate gene mutations preceding monosomy 7 in aplastic ane-
2013;160(3):359–362. mia. Blood. 2015;125(4):706–709.
14 P. C. BODDU AND T. M. KADIA

• Telomere attrition precedes chromosome 7 loss, implicating its 72. Kim HJ, Park CY, Park YH, et al. Successful allogeneic hematopoietic
role in the development of aneuploidy and paving way for AML. stem cell transplantation using triple agent immunosuppression in
55. Katagiri T, Sato-Otsubo A, Kashiwase K, et al. Frequent loss of HLA severe aplastic anemia patients. Bone Marrow Transplant. 2003;31
alleles associated with copy number-neutral 6pLOH in acquired (2):79–86.
aplastic anemia. Blood. 2011;118(25):6601–6609. 73. Kahl C, Leisenring W, Deeg HJ, et al. Cyclophosphamide and
56. Maciejewski JP, Selleri C. Evolution of clonal cytogenetic antithymocyte globulin as a conditioning regimen for allogeneic
abnormalities in aplastic anemia. Leuk Lymphoma. 2004;45 marrow transplantation in patients with aplastic anaemia: a long-
(3):433–440. term follow-up. Br J Haematol. 2005;130(5):747–751.
57. Maciejewski JP, Risitano A, Sloand EM, et al. Distinct clinical out- 74. Passweg JR, Pérez WS, Eapen M, et al. Bone marrow transplants
comes for cytogenetic abnormalities evolving from aplastic ane- from mismatched related and unrelated donors for severe aplastic
mia. Blood. 2002;99(9):3129–3135. anemia. Bone Marrow Transplant. 2006;37(7):641–649.
58. Mikhailova N, Sessarego M, Fugazza G, et al. Cytogenetic abnorm- 75. Kojima S, Matsuyama T, Kato S, et al. Outcome of 154 patients with
alities in patients with severe aplastic anemia. Haematologica. severe aplastic anemia who received transplants from unrelated
1996;81(5):418–422. donors: the Japan Marrow Donor Program. Blood. 2002;100(3):799–
59. Yoshizato T, Dumitriu B, Hosokawa K, et al. Somatic Mutations 803.
and Clonal Hematopoiesis in Aplastic Anemia. N Engl J Med. 76. Deeg HJ, O’Donnell M, Tolar J, et al. Optimization of conditioning
2015;373(1):35–47.. for marrow transplantation from unrelated donors for patients with
•• Characterization of somatic mutations and clonal hematopoi- aplastic anemia after failure of immunosuppressive therapy. Blood.
esis prevalent in AA 2006;108(5):1485–1491.
60. Frickhofen N, Heimpel H, Kaltwasser JP, et al. Antithymocyte glo- 77. Camitta BM, Thomas ED, Nathan DG, et al. A prospective study of
bulin with or without cyclosporin A: 11-year follow-up of a rando- androgens and bone marrow transplantation for treatment of
mized trial comparing treatments of aplastic anemia. Blood. severe aplastic anemia. Blood. 1979;53(3):504–514.
2003;101(4):1236–1242. 78. Kwon JH, Kim I, Lee YG, et al. Clinical course of non-severe aplastic
61. Rosenfeld S, Follmann D, Nunez O, et al. Antithymocyte globulin anemia in adults. Int J Hematol. 2010;91(5):770–775.
and cyclosporine for severe aplastic anemia: association between 79. Thomas ED, Storb R, Fefer A, et al. Aplastic anaemia treated by
hematologic response and long-term outcome. Jama. 2003;289 marrow transplantation. Lancet. 1972;1(7745):284–289.
(9):1130–1135. 80. Bacigalupo A, Brand R, Oneto R, et al. Treatment of acquired severe
62. Tichelli A, Schrezenmeier H, Socié G, et al. A randomized controlled aplastic anemia: bone marrow transplantation compared with
study in patients with newly diagnosed severe aplastic anemia immunosuppressive therapy–The European Group for Blood and
receiving antithymocyte globulin (ATG), cyclosporine, with or with- Marrow Transplantation experience. Semin Hematol. 2000;37
out G-CSF: a study of the SAA Working Party of the European (1):69–80.
Group for Blood and Marrow Transplantation. Blood. 2011;117 81. Gluckman E, Horowitz MM, Champlin RE, et al. Bone marrow trans-
(17):4434–4441. plantation for severe aplastic anemia: influence of conditioning
63. Kojima S, Hibi S, Kosaka Y, et al. Immunosuppressive therapy using and graft-versus-host disease prophylaxis regimens on outcome.
antithymocyte globulin, cyclosporine, and danazol with or without Blood. 1992;79(1):269–275.
human granulocyte colony-stimulating factor in children with 82. Bacigalupo A, Oneto R, Bruno B, et al. Current results of bone
acquired aplastic anemia. Blood. 2000;96(6):2049–2054. marrow transplantation in patients with acquired severe aplastic
64. Kadia TM, Borthakur G, Garcia-Manero G, et al. Final results of the phase anemia. Report of the European Group for Blood and Marrow
II study of rabbit anti-thymocyte globulin, ciclosporin, methylpredni- transplantation. On behalf of the Working Party on Severe
sone, and granulocyte colony-stimulating factor in patients with aplas- Aplastic Anemia of the European Group for Blood and Marrow
tic anaemia and myelodysplastic syndrome. Br J Haematol. 2012;157 Transplantation. Acta Haematol. 2000;103(1):19–25.
(3):312–320. 83. Schrezenmeier H, Passweg JR, Marsh JCW, et al. Worse outcome
65. Scheinberg P, Nunez O, Weinstein B, et al. Horse versus rabbit and more chronic GVHD with peripheral blood progenitor cells
antithymocyte globulin in acquired aplastic anemia. N Engl J than bone marrow in HLA-matched sibling donor transplants for
Med. 2011;365(5):430–438.. young patients with severe acquired aplastic anemia. Blood.
•• Randomized study showing upfront horse ATG to be superior 2007;110(4):1397–1400.
over rATG 84. Bacigalupo A, Socié G, Schrezenmeier H, et al. Bone marrow versus
66. Marsh JC, Bacigalupo A, Schrezenmeier H, et al. Prospective study peripheral blood as the stem cell source for sibling transplants in
of rabbit antithymocyte globulin and cyclosporine for aplastic acquired aplastic anemia: survival advantage for bone marrow in
anemia from the EBMT Severe Aplastic Anaemia Working Party. all age groups. Haematologica. 2012;97(8):1142–1148.
Blood. 2012;119(23):5391–5396. 85. Champlin RE, Perez WS, Passweg JR, et al. Bone marrow transplan-
67. Passweg JR, Socié G, Hinterberger W, et al. Bone marrow transplan- tation for severe aplastic anemia: a randomized controlled study of
tation for severe aplastic anemia: has outcome improved? Blood. conditioning regimens. Blood. 2007;109(10):4582–4585.
1997;90(2):858–864. 86. Champlin RE, Horowitz MM, Van Bekkum DW, et al. Graft failure
68. Locasciulli A, Oneto R, Bacigalupo A, et al. Outcome of patients following bone marrow transplantation for severe aplastic anemia:
with acquired aplastic anemia given first line bone marrow trans- risk factors and treatment results. Blood. 1989;73(2):606–613.
plantation or immunosuppressive treatment in the last decade: a 87. Deeg HJ, Self S, Storb R, et al. Decreased incidence of marrow graft
report from the European Group for Blood and Marrow rejection in patients with severe aplastic anemia: changing impact
Transplantation (EBMT). Haematologica. 2007;92(1):11–18. of risk factors. Blood. 1986;68(6):1363–1368.
69. Ahn M-J, Choi J-H, Lee -Y-Y, et al. Outcome of adult severe or very 88. Storb R, Champlin RE. Bone marrow transplantation for severe
severe aplastic anemia treated with immunosuppressive therapy aplastic anemia. Bone Marrow Transplant. 1991;8(2):69–72.
compared with bone marrow transplantation: multicenter trial. Int J 89. May WS, Sensenbrenner LL, Burns WH, et al. BMT for severe aplastic
Hematol. 2003;78(2):133–138. anemia using cyclosporine. Bone Marrow Transplant. 1993;11
70. Dulley FL, Vigorito AC, Aranha FJP, et al. Addition of low-dose (6):459–464.
busulfan to cyclophosphamide in aplastic anemia patients prior 90. Marsh JC, Gupta V, Lim Z, et al. Alemtuzumab with fludarabine and
to allogeneic bone marrow transplantation to reduce rejection. cyclophosphamide reduces chronic graft-versus-host disease after
Bone Marrow Transplant. 2004;33(1):9–13. allogeneic stem cell transplantation for acquired aplastic anemia.
71. Bai LY, Chiou TJ, Liu JH, et al. Hematopoietic stem cell transplanta- Blood. 2011;118(8):2351–2357.
tion for severe aplastic anemia–experience of an institute in • This conditioning regimen can be a very efficacious strategy
Taiwan. Ann Hematol. 2004;83(1):38–43. with low graft-versus-host disease and should be explored
EXPERT REVIEW OF HEMATOLOGY 15

further in haplo-SCT and other unrelated donor transplant 108. Frickhofen N, Kaltwasser JP, Schrezenmeier H, et al. Treatment of
types. aplastic anemia with antilymphocyte globulin and methylpredni-
91. Samarasinghe S, Steward C, Hiwarkar P, et al. Excellent outcome of solone with or without cyclosporine. The German Aplastic Anemia
matched unrelated donor transplantation in paediatric aplastic Study Group. N Engl J Med. 1991;324(19):1297–1304.
anaemia following failure with immunosuppressive therapy: a 109. Rosenfeld S, Kimball J, Vining D, et al. Intensive immunosuppres-
United Kingdom multicentre retrospective experience. Br J sion with antithymocyte globulin and cyclosporine as treatment for
Haematol. 2012;157(3):339–346. severe acquired aplastic anemia. Blood. 1995;85(11):3058–3065.
92. Killick SB, Bown N, Cavenagh J, et al. Guidelines for the diagnosis 110. Bacigalupo A, Chaple M, Hows J, et al. Treatment of aplastic anae-
and management of adult aplastic anaemia. Br J Haematol. mia (AA) with antilymphocyte globulin (ALG) and methylpredniso-
2016;172(2):187–207. lone (MPred) with or without androgens: a randomized trial from
93. Aljurf M, Al-Zahrani H, Van Lint MT, et al. Standard treatment of the EBMT SAA working party. Br J Haematol. 1993;83(1):145–151.
acquired SAA in adult patients 18-40 years old with an HLA-iden- 111. Gluckman E, Rokicka-Milewska R, Hann I, et al. Results and follow-
tical sibling donor. Bone Marrow Transplant. 2013;48(2):178–179. up of a phase III randomized study of recombinant human-granu-
94. Eapen M, Ramsay NK, Mertens AC, et al. Late outcomes after bone locyte stimulating factor as support for immunosuppressive ther-
marrow transplant for aplastic anaemia. Br J Haematol. 2000;111 apy in patients with severe aplastic anaemia. Br J Haematol.
(3):754–760. 2002;119(4):1075–1082.
95. Kim H, Lee K-H, Yoon -S-S, et al. Allogeneic hematopoietic stem cell 112. Olnes MJ, Scheinberg P, Calvo KR, et al. Eltrombopag and improved
transplant for adults over 40 years old with acquired aplastic hematopoiesis in refractory aplastic anemia. N Engl J Med.
anemia. Biol Blood Marrow Transplant. 2012;18(10):1500–1508. 2012;367(1):11–19.
96. Gupta V, Eapen M, Brazauskas R, et al. Impact of age on outcomes • Eltrombopag effective in refractory severe aplastic anemia,
after bone marrow transplantation for acquired aplastic anemia making it valuable salvage option in patient without a trans-
using HLA-matched sibling donors. Haematologica. 2010;95 plant option.
(12):2119–2125. 113. Desmond R, Townsley DM, Dumitriu B, et al. Eltrombopag restores
97. Dufour C, Veys P, Carraro E, et al. Similar outcome of upfront- trilineage hematopoiesis in refractory severe aplastic anemia that
unrelated and matched sibling stem cell transplantation in idio- can be sustained on discontinuation of drug. Blood. 2014;123
pathic paediatric aplastic anaemia. A study on behalf of the UK (12):1818–1825.
Paediatric BMT Working Party, Paediatric Diseases Working Party 114. Kadia T, Ravandi F, Daver N, et al. Single-center experience of
and Severe Aplastic Anaemia Working Party of EBMT. Br J immunosuppressive therapy with or without eltrombopag in
Haematol. 2015;171(4):585–594. patients with aplastic anemia. Blood. 2015;126(23):4779–4779.
• Equivalent outcomes, seen with both unrelated and related 115. Townsley DM, Dumitriu B, Young NS. Bone marrow failure and the
matched donor transplants in pediatric AA, should be explored telomeropathies. Blood. 2014;124(18):2775–2783.
in adult AA. 116. Scheinberg P, Nunez O, Wu C, et al. Treatment of severe aplastic
98. Miao M, Zhang X, Xu T, et al. Haploidentical hematopoietic stem anaemia with combined immunosuppression: anti-thymocyte glo-
cell transplantation for acquired severe aplastic anemia. Blood. bulin, ciclosporin and mycophenolate mofetil. Br J Haematol.
2015;126(23):3227–3227. 2006;133(6):606–611.
• Valuable transplant option in patients without matched trans- 117. Tisdale JF, Dunn DE, Geller N, et al. High-dose cyclophosphamide in
plant donor and should be explored further in future studies. severe aplastic anaemia: a randomised trial. Lancet (London,
99. Xu L-P, Jin S, Wang S-Q, et al. Upfront haploidentical transplant for England). 2000;356(9241):1554–1559.
acquired severe aplastic anemia: registry-based comparison with 118. Zhang F, Zhang L, Jing L, et al. High-dose cyclophosphamide
matched related transplant. J Hematol Oncol. 2017;10(1):25. compared with antithymocyte globulin for treatment of acquired
100. Luznik L, O’Donnell PV, Symons HJ, et al. HLA-haploidentical bone severe aplastic anemia. Exp Hematol. 2013;41(4):328–334.
marrow transplantation for hematologic malignancies using non- 119. Scheinberg P, Townsley D, Dumitriu B, et al. Moderate-dose cyclo-
myeloablative conditioning and high-dose, posttransplantation phosphamide for severe aplastic anemia has significant toxicity
cyclophosphamide. Biol Blood Marrow Transplant. 2008;14(6):641– and does not prevent relapse and clonal evolution. Blood.
650. 2014;124(18):2820–2823.
101. Im HJ, Koh KN, Choi ES, et al. Excellent outcome of haploidentical 120. Passweg JR, Tichelli A. Immunosuppressive treatment for aplastic
hematopoietic stem cell transplantation in children and adoles- anemia: are we hitting the ceiling?. Haematologica. 2009;94
cents with acquired severe aplastic anemia. Biol Blood Marrow (3):310–312.
Transplant. 2013;19(5):754–759. 121. Schrezenmeier H, Marin P, Raghavachar A, et al. Relapse of aplastic
102. Yagasaki H, Kojima S, Yabe H, et al. Tacrolimus/Methotrexate versus anaemia after immunosuppressive treatment: a report from the
cyclosporine/methotrexate as graft-versus-host disease prophylaxis European Bone Marrow Transplantation Group SAA Working
in patients with severe aplastic anemia who received bone marrow Party. Br J Haematol. 1993;85(2):371–377.
transplantation from unrelated donors: results of matched pair 122. Means RT Jr., Krantz SB, Dessypris EN, et al. Re-treatment of aplastic
analysis. Biol Blood Marrow Transplant. 2009;15(12):1603–1608. anemia with antithymocyte globulin or antilymphocyte serum. Am
103. Kuwatsuka Y, Kanda J, Yamazaki H, et al. A Comparison of J Med. 1988;84(4):678–682.
Outcomes for Cord Blood Transplantation and Unrelated Bone 123. Di Bona E, Rodeghiero F, Bruno B, et al. Rabbit antithymocyte
Marrow Transplantation in Adult Aplastic Anemia. Biol Blood globulin (r-ATG) plus cyclosporine and granulocyte colony stimu-
Marrow Transplant. 2016;22(10):1836–1843. lating factor is an effective treatment for aplastic anaemia patients
104. Socié G, Rosenfeld S, Frickhofen N, et al. Late clonal diseases of unresponsive to a first course of intensive immunosuppressive
treated aplastic anemia. Semin Hematol. 2000;37(1):91–101. therapy. Gruppo Italiano Trapianto di Midollo Osseo (GITMO). Br J
105. Speck B, Gluckman E, Haak HL, et al. Treatment of aplastic anaemia Haematol. 1999;107(2):330–334.
by antilymphocyte globulin with and without allogeneic bone- 124. Scheinberg P, Nunez O, Young NS. Retreatment with rabbit anti-
marrow infusions. Lancet. 1977;2(8049):1145–1148. thymocyte globulin and ciclosporin for patients with relapsed or
106. Champlin R, Ho W, Gale RP. Antithymocyte globulin treatment in refractory severe aplastic anaemia. Br J Haematol. 2006;133
patients with aplastic anemia: a prospective randomized trial. N (6):622–627.
Engl J Med. 1983;308(3):113–118. 125. Wei J, Huang Z, Guo J, et al. Porcine antilymphocyte globulin (p-
107. Marsh J, Hows JM, Bryett KA, et al. Survival after antilymphocyte ALG) plus cyclosporine A (CsA) treatment in acquired severe aplas-
globulin therapy for aplastic anemia depends on disease severity. tic anemia: a retrospective multicenter analysis. Ann Hematol.
Blood. 1987;70(4):1046–1052. 2015;94(6):955–962.
16 P. C. BODDU AND T. M. KADIA

126. Stein RS, Means RT, Krantz SB, et al. Treatment of aplastic anemia 129. Gómez-Almaguer D, Jaime-Pérez JC, Garza-Rodríguez V, et al.
with an investigational antilymphocyte serum prepared in rabbits. Subcutaneous alemtuzumab plus cyclosporine for the treatment
Am J Med Sci. 1994;308(6):338–343. of aplastic anemia. Ann Hematol. 2010;89(3):299–303.
127. Marsh J, Schrezenmeier H, Marin P, et al. Prospective Randomized 130. Scheinberg P, Nunez O, Weinstein B, et al. Activity of alemtuzumab
Multicenter Study Comparing Cyclosporin Alone Versus the monotherapy in treatment-naive, relapsed, and refractory severe
Combination of Antithymocyte Globulin and Cyclosporin for acquired aplastic anemia. Blood. 2012;119(2):345–354.
Treatment of Patients With Nonsevere Aplastic Anemia: A Report 131. Kim H, Min YJ, Baek JH, et al. A pilot dose-escalating study of
From the European Blood and Marrow Transplant (EBMT) Severe alemtuzumab plus cyclosporine for patients with bone marrow
Aplastic Anaemia Working Party. Blood. 1999;93(7):2191–2195. failure syndrome. Leuk Res. 2009;33(2):222–231.
128. Sloand EM, Olnes MJ, Weinstein B, et al. Long-term follow-up of 132. Podesta M, Piaggio G, Frassoni F, et al. The assessment of the hemato-
patients with moderate aplastic anemia and pure red cell aplasia poietic reservoir after immunosuppressive therapy or bone marrow
treated with daclizumab. Haematologica. 2010;95(3):382–387. transplantation in severe aplastic anemia. Blood. 1998;91(6):1959–1965.

You might also like