You are on page 1of 10

| |

Received: 4 April 2018    Revised: 23 July 2018    Accepted: 24 July 2018

DOI: 10.1111/ejh.13153

REVIEW ARTICLE

Aplastic anemia: Etiology, molecular pathogenesis, and


emerging concepts

Rory M. Shallis1  | Rami Ahmad1 | Amer M. Zeidan1,2

1
Division of Hematology/Medical
Oncology, Department of Medicine, Yale Abstract
University School of Medicine, New Haven, Aplastic anemia (AA) is rare disorder of bone marrow failure which if severe and not
Connecticut
2
appropriately treated is highly fatal. AA is characterized by morphologic marrow fea‐
Cancer Outcomes, Public Policy, and
Effectiveness Research (COPPER) tures, namely hypocellularity, and resultant peripheral cytopenias. The molecular
Center, Yale University, New Haven,
pathogenesis of AA is not fully understood, and a uniform process may not be the
Connecticut
culprit across all cases. An antigen‐driven and likely autoimmune dysregulated T‐cell
Correspondence: Amer M. Zeidan, Section
homeostasis is implicated in the hematopoietic stem cell injury which ultimately
of Hematology, Department of Internal
Medicine, Yale University, 333 Cedar Street, founds the pathologic features of the disease. Defective telomerase function and re‐
PO Box 208028, New Haven, CT (amer.
pair may also play a role in some cases as evidenced by recurring mutations in related
zeidan@yale.edu).
telomerase complex genes such as TERT and TERC. In addition, recurring mutations in
BCOR/BCORL, PIGA, DNMT3A, and ASXL1 as well as cytogenetic abnormalities,
namely monosomy 7, trisomy 8, and uniparental disomy of the 6p arm seem to be in‐
timately related to AA pathogenesis. The increased incidence of late clonal disease
has also provided clues to accurately describe plausible predispositions to the devel‐
opment of AA. The emergence of newer genomic sequencing and other techniques is
incrementally improving the understanding of the pathogenic mechanisms of AA, the
detection of the disease, and ultimately offers the potential to improve patient out‐
comes. In this comprehensive review, we discuss the current understanding of the
immunobiology, molecular pathogenesis, and future directions of such for AA.

KEYWORDS
aplastic anemia, cytogenetic, molecular, pathogenesis, review

1 |  I NTRO D U C TI O N dedicated treatment is not considered. Multiple etiologies for


AA have been proposed which in due course lead to an immune‐
Bone marrow failure is a characteristic finding in a number of mediated destruction of hematopoietic stem cells (HSCs) resid‐
inherited syndromes such as dyskeratosis congenita, Fanconi ing in the bone marrow, but the bulk of cases remain idiopathic.
anemia, Diamond‐Blackfan anemia, and Shwachman‐Diamond Allogeneic stem cell transplantation (alloSCT) offers much of the
syndrome; however, acquired etiologies of marrow aplasia may limited prospect of cure for patients (who do not respond to im‐
present similarly.1,2 Originally described by Dr. Paul Ehrlich in munosuppressive therapy), but unfortunately a significant subset
1888 and further defined by 1904 by Dr. Anatole Chauffard, of patients are not suitable candidates. Immunosuppressive ther‐
aplastic anemia (AA) is a nonmalignant hematologic disorder apies otherwise continue to be the mainstay for patients with AA
characterized by an injured, markedly hypocellular, and thus inef‐ not proceeding to alloSCT, but the increasing understanding of
fective bone marrow. 3 The resultant pancytopenia presents diag‐ the pathophysiology of HSC injury is expected to incrementally
nostic challenges and confers an extreme risk of mortality when broaden therapeutic options.

Eur J Haematol. 2018;1–10. wileyonlinelibrary.com/journal/ejh   © 2018 John Wiley & Sons A/S. |  1
Published by John Wiley & Sons Ltd
|
2       SHALLIS et al.

2 |  E TI O LO G Y A N D A NTI G E N I C manifests. In fact, several pathophysiologic pathways have been


E X P OS U R E proposed to explain the pathologic nature of the disease, but the
greatest proportion of cases is hypothesized to be a result of uni‐
Aplastic anemia is rare disease with an incidence of approximately form T cell‐mediated autoimmunity and marrow destruction lead‐
two to three cases per million per year based on dedicated studies, ing to defective and nearly absent hematopoiesis. Strong laboratory
but may be threefold higher in Asian populations. It is a disease of data suggest that suspected yet unidentified antigenic exposure
the young and typically that of the first three decades of life with leads to a polyclonal expansion of CD4+ T cells dysregulation at the
a median age of about 20 years old, though a second peak occurs genetic level and ultimately an overproduction of proinflammatory
around age 60.4-7 Geographic differences in incidence implicate cytokines such as interferon (IFN)‐γ as well as tumor necrosis factor
environmental and host‐related factors and ultimately corroborate (TNF)‐α. Oligoclonal expansion of a dysregulated cytotoxic CD8+ T‐
prior study into possible causative factors. Etiologic association with cell population has also been demonstrated in ex vivo bone marrow
occupational exposure to chemical haptens such as benzene and models of AA patients. 24-27 Increases in T helper 17 (Th17) cells, the
pesticides, specifically the organochlorines and organophosphates effector cells which produce the proinflammatory cytokine inter‐
has been demonstrated, although these are exceedingly rare causes leukin‐17 (IL‐17), are also found in the peripheral blood and bone
of AA when examining cases worldwide.7-9 In Asian populations spe‐ marrow of AA patients. 28 The degree of Th17 cell elevation in one
cifically, exposure to not only similar pesticides, but also animal fer‐ study was also found to correlate with the amount of IFN‐γ‐produc‐
tilizer and animals themselves have been linked to AA.10,11 ing cells and overall disease activity; Th17 cell populations also nega‐
Medication exposures with subsequent idiosyncratic reactions tively correlated with Treg populations. 28 The concept of aberrant,
are archetypal (yet still rare) causes of AA‐related marrow fail‐ disordered T‐cell populations initiating the pathology of AA is also
ure. Chloramphenicol is a classic example which is still without a supported by the finding that that T cell‐directed immunosuppres‐
known, culprit mechanism, although the myelosuppressive effect sive therapy (IST) such as the combination of antithymocyte globulin
is attributed to mitochondrial injury. The etiology of resultant AA and cyclosporine is able to induce response in up to 80% of patients
has been attributed to accumulation of drug metabolites toxic to with severe aplastic anemia. 29
12,13
HSCs. Ocular chloramphenicol and the antiepileptics carbamaz‐ AA patient marrows are also noted to have both marked quan‐
epine and valproic acid have been implicated as a cause of AA, al‐ titative and qualitative deficits of regulatory T‐cells (Tregs), which
though these are likewise exceedingly infrequent.14,15 A handful of normally suppress autoreactivity of other T‐cell populations to nor‐
other supposed offending medications are limited to case reports mal tissue including the bone marrow environment and HSCs.30,31
only. CD8+CD57+ cells, a subset of T‐cells known as effector memory
Aplastic anemia has also been associated with pregnancy, which cells which activate as a result of antigen affinity and stimulation,
interestingly enough was also the purported cause for the devel‐ are shown to be expanded in the peripheral blood of AA patients,
opment of AA of the case first described by Dr. Paul Ehrlich.3,16,17 with a majority demonstrating features of oligoclonality.32 Given the
Infection with human immunodeficiency virus (HIV) is associated role of CD8+CD57+ effector memory cells in immune surveillance,
with the development of AA, although this association is limited to their expansion may precipitate the abnormal oligoclonal expansion
case reports and small case series which may be a result of a de‐ seen in AA. A recent study of Treg population deep phenotyping
creasing incidence of HIV infection.18,19 Seronegative hepatitis has using mass cytometric analyses reproducibly defined two Treg sub‐
been implicated in approximately 5% of AA cases. 20,21 Association populations (deemed Treg A and Treg B) with specific phenotypes,
with eosinophilic fasciitis is limited to less than two dozen cases gene expression signature, and function.33 Dominance of a Treg B
reported in the literature, although many patients have concomi‐ subpopulation, defined in part by higher expression of specifically
tant autoimmune disorder and/or possible environmental exposure human leukocyte antigen (HLA)‐DR, FOXP3, CD95, and CCR4, lower
which could otherwise be implicated. 22,23 Clear infectious etiologies expression of CD45RA, and expression of the interleukin‐2 (IL‐2)/
beyond hepatitis and HIV have yet to be established and be con‐ STAT5 pathway were found in AA patient with response to IST.33
founded by the fact that undiagnosed postinfectious aplasia can be Earlier studies of HLA expression in AA have also supported their
clinically mistaken for AA. role in provoking the aberrant T‐cell homeostasis based on the find‐
ing of a higher frequency of both HLA‐DR2 and HLA‐DR15 (a split
of HLA‐DR2) presence in AA patients.34,35 The overrepresentation
3 |  TH E PATH O G E N I C I M M U N O B I O LO G Y of these specific HLA molecules in AA patient may effectively aug‐
ment the otherwise physiologic and constitutive expression of HLA
The injury to hematopoiesis observed in AA has primarily been class II molecules on antigen‐presenting cells to interact with CD4+
shown to be facilitated by an antigen‐driven immune response, T‐cells and ultimately present a HSC‐derived antigen resulting in
which may be related to the suspected etiologic factors discussed subsequent HSC immune‐mediated destruction.34 In addition, cells
previously. The broad and heterogeneous nature of these associated expressing the HLA‐DR2 antigen (after IFN‐γ priming) have been
etiologic factors may call into question whether a single mechanism previously shown to augment the release of TNF‐α, a known partic‐
is responsible for the resultant profound cytopenias with which AA ipant in the apparatus of this destruction.35 Together, the increasing
SHALLIS et al. |
      3

F I G U R E 1   Immunopathogenic mechanism of hematopoietic stem or progenitor cell apoptosis in the aplastic anemia bone marrow

data on the understanding of Treg and Th17 subsets or subpopula‐ illuminated. Increased incidence of proinflammatory cytokine poly‐
tions in the AA marrow or peripheral blood at diagnosis may soon morphisms, specifically in IFN‐γ, TNF‐α, and interleukin‐6, is repeat‐
solidify a reliable immune signature which predicts response to stan‐ edly found in cases of AA. HSC FasR expression is likely upregulated
dard IST and as a result guide front‐line treatment decisions. by this cytokine overproduction. Hypersecreting genotypes of the
Despite eventual antigen clearance which under physiologic multifunctional and regulatory transforming growth factor beta 1
circumstances would dictate abatement of the appropriate, proin‐ (TGFβ1) have also been demonstrated and taken together this may
flammatory immune response, the AA marrow milieu appears to be suggest a quicker and more robust immune response to antigenic
one of clonal persistence and continued aberrant immune response exposure and myelosuppression.39 MicroRNAs (miRNAs) including
leading to the observed pathology. Increasing understanding of the miR‐532‐5p, MiR‐4651, and miR‐126‐5p, which modulate down‐
culprit apoptotic mechanisms observed in AA has aided the identi‐ stream proinflammatory pathway components such as Toll‐like re‐
fication of the precise population of marrow cells affected by such ceptors and TNF‐α, are uniquely and commonly identified in AA.40,41
T‐cell dysregulation. Fas (first apoptosis signal) receptor (FasR), a miR‐126‐5p is also found to be decreased in patients with AA re‐
chief element in apoptotic signaling, has specifically been shown sponding to IST, suggesting miRNA interference of cytokine and
to be overexpressed in CD34+ progenitor cells and lymphocytes in growth factor‐related proinflammatory and immune signaling.41
marrow specimens of AA patients. Such data provide evidence that
the ensuing marrow failure is a consequence of the inflammatory
cytokine‐induced and Fas‐mediated apoptosis as well as that CD34+ 4 | TE LO M E R E H O M EOS TA S I S
progenitor cells appear to be the predominant population whose an‐ PATH O LO G Y
nihilation leads to HSC deficiency.36,37 Normal FasR expression in
such cells in patients with AA in remission further supports a Fas‐ Inherent HSC biological flaws are also suspected to play a role in the
38
based mechanism of apoptosis in AA. (Figure 1). pathogenesis of AA. Defective homeostasis of telomeres (the repeti‐
A genetic and molecular basis for why such clonal T‐cell ex‐ tive sequences of DNA which cap linear chromosomes and facilitate
pansion occurs and becomes pathogenic is being progressively cell proliferation) has also been implicated in a significant subset
|
4       SHALLIS et al.

of AA patients and is an emerging category of disease in general. foreseeable chromosomal instability) with androgen therapy averts
Telomere shortening (occurring if suppressor or other protective the molecular pathogenesis of clonal evolution.
genes are inactivated or by physiologic attrition) leads to cell pro‐
liferative arrest and eventual apoptosis.42 To combat the attrition,
germ line cells utilize telomerase reverse transcriptase (TERT), telom‐ 5 | TH E M YS TE R I O U S RO LE O F
erase RNA component (TERC), and the stabilizing protein dyskerin TH RO M B O P O I E TI N
(DKC1) to assemble the telomerase complex and maintain telomere
length, which is naturally and expectantly lengthy in HSCs.42 Human Increasing data support the role of thrombopoietin (TPO) and TPO
lymphocytes are the primary somatic cells which can upregulate signaling in hematopoiesis specifically related to HSC homeostasis,
TERT with resultant telomerase activity and provide a transient pro‐ proliferation, and survival. Knockout murine models lacking the c‐
liferative advantage, although this does not avert eventual replica‐ mpl gene (which encodes the TPO receptor which itself is also ex‐
tive senescence.43,44 pressed on HSCs) show significant deficiencies in HSCs and thus
Approximately 35% of AA patients are found to have periph‐ suggest that TPO is integral to HSC homeostasis.56,57 Higher levels
eral granulocyte and mononuclear cell telomere length shortening, of plasma TPO are observed in AA patients at diagnosis suggesting
pointing toward a progenitor cell defect.45,46 Critical loss of HSC a physiologic response to the diminished hematopoiesis and throm‐
telomere length in about 9% of acquired AA cases has been associ‐ bopoiesis. In addition, TPO levels decrease after response to IST.58
ated with mutations in TERC, TERT, and DKC1 which are known to be IFN‐γ, the principal proinflammatory cytokine responsible for the
the cause of inherited or constitutional aplastic anemia such as dys‐ HSC destruction in AA, is shown to decrease the interaction of TPO
keratosis congenita.47,48 Rare mutations in other telomerase genes and c‐MPL via direct TPO‐IFN‐γ heterodimerization and likely rep‐
such as TERF1 and TERF2 have been identified as possible driver mu‐ resent the key mechanism by which inflammatory cytokines directly
tations in AA.49 Biallelic mutations in regulator of telomere elonga‐ disrupt hematopoiesis in AA.59
tion helicase 1 (RTEL1), which encodes a protein critical to telomere Given the role of TPO in normal hematopoiesis and the re‐
homeostasis, is a known cause of congenital bone marrow failure, cently enlightened TPO‐related mechanism of pathology in AA, the
but has been identified in 1%‐2% of AA cases; most RTEL1 variants, use of TPO mimetics is garnering an increasing clinical role in AA.
however, are felt not to be pathogenic in the latter condition.50 Eltrombopag, a nonpeptide TPO mimetic and c‐MPL agonist, has
The degree of telomere length shortening or erosion has been previously shown response of 44% (including trilineage responses)
shown to correlate with the severity of AA, risk of relapse, overall sur‐ in patients with AA refractory to IST.60,61 Subsequent investigations
vival, and risk of clonal evolution (via acquisition of new cytogenetic noted to that eltrombopag interaction with c‐MPL is not affected
aberrancy) to conditions such as myelodysplastic syndrome (MDS), by direct TPO‐IFN‐γ heterodimerization and eventually abrogates
51
which will be discussed in more detail later. Telomere length short‐ the myelopoietic failure induced by such.59 Recently, eltrombopag
ening in cases of AA is likely insufficient to lead to AA and ultimately was combined with standard IST at varying doses and schedules in
calls into question the distinction between acquired and inherited newly‐diagnosed AA patients; induction of trilineage hematopoiesis
bone marrow failure syndromes such as AA given it frequently oc‐ as well as overall response and complete response rates of up to
curs in adult patients without a clear family history of such. 94% and 58%, respectively, was observed.62 Similarly, the TPO mi‐
Sex hormones have been shown to exert significant effects on metic romiplostim has been shown to stimulate primitive HSC, aug‐
telomere homeostasis. Previous in vitro evidence has shown that ment their differentiation into later progenitor cells, and ultimately
HSC exposure to several types of androgens induced telomerase demonstrate efficacy in patients with AA refractory to IST.63-65
activity and this correlated with higher levels of TERT mRNA.52 This Despite prior theoretical concerns, romiplostim therapy in AA has
effect was also seen in peripheral blood lymphocytes heterozygous not been shown to increase the subsequent development of cytoge‐
for loss‐of‐function TERT mutations which at baseline were associ‐ netic abnormalities or clonal evolution.64,65
ated with reduced telomerase, but with the addition of androgen TPO agonism likely promotes proliferation of the remaining,
52
were restored to normal. Subsequent murine models corroborated though distinctly reduced, HSC population. The HSC compartment
this observation of androgen‐induced telomere lengthening with has been shown to be markedly reduced (in the order of approx‐
eventual improved hematopoiesis.53 This was ultimately confirmed imately 10% of normal) even in patients who have responded to
in human studies using danazol, a strong androgen receptor agonist IST with appropriate count recovery and more robust responses to
which increased telomerase activity and correlated with upregu‐ IST have been observed in patients with less severe AA (and likely
lation of TERT.54 However, a recent retrospective study found no a larger and functional HSC compartment).66,67 These findings sug‐
difference in the telomere attrition rates of androgen‐treated or un‐ gest that a certain threshold for count recovery is required and is
treated patients with dyskeratosis congenita irrespective of the type hypothesized to be the result of a possible protective mechanism of
of androgen therapy used or leukocyte subset evaluated; such data HSC to prevent total exhaustion of the HSC pool, but may also be
imply a mechanism of clinical response other than androgen‐related the result of recurrently‐observed telomere length shortening in AA
effects on telomere homeostasis.55 It remains to be seen whether patients.60,66 TPO mimetics do not appear to abolish the pathogenic
or not abrogation of telomere length shortening seen in AA (and the autoimmunity of AA, and TPO biology to date has not been shown
SHALLIS et al. |
      5

to have any direct effect on cytotoxic or regulatory T‐cell expansion Karyotyping a hypocellular bone marrow specimen, however,
or perturbation. However, this impressive clinical activity might ad‐ can be problematic and underestimate an accurate percentage of
vocate for the incorporation of TPO mimetics such as eltrombopag cytogenic aberrancy in AA. The development of higher fidelity SNP
into front‐line therapies for AA. array karyotyping has revealed more subtle abnormalities below the
level of detection with conventional karyotyping and has elucidated
other means of CH in AA. Because of SNP array profiling, unipa‐
6 | C Y TO G E N E TI C A B N O R M A LITI E S A N D rental disomy (UPD) in 6p (6pUPD), which escaped conventional
TH E I M M U N E M EC H A N I S M karyotyping, can now be identified in up to 13% of AA patients.72,77
The most commonly involved region by acquired CN‐LOH in AA is
Cytogenetic aberrations commonly‐observed in other myeloid malig‐ the short arm of chromosome 6 at the site of the MHC locus, oc‐
nancies are recurrent in AA such as monosomy 7 and trisomy 8, oc‐ curring in about half of AA patients with acquired 6pUPD (Table 1).
curring in up to 13% and 7% of AA cases, respectively.68,69 Data from 6pUPD may develop via the immune‐escape of such HLA‐deficient
Sloand et  al70 have previously shown that bone marrow specimens HSCs.78 In a recent study combining targeted deep sequencing of
of patients with MDS (13% of which had progressed from AA) and class I HLA and SNP array in AA patients, 17% (11/66) of patients
evidence of trisomy 8 clonal disease showed a marked expansion of were found to have somatic HLA loss and those who inherited the
CD8+ T‐cell populations. Specific T‐cell receptor subfamilies, namely targeted HLA alleles, regardless of the mutation status, had a more
Vβ, demonstrated significant suppression of trisomy 8 myeloid cells.70 severe disease course with more frequent clonal complications, and
The same group also showed that trisomy 8 marrow HSCs are able to increased frequency of developing secondary MDS.79 Given these
escape destruction by CD8+ T‐cells via overexpression of prosurvival findings, AA patients with the acquisition of HSC HLA deficiencies
proteins cyclin D1 and survivin. Nontrisomy 8 HSCs are unable to such as 6pUPD or other somatic mutations may have class I HLA
mount this protection and are preferentially suppressed. No data to loss‐of‐function which could allow immune‐escape from cytotoxic T‐
date support a similar mechanism for monosomy 7 AA, but a study cell‐mediated destruction, as opposed to the typical HLA‐facilitated
by Dimitriu et  al71 demonstrated that progressive telomere loss is presentation of previously mentioned, plausible AA‐related antigens
noted before the detection of and thus may lead to eventual mono‐ to CD8+ T‐cells. Such findings also strengthen the assertion that
somy 7 in patient who develop MDS progressed from AA. These data understanding the pathogenesis of clonal evolution in AA may help
further support the relationship between cytogenetic predisposition explicate its autoimmune nature.
and the downstream immunobiologic HSC destruction. Less com‐
monly detected cytogenetic alterations in AA include del(13q), tri‐
somy 6, and trisomy 15 which occur in <5% of cases.72,73 The clinical 7 | S O M ATI C M U TATI O N S
relevance of cytogenetic abnormalities in AA is being gradually clari‐
fied. The identification of monosomy 7 in AA patients is associated The advent of next‐generation sequencing (NGS) has allowed the
with an increased risk of progression to MDS or AML, while trisomy identification of somatic mutations as a more dominant cause of
8 and del(13q) have been associated with more favorable response clonality in AA.72,78 PIGA, a gene essential for the synthesis of sur‐
to IST and improved clinical outcome, specifically improved progres‐ face glycosylphosphatidylinositol (GPI)‐linked anchoring proteins, is
sion‐free survival (PFS) and overall survival (OS).72-76 detected in 60% of patients with evidence of a paroxysmal nocturnal

TA B L E 1   Recurring cytogenetic
Abnormality Incidencea, % Prognostic impact Reference(s)
abnormalities in acquired aplastic anemia
75,85
6pUPD 13 Favorable response to IST
68,73,92
Monosomy 7/del(7q) 2.0‐13.3 Higher risk of progression
to MDS or AML
68,71,92,96
Trisomy 8 1.3‐6.7 Favorable response to IST,
lower risk of progression
to MDS or AML
75,85
Del(13q) 0.4‐1.8 Favorable response to IST,
possibly better survival
71
Trisomy 6 2.4 Unknown
71
Trisomy 15 2.4 Unknown

6pUPD, uniparental disomy of the 6p arm; AML, acute myeloid leukemia; IST, immunosuppressive
therapy; MDS, myelodysplastic syndrome.
a
Referenced studies had variable analyses either performed at diagnosis of aplastic anemia or later
in course of disease (without progression to MDS) as well as differences in analyses of peripheral
blood vs bone marrow specimens.
|
6       SHALLIS et al.

TA B L E 2   Frequently mutated genes in


Mutated gene Incidence, % Prognostic impact Reference(s)
acquired aplastic anemia
68,85
BCOR or BCORL 4.0‐9.3 Favorable response to IST, improved
PFS and OS
68,85
DNMT3A 5.3‐8.4 Poor response to IST, inferior PFS
and OS
68,85
ASXL1 6.2‐8.0 Poor response to IST, inferior PFS
and OS
85
PIGA 7.5‐40 Favorable response to IST, improved
PFS and OS
46
TERT 5.6 Unknown
47
TERC 3.3 Unknown
85
JAK2 and JAK3 1.8 Poor response to IST, inferior PFS
85
RUNX1 1.5 Poor response to IST, inferior PFS
and OS
85
TP53 1.5 Poor response to IST, inferior OS
85
CSMD1 1.0 Poor response to IST, inferior OS
68,85
TET2 0.6−0.7 Unknown
68
SRSF2 0.6 Unknown
68
U2AF1 0.6 Unknown
68
ERBB2 0.6 Unknown
68
MPL 0.6 Unknown
85
TERT 0.5 Unknown
48
TERF1 or TERF2 N/A Unknown

IST, immunosuppressive therapy; OS, overall survival; PFS, progression‐free survival.

hemoglobinuria (PNH) clone.80-82 Between 7.5% and 40% of AA pa‐ et  al72 also demonstrated a dominance of cytosine to thymine tran‐
tients are found to have mutations in PIGA, but PNH clones (though sition at CpG dinucleotides and a predilection for nonsense, frame‐
typically small) are found in up to 68% of patients at time of AA di‐ shift, and splice‐site alterations. Interestingly, the simple presence
agnosis and up to 19% will eventually develop overt PNH, another of an acquired somatic mutation (other than that of telomere main‐
clonal bone marrow failure disorder.72,78,83,84 GPI‐deficient HSCs tenance genes such as TERT and TERC) has been associated with
may in fact have some protective role in escape from the T‐cell‐me‐ shorter telomere length by a mechanism hypothesized to be the re‐
diated destruction which otherwise induces apoptosis of normal sult of hematopoietic stress.69
HSCs in the stem cell compartment. HSCs with these mutations may As with cytogenic abnormalities, outcomes in AA patients seem
have survival advantages by being less immunogenic as evidenced to be influenced by the presence of specific somatic mutations.
by more frequent PIGA mutations and may have a higher chance for Mutations in PIGA, BCOR, and BCORL1 are associated with better
recruitment especially when the pool of HSCs is depleted.85-87 response to IST as well as improved PFS and OS, while AA patients
Other than PIGA mutations, the most frequently detected somatic with mutated DNMT3A, ASXL1, JAK2/JAK3, or RUNX1 fare worse
mutations in AA are in that in BCOR, BCORL1, and the MDS‐related with regard to IST response and survival.69,72,88 AA patients with
somatic mutations in DNMT3A and ASXL1, which in one study cumu‐ mutations in DNMT3A or ASXL1 are shown to exhibit a 40% chance
latively accounted for 77% of all somatic mutation‐positive patients. of developing MDS, which is far more significant than the observed
About a third of AA patients have multiple, independent mutations, 10% incidence of such in AA patients in general.69,72,89
including that of the same gene, which supports a stout mechanism Primary or founder clones in AA appear to be consistently small
by which such clones are selected for expansion and evolution.72,87 at diagnosis, but clone sizes have been shown to be dynamic and
Median variant allelic frequency (VAF) seems to range from 9% to fluctuate without evidence of progression to MDS or AML or even
20% based on two strong studies detecting such, and three‐quarters worsened phenotypic disease. In some cases, initially detectable
of all detected somatic mutations have a VAF of <10%.69,72 Although mutations (specifically those clones with BCOR/BCORL or PIGA mu‐
less common, recurring mutations in JAK2/JAK3, RUNX1, TP53, tations) can even subsequently evade measurement during the dis‐
CSMD1, and TET2 have been recognized.72 Rare (in <3% of acquired ease course.72 The acquisition of new secondary or subclones (which
AA cases collectively) mutations in SRSF2, U2AF1, ERBB2, and MPL can be a product of the original or dominant HSC clone) has also
were identified in one of the more robust studies using NGS and shown to be variable, ranging from no appreciable clinical impact to
array‐based karyotyping of AA patients to date (Table 2).72 Yoshizato (in the case of clones with mutated ASXL1 or DNMT3A) a substantial
SHALLIS et al. |
      7

increase in clone size and even an occult metric preceding progres‐ abnormalities or somatic mutations, respectively) without dysplasia
sion to malignant myelopoiesis. ASXL1 and DNMT3A and to a lesser or a cytopenia is defined as clonal hematopoiesis of indeterminate
degree RUNX1 and U2AF1 clone sizes tend to increase through the potential (CHIP) and such an entity raises more questions than con‐
course of disease.72 Because of a lack of consistent predictability fidence in a pathogenic diagnosis.86,87 In addition, acquired genetic
and definitive understanding of clonal selection, most AA experts mutations (including those recurring in myeloid malignancy such as
recommend not basing therapeutic decision‐making on the presence MDS [ASXL1, DNMT3A, etc]) may arise out the hematopoietic stress
or size of clonal disease, although specific somatic mutations may induced by AA and become a substrate for clonal selection.
raise suspicion.90 Recent efforts have led to promising metrics to distinguish AA
from MDS irrespective of CH. Levels of circulating S100A8, a Toll‐
like receptor ligand which exerts its proinflammatory effect via
8 | E V I D E N C E I N C LO N A L E VO LU TI O N tumor necrosis factor receptor‐associated factor (TRAF) and nuclear
A N D QU E S TI O N S FRO M C LO N A L factor‐κB (NF‐κB)‐related transcription of TNF‐α and IL‐1b, were
H E M ATO P O I E S I S shown in a recent study to be elevated in patients with MDS, but not
those with AA (or healthy controls).98 Such a distinction may provide
Although AA is considered to be a nonmalignant disease with poten‐ a sensitive diagnostic tool to differentiate the two entities whose
tial cure using IST or alloSCT, it can be complicated by the develop‐ treatment varies in most circumstances. The presence of CH (even
ment of late clonal disease. This manifestation may provide insight in cases with a large VAF or somatic mutation associated with patho‐
as to further molecular or other biological pathogenic mechanisms genic significance in MDS) in a AA patient does not alter therapeutic
of AA development. AA patients have a higher incidence of AML decision‐making in current practice (such as for whom IST would be
and MDS, namely 7% and 10%, respectively, at 11‐year follow‐up preferred), but this may in fact change with improved understanding
after receiving IST.91 The increased prevalence of late clonal compli‐ of AA molecular pathogenesis.
cations like MDS and paroxysmal nocturnal hemoglobinuria (PNH) in
AA suggests a link between clonal hematopoiesis (CH) and AA.85 CH
(characterized by either a karyotypic abnormality or somatic muta‐ 9 | FU T U R E D I R EC TI O N S
tion) can be detected in approximately a quarter of adult AA cases
(and up to 70% in cohorts including children).72,78 Interestingly, T‐ There has been noticeable progress in dissecting the underlying and
cell‐directed IST itself has been associated with an increased risk of complex pathophysiology of AA over the past years. This has been
clonal evolution.80 in no small part due to the advancement of deep gene‐sequencing
Clonal disease is detected at time of AA diagnosis in a sub‐ techniques and SNP arrays for detecting genetic abnormalities along
stantial proportion of patients without morphologic evidence of with the application of multicolor flow cytometry and fluorescently
MDS.72,73,78,92 A more challenging task remains the distinction be‐ conjugated monoclonal antibodies for the assessment of protein ex‐
tween AA and an uncommon form of MDS known as hypoplastic pression and exploring distinct subpopulation of T‐cells. Single cell
MDS (hMDS). hMDS comprises approximately 15% of MDS cases RNA (scRNA) sequencing, which is a relatively new technique, allows
and is characterized by bone marrow cellularity <30% in patients direct analysis of the transcriptomes of individual cells, comparison
aged <70 years or <20% cellularity in those older than age 70 years.93 to a larger number of heterogeneous cells and thus will provide fur‐
The impossibility of morphologically distinguishing AA from hMDS ther characteristics of gene expression.99 Such a method may allow
has led many experts to identify this phenomenon as an overlap syn‐ new insight into AA pathophysiology, specifically the underlying
drome. A retrospective study of hMDS patients revealed that 62% recurring mutations and genetic instability as it related to clonal
of patients had normal cytogenetics and only 21% had mutations evolution. Ultimately this technique can potentially yield innova‐
in either ASXL1, DNMT3A, or BCOR; the overwhelming majority of tive experimental approaches in the future to explore gene‐targeted
hMDS patients with a somatic mutation had a prior history of AA.93 therapy.100
Because of these inconsistencies, the presence of CH does not aid One of the more significant challenges to the study of AA mo‐
in the distinction between AA and hMDS. In fact, the identification lecular pathophysiology has been the paucity of progenitor cell
of several cytogenetic aberrations (for instance, monosomy 7, which attainment to perform various biotechniques. The use of induced
is identified in up to 13% of AA cases at diagnosis) by conventional pluripotent stem cell (iPSC), which can be derived into hematopoi‐
karyotyping is diagnostic of MDS even in the absence of morphologic etic cell lineages, has recently provided some promise in overcom‐
dysplasia as per the World Health Organization (WHO) classification ing this barrier and represents a new perspective in the study of
of myeloid neoplasms.68,94,95 The risk of CH (as well as aneuploidy or AA biology and the pathological genotype. Combining the use of
copy‐neutral loss of heterozygosity) increases with age alone with up iPSC with the rapid improvement of RNA splicing and gene edit‐
to 10% of patients aged 70 years or old as per evidence reanalyzing ing techniques such as CRISPER/Cas9 may help with the induction
single nucleotide polymorphism (SNP) array data generated for indi‐ of mutations and discover genetic or epigenetic events related to
viduals with no history of hematological cancer.96,97 Such CH (quan‐ previously identified phenomena such as telomere shortening.
tified by least two metaphases or VAF of at least 2% for karyotypic This would ultimately provide a suitable animal model to study AA
|
8       SHALLIS et al.

in vitro as well as a therapeutic window by means of correcting 6. Kaufman DW, Kelly JP, Jurgelon JM, et al. Drugs in the aetiology
genetic mutations via gene editing. 101,102
Eventually, the above of agranulocytosis and aplastic anaemia. Eur J Haematol Suppl.
1996;60:23‐30.
should produce a more precise ability to distinguish founder mu‐
7. Hamerschlak N, Maluf E, Pasquini R, et al. Incidence of aplastic
tations among other acquired subcloncal populations and those anemia and agranulocytosis in Latin America–the LATIN study. Sao
that arise with age. Such may afford the ability to predict clini‐ Paulo Med J. 2005;123(3):101‐104.
cal outcomes of AA patients and thus allow a more appropriate 8. Smith MT. Overview of benzene‐induced aplastic anaemia. Eur J
Haematol Suppl. 1996;60:107‐110.
algorithm for therapeutic decisions including allogeneic stem cell
9. Fleming LE, Timmeny W. Aplastic anemia and pesticides. An etio‐
transplantation. logic association? J Occup Med. 1993;35(11):1106‐1116.
10. Issaragrisil S, Kaufman DW, Anderson T, et al. The epidemiology of
aplastic anemia in Thailand. Blood. 2006;107(4):1299‐1307.
11. Issaragrisil S, Leaverton PE, Chansung K, et al. Regional patterns in
10 | CO N C LU S I O N
the incidence of aplastic anemia in Thailand. The Aplastic Anemia
Study Group. Am J Hematol. 1999;61(3):164‐168.
The pathogenesis of AA is directly related to the destruction of he‐ 12. West BC, DeVault Jr GA, Clement JC, Williams DM. Aplastic ane‐
matopoietic stem cells. A myriad of environmental insults may drive mia associated with parenteral chloramphenicol: review of 10
cases, including the second case of possible increased risk with
the antigen‐driven and autoimmune means of this destruction. Strong
cimetidine. Rev Infect Dis. 1988;10(5):1048‐1051.
data support the hypothesis that a disturbance of T‐cell homeostasis
13. Yunis AA. Chloramphenicol toxicity: 25 years of research. Am J
is the link between these two assertions. Defective telomerase func‐ Med. 1989;87(3N):44N–48N.
tion, acquired cytogenetic aberrations, and recurring somatic muta‐ 14. Laporte JR, Vidal X, Ballarin E, Ibanez L. Possible association be‐
tions have been implicated as playing a significant role in aiding the tween ocular chloramphenicol and aplastic anaemia–the absolute
risk is very low. Br J Clin Pharmacol. 1998;46(2):181‐184.
autoimmunity leading the acquired bone marrow failure of AA. Our
15. Handoko KB, Souverein PC, van Staa TP, et al. Risk of aplas‐
current understanding of the biological and molecular mechanisms tic anemia in patients using antiepileptic drugs. Epilepsia.
for the development of AA has been deepened by the advent SNP 2006;47(7):1232‐1236.
arrays and NGS, which permitted the discovery of offending genomic 16. Choudhry VP, Gupta S, Gupta M, Kashyap R, Saxena R. Pregnancy
associated aplastic anemia–a series of 10 cases with review of lit‐
abnormalities. Future development of newer technologies and tech‐
erature. Hematology. 2002;7(4):233‐238.
niques may soon reveal a definitive genetic origin of the tolerance 17. Fleming AF. Pregnancy and aplastic anaemia. Br J Haematol.
for the inappropriate and persistent immune response seen in AA. 1999;105(1):315.
18. Grau JM, Bosch X, Salgado AC, Urbano‐Marquez A. Human im‐
munodeficiency virus (HIV) and aplastic anemia. Ann Intern Med.
C O N FL I C T O F I N T E R E S T 1989;110(7):576‐577.
19. Pagliuca S, Gerard L, Kulasekararaj A, et al. Characteristics
All authors report no relevant disclosures. and outcomes of aplastic anemia in HIV patients: a brief report
from the severe aplastic anemia working party of the European
Society of Blood and Bone Marrow Transplantation. Bone Marrow
AU T H O R C O N T R I B U T I O N S Transplant. 2016;51(2):313‐315.
20. Lu J, Basu A, Melenhorst JJ, Young NS, Brown KE. Analysis of
All authors wrote and approved the final manuscript. T‐cell repertoire in hepatitis‐associated aplastic anemia. Blood.
2004;103(12):4588‐4593.
21. Baumelou E, Guiguet M, Mary JY. Epidemiology of aplastic anemia
ORCID in France: a case‐control study. I. Medical history and medication
use. The French Cooperative Group for Epidemiological Study of
Rory M. Shallis  http://orcid.org/0000-0002-8542-2944 Aplastic Anemia. Blood. 1993;81(6):1471‐1478.
22. de Masson A, Bouaziz JD, Peffault de Latour R, et al. Severe aplastic
anemia associated with eosinophilic fasciitis: report of 4 cases and
REFERENCES review of the literature. Medicine (Baltimore). 2013;92(2):69‐81.
23. Bonnotte B, Chauffert B, Caillot D, Martin F, Lorcerie B. Successful
1. Alter BP, Giri N, Savage SA, Rosenberg PS. Telomere length treatment with antithymocyte globulin and cyclosporin A of a se‐
in inherited bone marrow failure syndromes. Haematologica. vere aplastic anaemia associated with an eosinophilic fasciitis. Br J
2015;100(1):49‐54. Rheumatol. 1998;37(12):1358‐1359.
2. Collins J, Dokal I. Inherited bone marrow failure syndromes. 24. Risitano AM, Maciejewski JP, Green S, Plasilova M, Zeng W, Young
Hematology. 2015;20(7):433‐434. NS. In‐vivo dominant immune responses in aplastic anaemia: mo‐
3. Ehrlich P. Uber einen fall von anaemie mit bemerkungen uber re‐ lecular tracking of putatively pathogenetic T‐cell clones by TCR
generative veranderungen des knochenmarks. Charité‐Annale. beta‐CDR3 sequencing. Lancet. 2004;364(9431):355‐364.
1888;13:300‐305. 25. Zeng W, Kajigaya S, Chen G, Risitano AM, Nunez O, Young NS.
4. Montane E, Ibanez L, Vidal X, et al. Epidemiology of aplas‐ Transcript profile of CD4+ and CD8+ T cells from the bone
tic anemia: a prospective multicenter study. Haematologica. marrow of acquired aplastic anemia patients. Exp Hematol.
2008;93(4):518‐523. 2004;32(9):806‐814.
5. Issaragrisil S, Sriratanasatavorn C, Piankijagum A, et al. Incidence 26. Sloand E, Kim S, Maciejewski JP, Tisdale J, Follmann D, Young
of aplastic anemia in Bangkok. The Aplastic Anemia Study Group. NS. Intracellular interferon‐gamma in circulating and mar‐
Blood. 1991;77(10):2166‐2168. row T cells detected by flow cytometry and the response to
SHALLIS et al. |
      9

immunosuppressive therapy in patients with aplastic anemia. 46. Brummendorf TH, Maciejewski JP, Mak J, Young NS, Lansdorp
Blood. 2002;100(4):1185‐1191. PM. Telomere length in leukocyte subpopulations of patients with
27. Demeter J, Messer G, Schrezenmeier H. Clinical relevance of the aplastic anemia. Blood. 2001;97(4):895‐900.
TNF‐alpha promoter/enhancer polymorphism in patients with 47. Yamaguchi H, Baerlocher GM, Lansdorp PM, et al. Mutations of
aplastic anemia. Ann Hematol. 2002;81(10):566‐569. the human telomerase RNA gene (TERC) in aplastic anemia and
28. de Latour RP, Visconte V, Takaku T, et al. Th17 immune responses myelodysplastic syndrome. Blood. 2003;102(3):916‐918.
contribute to the pathophysiology of aplastic anemia. Blood. 48. Yamaguchi H, Calado RT, Ly H, et al. Mutations in TERT, the gene
2010;116(20):4175‐4184. for telomerase reverse transcriptase, in aplastic anemia. N Engl J
29. Rosenfeld SJ, Kimball J, Vining D, Young NS. Intensive immu‐ Med. 2005;352(14):1413‐1424.
nosuppression with antithymocyte globulin and cyclospo‐ 49. Savage SA, Calado RT, Xin ZT, Ly H, Young NS, Chanock SJ. Genetic
rine as treatment for severe acquired aplastic anemia. Blood. variation in telomeric repeat binding factors 1 and 2 in aplastic
1995;85(11):3058‐3065. anemia. Exp Hematol. 2006;34(5):664‐671.
30. Solomou EE, Rezvani K, Mielke S, et al. Deficient CD4+ CD25+ 50. Marsh J, Gutierrez‐Rodrigues F, Cooper J, et al. Heterozygous
FOXP3+ T regulatory cells in acquired aplastic anemia. Blood. RTEL1 variants in bone marrow failure and myeloid neoplasms.
2007;110(5):1603‐1606. Blood Adv. 2018;2(1):36‐48.
31. Shi J, Ge M, Lu S, et al. Intrinsic impairment of CD4(+) 51. Scheinberg P, Cooper JN, Sloand EM, Wu CO, Calado RT, Young
CD25(+) regulatory T cells in acquired aplastic anemia. Blood. NS. Association of telomere length of peripheral blood leukocytes
2012;120(8):1624‐1632. with hematopoietic relapse, malignant transformation, and sur‐
32. Giudice V, Feng X, Lin Z, et al. Deep sequencing and flow cytomet‐ vival in severe aplastic anemia. JAMA. 2010;304(12):1358‐1364.
ric characterization of expanded effector memory CD8(+)CD57(+) 52. Calado RT, Yewdell WT, Wilkerson KL, et al. Sex hormones, acting
T cells frequently reveals T‐cell receptor Vbeta oligoclonality on the TERT gene, increase telomerase activity in human primary
and CDR3 homology in acquired aplastic anemia. Haematologica. hematopoietic cells. Blood. 2009;114(11):2236‐2243.
2018;103(5):759‐769. 53. Bar C, Huber N, Beier F, Blasco MA. Therapeutic effect of andro‐
33. Kordasti S, Costantini B, Seidl T, et al. Deep phenotyping of Tregs gen therapy in a mouse model of aplastic anemia produced by
identifies an immune signature for idiopathic aplastic anemia and short telomeres. Haematologica. 2015;100(10):1267‐1274.
predicts response to treatment. Blood. 2016;128(9):1193‐1205. 54. Townsley DM, Dumitriu B, Liu D, et al. Danazol treatment for telo‐
34. Saunthararajah Y, Nakamura R, Nam JM, et al. HLA‐DR15 (DR2) is mere diseases. N Engl J Med. 2016;374(20):1922‐1931.
overrepresented in myelodysplastic syndrome and aplastic anemia 55. Khincha PP, Bertuch AA, Gadalla SM, Giri N, Alter BP, Savage
and predicts a response to immunosuppression in myelodysplastic SA. Similar telomere attrition rates in androgen‐treated and
syndrome. Blood. 2002;100(5):1570‐1574. untreated patients with dyskeratosis congenita. Blood Adv.
35. Bendtzen K, Morling N, Fomsgaard A, et al. Association between 2018;2(11):1243‐1249.
HLA‐DR2 and production of tumour necrosis factor alpha and in‐ 56. Alexander WS, Roberts AW, Nicola NA, Li R, Metcalf D.
terleukin 1 by mononuclear cells activated by lipopolysaccharide. Deficiencies in progenitor cells of multiple hematopoietic lineages
Scand J Immunol. 1988;28(5):599‐606. and defective megakaryocytopoiesis in mice lacking the thrombo‐
36. Callera F, Falcao RP. Increased apoptotic cells in bone marrow poietic receptor c‐Mpl. Blood. 1996;87(6):2162‐2170.
biopsies from patients with aplastic anaemia. Br J Haematol. 57. Kimura S, Roberts AW, Metcalf D, Alexander WS. Hematopoietic
1997;98(1):18‐20. stem cell deficiencies in mice lacking c‐Mpl, the receptor for
37. Scopes J, Bagnara M, Gordon‐Smith EC, Ball SE, Gibson FM. thrombopoietin. Proc Natl Acad Sci U S A. 1998;95(3):1195‐1200.
Haemopoietic progenitor cells are reduced in aplastic anaemia. Br 58. Feng X, Scheinberg P, Wu CO, et al. Cytokine signature profiles
J Haematol. 1994;86(2):427‐430. in acquired aplastic anemia and myelodysplastic syndromes.
38. Callera F, Garcia AB, Falcao RP. Fas‐mediated apoptosis with nor‐ Haematologica. 2011;96(4):602‐606.
mal expression of bcl‐2 and p53 in lymphocytes from aplastic 59. Alvarado L, Huntsman HD, Cheng H, Knutson JR, Larochelle A.
anaemia. Br J Haematol. 1998;100(4):698‐703. Heterodimerization of TPO and IFNγ impairs human hematopoi‐
39. Gidvani V, Ramkissoon S, Sloand EM, Young NS. Cytokine gene etic stem/progenitor cell signaling and survival in chronic inflam‐
polymorphisms in acquired bone marrow failure. Am J Hematol. mation. Blood. 2017;130:4.
2007;82(8):721‐724. 60. Desmond R, Townsley DM, Dumitriu B, et al. Eltrombopag re‐
40. Hosokawa K, Kajigaya S, Feng X, et al. A plasma microRNA signa‐ stores trilineage hematopoiesis in refractory severe aplastic
ture as a biomarker for acquired aplastic anemia. Haematologica. anemia that can be sustained on discontinuation of drug. Blood.
2017;102(1):69‐78. 2014;123(12):1818‐1825.
41. Giudice V, Banaszak LG, Gutierrez‐Rodrigues F, et al. Circulating 61. Olnes MJ, Scheinberg P, Calvo KR, et al. Eltrombopag and im‐
exosomal microRNAs in acquired aplastic anemia and myelodys‐ proved hematopoiesis in refractory aplastic anemia. N Engl J Med.
plastic syndromes. Haematologica. 2018;103(7):1150‐1159. 2012;367(1):11‐19.
42. Calado RT, Young NS. Telomere diseases. N Engl J Med. 62. Townsley DM, Scheinberg P, Winkler T, et al. Eltrombopag added
2009;361(24):2353‐2365. to standard immunosuppression for aplastic anemia. N Engl J Med.
43. Liu K, Schoonmaker MM, Levine BL, June CH, Hodes RJ, Weng 2017;376(16):1540‐1550.
NP. Constitutive and regulated expression of telomerase reverse 63. Gill H, Leung GM, Lopes D, Kwong YL. The thrombopoietin mimet‐
transcriptase (hTERT) in human lymphocytes. Proc Natl Acad Sci U ics eltrombopag and romiplostim in the treatment of refractory
S A. 1999;96(9):5147‐5152. aplastic anaemia. Br J Haematol. 2017;176(6):991‐994.
44. Roth A, Baerlocher GM, Schertzer M, Chavez E, Duhrsen U, 64. Lee JW, Jang JH, Lee SE, Jung CW, Park S, Oh IH. Efficacy and
Lansdorp PM. Telomere loss, senescence, and genetic insta‐ safety of romiplostim in patients with aplastic anemia refractory
bility in CD4+ T lymphocytes overexpressing hTERT. Blood. to immunosuppressive therapy: 1‐year interim analysis of phase 2
2005;106(1):43‐50. clinical trial. Blood. 2016;128:3910.
45. Ball SE, Gibson FM, Rizzo S, Tooze JA, Marsh JC, Gordon‐Smith 65. Lee JW, Lee LS, Jung CW, et al. Hematologic response to romi‐
EC. Progressive telomere shortening in aplastic anemia. Blood. plostim treatment is associated with stimulation of primitive stem/
1998;91(10):3582‐3592. progenitor cells and stromal cells in patients with aplastic anemia
|
10       SHALLIS et al.

refractory to immunosuppressive therapy: a 2‐year interim explor‐ 86. Stanley N, Olson TS, Babushok DV. Recent advances in under‐
atory analysis of a phase 2 clinical trial. Blood. 2017;130:1167. standing clonal haematopoiesis in aplastic anaemia. Br J Haematol.
66. Maciejewski JP, Kim S, Sloand E, Selleri C, Young NS. Sustained 2017;177(4):509‐525.
long‐term hematologic recovery despite a marked quantita‐ 87. Ogawa S. Clonal hematopoiesis in acquired aplastic anemia. Blood.
tive defect in the stem cell compartment of patients with aplas‐ 2016;128(3):337‐347.
tic anemia after immunosuppressive therapy. Am J Hematol. 88. Marsh JC, Kulasekararaj AG. Management of the refractory aplas‐
2000;65(2):123‐131. tic anemia patient: what are the options? Hematology Am Soc
67. Young N, Griffith P, Brittain E, et al. A multicenter trial of anti‐ Hematol Educ Program. 2013;2013:87‐94.
thymocyte globulin in aplastic anemia and related diseases. Blood. 89. Marsh JC, Kulasekararaj AG. Management of the refrac‐
1988;72(6):1861‐1869. tory aplastic anemia patient: what are the options? Blood.
68. Keung YK, Pettenati MJ, Cruz JM, Powell BL, Woodruff RD, Buss 2013;122(22):3561‐3567.
DH. Bone marrow cytogenetic abnormalities of aplastic anemia. 90. Zeidan AM, Battiwalla M, Berlyne D, Winkler T. Aplastic ANEMIA
Am J Hematol. 2001;66(3):167‐171. and MDS International Foundation (AAMDSIF): bone marrow fail‐
69. Kulasekararaj AG, Jiang J, Smith AE, et al. Somatic mutations iden‐ ure disease scientific symposium 2016. Leuk Res. 2017;53:8‐12.
tify a subgroup of aplastic anemia patients who progress to myel‐ 91. Kojima S, Ohara A, Tsuchida M, et al. Risk factors for evolution of
odysplastic syndrome. Blood. 2014;124(17):2698‐2704. acquired aplastic anemia into myelodysplastic syndrome and acute
70. Sloand EM, Mainwaring L, Fuhrer M, et al. Preferential suppression myeloid leukemia after immunosuppressive therapy in children.
of trisomy 8 compared with normal hematopoietic cell growth by Blood. 2002;100(3):786‐790.
autologous lymphocytes in patients with trisomy 8 myelodysplas‐ 92. Lane AA, Odejide O, Kopp N, et al. Low frequency clonal muta‐
tic syndrome. Blood. 2005;106(3):841‐851. tions recoverable by deep sequencing in patients with aplastic
71. Dumitriu B, Feng X, Townsley DM, et al. Telomere attrition and anemia. Leukemia. 2013;27(4):968‐971.
candidate gene mutations preceding monosomy 7 in aplastic ane‐ 93. Hunt A, Potter VT, et al. Hypoplastic MDS is a distinct clinico‐
mia. Blood. 2015;125(4):706‐709. pathological entity with somatic mutations frequent in patients
72. Yoshizato T, Dumitriu B, Hosokawa K, et al. Somatic muta‐ with prior aplastic anaemia with favorable clinical outcome. Blood.
tions and clonal hematopoiesis in aplastic anemia. N Engl J Med. 2014;124:3269.
2015;373(1):35‐47. 94. Arber DA, Orazi A, Hasserjian R, et al. The 2016 revision to the
73. Maciejewski JP, Risitano A, Sloand EM, Nunez O, Young NS. World Health Organization classification of myeloid neoplasms
Distinct clinical outcomes for cytogenetic abnormalities evolving and acute leukemia. Blood. 2016;127(20):2391‐2405.
from aplastic anemia. Blood. 2002;99(9):3129‐3135. 95. Vardiman JW, Thiele J, Arber DA, et al. The 2008 revision of the
74. Gupta V, Brooker C, Tooze JA, et al. Clinical relevance of cyto‐ World Health Organization (WHO) classification of myeloid neo‐
genetic abnormalities at diagnosis of acquired aplastic anaemia in plasms and acute leukemia: rationale and important changes.
adults. Br J Haematol. 2006;134(1):95‐99. Blood. 2009;114(5):937‐951.
75. Saitoh T, Saiki M, Kumagai T, Kura Y, Sawada U, Horie T. Spontaneous 96. Jacobs KB, Yeager M, Zhou W, et al. Detectable clonal mo‐
clinical and cytogenetic remission of aplastic anemia in a patient saicism and its relationship to aging and cancer. Nat Genet.
with del(13q). Cancer Genet Cytogenet. 2002;136(2):126‐128. 2012;44(6):651‐658.
76. Ishiyama K, Karasawa M, Miyawaki S, et al. Aplastic anaemia with 97. Xie M, Lu C, Wang J, et al. Age‐related mutations associated
13q‐: a benign subset of bone marrow failure responsive to immu‐ with clonal hematopoietic expansion and malignancies. Nat Med.
nosuppressive therapy. Br J Haematol. 2002;117(3):747‐750. 2014;20(12):1472‐1478.
77. Katagiri T, Sato‐Otsubo A, Kashiwase K, et al. Frequent loss of 98. Giudice V, Wu Z, Kajigaya S, et al. Circulating S100A8 and
HLA alleles associated with copy number‐neutral 6pLOH in ac‐ S100A9 protein levels in plasma of patients with acquired aplas‐
quired aplastic anemia. Blood. 2011;118(25):6601‐6609. tic anemia and myelodysplastic syndromes. Cytokine. 2018;pii:
78. Babushok DV, Perdigones N, Perin JC, et al. Emergence of clonal S1043-4666(18)30274-6.
hematopoiesis in the majority of patients with acquired aplastic 99. Haque A, Engel J, Teichmann SA, Lonnberg T. A practical guide
anemia. Cancer Genet. 2015;208(4):115‐128. to single‐cell RNA‐sequencing for biomedical research and clinical
79. Babushok DV, Duke JL, Xie HM, et al. Somatic HLA mutations ex‐ applications. Genome Med. 2017;9(1):75.
pose the role of class I‐mediated autoimmunity in aplastic anemia 100. Zhao X, Gao S, Wu Z, et al. Single‐cell RNA‐seq reveals a distinct
and its clonal complications. Blood Adv. 2017;1(22):1900‐1910. transcriptome signature of aneuploid hematopoietic cells. Blood.
80. Li Y, Li X, Ge M, et al. Long‐term follow‐up of clonal evolutions 2017;130(25):2762‐2773.
in 802 aplastic anemia patients: a single‐center experience. Ann 101. Melguizo‐Sanchis D, Xu Y, Taheem D, et al. iPSC modeling of se‐
Hematol. 2011;90(5):529‐537. vere aplastic anemia reveals impaired differentiation and telomere
81. Li LY, Liu ZY, Liu H, Liu CY, Shao ZH, Fu R. Deep sequencing of shortening in blood progenitors. Cell Death Dis. 2018;9(2):128.
whole genome exon in paroxysmal nocturnal hemoglobinuria. Am 102. Banaszak LG, Giudice V, Zhao X, et al. Abnormal RNA splicing
J Hematol. 2017;92(4):E51–E53. and genomic instability after induction of DNMT3A mutations by
82. Shen W, Clemente MJ, Hosono N, et al. Deep sequencing reveals CRISPR/Cas9 gene editing. Blood Cells Mol Dis. 2018;69:10‐22.
stepwise mutation acquisition in paroxysmal nocturnal hemoglo‐
binuria. J Clin Invest. 2014;124(10):4529‐4538.
83. Afable 2nd MG, Tiu RV, Maciejewski JP. Clonal evolution in
How to cite this article: Shallis RM, Ahmad R, Zeidan AM.
aplastic anemia. Hematology Am Soc Hematol Educ Program.
Aplastic anemia: Etiology, molecular pathogenesis, and
2011;2011:90‐95.
84. Sugimori C, Chuhjo T, Feng X, et al. Minor population of CD55‐ emerging concepts. Eur J Haematol. 2018;00:1–10. https://
CD59‐ blood cells predicts response to immunosuppressive doi.org/10.1111/ejh.13153
therapy and prognosis in patients with aplastic anemia. Blood.
2006;107(4):1308‐1314.
85. Young NS. The problem of clonality in aplastic anemia: Dr
Dameshek’s riddle, restated. Blood. 1992;79(6):1385‐1392.

You might also like