You are on page 1of 19

review

Advances in understanding the pathogenesis of acquired


aplastic anaemia

Lucio Luzzatto1 and Antonio M. Risitano2


1
Muhimbili University of Health and Allied Sciences, Dar-es-Salaam, Tanzania, and 2Department of Clinical Medicine and Surgery,
Federico II University, Naples, Italy

marrow of another patient (Marmont, 1995), although the


Summary
title of their article, published in 1904, used the term perni-
This review examines the evidence that bone marrow failure cious anaemia. In what is perhaps the first major haematol-
(BMF) in aplastic anaemia (AA) is due to loss of ogy textbook, Le Emopatie (Ferrata, 1934), it is instead stated
haematopoietic stem cells (HSCs), which, in turn, is caused emphatically – and of course correctly – that AA should not
by deranged immunity and inflammation. We also consider be confused with pernicious anaemia: today this terminology
how the course of the disease and the response to immuno- seems strangely twisted, because pernicious anaemia is emi-
suppressive therapy are influenced by the nature and speci- nently curable, whereas AA is still a pernicious threat. From
ficity of the pathogenic process. A somatic mutation of the the point of view of haematopoietic function, the regenera-
PIGA gene underlies the clonal disease paroxysmal nocturnal tion of effete blood cells is woefully inadequate in AA: hence,
haemoglobinuria (PNH): there is direct evidence that the the term bone marrow failure (BMF) is often used in prac-
expansion of the PIGA mutant clone results from Darwinian tice as synonymous with AA.
selection exerted by a glycosyl-phosphatidyl-inositol -specific AA can be inherited or acquired, and there are three
auto-immune attack. Thus, PNH patients are a unique subset major groups of inherited AA (Wegman-Ostrosky & Savage,
of patients with AA, in whom haematopoiesis recovers 2017); Table I.
through this escape mechanism. A similar process, although Among several possible causes of acquired AA, the one for
less effective, may operate when the auto-immune attack is which there is most definitive evidence is a heavy dose of
against a human leucocyte antigen (HLA) molecule and an radiation. Indeed, an industrial accident near Belgrade (Ser-
HLA mutation has produced a clone missing that molecule. bia, at that time Yugoslavia) caused severe BMF in at least 6
We then discuss the significance of other mutant clones that men who worked at a nuclear facility (Champlin, 1989). This
are frequently found in AA, presumably due to a combina- accident was an urgent stimulus to treating BMF by bone
tion of genetic drift and selection. These clones are not cau- marrow transplantation [BMT: (Mathe et al, 1958); in the
sative of AA, but they emerge in AA and they may be pre- event, the procedure was unsuccessful because the crucial
leukaemic: unlike a PIGA mutant clone, in general they are histocompatibility requirements were not yet known]. Not
unable to effectively reconstitute haematopoiesis. surprisingly, cytotoxic agents, such as cyclophosphamide,
busulphan or fludarabine, also cause dose-dependent BMF,
Keywords: aplastic anaemia, PNH, MDS, clonality, clonal and are indeed used deliberately for the purpose of bone
expansion, clonal evolution. marrow ablation as part of ‘conditioning’ prior to BMT. In
these situations, bone marrow aplasia is clearly secondary to a
physical or chemical myelo-toxic insult, and whether we
Introduction
should call it AA is a matter of semantics; but the majority
Aplastic anaemia is a rare disease and a serious disease. Paul of patients diagnosed with AA have not received heavy radia-
Ehrlich is credited with first reporting a patient with this tion or cytotoxic drugs. Idiosyncrasy to non-cytotoxic drugs,
condition in 1888; and Vaquez and Aubertin were probably e.g. chloramphenicol, is often quoted as a possible cause of
first in coining the word aplasique when describing the bone AA (Young, 1994); whereas among infectious causes the best
known association is with viral hepatitis (Brown et al, 1997):
however, the majority of the millions of subjects who have
Correspondence: Lucio Luzzatto, Professor of Haematology, suffered hepatitis do not go on to develop AA. In the major-
Department of Haematology and Blood Transfusion, Muhimbili ity of cases of acquired AA (or primary BMF) we are unable
University of Health and Allied Sciences, P.O.Box 65001, to identify a specific cause, and therefore it is termed idio-
Dar-es-Salaam, Tanzania pathic. For the sake of brevity in this paper, unless otherwise
E-mails: lluzzatto@blood.ac.tz; lucio.luzzatto@unifi.it indicated, AA means idiopathic acquired AA.

First published online 5 July 2018 ª 2018 British Society for Haematology and John Wiley & Sons Ltd
doi: 10.1111/bjh.15443 British Journal of Haematology, 2018, 182, 758–776
Review

Schwachman-Diamond syndrome

AA, aplastic anaemia; GPI, glycosyl-phosphatidyl-inositol; HSCs, haematopoietic stem cells; IFNG, interferon-c; MDS, myelodysplastic syndrome; PNH, paroxysmal nocturnal haemoglobinuria; TNF,
General concepts

PNH, AA/PNH. Idiopathic AA


Pancytopenia in the peripheral blood before aplasia is often

Dyskeratosis congenita
the first finding in patients with AA; and it is, in first

MDS in the elderly?


approximation, an indictment of the haematopoietic stem

Post-radiation AA
Post-hepatitis AA
Fanconi anaemia
Disease example

cell (HSC).

Idiopatic AA
Idiopatic AA
When allogeneic BMT was introduced as a rational treat-
ment for patients with BMF – whether primary or secondary

CHIP?*
– it was observed that sometimes the transplanted bone mar-
row was rejected, but later the patient recovered (Thomas
et al, 1972; Mathe & Schwarzenberg, 1976). This remarkable
phenomenon, autologous reconstitution or regeneration of
Defective ribosome assembly/function (ribosomoapthy)

haematopoiesis [which often entails a good prognosis, (Pic-


Other target-specific auto-immune attack on HSCs
HLA-B4002-specific auto-immune attack on HSCs

cin et al, 2010)], was promptly attributed to the ‘condition-


ing’ regimen that, aiming to prevent rejection, was strongly
Abnormal telomere attrition (telomeropathy)

GPI-specific auto immune attack on HSCs

immune-suppressive (Speck et al, 1976a, 1978). This led to


the introduction of intensive immuno-suppressive treatment
(IST),including anti-lymphocyte globulin (Speck et al, 1976b,
Loss of stem cells/loss of stemness
Damage mediated by IFNG, TNF

1978, 1981; Gluckman et al, 1982; Champlin et al, 1983),


methylprednisolone(Doney et al, 1992) and cyclosporine
Direct damage to HSCs

(Frickhofen et al, 1991, 2003; Rosenfeld et al, 1995), that are


Table I. Various modalities that can result in depletion of haematopoietic stem cells and eventually in bone marrow failure.

Impaired DNA repair

still in use today. Given that IST produces remission and,


Main mechanism

often, cure of AA in over 70% of cases (Rosenfeld et al,


2003; Teramura et al, 2007; Scheinberg et al, 2011; Tichelli
*CHIP: clonal haematopoiesis of indeterminate potential, which at the moment must not be considered a disease.

et al, 2011), we can legitimately infer that, at least in a large


majority of cases, the primary mechanism of BMF in AA is
an auto-immune process. With this in mind, we must review
three steps in pathogenesis: (i) whether and in which way
stem cells are damaged; (ii) the effectors of damage; (iii) the
Mutations in genes of the Fanconi complex

source of the damage [see also (Risitano et al, 2007)]. But


first, we briefly outline the relationship between AA and
Mutation in DKC1, TERT or TERC†

paroxysmal nocturnal haemoglobinuria (PNH), because it


Many previous cell division cycles

provides a key to understanding the pathophysiology of AA.


Severe inflammatory process

AA and PNH
Main causative factor

†Less frequent genes causing dyskeratosis congenita are not listed here.
Auto reactive T cells
Mutations in SDBS

If acquired AA is rare, PNH is even more rare and, being an


acquired haemolytic anaemia, it features in a different chap-
ter from AA in textbooks. Nevertheless, AA and PNH over-
Radiation

lap in many patients and, given the rarity of both diseases,


this cannot be a coincidence (Dameshek, 1967). PNH is
defined by the presence of a large population of blood cells
that, as a result of an inactivating somatic mutation in the
X-linked gene PIGA, are deficient in all proteins that are nor-
Inherited bone marrow failure syndromes

mally tethered to the cell membrane through a glycosyl-phos-


phatidyl-inositol (GPI) molecule: because these include the
Acquired Aplastic Anaemia (AA)

complement regulatory surface proteins CD59 and CD55,


GPI-deficient red cells are exquisitely sensitive to lysis by
activated complement (Luzzatto & Araten, 2006). Clinically,
tumour necrosis factor.

PNH is typified by the triad of severe haemolysis, thrombosis


and cytopenias with high reticulocytosis, very high serum
Patient group

lactate dehydrogenase and erythroid hyperplasia in the bone


marrow, it could be hardly more different from AA. How-
Elderly

ever, cytopenias are reminiscent of AA; and the first hint to


a relationship between PNH and AA was that the clinical

ª 2018 British Society for Haematology and John Wiley & Sons Ltd 759
British Journal of Haematology, 2018, 182, 758–776
Review

picture may shift, over time, from one to the other (or vicev- two ancillary issues: (1) whether HSC failure is endogenous
ersa) (Dacie & Lewis, 1961; Lewis & Dacie, 1967). Later, the or exogenous; (2) whether it is reversible or irreversible.
development of PNH in AA patients was regarded as a ‘com- Regarding (1), given that mutations in any of several genes
plication’ of IST (Tichelli et al, 1994): more probably, IST can cause inherited AA (see above and Table I), it is always
had allowed AA patients to survive and to develop PNH of possible that what appeared to be acquired AA is in fact inher-
their own accord, as discussed below. Another interesting ited AA (Wegman-Ostrosky & Savage, 2017); or that a muta-
link was the finding that red cells with high sensitivity to tion in one of the same genes that can cause inherited AA, or
complement, now known to be GPI-deficient, are not even in other genes, may make HSCs more susceptible to
uncommon in AA patients, as first shown by Ben-Bassat et al developing acquired AA (Calado & Young, 2009). An analogy
(1975), and subsequently confirmed by other groups (Fores between BMF in dyskeratosis congenita (DC) and in acquired
et al, 1995; Wang et al, 2002; Scheinberg et al, 2010). In AA is that significant shortening of telomeres is found in both
1989 it was suggested that PNH develops when a somatic (Ball et al, 1998; Brummendorf et al, 2001). In DC this results
mutation, now known to be in PIGA (Takeda et al, 1993), from specific mutations (Calado & Young, 2009), whereas the
co-exists with BMF: indeed, that PNH develops because cause is not known in acquired AA, but it signifies that the
PIGA mutant stem cells can escape the damage suffered by stem cells have become abnormal. Although the possible role
PIGA-normal stem cells (Rotoli & Luzzatto, 1989a,b) of the bone marrow stroma in causing AA has been investi-
(Fig 1C, Fig 2). In keeping with this notion, GPI-deficient gated [e.g., the physiological immunomodulatory activity of
PIGA mutant granulocytes were found in normal subjects mesenchymal stem cells may be impaired in AA (Bacigalupo
(Araten et al, 1999): they may derive from a mutant HSC or et al, 2005)], patients can recover from AA following allo-
from a progenitor cell downstream of a HSC. Based on the geneic BMT while retaining their own bone marrow stroma
large dataset of an international registry (Schrezenmeier et al, (Spyridonidis et al, 2005); and stromal cells from AA marrows
2014), about one-half of PNH patients have a previous his- can form a functional haematopoietic stem cell niche (Miche-
tory of AA (and this is certainly an under-estimate, because a lozzi et al, 2017). Thus, there is overwhelming evidence that
diagnosis of mild AA preceding PNH might have been the central problem is in the HSCs themselves (Fig 1A. Marsh
missed). PNH is so different from AA that it must retain its et al, 1991). Transcriptome analysis of CD34+ cells from AA
distinct status in clinical nosography: but from the point of patients has shown increased expression of genes implicated in
view of pathogenesis, the majority of PNH patients are a apoptosis, cytokine/chemokine signalling, stress response, and
subset of AA patients (Young, 1992). defence/immune response; whereas the expression of cell cycle
progress-enhancing genes was decreased (Zeng et al, 2004).
Regarding (2), clinical experience tells us that patients can
Stem cells are damaged in AA
recover from AA without BMT, i.e., without a supply of allo-
A stem cell is defined by its ability to undergo asymmetric geneic HSCs: at least in these cases, either not all HCSs were
division, whereby one daughter cell differentiates and the killed, or the damage was reversible.
other daughter cell is still a stem cell. The stem cell status
of an HSC depends on its niche (Zhang et al, 2003; Crane
The effectors of damage
et al, 2017), which comprises bone marrow stromal cells,
the extracellular matrix and the local levels of cytokines Antibodies have been detected in patients with AA against a
(Riether et al, 2015; Crane et al, 2017). In experimental ani- number of proteins (e.g., kinectin, moesin, post-meiotic seg-
mals, where HSCs can be quantified by in vivo assay, it has regation 1, diazepam-binding inhibitor-related protein 1)
been amply demonstrated that the shortage of stem cells is (Hirano et al, 2003, 2005; Feng et al, 2004; Takamatsu et al,
associated with AA (Scheinberg & Chen, 2013). In humans, 2007; Espinoza et al, 2009), which could be auto-antigens:
the closest surrogate of HSCs are the so–called long-term however, these auto-antibodies have not been shown to be
culture initiating cells (LTC-IC): their numbers in the cytotoxic and, rather than being pathogenic, they seem more
peripheral blood and in the bone marrow of patients with likely to be an epiphenomenon, as in other auto-immune
AA are drastically reduced (Fig 1B; Marsh et al, 1990; diseases.
Maciejewski et al, 1996). CD34+ cells are also decreased in Stem cells are maintained by a precise genetic and epige-
AA (Maciejewski et al, 1996; Manz et al, 1996). We infer netic programme. Retaining stemness whilst at the same time
therefore that BMF results from stem cell failure. In princi- being capable of differentiation through asymmetric divisions
ple, stem cells might be damaged in at least 3 different ways is a bit like walking a tightrope: it is not surprising that the
(Fig 3). They might be (i) killed, (ii) inhibited, or (iii) delicate balance can be disturbed, probably in more ways
induced to undergo symmetric rather than asymmetric divi- than one. Among several extracellular regulatory molecules,
sion: if both daughter cells differentiate the HSC pool will interferons, which are normally produced in response to
be depleted. infection, have emerged as powerful and potentially disturb-
At the moment we don’t know which of these mecha- ing. In the short term, interferons enhance the production of
nisms predominate(s). In the meantime, we must deal with neutrophils and lymphocytes, which is advantageous for an

760 ª 2018 British Society for Haematology and John Wiley & Sons Ltd
British Journal of Haematology, 2018, 182, 758–776
Review

(A) (B)

Fig 1. Historic steps in understanding the pathogenesis of idiopathic acquired aplastic anaemia (AA). (A) Stem cells failure in AA (from Marsh
et al, 1991). One sees a marked decrease in progenitor cells (granulocyte-macrophage colony-forming units; CFU-GM) generated by long-term
culture-initiating cells in patients with AA. The figure also shows that normal stroma does not correct the stem cell defect of AA patients; con-
versely, stroma for AA does not affect stem cells from normal controls. (B) In vitro growth of hematopoietic progenitors in AA. Anti-interferon-c
(black bars), compared to controls (white bars) markedly increases formation of colonies (colony-forming unit cells; CFU-C) from peripheral
blood mononuclear cells from a majority of 10 patients with severe AA (Zoumbos et al, 1985a). Different columns represent bone marrow
mononuclear cells grown in culture medium containing (a) no antiserum, (b) control hyperimmune sheep antiserum, (c) control hyperimmune
sheep antiserum, (d) sheep antiserum to human a-interferon (neutralizing antibodies, (e) sheep antiserum to human a-interferon (neutralizing
antibodies). C. Close relationship between the pathophysiology of AA and paroxysmal nocturnal haemoglobinuria (PNH). Rotoli and Luzzatto
(1989a) explicitly suggested that in PNH, a disease already known to be clonal, glycosyl-phosphatidyl-inositol-deficient haematopoietic stem cells
expand and take over haematopoiesis in the bone marrow of a patient with AA. The different panels represented different stages/clinical situa-
tions: an important implication was that patients with PNH are a subset of patients with AA. AAA, acquired aplastic anaemia; AL, acute leukae-
mia. (A) reproduced with permission from Marsh J.C., Chang J., Testa N.G., Hows J.M. and Dexter T.M. In vitro assessment of marrow ‘stem
cell’ and stromal cell function in aplastic anaemia. British Journal of Haematology 78, 258-267. © 1991 John Wiley & Sons, Inc. (B) reproduced
from Zoumbos, N.C., Gascon P., Djeu J.Y. and Young N.S. (1985). Interferon is a mediator of hematopoietic suppression in aplastic anemia
in vitro and possibly in vivo. Proc Natl Acad Sci U S A 82(1): 188-192. (C) Reprinted from Bailliere’s Clin Haematol, 2 (1), Rotoli, B. and Luz-
zatto, L., Paroxysmal nocturnal haemoglobinuria, 113-138, © 1989, with permission from Elsevier.

effective response to infection. A major advance in character- (Alvarado et al, 2017). Indirectly, IFNG acts on IFNG
izing how haematopoiesis is disturbed in patients with AA receptor-expressing macrophages and stromal cells, that in
was the discovery that the level of interferon-c (IFNG) was turn may affect stem cell quiescence and exert a pull towards
raised in their sera, and that addition of anti-IFNG markedly producing progenitor cells (Smith et al, 2016). A polymor-
increased numbers of in vitro haematopoietic colonies from phic site in the IFNG gene may increase susceptibility to
bone marrow cells of AA patients, but not from normal con- developing AA, as well as its severity (Bestach et al, 2015),
trols (Zoumbos et al, 1985a) (Fig 1B). These results have and may influence response to IST (Zeng et al, 2015). AA
been extensively confirmed (Selleri et al, 1996). Of course, all develops in mice genetically modified to express constitu-
conventional in vitro colonies are generated by progenitor tively low levels of IFNG (Lin et al, 2014). Tumour necrosis
cells that are downstream of HSCs, and therefore we cannot factor-a (TNFa could also affect HSCs (Riether et al, 2015):
automatically extrapolate to the latter from the former. IFNG TNFa-producing lymphocytes are increased in AA patients
may act on HSCs both directly and indirectly: directly, once (Schultz & Shahidi, 1994; Selleri et al, 1995), and their disap-
bound to the IFNG receptor, IFNG modulates the STAT and pearance tends to correlate with clinical response to IST
SOCS2 pathways (Krebs & Hilton, 2001), thus potentially (Dufour et al, 2001).
affecting proliferation and retention of stemness; in addition, The finding that these cytokines are increased in AA signi-
IFNG up-regulates FAS expression, thus making cells more fies that an inflammatory process is on-going (see below and
prone to apoptosis (Maciejewski et al, 1995). Very recent Table II). Multiple threads of evidence both in mice (Schein-
data suggest that IFNG may compromise the survival of berg & Chen, 2013) and in humans (Sloand et al, 2002),
HSCs by forming a heterodimer with thrombopoietin (TPO), may not be definitive proof that IFNG is a culprit, but they
with consequent impairment of TPO-cMPL signalling indict it as a prime suspect.

ª 2018 British Society for Haematology and John Wiley & Sons Ltd 761
British Journal of Haematology, 2018, 182, 758–776
Review

Fig 1. Continued.

with AA (reviewed in Young et al, 2006). Indirect evidence


The source of damage
for involvement of CD4+ T cells was the over-representation
Spurred by the fact that IST can overcome AA, a large body of certain human leucocyte antigen (HLA) alleles in AA
of accumulated evidence indicates that the damage to HSCs patients (Nakao et al, 1994). Activated CD8+ T cells are
originates from T cells (Young et al, 2006; Risitano et al, cytotoxic for bone marrow cells (Zoumbos et al, 1985b), lar-
2007). Here we will focus on two extreme possibilities. (i) gely through IFNG secretion. IFNG can be produced by
On one hand, a variety of abnormalities in the bone marrow CD4+ T cells, CD8+ T cells and Natural Killer T (NKT) cells,
can produce, as their final common pathway, a local level of all of which are increased in AA patients. Disorders of NKT
IFNG and other cytokines sufficient to cause serious damage or NKT-like cells (large granular lymphocytic cells, LGL) can
to a majority of HSCs. (ii) On the other hand, a small subset be associated with single or multiple cytopenias. LGLs secrete
of T cells, possible a ‘forbidden clone’, may recognize a soluble Fas receptors that are elevated in the serum of AA
specific molecular target in HSCs, whereby IFNG may be still patients; in addition, Fas-Receptor is overexpressed on
relevant in an ancillary capacity. These two possibilities are CD34+ cells of AA patients, consistent with susceptibility to
not mutually exclusive. apoptosis of HSCs (Maciejewski et al, 1995).
The combination of locally increased levels of IFNG in the
bone marrow and increased susceptibility to apoptosis could
Evidence for deranged immunity and inflammation
cause sufficient damage to HSCs to produce the clinical pic-
Abnormalities in relative proportions of T cells subsets and ture of AA. T-helper cell type 1 (Th1) polarization of T cells
in the T cell transcriptome have been reported in patients in AA patients is associated with TBX21 over-expression,

762 ª 2018 British Society for Haematology and John Wiley & Sons Ltd
British Journal of Haematology, 2018, 182, 758–776
Review

PIGA+ blood cell PIGA– blood cell

PIGA+ ‘crippled’ blood cell Noxious agent

Fig 2. Somatic mutation and Darwinian selection of mutant somatic cells in paroxysmal nocturnal haemoglobinuria (PNH). This cartoon pre-
dicted that a PIGA mutation, by preventing formation of glycosyl-phosphatidyl-inositol (GPI) and GPI-linked proteins, probably pre-existing,
provided a clone invulnerable to GPI-specific auto-immune attack. The 3 panels on the right depict possible outcomes. Top: PNH, with ongoing
auto-immune process; centre: PNH after demise of normal haematopoietic stem cells (HSCs); bottom: PNH with survival of some normal HSCs,
a situation that explains rare cases of spontaneous recovery from PNH. Modified from Luzzatto, L., Bessler M. and Rotoli B. Somatic mutations
in paroxysmal nocturnal hemoglobinuria: a blessing in disguise? Cell 88(1): 1-4. © 1997 John Wiley & Sons, Inc.

potentially accounting for active transcription of IFNG other presenting molecule. In this case, the T cells involved
(Solomou et al, 2006), as well as an increase in Th17 cells at would probably be a very small number amongst all T cells:
diagnosis: again these decline in good responders to IST they cannot be identified by analysis of conventional T cell
(de Latour et al, 2010). The number of type-1 cytokine subsets, but only by ad hoc methodologies. The first such
producing cells is inversely correlated with the number of methodology, spectratyping analysis, measures the size of the
regulatory T (Treg) cells, which tend to be decreased at diag- TCR-b chain complementarity determining region 3 (CDR3)
nosis and are eventually restored after effective IST (Solomou of each of the 24 b chain families. By this analysis a skewed
et al, 2007). A broad imbalance between increased Th1, Th2 size distribution – suggesting the presence of sizable T cell
and Th17 T cells versus decreased Tregs seem characteristic clones (clonotypes) – was first observed in a group of
of patients with AA (Kordasti et al, 2012). A detailed analysis patients with PNH (Karadimitris et al, 2000), and then in a
of different subsets of T cells has revealed subtle changes larger series of patients with AA (Risitano et al, 2004), as
(Kordasti et al, 2016) in sub-populations of Treg cells, well as in other studies (Zeng et al, 1999, 2001). Subse-
regarded as the master regulators of the T cell network, quently, sequence analysis of the TCR-b chain confirmed the
adding further support to the notion that AA is a existence of recurring clonotypes in patients with PNH (Gar-
cell-mediated auto-immune disease. giulo et al, 2007) and in children with hepatitis-associated
severe AA (Krell et al, 2013). Both AA and PNH can be asso-
ciated with LGL (Karadimitris et al, 2001; Risitano et al,
Evidence for specific (clonotypic) T cell populations and
2005), a disease that consists of an expanded NKT or NKT-
possible molecular targets
like cell clone.
If an auto-immune attack recognizes a specific antigenic Recurring T cell clonotypes may recognize specific mole-
molecule, the T cells involved must have the appropriate cules on HSCs. If a putative antigen were HLA-restricted, the
T-cell receptor (TCR) for recognition of that molecule within TCR structure recognizing it would be different, depending
the patient’s major histocompatibility complex (MHC), or on the HLA genotype of the patient, possibly explaining why

ª 2018 British Society for Haematology and John Wiley & Sons Ltd 763
British Journal of Haematology, 2018, 182, 758–776
Review

(A) Normal haematopoiesis (B) Direct killing of HSCs

XXX Direct killing

Self- Self-
XXX
HSCs HSCs
renewal renewal

HPCs HPCs

Granulocytes, platelets, RBCs Pancytopenia


(C) Functional inhibition of HSCs (D) Depletion consequent to loss of asymmetric division

X
Inhibition of
proliferation Inhibition of
asymmetric division
Self- No Self-
HSCs HSCs
renewal renewal

HPCs HPCs

Pancytopenia Pancytopenia

Fig 3. Different modes of injury to haematopoietic stem cells (HSCs) and haematopoietic progenitor cells (HPCs). (A) Normal haematopoiesis:
HSCs undergo asymmetric division, generating HPC which eventually produce mature blood cells [granulocytes, platelets and red blood cells
(RBCs)], while the HSC pool is preserved. (B) Direct killing. HSCs may be killed by a noxious agent (e.g. cytotoxic T cells): the HSC pool is
gradually depleted, with consequent impaired production of HPCs and of mature blood cells. (C) Functional inhibition. A noxious agent (e.g.,
interferon-c) may inhibit the proliferation of HSCs: the consequences will be as in (B). D. Loss of asymmetric division. HSCs (perhaps as a result
of an epigenetic change) may undergo symmetric rather than asymmetric division: if both daughter cells differentiate, the HSC pool will be
depleted, finally resulting in pancytopenia.

the same clonotype is not found in all AA patients (Risitano these findings support the escape model outlined above,
et al, 2004). Alternatively, the antigen may not be HLA- because GPI-positive HSCs (the majority) would suffer dam-
restricted: e.g., if it were a glycolipid it would be CD1d- age, and only PIGA mutant GPI-negative HSCs would be
restricted instead. This hypothesis has been tested in a subset spared (positive Darwinian selection: Table III, Fig 4). A pos-
of AA patients who have PNH, by using CD1d dimer tech- sible objection to this mechanism is that, because GPI is
nology and human GPI, obtained ad hoc by organic synthesis ubiquitous, GPI-specific T cells might damage cells other
(Richichi et al, 2011), as the putative antigen. By this than HSCs: however, only cells expressing CD1d would be
approach, GPI-specific T cells have been detected in the susceptible to such damage and, in addition, GPI-specific T
peripheral blood of all PNH patients: the numbers were very cells may be more abundant in the bone marrow than else-
small (usually less than 1% of all CD8 + cells), but signifi- where. Of course, GPI may not be the target in all cases: the
cantly above the control background (Gargiulo et al, 2013). escape model is versatile (Risitano, 2017; Luzzatto & Notaro,
This finding points to a specific molecule, GPI, as the target 2018) and if, as a result of mutation in a gene other than
of the auto-immune attack in the pathogenesis of those AA PIGA, a different target molecule were missing from a HSC,
patients who have PNH. Although direct damage to GPI(+) its progeny might be able to escape damage. This seems to
HSC by GPI-specific T cells has not yet been demonstrated, be the case in a group of patients with AA who are

764 ª 2018 British Society for Haematology and John Wiley & Sons Ltd
British Journal of Haematology, 2018, 182, 758–776
Review

Table II. Genes frequently observed to have somatic mutations in residual haematopoiesis in patients with aplastic anaemia.

Map Major function of Most frequent Possible mechanism of somatic Disease caused by germ-line
Gene location protein encoded mutations mutant clone persistence/expansion mutations

BCOR Xp114 Transcriptional co-repressor Inactivating Uncertain* Oculo-facial-cardio-dental syndrome


via histone deacetylases
ASXL1 20q1121 Chromatin conformation Inactivating Uncertain† Boring-Opitz syndrome†
(polycomb family)
DNMT3A 2p233 De novo DNA methylation Missense Uncertain* Tatton-Brown-Rahman syndrome‡
PIGA Xp222 Biosynthesis of GPI anchor Inactivating Escape from GPI-targeted Multiple congenital abnormalities-
auto-immune attack hypotonia-seizures
HLA locus 6p2131 Antigen presentation Inactivating Escape from auto-immune attack None

GPI, glycosyl-phosphatidyl-inositol.
*Could be resistance to apoptosis, increased proliferation, other.
†Multiple severe congenital malformations thought to be related to HOX genes dysfunction.
‡Tall stature and severe cognitive impairment.

heterozygous for the HLA-B allele 4002, and who have lost mutations are in only 5 genes: (Table II). Third, in each
its expression through a somatic point mutation or deletion individual patient the clones identified by mutant genes
of this allele (Zaimoku et al, 2017); and very recently autolo- make up a very variable proportion of total residual haema-
gous cytotoxic T cells that specifically recognize induced topoiesis: less than 10% in some cases, but up to 90% in
pluripotent stem cell-derived B4002+ CD34+ cells have been others.
obtained from these patients (Espinoza et al, 2018). Patients Three of these genes (Table II) are concerned with chro-
who have B4002 are relatively few and, therefore, their pre- matin structure and/or transcriptional regulation. BCOR has
sumed escape mechanism cannot account for what happens been so-called because it is a co-repressor of BCL6 (known
in the majority of patients with AA; but a similar mechanism to be involved in B cell development and lymphomagenesis).
of selection may take place with respect to other HLA alleles In AA, BCOR, an X-linked gene, has mostly inactivating
(Babushok et al, 2017a). At the same time, GPI-specific mutations: if the only copy of BCOR is inactivated, BCL6
CD1d-restricted T cells have been found at diagnosis in 75% will be less repressed. Loss-of-function mutations of BCOR
of Japanese AA patients, most of whom had GPI-negative are associated with enhanced cell proliferation and myeloid
blood cells above the noise level, but did not have PNH differentiation (Cao et al, 2016). ASXL1 belongs to the poly-
(Gargiulo et al, 2017). comb gene family: it too has inactivating mutations causing
loss of polycomb repressive complex 2 (PRC2)-dependent
histone H3 methylation (Abdel-Wahab et al, 2012), presum-
Mutant clones in AA
ably through haploinsufficieny. The ASXL1 protein forms,
Amongst AA patients it is clear that the subset of PNH with BAP1 protein, a complex required for the deubiquitina-
patients has a clonal disorder (Oni et al, 1970) that is conse- tion of mono-ubiquitinated histone 2A (Balasubramani et al,
quent upon a specific somatic mutation in the PIGA gene; 2015), whereby gain of function mutations found in AA will
and about one-half of all patients with AA also have a PIGA influence chromatin structure and transcription. DNMT3A
mutation, even though a mutant clone has not expanded (as encodes one subunit of an enzyme responsible for de novo
it has in PNH). Cytogenetic abnormalities (Appelbaum et al, methylation of DNA. Here one finds mostly missense muta-
1987; Socie, 1996) and skewed X chromosome inactivation tions, with a recurrent mutation at codon 882: this has been
patterns (van Kamp et al, 1991) in AA have been known for shown to act as dominant-negative by interfering with the
a long time; but only over the last few years has massive par- dimerization of the DNMT3A protein (Russler-Germain
allel DNA sequencing technology made the detection of et al, 2014), but other mutations may exert their effect
somatic mutations possible, even if present only in a minor- through haploinsufficiency (Cole et al, 2017). Each one of
ity of cells. Patients with AA have been tested for mutations these genes must have an important role in development:
in a set of candidate genes (Lane et al, 2013; Heuser et al, indeed, germ-line mutations cause multiple complex devel-
2014; Kulasekararaj et al, 2014), or by full exome sequencing opmental abnormalities (Table II).
(Babushok et al, 2015; Yoshizato et al, 2015). Reviews of this
work (Ogawa, 2016; Cooper & Young, 2017; Stanley et al,
Clonal haematopoiesis versus haematopoiesis
2017) have raised several points of interest. First, about one-
with clones
third of AA patients have mutant clones, either single or
multiple. Second, although mutations have been found in In the subset of AA patients who have PNH, haematopoiesis
some 30 different genes, the large majority of recurrent is supported effectively by a PIGA mutant clone: therefore,

ª 2018 British Society for Haematology and John Wiley & Sons Ltd 765
British Journal of Haematology, 2018, 182, 758–776
Review

Table III. Population genetics of organisms and of somatic cells

In a population of organisms In a population of somatic cells

Event/feature Consequences Examples Consequences Examples

Mutation
Any Creates a mutant Many Creates a mutant Many
individual/family cell/clone
Lethal No offspring Many No clone Many
Neutral No visible effect Many No visible effect Many
Genetic drift
neutral or not Frequency of a certain Unusually high A mutant clone can Mutant clones in AA
mutant gene increases frequency of more easily expand
when founder population inherited if original normal
small (bottleneck) polycythaemia cells scarce
due to germ-line
VHL mutations*
Selection†
With absolute growth Mutant progeny will Fixation of Clone will expand/ Leukaemogenic mutations
advantage gradually take over species-specific become dominant
genes
With conditional growth Mutant progeny will HbS gene (HBB Clone will expand in a PIGA mutant clone in the
advantage increase in a certain E7V) in certain environment presence of GPI-specific
environment malaria-endemic T cells (PNH)
area
Convergent evolution
Involving different genes Increase in frequency of Several genes related Clones with mutations in Clones with JAK2, CALR,
mutant alleles to melanogenesis in different genes yielding MPL mutations in MPNs
tropical areas similar disease
Involving the same gene Increase in frequency of Many different G6PD Clones with different Clones with different PIGA
mutant alleles mutant alleles mutations in the same mutations may co-exist in PNH
underlying G6PD gene yield similar disease
deficiency in may concurrently expand
malaria-endemic
areas

AA, aplastic anaemia; GPI, glycosyl-phosphatidyl-inositol; PNH, paroxysmal nocturnal haemoglobinuria.


*One VHL mutation (Arg200Trp mutation (C598T) has a high prevalence in the Chuvash region of Russia (Sergeyeva et al, 1997); the same VHL
mutation has a high prevalence on the island of Ischia (Italy), resulting in typical Chuvash polycythaemia (Perrotta et al, 2006).
†Unlike with genetic drift, population size is not influential.

clonal haematopoiesis describes this situation well. In other whether each generation is a representative sample of the
AA patients, haematopoiesis remains inadequate in spite of previous generation (i.e., genetic drift, still a stochastic phe-
the presence of mutant clones. The phrase ‘clonal disease’ nomenon), and on whether the environment is selective for
might convey the impression that the disease results from or against the mutant. In this light, the clonal composition
mutant clone(s); in fact, they are prominent as a conse- of an aplastic marrow probably results from a mix of drift
quence of the disease, the pathogenic cause of which consists and selection (see also Box 2, Fig 4). Drift is favoured by the
instead, as abundantly outlined above, in the demise of nor- overall paucity of cells, whereby a pre-existing mutant clone
mal HSCs. The clones that we see are reminiscent of residual has a greater chance to expand than the same mutant clone
vegetation in a de-populated landscape. would have in a non-aplastic marrow. At the same time, by
In order to outline the pathogenic significance of these the very fact that these clones are present in AA, we must
clones, currently a subject of lively debate (Cooper & Young, postulate they have preferentially survived at the time of
2017), we resort to the principles of populations and evolu- onset of the disease: i.e., they have been selected for. It does
tionary genetics (see Box 1 for definitions) – classically devel- not seem likely that selection is mediated by escape from a
oped in studying populations of organisms – to the extent target-specific attack (as is the case for PIGA mutant cells):
that they can apply to population of somatic cells (see rather, cells with mutations of ASXL1, BCOR, DNMT3A or
Table III). The engine of evolution consists of mutations: other genes may be overall more resistant to attack, be it T
these are stochastic events. The fate of a mutant depends on cell-mediated or mediated by inflammatory cytokines.

766 ª 2018 British Society for Haematology and John Wiley & Sons Ltd
British Journal of Haematology, 2018, 182, 758–776
Review

(A)

Time

Normal (polyclonal) set of HSC providing normal haematopoiesis

(B) Inflammation Persistant


and myelotoxic inflammatory
cytokines milieu

IFNG
IFNG

TGF-β TGF-β

TNF-α
TNF-α

Time

Normal HSC depletion Overt bone marrow failure: AA


(polyclonal) → oligoclonal
set of HSC haematopoiesis

(C)
Protracted GPI-targeted autoimmune
T-cell attack

±IST

PIGA
mutation

Time
Normal Loss of non-mutant HSCs and immune Rescue of haematopoiesis
(polyclonal) selection for GPI-deficient mutant HSC by expansion - clonal dominance -
set of HSC of PIGA mutant HSC: PNH

Fig 4. Dynamics of haematopoiesis in different situations. (A) Normal steady haematopoiesis in a normal bone marrow. (B) AA caused by a persis-
tent inflammatory process. (C) AA with PIGA mutants. In patients with PIGA mutant HSC and GPI-specific T cells AA converts to PNH: a PIGA
mutation seems to be the only case where haematopoiesis can be fully restored in a patient with AA. Given that some HSCs without PIGA muta-
tion may still be present, this diagram also accounts for spontaneous recovery from PNH, as previously suggested (Hillmen et al, 1995) and
recently confirmed (Babushok et al, 2017b). (D) AA with HLA-deficient mutant clones. In patients with loss of an HLA allele targeted by an auto-
immune process the HLA-deficient mutant clone can support haematopoiesis until IST helps recovery. (E) AA with other mutant clones: although
for clarity these are shown only in this panel, they may coexist also in the situations depicted in panels B–D. Note that mutant clones, which can
arise in normal people (as in panel E), are not the cause of AA. The emergence of mutant clones is favoured by the overall paucity of cells in the
bone marrow of AA patients (genetic drift: see Table III), it implies that the mutations most frequently seen must have an intrinsic slight survival
or growth advantage. The mutations of BCOR, ASXL1, DNMT3A (as well as others) are not leukaemogenic per se, because they are commonly
seen in the absence of leukaemia: however, they are probably in some cases the first step in a sequence of mutations that can cause leukaemia.
For additional explanations see Box 2. AA, aplastic anaemia; HLA, human leucocyte antigen; HSCs, haematopoietic stem cells; IFNG, interferon-c;
IST, immuno-suppressive treatment; PNH, paroxysmal nocturnal haemoglobinuria; TGF-b, transforming growth factor b; TNF-a, tumour necrosis
factor a

ª 2018 British Society for Haematology and John Wiley & Sons Ltd 767
British Journal of Haematology, 2018, 182, 758–776
Review

(D)
Protracted HLA-targeted autoimmune
T-cell attack

Successful
IST

6p LOH
(or B*4002)
mutation

Normal Loss of non-mutant HSCs and immune Restoration of haematopoiesis


(polyclonal) privilege of HLA-deficient mutant HSC without clonal dominance of
set of HSC HLA-deficient mutant HSC

(E) Other situations with mutant clones

DNMT3A ASXL1
mutation BCOR mutation
Clonal haematopoiesis
mutation
of indetermined potential
Slightly increased
HSC ‘fitness’?

Autoimmune T-
cell attack and/or Aplastic anaemia
with mutant clone(s)
inflammation

Additional
mutations
Myeloid malignancy

T cells Cytokines

Normal
(polyclonal)
set of HSC

Fig 4. Continued.

mutated in more than one patient. This is a compelling


AA and PNH are not cancer
confirmation, in a totally unbiased analysis, of the unique
The question of how PNH and AA relate to leukaemia has PNH-genic role of inactivating mutations of PIGA. When
been recurrent in the literature since the landmark editorial serial samples were available, it emerged that when the first
written by William Dameshek half a century ago, as a com- mutation is in PIGA, it is sufficient to cause clonal expansion,
mentary to three case reports on patients with PNH who whereby the superimposition of further mutations does not
developed acute myeloid leukaemia (AML) (Dameshek, affect the process much. On the other hand, when the PIGA
1967). Although more similar case reports have appeared mutation takes place in a cell belonging to a clone that has
subsequently, it is clear that they are exceptions rather than already a mutation in another gene, it is the clone that has
the rule: the majority of patients with PNH have not devel- both mutations that expands much more than the clone that
oped AML. This is particularly significant because PNH is a has the non-PIGA mutation alone. In a separate study (Yoshi-
very long-term disease: we and others have followed for dec- zato et al, 2015), again a PIGA mutation was found in every
ades individual patients who are still alive. The proportion of case. Thus, the two previously reported patients with an
patients with PNH who develop AML is estimated to be less HMGA2 mutation in addition to a PIGA mutation (Inoue
than 2% (de Latour et al, 2008); it is rather higher, perhaps et al, 2006) remain exceptional: in almost all PNH patients a
around 15% (Socie et al, 2000; Scheinberg et al, 2011), for PIGA mutation, apart from being responsible for GPI-negative
patients with AA who do not have PNH. haematopoiesis, is also the key to clonal expansion.
These clinical facts have been corroborated by full exome Unlike other clones in AA, which may survive and expand
sequencing of blood cells from patients with PNH (Shen et al, by a combination of drift and selection, a PIGA mutant clone
2014; Asad et al, 2017). The most consistent finding was that is able to expand by a specific selective mechanism (Luzzatto
PIGA is always mutated. Additional somatic mutations were et al, 1997; Young & Maciejewski, 2000). It is unfortunate that
found in 1 to 6 other genes, but only two of the genes were an editorial (Lee & Abdel-Wahab, 2014) accompanying the

768 ª 2018 British Society for Haematology and John Wiley & Sons Ltd
British Journal of Haematology, 2018, 182, 758–776
Review

Box 1. Clones and somatic mutations

1 A clone is the progeny of a single cell. Clonal population is used to designate a part of a clone: for instance, the GPI-
negative erythrocytes in a PNH patient are a clonal population (the entire PNH clone also includes other GPI-negative
blood cells and their precursors). Sometimes clonal population is used as synonym of clone.
2 A clone is of course not in itself pathological: for instance, the epithelial cells of a single intestinal crypt are a clonal pop-
ulation arising from a stem cell located at the base of the crypt.
3 A mutant clone is the progeny of a cell in which a mutation has taken place: it may or may not have a recognizable
phenotype, depending on the consequences of the mutation. Again, a mutant clone is not necessarily pathological: a
freckle or a naevus is a clone of cells with different pigmentation from surrounding skin.
4 Given that immunoglobulin and T-cell receptor rearrangements are part of the physiological development of the immune
system, there are myriads of normal B cell clones and T cell clones in our body.
5 A sub-clone is again the progeny of a single cell (see 1 above). This term is often used when a second mutation takes
place in one cell of a mutant clone.
6 Clonal haematopoiesis has been known for a long time in patients with leukaemia or PNH. Recently this term has been
used to indicate that, in contrast to normal haematopoiesis to which many stem cells contribute, in AA few clones aris-
ing from mutant HSCs may account for the production of some or even most most mature blood cells.
7 Clonal expansion indicates growth of a clone. Given that growth in the haematopoietic tissue is a normal process, the
term clonal expansion is appropriate only when the clone is growing at a higher rate than neighbouring non-mutant
cells. In AA, clonal expansion may be favoured by the vacuum created through the demise of non-mutant HSCs, but as
marrow aplasia is not corrected, the expansion is in relative, not in absolute terms.
8 The term clonal dominance is used when a clone or a sub-clone has expanded in size to the extent of over-shadowing
other cell populations in a particular tissue: for instance, a leukaemic clone will dominate or even overwhelm normal
haematopoiesis. In other cases, like in AA, dominance may be only relative (see clonal expansion above).
9 Clonal evolution* means that one or more sub-clones can develop from a clone; it is not synonymous with leukaemoge-
nesis, because many clones with two or even more mutations may not be leukaemic. On the other hand, it is true that a
mutation causing blast crisis constitutes the clonal evolution of chronic myeloid leukaemia.
10 A clonal disease is any disease caused by a mutant clone: it is not necessarily malignant (e.g., PNH). However, it is true that
a clonal disease can evolve to malignancy (e.g., MGUS to MM). Although there is clonal haematopoiesis in AA, it does not
seem appropriate or useful to call it a clonal disease, because the clones are not the cause of the bone marrow failure charac-
teristic of AA: rather, they are a consequence: in fact, not all AA patients have clones, indicating that they are not a must.

* The term clonal architecture is sometimes used to describe, in a particular condition, the set of clones, whether indepen-
dently arisen or evolved from one another, that are present, as well as their sizes. We think this phrase is not ideal and may
be misleading, because architecture conveys the notion of a deliberate plan: instead, mutations are random events and the
fate of mutant clones depends on genetic drift and selection (see Table III). If at all, clonal composition or clonal landscape
may be better.

paper by Shen et al (2014) gave the impression that PNH is pre-leukaemic clones (Luzzatto & Pandolfi, 2015). In AA
‘strikingly similar to other myeloid neoplasms’. Not only this patients, sizable mutant clones of BCOR, ASXL1 and
is not the case, but we can also visualize why. In Table II, PIGA DNMT3A may persist even after IST: hence, following addi-
stands out because it has a fundamentally different function tional mutations, some 15% of AA patients eventually
from some of the others listed as examples of genes mutated in develop AML. However, in PNH, most haematopoiesis has
AA. Mutant clones of BCOR, ASXL1 and DNMT3A, with their been taken over by the PIGA mutant clone: indeed, a single
altered chromatin arrangement, may evolve to malignant PIGA mutant HSC may, on its own, sustain haematopoiesis
sub-clones through additional mutations; in contrast, there is even for decades (Nishimura Ji et al, 2002). Independent
no biological basis for regarding a PIGA mutant clone as (non-PIGA mutant) clones, if any, or PIGA-mutant sub-
pre-leukaemic. clones that have other mutations, if any, are on average
We can probably also explain why the development of smaller: hence the much lower probability of AML.
AML is so much rarer in PNH patients compared to other
AA patients. Given that mutations are stochastic events,
AA and myelodysplastic syndrome
leukaemogenesis is not a clockwise process (see also Box 2),
and it may not happen; but it is a stepwise process, and the Myelodysplastic syndrome (MDS) is not within our remit; but
probability of further mutations is proportional to the size of it has been noticeable for a long time that AA and ‘hypoplastic

ª 2018 British Society for Haematology and John Wiley & Sons Ltd 769
British Journal of Haematology, 2018, 182, 758–776
Review

et al, 2017). In addition, there is wide overlap in marrow mor-


Box 2. Clones and their fate in aplastic anaemia
phology between MDS and PNH (Araten et al, 2001).
1 Random mutations probably occur just as frequently That there is BMF in MDS is not a new concept (Galton,
in AA patients as in normal people (Rondelli et al, 1989): indeed, when patients with MDS were treated as
2013). though they had AML, the outcome was grim (Tricot &
2 Mutant clones found in AA patients almost certainly Boogaerts, 1986; Fenaux et al, 1988; Loffler et al, 1992; Ber-
pre-existed AA, as they can be found also in normal nasconi et al, 1998), because normal haematopoiesis failed to
persons (Genovese et al, 2014; Jaiswal et al, 2014; Xie regenerate, suggesting that there were few, if any, viable nor-
et al, 2014). mal HSCs. It is also known that, in the age distribution of
3 Survival advantage may be absolute (as for BCR- AA there are two peaks: one in young people, the other
ABL1 mutant cells in chronic myeloid leukaemia), or around 65 years old (Montane et al, 2008), within the typical
it may be relative to a particular situation, e.g. an age group of MDS. The differential diagnosis between AA
inflammatory environment or an auto-immune and hypoplastic MDS is hard, because they are probably the
attack. same thing (Barrett et al, 2000). If the bone marrow is exam-
4 The epigenetic profile of mutant cells may be critical ined at an early stage the diagnosis will be AA; if it is exam-
to survival, e.g. by making cells less prone to apopto- ined when some of the clones arising in AA have already
sis: this may be why several of the most common established themselves, the diagnosis will be MDS. The signif-
mutations in clones found in AA (see Table II) are in icant beneficial results of IST in a subset of patients with
genes related to chromatin structure. MDS (Saunthararajah et al, 2002, 2003; Sloand & Barrett,
5 Growth advantage does not necessarily follow survival 2010) supports this notion.
advantage: in AA, when IST is successful and normal At the same time, in elderly people there are at least two
haematopoiesis is restored, mutant clones may retain hypothetical but plausible mechanisms other than auto-
the percentage of space they had acquired, but expand immune attack that could cause BMF. One of them is telom-
very little further or not at all (Yoshizato et al, 2015). ere attrition (Calado & Young, 2008; Alter et al, 2015): the
6 The phrase ‘clonal haematopoiesis of indeterminate distribution of telomere length at birth is wide, but all of us
potential (CHIP)’ was coined (Steensma et al, 2015) lose telomere length as we age. In some people, by 70 years
following the finding that in normal subjects, particu- of age, the telomeres may be sufficiently worn out as to com-
larly the elderly (Genovese et al, 2014; Jaiswal et al, promise ‘normal’ HSCs, whereas some mutant clones might
2014; Xie et al, 2014), somatic mutations may be pre- express telomerase activity or alternative lengthening of
sent that are identical to some found in MDS and telomeres (ALT) mechanisms to enable their survival.
leukaemia. Another possibility is loss of stemness: at every cell division
7 In most cases, leukaemogenesis is a multi-step pro- there is a risk that both daughter cells will differentiate,
cess, and a mutation of ASXL1 (or BCOR or entailing the loss of one HSC (see Fig 3). A mutant clone
DNMT3A) may be one step. Thus, if n steps are might provide abnormal stem cells that are more capable of
required for a particular type of leukaemia to develop, retaining stemness. These hypothetical mechanisms might
a cell with one of these mutations will now only need explain why the mutational landscape in MDS is wider than
n-1 steps. in AA: there may be a greater variety of selective mechanisms
8 Multi-step versus clockwise. Since mutations are at play.
stochastic events, the n steps do not take place in a
clockwise manner: rather, each event happens by
Concluding remarks
chance. Therefore, an elderly person or a patient with
AA who has a very small ASXL1 mutant clone is at an Our understanding of inherited syndromes leading to bone
increased risk of leukaemia (compared to a person marrow failure has made great strides with the identification
who does not have that small clone); however, that of a discrete set of genes, the mutation of which underlie
person does not have leukaemia, and may not develop these syndromes (see Table I). Similarly, it is clear that heavy
leukaemia, because the next mutation(s) may not take radiation (which may be accidental) or high doses of cyto-
place within the person’s lifetime. toxic agents (such as those used for deliberate pre-transplant
haematopoietic ablation) cause acquired bone marrow aplasia
by eliminating HSCs.
In contrast, unravelling the pathogenesis of acquired ‘idio-
MDS’ overlap: molecular analysis has only reinforced the rela- pathic’ AA has proven harder, because it is a subtler process.
tionship, as clones with the very same somatic mutations may The beneficial and often curative effect of intensive IST
be found in patients who had been diagnosed with one or the remains, to date, the most compelling evidence that AA is, in
other condition. The nature and number of these somatic most cases, a disease of deranged immunity: but by now this
mutations are predictive of transformation to AML (Malcovati evidence is further corroborated by a multitude of studies

770 ª 2018 British Society for Haematology and John Wiley & Sons Ltd
British Journal of Haematology, 2018, 182, 758–776
Review

demonstrating in this condition consistent changes in the no PIGA-mutant HSC, and therefore AA does not get cured
levels of certain cytokines, the relative proportions of T cell by PNH. In some cases, an escape process similar to that of
subsets and their function, as assessed by analysis of their tran- PNH may apply, as in patients with loss or mutation of
scriptome. It is conceivable that, in some cases, such changes HLA-B4002, where the target of an auto-immune attack
might amount to an inflammatory state sufficiently powerful might be a peptide from that allele: this would be MHC-
to damage HSCs as innocent bystanders. However, the efficacy restricted rather than CD1d-restricted. In these cases, the cell
of an agent targeting T cells specifically, such as alemtuzumab population with the mutant HLA-B4002 allele tends to
(Risitano et al, 2010; Scheinberg et al, 2012), makes one won- decrease upon IST, meaning that, unlike in PNH, recovery is
der whether this may be true in the majority of cases. not mediated by expansion of that mutant clone, but by
PNH, being even more rare than AA, has seemed for a residual non-mutant HSCs.
long time to be an accessory to the crime. There is now It is always tempting to conclude by considering therapeu-
overwhelming evidence that, in fact, PNH is a subset of AA; tic implications, and we will not resist this temptation. IST
and because of the unique molecular lesion – a PIGA muta- (Scheinberg et al, 2011), recently supplemented by the use of
tion – these patients have provided us with a special insight eltrombopag (Townsley et al, 2017), as well as allogeneic
into pathogenesis. In PNH, normal haematopoiesis is largely BMT (Marsh et al, 2011) have a high rate of success in AA:
and sometimes completely replaced by clonal (phenotypically both procedures subjugate or annihilate immune cells, and
GPI-negative) haematopoiesis, driven by a single PIGA- the latter provides healthy HSCs in addition. However, these
mutant stem cell (Nishimura Ji et al, 2002): meaning that it therapeutic strategies seem rather blunt instruments: a more
must have been spared by whatever has injured the other targeted intervention would seem preferable. If and when the
HSCs. Given that there is no difference between PIGA- pathogenic agent is a specific population of T cells, this pop-
mutant and non-mutant HSC in all the essential biological ulation could be isolated, and a specific antiserum produced:
characteristics of HSCs, including stem cell renewal and for instance, against GPI-specific T cells or even against their
potential for tri-lineage differentiation, it is not easy to imag- TCR. Such an antiserum can be expected to have far fewer
ine a damage effector mechanism that is sufficiently sophisti- side effects than antithymocyte globulin or BMT, and would
cated to discriminate between the two, if not by immune introduce a new type of precision medicine.
recognition. Thus, at least for PNH, at the time the ‘escape’
model was formulated nearly three decades ago, the notion
Acknowledgements
of a non-specific inflammatory process had to be rejected:
the target of the immune attack had to be specific, although We are very grateful to Dr Rosario Notaro and Dr Regis Pef-
at that time we had no clue as to whether it was GPI or any fault de Latour for reviewing the manuscript and for
of the GPI-linked proteins. Now we know that PNH has thoughtful suggestions. This paper is dedicated to the mem-
GPI-specific T cells that recognize GPI in the CD1d groove ory of Bruno ROTOLI: a great haematologist, an invaluable
(Gargiulo et al, 2007), whereby it appears that GPI is the real colleague and collaborator of some 30 years to one of us, an
target. With the expansion of the GPI-negative clone, AA is exceptionally inspiring mentor to one of us, and a dear
cured and the diagnosis becomes PNH: the escape model is friend for us both.
fully vindicated (Fig 2).
But what about the other patients with AA? In some cases,
Author contribution
the demise of HSCs may be caused by a non-targeted inflam-
matory process. But in a substantial fraction of cases the tar- LL and AMR have written the paper together and agreed on
get of auto-reactive T cells may be still GPI, except there is the final text.

References Alvarado, L.J., Andreoni, A., Huntsman, H.D., present in normal individuals. Proceedings of the
Cheng, H., Knutson, J.R. & Larochelle, A. National Academy of Sciences of the United States
Abdel-Wahab, O., Adli, M., LaFave, L.M., Gao, J., (2017) Heterodimerization of TPO and IFNc of America, 96, 5209–5214.
Hricik, T., Shih, A.H., Pandey, S., Patel, J.P., impairs human hematopoietic stem/progenitor Araten, D.J., Swirsky, D., Karadimitris, A., Notaro,
Chung, Y.R., Koche, R., Perna, F., Zhao, X., cell signaling and survival in chronic inflamma- R., Nafa, K., Bessler, M., Thaler, H.T., Castro-
Taylor, J.E., Park, C.Y., Carroll, M., Melnick, A., tion. Blood, 130, 4. Malaspina, H., Childs, B.H., Boulad, F., Weiss,
Nimer, S.D., Jaffe, J.D., Aifantis, I., Bernstein, Appelbaum, F.R., Barrall, J., Storb, R., Ramberg, M., Anagnostopoulos, N., Kutlar, A., Savage,
B.E. & Levine, R.L. (2012) ASXL1 mutations R., Doney, K., Sale, G.E. & Thomas, E.D. (1987) D.G., Maziarz, R.T., Jhanwar, S. & Luzzatto, L.
promote myeloid transformation through loss of Clonal cytogenetic abnormalities in patients with (2001) Cytogenetic and morphological abnor-
PRC2-mediated gene repression. Cancer Cell, 22, otherwise typical aplastic anemia. Experimental malities in paroxysmal nocturnal haemoglobin-
180–193. Hematology, 15, 1134–1139. uria. British Journal of Haematology, 115, 360–
Alter, B.P., Giri, N., Savage, S.A. & Rosenberg, P.S. Araten, D.J., Nafa, K., Pakdeesuwan, K. & Luz- 368.
(2015) Telomere length in inherited bone mar- zatto, L. (1999) Clonal populations of Asad, M.F.B., Goyal, A., Awada, H., Aly, M.,
row failure syndromes. Haematologica, 100, 49– hematopoietic cells with paroxysmal nocturnal Hirsch, C., Clemente, M.J., Nazha, A., Carraway,
54. hemoglobinuria genotype and phenotype are H.E., Mukherjee, S., Nagata, Y. & Maciejewski,

ª 2018 British Society for Haematology and John Wiley & Sons Ltd 771
British Journal of Haematology, 2018, 182, 758–776
Review

J.P. (2017) Extent and clinical implications of Brummendorf, T.H., Maciejewski, J.P., Mak, J., from patients with aplastic anemia stimulate
subclonal diversity in paroxysmal nocturnal Young, N.S. & Lansdorp, P.M. (2001) Telomere monocytic cells to secrete TNF-alpha through
hemoglobinuria. Blood, 130, 779. length in leukocyte subpopulations of patients an ERK1/2-dependent pathway. International
Babushok, D.V., Olson, T.S. & Bessler, M. (2015) with aplastic anemia. Blood, 97, 895–900. Immunology, 21, 913–923.
Somatic mutations and clonal hematopoiesis in Calado, R.T. & Young, N.S. (2008) Telomere Espinoza, J.L., Elbadry, M.I., Chonabayashi, K.,
aplastic anemia. New England Journal of Medi- maintenance and human bone marrow failure. Yoshida, Y., Katagiri, T., Harada, K., Nakagawa,
cine, 373, 1673. Blood, 111, 4446–4455. N., Zaimoku, Y., Imi, T., Hassanein, H.A., Khal-
Babushok, D.V., Duke, J.L., Xie, H.M., Stanley, N., Calado, R.T. & Young, N.S. (2009) Telomere dis- ifa, A.N.A., Takenaka, K., Akashi, K., Hamana,
Atienza, J., Perdigones, N., Nicholas, P., Ferriola, eases. New England Journal of Medicine, 361, H., Kishi, H., Akatsuka, Y. & Nakao, S. (2018)
D., Li, Y., Huang, H., Ye, W., Morrissette, J.J.D., 2353–2365. Hematopoiesis by iPSC-derived hematopoietic
Kearns, J., Porter, D.L., Podsakoff, G.M., Eisen- Cao, Q., Gearhart, M.D., Gery, S., Shojaee, S., stem cells of aplastic anemia that escape cyto-
lohr, L.C., Biegel, J.A., Chou, S.T., Monos, D.S., Yang, H., Sun, H., Lin, D.C., Bai, J.W., Mead, toxic T-cell attack. Blood Advances, 2, 390–400.
Bessler, M. & Olson, T.S. (2017a) Somatic HLA M., Zhao, Z., Chen, Q., Chien, W.W., Alkan, S., Fenaux, P., Lai, J.L., Jouet, J.P., Pollet, J.P. & Bau-
mutations expose the role of class I-mediated Alpermann, T., Haferlach, T., Muschen, M., ters, F. (1988) Aggressive chemotherapy in adult
autoimmunity in aplastic anemia and its clonal Bardwell, V.J. & Koeffler, H.P. (2016) BCOR primary myelodysplastic syndromes. A report on
complications. Blood Advances, 1, 1900–1910. regulates myeloid cell proliferation and differen- 29 cases. Blut, 57, 297–302.
Babushok, D.V., Stanley, N., Xie, H.M., Huang, H., tiation. Leukemia, 30, 1155–1165. Feng, X., Chuhjo, T., Sugimori, C., Kotani, T., Lu,
Bagg, A., Olson, T.S. & Bessler, M. (2017b) Clonal Champlin, R. (1989) Bone marrow aplasia due to X., Takami, A., Takamatsu, H., Yamazaki, H. &
replacement underlies spontaneous remission in radiation accidents: pathophysiology, assessment Nakao, S. (2004) Diazepam-binding inhibitor-
paroxysmal nocturnal haemoglobinuria. British and treatment. Baillieres Clinical Haematology, 2, related protein 1: a candidate autoantigen in
Journal of Haematology, 176, 487–490. 69–82. acquired aplastic anemia patients harboring a
Bacigalupo, A., Valle, M., Podesta, M., Pitto, A., Champlin, R., Ho, W. & Gale, R.P. (1983) Antithy- minor population of paroxysmal nocturnal
Zocchi, E., De Flora, A., Pozzi, S., Luchetti, S., mocyte globulin treatment in patients with aplas- hemoglobinuria-type cells. Blood, 104, 2425–
Frassoni, F., Van Lint, M.T. & Piaggio, G. tic anemia: a prospective randomized trial. New 2431.
(2005) T-cell suppression mediated by mes- England Journal of Medicine, 308, 113–118. Ferrata, A. (ed.) (1934) Le Emopatie. Societa edi-
enchymal stem cells is deficient in patients with Cole, C.B., Russler-Germain, D.A., Ketkar, S., Ver- trice libraria, Milan.
severe aplastic anemia. Experimental Hematology, doni, A.M., Smith, A.M., Bangert, C.V., Helton, Fores, R., Alcocer, M., Diez-Martin, J.L. & Fernan-
33, 819–827. N.M., Guo, M., Klco, J.M., O’Laughlin, S., Fron- dez, M.N. (1995) Flow cytometric analysis of
Balasubramani, A., Larjo, A., Bassein, J.A., Chang, ick, C., Fulton, R., Chang, G.S., Petti, A.A., decay-accelerating factor (CD55) on neutrophils
X., Hastie, R.B., Togher, S.M., Lahdesmaki, H. Miller, C.A. & Ley, T.J. (2017) Haploinsuffi- from aplastic anaemia patients. British Journal of
& Rao, A. (2015) Cancer-associated ASXL1 ciency for DNA methyltransferase 3A predis- Haematology, 90, 728–730.
mutations may act as gain-of-function muta- poses hematopoietic cells to myeloid Frickhofen, N., Kaltwasser, J.P., Schrezenmeier, H.,
tions of the ASXL1-BAP1 complex. Nature Com- malignancies. J Clin Invest, 127, 3657–3674. Raghavachar, A., Vogt, H.G., Herrmann, F., Fre-
munications, 6, 7307. Cooper, J.N. & Young, N.S. (2017) Clonality in und, M., Meusers, P., Salama, A. & Heimpel, H.
Ball, S.E., Gibson, F.M., Rizzo, S., Tooze, J.A., context: hematopoietic clones in their marrow (1991) Treatment of aplastic anemia with anti-
Marsh, J.C. & Gordon-Smith, E.C. (1998) Pro- environment. Blood, 130, 2363–2372. lymphocyte globulin and methylprednisolone
gressive telomere shortening in aplastic anemia. Crane, G.M., Jeffery, E. & Morrison, S.J. (2017) with or without cyclosporine. The German
Blood, 91, 3582–3592. Adult haematopoietic stem cell niches. Nature Aplastic Anemia Study Group. New England
Barrett, J., Saunthararajah, Y. & Molldrem, J. Reviews Immunology, 17, 573–590. Journal of Medicine, 324, 1297–1304.
(2000) Myelodysplastic syndrome and aplastic Dacie, J.V. & Lewis, S.M. (1961) Paroxysmal noc- Frickhofen, N., Heimpel, H., Kaltwasser, J.P. &
anemia: distinct entities or diseases linked by a turnal haemoglobinuria: variation in clinical Schrezenmeier, H.; German Aplastic Anemia
common pathophysiology? Seminars in Hematol- severity and association with bone-marrow Study Group. (2003). “Antithymocyte globulin
ogy, 37, 15–29. hypoplasia. British Journal of Haematology, 7, with or without cyclosporin A: 11-year follow-
Ben-Bassat, I., Brok-Simoni, F. & Ramot, B. 442–457. up of a randomized trial comparing treatments
(1975) Complement-sensitive red cells in aplas- Dameshek, W. (1967) Riddle: what do aplastic of aplastic anemia.” Blood 101, 1236–1242.
tic anemia. Blood, 46, 357–361. anemia, paroxysmal nocturnal hemoglobinuria Galton, D.A. (1989) The myelodysplastic syn-
Bernasconi, C., Alessandrino, E.P., Bernasconi, P., (PNH) and “hypoplastic” leukemia have in drome. BMJ, 299, 582.
Bonfichi, M., Lazzarino, M., Canevari, A., Cas- common? Blood, 30, 251–254. Gargiulo, L., Lastraioli, S., Cerruti, G., Serra, M.,
telli, G., Brusamolino, E., Pagnucco, G. & Doney, K., Pepe, M., Storb, R., Bryant, E., Ana- Loiacono, F., Zupo, S., Luzzatto, L. & Notaro,
Castagnola, C. (1998) Randomized clinical study setti, C., Appelbaum, F.R., Buckner, C.D., San- R. (2007) Highly homologous T-cell receptor
comparing aggressive chemotherapy with or ders, J., Singer, J., Sullivan, K., Weiden, P. & beta sequences support a common target for
without G-CSF support for high-risk myelodys- Hansen, J.A. (1992) “Immunosuppressive ther- autoreactive T cells in most patients with parox-
plastic syndromes or secondary acute myeloid apy of aplastic anemia: results of a prospective, ysmal nocturnal hemoglobinuria. Blood, 109,
leukaemia evolving from MDS. British Journal of randomized trial of antithymocyte globulin 5036–5042.
Haematology, 102, 678–683. (ATG), methylprednisolone, and oxymetholone Gargiulo, L., Papaioannou, M., Sica, M., Talini, G.,
Bestach, Y., Sieza, Y., Attie, M., Riccheri, C., Verri, to ATG, very high-dose methylprednisolone, Chaidos, A., Richichi, B., Nikolaev, A.V., Nativi,
V., Bolesina, M., Bengio, R., Larripa, I. & Belli, and oxymetholone”. Blood, 79, 2566–2571. C., Layton, M., de la Fuente, J., Roberts, I., Luz-
C. (2015) Polymorphisms in TNF and IFNG are Dufour, C., Corcione, A., Svahn, J., Haupt, R., zatto, L., Notaro, R. & Karadimitris, A. (2013)
associated with clinical characteristics of aplastic Battilana, N. & Pistoia, V. (2001) Interferon Glycosylphosphatidylinositol-specific, CD1d-
anemia in Argentinean population. Leukaemia & gamma and tumour necrosis factor alpha are restricted T cells in paroxysmal nocturnal
Lymphoma, 56, 1793–1798. overexpressed in bone marrow T lymphocytes hemoglobinuria. Blood, 121, 2753–2761.
Brown, K.E., Tisdale, J., Barrett, A.J., Dunbar, C.E. from paediatric patients with aplastic anaemia. Gargiulo, L., Zaimoku, Y., Scappini, B., Mar-
& Young, N.S. (1997) Hepatitis-associated aplas- British Journal of Haematology, 115, 1023–1031. uyama, H., Ohumi, R., Luzzatto, L., Nakao, S. &
tic anemia. New England Journal of Medicine, Espinoza, J.L., Takamatsu, H., Lu, X., Qi, Z. & Notaro, R. (2017) Glycosylphosphatidylinositol-
336, 1059–1064. Nakao, S. (2009) Anti-moesin antibodies derived specific T cells, IFN-gamma-producing T cells,

772 ª 2018 British Society for Haematology and John Wiley & Sons Ltd
British Journal of Haematology, 2018, 182, 758–776
Review

and pathogenesis of idiopathic aplastic anemia. hematopoiesis in patients with acquired aplastic Young Hematologists. (2008). “Paroxysmal noc-
Blood, 129, 388–392. anemia. Blood, 78, 3209–3214. turnal hemoglobinuria: natural history of disease
Genovese, G., Kahler, A.K., Handsaker, R.E., Lind- Karadimitris, A., Manavalan, J.S., Thaler, H.T., subcategories.” Blood 112, 3099–3106.
berg, J., Rose, S.A., Bakhoum, S.F., Chambert, Notaro, R., Araten, D.J., Nafa, K., Roberts, I.A., de Latour, R.P., Visconte, V., Takaku, T., Wu, C.,
K., Mick, E., Neale, B.M., Fromer, M., Purcell, Weksler, M.E. & Luzzatto, L. (2000) Abnormal Erie, A.J., Sarcon, A.K., Desierto, M.J., Schein-
S.M., Svantesson, O., Landen, M., Hoglund, M., T-cell repertoire is consistent with immune pro- berg, P., Keyvanfar, K., Nunez, O., Chen, J. &
Lehmann, S., Gabriel, S.B., Moran, J.L., Lander, cess underlying the pathogenesis of paroxysmal Young, N.S. (2010) Th17 immune responses
E.S., Sullivan, P.F., Sklar, P., Gronberg, H., nocturnal hemoglobinuria. Blood, 96, 2613– contribute to the pathophysiology of aplastic
Hultman, C.M. & McCarroll, S.A. (2014) Clonal 2620. anemia. Blood, 116, 4175–4184.
hematopoiesis and blood-cancer risk inferred Karadimitris, A., Li, K., Notaro, R., Araten, D.J., Lee, S.C. & Abdel-Wahab, O. (2014) The muta-
from blood DNA sequence. New England Jour- Nafa, K., Thertulien, R., Ladanyi, M., Stevens, tional landscape of paroxysmal nocturnal
nal of Medicine, 371, 2477–2487. A.E., Rosenfeld, C.S., Roberts, I.A. & Luzzatto, hemoglobinuria revealed: new insights into clo-
Gluckman, E., Devergie, A., Poros, A. & Degoulet, L. (2001) Association of clonal T-cell large gran- nal dominance. The Journal of Clinical Investiga-
P. (1982) Results of immunosuppression in 170 ular lymphocyte disease and paroxysmal noctur- tion, 124, 4227–4230.
cases of severe aplastic anaemia. Report of the nal haemoglobinuria (PNH): further evidence Lewis, S.M. & Dacie, J.V. (1967) The aplastic
European Group of Bone Marrow Transplant for a pathogenetic link between T cells, aplastic anaemia–paroxysmal nocturnal haemoglobinuria
(EGBMT). British Journal of Haematology, 51, anaemia and PNH. British Journal of Haematol- syndrome. British Journal of Haematology, 13,
541–550. ogy, 115, 1010–1014. 236–251.
Heuser, M., Schlarmann, C., Dobbernack, V., Kordasti, S., Marsh, J., Al-Khan, S., Jiang, J., Lin, F.C., Karwan, M., Saleh, B., Hodge, D.L.,
Panagiota, V., Wiehlmann, L., Walter, C., Beier, Smith, A., Mohamedali, A., Abellan, P.P., Veen, Chan, T., Boelte, K.C., Keller, J.R. & Young,
F., Ziegler, P., Yun, H., Kade, S., Kirchner, A., C., Costantini, B., Kulasekararaj, A.G., Benson- H.A. (2014) IFN-gamma causes aplastic anemia
Huang, L., Koenecke, C., Eder, M., Brummen- Quarm, N., Seidl, T., Mian, S.A., Farzaneh, F. & by altering hematopoietic stem/progenitor cell
dorf, T.H., Dugas, M., Ganser, A. & Thol, F. Mufti, G.J. (2012) Functional characterization of composition and disrupting lineage differentia-
(2014) Genetic characterization of acquired CD4 + T cells in aplastic anemia. Blood, 119, tion. Blood, 124, 3699–3708.
aplastic anemia by targeted sequencing. Haema- 2033–2043. Loffler, H., Schmitz, N. & Gassmann, W. (1992)
tologica, 99, e165–e167. Kordasti, S., Costantini, B., Seidl, T., Perez Abel- Intensive chemotherapy and bone marrow trans-
Hillmen, P., Lewis, S.M., Bessler, M., Luzzatto, L. lan, P., Martinez Llordella, M., McLornan, D., plantation for myelodysplastic syndromes.
& Dacie, J.V. (1995) Natural history of paroxys- Diggins, K.E., Kulasekararaj, A., Benfatto, C., Hematology/Oncology Clinics of North America,
mal nocturnal hemoglobinuria. New England Feng, X., Smith, A., Mian, S.A., Melchiotti, R., 6, 619–631.
Journal of Medicine, 333, 1253–1258. de Rinaldis, E., Ellis, R., Petrov, N., Povoleri, Luzzatto, L. & Araten, D. (2006). Paroxysmal noctur-
Hirano, N., Butler, M.O., Von Bergwelt-Baildon, G.A., Chung, S.S., Thomas, N.S., Farzaneh, F., nal hemoglobinuria. In: Clinical Hematology. (ed.
M.S., Maecker, B., Schultze, J.L., O’Connor, Irish, J.M., Heck, S., Young, N.S., Marsh, J.C. & by N. S. Young, S. L. Gerson & K. A. High), pp.
K.C., Schur, P.H., Kojima, S., Guinan, E.C. & Mufti, G.J. (2016) Deep phenotyping of Tregs 326–339. Elsevier, Philadelphia, PA.
Nadler, L.M. (2003) Autoantibodies frequently identifies an immune signature for idiopathic Luzzatto, L. & Notaro, R. (2018). “The “escape”
detected in patients with aplastic anemia. Blood, aplastic anemia and predicts response to treat- model: a versatile mechanism for clonal expan-
102, 4567–4575. ment. Blood, 128, 1193–1205. sion.” British Journal of Haematology. https://
Hirano, N., Butler, M.O., Guinan, E.C., Nadler, Krebs, D.L. & Hilton, D.J. (2001) SOCS proteins: doi.org/doi: 10.1111/bjh.15111. [Epub ahead of
L.M. & Kojima, S. (2005) Presence of anti- negative regulators of cytokine signaling. Stem print].
kinectin and anti-PMS1 antibodies in Japanese Cells, 19, 378–387. Luzzatto, L. & Pandolfi, P.P. (2015) Causality and
aplastic anaemia patients. British Journal of Hae- Krell, P.F., Reuther, S., Fischer, U., Keller, T., chance in the development of cancer. New Eng-
matology, 128, 221–223. Weber, S., Gombert, M., Schuster, F.R., Asang, land Journal of Medicine, 373, 84–88.
Inoue, N., Izui-Sarumaru, T., Murakami, Y., C., Stepensky, P., Strahm, B., Meisel, R., Stoye, J. Luzzatto, L., Bessler, M. & Rotoli, B. (1997) Somatic
Endo, Y., Nishimura, J., Kurokawa, K., & Borkhardt, A. (2013) Next-generation-sequen- mutations in paroxysmal nocturnal hemoglobin-
Kuwayama, M., Shime, H., Machii, T., Kana- cing-spectratyping reveals public T-cell receptor uria: a blessing in disguise? Cell, 88, 1–4.
kura, Y., Meyers, G., Wittwer, C., Chen, Z., repertoires in pediatric very severe aplastic ane- Maciejewski, J., Selleri, C., Anderson, S. & Young,
Babcock, W., Frei-Lahr, D., Parker, C.J. & mia and identifies a beta chain CDR3 sequence N.S. (1995) Fas antigen expression on CD34 +
Kinoshita, T. (2006) Molecular basis of clonal associated with hepatitis-induced pathogenesis. human marrow cells is induced by interferon
expansion of hematopoiesis in 2 patients with Haematologica, 98, 1388–1396. gamma and tumor necrosis factor alpha and
paroxysmal nocturnal hemoglobinuria (PNH). Kulasekararaj, A.G., Jiang, J., Smith, A.E., Moha- potentiates cytokine-mediated hematopoietic
Blood, 108, 4232–4236. medali, A.M., Mian, S., Gandhi, S., Gaken, J., suppression in vitro. Blood, 85, 3183–3190.
Jaiswal, S., Fontanillas, P., Flannick, J., Manning, Czepulkowski, B., Marsh, J.C. & Mufti, G.J. Maciejewski, J.P., Selleri, C., Sato, T., Anderson, S.
A., Grauman, P.V., Mar, B.G., Lindsley, R.C., (2014) Somatic mutations identify a subgroup & Young, N.S. (1996) A severe and consistent
Mermel, C.H., Burtt, N., Chavez, A., Higgins, of aplastic anemia patients who progress to deficit in marrow and circulating primitive
J.M., Moltchanov, V., Kuo, F.C., Kluk, M.J., myelodysplastic syndrome. Blood, 124, 2698– hematopoietic cells (long-term culture-initiating
Henderson, B., Kinnunen, L., Koistinen, H.A., 2704. cells) in acquired aplastic anemia. Blood, 88,
Ladenvall, C., Getz, G., Correa, A., Banahan, Lane, A.A., Odejide, O., Kopp, N., Kim, S., Yoda, 1983–1991.
B.F., Gabriel, S., Kathiresan, S., Stringham, A., Erlich, R., Wagle, N., Abel, G.A., Rodig, S.J., Malcovati, L., Galli, A., Travaglino, E., Ambaglio,
H.M., McCarthy, M.I., Boehnke, M., Tuomile- Antin, J.H. & Weinstock, D.M. (2013) Low fre- I., Rizzo, E., Molteni, E., Elena, C., Ferretti,
hto, J., Haiman, C., Groop, L., Atzmon, G., Wil- quency clonal mutations recoverable by deep V.V., Catricala, S., Bono, E., Todisco, G., Bian-
son, J.G., Neuberg, D., Altshuler, D. & Ebert, sequencing in patients with aplastic anemia. chessi, A., Rumi, E., Zibellini, S., Pietra, D.,
B.L. (2014) Age-related clonal hematopoiesis Leukemia, 27, 968–971. Boveri, E., Camaschella, C., Toniolo, D.,
associated with adverse outcomes. New England deLatour, R.P., Mary, J.Y., Salanoubat, C., Terriou, Papaemmanuil, E., Ogawa, S. & Cazzola, M.
Journal of Medicine, 371, 2488–2498. L., Etienne, G., Mohty, M., Roth, S., deGuibert, (2017) Clinical significance of somatic mutation
van Kamp, H., Landegent, J.E., Jansen, R.P., Will- S., Maury, S., Cahn, J.Y. & Socie, G.; French in unexplained blood cytopenia. Blood, 129,
emze, R. & Fibbe, W.E. (1991) Clonal Society of Hematology; French Association of 3371–3378.

ª 2018 British Society for Haematology and John Wiley & Sons Ltd 773
British Journal of Haematology, 2018, 182, 758–776
Review

Manz, C.Y., Nissen, C. & Wodnar-Filipowicz, A. Oni, S.B., Osunkoya, B.O. & Luzzatto, L. (1970) Rosenfeld, S.J., Kimball, J., Vining, D. & Young,
(1996) Deficiency of CD34 + c-kit+ and Paroxysmal nocturnal hemoglobinuria: evidence N.S. (1995) Intensive immunosuppression with
CD34 + 38- hematopoietic precursors in aplastic for monoclonal origin of abnormal red cells. antithymocyte globulin and cyclosporine as
anemia after immunosuppressive treatment. Blood, 36, 145–152. treatment for severe acquired aplastic anemia.
American Journal of Hematology, 52, 264–274. Perrotta, S., Nobili, B., Ferraro, M., Migliaccio, C., Blood, 85, 3058–3065.
Marmont, A.M. (1995) Who really discovered Borriello, A., Cucciolla, V., Martinelli, V., Rossi, Rosenfeld, S., Follmann, D., Nunez, O. & Young,
aplastic anemia? Haematologica, 80, 294. F., Punzo, F., Cirillo, P., Parisi, G., Zappia, V., N.S. (2003) Antithymocyte globulin and cyclos-
Marsh, J.C., Chang, J., Testa, N.G., Hows, J.M. & Rotoli, B. & Della Ragione, F. (2006) Von Hip- porine for severe aplastic anemia: association
Dexter, T.M. (1990) The hematopoietic defect pel-Lindau-dependent polycythemia is endemic between hematologic response and long-term
in aplastic anemia assessed by long-term marrow on the island of Ischia: identification of a novel outcome. JAMA, 289, 1130–1135.
culture. Blood, 76, 1748–1757. cluster. Blood, 107, 514–519. Rotoli, B. & Luzzatto, L. (1989a) Paroxysmal noc-
Marsh, J.C., Chang, J., Testa, N.G., Hows, J.M. & Piccin, A., McCann, S., Socie, G., Oneto, R., Baci- turnal haemoglobinuria. Baillieres Clinical Hae-
Dexter, T.M. (1991) In vitro assessment of mar- galupo, A., Locasciulli, A., Marsh, J., Schrezenmeier, matology, 2, 113–138.
row ‘stem cell’ and stromal cell function in H., Tichelli, A., Hand, E., Lawler, M. & Passweg, J.; Rotoli, B. & Luzzatto, L. (1989b) Paroxysmal noc-
aplastic anaemia. British Journal of Haematology, Aplastic Anaemia Working Party of the European turnal hemoglobinuria. Seminars in Hematology,
78, 258–267. Group for Blood and Marrow Transplantation. 26, 201–207.
Marsh, J.C., Gupta, V., Lim, Z., Ho, A.Y., Ireland, (2010). “Survival of patients with documented Russler-Germain, D.A., Spencer, D.H., Young,
R.M., Hayden, J., Potter, V., Koh, M.B., Islam, autologous recovery after SCT for severe aplastic M.A., Lamprecht, T.L., Miller, C.A., Fulton, R.,
M.S., Russell, N., Marks, D.I., Mufti, G.J. & anemia: a study by the WPSAA of the EBMT.” Bone Meyer, M.R., Erdmann-Gilmore, P., Townsend,
Pagliuca, A. (2011) Alemtuzumab with fludara- Marrow Transplantation 45, 1008–1013. R.R., Wilson, R.K. & Ley, T.J. (2014) The
bine and cyclophosphamide reduces chronic Richichi, B., Luzzatto, L., Notaro, R., la Marca, G. R882H DNMT3A mutation associated with
graft-versus-host disease after allogeneic stem & Nativi, C. (2011) Synthesis of the essential AML dominantly inhibits wild-type DNMT3A
cell transplantation for acquired aplastic anemia. core of the human glycosylphosphatidylinositol by blocking its ability to form active tetramers.
Blood, 118, 2351–2357. (GPI) anchor. Bioorganic Chemistry, 39, 88–93. Cancer Cell, 25, 442–454.
Mathe, G. & Schwarzenberg, L. (1976) Treatment Riether, C., Schurch, C.M. & Ochsenbein, A.F. Saunthararajah, Y., Nakamura, R., Nam, J.M.,
of bone marrow aplasia by mismatched bone (2015) Regulation of hematopoietic and leuke- Robyn, J., Loberiza, F., Maciejewski, J.P., Simo-
marrow transplantation after conditioning with mic stem cells by the immune system. Cell nis, T., Molldrem, J., Young, N.S. & Barrett, A.J.
antilymphocyte globulin–long-term results. Death and Differentiation, 22, 187–198. (2002) HLA-DR15 (DR2) is overrepresented in
Transplantation Proceedings, 8, 595–602. Risitano, A.M. (2017) Immune insights into AA. myelodysplastic syndrome and aplastic anemia
Mathe, G., Hartmann, L., Loverdo, A. & Bernard, Blood, 129, 2824–2826. and predicts a response to immunosuppression
J. (1958) Attempt at protection against radio- Risitano, A.M., Maciejewski, J.P., Green, S., Plasi- in myelodysplastic syndrome. Blood, 100, 1570–
gold-induced mortality by injection of isologous lova, M., Zeng, W. & Young, N.S. (2004) In- 1574.
or homologous bone marrow cells. Revue vivo dominant immune responses in aplastic Saunthararajah, Y., Nakamura, R., Wesley, R.,
Francßaise D’etudes Cliniques et Biologiques, 3, anaemia: molecular tracking of putatively patho- Wang, Q.J. & Barrett, A.J. (2003) A simple
1086–1087. genetic T-cell clones by TCR beta-CDR3 method to predict response to immunosuppres-
Michelozzi, I.M., Pievani, A., Pagni, F., Antolini, sequencing. Lancet, 364, 355–364. sive therapy in patients with myelodysplastic
L., Verna, M., Corti, P., Rovelli, A., Riminucci, Risitano, A.M., Maciejewski, J.P., Muranski, P., syndrome. Blood, 102, 3025–3027.
M., Dazzi, F., Biondi, A. & Serafini, M. (2017) Wlodarski, M., O’Keefe, C., Sloand, E.M. & Scheinberg, P. & Chen, J. (2013) Aplastic anemia:
Human aplastic anaemia-derived mesenchymal Young, N.S. (2005) Large granular lymphocyte what have we learned from animal models and
stromal cells form functional haematopoietic (LGL)-like clonal expansions in paroxysmal noc- from the clinic. Seminars in Hematology, 50,
stem cell niche in vivo. British Journal of Haema- turnal hemoglobinuria (PNH) patients. Leuke- 156–164.
tology, 179, 669–673. mia, 19, 217–222. Scheinberg, P., Marte, M., Nunez, O. & Young,
Montane, E., Ibanez, L., Vidal, X., Ballarin, E., Risitano, A.M., Maciejewski, J.P., Selleri, C. & N.S. (2010) Paroxysmal nocturnal hemoglobin-
Puig, R., Garcia, N. & Laporte, J.R.; Catalan Rotoli, B. (2007) Function and malfunction of uria clones in severe aplastic anemia patients
Group for Study of Agranulocytosis and Aplastic hematopoietic stem cells in primary bone mar- treated with horse anti-thymocyte globulin plus
Anemia. (2008). “Epidemiology of aplastic ane- row failure syndromes. Current Stem Cell cyclosporine. Haematologica, 95, 1075–1080.
mia: a prospective multicenter study.” Haemato- Research & Therapy, 2, 39–52. Scheinberg, P., Nunez, O., Weinstein, B., Schein-
logica 93, 518–523. Risitano, A.M., Selleri, C., Serio, B., Torelli, G.F., berg, P., Biancotto, A., Wu, C.O. & Young, N.S.
Nakao, S., Takamatsu, H., Chuhjo, T., Ueda, M., Kulagin, A., Maury, S., Halter, J., Gupta, V., (2011) Horse versus rabbit antithymocyte globu-
Shiobara, S., Matsuda, T., Kaneshige, T. & Bacigalupo, A., Socie, G., Tichelli, A., Schrezen- lin in acquired aplastic anemia. New England
Mizoguchi, H. (1994) Identification of a specific meier, H., Marsh, J., Passweg, J. & Rotoli, B.; Journal of Medicine, 365, 430–438.
HLA class II haplotype strongly associated with Working Party Severe Aplastic Anaemia Scheinberg, P., Nunez, O., Weinstein, B., Schein-
susceptibility to cyclosporine-dependent aplastic (WPSAA) of the European Group for Blood and berg, P., Wu, C.O. & Young, N.S. (2012) Activ-
anemia. Blood, 84, 4257–4261. Marrow Transplantation (EBMT) (2010). ity of alemtuzumab monotherapy in treatment-
Nishimura Ji, J., Hirota, T., Kanakura, Y., Machii, “Alemtuzumab is safe and effective as immuno- naive, relapsed, and refractory severe acquired
T., Kageyama, T., Doi, S., Wada, H., Masaoka, suppressive treatment for aplastic anaemia and aplastic anemia. Blood, 119, 345–354.
T., Kanayama, Y., Fujii, H., Inoue, N., single-lineage marrow failure: a pilot study and Schrezenmeier, H., Muus, P., Socie, G., Szer, J.,
Kuwayama, M., Inoue, N., Ohishi, K. & a survey from the EBMT WPSAA.” British Jour- Urbano-Ispizua, A., Maciejewski, J.P., Brodsky,
Kinoshita, T. (2002) Long-term support of nal of Haematology 148, 791–796. R.A., Bessler, M., Kanakura, Y., Rosse, W.,
hematopoiesis by a single stem cell clone in Rondelli, T., Berardi, M., Peruzzi, B., Boni, L., Capo- Khursigara, G., Bedrosian, C. & Hillmen, P.
patients with paroxysmal nocturnal hemoglobin- rale, R., Dolara, P., Notaro, R. & Luzzatto, L. (2014) Baseline characteristics and disease bur-
uria. Blood, 99, 2748–2751. (2013) The frequency of granulocytes with sponta- den in patients in the International Paroxysmal
Ogawa, S. (2016) Clonal hematopoiesis in acquired neous somatic mutations: a wide distribution in a Nocturnal Hemoglobinuria Registry. Haemato-
aplastic anemia. Blood, 128, 337–347. normal human population. PLoS ONE, 8, e54046. logica, 99, 922–929.

774 ª 2018 British Society for Haematology and John Wiley & Sons Ltd
British Journal of Haematology, 2018, 182, 758–776
Review

Schultz, J.C. & Shahidi, N.T. (1994) Detection of patient with severe aplastic anemia. Experimental Hallek, M., Thiel, E., Wilhelm, M., Hochsmann,
tumor necrosis factor-alpha in bone marrow Hematology, 4, 131–137. B., Barrois, A., Champion, K. & Passweg, J.R.
plasma and peripheral blood plasma from Speck, B., Buckner, C.D., Cornu, P. & Jeannet, B. (2011) A randomized controlled study in
patients with aplastic anemia. American Journal (1976b) Rationale for the use of ALG as sole patients with newly diagnosed severe aplastic
of Hematology, 45, 32–38. immunosuppressant in allogeneic bone marrow anemia receiving antithymocyte globulin (ATG),
Selleri, C., Sato, T., Anderson, S., Young, N.S. & transplantation for aplastic anemia. Transplanta- cyclosporine, with or without G-CSF: a study of
Maciejewski, J.P. (1995) Interferon-gamma and tion Proceedings, 8, 617–622. the SAA Working Party of the European Group
tumor necrosis factor-alpha suppress both early Speck, B., Gluckman, E., Haak, H.L. & van Rood, for Blood and Marrow Transplantation. Blood,
and late stages of hematopoiesis and induce pro- J.J. (1978) Treatment of aplastic anaemia by 117, 4434–4441.
grammed cell death. Journal of Cellular Physiol- antilymphocyte globulin with or without mar- Townsley, D.M., Scheinberg, P., Winkler, T., Des-
ogy, 165, 538–546. row infusion. Clinics in Haematology, 7, 611– mond, R., Dumitriu, B., Rios, O., Weinstein, B.,
Selleri, C., Maciejewski, J.P., Sato, T. & Young, 621. Valdez, J., Lotter, J., Feng, X., Desierto, M.,
N.S. (1996) Interferon-gamma constitutively Speck, B., Gratwohl, A., Nissen, C., Leibundgut, Leuva, H., Bevans, M., Wu, C., Larochelle, A.,
expressed in the stromal microenvironment of U., Ruggero, D., Osterwalder, B., Burri, H.P., Calvo, K.R., Dunbar, C.E. & Young, N.S. (2017)
human marrow cultures mediates potent Cornu, P. & Jeannet, M. (1981) Treatment of Eltrombopag added to standard immunosup-
hematopoietic inhibition. Blood, 87, 4149–4157. severe aplastic anaemia with antilymphocyte pression for aplastic anemia. New England Jour-
Sergeyeva, A., Gordeuk, V.R., Tokarev, Y.N., Sokol, globulin or bone-marrow transplantation. British nal of Medicine, 376, 1540–1550.
L., Prchal, J.F. & Prchal, J.T. (1997) Congenital Medical Journal (Clinical Research Ed), 282, Tricot, G. & Boogaerts, M.A. (1986) The role of
polycythemia in Chuvashia. Blood, 89, 2148– 860–863. aggressive chemotherapy in the treatment of the
2154. Spyridonidis, A., Kuttler, T., Wasch, R., Samek, E., myelodysplastic syndromes. British Journal of
Shen, W., Clemente, M.J., Hosono, N., Yoshida, Waterhouse, M., Behringer, D., Bertz, H. & Haematology, 63, 477–483.
K., Przychodzen, B., Yoshizato, T., Shiraishi, Y., Finke, J. (2005) Reduced intensity conditioning Wang, H., Chuhjo, T., Yasue, S., Omine, M. &
Miyano, S., Ogawa, S., Maciejewski, J.P. & Mak- compared to standard conditioning preserves Nakao, S. (2002) Clinical significance of a minor
ishima, H. (2014) Deep sequencing reveals step- the in vitro growth capacity of bone marrow population of paroxysmal nocturnal
wise mutation acquisition in paroxysmal stroma, which remains of host origin. Stem Cells hemoglobinuria-type cells in bone marrow fail-
nocturnal hemoglobinuria. The Journal of Clini- and Development, 14, 213–222. ure syndrome. Blood, 100, 3897–3902.
cal Investigation, 124, 4529–4538. Stanley, N., Olson, T.S. & Babushok, D.V. (2017) Wegman-Ostrosky, T. & Savage, S.A. (2017) The
Sloand, E.M. & Barrett, A.J. (2010) Immunosup- Recent advances in understanding clonal haema- genomics of inherited bone marrow failure:
pression for myelodysplastic syndrome: how topoiesis in aplastic anaemia. British Journal of from mechanism to the clinic. British Journal of
bench to bedside to bench research led to suc- Haematology, 177, 509–525. Haematology, 177, 526–542.
cess. Hematology/Oncology Clinics of North Steensma, D.P., Bejar, R., Jaiswal, S., Lindsley, Xie, M., Lu, C., Wang, J., McLellan, M.D., John-
America, 24, 331–341. R.C., Sekeres, M.A., Hasserjian, R.P. & Ebert, son, K.J., Wendl, M.C., McMichael, J.F., Sch-
Sloand, E., Kim, S., Maciejewski, J.P., Tisdale, J., B.L. (2015) Clonal hematopoiesis of indetermi- midt, H.K., Yellapantula, V., Miller, C.A.,
Follmann, D. & Young, N.S. (2002) Intracellular nate potential and its distinction from Ozenberger, B.A., Welch, J.S., Link, D.C., Wal-
interferon-gamma in circulating and marrow T myelodysplastic syndromes. Blood, 126, 9–16. ter, M.J., Mardis, E.R., Dipersio, J.F., Chen, F.,
cells detected by flow cytometry and the Takamatsu, H., Feng, X., Chuhjo, T., Lu, X., Sugi- Wilson, R.K., Ley, T.J. & Ding, L. (2014) Age-
response to immunosuppressive therapy in mori, C., Okawa, K., Yamamoto, M., Iseki, S. & related mutations associated with clonal
patients with aplastic anemia. Blood, 100, 1185– Nakao, S. (2007) Specific antibodies to moesin, hematopoietic expansion and malignancies. Nat-
1191. a membrane-cytoskeleton linker protein, are fre- ure Medicine, 20, 1472–1478.
Smith, J.N., Kanwar, V.S. & MacNamara, K.C. quently detected in patients with acquired aplas- Yoshizato, T., Dumitriu, B., Hosokawa, K., Mak-
(2016) Hematopoietic stem cell regulation by tic anemia. Blood, 109, 2514–2520. ishima, H., Yoshida, K., Townsley, D., Sato-
type I and II interferons in the pathogenesis of Takeda, J., Miyata, T., Kawagoe, K., Iida, Y., Endo, Otsubo, A., Sato, Y., Liu, D., Suzuki, H., Wu,
acquired aplastic anemia. Frontiers in Immunol- Y., Fujita, T., Takahashi, M., Kitani, T. & C.O., Shiraishi, Y., Clemente, M.J., Kataoka, K.,
ogy, 7, 330. Kinoshita, T. (1993) Deficiency of the GPI Shiozawa, Y., Okuno, Y., Chiba, K., Tanaka, H.,
Socie, G. (1996) Could aplastic anaemia be consid- anchor caused by a somatic mutation of the Nagata, Y., Katagiri, T., Kon, A., Sanada, M.,
ered a pre-pre-leukaemic disorder? European PIG-A gene in paroxysmal nocturnal Scheinberg, P., Miyano, S., Maciejewski, J.P.,
Journal of Haematology. Supplementum, 60, 60– hemoglobinuria. Cell, 73, 703–711. Nakao, S., Young, N.S. & Ogawa, S. (2015)
63. Teramura, M., Kimura, A., Iwase, S., Yonemura, Somatic mutations and clonal hematopoiesis in
Socie, G., Rosenfeld, S., Frickhofen, N., Gluckman, Y., Nakao, S., Urabe, A., Omine, M. & Mizogu- aplastic anemia. New England Journal of Medi-
E. & Tichelli, A. (2000) Late clonal diseases of chi, H. (2007) Treatment of severe aplastic ane- cine, 373, 35–47.
treated aplastic anemia. Seminars in Hematology, mia with antithymocyte globulin and Young, N.S. (1992) The problem of clonality in
37, 91–101. cyclosporin A with or without G-CSF in adults: aplastic anemia: Dr Dameshek’s riddle, restated.
Solomou, E.E., Keyvanfar, K. & Young, N.S. a multicenter randomized study in Japan. Blood, Blood, 79, 1385–1392.
(2006) T-bet, a Th1 transcription factor, is up- 110, 1756–1761. Young, N. S. (1994). Drugs and chemicals. In:
regulated in T cells from patients with aplastic Thomas, E.D., Storb, R., Fefer, A., Slichter, S.J., Aplastic Anemia, Acquired and Inherited. (ed.
anemia. Blood, 107, 3983–3991. Bryant, J.I., Buckner, C.D., Neiman, P.E., Clift, by N. S. Young & B. P. Alter), pp. 100–132. WB
Solomou, E.E., Rezvani, K., Mielke, S., Malide, D., R.A., Funk, D.D. & Lerner, K.E. (1972) Aplastic Saunders, Philadelphia, PA.
Keyvanfar, K., Visconte, V., Kajigaya, S., Barrett, anaemia treated by marrow transplantation. Young, N.S. & Maciejewski, J.P. (2000) Genetic
A.J. & Young, N.S. (2007) Deficient CD4 + Lancet, 1, 284–289. and environmental effects in paroxysmal noctur-
CD25 + FOXP3 + T regulatory cells in Tichelli, A., Gratwohl, A., Nissen, C. & Speck, B. nal hemoglobinuria: this little PIG-A goes
acquired aplastic anemia. Blood, 110, 1603–1606. (1994) Late clonal complications in severe aplas- “Why? Why? Why?”. The Journal of Clinical
Speck, B., Cornu, P., Jeannet, M., Nissen, C., tic anemia. Leukaemia & Lymphoma, 12, 167– Investigation, 106, 637–641.
Burri, H.P., Groff, P., Nagel, G.A. & Buckner, 175. Young, N.S., Calado, R.T. & Scheinberg, P. (2006)
C.D. (1976a) Autologous marrow recovery fol- Tichelli, A., Schrezenmeier, H., Socie, G., Marsh, Current concepts in the pathophysiology and treat-
lowing allogeneic marrow transplantation in a J., Bacigalupo, A., Duhrsen, U., Franzke, A., ment of aplastic anemia. Blood, 108, 2509–2519.

ª 2018 British Society for Haematology and John Wiley & Sons Ltd 775
British Journal of Haematology, 2018, 182, 758–776
Review

Zaimoku, Y., Takamatsu, H., Hosomichi, K., Zeng, W., Maciejewski, J.P., Chen, G. & Young, Zhang, J., Niu, C., Ye, L., Huang, H., He, X.,
Ozawa, T., Nakagawa, N., Imi, T., Maruyama, N.S. (2001) Limited heterogeneity of T cell Tong, W.G., Ross, J., Haug, J., Johnson, T.,
H., Katagiri, T., Kishi, H., Tajima, A., Mura- receptor BV usage in aplastic anemia. The Jour- Feng, J.Q., Harris, S., Wiedemann, L.M., Mis-
guchi, A., Kashiwase, K. & Nakao, S. (2017) nal of Clinical Investigation, 108, 765–773. hina, Y. & Li, L. (2003) Identification of the
Identification of an HLA class I allele closely Zeng, W., Chen, G., Kajigaya, S., Nunez, O., haematopoietic stem cell niche and control of
involved in the autoantigen presentation in Charrow, A., Billings, E.M. & Young, N.S. the niche size. Nature, 425, 836–841.
acquired aplastic anemia. Blood, 129, 2908– (2004) Gene expression profiling in CD34 cells Zoumbos, N.C., Gascon, P., Djeu, J.Y. & Young,
2916. to identify differences between aplastic anemia N.S. (1985a) Interferon is a mediator of
Zeng, W., Nakao, S., Takamatsu, H., Yachie, A., patients and healthy volunteers. Blood, 103, hematopoietic suppression in aplastic anemia
Takami, A., Kondo, Y., Sugimori, N., Yamazaki, 325–332. in vitro and possibly in vivo. Proceedings of the
H., Miura, Y., Shiobara, S. & Matsuda, T. (1999) Zeng, Q., Wang, H., Chang, H. & Liu, T. (2015) National Academy of Sciences of the United States
Characterization of T-cell repertoire of the bone Relationship between single nucleotide polymor- of America, 82, 188–192.
marrow in immune-mediated aplastic anemia: phism of interferon-gamma Gene +874 T/A and Zoumbos, N.C., Gascon, P., Djeu, J.Y., Trost, S.R.
evidence for the involvement of antigen-driven efficacy of immunosuppressive therapy in & Young, N.S. (1985b) Circulating activated
T-cell response in cyclosporine-dependent aplas- patients with aplastic anemia. Zhongguo Shi Yan suppressor T lymphocytes in aplastic anemia.
tic anemia. Blood, 93, 3008–3016. Xue Ye Xue Za Zhi, 23, 1375–1379. New England Journal of Medicine, 312, 257–265.

776 ª 2018 British Society for Haematology and John Wiley & Sons Ltd
British Journal of Haematology, 2018, 182, 758–776

You might also like